Vous êtes sur la page 1sur 7

Neurochemistry International 60 (2012) 690–696

Contents lists available at SciVerse ScienceDirect

Neurochemistry International
journal homepage: www.elsevier.com/locate/nci

Review

Pathogenesis of hepatic encephalopathy and brain edema in acute liver failure:


Role of glutamine redefined
Paul Desjardins a, Ting Du b, Wenlei Jiang a, Liang Peng b, Roger F. Butterworth a,⇑
a
Neuroscience Research Unit, Saint-Luc Hospital (CHUM), University of Montreal, Montreal, Quebec, Canada
b
Department of Clinical Pharmacology, China Medical University, Shenyang, PR China

a r t i c l e i n f o a b s t r a c t

Article history: Acute liver failure (ALF) is characterized neuropathologically by cytotoxic brain edema and biochemically
Available online 21 February 2012 by increased brain ammonia and its detoxification product, glutamine. The osmotic actions of increased
glutamine synthesis in astrocytes are considered to be causally related to brain edema and its complications
Keywords: (intracranial hypertension, brain herniation) in ALF. However studies using multinuclear 1H- and 13C-NMR
Acute liver failure spectroscopy demonstrate that neither brain glutamine concentrations per se nor brain glutamine synthesis
Ammonia rates correlate with encephalopathy grade or the presence of brain edema in ALF. An alternative mechanism
Glutamine
is now proposed whereby the newly synthesized glutamine is trapped within the astrocyte as a conse-
Small neutral amino acid transporters
Brain edema
quence of down-regulation of its high affinity glutamine transporter SNAT5 in ALF. Restricted transfer
Hepatic encephalopathy out of the cell rather than increased synthesis within the cell could potentially explain the cell swelling/
In vivo cerebral microdialysis brain edema in ALF. Moreover, the restricted transfer of glutamine from the astrocyte to the adjacent gluta-
NMR spectroscopy matergic nerve terminal (where glutamine serves as immediate precursor for the releasable/transmitter
pool of glutamate) could result in decreased excitatory transmission and excessive neuroinhibition that
is characteristic of encephalopathy in ALF. Paradoxically, in spite of renewed interest in arterial ammonia
as a predictor of raised intracranial pressure and brain herniation in ALF, ammonia-lowering agents aimed
at reduction of ammonia production in the gut have so far been shown to be of limited value in the preven-
tion of these cerebral consequences. Mild hypothermia, shown to prevent brain edema and intracranial
hypertension in both experimental and human ALF, does so independent of effects on brain glutamine syn-
thesis; whether or not hypothermia restores expression levels of SNAT5 in ALF awaits further studies. While
inhibitors of brain glutamine synthesis such as methionine sulfoximine, have been proposed for the preven-
tion of brain edema in ALF, potential adverse effects have so far limited their applicability.
Ó 2012 Elsevier Ltd. All rights reserved.

1. Introduction been reported in experimental animal models of ALF at coma/ede-


ma stages of encephalopathy (Swain et al., 1992; Mans et al., 1979).
Hepatic Encephalopathy (HE) and brain edema and its complica- The advent of modern techniques such as in vivo cerebral micro-
tions (intracranial hypertension, brain herniation) remain serious dialysis, multi-nuclear magnetic resonance (NMR) spectroscopy
cerebral complications of acute liver failure (ALF). The occurrence and related molecular approaches have provided new impetus to
of HE in ALF heralds the need for liver transplantation and compli- studies on pathophysiologic mechanisms in ALF, helping to resolve
cations of brain edema remain one of the major causes of mortality existing controversies and to point the way to new diagnostic and
in ALF patients. The precise pathophysiological mechanisms therapeutic strategies in this area of investigation. The current re-
responsible for these complications have not been completely eluci- view is both a critical appraisal of the role of intracellular accumu-
dated. However, studies related to the cytotoxic effects of ammonia lation of glutamine in the pathogenesis of HE and brain edema in
continue to be a focus of research in this area. Hyperammonemia ALF and also an attempt to identify new areas of investigation
occurs in >90% of ALF patients (Ferenci et al., 1996; Brusilow et al., and novel treatment opportunities for ALF.
2010) and brain ammonia concentrations in the 1–5 mM range have

2. Arterial ammonia and brain edema in acute liver failure

⇑ Corresponding author. Address: Neuroscience Research Unit, CHUM, Campus Although not reliable for diagnosis purposes due undoubtedly
Saint-Luc, University of Montreal, 1058 St-Denis Street, Montreal, Quebec, Canada to difficulties in measurement, arterial ammonia concentrations
H2X 3J4. Tel.: +1 514 890 8000x35759; fax: +1 514 412 7377. in excess of 300 lM are reliable predictors of brain herniation in
E-mail address: roger.butterworth@umontreal.ca (R.F. Butterworth). ALF patients (Clemmesen et al., 1999; Bernal et al., 2007). In a

