Brain Glutamate Metabolism

Download as pdf or txt
Download as pdf or txt
You are on page 1of 20

HHS Public Access

Author manuscript
Adv Neurobiol. Author manuscript; available in PMC 2015 December 02.
Author Manuscript

Published in final edited form as:


Adv Neurobiol. 2014 ; 11: 13–30. doi:10.1007/978-3-319-08894-5_2.

Glutamate Metabolism in the Brain Focusing on Astrocytes


Arne Schousboe1, Susanna Scafidi2, Lasse K. Bak1, Helle S. Waagepetersen1, and Mary C.
McKenna3,#
1Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences,
University of Copenhagen, DK-2100 Copenhagen, Denmark
2Department of Anesthesiology and Critical Care Medicine, Johns Hopkins University School of
Author Manuscript

Medicine, Baltimore, MD 21287, USA


3Department of Pediatrics and Program in Neuroscience, University of Maryland School of
Medicine, Baltimore, MD 21201, USA

Abstract
Metabolism of glutamate, the main excitatory neurotransmitter and precursor of GABA, is
exceedingly complex and highly compartmentalized in brain. Maintenance of these
neurotransmitter pools is strictly dependent on the de novo synthesis of glutamine in astrocytes
which requires both the anaplerotic enzyme pyruvate carboxylase and glutamine synthetase.
Glutamate is formed directly from glutamine by deamidation via phosphate activated glutaminase
a reaction that also yields ammonia. Glutamate plays key roles linking carbohydrate and amino
acid metabolism via the tricarboxylic acid (TCA) cycle, as well as in nitrogen trafficking and
Author Manuscript

ammonia homeostasis in brain. The anatomical specialization of astrocytic endfeet enables these
cells to rapidly and efficiently remove neurotransmitters from the synaptic cleft to maintain
homeostasis, and to provide glutamine to replenish neurotransmitter pools in both glutamatergic
and GABAergic neurons. Since the glutamate-glutamine cycle is an open cycle that actively
interfaces with other pathways, the de novo synthesis of glutamine in astrocytes helps to maintain
the operation of this cycle. The fine-tuned biochemical specialization of astrocytes allows these
cells to respond to subtle changes in neurotransmission by dynamically adjusting their anaplerotic
and glycolytic activities, and adjusting the amount of glutamate oxidized for energy relative to
direct formation of glutamine, to meet the demands for maintaining neurotransmission. This
chapter summarizes the evidence that astrocytes are essential and dynamic partners in both
glutamatergic and GABAergic neurotransmission in brain.
Author Manuscript

Keywords
Glutamate; Glutamine; GABA; Metabolic compartmentation; Astrocytes; Glutamate-glutamine
cycle; Pyruvate carboxylase; Glutamine synthetase; Phosphate activated glutaminase; Glutamate
dehydrogenase

#
Corresponding author: Prof. Mary C. McKenna, Department of Pediatrics and Program in Neuroscience, University of Maryland
School of Medicine, 655 W. Baltimore St., Room 13-019, Baltimore, MD 21201, USA, Phone: 410-706-1990, FAX: 410-706-2611,
[email protected].
Schousboe et al. Page 2

Introduction
Author Manuscript

Glutamate plays a key role in intermediary metabolism in all organs and tissues linking
carbohydrate and amino acid metabolism via the tricarboxylic acid (TCA) cycle. The reason
is that it serves as a co-substrate in all reactions catalyzed by aminotransferases being
converted to α-ketoglutarate, a key intermediate in the TCA cycle (McKenna et al. 2012). In
the brain, glutamate metabolism extends beyond this general view as it serves as the
immediate precursor for γ-aminobutyric acid (GABA) which is formed by decarboxylation
of glutamate catalyzed by glutamate decarboxylase (GAD) as first shown by Roberts and
Frankel (1950). The fact that GABA is metabolized by GABA-transaminase (GABA-T) and
further to the TCA cycle intermediate succinate, catalyzed by succinic semialdehyde
dehydrogenase, provides a deviation of the TCA cycle reactions called the GABA-shunt
which circumvents succinyl CoA (see Schousboe et al. 2013). As both glutamate and GABA
serve dual roles in the brain as metabolites and important neurotransmitters mediating
Author Manuscript

excitatory and inhibitory signals, respectively (for references see Schousboe et al. 2013,
Schousboe et al. 2012), their metabolic pathways are of significant interest. The immediate
precursor for neuronal synthesis of glutamate is glutamine. This reaction is catalyzed by
phosphate activated glutaminase (PAG) which hydrolytically deamidates glutamine to form
glutamate and ammonia (for further details, see below). Interestingly, this enzymatic
reaction was extensively investigated by Krebs (1935) in several tissues including the brain.
Later, detailed studies of glutamate and glutamine metabolic pathways in the brain
performed in the laboratories of H. Waelsch and J.H. Quastel (e.g. Quastel 1975, Berl &
Clarke 1969) provided evidence that glutamate metabolism in the brain is extremely
complex. It was noted that using radioactively labeled glucose as a precursor higher specific
radioactivity was seen in glutamate; whereas, with the radioactive precursors leucine, acetate
and bicarbonate, glutamine exhibited a higher specific radioactivity than its precursor
Author Manuscript

glutamate (for references, see Berl & Clarke 1969, Quastel 1975). A higher specific
radioactivity in a compound (e.g. glutamine) than that seen in its precursor (e.g. glutamate)
indicates that the precursor exists in separate metabolic pools having different turn-over rates
or in other words this is referred to as “metabolic compartmentation” (for further details, see
McKenna et al. 2012). This finding led to the concept of metabolic compartmentation of
glutamate in the brain, with at least two compartments (van den Berg & Garfinkel 1971,
Garfinkel 1966) which were subsequently defined as representing neurons and astrocytes.
This concept is based on the finding that glutamine synthetase (GS), the enzyme that
converts glutamate to glutamine, is exclusively localized in astrocytes (Norenberg &
Martinez-Hernandez 1979), together with the finding that higher specific radioactivity in
glutamine is observed with the precursors acetate, bicarbonate and leucine (see above). It is
of interest that the de novo synthesis of glutamate, the precursor of glutamine, is also
Author Manuscript

restricted to astrocytes as the anaplerotic enzyme pyruvate carboxylase (PC) is exclusively


localized in astrocytes (Shank et al. 1985, Yu et al. 1983). This will be discussed in further
detail below.

Adv Neurobiol. Author manuscript; available in PMC 2015 December 02.


