You are on page 1of 22

PRIMER

Hepatic encephalopathy
Dieter Häussinger   1 ✉, Radha K. Dhiman   2, Vicente Felipo   3, Boris Görg   1,
Rajiv Jalan4,5, Gerald Kircheis   6, Manuela Merli   7, Sara Montagnese   8,
Manuel Romero-Gomez   9, Alfons Schnitzler   10, Simon D. Taylor-Robinson   11
and Hendrik Vilstrup12
Abstract | Hepatic encephalopathy (HE) is a prognostically relevant neuropsychiatric syndrome
that occurs in the course of acute or chronic liver disease. Besides ascites and variceal bleeding,
it is the most serious complication of decompensated liver cirrhosis. Ammonia and inflammation
are major triggers for the appearance of HE, which in patients with liver cirrhosis involves patho-
physiologically low-grade cerebral oedema with oxidative/nitrosative stress, inflammation and
disturbances of oscillatory networks in the brain. Severity classification and diagnostic approaches
regarding mild forms of HE are still a matter of debate. Current medical treatment predominantly
involves lactulose and rifaximin following rigorous treatment of so-called known HE precipitating
factors. New treatments based on an improved pathophysiological understanding are emerging.

Hepatic encephalopathy (HE) is a neuropsychiatric syn- somnolence is present in grade III HE and a coma-like
drome that occurs in patients with acute or chronic liver state in grade IV. HE can also be classified according to
disease. Symptoms of HE largely comprise cognitive and the time course of mental alteration as episodic, recur-
fine-motor disturbances of varying severity, which can rent (more than one bout of overt HE within 6 months)
be ascribed to a slowing of cerebral oscillatory networks. or persistent (if the patient does not return to normal
HE is categorized as type A, B or C, and graded as mental performance between bouts of overt HE).
minimal, grades I, II, III and IV1. HE in patients with Most cases of HE occur in patients with liver cirrhosis.
acute liver failure is classified as HE type A. HE pri- In these patients, HE is indicative of a poor progno-
marily or exclusively caused by portosystemic shunts sis and is associated with a reduction of health-related
is classified as type B HE; such shunts arise spontane- QOL (HRQOL). HE in patients with liver cirrhosis is
ously as a result of portal vein hypertension and allow accompanied by low-grade cerebral oedema. By contrast,
blood from portal vein-drained viscera to bypass the overt brain oedema is an extremely serious and often
liver and directly enter the systemic circulation1,2. HE fatal complication of acute liver failure or congenital
caused primarily or exclusively by loss of functional liver hyperammonaemic disorders.
mass due to cirrhosis is classified as type C HE1, and HE HE disrupts personality, self-reliance and capability
in patients with acute-on-chronic liver failure (ACLF) for everyday living. The experience of HE is highly dis-
is also considered type C HE. However, HE in patients tressing and perceived by the patient as the experience
with ACLF is clinically, pathophysiologically and prog- of multiple losses, such as of independency and social
nostically distinct from HE in patients without ACLF. interaction, and a sustained fear of recurrence8. This,
Typically, HE in those with ACLF is characterized by together with the weakening of cognitive coherence,
more frequent cerebral oedema, marked disturbances gives rise to a serious loss in QOL. The personality dis-
in cerebral oxygenation, systemic inflammation and ruption also poses widespread distress, uncertainty and
neuroinflammation, higher ammonia levels, and risk anxiety to caregivers, particularly those in the family9,10.
of short-term mortality3–6. This type of liver failure is Ammonia and inflammation are involved in the
defined by the occurrence of additional organ failure pathogenesis of HE, and recent studies have provided
such as renal, immunological or circulatory failure3. novel mechanistic insight into ammonia toxicity and the
Minimal HE (MHE) is clinically undiscernible and pathobiochemistry/pathophysiology of HE. This Primer
requires psychometric methods for detection. However, largely focuses on HE in the patient with cirrhosis
despite its innocuous presentation, it is closely associ- because most HE cases are found in patients with liver
✉e-mail: haeussin@ ated with severe loss of quality of life (QOL)7. Grade I cirrhosis in whom low-grade cerebral oedema in combi-
uni-duesseldorf.de HE is the mildest clinically detectable form. Grade II nation with oxidative/nitrosative stress play a prominent
https://doi.org/10.1038/ or higher is referred to as clinically manifest or overt pathogenetic role. Although many studies on HE and
s41572-022-00366-6 HE and is distinguished by disorientation, whereas ammonia toxicity were conducted in animal models, key


NATURE REVIEwS | DISEASE PRIMERS | Article citation ID: (2022) 8:43 1

0123456789();:
Primer

Author addresses The exact number of these patients who develop HE


is not known. The prevalence of MHE is 40–60%
1
Department of Gastroenterology, Hepatology and Infectious Diseases, Medical Faculty in patients with cirrhosis16,17; within 1 year, ~33% of
and University Hospital Düsseldorf, Heinrich Heine University Düsseldorf, Düsseldorf, patients with cirrhosis and MHE progress to clinically
Germany. manifest HE. The estimated prevalence of grade I HE is
2
Department of Hepatology, Sanjay Gandhi Postgraduate Institute of Medical Sciences,
15–25% in patients with cirrhosis16, of whom 50% pro-
Lucknow, (Uttar Pradesh), India.
3
Laboratory of Neurobiology, Centro de Investigación Principe Felipe, Valencia, Spain. gress to higher grades of clinically manifest HE within
4
Liver Failure Group ILDH, Division of Medicine, UCL Medical School, Royal Free Campus, 1 year16. Clinically manifest HE is present in 10–15% of
London, UK. patients with cirrhosis at diagnosis. In 40% of patients
5
European Foundation for the Study of Chronic Liver Failure, Barcelona, Spain. with cirrhosis and HE grade II or above, HE will recur
6
Department of Gastroenterology, Diabetology and Hepatology, University Hospital within 1 year18,19. Persistent HE is the rarest form of HE.
Brandenburg an der Havel, Brandenburg Medical School, Brandenburg an der Havel, No good data on the prevalence of this type are availa-
Germany. ble but patients are intensively pharmacologically treated
7
Department of Translational and Precision Medicine, Universita’ degli Studi di and, in some cases, need liver transplantation. Most
Roma – Sapienza, Roma, Italy. patients with persistent HE have advanced cirrhosis with
8
Department of Medicine, University of Padova, Padova, Italy.
extensive portosystemic shunting.
9
UCM Digestive Diseases, Virgen del Rocío University Hospital, Institute of Biomedicine
of Seville (HUVR/CSIC/US), University of Seville, Seville, Spain. HE occurs in ~60% of patients with ACLF and is con-
10
Institute of Clinical Neuroscience and Medical Psychology, Heinrich Heine University sidered part of the organ failure count that defines the
Düsseldorf, Düsseldorf, Germany. diagnosis and prognosis of ACLF20.
11
Department of Surgery and Cancer, St. Mary’s Hospital Campus, Imperial College
London, London, UK. Burden of disease
12
Department of Hepatology and Gastroenterology, Aarhus University Hospital, The institutional health-care cost of HE is high owing
Aarhus, Denmark. to the large number of patients at risk, frequent hospital
admissions, and the need for close monitoring, inten-
findings from these studies have also been confirmed in sive care support and prolonged periods of hospital-
the human brain. HE symptoms are of varying severity ization. Estimates of the cost of HE are available only
and there is an ongoing debate regarding severity assess- from the USA with limited data from low-income and
ment and nomenclature of HE. This, and the dynamics middle-income countries. In the USA, costs of HE rose
of HE, which can resolve after correction of precipitating by 30% from 2010 to 2014, to US$ 12 billion per year21.
factors, make clinical studies on the efficacy of medical Costs will continue to grow owing to the increasing
treatment options more difficult. incidence of cirrhosis. The hospital cost of admissions
with HE exceeds that of cardiac failure and chronic
Epidemiology pulmonary disease by 50%22.
The epidemiology of HE is not well established as diag-
nostic criteria are not unanimous, patients can have dif- Risk factors
ferent types of liver disease, HE does not have a WHO Risk factors of HE (Table 1) can be classified according to
International Classification of Diseases code, and few the organs involved in HE development, including liver,
population-based studies of relevant format have been portal hypertension, kidney, gut–liver axis, genetic back-
carried out. Furthermore, prevalence estimates may be ground, drugs and accompanying diseases (reviewed in
inaccurate as HE is variable over time. Nonetheless, the ref.18). Risk factors can be stratified as predisposing or
available data show that HE is the most common, dev- precipitating factors.
astating, resource-demanding complication of cirrhosis
and is closely associated with a poor prognosis11. Predisposing risk factors. Levels of albumin (<3.5 g/dl)
and bilirubin (≥2.1 mg/dl) and the Model for End-
Prevalence and incidence stage Liver Disease (MELD) score can predict HE23–25.
Type A HE is the most important clinical event defin- A patient without previous episodes of HE or any
ing acute liver failure; therefore, the incidence of HE genetic risk has an ~44% risk of an HE episode within
type A is largely the same as that of acute liver failure12. 3 years with albumin ≥40 g/dl and bilirubin >8 mg/dl or
Acute liver failure is rare, with a reported incidence of with albumin <20 g/dl and bilirubin ≤1.8 mg/dl (ref.25).
~0.5 cases per 100,000 individuals per year12. Nevertheless, the MELD score, which is widely used
Type B HE is uncommon but occurs more frequently for prognostication of patients with cirrhosis, does not
than type A HE. Incidence estimates of type B HE are correlate with HE severity26.
based on the conditions that most often cause porto- A cross-sectional area (that is, sum of cross-sectional
systemic shunting such as portal vein thrombosis. Of area of all detected shunts) of >83 mm2 of portosystemic
these patients, 6–13% experience HE at some point13. shuntings is related to risk of HE and shorter survival27.
HE after placement of a transjugular intrahepatic porto- In patients with liver impairment, a spontaneous porto­
systemic stent shunt (TIPSS) to ameliorate severe portal systemic shunt increased morbidity and mortality27.
hypertension, particularly in patients with cirrhosis, is Although TIPSS increased the incidence of HE by
considered a mixture of HE types B and C. The 1-year 10–50% at 1 year28, covered stents29 and placement of
post-TIPSS incidence of HE ranges from 10% to 50%14. early (pre-emptive) TIPSS, to avoid variceal bleeding in
Type C HE is the most common subtype. The global patients at high risk, seem to reduce the incidence of new
burden of cirrhosis is of at least 125 million patients15. bouts of HE30,31.