0197-0186/$ - see front matter Ó 2012 Elsevier Ltd. All rights reserved.
doi:10.1016/j.neuint.2012.02.001
P. Desjardins et al. / Neurochemistry International 60 (2012) 690–696 691

recent study using the in vivo microdialysis technique in 17 pa- (Michalak et al., 1996). Although these findings (as well as those
tients with ALF resulting primarily from acetaminophen overdose, reported by Zwingmann et al., 2003) support a role for glutamine
a significant positive correlation between arterial ammonia con- in the pathogenesis of early encephalopathy in ALF, measurement
centrations and intracranial pressure was observed with r = 0.73, of intracellular (astrocytic) concentrations of glutamine may be re-
p < 0.05 (Fig. 1) (Tofteng et al., 2006) further suggestive of a role quired in order to assess its role in the pathogenesis of severe
for ammonia in the pathogenesis of this complication of brain ede- encephalopathy (coma) or brain edema in ALF. The issue was par-
ma in these patients. tially addressed using in vivo cerebral microdialysis to measure
extracellular brain glutamine in patients with ALF (Tofteng et al.,
2006). Patients with high intracranial pressure (ICP) were found
3. Brain glutamine in acute liver failure
to have significantly higher brain glutamine compared to patients
with normal ICP (with increased ICP: 6.54 lM, without increased
Two enzyme systems are theoretically capable of removing
ICP: 3.52 lM; p < 0.05) (Fig. 3). The authors suggested that the rise
ammonia in brain. These enzymes are glutamate dehydrogenase
in glutamine was a prerequisite for the development of increased
(GDH) and glutamine synthetase (GS). GDH catalyses the conver-
cerebral blood flow and high ICP in ALF patients.
sion of alpha-ketoglutarate to glutamate, a reaction shown to occur
in both neurons and astrocytes, according to reaction (1); GS catal-
yses the conversion of glutamate to glutamine, a reaction that oc- 4. Brain ammonia removal (glutamine synthesis) in acute liver
curs primarily if not exclusively in astrocytes (Norenberg, 1979), failure
according to reaction (2):
Although increased brain glutamine is a consistent feature of
Alpha  ketoglutarate þ NHþ4 þ NADðPÞH þ Hþ ¢ Glutamate
both experimental and human ALF, activities of GS are either un-
þ NADðPÞþ þ H2 O ð1Þ changed or slightly reduced in acute hyperammonemia (Cooper
et al., 1985) and in experimental ALF in the rat resulting from he-
NH3 þ Glutamate þ ATP ! Glutamine þ ADP þ Pi ð2Þ patic devascularisation where a small but significant 18% reduction
in GS activity was noted in cerebral cortex (Chatauret et al., 2006).
A limited capacity for brain glutamine synthesis via GS was con-
Studies using 13N-ammonia have shown convincingly that cluded by Deutz et al. (1988) who noted a faster rate of increase
blood-borne ammonia is incorporated almost exclusively into glu- of brain ammonia compared to brain glutamine in an ALF model
tamine by reaction (2) in the brain under both normal and hyper- and by Bosman et al. (1990) who observed that increased gluta-
ammonemic conditions (Cooper et al., 1985). Consequently it is the mine synthesis was not sustained in experimental ALF and, at
astrocyte that is called upon to remove both blood-borne and lo- coma/edema stages of HE, brain glutamine synthesis was actually
cally synthesized ammonia in the brain in ALF. It is not surprising, decreased. Similar findings were reported by Michalak et al.
therefore, that brain glutamine concentrations have consistently (1996). Finally, a lack of correlation between HE grade and
been found to be elevated up to 5-fold in experimental ALF using in vivo GS activities was reported by Kanamori et al. (1996) using
classical biochemical procedures (Traber et al., 1989; Swain et al., 1
H-NMR spectroscopy in a hyperammonemia model. One possible
1992; Mans et al., 1994) and subsequently confirmed by 1H-NMR explanation for the observed loss of GS activity in brain in ALF may
spectroscopy (Zwingmann et al., 2003). Both CSF and brain concen- relate to inactivation of the enzyme due to protein tyrosine nitra-
trations of glutamine are also increased in patients with ALF due to tion. Exposure of primary astrocyte cultures to millimolar concen-
acetaminophen hepatotoxicity (Record et al., 1976). More recently, trations of ammonia results in significant nitration of GS protein
using the technique of in vivo cerebral microdialysis, glutamine leading to loss of enzyme activity (Schliess et al., 2002). However,
concentrations were found to be increased up to 5-fold in brain whether or not such mechanisms are applicable to GS loss in brain
extracellular fluid starting early (prior to the onset of HE and brain in ALF remains a matter of conjecture. The findings of a lack of
edema) and remaining high until coma/edema stages of HE (Fig. 2) induction of GS in brain underscores the vulnerability of the brain
to hyperammonemic conditions in general and to ALF, in
particular.
In contrast to brain, skeletal muscle of ALF animals manifested a
significant 27% increase of GS activity resulting from a post-trans-
lational induction of GS expression (Fig. 4). Increased de novo
synthesis of glutamine in muscle of ALF animals was confirmed

400

* *
Glutamine (µM)