Schousboe et al. Page 3

Enzymatic reactions involving glutamate as substrate or product


Author Manuscript

Aspartate aminotransferase
This enzyme catalyzing the reversible interconversion of aspartate, α-ketoglutarate,
oxaloacetate and glutamate (Fig.1) is present in all tissues, and has the highest specific
activity among aminotransferases in the brain (for references, see Cooper 1988). The
equilibrium constant for the enzyme is close to unity (Krebs 1953), hence, the reaction
proceeds easily in both directions and is generally considered an exchange reaction. This is
important considering the fact that the two keto acid substrates (oxaloacetate and α-
ketoglutarate) are constituents of the TCA cycle. Thus, the corresponding amino acids
aspartate and glutamate are in equilibrium with these TCA cycle intermediates and reflect
the metabolic status of the keto acids at any given time due to the high activity of this
enzyme (for references, see McKenna et al. 2012). This forms the basis for using the
appearance of labeled carbon in glutamate and aspartate, from labeled precursors that
Author Manuscript

provide substrates for the TCA cycle as a surrogate for determining the activity of the TCA
cycle (see, McKenna et al. 2012). It should be noted, that aspartate aminotransferase (AAT)
can reversibly bind to the inner mitochondrial membrane, which influences the functional
activity of this enzyme (for futher details, see McKenna et al. 2000, 2006).

Alanine aminotransferase
This aminotransferase catalyzes the reversible interconversion of alanine, α-ketoglutarate,
pyruvate and glutamate (Fig.1) and its activity in the brain as well as in cultured brain cells
is two orders of magnitude lower than that of aspartate aminotransferase (Benuck et al.
1972, Larsson et al. 1985, Westergaard et al. 1993). The equilibrium constant for alanine
aminotransferase (ALAT) is close to unity (Krebs 1953). Since pyruvate, one of the
substrates for this enzyme is a product of glycolysis, ALAT couples the glycolytic pathway
Author Manuscript

to amino acid metabolism and carbon label from glucose can be monitored by its appearance
in alanine, which has a slower turnover rate than pyruvate. This provides a method to
monitor glycolytic activity although it should be kept in mind that this relationship is not a
direct correlation since the pyruvate pool has been shown to be compartmentalized (for
further details, see Bakken et al. 1998, Obel et al. 2012, Waagepetersen et al. 2000).

Alanine aminotransferase is present in both astrocytes and neurons and may have a role in
transfer of ammonia nitrogen between these cells as suggested by Waagepetersen et al.
(2000). The ALAT reaction in concert with glutamate dehydrogenase (GDH) may be a
particularly important mechanism for fixation of ammonia during hyperammonemic
conditions when the normal activity of glutamine synthetase (see below) is inhibited
(Dadsetan et al. 2011, 2013). This aspect will be discussed in further detail below.
Author Manuscript

Branched chain amino acid aminotransferase


The three branched chain amino acids (valine, leucine and isoleucine) are metabolized by
transamination with α-ketoglutarate, catalyzed by a common aminotransferase forming
glutamate (Fig.1) and the three keto acids α-ketoisovalerate, α-ketoisocaproate and α-keto-
β-methylvalerate, respectively (Cooper 1988). The branched chain amino acid
aminotransferase isozymes (BCATs) are compartmentalized in brain cells; with the

Adv Neurobiol. Author manuscript; available in PMC 2015 December 02.


Schousboe et al. Page 4

mitochondrial form being selectively localized in astrocytes and the cytosolic form localized
in neurons (see, Lieth et al. 2001). This selective localization plays an important functional
Author Manuscript

role as the branched chain amino acids also mediate shuttling of ammonia nitrogen between
astrocytes and neurons (Bak et al. 2013, Lieth et al. 2001). This reaction may also contribute
the amino group for de novo glutamate synthesis in astrocytes, which requires CO2 fixation
(Lieth et al. 2001). The branched chain amino acids have also been proposed to play a role
in removal of ammonia during hyperammonemic conditions as seen in hepatic
encephalopathy (Ott et al. 2005). Additionally, since these amino acids ultimately are
metabolized to propionyl-CoA, acetyl-CoA, succinyl-CoA and acetoacetate, the latter three
compounds can fuel the TCA cycle and facilitate glutamine production during
hyperammonemia thereby ameliorating some of the excess ammonia (Ott et al. 2005). This
functional role has been questioned since studies using isolated neurons and astrocytes
suggest that the branched chain amino acids are only modestly metabolized in brain (see,
Bak et al. 2013). However, it should be noted that supplementation with branched chain
Author Manuscript

amino acids was neuroprotective after traumatic brain injury (Cole et al. 2010).

Glutamate dehydrogenase
This mitochondrially localized (Salganicoff & Derobertis 1965) enzyme catalyzes the redox-
based interconversion of glutamate and α-ketoglutarate using NAD(P)+ as coenzyme (Fig.1).
The thermodynamic equilibrium constant (6 × 10−15 M) favors the reductive amination of
glutamate (Engel & Dalziel 1967). However, in the brain the normal direction of the reaction
favors oxidative deamination of glutamate since the NAD+/NADH ratio is high and the Km,
for ammonia (14–26 mM) is orders of magnitude higher than the prevailing ammonia
concentration (Zaganas et al. 2001, 2009). The enzyme is highly regulated by ADP and
leucine both of which function as allosteric activators and by GTP, which acts as an
allosteric inhibitor (Spanaki et al. 2012). Emerging evidence suggests that GDH can be
Author Manuscript

allosterically inhibited by mitochondrial SIRT4 (silent information regulator 4) that is highly


expressed during brain development (Komlos et al. 2013, Lavu et al. 2008). Humans express
two isoforms of the enzyme (GDH1 and GDH2) that differ dramatically with regard to the
allosteric regulation; other mammals express only the housekeeping isoform, GDH1
(Spanaki et al. 2012). The enzyme is present in both neurons and astrocytes albeit the
expression may be higher in astrocytes (Lovatt et al. 2007), particularly those from brain
regions with high glutamatergic activity (Aoki et al. 1987).

As pointed out above, glutamate dehydrogenase in combination with aminotransferases is


important for transferring ammonia to form amino acids from keto acids. Since reductive
amination can only occur when the ammonia concentration approaches the Km value for
ammonia, it is likely that enzymatic complexes between GDH and the respective
Author Manuscript

aminotransferases must exist to facilitate this reaction in vivo and there is some experimental
evidence to suggest that this is indeed the case (Fahien et al. 1977, Islam et al. 2010,
McKenna 2011).

Phosphate activated glutaminase


This enzyme which hydrolyzes glutamine to glutamate (Fig.1) was first described by Krebs
(1935), and its phosphate dependence was reported shortly thereafter by Errera and

Adv Neurobiol. Author manuscript; available in PMC 2015 December 02.


Schousboe et al. Page 5

Greenstein (1949). The name, phosphate activated glutaminase or PAG, originates from this
latter publication. The brain enzyme has been purified and extensively characterized by
Author Manuscript

Kvamme and co-workers and is highly enriched in neurons (for review, see Kvamme et al.
2001). Using a preparation of cultured astrocytes, which may reflect the properties of these
cells in situ (Lange et al. 2012), it was shown by Schousboe et al. (1979) that this enzyme is
also found in astrocytes. This finding has been controversial but a transcriptomic analysis of
acutely isolated astrocytes has recently confirmed that PAG is indeed expressed by
astrocytes (Lovatt et al. 2007). PAG is essential for synthesis of transmitter glutamate in
glutamatergic neurons and it also plays a significant role in the synthesis of neurotransmitter
GABA, since NMR studies revealed that glutamine synthesized in astrocytes and
deamidated by neuronal PAG is preferentially used for GABA synthesis (see, Schousboe et
al. 2013, Sonnewald et al. 1993c).