2 | Article citation ID: (2022) 8:43 www.nature.com/nrdp

0123456789();:
Primer

Genetic risk factors for HE have been identified. whereas cerebral oedema is low grade in HE in chronic
Patients bearing variants in the promoter of GLS1 liver failure 49–51. Moreover, glutamatergic neuro­
(encoding kidney-type glutaminase) showed increased transmission is enhanced in HE in acute liver failure,
incidence of overt HE, linking genes with an increased resulting in neuroexcitation, whereas it is impaired in
risk in patients with previous bouts of overt HE18,30. chronic liver failure and inhibitory neurotransmission
(neurodepression) tends to be enhanced52.
Precipitating risk factors. Several precipitating risk Ammonia (which can readily cross the blood–brain
factors of HE have been identified. These factors barrier (BBB) in its protonated and deprotonated form53)
include various diseases such as acute kidney injury, is insufficiently eliminated in those with liver cirrhosis,
hepatorenal syndrome, gastrointestinal bleeding3, dia- and together with inflammation, has a major role in the
betes mellitus32–35, epilepsy36, and malnutrition and pathogenesis of HE. Other involved mechanisms are
sarcopenia37. Other precipitating factors are systemic hyponatraemia, benzodiazepines, neurosteroids, man-
inflammation38–40, disruption of gut–liver axis41,42, consti- ganese, mercaptans, bilirubin, zinc, phenols, short-chain
pation and small intestinal bacterial overgrowth43, drug fatty acids, bile acids and amino acid imbalances (low
use (alcohol consumption, proton-pump inhibitors44,45 branched-chain amino acid to aromatic amino acid
and drugs targeting the central nervous system, mainly ratio) (for reviews, see refs11,54–62).
benzodiazepines, GABAergic drugs and opioids)18 and
more advanced age46 (Table 1). Pathobiochemistry of hyperammonaemia
Ammonia is predominantly produced in the small
Mechanisms/pathophysiology bowel through the action of enterocytic glutaminase as
Several animal models of HE have been established47,48. well as in the colon through the action of gut bacteria.
Hallmarks of HE in acute and chronic liver failure are This ammonia is removed primarily by the liver with its
astrocyte swelling, cerebral oxidative stress, microglial sophisticated structural and functional organization of
activation and altered neurotransmission. However, ammonia-metabolizing pathways in the liver acinus, which
cerebral oedema is frequent in HE in acute liver failure, is the functional unit of the liver (for reviews, see refs63,64).

Table 1 | Risk factors for HE


Organs Risk factors Association with HE Average risk HR Refs
involved (95% CI)
Predisposing factors
Liver Liver dysfunction Bilirubin level 1.184 (1.04–1.36) 28

Albumin level 0.93 (0.89–0.97)


SPSS Spontaneous portosystemic shunts Total area >83 mm2 1.83 (1.14–2.93) 27

Genes Genetic background Glutaminase gene 2.1 (1.17–3.79) 25,326

Previous episode of overt HE Personal risk 4.22 (3.30–5.41) 3

Precipitating factors
Gut–liver axis Variceal bleeding Not associated (?) 0.52 (0.37–0.73) 3

Constipation and SIBO Hydrogen-SIBO 3.14 (1.05–9.34) 43,327

Infections SBP, pneumonia, cellulitis, UTI 3.0 (2.4–3.8) 39

Dysbiosis Cirrhosis to dysbiosis ratio Controls: 2.05 vs 42

decreased cirrhosis: 1.2 vs HE: 0.42


Kidneys Renal insufficiency AKI–HRS 1.01 (1.00–1.02) 28

Hyponatraemia 0.08 for each mmol/l decrease 10.7 (4.4–26.0) 24

TIPSS TIPS placement, TIPSS Associated 1.66 (1.31–2.11) 29,31,


33,328
Early TIPSS and covered stent Not associated 1.08 (0.84–1.38)
Drugs CNS, drug use Benzodiazepines 1.24 (1.21–1.27) 18,44,45

GABAergics 1.17 (1.14–1.21)


Opioids 1.24 (1.21–1.27)
Proton-pump inhibitors 1.41 (1.38–1.46)
Alcohol Alcohol consumption 1.44 (1.40–1.47)
Pancreas T2DM control HBA1c% 4.11 (1.79–9.46) 32,33,329

Brain Epilepsy Definite epilepsy 6.12 (1.47–25.44) 36

Skeletal Sarcopenia Skeletal muscle index 18.3 (2.82–29.3) 37

muscle
Age Age Older age 1.034 (1.016–1.053) 46

AKI, acute kidney injury; CNS, central nervous system; HE, hepatic encephalopathy; HRS, hepatorenal syndrome; SBP, spontaneous
bacterial peritonitis; SIBO, small intestinal bacterial overgrowth; SPSS, spontaneous portosystemic shunt; T2DM, type 2 diabetes
mellitus; TIPS, transjugular intrahepatic portosystemic stent; TIPSS, transjugular intrahepatic portosystemic stent shunt; UTI, urinary
tract infection.


NATURE REVIEwS | DISEASE PRIMERS | Article citation ID: (2022) 8:43 3

0123456789();:
Primer

Periportal hepatocytes eliminate ammonia by urea glutamine synthetase-mediated condensation of ammo-