300
*
200

100

0
SHAM ALF-6h PRECOMA COMA

Fig. 1. Correlation between arterial ammonia concentration (lM) and intracranial Fig. 2. Extracellular concentrations of glutamine in the frontal cortex of rats with
pressure (mm Hg) in patients (n = 17) with fulminant hepatic failure. Regression ALF resulting from hepatic devascularization as a function of progression of
line and 95% confidence intervals are shown (p < 0.05 ; r = 0.73) (adapted from encephalopathy compared to sham-operated controls (⁄p < 0.05 vs. sham-operated
Tofteng et al., 2006). controls) (adapted from Michalak et al., 1996).
692 P. Desjardins et al. / Neurochemistry International 60 (2012) 690–696

been undertaken in brain in experimental ALF using the technique


of 13C-NMR. Using this approach, it was demonstrated that the
synthesis of glutamine from 13C-glucose via both the pyruvate
dehydrogenase and pyruvate carboxylase metabolic pathways
was increased at early time points following onset of ALF but that
this increase was not maintained at coma/edema time points again
suggesting a limit on the capacity of the astrocytes to remove
ammonia in ALF (Fig. 5) (Zwingmann et al., 2003).
As with the findings of a lack of correlation between brain glu-
tamine levels and HE severity or presence of brain edema, these
findings cast doubt on the role of glutamine synthesis in the path-
ogenesis of these complications.
On the other hand, it has been known for almost half a decade
since the pioneering work of Warren and Schenker (1964) who
showed that GS inhibition by methionine sulfoximine (MSO) pro-
tected against the lethal effects of acute ammonia toxicity. Since
that time MSO has been shown to be partially protective against
the CNS effects of hyperammonemia in a variety of experimental
paradigms. For example, MSO treatment results in decreased
Fig. 3. Correlation between extracellular glutamine concentration (lM) and swelling of cortical astrocytes in ammonium acetate-treated rats
intracranial pressure (mm Hg) in patients (n = 17) with fulminant hepatic failure. (Willard-Mack et al., 1996) and MSO treatment is protective in
Regression line and 95% confidence intervals are shown (p < 0.05; r = 0.58) (adapted ammonia-precipitated brain edema in portacaval-shunted rats
from Tofteng et al., 2006).
(Blei et al., 1994). It was concluded that increased glutamine pro-
duction in the astrocyte contributes to the pathogenesis of ammo-
by 13C-NMR spectroscopy in this study (Chatauret et al., 2006). nia-induced brain edema in the context of liver failure.
Skeletal muscle also plays an important role in the detoxification
of ammonia to glutamine in patients with liver disease (Lockwood 5. Inter-cellular trafficking of glutamine in brain in acute liver
et al., 1979). failure
These findings provide a molecular explanation for the view
that, in ALF, skeletal muscle becomes the major organ responsible Unequivocal evidence of the cellular localization of synthetic
for ammonia detoxification and suggests fundamentally distinct and degradative enzymes together with transporters for substrates
mechanisms for GS regulation in muscle and brain. and release of products implicated in glutamine synthesis and
Although measurement of activities of GS in vitro may be indic- metabolism is now available. This evidence has given rise to the
ative of metabolic capacity, measurement of metabolic fluxes concept of the ‘‘Glutamate–Glutamine Cycle’’ in brain (Fig. 6)
through the enzymic pathway are required in order to adequately whereby glutamine formed in the astrocyte by the action of GS is
assess the dynamics of glutamine synthesis. Such studies have released from the cell via the glutamine transporter SNAT5 (Cube-
los et al., 2005) into the extracellular space where a portion of it
is available for uptake into the surrounding neurons via SNAT1
and SNAT2 (Melone et al., 2004, 2006). Once inside the neuron, glu-
GS expression (units)

1 tamine is transformed by the action of glutaminase into glutamate.


0.8 * sham-operated
In this way, glutamine serves as the immediate precursor of the
0.6 controls
releasable pool of the principal excitatory neurotransmitter (gluta-
* ALF
mate). The glutamate is released into the synaptic space where it
0.4
stimulates post-synaptic receptors. Surplus glutamate is cleared
0.2

0
brain muscle

GS activity
(µmole/mg protein/h)
Tissue Sham-operated ALF
controls

Cortex 5.86+/-0.25 4.76+/-0.35 *

Muscle 0.81+/-0.03 1.12+/-0.11 *

Fig. 4. GS expression and activities in cerebral cortex and skeletal muscle of rats Fig. 5. Incorporation of 13C-label into glutamine (lmole/g tissue) as calculated from
13
with ALF resulting from hepatic devascularization. (A) Expression levels of GS (as C-NMR spectra of brain extracts from sham-operated controls (white bars), ALF
determined by Western blot analysis) (n = 8) in ALF rats and sham-operated rats resulting from hepatic devascularization at precoma (grey bars) and coma
controls; (B) GS activities in cerebral cortex and skeletal muscle of ALF rats and (black bars) stages of encephalopathy. Values represent means of n = 4 animals +/
sham-operated controls (⁄p < 0.05 vs. Sham-operated controls) (adapted from SD (⁄p < 0.001 vs. sham-operated controls;  p < 0.01 vs. precoma) (adapted from
Chatauret et al., 2006). Zwingmann et al., 2003).
P. Desjardins et al. / Neurochemistry International 60 (2012) 690–696 693

glutamatergic nerve terminal

astrocyte

GLN
(to CSF)

GLN SNAT-1 SNAT-5 GLN

GLNase Glutamine
synthetase
NH3 NH3

GLU NH3

EAAT2 GLU
GLU

NH3
GLU (Blood-borne)

Fig. 6. Schematic representation of a glutamate–glutamine cycle. The cartoon illustrates position of the astrocytic glutamine synthetase (GS), neuronal glutaminase (GLNase),
the astrocytic glutamate transporter EAAT-2 and the astrocytic and neuronal glutamine transporters SNAT-5 and SNAT-1.