Glutamate decarboxylase
Author Manuscript

The presence of GABA in the brain and its synthesis by α-decarboxylation of glutamate (see
Fig.1) was first described by Roberts and Frankel (1950). An enzyme, glutamic acid
decarboxylase, catalyzing the conversion of glutamate to GABA was first identified in plants
and bacteria. Therefore, it was concluded that formation of GABA from glutamate by α-
decarboxylation in the brain was likely to be catalyzed by this enzyme. Glutamic acid
decarboxylase (GAD) was finally purified to homogeneity from mouse brain more than
twenty years later by Wu et al. (1973). Cloning studies subsequently showed that GAD
exists in two isoforms with molecular weights of 65 kD and 67 kD; these isozymes are
referred to as GAD65 and GAD67, respectively (for references, see Soghomonian & Martin
1998). In the brain these GAD isozymes are restricted to neurons and primarily GABAergic
cells (Saito et al. 1974). The enzymes are also expressed in other tissues such as the pancreas
(Okada et al. 1976). Both isoforms of the enzyme are located in the cytosol, but their
Author Manuscript

subcellular localization differs, with the GAD67 isoform having a widespread expression
compatible with a function related to general metabolism of the GABA pool; whereas the
GAD65 isoform is mainly present in nerve endings and thought to be involved in the
synthesis of neurotransmitter GABA (Walls et al. 2011, Martin & Rimvall 1993).

Glutamine synthetase
As already pointed out, glutamate metabolism in the brain is highly complex and
compartmentalized with different enzymes located in different cell types. The enzyme
glutamine synthetase (GS) is responsible for conversion of glutamate to glutamine (Fig.1)
and is expressed exclusively in astrocytes (Norenberg & Martinez-Hernandez 1979). The
functional importance of GS can most conveniently be assessed by use of the irreversible
inhibitor methionine sulfoximine (MSO; Ronzio et al. 1969). The GS reaction adds a second
Author Manuscript

nitrogen atom in the form of ammonia to glutamate to form glutamine and requires ATP for
energy (Fig 1). The Km values for the three substrates glutamate, ammonia and ATP are 2.5
mM, 0.2 mM and 2.3 mM, respectively (Pamiljans et al. 1962), which means that the
enzyme in situ is unlikely to be saturated with any of the substrates (see, Schousboe et al.
2012).

Adv Neurobiol. Author manuscript; available in PMC 2015 December 02.


Schousboe et al. Page 6

One of the most essential roles of astrocytes is removal of neurotransmitter glutamate from
the synaptic cleft after depolarization (to maintain the low resting glutamate concentration of
Author Manuscript

1–10 µM) and conversion to glutamine (Bergles et al. 1999, Matsui et al. 2005). It should be
noted, that the de novo synthesis of glutamine takes place only in astrocytes. De novo
synthesis of glutamine from glucose requires the concerted action of glycolytic enzymes,
pyruvate carboxylase, the TCA cycle and conversion of α-ketoglutarate to glutamate, which
via the action of GS can be converted to glutamine (Fig. 2). Pyruvate carboxylase like GS is
dependent on ATP and is only expressed in astrocytes (Yu et al. 1983). Pyruvate
carboxylation adds a carbon to pyruvate to form oxaloacetate and serves to replenish
intermediates that would otherwise be drained from the TCA cycle by the conversion of α-
ketoglutarate to glutamate and subsequently glutamine. 13C-NMR studies show
compartmentation in this process as the carbon added via pyruvate carboxylase is
preferentially found in newly synthesized glutamine (Sonnewald et al. 1993a). Due to the
selective cellular localization of glutamine synthetase and pyruvate carboxylase only
Author Manuscript

astrocytes are able to perform a net synthesis of glutamine, which is the major precursor for
the two most important neurotransmitters, glutamate and GABA (see, McKenna 2007,
McKenna et al. 2012). A further discussion of this important aspect of brain function based
on cellular specialization is provided below.

Overview of the glutamate-glutamine cycle and its role in glutamatergic


neurotransmission
Based on a series of elegant studies of radioactive labeling of glutamate and glutamine using
different 14C-labeled precursors such as glucose, acetate and bicarbonate it was concluded
that the brain contains at least two separate cellular compartments of these amino acids each
having a distinct turn-over of the amino acid pools (for references, see McKenna et al. 2012,
Author Manuscript

Schousboe 2012). The seminal demonstration of the astrocyte specific localization of


glutamine synthetase (Norenberg & Martinez-Hernandez 1979) together with the multiple
compartments of glutamate revealed by the labeling studies, led to the firm conclusion that
the proposed glutamate-glutamine cycle (see Figure 3) must be the mechanism allowing
inter-cellular exchange of these amino acids (see, Öz et al. 2012, and Schousboe et al. 2012
for references). The selective cellular localization of specific glutamate and glutamine
transporters on neuronal and glial plasma membranes is consistent with the functional
importance of this cycle (see, Schousboe et al. 2012).

When the concept of the glutamate-glutamine cycle was first introduced it was thought that
essentially all glutamate being released as neurotransmitter would be transferred to the
astrocytes where it would quantitatively be converted to glutamine and subsequently
Author Manuscript

transferred back to the glutamatergic neurons (Cotman et al. 1981). The fact that exogenous
glutamate taken up by astrocytes can be oxidatively metabolized (see below for further
details) demonstrates that the glutamate-glutamine cycle does not operate in a stoichiometric
manner as first proposed (see, McKenna 2007, McKenna et al. 2012). If the glutamate-
glutamine cycle exclusively operated in a stoichiometric fashion, then none of the glutamate
taken up would be used to offset the high energy cost of glutamate transport into astrocytes,

Adv Neurobiol. Author manuscript; available in PMC 2015 December 02.


Schousboe et al. Page 7

i.e. 3 ATP are required for transport of each glutamate molecule and activity of Na+-K+-
ATPase to restore the sodium gradient (Attwell & Laughlin 2001, McKenna 2013).
Author Manuscript

Since the glutamate-glutamine cycle is an open cycle that loses intermediates to other
pathways, the de novo synthesis of glutamine in astrocytes helps to maintain the operation of
this cycle. Numerous studies have been aimed at obtaining a quantitative measure of the
anaplerotic de novo synthesis of glutamate and glutamine; this information has been
summarized by Öz et al. (2012). The rate of anaplerosis mediated by pyruvate carboxylase
ranges from 6 to 35 % of the rate of the TCA cycle and fulfills the need for de novo
synthesis of glutamine (Öz et al. 2004, 2012). Since glutamine also serves as the main
precursor for GABA (Sonnewald et al. 1993c) and some of the GABA released by neurons
is taken up and metabolized by astrocytes, maintenance of GABAergic neurotransmission
also relies on anaplerosis (Schousboe et al. 2013).
Author Manuscript