synthesis, which requires ammonia amplification by nia and glutamate to glutamine66. Hyperammonaemia
mitochondrial glutaminase (GLS2) because of the low in liver cirrhosis leads to osmotic stress, in part due to
ammonia affinity of carbamoylphosphate synthetase. intra-astrocytic glutamine accumulation with develop-
Upregulation of this amplification process in liver cir- ment of low-grade cerebral oedema, which is counter-
rhosis maintains a life-compatible rate of urea synthe- acted by the compensatory release of organic osmolytes,
sis, despite an 80% decrease in the ureogenic capacity such as myo-inositol, by astrocytes49,67. However, deple-
(for details, see ref.64). Ammonia that escapes periportal tion of this osmolyte pool in astrocytes restricts the cell
urea synthesis is eliminated with high affinity by a small volume-regulatory capacity and renders astrocytes vul-
perivenous hepatocyte population (so-called perivenous nerable to other cell volume challenging agents, which
scavenger cells) via glutamine synthesis. In liver cirrho- exacerbate the low-grade cerebral oedema and thereby
sis, the capacity of this hepatocyte population to elim- induce oxidative and nitrosative stress in astrocytes.
inate ammonia before sinusoidal blood reaches the Evidence supporting low-grade cerebral oedema in
systemic circulation is strongly impaired and hyperam- patients with liver cirrhosis and HE came from studies
monaemia ensues. In line with this, liver-specific dele- employing 1H-MR spectroscopy49–51,67 and quantitative
tion of glutamine synthetase in mice results in lifelong water imaging of the brain68,69, whereas the presence
systemic hyperammonaemia47. These scavenger cells of oxidative/nitrosative stress in brains from patients
not only exclusively express glutamine synthetase in the with HE was shown in studies using post mortem brain
liver but also specifically express ornithine aminotrans- samples70 (for a review, see ref.71).
ferase and uptake systems for aspartate, glutamate and Based on available knowledge, HE can be viewed as
related dicarboxylates65, which can provide the carbon the clinical manifestation of a pathogenetic interplay
skeleton for glutamine synthesis and thereby a rationale between osmotic stress and oxidative and nitrosative
for their supplementation. Additionally, muscles contain stress in astrocytes49,71,72. This interplay is triggered by
glutamine synthetase and can contribute to ammonia ammonia, pro-inflammatory cytokines, hyponatraemia
detoxication. (low serum sodium levels) and benzodiazepines73–77.
Accordingly, astrocyte swelling and the formation of
Osmotic and oxidative/nitrosative stress reactive oxygen and nitrogen species (RONS) represent
Overwhelming in vitro and in vivo evidence points to a a final common path of action of conditions precipitat-
central role of astrocytes in the pathogenesis of ammo- ing HE (including excessive protein intake, bleeding,
nia toxicity and HE. Astrocytes are the major cell type trauma, infections, sedatives, metabolic acidosis, diuretic
involved in the removal of ammonia, which occurs via overdose, renal insufficiency and hyponatraemia)49,73,74,78.
Swelling and RONS formation mutually amplify each
other in astrocytes, and HE precipitating factors act
Hyponatraemia Inflammatory cytokines Ammonia Benzodiazepines synergistically in the induction of astrocyte swelling
and RONS formation78,79. RONS originating from other
cell types in the brain, such as neurons, microglia and
Swelling endothelial cells80–82, or from outside the brain83 can
also contribute to astrocyte swelling. The interplay
between osmotic and oxidative and nitrosative stress
Astrocyte triggers post-translational protein modifications, RNA
oxidation, senescence, altered signalling and altered
RONS gene expression (Fig. 1). These alterations have been
found in brains of patients with liver cirrhosis and HE
but not in brains of patients with liver cirrhosis without
HE. These alterations are thought to affect astrocytic
• Signalling
Gene expressiona RNA oxidation • Senescence Protein modifications and neuronal function and synaptic plasticity and trig-
ger disturbances of oscillatory networks in the brain,
Astrocytic/neuronal dysfunction which, ultimately, lead to cognitive and motor symptoms
of HE72,84,85.
Synaptic plasticity
Origin of oxidative/nitrosative stress
Oscillatory networks
Induction of oxidative and nitrosative stress by pre-
Symptoms cipitating factors for HE in cultured rat astrocytes is
triggered by N-methyl-d-aspartate receptor (NMDAR)-
Fig. 1 | Model for the pathogenesis of HE. Precipitating factors for hepatic encephalopathy dependent elevation of the intracellular calcium con-
(HE) trigger astrocyte swelling and oxidative/nitrosative stress in astrocytes, which mutually centration [Ca2+]i (refs75–77,86) (Fig.  2). This NMDAR
enhance each other. This results in covalent modification of proteins and RNA, senescence,
activation may originate from membrane depolari-
and changes in cerebral gene expression, which lead to astrocyte and neuronal dysfunction,
impaired synaptic plasticity, and disturbance of oscillatory networks in the brain, which zation87 and/or mechanical tension of the membrane
account for HE symptoms. RONS, reactive oxygen and nitrogen species. aGenes with altered triggered by astrocyte swelling 88. Cytosolic phos-
expression in post mortem brain samples from patients with liver cirrhosis with HE relate pholipase A 2 (cPLA 2)-dependent arachidonic acid
to oxidative stress, inflammatory pathways, microglial activation, receptor signalling, release further amplifies [Ca2+]i through exocytosis of
cell proliferation, apoptosis and others97. Adapted from ref.330, Springer Nature Limited. l-glutamate-containing vesicles89 (Fig. 2).

4 | Article citation ID: (2022) 8:43 www.nature.com/nrdp

0123456789();:
Primer

HE-precipitating factors (for example, ammonia, inflammatory cytokines)


Mg2+
Glu

MRP4 NMDAR 1
TGR5 Extracellular
GS Cytosol
Membrane
AC stretch cPLA2
Neurosteroids
Ca2+ Arachidonic acid
PBR
Prostanoids 2
GLS
Gln Glu + NH4+
cAMP NOX2 iNOS nNOS 3

Mitochondrion
Phagosome O2– 4 4
NO•
Inactivation

SOD
Proteasome
Activation
6 NOX4 H2O2 4 ONOO– PPARα MTF1 SP1

7 HO1 Fe(II) 8 4, 5
P53
OH• Protein tyrosine nitration
MRP4 MT1/2 PBR
TGR5
4

18S-rRNA- 8-OHG RNA oxidation


↑ p21
↑ GADD45α Translation inhibition?

Senescence GLAST-mRNA- 8-OHG GS- Tyr-NO2 NKCC1- Tyr-NO2


↓ GLAST Degradation? Inactivation Activation

Glu NH4+

Fig. 2 | Mechanisms and consequences of oxidative/nitrosative stress in astrocytes in HE. Precipitating factors for
hepatic encephalopathy (HE), such as ammonia and inflammatory cytokines, trigger an N-methyl-d-aspartate receptor
(NMDAR)-dependent elevation of the intracellular calcium concentration in astrocytes75–77,86. This change leads to the
formation of a variety of reactive oxygen and nitrogen species that modify proteins and RNA species, alter gene expression
and signalling, and induce senescence. NADPH oxidase 4 (NOX4) and haem oxygenase 1 (HO1) play an important role
in generating oxidative stress. Both enzymes are upregulated by ammonia in a glutamine synthesis-dependent manner
involving O-GlcNAcylation-dependent downregulation of miR326-3p71,108. Potential sites of inhibition of oxidative and
nitrosative stress are as follows: (1) NMDAR antagonists (such as MK801 (refs75–77,86,121)); (2) cyclooxygenase inhibitors
(such as indometacin89,94); (3) nitric oxide synthase (NOS) inhibitors (such as NG-monomethyl-l-arginine75–77,86,91,93,102);
(4) antioxidants/reactive oxygen and nitrogen species scavengers (such as epigallocatechin gallate91,121); (5) uric acid86;
(6) NAPDH oxidase inhibitors (such as apocynin90,107,108,117,120); (7) HO1 inhibitors (such as zinc protoporphyrin IX107,108); and
(8) iron chelators (such as bipyridine108). Of note, pharmacological interventions at these sites have not yet been tested
clinically. cPLA2, cytosolic phospholipase A2; GADD45α, growth arrest and DNA damage-inducible protein 45α; GLAST,
sodium-dependent glutamate/aspartate transporter 1; GLS, glutaminase; Glu, glutamate; iNOS, inducible NOS; MRP4,
multidrug resistance-associated protein 4; MT, metallothionein; MTF1, metal regulatory transcription factor 1; NKCC1,
Na+-K+-2Cl− cotransporter 1; nNOS, neuronal NOS; NO, nitric oxide; PBR, peripheral-type benzodiazepine receptor;
PPARα, peroxisome proliferator-activated receptor-α; SP1, specificity protein 1.

