You are on page 1of 7

Hepatic Encephalopathy 1087

Hepatic Encephalopathy
A S Basile, DOV Pharmaceutical, Inc., Somerset, surgically constructed, or formed spontaneously as a
NJ, USA result of portal hypertension associated with cirrhosis.
K D Mullen, Case Western Reserve University, Once these gut-derived substances reach the brain, they
Cleveland, OH, USA modify CNS function either by altering metabolic
ã 2009 Elsevier Ltd. All rights reserved. processes such as oxidative metabolism or neuro-
transmitter synthesis, or by interacting directly with
neurotransmitter receptors.

Description
Hyperammonemia and HE
Hepatic encephalopathy (HE) is a complex neuro-
psychiatric syndrome characterized by the global Ammonia is produced systemically by deamination of
depression of central nervous system (CNS) activity, amino acids, in the gut wall by the action of gluta-
with a gradual deterioration of higher mental and minase, and by degradation of amines, amino acids,
motor functions (Table 1). The syndrome is asso- and urea by gut bacteria. Ammonia diffuses into the
ciated either with acute liver failure, commonly preci- portal circulation, where it would normally be con-
pitated by drug overdose or viral hepatitis, or with verted to glutamine and urea via the Krebs–Henseleit
chronic liver failure, frequently caused by alcoholic cycle. However, this process is impaired in liver fail-
cirrhosis. Liver disease is the fourth largest cause ure, resulting in elevated concentrations of ammonia
of death in Americans between 45 and 54 years of in the blood. Ammonia in the blood readily diffuses
age, and HE (including minimal HE) is associated across the blood–brain barrier in its uncharged form.
with 50–70% of all patients with cirrhosis. Moreover, Levels of ammonia in the cerebrospinal fluid (CSF) of
HE is more prevalent in nations with higher rates of patients and animals with clinically significant mani-
liver disease than occur in the United States. festations of encephalopathy secondary to liver failure
Although HE has been recognized since the begin- range from 70 to 850 mM, compared to concentrations
ning of recorded medical history, the precise patho- <50 mM in normal humans. Brain ammonia concen-
genic mechanisms responsible for the syndrome trations may reach as high as 1–5 mM in animal mod-
remain complex and poorly defined. Thus, the mod- els of acute liver failure. While total ammonia levels in
alities for treating HE are still evolving. A profusion of the blood are not well correlated with the severity of
metabolic abnormalities and gut-derived toxins result HE, brain ammonia uptake, as indicated by glutamine
from hepatic failure, resulting in significant alterations levels in CSF and by 1H magnetic resonance spectros-
in neurotransmission and CNS energy metabolism, copy, is well correlated with the severity of HE asso-
superimposed upon a characteristic astrocyte patho- ciated with cirrhosis.
logy (Alzheimer type II degeneration). Thus, it is diffi- Acute exposure to elevated levels of ammonia causes
cult to ascribe the neurological and psychiatric a number of biochemical and electrophysiological
manifestations of HE to the presence of a single agent derangements in the CNS, including suppression of
or to changes in a particular metabolic pathway. How- oxidative metabolism, inhibition of Naþ/Kþ-ATPase
ever, since HE is a metabolic encephalopathy, with few activity, inactivation of Cl extrusion pumps, and
neuroanatomical abnormalities observed in patients decreases in the equilibrium potential of the neuronal
who die of either acute or chronic liver failure, it has inhibitory postsynaptic potential (IPSP). More recent
the potential to be completely reversible. investigations indicate a significant impact of ammo-
The neurological manifestations of HE (Table 1) are nia on astrocytic function. In the presence of ammonia,
determined by two principal components of the under- astrocytes swell (Figure 2), contributing to cerebral
lying liver disease: hepatocellular failure and systemic edema in acute hepatic failure, while persistent eleva-
shunting of portal venous contents (Figure 1). Nor- tions of brain ammonia levels, as observed in chronic
mally, potentially neurotoxic substances are absorbed hepatic failure, are associated with the development
from the gut and detoxified by the liver. However, in of Alzheimer type II astrocytosis. In the absence of
hepatocellular failure these agents may avoid catabo- a functional hepatic mechanism for converting ammo-
lism by passing through the diseased liver unmetabo- nia to urea, astrocytes remove ammonia by converting
lized and into the systemic circulation (portal–systemic it to glutamine. Exposing astrocytes in vitro to elevated
shunting). Alternatively, these toxins may bypass concentrations of either ammonia or glutamine results
the liver altogether and enter the systemic circula- in increased free radical production, possibly by
tion through portal-venous shunts that are either activation of the mitochondrial permeability transition.
1088 Hepatic Encephalopathy