Influence of the concentration of glutamate on the metabolic pathways for


glutamate metabolism
Early studies of glutamate metabolism in astrocytes using tracer concentrations of 14C-
labeled precursors reported a high proportion of the glutamate was converted to glutamine
(Farinelli & Nicklas 1992, Zielke et al. 1990). However, a study by Yu et al. (1982) showed
that a substantial fraction of exogenous glutamate was metabolized via GDH and
subsequently oxidized to CO2 in astrocytes. Sonnewald et al. (1993b) using 13C-NMR
spectroscopy reported that a significant proportion of the label from glutamate metabolism
in astrocytes was found in lactate. This labeling can only occur when glutamate is converted
to α-ketoglutarate and further metabolized via the TCA cycle to malate, which can be
converted to pyruvate and subsequently to lactate by a partial pyruvate recycling pathway
Author Manuscript

(Fig. 4). These studies confirmed that a significant amount of glutamate was oxidatively
metabolized for energy in astrocytes (Sonnewald et al. 1993b, McKenna 2013). McKenna et
al. (1996) demonstrated that when the exogenous glutamate concentration was increased
from 0.1 mM to 0.5 mM the proportion of glutamate oxidized by the TCA cycle in
astrocytes greatly increased (from ~ 15% to 43%) and the percent converted to glutamine
decreased correspondingly (from ~ 85% to 57%). This demonstrated that the metabolic fate
of glutamate was shifted away from conversion to glutamine and towards oxidative
metabolism as the concentration of glutamate in the extracellular milieu increased. Since the
amount of glutamate in the synaptic cleft increases several orders of magnitude during
depolarization (from ~ 10 µM to 1 mM) this provides astrocytes with the metabolic
flexibility to readily form ATP from glutamate oxidation in the TCA cycle which offsets the
high cost of glutamate transport into astrocytes (for further details, see McKenna 2013).
Author Manuscript

Recent studies show that the astrocyte glutamate transporter GLT-1 (in humans EAAT2)
forms a complex with other proteins including hexokinase, several specific mitochondrial
proteins including GDH and mitochondria that together serves to effectively channel
glutamate towards energy producing oxidative metabolism rather than the energy utilizing
reaction of conversion to glutamine in the cytosol (Genda et al. 2011).

Adv Neurobiol. Author manuscript; available in PMC 2015 December 02.


Schousboe et al. Page 8

Glutamate and ammonia homeostasis under normal and hyperammonemic


Author Manuscript

conditions
Physiological conditions
In glutamatergic and to some extent in GABAergic neurons ammonia is generated by the
action of PAG, which removes the amido nitrogen from glutamine to form glutamate and
subsequently GABA in GABAergic neurons (Fig. 5). Since GS, the primary enzyme capable
of ammonia fixation in the brain, is located in astrocytes there needs to be a way by which
ammonia nitrogen can be transferred between the neurons and the surrounding astrocytes.
The specific mechanism(s) for returning ammonia removed by PAG in neurons to astrocytes
has not been defined. For years it was assumed that the ammonium ion (being the prevailing
form of ammonia) can diffuse freely through plasma membranes or perhaps through
channels (Cooper & Plum 1987). Recently, two shuttle mechanisms based on exchange of an
Author Manuscript

amino acid have been proposed (Lieth et al. 2001, Waagepetersen et al. 2000, Yudkoff et al.
1996, Zwingmann et al. 2000). Both models operate on the basis of ammonia being fixed
into the amino group of glutamate by the action of glutamate dehydrogenase in neurons, and
subsequently being transferred to either alanine or a branched chain amino acid, which
serves to return the amino group to astrocytes (Fig. 5). In astrocytes the amino groups are
transferred to glutamate by the respective aminotransferases (see above and Fig. 1) forming
branched chain keto acids and pyruvate from the branch chain amino acids and alanine,
respectively. The amino group transferred to glutamate is subsequently removed by
oxidative deamination via GDH making it available to glutamine synthetase for glutamine
formation (see Figs. 1 and 5). A careful analysis of these mechanisms by modeling the rates
of the reactions involved has, however, failed to provide evidence that the activity of these
amino acid shuttles would be sufficient to match the actual need for transfer of ammonia
Author Manuscript

back to astrocytes (Rothman et al. 2012). Hence, at the present time the quantitative
importance of these proposed mechanisms for ammonia transfer in vivo has not been
resolved.

Hyperammonemia
The most efficient pathway for ammonia fixation in the brain is the GS catalyzed conversion
of glutamate to glutamine and a classical experiment by Cooper et al. (1979) using infusion
of trace amounts of [13N]ammonia into the brain showed that at least 98% of the labeled
ammonia could be found in the amide group of glutamine within seconds. When GS was
inhibited by MSO, ammonia radioactivity was recovered in glutamate, albeit the amounts
were small. This does, however, show that under appropriate conditions reductive amination
mediated by GDH can occur (see above). In this context, it may be noted that during
Author Manuscript

hyperammonemic conditions in neuronal-astrocytic co-cultures, the formation of both


alanine and glutamine is increased (Leke et al. 2011). As pointed out above, de novo
synthesis of glutamine requires CO2 fixation by pyruvate carboxylase to form oxaloacetate;
interestingly, hyperammonemic conditions have been shown to stimulate this pathway (Leke
et al. 2011). On the other hand, when GS was inhibited and ammonia was fixed in alanine,
the glycolytic pathway was accelerated while the anaplerotic pathway did not increase
(Dadsetan et al. 2011, 2013). This shows that astrocytes are able to switch back and forth

Adv Neurobiol. Author manuscript; available in PMC 2015 December 02.


Schousboe et al. Page 9

between anaplerosis and glycolysis depending on whether pyruvate or a TCA cycle


intermediate is needed. This concept has been confirmed in cultures and in the brain in vivo
Author Manuscript

since inhibition of GS by MSO during hyperammonemic conditions leads to a decrease in


glutamine and an increase in alanine production (Dadsetan et al. 2011, 2013, Fries et al.
2013). This may be due in part to the fact that alanine does not function as an intracellular
osmolyte in astrocytes, whereas glutamine does perform this function. It has been proposed
that inhibition of GS may be a potentially useful therapeutic strategy to prevent the
glutamine-associated glial swelling and the brain edema observed in hepatic encephalopathy
(Brusilow et al. 2010, Dadsetan et al. 2011, 2013).

Concluding remarks
It should be noted that glutamate metabolism is exceedingly complex and highly
compartmentalized. Maintenance of the glutamate and GABA neurotransmitter pools is
Author Manuscript

strictly dependent on the de novo synthesis of glutamine in astrocytes. The exquisite


anatomical specialization of astrocytic endfeet enables these cells to rapidly and efficiently
remove neurotransmitters from the synaptic cleft and provide glutamine to replenish
neurotransmitter pools in glutamatergic and GABAergic neurons. This is possible due to the
fine-tuned biochemical machinery including the distribution of pertinent transporters, as
well as the presence of mitochondria and specific cytosolic enzymes in the astrocytic
processes which ensheathe the synapses. This allows astrocytes to respond to subtle changes
in neurotransmission by dynamically adjusting their anaplerotic and glycolytic activities to
meet the demands for maintaining neurotransmission. Additionally, astrocytes can readily
shift the metabolic fate of glutamate removed from the synaptic cleft towards increased
oxidative energy metabolism relative to direct formation of glutamine.