Increased [Ca2+]i triggers the rapid formation of super- (iNOS)86,92,93; however, the pathogenetic relevance of
oxide anion radicals (O2–) by NADPH oxidase 2 (NOX2)90 iNOS in HE is unclear as iNOS is not consistently upregu-
and the synthesis of nitric oxide (NO) through activation lated in animal models of HE94–96 or in post mortem brain
of the neuronal nitric oxide synthase (nNOS)75,77,86,91. samples from patients with liver cirrhosis and HE70,82,97.
Another source of NO in ammonia-exposed astrocytes Mitochondria contribute to O 2– formation in
is the upregulation of inducible nitric oxide synthase ammonia-exposed cultured astrocytes and in animal


NATURE REVIEwS | DISEASE PRIMERS | Article citation ID: (2022) 8:43 5

0123456789();:
Primer

Ammonia and in brains of patients with liver cirrhosis and HE, which
GS may reflect an adaption to ameliorate ROS formation106
Glutamine (Fig. 2). However, TGR5 downregulation in the brain of
Hexosamine biosynthetic patients with liver cirrhosis and HE may also compro-
pathway mise the anti-inflammatory actions of TGR5 with so far
UDP-GlcNAc
unknown consequences. Of note, neurosteroid release
may be facilitated by multidrug resistance-associated
OGT
protein 4 (MRP4), which is upregulated by ammonia
Protein O-GlcNAcylation in astrocytes through RONS-mediated activation of the
peroxisome proliferator-activated receptor-γ (PPARγ)101.
↓ Pri-miR326-3p Importantly, MRP4 mRNA and protein levels are ele-
vated in post mortem brain tissue from patients with liver
cirrhosis and HE but not in those without HE101.
Ammonia also upregulates NOX isozyme 4
↓ miR326-3p (NOX4) and haem oxygenase 1 (HO1) through
O-GlcNAcylation-dependent transcription inhibition
↑ HO1 ↑ NOX4
of HO1-targeting and NOX4-targeting microRNAs107,108,
leading to increased intracellular levels of free ferrous
iron and H2O2. Increased free ferrous iron and H2O2 are
Fe(II) H2O2 suggested to trigger the formation of hydroxyl radicals
Fenton
reaction (OH•) presumably via induction of the Fenton reaction108.
OH• OH• formation subsequently leads to RNA oxidation and
induction of astrocyte senescence through the activation
of the cell cycle master regulator p53 (Fig. 3). This path-
RNA oxidation Senescence way requires glutamine-dependent O-GlcNAcylation,
Fig. 3 | Glutamine formation triggers oxidative
which again underlines the importance of glutamine
stress in astrocytes through protein O-GlcNAcylation. formation in the pathogenesis of ammonia toxicity.
In astrocytes, ammonia is used by glutamine synthetase
(GS) to form glutamine, which is a rate-limiting substrate Effects of oxidative/nitrosative stress
for the synthesis of activated N-acetyl-d-glucosamine Covalent protein modifications. RONS formation
(UDP-GlcNAc) within the hexosamine biosynthetic can lead to post-translational modifications of several
pathway. UDP-GlcNAc is a substrate of O-GlcNAc- proteins75–77,86,99,109 (Fig. 2). Ammonia-induced RONS
transferase (OGT), which attaches GlcNAc moieties formation triggers protein tyrosine nitration (PTN) of
onto serine or threonine residues in selected proteins. a variety of proteins, such as glutamine synthetase (GS),
This inhibits the transcription of the haem oxygenase 1
Na+-K+-2Cl− cotransporter 1 (NKCC1), extracellular
(HO1) and NADPH oxidase 4 (NOX4) mRNA-repressing
microRNA 326-3p. The resulting upregulation of HO1 signal-regulated protein kinase 1 (ERK1) and PBR, in
and NOX4 proteins elevates intracellular levels of free rat astrocytes75–77,86,109. Importantly, increased PTN and
ferrous iron and H2O2, respectively, and thereby triggers tyrosine nitration of GS have also been found in brains
the formation of hydroxyl radicals (OH•) in the Fenton from animal models of HE and in post mortem brain tis-
reaction. Consequences of the enhanced OH• formation sue from patients with liver cirrhosis and HE but not in
are RNA oxidation and astrocyte senescence. Adapted patients with cirrhosis without HE70,86. Catalytic activity
with permission from ref.108, Elsevier. of GS was inhibited by tyrosine nitration whereas the
transport activity of NKCC1 was enhanced63,86,109. The
models of HE98,99 presumably through a glutaminase latter can contribute to astrocyte swelling in response
(GLS)-mediated mitochondrial hydrolysis of glutamine81. to ammonia, and the interplay between osmotic and
Although studies have confirmed the expression of oxidative and nitrosative stress. PTN was also observed
GLS1 and GLS2 by cultured astrocytes and in rat and in astrocytes constituting the BBB in animal models of
human brain in situ100, the mechanisms underlying the HE, with unknown consequences for the integrity of the
GLS-induced mitochondrial reactive oxygen species BBB86,110. Moreover, ammonia-induced ROS formation
(ROS) formation still need to be established. Mitochondria triggers the carbonylation of proteins in cultured astro-
may also contribute to ROS formation by astrocytes cytes and brains of animal models of HE99,109,111. Similar to
in HE through synthesis of neurosteroids owing to nitration, carbonylation of NKCC1 in ammonia-exposed
ammonia-induced upregulation of the peripheral-type astrocytes enhanced its transport activity109.
benzodiazepine receptor (PBR)101–103. In line with this
finding, increased levels of GABAA receptor-modulating Protein homeostasis. Ammonia-induced ROS forma-
neuro­steroids (such as pregnenolone and allopregnano- tion in astrocytes also affects mechanisms involved in
lone) were found in brains from animal models of HE and protein homeostasis (proteostasis) such as protea­somal
in post mortem brain samples from patients with liver cir- degradation, which is enhanced99, and autophagy, which
rhosis and HE104,105. Importantly, neurosteroids triggered is impaired in cultured astrocytes and in brains from
ROS formation in cultured astrocytes through activation animal models of HE112,113 (Fig. 2). Importantly, surro-
of the G protein-coupled bile acid receptor TGR5 (ref.106). gate markers for autophagy were also upregulated in post
TGR5 is downregulated by ammonia in cultured astrocytes mortem brain tissue from patients with liver cirrhosis

6 | Article citation ID: (2022) 8:43 www.nature.com/nrdp

0123456789();:
Primer

and HE, and nuclear accumulation of the autophagy cirrhosis without HE47,70,94,121. Interestingly, surrogate
adaptor protein p62, which is known to be degraded dur- markers for endoplasmic reticulum stress are upregulated
ing autophagy, was found in some astrocytes in HE114,115. in ammonia-exposed cultured astrocytes and in post
These findings strongly suggest an impairment mortem brain samples from patients with liver cirrhosis
of autophagy in astrocytes in HE. and HE70,108,121. Endoplasmic reticulum stress might be
triggered by an improper translation of oxidized RNA.
Mitochondria. Mild oxidative stress can trigger mito- Oxidized RNA was also found at synapses and in RNA
phagy (mitochondrial degradation by autophagy) in a granules of neurons from ammonium acetate-treated
mitochondrial fission-dependent manner116. In line with rats, whereby it may disturb local postsynaptic protein
this, ammonia triggers mitochondrial swelling117 and synthesis and, consequently, impair related functions
reversible fragmentation of mitochondria and inhib- such as synaptic plasticity and memory formation121.
its energy metabolism in astrocytes112,118 (for reviews,
see refs119,120). Senescence. Ammonia also triggers astrocyte senescence
through ROS-dependent p53-activation and transcrip-
RNA oxidation. RNA is modified by ROS in cultured tion of p21 and growth arrest and DNA damage-inducible
astrocytes exposed to HE-relevant factors (ammonia, protein 45α (GADD45α)108,117 (Fig. 2). Importantly, bio-
inflammatory cytokines, benzodiazepines and hypo­ markers for senescence were also increased in post
natraemia)108,121. As RNA oxidation can affect protein mortem brain tissue from patients with liver cirrho-
translation, oxidation of sodium-dependent glutamate/ sis and HE but not in those without HE117. As astro-
aspartate transporter 1 (GLAST, which is responsible for cyte senescence decouples neuronal networks and/or
glutamate uptake from the extracellular space) mRNA inhibits neurogenesis, it may contribute to cognitive
may underlie the downregulation of GLAST in cultured impairment in patients with cirrhosis117,124, which persists
astrocytes and in brains of animal models of HE121–123. after resolution of an acute episode of overt HE125,126.
However, the relevance of GLAST downregulation for
synaptic glutamate clearance in HE remains to be inves- Gene expression. RONS formation in astrocytes exposed
tigated. Moreover, the 18S ribosomal RNA subunit is oxi- to HE-relevant factors also affects gene expression
dized in ammonia-exposed astrocytes, which may impair owing to the release of zinc ions from proteins in a NO
translation efficacy121. Increased levels of oxidized RNA synthesis-dependent way91,102, thereby activating metal
were also observed in brains from animal models of HE regulatory transcription factor 1 (MTF1)-dependent
and in post mortem brain samples from patients with and specificity protein 1 (SP1)-dependent gene
liver cirrhosis and HE but not from patients with liver transcription 91,102. The resulting upregulation of
zinc-chelating metallothioneins may protect astrocytes
Liver damage from the cytotoxic effects of free zinc ions. Moreover,
activation of SP1 may enhance the transcription of
PBR91,102 (Fig. 2). Importantly, comprehensive HE-specific
Hyperammonaemia Peripheral inflammation Altered microbiome gene expression changes, such as upregulation of sev-
eral metallothionein isoforms, were also detected in
Changes in the cerebral cortex of patients with liver cirrhosis97,127.
• Immunophenotype These transcriptome analyses identified ~600 genes with
• Extracellular vesicles altered expression in patients with liver cirrhosis with HE
but not in those without HE compared with controls.
Transmission to brain
Genes with altered expression include those involved in
oxidative stress, zinc homeostasis, microglial activation,
Neuroinflammation
and counteraction of pro-inflammatory signalling and
inflammatory cytokine expression97. Chronic hyper-
Altered neurotransmission
ammonaemia also alters cerebral protein expression as
shown by proteome analysis of brains from liver-specific
Cognitive and motor impairment
glutamine synthetase knockout mice128.
Fig. 4 | Steps involved in the process by which liver damage leads to cognitive
and motor impairment in MHE and HE. Hyperammonaemia and inflammatory Inflammation in HE
factors induce astrocytic and neuronal dysfunction, which alters synaptic plasticity MHE is associated with a shift in the immune system,
and oscillatory networks leading to the neurological symptoms in hepatic encephalopathy with the expansion and activation of T helper 17 (TH17),
(HE). Patients with liver cirrhosis have liver damage and inflammation, chronic TH22 and T follicular helper CD4+ lymphocytes as well as
hyperammonaemia, and altered microbiota. Each of these factors is enough to induce increased serum levels of pro-inflammatory cytokines,
peripheral inflammation, and changes in the immunophenotype and cargo of extracellular such as IL‐6, IL‐21, IL‐17, IL‐18, TNF, IL‐1β, IL‐15
vesicles, although the features of these changes are different for each factor. These and IL‐22, as well as of CCL20, CXCL13 and CX3CL1.
peripheral changes are transmitted to the brain by different mechanisms and result in
These alterations are transmitted to the brain and lead
induction of neuroinflammation, which alters neurotransmission and neuronal connectivity.
Moreover, chronic hyperammonaemia may also alter neurotransmission and induce to neuro­inflammation, which can alter neurotransmis-
neuroinflammation. Altered neurotransmission leads to impairment of cognitive and motor sion and lead to cognitive and motor impairment129.
function. Characterization of the molecular mechanisms involved in these processes could Studies in patients and in animal models support that
allow the identification of therapeutic targets on which to act to reverse the cognitive and the sequence of events involved in triggering the early
motor alterations in minimal HE (MHE) and HE. stages of MHE is that summarized in Fig. 4. At advanced