Table 1 Clinical stages of HE

Stage Mental state Neuromuscular state

I Mild confusion, euphoria, depression, decreased attention, slowed analytical Mild incoordination, impaired handwriting
ability, irritability, sleep inversion
II Drowsiness, lethargy, gross deficits in analytical ability, obvious personality Asterixis, ataxia, dysarthria, paratonia, apraxia
changes, inappropriate behavior, intermittent disorientation;
electroencephalogram abnormalities (high-amplitude, low-frequency
waves with nonfocal changes)
III Somnolent but rousable, unable to perform analytical tasks, disorientation Hyperreflexia, muscle spasticity, Babinski’s
with respect to time and/or place, amnesia, rage, slurred speech sign present, seizures (rare)
IVA Coma Thalamic–subcortical spasticity of entire body;
no response to painful stimuli
IVB Deep coma Death imminent

CNS
dysfunction

Astrocyte process
Brain capillary

Tight junction

Carotid
Vesicle

Inferior Simple diffusion


vena ( )
through cell, or
cava across tight
junction
( ) Vesicular transport
Facilitated diffusion
Blood-to-brain
Portacaval transport
Heptocellular
disease shunt
Food protein
Nitrogen GI hemorrhage
Portal load Drugs +
vein
Bacterial action

Intestine

Figure 1 The concept of portal–systemic shunting as it contributes to the development of HE. Nitrogenous substances arising from the
gastrointestinal (GI) system normally are absorbed into the portal venous system and delivered to the liver, where they are extracted and
metabolized. However, in liver disease these substances may pass directly into the systemic circulation or bypass the liver completely via
portal–venous shunts. Once these compounds enter the systemic circulation, they may gain entry to the central nervous system (CNS) by
a variety of mechanisms, including high-capacity active transport or facilitated diffusion, resulting in abnormal brain function. Reprinted
from Basile AS, Jones EA, Skolnick P, et al. (1991) The pathogenesis and treatment of hepatic encephalopathy: Evidence for the
involvement of benzodiazepine receptor ligands. Pharmacological Reviews 43: 27–71, with permission of the American Society for
Pharmacology and Experimental Therapeutics.
Hepatic Encephalopathy 1089

GABA

GABA-X
Glutamate

GLAST Inhibit uptake

MAPK
Glu Gln Swelling
GS
ROS

PAG
NH3 MPT

Mitochondrion

Astrocyte

Figure 2 Alterations in astrocytic function induced by hyperammonemia. Alzheimer type II astrocytosis and cerebral edema are often
observed in acute liver failure and contribute to the end stages of hepatic encephalopthy. Exposing astrocytes in culture to elevated
concentrations (5 mM) of ammonia (NH3) for 1–3 days causes them to swell. Astrocytes will try to reduce ammonia levels by fixing
ammonia to glutamate (Glu) using glutamine synthase (GS), yielding glutamine (Gln). However, ammonia may be released from
glutamine by mitochondrial phosphate-activated glutaminase (PAG), causing the production of reactive oxygen species (ROS) by the
mitochondria and induction of the mitochondrial permeability transition (MPT). Elevated ROS levels increase the phosphorylation of a
number of mitogen-activated protein kinases (MAPK), such as ERK-1/2, p39MAPK, and JNK-1/2/3. Treating astrocytes with appropriate
MAPK inhibitors or antioxidants will prevent the astrocytic swelling, glutamate uptake inhibition, and decline in glutamate–aspartate
transporter (GLAST) mRNA and protein expression. Suppression of GABA transport (GABA-X), which is reported in experimental
hyperammonemic conditions, may also be influenced by these processes and contribute to some of the manifestations of hepatic
encephalopathy.