In conclusion, this chapter has summarized the evidence that astrocytes are dynamic partners
Author Manuscript

in both glutamatergic and GABAergic neurotransmission in brain. Protoplasmic astrocytes


cover and actively interact with hundreds of thousands to millions synapses in human brain
(Oberheim et al. 2006, 2009). Hence, astrocytes are essential partners in neurotransmission
since they continuously monitor the extracellular milieu and modulate their metabolic
activity in response to fluctuations in neuronal activity.

References
Aoki C, Milner TA, Berger SB, Sheu KF, Blass JP, Pickel VM. Glial glutamate dehydrogenase:
ultrastructural localization and regional distribution in relation to the mitochondrial enzyme,
cytochrome oxidase. J. Neurosci. Res. 1987; 18:305–318. [PubMed: 2826798]
Attwell D, Laughlin SB. An energy budget for signaling in the grey matter of the brain. J. Cereb.
Blood Flow Metab. 2001; 21:1133–1145. [PubMed: 11598490]
Author Manuscript

Bak LK, Waagepetersen HS, Sorensen M, Ott P, Vilstrup H, Keiding S, Schousboe A. Role of
branched chain amino acids in cerebral ammonia homeostasis related to hepatic encephalopathy.
Metab. Brain Dis. 2013; 28:209–215. [PubMed: 23371316]
Bakken IJ, White LR, Aasly J, Unsgard G, Sonnewald U. [U-13C]Aspartate metabolism in cultured
cortical astrocytes and cerebellar granule neurons studied by NMR spectroscopy. Glia. 1998;
23:271–277. [PubMed: 9633811]
Benuck M, Stern F, Lajtha A. Regional and subcellular distribution of aminotransferases in rat brain. J.
Neurochem. 1972; 19:949–957. [PubMed: 5019591]

Adv Neurobiol. Author manuscript; available in PMC 2015 December 02.


Schousboe et al. Page 10

Bergles DE, Diamond JS, Jahr CE. Clearance of glutamate inside the synapse and beyond. Curr. Opin
Neurobiol. 1999; 9:293–298. [PubMed: 10395570]
Author Manuscript

Berl, S.; Clarke, DD. Compartmentation of amino acid metabolism. In:. In: Lajtha, A., editor.
Handbook of neurochemistry. Vol. 2. New York, USA: Plenum Publishing Corporation; 1969. p.
447-472.
Brusilow SW, Koehler RC, Traystman RJ, Cooper AJ. Astrocyte glutamine synthetase: importance in
hyperammonemic syndromes and potential target for therapy. Neurotherapeutics. 2010; 7:452–470.
[PubMed: 20880508]
Cole JT, Mitala CM, Kundu S, Verma A, Elkind JA, Nissim I, Cohen AS. Dietary branched chain
amino acids ameliorate injury-induced cognitive impairment. Proc. Nat. Acad. Sci. U.S.A. 2010;
107:366–371.
Cooper, AJ. L-Glutamate(2-oxoglutarate) aminostransferases. In: Kvamme, E., editor. Glutamine and
glutamate in mammals. Vol. I. Boca Raton, FL, USA: CRC Press; 1988. p. 123-152.
Cooper AJ, McDonald JM, Gelbard AS, Gledhill RF, Duffy TE. The metabolic fate of 13N-labeled
ammonia in rat brain. J. Biol. Chem. 1979; 254:4982–4992. [PubMed: 36379]
Cooper AJ, Plum F. Biochemistry and physiology of brain ammonia. Physiol. Rev. 1987; 67:440–519.
Author Manuscript

[PubMed: 2882529]
Cotman CW, Foster A, Lanthorn T. An overview of glutamate as a neurotransmitter. Advan. Biochem.
Psychopharm. 1981; 27:1–27.
Dadsetan S, Bak LK, Sorensen M, Keiding S, Vilstrup H, Ott P, Leke R, Schousboe A, Waagepetersen
HS. Inhibition of glutamine synthesis induces glutamate dehydrogenase-dependent ammonia
fixation into alanine in co-cultures of astrocytes and neurons. Neurochem. Int. 2011; 58:482–488.
[PubMed: 21397649]
Dadsetan S, Kukolj E, Bak LK, Sorensen M, Ott P, Vilstrup H, Schousboe A, Keiding S,
Waagepetersen HS. Brain alanine formation as an ammonia-scavenging pathway during
hyperammonemia: effects of glutamine synthetase inhibition in rats and astrocyte-neuron co-
cultures. J. Cereb. Blood Flow Metab. 2013; 33:1235–1241. [PubMed: 23673435]
Engel PC, Dalziel K. The equilibrium constants of the glutamate dehydrogenase systems. Biochem. J.
1967; 105:691–695. [PubMed: 4384597]
Errera M, Greenstein JP. Phosphate-activated glutaminase in kidney and other tissues. J. Biol. Chem.
1949; 178:495–502. [PubMed: 18112132]
Author Manuscript

Fahien LA, Hsu SL, Kmiotek E. Effect of aspartate on complexes between glutamate dehydrogenase
and various aminotransferases. J. Biol. Chem. 1977; 252:1250–1256. [PubMed: 14147]
Farinelli SE, Nicklas WJ. Glutamate metabolism in rat cortical astrocyte cultures. J. Neurochem. 1992;
58:1905–1915. [PubMed: 1348525]
Fries A, Dadsetan S, Keiding S, et al. Effect of glutamine synthetase inhibition on brain and interorgan
ammonia metabolism in bile duct ligated rats. J. Cereb. Blood Flow Metab. 2013 In Press.
Garfinkel D. A simulation study of the metabolism and compartmentation in brain of glutamate,
aspartate, the Krebs cycle, and related metabolites. J. Biol. Chem. 1966; 241:3918–3929.
[PubMed: 5920803]
Genda EN, Jackson JG, Sheldon AL, et al. Co-compartmentalization of the astroglial glutamate
transporter, GLT-1, with glycolytic enzymes and mitochondria. J. Neurosci. 2011; 31:18275–
18288. [PubMed: 22171032]
Islam MM, Nautiyal M, Wynn RM, Mobley JA, Chuang DT, Hutson SM. Branched-chain amino acid
metabolon: interaction of glutamate dehydrogenase with the mitochondrial branched-chain
Author Manuscript

aminotransferase (BCATm). J. Biol. Chem. 2010; 285:265–276. [PubMed: 19858196]


Komlos D, Mann KD, Zhuo Y, Ricupero CL, Hart RP, Liu AY, Firestein BL. Glutamate dehydrogenase
1 and SIRT4 regulate glial development. Glia. 2013; 61:394–408. [PubMed: 23281078]
Krebs HA. Metabolism of amino-acids: The synthesis of glutamine from glutamic acid and ammonia,
and the enzymic hydrolysis of glutamine in animal tissues. Biochem. J. 1935; 29:1951–1969.
[PubMed: 16745865]
Krebs HA. Equilibria in transamination systems. Biochem. J. 1953; 54:82–86. [PubMed: 13058836]
Kvamme E, Torgner IA, Roberg B. Kinetics and localization of brain phosphate activated glutaminase.
J. Neurosci. Res. 2001; 66:951–958. [PubMed: 11746423]

Adv Neurobiol. Author manuscript; available in PMC 2015 December 02.