NATURE REVIEwS | DISEASE PRIMERS | Article citation ID: (2022) 8:43 7

0123456789();:
Primer

stages of HE, all these processes occur simultaneously. and transmission of signals to the brain. These mecha-
Understanding the sequence of events involved in trig- nisms ultimately lead to neuroinflammation with activa-
gering MHE is essential to the design and development tion of microglia and astrocytes and increased synthesis
of treatments to reverse MHE and HE. of pro-inflammatory factors, which alter neurotransmis-
Hyperammonaemia and peripheral inflammation sion and trigger neurological impairment129. Microglial
act synergistically to induce HE38,130,131. Wild-type mice activation has been reported in post mortem brains from
treated with TNF are sensitized to ammonia toxicity, patients with HE82,151,152. However, microglial reactivity
whereas TNF-deficient or TNF-receptor 1-deficient mice (pro-inflammatory or anti-inflammatory) has not been
are protected from ammonia toxicity132. The protection consistently shown. Such discrepant results from differ-
of TNF-deficient mice was paralleled by decreased cer- ent studies may be explained because different stages of
ebral expression of NKCC1, which is expected to coun- HE progression and also different brain areas at simi-
teract ammonia-induced glial swelling110,133. In line with lar stages of HE were investigated82,97,133,151,152. Of note,
this finding, TNF induces synergistically with ammo- enhanced mRNA levels of pro-inflammatory cytokines,
nia astrocyte swelling and protein tyrosine nitration of IL-1β and TNF, or increased protein levels of IL-1β, TNF,
cultured astrocytes73,78,79. IFNγ, IL-4 and IL-10, have not been detected in post
Hyperammonaemia is enough to induce systemic mortem cerebral cortex from patients with HE82,97,151.
inflammation, neuroinflammation and neurological
impairment 133,134. Conversely, systemic inflamma- Neurotransmission in HE
tion induced by lipopolysaccharide treatment of rats Previous attempts to ascribe HE to alterations of a sin-
impaired hepatic ammonia detoxification by glutamine gle neurotransmitter and/or its receptors have failed.
synthesis135. Alterations in the gut–liver–brain axis also Multiple neurotransmitter and receptor systems are
contribute to HE. For example, liver disease is associ- altered in HE, and many of these changes were brain
ated with changes in intestinal microbiota. Indeed, the region specific and possibly stage specific (for reviews,
colonic mucosal microbiota is altered in patients with see refs55,153).
cirrhosis and is linked with inflammation and impaired Neuroinflammation affects glutamatergic and
cognition136. These changes can also be involved in GABAergic neurotransmission129. Additionally, other
MHE136–139. Systemic inflammation is also suggested neurotransmitter systems are altered in HE, such as
to have a role in triggering MHE140,141 as prevention of glycinergic 154, serotoninergic 155, cholinergic 156 and
systemic inflammation (by intravenous infliximab, an dopaminergic157 systems, but the mechanisms involved
anti-TNF antibody) in animal models of HE prevents are not well known. Previously, a generally increased
neurological impairment134,142. Furthermore, MHE onset GABAergic tone was considered characteristic of HE;
is associated with a shift in peripheral inflammation and  however, this concept was challenged by findings of
immunophenotype, with increased differentiation increased GABAergic tone in the rat cerebellum with
and activation of TH22, T follicular helper and TH17 CD4+ decreased GABAergic tone in the rat cortex in response
T lymphocytes as well as changes in specific cytokines143. to hyperammonaemia153. Increased GABAergic tone
Studies have suggested that changes in extracellular in the cerebellum is mediated by neuroinflamma-
vesicles may contribute to liver disease development tion that increases NF-κB-mediated transcription of
and could be useful as biomarkers for diagnosis144,145. pro-inflammatory molecules and glutaminase, leading
Damaged hepatocytes, non-parenchymal cells and to increased glutamate and sodium uptake by astrocytes
infiltrated inflammatory cells in the liver release large and, ultimately, GABA release and increased extracellu-
amounts of extracellular vesicles that contain altered lar GABA levels, which leads to motor incoordination129.
cargo, which contributes to the pathogenesis of liver dis- Enhanced GABAergic neurotransmission in the cer-
ease and MHE. Moreover, extracellular vesicles from out- ebellum and reduced cortical GABAergic tone have
side the liver can also contribute to progression of liver also been reported in patients with HE158,159. However,
disease. Cargo alterations are different at different stages a similar study found increased GABAergic tone in the
of liver disease146. Cargo alterations in liver cirrhosis and cortex of patients with MHE160, suggesting that motor
hyperammonaemia affect proteins involved in immune cortical GABAergic tone decreases with increasing HE
function such as increased TNF, TNFR1, HSP70, TIMP3 severity and that alterations in neurotransmission may
and glutamine synthetase147. These extracellular vesicles be different in different brain areas. In a pilot study gol-
may reach the brain and their changes contribute to exanolone, a GABAA receptor-modulating steroid antag-
MHE induction147. The source of extracellular vesicles onist, improved the cognitive performance of patients
inducing cognitive impairment remains unclear and it with covert HE161.
is not known if they are generated in the liver. Increased Increased levels of TNF and IL-1β have been found
bile acids may also contribute to HE through sphin- in the hippocampus of rats with hyperammonaemia and
gosine 1-phosphate receptor 2 (S1PR2) and TGR5 acti- MHE, and may be associated with altered phosphoryl-
vation and increased CCL2, which activates microglia ation and expression of AMPA and NMDA receptors
and contributes to cognitive impairment148. and impaired learning and memory. Normalizing TNF
In summary, peripheral alterations can induce MHE levels or blocking IL-1 receptors restore different aspects
via several mechanisms, including infiltration of periph- of hippocampal neurotransmission and cognitive func-
eral lymphocytes and monocytes into the brain149,150, tion, indicating that different pro-inflammatory fac-
infiltration of extracellular vesicles from plasma147, and tors induce different cognitive alterations by varying
activation of cytokine receptors in endothelial cells mechanisms129,162.