from the synapse by the action of a high affinity, high capacity glu- It is true that total brain glutamate is decreased in a range of
tamate transporter EAAT-2. Once inside the astrocyte, glutamate hyperammonemias including those associated with liver failure
serves as the substrate for GS and the cycle goes on. A similar inter- (Fitzpatrick et al., 1989; Lavoie et al., 1987; Mans et al., 1994; Swain
cellular cycle exists for GABA with distinct enzymes and transport- et al., 1992) and, although increased concentrations of glutamate
ers; in the case of the GABA system, the astrocyte glutamine have been reported in brain extracellular fluid in experimental
transporter involved is primarily SNAT3 rather than SNAT5 ALF (Michalak et al., 1996) using the technique of in vivo microdi-
(Boulland et al., 2002). alysis, it is important to bear in mind that microdialysates may
Studies using molecular techniques reveal a reduction in not accurately reflect true presynaptic, Ca2+-dependent, evoked re-
expression of the astrocytic glutamate transporter EAAT-2 in brain lease of glutamate from nerve terminals. Further studies are re-
in experimental ALF due to hepatic devascularization (Knecht et al., quired in order to address this important issue.
1997). Preliminary evidence shows that HE and brain edema in this
same animal model of ALF results in a selective loss of expression 6. Glutamine metabolism in brain in acute liver failure
of the gene coding for SNAT5 in brain (Fig. 7) (Du et al., 2010).
Loss of SNAT5 expression could effectively trap glutamine inside Phosphate-activated glutaminase (GLNase) is a mitochondrial
the astrocyte and this action (glutamine ‘‘trapping’’) rather than in- enzyme responsible for the deamination of glutamine to glutamate
creased glutamine synthesis could itself result in cell swelling and with the production of ammonia according to the reaction:
brain edema in ALF. Moreover it is important to bear in mind that
glutamine functions not only as an osmoactive substance in brain Glutamine þ H2 O ! Glutamate þ NH3
but is also intimately involved in the cell–cell interactions that Primarily neuronal in localization, GLNase is an integral compo-
are key to neurotransmitter regulation. In this regard, it is well nent of the Glutamate–Glutamine cycle (Fig. 6). Thus one turn of
established from classical biochemical techniques (Rothstein and the cycle removes one molecule of ammonia in the astrocyte due
Tabakoff, 1985) as well as immunogold histochemistry (Laake to GS and produces one molecule of ammonia in the neuron due
et al., 1995) that glutamine synthesized in the astrocyte is essential to GLNase. It should be borne in mind, however that the steps de-
for the maintenance of the releasable (neurotransmitter) pool of picted in the cycle are not meant to imply that the steps are con-
glutamate in the adjoining glutamatergic neuron (Fig. 6). Conse- certed; in fact intermediates enter and leave the cycle at various
quently, trapping of glutamine in the astrocyte resulting from steps dependent upon the nature of the intermediate. It is also
down-regulation of SNAT5 in ALF has the potential to result in important to recall that brain GLNase is inhibited by NH3 (product
impairment of glutamatergic (excitatory) neurotransmission in inhibition) (Matheson and van den Berg, 1975) and, in view of the
brain leading to the excessive neuroinhibition that is characteristic increased brain ammonia concentrations in ALF, it has been sug-
of encephalopathy in ALF. Although attractive, this hypothesis has gested that the increased brain glutamine results, at least in part,
not yet been totally validated and other astrocytic and/or neuronal from decreased activity of GLNase (Felipo and Butterworth, 2002).
glutamine transporters may also be implicated. These mechanisms A novel hypothesis was recently proposed whereby glutamine,
may also apply in a brain region-dependent manner. acting as a ‘‘Trojan horse’’ was transported into the mitochondrion
694 P. Desjardins et al. / Neurochemistry International 60 (2012) 690–696

(A) STD
SHAM ALF
(bp)

310

SNAT1
234 (249bp)

194

SNAT5
(149bp)
118

603

β-actin
310 (360bp)

1 2 3 4 5 6

SNAT5
(B) 150

SNAT1
mRNA expression
(% beta actin)

100
*

50

0
SHAM ALF SHAM ALF

Fig. 7. Expression of the neuronal amino acid transporter SLC38A1 (SNAT1) and the astrocytic transporters SLC38A5 (SNAT5) in the brain of rats with ALF resulting from
hepatic devascularization. (A) Total RNA was extracted from the frontal cortex of ALF rats at coma stage of encephalopathy (ALF) (lanes 5–6) or from sham-operated controls
(SHAM) (lanes 3–4). SNAT1 (279 bp) and SNAT5 (149 bp) mRNAs were reverse-transcribed and amplified by PCR. Expression were normalized to beta-actin housekeeping
gene. Reverse transcriptase was omitted from the reaction as a negative control (lane 2). (B) Graph bar representation of data shown in panel A. Data represent mean ± SEM of
four animals in each group (⁄p < 0.001 vs. SHAM by Student t test).