Schousboe et al. Page 11

Lange SC, Bak LK, Waagepetersen HS, Schousboe A, Norenberg MD. Primary Cultures of
Astrocytes: Their Value in Understanding Astrocytes in Health and Disease. Neurochem. Res.
Author Manuscript

2012; 37:2569–2588. [PubMed: 22926576]


Larsson OM, Drejer J, Kvamme E, Svenneby G, Hertz L, Schousboe A. Ontogenetic development of
glutamate and GABA metabolizing enzymes in cultured cerebral cortex interneurons and in
cerebral cortex in vivo. Int. J. Dev. Neurosci. 1985; 3:177–185. [PubMed: 24874599]
Lavu S, Boss O, Elliott PJ, Lambert PD. Sirtuins--novel therapeutic targets to treat age-associated
diseases. Nat. Rev. Drug Discov. 2008; 7:841–853. [PubMed: 18827827]
Leke R, Bak LK, Anker M, et al. Detoxification of Ammonia in Mouse Cortical GABAergic Cell
Cultures Increases Neuronal Oxidative Metabolism and Reveals an Emerging Role for Release of
Glucose-Derived Alanine. Neurotox. Res. 2011; 19:496–510. [PubMed: 20480276]
Lieth E, LaNoue KF, Berkich DA, Xu B, Ratz M, Taylor C, Hutson SM. Nitrogen shuttling between
neurons and glial cells during glutamate synthesis. J. Neurochem. 2001; 76:1712–1723. [PubMed:
11259489]
Lovatt D, Sonnewald U, Waagepetersen HS, et al. The transcriptome and metabolic gene signature of
protoplasmic astrocytes in the adult murine cortex. J. Neurosci. 2007; 27:12255–12266. [PubMed:
Author Manuscript

17989291]
Martin DL, Rimvall K. Regulation of gamma-aminobutyric acid synthesis in the brain. J. Neurochem.
1993; 60:395–407. [PubMed: 8419527]
Matsui K, Jahr CE, Rubio ME. High-concentration rapid transients of glutamate mediate neural-glial
communication via ectopic release. J. Neurosci. 2005; 25:7538–7547. [PubMed: 16107641]
McKenna M. Glutamate pays its own way in astrocytes. Front. Endocrinol. 2013 In revision.
McKenna, M.; Dienel, GA.; Sonnewald, U.; Waagepetersen, HS.; Schousboe, A. Energy metabolism
of the brain. In:. In: Brady, ST.; Siegel, GJ.; Albers, RW.; Price, DI., editors. Basic Neurochmistry.
Waltham, MS, USA: Academic Press, Elsevier; 2012. p. 200-231.
McKenna MC. The glutamate-glutamine cycle is not stoichiometric: fates of glutamate in brain. J.
Neurosci. Res. 2007; 85:3347–3358. [PubMed: 17847118]
McKenna MC. Glutamate dehydrogenase in brain mitochondria: do lipid modifications and transient
metabolon formation influence enzyme activity? Neurochem. Int. 2011; 59:525–533. [PubMed:
21771624]
Author Manuscript

McKenna MC, Hopkins IB, Lindauer SL, Bamford P. Aspartate aminotransferase in synaptic and
nonsynaptic mitochondria: differential effect of compounds that influence transient hetero-enzyme
complex (metabolon) formation. Neurochem. Int. 2006; 48:629–636. [PubMed: 16513215]
McKenna MC, Sonnewald U, Huang X, Stevenson J, Zielke HR. Exogenous glutamate concentration
regulates the metabolic fate of glutamate in astrocytes. J. Neurochem. 1996; 66:386–393.
[PubMed: 8522979]
McKenna MC, Stevenson JH, Huang X, Hopkins IB. Differential distribution of the enzymes
glutamate dehydrogenase and aspartate aminotransferase in cortical synaptic mitochondria
contributes to metabolic compartmentation in cortical synaptic terminals. Neurochem. Int. 2000;
37:229–241. [PubMed: 10812208]
Norenberg MD, Martinez-Hernandez A. Fine structural localization of glutamine synthetase in
astrocytes of rat brain. Brain Res. 1979; 161:303–310. [PubMed: 31966]
Obel LF, Andersen KMH, Bak LK, Schousboe A, Waagepetersen HS. Effects of adrenergic agents on
intracellular ca2+ homeostasis and metabolism of glucose in astrocytes with an emphasis on
pyruvate carboxylation, oxidative decarboxylation and recycling: implications for glutamate
Author Manuscript

neurotransmission and excitotoxicity. Neurotox. Res. 2012; 21:405–417. [PubMed: 22194159]


Oberheim NA, Takano T, Han X, et al. Uniquely hominid features of adult human astrocytes. J.
Neurosci. 2009; 29:3276–3287. [PubMed: 19279265]
Oberheim NA, Wang X, Goldman S, Nedergaard M. Astrocytic complexity distinguishes the human
brain. Trends Neurosci. 2006; 29:547–553. [PubMed: 16938356]
Okada Y, Taniguchi H, Schimada C. High concentration of GABA and high glutamate decarboxylase
activity in rat pancreatic islets and human insulinoma. Science. 1976; 194:620–622. [PubMed:
185693]

Adv Neurobiol. Author manuscript; available in PMC 2015 December 02.


Schousboe et al. Page 12

Ott P, Clemmesen O, Larsen FS. Cerebral metabolic disturbances in the brain during acute liver failure:
from hyperammonemia to energy failure and proteolysis. Neurochem. Int. 2005; 47:13–18.
Author Manuscript

[PubMed: 15921824]
Öz G, Berkich DA, Henry PG, Xu Y, LaNoue K, Hutson SM, Gruetter R. Neuroglial metabolism in the
awake rat brain: CO2 fixation increases with brain activity. J. Neurosci. 2004; 24:11273–11279.
[PubMed: 15601933]
Öz, G.; Okar, DA.; Henry, PG. Glutamate-glutamine cycle and anaplerosis. In: Choi, I.; Gruetter, R.,
editors. Neural metabolism in vivo, Adv. Neurobiol. Vol. 4. New York, USA: Springer; 2012. p.
921-946.
Pamiljans V, Krishnaswamy PR, Dumville G, Meister A. Studies on the mechanism of glutamine
synthesis; isolation and properties of the enzyme from sheep brain. Biochemistry. 1962; 1:153–
158. [PubMed: 14483465]
Quastel, JH. Metabolic compartmentation in the brain and effects of metabolic inhibitors. In: Berl, S.;
Clarke, DD.; Scheider, D., editors. Metabolic compartmentation and neurotransmission. Vol. 6.
New York, USA: Plenum Publishing Corporation; 1975. p. 337-361.
Roberts E, Frankel S. gamma-Aminobutyric acid in brain: its formation from glutamic acid. J. Biol.
Author Manuscript

Chem. 1950; 187:55–63. [PubMed: 14794689]