8 | Article citation ID: (2022) 8:43 www.nature.com/nrdp

0123456789();:
Primer

In support of a role of inflammation in HE, targeting Box 1 | Pathophysiological changes in HE at the


inflammation in animal models of HE can restore cog- system level
nitive and motor function. These are restored by treat-
ments that reduce peripheral inflammation (anti-TNF Primary motor cortex
antibodies or ibuprofen), reduce microglial activation • Corticomuscular coherence is slowed in hepatic
and neuroinflammation (sulforaphane, inhibitors of encephalopathy (HE) in parallel with a slowed critical
p38 or S1PR2 antagonists), reduce GABAA receptor acti- flicker frequency (CFF)167
vation (bicuculline, pregnenolone sulfate or GR3027), • Slowed motor performance85
or increase extracellular cyclic guanosine monophos- • Reduced GABAergic tone in HE159
phate (cGMP) per se or total cGMP with sildenafil129,148.
Primary somatosensory cortex
Although not yet tested in humans, such treatments
• Slowed stimulus-related alpha-band activity in HE in
could be beneficial in patients with HE.
parallel to the slowed CFF170
• Impaired processing of temporal tactile stimuli171
Cerebral oscillatory networks in HE
Oscillatory neuronal activity serves as a key mechanism • Impaired thermal perception331
of large-scale functional communication and integra- Occipital cortex
tion across different brain regions and forms the basis of • Impaired temporal processing of visual stimuli reflected
cerebral network interactions163. Clinical HE symptom- by a slowed CFF203
atology is associated with alterations of oscillatory brain • Slowed spontaneous magnetoencephalography and
activity across different frequency bands and functional electroencephalography activity in the alpha-band
subsystems in patients with HE164 (Box 1). For example, and oscillatory activity174,332
mini-asterixis (a postural tremor-like phenomenon) in • Slowed attention-related oscillatory activity in the
the upper limbs arises from an abnormally slow thalam- gamma band168
ocortical and corticomuscular oscillatory drive165,166. The • Decreased levels of GABA333
frequency decrease in corticomuscular drive is paralleled • Increased ammonia levels334
by a decrease in the critical flicker frequency (CFF) in
HE167, suggesting that slowing of oscillatory activity rep- Cerebellum
resents a common pathophysiological mechanism across • Less cerebellar inhibition in HE158
modalities underlying diverse clinical HE symptoms. • Increased ammonia levels334
In agreement with this concept, attentional deficits
in HE are also directly related to changes in oscillatory
brain activity. Patients with HE have a marked negative state97,173,174. Occipital alpha-band peak frequency posi-
correlation between occipital gamma-band oscillations tively correlates in a linear manner with the CFF, sug-
to visual stimuli and HE disease severity (assessed by gesting a connection between spontaneous alpha-band
the CFF)168. Moreover, patients with HE lacked the activity and visual temporal resolution. Similar to
physiological power modulation of visual oscillatory the link between slowed alpha-band oscillations in the
activity in the gamma band around 60 Hz associated somatosensory cortex and impaired temporal tactile
with attentional shifts. As gamma-band oscillations are discrimination, this connection between alpha-band
instrumental in directing attention to stimuli169, these activity and visual temporal resolution can be explained
findings indicate that slowing of occipital gamma-band by current models of perceptual cycles in the visual
oscillations mediates an impairment of top-down atten- system175. As a possible neurochemical mechanism,
tional mechanisms of patients with HE in this task. occipital alpha-band peak frequencies are positively cor-
Thus, attentional deficits in HE are related to changes related with occipital GABA levels measured with MR
of the oscillatory gamma-band activity. However, to spectroscopy. Together, these findings revealed distinct
what extent the reported gamma-band changes are also disturbances of oscillatory brain activities in patients
specific for HE is unclear. with HE which, in turn, are related to neurochemical
Disease-related slowing of oscillatory brain activity changes and result in clinically relevant behavioural
also affects the somatosensory system. A slowed peak disturbances. However, further research is needed to
frequency of alpha-band oscillations and a slowing of unravel molecular and cellular mechanisms of oscilla-
stimulus-induced modulation of oscillatory activity tory dysfunction and to provide more direct evidence
have been found in the primary somatosensory cortex for a causal rather than correlational connection with
of patients with HE170. At the behavioural level, patients clinical symptomatology.
with HE are impaired in the temporal discrimination
of tactile stimuli, which correlates negatively with Cerebral vessel alterations in HE
the CFF171. This behavioural deficit is likely related to the Endothelial cell dysfunction is also suggested to con-
slowing of oscillations in the primary somatosensory tribute to the pathogenesis of HE80. Evidence from
cortex as these oscillations define perceptual cycles animal models of HE suggests impaired cerebral blood
underlying discrete processing of sensory input; flow, enhanced permeability of the BBB, and reduced
however, this has not yet been directly investigated172. clearance of metabolites and toxins from the brain
Patients with HE also exhibit a more global slow- through the glymphatic system176. Some have suggested
ing of oscillatory activity with prominent effects at the describing HE as a global dysfunction of the so-called
occipital alpha-band peak frequency during the resting neurogliovascular unit owing to the involved cell types177.


NATURE REVIEwS | DISEASE PRIMERS | Article citation ID: (2022) 8:43 9

0123456789();:
Primer

Circadian rhythm alterations in HE myoclonus. By contrast, grade I HE abnormalities, which


Patients with liver cirrhosis and HE frequently exhibit are usually detected by caregivers or doctors who know
disturbances of the sleep–wake cycle as reflected by the patient well, are grouped with abnormalities on test-
both insomnia and excessive daytime sleepiness, in ing (MHE) and are referred to as covert HE by some.
addition to altered circadian rhythmicity of melatonin The use of the term ‘covert HE’ is debated as this term
and cortisol blood levels178. As astrocytes together with implies ‘hidden’ disease; however, it includes patients
clock neurons are involved in the generation of circadian with grade I HE who have symptoms. Furthermore, cov-
rhythms in the suprachiasmatic nucleus of the hypo- ert HE is a heterogeneous syndrome186 that is prognos-
thalamus, altered circadian rhythmicity in HE has been tically distinct from MHE187 and there is no sub-grading
recently suggested to be, at least in part, a consequence of severity. What is certainly of value in the covert/overt
of astrocyte dysfunction178. HE model is the fact that disorientation to time and/or
asterixis (flapping tremor) identify grade II overt HE1,185.
Sarcopenia in HE A strong argument against the covert/overt HE model
Sarcopenia due to increased loss and decreased gain definitions is that patients with grade I HE already have
of muscle mass is a frequent complication in patients manifest symptoms and signs; thus, the word ‘covert’
with liver cirrhosis (for a review, see ref.179) owing to may not be appropriate and unfortunately adds to con-
physical inactivity and ammonia-induced upregula- fusion regarding nomenclature. Figure 5 summarizes the
tion of myostatin180. As glutamine synthetase activity currently used severity classifications of HE.
in muscle may compensate for the impaired ammo- The diagnosis of MHE is important because the
nia detoxification in the liver of patients with cirrho- condition is common and may be associated with an
sis, sarcopenia may contribute to the development of increased likelihood of subsequent overt HE episodes188
hyperammonaemia and HE (for a review, see ref.179). and is associated with poorer QOL 189. As a group,
patients with MHE and grade I HE have also been shown
Diagnosis, screening and prevention to drive worse than their counterparts with cirrhosis
Classification and no neuropsychiatric impairment190. As HE affects
Four items are used for the classification of HE1: the multiple components of mental functioning, probably
underlying condition (type A–C), the severity of men- to a different degree at any given moment in time, the
tal alterations (Fig. 5), time course (episodic, recurrent, International Society for Hepatic Encephalopathy and
persistent) and precipitating events. Nitrogen Metabolism suggested that the diagnosis be
In clinical practice, the grade of HE can be deter- based on more than one test to be chosen depending
mined using the West Haven criteria, which considers on local experience184. However, limited information
alterations in consciousness, intellectual function, per- is available on how to combine different test strategies/
sonality–behaviour and neuromuscular abnormalities1. results, and concordance between tests has been generally
As the clinical diagnosis of grade I HE, according to the reported as low186.
West Haven criteria181,182, is operator dependent183, some It has also been proposed to replace categorical
have suggested1,184 referring to ‘overt HE’ when at least classifications such as the West Haven criteria with
temporal disorientation or flapping tremor are present continuous classification schemes, considering neuro­
(≥grade II according to the West Haven criteria1,181,182); psychiatric changes as a spectrum, which they effec-
for a full grading algorithm, see refs1,182,185. Of note, flap- tively are. This continuum (low–high-grade HE) may
ping tremor is actually not tremor but is rather a negative capture neuropsychiatric changes from normality