of the astrocyte and deaminated to ammonia which, in turn, (1995) that, under hyperammonemic conditions, brain GLNase
caused cellular dysfunction and cell swelling/edema (Albrecht operates at less than 1% of maximal levels.
and Norenberg, 2006). Evidence for this was based primarily on On a related topic, transamination of glutamine (with a suitable
the results of experiments performed in cultured astrocytes which alpha-ketoacid co-substrate) in brain results in the formation of an
are known to express much higher concentrations of GLNase than interesting neuroactive/neurotoxic molecule known as alpha-keto-
those observed in astrocytes in situ and on these grounds together glutaramate (KGM). CSF concentrations of KGM are increased over
with other fundamental differences in astrocyte phenotype be- 10-fold in patients with liver failure (Vergara et al., 1974) and CSF
tween the in vitro and in vivo preparations, the theory has been se- levels correlate better with severity of HE than did CSF glutamine.
verely criticised (Brusilow et al., 2010). A major contentious issue It was proposed that this compound may represent a putative
was that of the existence and function of GLNase in the astrocyte. endogenous neurotoxin or, alternatively, act as a biomarker of
It is well established that GLNase is present in mouse brain astro- CNS damage in liver failure. Urinary KGM is also markedly in-
cytes in culture (Kvamme et al., 1982), a finding that may relate to creased in urea cycle disorders and other primary hyperammone-
culture conditions. However, it was recently shown, using immu- mic diseases (Kuhara et al., 2011).
nohistochemical techniques, that GLNase is also expressed by a
population of astrocytes in human brain (Olalla et al., 2008). A 7. Therapeutic implications
novel form of GLNase has been reported in cultured astrocytes
(Kvamme et al., 2008). However, in contrast to neuronal GLNase, Given the impressive correlation between arterial ammonia and
the astrocytic enzyme is not subject to inhibition by the product raised ICP in patients with ALF (Fig. 1) coupled to the findings of a
of the reaction, ammonia (Kvamme et al., 1982). Precisely how significant value of raised arterial ammonia for the prediction of
the ‘‘Trojan horse’’ (glutamine) would result in the production of brain herniation in ALF patients (Clemmesen et al., 1999; Bernal
sufficient ammonia to cause cell swelling and HE rather than et al., 2007), it would be anticipated that ammonia-lowering strat-
undergoing uncontrolled futile cycling to and from glutamate with egies using non-absorbable disaccharides, antibiotics, branched-
production and release of ammonia is difficult to reconcile with chain amino acids or L-ornithine L-aspartate (LOLA) would be effec-
these observations and with the report from Kanamori and Ross tive in the prevention of brain edema and its complications in ALF.
P. Desjardins et al. / Neurochemistry International 60 (2012) 690–696 695