Ronzio RA, Rowe WB, Meister A. Studies on the mechanism of inhibition of glutamine synthetase by
methionine sulfoximine. Biochemistry. 1969; 8:1066–1075. [PubMed: 4305484]
Rothman DL, De Feyter HM, Maciejewski PK, Behar KL. Is there in vivo evidence for amino acid
shuttles carrying ammonia from neurons to astrocytes? Neurochem. Res. 2012; 37:2597–2612.
[PubMed: 23104556]
Saito K, Barber R, Wu J, Matsuda T, Roberts E, Vaughn JE. Immunohistochemical localization of
glutamate decarboxylase in rat cerebellum. Proc. Nat. Acad. Sci. U.S.A. 1974; 71:269–273.
Salganicoff L, Derobertis E. Subcellular Distribution of the Enzymes of the Glutamic Acid, Glutamine
and Gamma-Aminobutyric Acid Cycles in Rat Brain. J. Neurochem. 1965; 12:287–309. [PubMed:
14336230]
Schousboe, A. Studies of brain metabolism: A historical perspective. In: Choi, I.; Gruetter, R., editors.
Neural metabolism in vivo, Adv. Neurobiol. Vol. 4. New York, USA: Springer; 2012. p. 909-920.
Schousboe, A.; Bak, LK.; Madsen, KK.; Waagepetersen, HS. Amino acid neurotransmitter synthesis
and removal. In: Kettenmann, H.; Ransom, B., editors. Neuroglia. Oxford, UK: Oxford University
Author Manuscript

Press; 2012. p. 443-456.


Schousboe A, Bak LK, Waagepetersen HS. Astrocytic Control of Biosynthesis and Turnover of the
Neurotransmitters Glutamate and GABA. Front. Endocrinol. 2013; 4:102.
Schousboe A, Hertz L, Svenneby G, Kvamme E. Phosphate activated glutaminase activity and
glutamine uptake in primary cultures of astrocytes. J. Neurochem. 1979; 32:943–950. [PubMed:
430071]
Shank RP, Bennett GS, Freytag SO, Campbell GL. Pyruvate carboxylase: an astrocyte-specific enzyme
implicated in the replenishment of amino acid neurotransmitter pools. Brain Res. 1985; 329:364–
367. [PubMed: 3884090]
Soghomonian JJ, Martin DL. Two isoforms of glutamate decarboxylase: why? Trends Pharmacol. Sci.
1998; 19:500–505. [PubMed: 9871412]
Sonnewald U, Westergaard N, Hassel B, Muller TB, Unsgard G, Fonnum F, Hertz L, Schousboe A,
Petersen SB. NMR spectroscopic studies of 13C acetate and 13C glucose metabolism in
neocortical astrocytes: evidence for mitochondrial heterogeneity. Devel. Neurosci. 1993a; 15:351–
Author Manuscript

358. [PubMed: 7805589]


Sonnewald U, Westergaard N, Petersen SB, Unsgard G, Schousboe A. Metabolism of
[U-13C]glutamate in astrocytes studied by 13C NMR spectroscopy: incorporation of more label
into lactate than into glutamine demonstrates the importance of the tricarboxylic acid cycle. J.
Neurochem. 1993b; 61:1179–1182. [PubMed: 8103082]
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. 1993c; 22:19–29. [PubMed: 8095170]

Adv Neurobiol. Author manuscript; available in PMC 2015 December 02.


Schousboe et al. Page 13

Spanaki C, Zaganas I, Kounoupa Z, Plaitakis A. The complex regulation of human glud1 and glud2
glutamate dehydrogenases and its implications in nerve tissue biology. Neurochem. Int. 2012;
Author Manuscript

61:470–481. [PubMed: 22658952]


van den Berg CJ, Garfinkel D. A stimulation study of brain compartments. Metabolism of glutamate
and related substances in mouse brain. Biochem. J. 1971; 123:211–218. [PubMed: 5164952]
Waagepetersen HS, Sonnewald U, Larsson OM, Schousboe A. A possible role of alanine for ammonia
transfer between astrocytes and glutamatergic neurons. J. Neurochem. 2000; 75:471–479.
[PubMed: 10899921]
Walls AB, Eyjolfsson EM, Smeland OB, Nilsen LH, Schousboe I, Schousboe A, Sonnewald U,
Waagepetersen HS. Knockout of GAD65 has major impact on synaptic GABA synthesized from
astrocyte-derived glutamine. J. Cereb. Blood Flow Metab. 2011; 31:494–503. [PubMed:
20664610]
Westergaard N, Varming T, Peng L, Sonnewald U, Hertz L, Schousboe A. Uptake, release, and
metabolism of alanine in neurons and astrocytes in primary cultures. J. Neurosci. Res. 1993;
35:540–545. [PubMed: 8377225]
Wu JY, Matsuda T, Roberts E. Purification and characterization of glutamate decarboxylase from
Author Manuscript

mouse brain. J. Biol. Chem. 1973; 248:3029–3034. [PubMed: 4700449]


Yu AC, Drejer J, Hertz L, Schousboe A. Pyruvate carboxylase activity in primary cultures of astrocytes
and neurons. J. Neurochem. 1983; 41:1484–1487. [PubMed: 6619879]
Yu AC, Schousboe A, Hertz L. Metabolic fate of 14C-labeled glutamate in astrocytes in primary
cultures. J. Neurochem. 1982; 39:954–960. [PubMed: 6126524]
Yudkoff M, Daikhin Y, Nelson D, Nissim I, Erecinska M. Neuronal metabolism of branched-chain
amino acids: flux through the aminotransferase pathway in synaptosomes. J. Neurochem. 1996;
66:2136–2145. [PubMed: 8780046]
Zaganas I, Kanavouras K, Mastorodemos V, Latsoudis H, Spanaki C, Plaitakis A. The human GLUD2
glutamate dehydrogenase: localization and functional aspects. Neurochem. Int. 2009; 55:52–63.
[PubMed: 19428807]
Zaganas I, Waagepetersen HS, Georgopoulos P, Sonnewald U, Plaitakis A, Schousboe A. Differential
expression of glutamate dehydrogenase in cultured neurons and astrocytes from mouse cerebellum
and cerebral cortex. J. Neurosci. Res. 2001; 66:909–913. [PubMed: 11746418]
Zielke HR, Tildon JT, Landry ME, Max SR. Effect of 8-bromo-cAMP and dexamethasone on
Author Manuscript

glutamate metabolism in rat astrocytes. Neurochem. Res. 1990; 15:1115–1122. [PubMed:


1982459]
Zwingmann C, Richter-Landsberg C, Brand A, Leibfritz D. NMR spectroscopic study on the metabolic
fate of [3-(13)C]alanine in astrocytes, neurons, and cocultures: implications for glia-neuron
interactions in neurotransmitter metabolism. Glia. 2000; 32:286–303. [PubMed: 11102969]
Author Manuscript

Adv Neurobiol. Author manuscript; available in PMC 2015 December 02.