The resulting increase in oxidative stress causes and catecholamines/false neurotransmitters. The rel-
phosphorylation of extracellular signal-regulated evance of these agents to the pathogenesis of HE
kinase-1/2 (ERK-1/2), p38 mitogen-activated protein is now mostly historical. Mercaptans, phenol, and
kinase (p38MAPK), and c-Jun N-terminal kinase-1/2/3 medium-chain-length fatty acids are present in ele-
(JNK-1/2/3). Ammonia-induced astrocytic swelling, vated concentrations in patients with liver disease.
which can be blocked in vitro by antioxidants and However, the neurotoxic concentrations of these
MAPK inhibitors, may have a number of secondary substances are 8–15 times higher than the levels
effects on neuronal function and neurotransmitter commonly observed in patients with HE. While it
levels, as detailed in following sections. has been proposed that the toxicity of these agents is
enhanced in the presence of ammonia, the concept of
‘synergistic neurotoxicity’ clearly applies to the
Other Neurotoxins
effects of all of the metabolic abnormalities and
Hyperammonemia plays a significant role in the patho- toxic compounds that arise in liver failure, and is
genesis of HE, but other conditions/agents are pres- not unique to these agents.
ent in hepatic failure/portal–systemic shunting that CNS levels of aromatic amino acids (e.g., tryptophan,
can alter CNS function. Early work attempting to phenylalanine, and tyrosine) are increased between
identify and characterize the potential neurotoxins 70% and 300% in patients with cirrhosis. These
involved in HE focused on the following classes of amino acids are precursors to several monoaminergic
agents: mercaptans, fatty acids, imbalances in the neurotransmitters, including serotonin (Table 2). The
ratio of branched-chain to aromatic amino acids, rate of serotonin turnover in discrete brain regions
1090 Hepatic Encephalopathy

Table 2 CNS levels of selected neurotransmitters in HEa

Neurotransmitter Control Liver failure Liver failure with HE

Dopamineb 2698 ng g1 caudate 2852 4774


Noradrenalineb 92 ng g1 caudate 94 154
Serotoninc 0.281 ng mg1 protein 1.397
Glutamated 9.54 mmol g1 brain 7.59**
g-Aminobutyric acid 1.46 mmol g1 braind 1.21
0.38 mmol l1 plasmae 3.37**
Benzodiazepine receptor ligandsf 78 ng DZ equiv g1 brain 405*
a
Values represent mean concentrations per unit of brain obtained during autopsy, or plasma from patients with acute or chronic liver
failure, with or without HE, or from patients with diseases other than liver failure (controls). DZ equiv., diazepam equivalents. *, **, P < 0.05,
0.01 vs. control, respectively.
b
Levels measured in samples of frontal cortex (Cuilleret et al., 1980).
c
Levels measured in samples of caudate nucleus (Bergeron et al., 1989).
d
Levels measured in samples of prefrontal cortex (Lavoie et al., 1987).
e
Median values of GABA levels measured in plasma samples from controls and patients with acute liver failure, stage IV HE (Levy and
Losowsky, 1989).
f
Levels measured in samples of frontal cortex from patients with acute liver failure (Basile et al., 1991).