including effects on brain energy metabolism, brain lactate accu-


mulation (Chatauret et al., 2003), and anti-inflammatory proper-
ties (Jiang et al., 2009).
In summary, ALF, in common with other acute hyperammone-
mic disorders, is characterized by increased CSF and brain gluta-
mine. The osmotic consequences of increased glutamine synthesis
in astrocytes have traditionally been considered to be causally re-
lated to the cytotoxic brain edema and its complications (intracra-
nial hypertension, brain herniation) in ALF. However studies using
multinuclear 1H- and 13C-NMR spectroscopy demonstrate that nei-
ther brain glutamine concentrations nor brain glutamine synthesis
rates correlate with HE grade or the presence of brain edema in ALF.
An alternative mechanism is now proposed whereby glutamine is
trapped within the astrocyte as a consequence of down-regulation
Fig. 8. Effect of hypothermia on synthesis of brain glutamine in rats with ALF
resulting from hepatic devascularization. Percentage of 13C enrichments in of the glutamine transporter SNAT5 in ALF. Restricted transfer out
individual carbon position via the pyruvate carboxylase (PC) and pyruvate of the cell rather than increased synthesis within the cell could ex-
dehydrogenase (PDH) pathways calculated from 13C-NMR spectra of extracts from plain the cell swelling/brain edema and restricted transfer of gluta-
frontal cortex of normothermic (37 °C; black bars) and hypothermic (35 °C; hatched
mine from the astrocyte to the adjacent glutamatergic nerve
bars) ALF rats compared to sham-operated controls (white bars). Values represent
means of n = 6 animals +/ SD (⁄p < 0.001 vs. sham-operated controls;  p < 0.001 vs terminal (where glutamine serves as immediate precursor for the
normothermic by ANOVA) (adapted from Chatauret et al., 2003). releasable/transmitter pool of glutamate) could result in decreased
excitatory transmission and excessive neuroinhibition that is
characteristic of HE in ALF.
However, to date, clinical trials using lactulose (Stravitz et al., In spite of renewed interest in arterial ammonia as a predictor
2007) or LOLA (Acharya et al., 2009) have failed to show a robust of raised intracranial pressure and brain herniation in ALF, ammo-
beneficial effect. Further clinical trials with other agents are clearly nia-lowering agents aimed at reduction of ammonia production in
warranted. the gut are of limited value in the prevention of these cerebral con-
Reduction of brain glutamine concentrations would theoreti- sequences. Moreover, mild hypothermia, shown to prevent brain
cally reduce astrocyte swelling and, in this way, prevent brain ede- edema and intracranial hypertension in both experimental and hu-
ma and its complications in ALF. With this principal in mind, man ALF, does so independent of effects on brain glutamine syn-
several studies have demonstrated the effective use of MSO in both thesis. While inhibitors of brain glutamine synthesis have been
in vivo (Willard-Mack et al., 1996) and in vitro (Norenberg and proposed for the prevention of brain edema in ALF, potential ad-
Bender, 1994) preparations in the reduction of brain glutamine verse effects have so far limited their applicability.
and the concomitant prevention of swelling caused by ammonia.
This approach was pioneered by Warren and Schenker (1964)
almost 50 years ago and continues to garner interest today (Brusilow Acknowledgment
et al., 2010). MSO treatment attenuates brain edema in portacaval
rats with ammonia-induced brain edema (Blei et al., 1994). MSO Funded by the Canadian Institutes of Health Research
decreases the size of the astrocytic glutamine pool (Laake et al., (China–Canada Joint Health Research Initiative).
1995) and also restores the loss of control of cerebral blood flow
(Master et al., 1999) and cerebral metabolic rate for glucose
(Hawkins and Jessy, 1991) caused by acute hyperammonemia. Fur- References
thermore, studies in mice demonstrate no impairment of memory
Acharya, S.K., Bhatia, V., Sreenivas, V., Khanal, S., Panda, S.K., 2009. Efficacy of L-
function or other behavioural markers (Blin et al., 2002) which was ornithine L-aspartate in acute liver failure: a double-blind, randomized,
taken to indicate lack of neurotoxicity. However, the concept that placebo-controlled study. Gastroenterology 136, 2159–2168.
GS inhibition may afford a new therapeutic strategy in ALF has Albrecht, J., Norenberg, M.D., 2006. Glutamine: a Trojan horse in ammonia
neurotoxicity. Hepatology 44, 788–794.
been challenged (Zielińska et al., 2004) on the basis of its action Bernal, W., Hall, C., Karvellas, C.J., Auzinger, G., Sizer, E., Wendon, J., 2007. Arterial
as a convulsant which may or may not relate to inhibition of GS, ammonia and clinical risk factors for encephalopathy and intracranial
to the fact that MSO is metabolized to products of unknown toxic- hypertension in acute liver failure. Hepatology 46, 1844–1852.
Blei, A.T., Olafsson, S., Therrien, G., Butterworth, R.F., 1994. Ammonia-induced brain
ity (Rao and Meister, 1972; Cooper et al., 1976) and to the known
edema and intracranial hypertension in rats after portacaval anastomosis.
species-selectivity of its actions. In addition, given the evidence Hepatology 19, 1437–1444.
that astrocyte-derived glutamine is required for the replenishment Blin, M., Crusio, W.E., Hévor, T., Cloix, J.F., 2002. Chronic inhibition of glutamine
synthetase is not associated with impairment of learning and memory in mice.
of the releasable (neurotransmitter) pool of glutamate, restriction
Brain Res. Bull. 57, 11–15.
on availability of this precursor has the potential to result in im- Bosman, D.K., Deutz, N.E., De Graaf, A.A., vd Hulst, R.W., Van Eijk, H.M., Bovée, W.M.,
paired excitatory transmission and worsening of HE. Maas, M.A., Jörning, G.G., Chamuleau, R.A., 1990. Changes in brain metabolism
Mild hypothermia delays the onset of HE and prevents brain ede- during hyperammonemia and acute liver failure: results of a comparative 1H-
NMR spectroscopy and biochemical investigation. Hepatology (12) 281–290.
ma and intracranial hypertension in both humans (Jalan et al., 1999) Boulland, J.L., Osen, K.K., Levy, L.M., Danbolt, N.C., Edwards, R.H., Storm-Mathisen, J.,
and experimental animals (Traber et al., 1989; Rose et al., 2000; Chaudhry, F.A., 2002. Cell-specific expression of the glutamine transporter SN1
Chatauret et al., 2003) with ALF. However, 13C-NMR studies in ALF suggests differences in dependence on the glutamine cycle. Eur. J. Neurosci. 15,
1615–1631.
animals demonstrate that, although mild hypothermia prevents Brusilow, S.W., Koehler, R.C., Traystman, R.J., Cooper, A.J., 2010. Astrocyte glutamine
brain edema, this protective effect is not accompanied by significant synthetase: importance in hyperammonemic syndromes and potential target
decreases of glutamine synthesis via either of the two metabolic for therapy. Neurotherapeutics 7, 452–470.
Chatauret, N., Desjardins, P., Zwingmann, C., Rose, C., Rao, K.V., Butterworth, R.F.,
pathways involving either pyruvate dehydrogenase or pyruvate car- 2006. Direct molecular and spectroscopic evidence for increased ammonia
boxylase (Fig. 8) (Chatauret et al., 2003). removal capacity of skeletal muscle in acute liver failure. J. Hepatol. 44, 1083–
Consequently, mild hypothermia does not appear to prevent 1088.
Chatauret, N., Zwingmann, C., Rose, C., Leibfritz, D., Butterworth, R.F., 2003. Effects
brain edema and its complications in ALF by prevention of the rise
of hypothermia on brain glucose metabolism in acute liver failure: a H/C-
in astrocytic glutamine. Other mechanisms have been identified nuclear magnetic resonance study. Gastroenterology 125, 815–824.
696 P. Desjardins et al. / Neurochemistry International 60 (2012) 690–696