Schousboe et al. Page 14
Author Manuscript
Author Manuscript
Author Manuscript

Figure 1.
Reactions involving glutamate (Glu) in astrocytes. Ala, Alanine; α-KG, α-ketoglutarate;
Asp, aspartate; BCAA, branched chain amino acid; Gln, glutamine. See text for other
abbreviations.
Author Manuscript

Adv Neurobiol. Author manuscript; available in PMC 2015 December 02.


Schousboe et al. Page 15
Author Manuscript
Author Manuscript
Author Manuscript

Figure 2.
Schematic representation of complete oxidation of glutamate via the ”pyruvate recycling
pathway” (essential enzymes in red) and de novo synthesis of glutamate and glutamine from
glucose via pyruvate carboxylase (PC) (essential enzymes in blue) in astrocytes.
For de novo synthesis of glutamate and glutamine, glucose is metabolized to acetyl CoA and
oxaloacetate via pyruvate dehydrogenase and pyruvate carboxylase, respectively. Acetyl
CoA and oxaloacetate condenses to citrate and subsequently, α-ketoglutarate (α-KG) is
formed. α-Ketoglutarate is converted to glutamate catalyzed by aspartate aminotransferase
(AAT) and via an amidation, catalyzed by the energy requiring enzyme glutamine synthetase
(GS), glutamine may be formed. The complete oxidation of glutamate is initiated by
oxidative deamination catalyzed by glutamate dehydrogenase (GDH) and oxidative
Author Manuscript

decarboxylation of α-ketoglutarate to succinyl CoA catalyzed by α-ketoglutarate


dehydrogenase (KGDH). The carbon skeleton is subsequently converted into malate via
multiple steps in the tricarboxylic acid (TCA) cycle and pyruvate via malic enzyme (ME).
Alternatively, the concerted action of phosphoenolpyruvate carboxykinase (PEPCK) and
pyruvate kinase (PK) converts oxaloacetate (OAA) into pyruvate. The operation of ME or
PEPCK plus PK in the direction of pyruvate is necessary for complete oxidation of
glutamate. Pyruvate re-enters the TCA cycle via pyruvate dehydrogenase (PDH) and the

Adv Neurobiol. Author manuscript; available in PMC 2015 December 02.


Schousboe et al. Page 16

carbon skeleton originating from glutamate may through this pathway be completely
oxidized to CO2 in the TCA cycle. Asp, aspartate.
Author Manuscript
Author Manuscript
Author Manuscript
Author Manuscript

Adv Neurobiol. Author manuscript; available in PMC 2015 December 02.


Schousboe et al. Page 17
Author Manuscript
Author Manuscript

Figure 3.
Neurotransmitter glutamate (GLU) is mainly taken up by surrounding astrocytes subsequent
to interaction with receptors in the synapse. In the astrocyte, glutamate is either converted to
Author Manuscript

glutamine (GLN) catalyzed by glutamine synthetase (GS) as part of the glutamate-glutamine


cycle or metabolized in the tricarboxylic acid (TCA) cycle. Glutamine is transferred to the
glutamatergic neuron to be used for synthesis of glutamate catalyzed by phosphate activated
glutaminase (PAG). Glutamate enters the TCA cycle by the activity of glutamate
dehydrogenase (GDH) or an aminotransferase (AT), and the carbon skeleton may either be
completely oxidatively metabolized via pyruvate recycling including malic enzyme (ME)
activity or phosphoenolpyruvate carboxykinase and pyruvate kinase. Alternative, the carbon
skeleton supports the pool of TCA cycle intermediates and in that way potentially increases
the oxidation of acetyl CoA in the TCA cycle. De novo synthesis of glutamate and
glutamine from glucose occurs via the concerted action of pyruvate dehydrogenase (PDH)
and pyruvate carboxylase (PC) making a net synthesis of TCA cycle intermediates. CIT,
citrate; OAA, oxaloacetate; PYR, pyruvate.
Author Manuscript

Adv Neurobiol. Author manuscript; available in PMC 2015 December 02.


Schousboe et al. Page 18
Author Manuscript
Author Manuscript
Author Manuscript

Figure 4.
Author Manuscript

[U-13C]Glutamate (GLU) may be completely metabolized to CO2 via the “pyruvate


recycling pathway”. Initially [U-13C]glutamate is converted to α-ketoglutarate (α-KG)
catalyzed mainly by glutamate dehydrogenase (GDH). Via the tricarboxylic acid (TCA)
cycle [U-13C]malate (MAL)/oxaloacetate (OAA) is formed. [U-13C]Malate may be
oxidatively decarboxylated to [U-13C]pyruvate (PYR) catalyzed by malic enzyme (ME).
Alternatively, [U-13C]oxaloacetate may be metabolized to pyruvate via
[U-13C]phosphoenolpyruvate catalyzed by the concerted action of phosphoenolpyruvate

Adv Neurobiol. Author manuscript; available in PMC 2015 December 02.


Schousboe et al. Page 19

carboxykinase (PEPCK) and pyruvate kinase (PK). The carbons originating from glutamate
can subsequently re-enter the TCA cycle by oxidative decarboxylation of [U-13C]pyruvate to
Author Manuscript

[1,2-13C]acetyl CoA catalyzed by pyruvate dehydrogenase (PDH). Unlabeled oxaloacetate


may condense with [1,2-13C]acetyl CoA forming [1,2-13C]citrate (CIT). From metabolism
in the TCA cycle [4,5-13C] α-ketoglutarate is formed and consequently [4,5-13C]glutamate
due to the activity of aspartate aminotransferase (AAT). The symmetric succinate molecule
causes scrambling of labeling and therefore both [1,2-13C] and [3,4-13C] oxaloacetate are
formed. Oxaloacetate is in equilibrium with [1,2-13C] and [3,4-13C]aspartate due to the
activity of aspartate aminotransferase. Alternative to oxidative decarboxylation of pyruvate,
[U-13C]pyruvate may be converted to [U-13C]lactate (LAC) catalyzed by lactate
dehydrogenase (LDH).
Author Manuscript
Author Manuscript
Author Manuscript

Adv Neurobiol. Author manuscript; available in PMC 2015 December 02.


Schousboe et al. Page 20
Author Manuscript
Author Manuscript
Author Manuscript

Figure 5.
Neurotransmitter glutamate is subsequent to receptor interaction primarily taken up mainly
by astrocytes. Glutamate (GLU) is amidated to glutamine (GLN) via glutamine synthetase
(GS) and glutamine is subsequently released from the astrocyte via specific transporters
followed by uptake into the neuron. In the neuron glutamine is deamidated by phosphate
activated glutaminase (PAG) to glutamate, which completes the glutamate - glutamine cycle.
A net transfer of nitrogen from the astrocyte to the neuron occurs as part of the glutamate -
glutamine cycle. This nitrogen transfer may be counteracted by transport of a neuro-inactive
amino acid (AA) likely either alanine or one of the branched chain amino acids. The
glutamate - glutamine cycle is coupled to an amino acid – keto acid (KA) cycle via the
Author Manuscript

action of glutamate dehydrogenase (GDH) and the relevant aminotransferase (AT).

Adv Neurobiol. Author manuscript; available in PMC 2015 December 02.

You might also like