increases in liver failure, possibly as a result of increased receptor complex are observed in animal models of HE
concentrations of tryptophan associated with liver fail- or in humans with liver failure. While plasma GABA
ure. However, serotonin may play only a minor role in levels are reportedly increased in liver failure (Table 2),
the development of HE (possibly in the development of they may not impact basal levels of free GABA in the
sleep inversion), since there is no correlation between CNS. Nonetheless, there is evidence that synaptic con-
the severity of HE and the concentration of serotonin or centrations of GABA may be increased, possibly as a
its metabolites. Alternatively, it has been proposed that result of ammonia-induced decreases in glial reuptake
the elevations in CNS tyrosine and phenylalanine con- (Figures 2 and 3) or alterations in release parameters.
centrations could enhance the synthesis of ‘false’ neuro- GABAergic neurotransmission may also be
transmitters, such as octopamine or tyramine, which enhanced by the presence of endogenous, positive
may serve to deplete catecholamine neurotransmitters. modulators of the GABAA receptor, including ago-
However, the levels of the false neurotransmitter octo- nists of the benzodiazepine-binding site on the
pamine decrease in the brains of patients with HE, while GABAA receptor complex and neurosteroids. Benzo-
noradrenaline and dopamine levels are generally diazepine binding site agonists are increased in sev-
unchanged or increased (Table 2). eral animal models of the syndrome and in humans
Magnetic resonance imaging (MRI) of some cir- suffering from HE due to either acute or chronic liver
rhotic patients reveals hyperintense T1 signals in the failure (Table 2, Figure 3). Tissues and body fluids
globus pallidus, suggestive of manganese accumula- from humans and animals with HE, who were other-
tion. Subsequent analysis of necropsy samples from wise never exposed to 1,4-substituted benzodiaze-
cirrhotics indicated a 128% increase in manganese pines, contain numerous benzodiazepine binding
levels. Manganese is neurotoxic and causes Alzheimer site ligands, as determined by radiometric techni-
type II astrocytic changes. However, the degree of ques. Only two of these were chemically and
pallidal hyperintensity resulting from liver failure immunochemically identified as 1,4-benzodiazepines.
does not, in itself, appear to be associated with the However, these compounds comprised less than 20%
severity of HE. of the total concentration of benzodiazepine binding
site ligands present in HE in both animal models and
Enhancement of GABAergic humans (Table 2, Figure 3). The identification and
source of the remaining benzodiazepine binding-site
Neurotransmission and HE
ligands is unclear, but metabolites of hemoglobin,
g-Aminobutyric acid (GABA) is the primary inhibi- including hemin and protoporphyrin IX, are possible
tory neurotransmitter in the CNS, and overactivation candidates.
of this pathway results in behavioral changes consistent Alterations in astrocytic function induced by
with the primary manifestations of HE, specifically ammonia may increase the production of neuroster-
motor dysfunction, cognitive deficits, and altered oids such as allopregnanolone, a potent inhibitor of
consciousness. No consistent changes in the GABAA neuronal activity acting through the GABAA receptor.
Hepatic Encephalopathy 1091

Plasma GABA Ammonia BZR agonists

CNS

D Astrocytic
function

BBB permeability (FHF)


Neurosteroid
GABA uptake
synthesis

GABA concentrations
in synaptic cleft

GABAA receptor
activation

Neuronal depression

Hepatic encephalopathy
Figure 3 GABAergic neurotransmission is enhanced in hepatic failure through a variety of mechanisms. g-Aminobutyric acid (GABA),
benzodiazepine receptor (BZR) agonists, and particularly ammonia, originating from outside of the brain (top tier), make their way into the
central nervous system (CNS) either through diffusion or via a permeabilized blood–brain barrier (BBB). Elevations in CNS ammonia
levels alter astrocytic function, which may lead to swelling and permeabilization of the BBB and fulminant hepatic failure (FHF), often
observed in acute liver failure. Astrocytes in a hyperammonemic state also show a reduction in GABA uptake. The increased access of
plasma GABA to the CNS, combined with decreased GABA uptake by astrocytes and evidence of enhanced GABA release, increases the
availability of GABA at the synapse. In addition, the presence of positive modulators of the GABAA receptor, such as neurosteroid and
benzodiazepine receptor agonists, as well as ammonia itself, yields a net increase in GABAergic neurotransmission. These changes, in
combination with the suppression of oxidative metabolism of neurons by elevated ammonia levels, cause neuronal depression, which is
expressed as the core manifestation of HE (see Table 1).