West Haven criteria and clinical description


HE 0 MHE HE 1 HE 2 HE 3 HE 4
No abnormalities No clinical signs, Mild Disorientation Somnolence Coma-like
abnormal recognizable but arousable
psychometry clinical signs confusion

Covert HE Overt HE

Low-grade HE High-grade HE
• No hospitalization required • Sub-gradation by CFF or PHES • Hospitalization required

CFF (Hz) 48 44 40 39 38 32 28
Glasgow Coma Scale
PHES pts. +4 0 –4 –5 –8 –12 –16

Fig. 5 | Classification of severity of HE. According to the West Haven criteria181, hepatic encephalopathy (HE) is classified
into four stages (HE 1–4) in addition to minimal HE (MHE), which only shows abnormalities in psychometric testing1,182.
The covert/overt classification summarizes MHE and HE 1 as covert and HE 2–4 as overt HE, whereby the presence of
disorientation defines overt HE. The low-grade/high-grade classification defines low-grade HE as disease that does not
require hospitalization and describes severity within the low-grade HE range by means of critical flicker frequency (CFF)
or Psychometric Hepatic Encephalopathy Score (PHES) test results. High-grade HE corresponds to patients requiring
hospitalization and is further characterized by the Glasgow Coma Scale.

10 | Article citation ID: (2022) 8:43 www.nature.com/nrdp

0123456789();:
Primer

to unambiguous pathology191,192, using objective and have documented its reduction with worsening HE
reproducible parameters. Within the low-grade HE, HE and improvement after treatment191,203,204; it has been
0 to HE 2 are subsumed and continuously recorded and shown to be useful in predicting overt HE and sur-
tracked in their course using objective, reproducible vival in patients with cirrhosis as well as in predicting
and change-sensitive parameters such as the Psychometric post-TIPSS HE205–208.
Hepatic Encephalopathy Score or the CFF191,192. Severe HE
can be graded according to the Glasgow Coma Scale and Choice of test. The choice of neuropsychological and
a pragmatic criterion for separating between low-grade psychophysical tests depends on local experience,
and high-grade forms is the need for hospitalization due availability of appropriate normal reference values
to neuropsychiatric symptoms191,192. (to allow adjustment for age and educational attain-
ment), and the clinical inpatient and outpatient set-up.
Diagnostic approaches Neuropsychological and psychophysical tests can be
Neuropsychological, neurophysiological and psycho- used to both diagnose MHE and covert HE and to
physical tests are used for the diagnosis of MHE and/or quantify mild forms of overt HE, as they mostly require
to quantify overt HE. Neuropsychological tests are some degree of cooperation from the patient. EEG can
closer to the phenotype reality but they are prone to be used in uncooperative patients, and thus across the
learning effects, and the existence of local reference whole HE spectrum.
values is crucial as age and educational attainment
are major confounders. Neurophysiological tests like Serum biomarkers. 3-Nitrotyrosine is an oxidative stress
the electroencephalogram (EEG) can be obtained in marker in neurodegenerative diseases (for a review, see
patients with any severity of HE (also in uncooperative ref.209). A pilot study showed that serum 3-nitrotyrosine
patients) but they are further away from the pheno- levels (a degradation product of tyrosine nitrated pro-
type, and their recording/analysis requires equipment teins) could be a peripheral biomarker of MHE210. In this
and expertise that may not be necessarily available to study, 3-nitrotyrosine levels had good sensitivity, spec-
hepato-gastroenterology departments. ificity, and positive and negative predictive values for
MHE; however, these findings need validation in a larger
Neuropsychological, paper and pencil, or bedside tests. cohort. IL-6 has been suggested as another biomarker for
The Psychometric Hepatic Encephalopathy Score is a MHE, with one study finding twice as high serum IL-6
combination of five paper and pencil tests that assess levels in patients with liver cirrhosis and MHE compared
cognitive and psychomotor processing, speed, and with those without MHE211. Furthermore, serum IL-6
visuomotor coordination193. The tests are relatively easy levels may identify patients with liver cirrhosis at high
to administer and have been translated into several risk for overt HE212.
languages and validated in many countries.
The Animal Naming Test, during which patients Abdominal imaging. Abdominal imaging (CT/MRI)
are asked to name as many animals as possible in 60 s, should be carried out in patients with HE and less severe
compares favourably with more established and more liver disease to rule out spontaneous portosystemic
complex MHE or covert HE measures and can predict shunts, which could be subject to embolization.
overt HE194.
Brain imaging. CT and MRI of the brain can exclude
Neuropsychological, computerized tests. The Continuous other neurological diagnoses that may be confused
Reaction Time test assesses motor reaction time to with HE or that coexist with it213,214. However, most of
auditory stimuli delivered via headphones. Age, sex and the imaging techniques listed below are not suitable for
learning/tiring seem to have limited influence195,196. The routine examinations and are accordingly reserved
Inhibitory Control Test assesses response inhibition and for research purposes. Cerebral MRI with volumetric
working memory and has good validity for HE, although analysis, diffusion tensor imaging, magnetization trans-
it requires patients who are highly functional197,198. The fer imaging and functional MRI (fMRI) sequences can
Stroop test assesses psychomotor speed and cognitive facilitate assessment of brain water, atrophy, neuronal
flexibility by the interference between recognition reac- damage and functional connectivity in patients with
tion to a coloured field and a written colour name, and chronic liver disease215, whereas PET can be used to study
is available as an app199. The SCAN test measures the neurotransmitter imbalances and neuroinflammatory
speed and accuracy of a working memory task (digit status216.
recognition) of increasing difficulty/cognitive load and MRI is the most popular imaging technique in HE
has prognostic value for HE200. research studies. Cerebral oedema is often low grade
and may be radiologically undetectable49. However,
Neurophysiological tests. EEG can detect changes in cor- MRI measurement of total brain volume can be useful
tical cerebral activity in patients with any degree of HE to detect low-grade oedema in HE217,218. Magnetization
and its reliability increases if evaluated by quantitative transfer imaging can be used to assess neuronal dam-
semi-automated spectral analysis rather than visually201,202. age and increases in brain water content or membrane
permeability219–221. fMRI can be used to study brain
Psychophysical test. The CFF is the frequency at which function222,223 and, unlike PET, which involves radio­
a flickering light (from 60 Hz downwards) appears to be active tracer injections, does not require radiotracer use,
flickering (as opposed to fixed) to the observer. Studies thus allowing safe, longitudinal studies before and after


NATURE REVIEwS | DISEASE PRIMERS | Article citation ID: (2022) 8:43 11

0123456789();:
Primer

a
Assessment for HE and treatment of stable outpatient with cirrhosis

History of overt HE No history of overt HE

Neurological examination

Normal Abnormal

Search and treat risk factors for HE • Test for MHEa CT of the head
• Nutrition • PHES, CFF, EEG, other
• Lactulose ± rifaximin
• Secondary prophylaxis

Normal Abnormal Normal Abnormal

Stable Recurrent or No MHE MHE Grade I/II Neurological


• Follow-up persistent • Follow-up HE? disease
• Refer to
neurology
Search and treat risk factors for HE
Large shunts • Nutrition
• Embolize Consider liver • Lactulose
• Reduce TIPSS transplantation • Primary prophylaxisb

b Assessment for HE and treatment of hospitalized patients with cirrhosis

Assess airway, breathing and circulation

Neurological examination

Abnormal
Normal
CT scan

Abnormal Normal

Assessment of ammonia levelsc

Normal Abnormal

Manage Refer to Hepatic encephalopathy


complications neurology Grade I–II Grade III–IV Specific Rx
• Monitor • Admit to ICU • First line: lactulose or rifaximin
• Search for and treat • Protect airway • Second line: polyethylene
precipitating events • Intubate glycol/LOLA or LOLA
• Nutritional support • Third line: albumin dialysis or
adequate protein: check SPSS, consider occlusion
enteral/parenteral • Fourth line: liver transplantation

treatment intervention224. One fMRI study has demon- composite resonance67,228–230. This pattern of metabolite
strated reduced BOLD signal in the default-mode net- disturbance correlates with the severity of psychometric
work in patients with HE (more specifically, in the right test dysfunction in patients with HE231.
middle frontal gyrus and in the left posterior cingulate
cortex, which are involved in maintaining attention and Ammonia levels. As ammonia is central to the pathogen-
are normally highly active in alert individuals)225,226. MR esis of HE, the diagnosis of HE should not be sustainable
spectroscopy can be used to evaluate brain neurochem- in the absence of hyperammonaemia. Although there is
istry, including osmolyte status227(reflecting brain swell- no direct relationship between hyperammonaemia sever-
ing). The characteristic spectral appearance of HE using ity and HE severity11, elevated blood ammonia levels can
MR spectroscopy is a reduction in the myo-inositol predict HE-related hospitalization, and ammonia levels
and choline resonances with increases in measura- of >150 µmol/l and >80 µmol/l define the risk of mortal-
ble glutamine, which may overlap with glutamate in a ity in acute liver failure and cirrhosis, respectively232–235.