Clemmesen, J.O., Larsen, F.S., Kondrup, J., Hansen, B.A., Ott, P., 1999. Cerebral Matheson, D.F., van den Berg, C.J., 1975. Ammonia and brain glutamine: inhibition
herniation in patients with acute liver failure is correlated with arterial of glutamine degradation by ammonia. Biochem. Soc. Trans. 3, 525–528.
ammonia concentration. Hepatology 29, 648–653. Melone, M., Varoqui, H., Erickson, J.D., Conti, F., 2006. Localization of the Na(+)-
Cooper, A.J., Mora, S.N., Cruz, N.F., Gelbard, A.S., 1985. Cerebral ammonia coupled neutral amino acid transporter 2 in the cerebral cortex. Neuroscience
metabolism in hyperammonemic rats. J. Neurochem. 44, 1716–1723. 140, 281–292.
Cooper, A.J., Stephani, R.A., Meister, A., 1976. Enzymatic reactions of methionine Melone, M., Quagliano, F., Barbaresi, P., Varoqui, H., Erickson, J.D., Conti, F., 2004.
sulfoximine. Conversion to the corresponding alpha-imino and alpha-keto acids Localization of the glutamine transporter SNAT1 in rat cerebral cortex and
and to alpha-ketobutyrate and methane sulfinimide. J. Biol. Chem. 251, 6674– neighboring structures, with a note on its localization in human cortex. Cereb.
6682. Cortex 14, 562–574.
Cubelos, B., González-González, I.M., Giménez, C., Zafra, F., 2005. Amino acid Michalak, A., Rose, C., Butterworth, J., Butterworth, R.F., 1996. Neuroactive amino
transporter SNAT5 localizes to glial cells in the rat brain. Glia 49, 230–244. acids and glutamate (NMDA) receptors in frontal cortex of rats with
Deutz, N.E., Chamuleau, R.A., de Graaf, A.A., Bovée, W.M., de Beer, R., 1988. In vivo experimental acute liver failure. Hepatology 24, 908–913.
31
P NMR spectroscopy of the rat cerebral cortex during acute hepatic Norenberg, M.D., Bender, A.S., 1994. Astrocyte swelling in liver failure: role of
encephalopathy. NMR Biomed. 1, 101–106. glutamine and benzodiazepines. Acta Neurochir. Suppl. (Wien) 60, 24–27.
Du, T., Grimm, A., Jiang, W., Desjardins, P., Butterworth, R.F., 2010. Pathogenesis of Norenberg, M.D., 1979. Distribution of glutamine synthetase in the rat central
Brain Edema in Acute Liver Failure: Key Role of the Astrocytic Glutamine nervous system. J. Histochem. Cytochem. 27, 756–762.
Transporter SNAT-3. Hepatology (52) 271A (abstract). Olalla, L., Gutiérrez, A., Jiménez, A.J., López-Téllez, J.F., Khan, Z.U., Pérez, J., Alonso,
Felipo, V., Butterworth, R.F., 2002. Neurobiology of ammonia. Prog. Neurobiol. 67, F.J., de la Rosa, V., Campos-Sandoval, J.A., Segura, J.A., Aledo, J.C., Márquez, J.,
259–279. 2008. Expression of the scaffolding PDZ protein glutaminase-interacting protein
Ferenci, P., Herneth, A., Steindl, P., 1996. Newer approaches to therapy of hepatic in mammalian brain. J. Neurosci. Res. 86, 281–292.
encephalopathy. Semin. Liver Dis. 16, 329–338. Rao, S.L., Meister, A., 1972. In vivo formation of methionine sulfoximine phosphate,
Fitzpatrick, S.M., Hetherington, H.P., Behar, K.L., Shulman, R.G., 1989. Effects of acute a protein-bound metabolite of methionine sulfoximine. Biochemistry 11, 1123–
hyperammonemia on cerebral amino acid metabolism and pHi in vivo, 1127.
measured by 1H and 31P nuclear magnetic resonance. J. Neurochem. 52, 741– Record, C.O., Buxton, B., Chase, R.A., Curzon, G., Murray-Lyon, I.M., Williams, R.,
749. 1976. Plasma and brain amino acids in fulminant hepatic failure and their
Jalan, R., O Damink, S.W., Deutz, N.E., Lee, A., Hayes, P.C., 1999. Moderate relationship to hepatic encephalopathy. Eur. J. Clin. Invest. 6, 387–394.
hypothermia for uncontrolled intracranial hypertension in acute liver failure. Rose, C., Michalak, A., Pannunzio, M., Chatauret, N., Rambaldi, A., Butterworth, R.F.,
Lancet (354) 1164–1168. 2000. Mild hypothermia delays the onset of coma and prevents brain edema
Jiang, W., Desjardins, P., Butterworth, R.F., 2009. Direct evidence for central and extracellular brain glutamate accumulation in rats with acute liver failure.
proinflammatory mechanisms in rats with experimental acute liver failure: Hepatology 31, 872–877.
protective effect of hypothermia. J. Cerebl. Blood Flow Metab. 29, 944–952. Rothstein, J.D., Tabakoff, B., 1985. Glial and neuronal glutamate transport following
Hawkins, R.A., Jessy, J., 1991. Hyperammonaemia does not impair brain function in glutamine synthetase inhibition. Biochem. Pharmacol. 34, 73–79.
the absence of net glutamine synthesis. Biochem. J. 277, 697–703. Schliess, F., Görg, B., Fischer, R., Desjardins, P., Bidmon, H.J., Herrmann, A.,
Kanamori, K., Ross, B.D., Chung, J.C., Kuo, E.L., 1996. Severity of hyperammonemic Butterworth, R.F., Zilles, K., Häussinger, D., 2002. Ammonia induces MK-801-