The peripheral-type benzodiazepine receptor (PBR), binding to sites on the GABAA receptor confirmed and
located on glial mitochondria, plays a role in neuro- extended these observations. Pathophysiologically rel-
steroid synthesis. The density of PBRs is increased in evant concentrations of ammonia selectively increased
animal models of HE, and in postmortem samples the density of agonist (flunitrazepam) ligand binding
from humans with hepatic failure, but interestingly to the GABA-binding site of the GABAA receptor com-
is not observed using positron emission tomography plex, with no effect on antagonist (flumazenil) ligand
(PET) imaging in patients with HE. Nonetheless, allo- binding. Further, ammonia increased the affinity of
pregnanolone has been measured in postmortem sam- agonist binding to the benzodiazepine-binding site on
ples of brain from patients with HE in concentrations the GABAA receptor complex, without altering antag-
sufficient to enhance GABA-gated chloride currents, onist binding. The latter effect was further enhanced
which may serve to depress neuronal activity. (threefold) in the presence of GABA. It appears that
Evidence also indicates that ammonia can enhance the ability of ammonia to modulate ligand binding to
GABAergic neurotransmission through direct actions the GABAA receptors (a heterooligomeric complex
at the GABAA receptor. An electrophysiological study composed of members from five families of subunits)
using isolated cortical neurons indicated that the depends on the presence of the b subunit, as ammonia
maximal level of GABA-activated current is enhanced can enhance [3H]flunitrazepam binding to recombi-
by pathophysiologically relevant concentrations of nant GABAA receptors composed of a1b2g2 subunits
ammonia, as is the potency of GABA. Subsequent (the pharmacology of which most resembles that of the
investigations of the effects of ammonia on radioligand native GABAA receptor) but has no effect on binding
1092 Hepatic Encephalopathy

to a1g2 constructs. Taken together, the numerous from the gut. Restricting oral protein consumption in
neurochemical changes present in hepatic failure may order to reduce a source of ammonia was a primary
enhance GABAergic neurotransmission, leading to the treatment for decades, but is now discouraged because
motoric and behavioral changes characteristic of HE. it aggravates lean body wasting in cirrhosis. Another
approach to reducing gut ammonia production from
dietary sources is to prescribe less encephalopatho-
Suppression of Glutamatergic
genic, vegetable-based protein diets. Nonabsorbable
Neurotransmission and HE antibiotics and cathartics (lactulose and lactitol) have
Contrasting with GABA, glutamate is the primary also been used to reduce ammonia production and
excitatory neurotransmitter. Accumulating evidence absorption. In addition to its antibiotic actions, neo-
suggests that there are alterations in glutamatergic mycin may reduce ammonia production by inhibiting
neurotransmission in acute liver failure. Total brain intestinal glutaminase. Bacterial overgrowth in the
glutamate levels are decreased under hyperammone- small intestine and other disturbances of the micro-
mic conditions, including liver failure, consistent with ecology of indigenous gut flora are common in cirrho-
the conversion of glutamate to glutamine by astro- sis, and may impact toxin/ammonia production. The
cytes in order to reduce ammonia levels. Basal extra- ammonia-lowering actions of lactulose/lactitol may
cellular glutamate levels increase in models of acute be due in part to their effects on gut flora. However,
hepatic failure, but not in the portal–systemic shunted despite their widespread use, controlled trials have
rat model of chronic hepatic failure. In the latter model, indicated that lactulose/lactitol and neomycin therapies
stimulated release of glutamate is variably increased. are not superior to placebo in the treatment of HE. The
Glial glutamate transporter (GLT-1 and/or GLAST) utility of these agents in the treatment of HE is cur-
expression is transiently decreased in portal–systemic rently being reassessed using randomized, placebo-
shunted rats and is more consistently decreased in rats controlled trials. Similarly, the efficacy of modulating
with acute liver failure. Glutamate uptake by astrocytes intestinal flora, such as through the administration of
in vitro is also reduced in the presence of ammonia. Enterobacter faecium SF 68, in treating HE is receiving
Thus, there is evidence for increased extracellular renewed interest.
glutamate levels in acute liver failure, resulting from Another mode of therapy is to enhance the excre-
either increased release or decreased reuptake. There is tion of waste nitrogen from the body in patients
no consistent evidence of a change in N-methyl-D- with liver failure. Sodium benzoate, phenylacetate,
aspartate (NMDA) receptor density in animal models and L-ornithine-L-aspartate (LOLA) promote this
of acute liver failure. process through a variety of mechanisms. LOLA
The specific contribution(s) of the alterations in glu- may improve HE by lowering ammonia levels and
tamatergic neurotransmission to the manifestations of suppressing ammonia-induced neurotoxicity by
HE is unclear. It has been proposed that increased stimulating urea cycle function. Moreover, LOLA is
glutamate levels may contribute to glial swelling and more effective than placebo in clinical trials.
cerebral edema, but there is no correlation between the Other therapies for HE shown to be effective in
onset of glutamate elevations and increased intracranial placebo-controlled trials include acarbose and the
pressure. Similarly, changes in glutamate levels may centrally acting benzodiazepine receptor antagonist,
contribute to the sleep-inversion, extrapyramidal disor- flumazenil. There are a number of anecdotal reports
ders and to the cognitive deficits observed in HE, par- of the efficacy of flumazenil in treating patients
ticularly when associated with chronic liver failure. suffering from HE. It has been claimed that the rever-
However, the evidence for alterations in extracellular sal of coma by flumazenil in these reports reflects the
glutamate levels in animal models of chronic liver abuse of benzodiazepines by patients with liver dis-
failure is variable, and the role of glutamatergic neuro- ease. However, these claims do not take into account
transmission in the aforementioned behavioral changes that endogenous benzodiazepine receptor ligands
is weak. Nonetheless, the significance of glutamatergic have been found in multiple animal models of HE.
neurotransmission to CNS function is so great as to Moreover, a number of placebo-controlled trials of
warrant continued investigation of its role in HE. the efficacy of flumazenil in controlled patient popu-
lations have been conducted. Typically, low doses
(1–5 mg) of flumazenil reverse HE in 30% of patients
Therapy
within 0.5–5 min of administration. In a meta-analy-
Therapeutic strategies for allaying the manifestations sis of 12 randomized trials, eight with cross-over
of HE may focus on the target organ, the CNS, and the design, flumazenil was associated with a significant
gut. Current, standard therapeutic regimens are dir- improvement of HE (30%) compared to placebo
ected at reducing ammonia production and absorption (7%), particularly in a subpopulation of patients
Hepatic Encephalopathy 1093