encephalopathy correlates with brain ammonia level and saturation of sensitive nitration and phosphorylation of protein tyrosine residues in rat
glutamine synthetase in vivo. J. Neurochem. 67, 1584–1594. astrocytes. FASEB J. 16, 739–741.
Kanamori, K., Ross, B.D., 1995. In vivo activity of glutaminase in the brain of Stravitz, R.T., Kramer, A.H., Davern, T., Shaikh, A.O., Caldwell, S.H., Mehta, R.L., Blei,
hyperammonaemic rats measured by 15N nuclear magnetic resonance. A.T., Fontana, R.J., McGuire, B.M., Rossaro, L., Smith, A.D., Lee, W.M; Acute Liver
Biochem. J. 305, 329–336. Failure Study Group, 2007. Intensive care of patients with acute liver failure:
Knecht, K., Michalak, A., Rose, C., Rothstein, J.D., Butterworth, R.F., 1997. Decreased recommendations of the U.S. Acute Liver Failure Study Group. Crit Care Med
glutamate transporter (GLT-1) expression in frontal cortex of rats with acute (35) 2498–2508.
liver failure. Neurosci. Lett. 229, 201–203. Swain, M., Butterworth, R.F., Blei, A.T., 1992. Ammonia and related amino acids in
Kuhara, T., Inoue, Y., Ohse, M., Krasnikov, B.F., Cooper, A.J., 2011. Urinary 2-hydroxy- the pathogenesis of brain edema in acute ischemic liver failure in rats.
5-oxoproline, the lactam form of a-ketoglutaramate, is markedly increased in Hepatology 15, 449–453.
urea cycle disorders. Anal. Bioanal. Chem. 400, 1843–1851. Tofteng, F., Hauerberg, J., Hansen, B.A., Pedersen, C.B., Jørgensen, L., Larsen, F.S.,
Kvamme, E., Nissen-Meyer, L.S., Roberg, B.A., Torgner, I.A., 2008. Novel form of 2006. Persistent arterial hyperammonemia increases the concentration of
phosphate activated glutaminase in cultured astrocytes and human glutamine and alanine in the brain and correlates with intracranial pressure in
neuroblastoma cells, PAG in brain pathology and localization in the patients with fulminant hepatic failure. J. Cerebl. Blood Flow Metab. 26, 21–27.
mitochondria. Neurochem Res 33, 1341–1345. Traber, P., DalCanto, M., Ganger, D., Blei, A.T., 1989. Effect of body temperature on
Kvamme, E., Svenneby, G., Hertz, L., Schousboe, A., 1982. Properties of phosphate brain edema and encephalopathy in the rat after hepatic devascularization.
activated glutaminase in astrocytes cultured from mouse brain. Neurochem. Gastroenterology 96, 885–891.
Res. 7, 670–761. Vergara, F., Plum, F., Duffy, T.E., 1974. Alpha-ketoglutaramate: increased
Laake, J.H., Slyngstad, T.A., Haug, F.M., Ottersen, O.P., 1995. Glutamine from glial concentrations in the cerebrospinal fluid of patients in hepatic coma. Science
cells is essential for the maintenance of the nerve terminal pool of glutamate: 183, 81–83.
immunogold evidence from hippocampal slice cultures. J. Neurochem. 65, 871– Warren, K.S., Schenker, S., 1964. Effect of an inhibitor of glutamine synthesis
881. (Methionine sulfoximine) on ammonia toxicity and metabolism. J. Lab. Clin.
Lavoie, J., Giguère, J.F., Layrargues, G.P., Butterworth, R.F., 1987. Amino acid changes Med. 64, 442–449.
in autopsied brain tissue from cirrhotic patients with hepatic encephalopathy. J. Willard-Mack, C.L., Koehler, R.C., Hirata, T., Cork, L.C., Takahashi, H., Traystman, R.J.,
Neurochem. 49, 692–697. Brusilow, S.W., 1996. Inhibition of glutamine synthetase reduces ammonia-
Lockwood, A.H., McDonald, J.M., Reiman, R.E., Gelbard, A.S., Laughlin, J.S., Duffy, T.E., induced astrocyte swelling in rat. Neuroscience 71, 589–599.
Plum, F., 1979. The dynamics of ammonia metabolism in man. Effects of liver Zielińska, M., Stafiej, A., Law, R.O., Albrecht, J., 2004. Effects of methionine
disease and hyperammonemia. J. Clin. Invest. 63, 449–460. sulfoximine on the glutamine and glutamate content and cell volume in rat
Mans, A.M., DeJoseph, M.R., Hawkins, R.A., 1994. Metabolic abnormalities and grade cerebral cortical slices: involvement of mechanisms not related to inhibition of
of encephalopathy in acute hepatic failure. J. Neurochem. 63, 1829–1838. glutamine synthesis. Neurotoxicology 25, 443–449.
Mans, A.M., Saunders, S.J., Kirsch, R.E., Biebuyck, J.F., 1979. Correlation of plasma Zwingmann, C., Chatauret, N., Leibfritz, D., Butterworth, R.F., 2003. Selective
and brain amino acid and putative neurotransmitter alterations during acute increase of brain lactate synthesis in experimental acute liver failure: results
hepatic coma in the rat. J. Neurochem. 32, 285–292. of a [H–C] nuclear magnetic resonance study. Hepatology 37, 420–428.
Master, S., Gottstein, J., Blei, A.T., 1999. Cerebral blood flow and the development of
ammonia-induced brain edema in rats after portacaval anastomosis.
Hepatology 30, 876–880.

Vous aimerez peut-être aussi