with low-grade encephalopathy. While no follow-up Bai G, Rama Rao KV, Murthy CRK, et al. (2001) Ammonia
therapy was employed in these studies, long-term induces the mitochondrial permeability transition in primary
cultures of rat astrocytes. Journal of Neuroscience Research
suppression of HE has been reported with chronic 66: 981–991.
oral administration of flumazenil (25 mg). Develop- Barbaro G, DiLorenzo G, Soldini M, et al. (1998) Flumazenil for
ment of a long-acting, orally available form of a benzo- hepatic encephalopathy grade III and IVa in patients with cir-
diazepine antagonist should enhance the use of this rhosis: An Italian multicenter double-blind, placebo-controlled,
cross-over study. Hepatology 28: 374–378.
therapy for HE.
Basile AS (2002) Direct and indirect enhancement of GABAergic
Finally, can the evidence implicating ammonia in neurotransmission by ammonia: Implications for the pathogen-
the enhancement of GABAergic neurotransmission esis of hyperammonemic syndromes. Neurochemistry Inter-
be used to develop therapeutic modalities for treating national 41: 115–122.
HE? An agent that can reverse ammonia’s enhance- Basile AS, Hughes R, Harrison P, et al. (1991) Brain concentra-
ment of GABA and benzodiazepine receptor function tions of 1,4 benzodiazepines are elevated in patients with fulmi-
nant hepatic failure. New England Journal of Medicine 325:
with minimal convulsant activity would be desirable. 473–478.
Such an agent may be an antagonist of the neuroster- Basile AS, Jones EA, and Skolnick P (1991) The pathogenesis and
oid-binding site on the GABAA receptor complex, treatment of hepatic encephalopathy: Evidence for the involve-
such as pregnenolone. As previously noted, ammonia ment of benzodiazepine receptor ligands. Pharmacological
Reviews 43: 27–71.
enhances the binding of [3H]flunitrazepam. Perfor-
Bergeron M, Reader TA, Layrargues GP, et al. (1989) Monoamines
ming these assays in the presence of pregnenolone and metabolites in autopsied brain tissue from cirrhotic patients
sulfate completely abolished the ability of ammonia with hepatic encephalopathy. Neurochemical Research 14:
to enhance the binding of [3H]flunitrazepam. Similar 853–859.
effects were seen on ligand binding to the GABA site Conn HO and Burcher J (1989) Hepatri Encephalopathy: Manage-
on the GABAA receptor complex. These preliminary ment with Lactulose and Related carbohydrates. East lansing,
MI: MediEd Press.
observations suggest a potential use for neurosteroid Cooper AJ and Plum F (1987) Biochemistry and physiology of
antagonists in reversing some of the psychomotor brain ammonia. Physiological Review 67: 440–519
symptoms of HE. Interestingly, present therapeutic Cuilleret G, PomierLayrargues G, Pons F, et al. (1980) Changes in
protocols aimed at reducing ammonia levels by restrict- brain catecholamine levels in human cirrhotic hepatic encephalo-
ing dietary intake and modifying colonies of enteric pathy. Gut 21: 565–569.
Jayakumar AR, Panickar KS, Murthy CRK, et al. (2006) Oxidative
bacteria may also reduce the synthesis or uptake of stress and mitogen-activated protein kinase phosphorylation
benzodiazepine receptor ligands or their precursors. mediate ammonia-induced cell swelling and glutamate uptake
inhibition in cultured astrocytes. Journal of Neuroscience 26:
4774–4784.
Conclusion Klos KJ, Ahlskog JE, Josephs KA, et al. (2005) Neurologic spec-
In summary, multiple factors contribute to the patho- trum of chronic liver failure and basal ganglia T1 hyperintensity
on magnetic resonance imaging: Probable manganese neuro-
genesis of HE, including agents that impair oxidative toxicity. Archives of Neurology 62: 1385–1390.
metabolism or neurotransmitter synthesis in the CNS, Lavoie J, Giguere JF, Layrargues GP, et al. (1987) Amino-acid
or which interact directly with neurotransmitter recep- changes in autopsied brain tissue from cirrhotic patients
tors. The principal effect of this is to suppress neuronal with hepatic encephalopathy. Journal of Neurochemistry 49:
activity, in part by enhancing GABAergic neuro- 692–697.
Levy LJ and Losowsky MS (1989) Plasma gamma aminobutyric
transmission. The renewed emphasis on the role of acid concentrations provide evidence of different mechanisms in
abnormal CNS function in the manifestations of the pathogenesis of hepatic encephalopathy in acute and chronic
HE suggests that future therapeutic modalities for this liver disease. Hepatogastroenterology 36: 494–498.
disorder will focus upon the brain as well as the gut. Norenberg MD (2001) Astrocytes and ammonia in hepatic enceph-
alopathy. In: de Vellis J (ed.) Neuroglia in the Aging Brain, pp.
See also: GABAA Receptors: Developmental Roles; 477–496. Totowa, NJ: Humana Press.
Olasmaa M, Rothstein JD, Guidotti A, et al. (1990) Endogenous
Gamma-Aminobutyric Acid (GABA); Glutamatergic and
benzodiazepine receptor ligands in human and animal hepatic
Gabaergic Systems; Neural Stem Cells and CNS
encephalopathy. Journal of Neurochemistry 55: 2015–2023.
Diseases; Neurotoxins; Neurotoxins and their Rose C, Butterworth RF, Zayed J, et al. (1999) Manganese deposi-
Neurotoxicology. tion in basal ganglia structures results from both portal-
systemic shunting and liver dysfunction. Gastroenterology
117: 640–644.
Further Reading Ruscito BJ and Harrison NL (2003) Hemoglobin metabolites
mimic benzodiazepines and are possible mediators of hepatic
Als-Nielsen B, Kjaergard LL, and Gluud C (2004) Benzodiazepine encephalopathy. Blood 102: 1525–1528.
receptor antagonists for acute and chronic hepatic encephalo- Scollo-Lavizzari G and Steinmann E (1985) Reversal of hepatic coma
pathy. Cochrane Database of Systematic Reviews 2CD002798. by benzodiazepine antagonist (Ro 15-1788). Lancet 1: 1324.

You might also like