12 | Article citation ID: (2022) 8:43 www.nature.com/nrdp

0123456789();:
Primer

◀ Fig. 6 | Algorithms for assessment of HE and treatment in stable outpatients and artefact leads to overestimates in blood drawn from the
hospitalized patients with cirrhosis. a | Clinically stable patients with cirrhosis who fingertip240,241. Appropriate reference values should be
have a history of overt hepatic encephalopathy (HE) should be assessed and treated used if arterial or capillary ammonia are utilized.
for prevention of HE recurrence. Patients without a history of overt HE should undergo
neuropsychometric testing to detect minimal HE (MHE). Selected patients with MHE
Prognostic relevance
may be given primary prophylaxis with lactulose although the data supporting primary
prophylaxis is relatively poor. b | Patients with cirrhosis that are hospitalized for any
MHE is a predictor of overt HE188,242–244; thus, its detec-
complication associated with cirrhosis should be assessed for overt HE. The diagnosis tion should raise attention to the possibility of the occur-
of overt HE is based on the exclusion of other causes of neurological dysfunction in a rence of overt HE. While it has not been proven that
patient showing neurological disturbances compatible with HE. Patients with severe HE treatment of MHE or covert HE prevents the occurrence
should be managed in the intensive care unit (ICU). A careful search for and correction of overt HE, this assumption seems reasonable. Overt
of precipitating factors should be made. Specific therapies for the treatment of HE HE is a predictor of death3,245, and its appearance gen-
are introduced sequentially depending upon response. The mainstay of treatment is erally marks worsening of both hepatic function and
lactulose and/or rifaximin. If these fail, other treatments with less robust evidence, such prognosis19,246. Therefore, after a first bout of overt HE,
as polyethylene glycol or l-ornithine l-aspartate (LOLA), can be introduced. Patients the patient should probably be referred to a liver trans-
who remain refractory with treatment of precipitating events and other first-line and
plant centre. In the transplant setting, an episode of overt
second-line therapies can be considered for albumin dialysis using the molecular
adsorbents recirculating system. Simultaneously, a search for a large spontaneous HE increases mortality in patients with the same MELD
portosystemic shunt (SPSS) using CT scan of the abdomen should be initiated. If present, score247. As outlined above, even after resolution follow-
depending upon the clinical condition of the patient, this can be occluded radiologically. ing an attack of overt HE, patients may show persistent
Liver transplantation should be considered in all patients that have had an episode of cognitive deficits (for a review, see ref.248).
overt HE. CFF, critical flicker frequency; EEG, electroencephalogram; PHES, Psychometric
Hepatic Encephalopathy Score; Rx, prescription; TIPSS, transjugular intrahepatic Differential diagnosis
portosystemic stent shunt. aRoutine testing for MHE is not recommended in routine Although the clinical symptoms of HE in a patient with
clinical practice. bRoutine treatment is not recommended in patients with MHE. liver cirrhosis are fairly typical, other neuropsychiatric
c
Routine measurement of ammonia can be difficult and its use is debated. diseases also need to be considered. The main differen-
tial diagnoses are sleep apnoea, Wernicke encephalopa-
Moreover, increasing ammonia levels are associated thy, alcohol withdrawal syndrome, infections and septic
with increased risk of death, whereas a reduction is encephalopathy, hypoglycaemia and hyperglycaemia,
associated with survival4,234,236, suggesting that sequen- Wilson disease, sedative overdose, dementia, electrolyte
tial measurements of ammonia may be useful in imbalances, uraemia and hepatocerebral degeneration.
clinical practice. Whether ammonia levels are of value Moreover, hyperammonaemic conditions as a result of
in outpatients with stable cirrhosis to predict episodes of inborn errors of metabolism, such as urea cycle enzyme
overt HE is unknown. defects, must be considered (for reviews, see refs249,250).
Owing to the evidence in support of a potential role of Another major and important differential diagnosis
ammonia measurement in the diagnosis and prognosis is intracerebral bleeding following falls and trauma in
of HE, it is intriguing that assessment of ammonia levels patients with cirrhosis, who frequently have compro-
is not uniformly recommended for routine clinical use mised blood coagulation. Such bleeding may present
for patients with HE. Reasons for this reluctance may with somnolence and is detected by cerebral imaging.
be that peripheral ammonia levels will not necessarily
reflect those inside the brain and the growing evidence Management
that neurotoxicity of ammonia is primarily mediated by General approach and nutrition
ammonia-derived glutamine81,108 in the brain. In line with Guidelines for the clinical management of HE have been
this, plasma ammonia levels poorly correlate with the published jointly by the European Association for the Study
glutamine signal in 1H-MR spectroscopy228, whereas of the Liver and the American Association for the Study of
the latter correlates with HE severity228,232. The main issues Liver Diseases, by the Italian Association for the Study
with interpreting ammonia levels in clinical practice are of the Liver, by the Japanese Society of Hepatology, by the
the lack of standardization of procedures for blood sam- International Society for Hepatic Encephalopathy and
pling and storage and of its measurement, which can Nitrogen Metabolism, and by the European Society for
lead to erroneous results237. Although the measurement Parenteral and Enteral Nutrition1,12,185,251–254.
of ammonia is relatively straightforward, normal values Approaches to the management of HE in patients
vary considerably between hospitals, making it difficult with cirrhosis (that is, type C HE) comprise primary
to interpret absolute values across different hospitals. and secondary prophylaxis and treating HE episodes.
Arterial levels tend to be higher than venous levels, and The pathophysiology of type A and B HE is different,
there seems to be limited advantage in obtaining arte- approaches and goals of therapy are distinctive, and are
rial samples as opposed to venous samples for ammonia not considered further herein.
measurement238,239. Venous blood should be prefera-
bly drawn when the patient is fasting and, for accurate Primary prophylaxis
measurements, blood samples need to be collected in Primary prophylaxis aims to prevent the occurrence of
pre-cooled tubes, transported to the laboratory on ice, a first attack of overt HE in patients with liver cirrhosis,
and measurements made within 30 min. Of note, reliable especially in those with MHE. All patients with liver cir-
ammonia levels can be obtained only if standard operat- rhosis should undergo regular screening for complica-
ing procedures are in place. Capillary ammonia is best tions of cirrhosis such as hepatocellular carcinoma and
measured on blood obtained from the earlobe, as sweat HE (Fig. 6a). Lactulose, administered orally or as enemas,


NATURE REVIEwS | DISEASE PRIMERS | Article citation ID: (2022) 8:43 13

0123456789();:
Primer

has the best supporting evidence for primary prophy- may be poor in patients with recurrence or persis-
laxis of HE. Tolerability may be compromised by the tence of cognitive symptoms and should be reinforced
sweet taste of orally administered lactulose in addition through a multidisciplinary approach and repeated diet­
to flatulence and diarrhoea, which can be avoided by ary counselling252. The involvement of the caregiver is
proper dosage. Additionally, nutrition is an important crucial to increasing patients’ motivation and fulfilment
adjunct; consuming small meals during the day and a late of the nutritional regimen264. An algorithm describing
evening snack is recommended to reduce fasting peri- the treatment of patients with an acute episode of HE is
ods, which may cause protein catabolism255. In support shown in Fig. 6b.
of a role of nutrition, one randomized trial demonstrated
an improvement in cognitive performance with nutri- Secondary prophylaxis
tional therapy (30–35 kcal/kg/day, 1.0–1.5 g vegetable Risk of recurrence after recovery from an episode of
protein/kg/day) compared with no nutritional therapy in overt HE is high, which provides a rationale for second-
patients with cirrhosis and MHE, with reversal of MHE ary prophylaxis. Lactulose and probiotics are effective in
in 71% versus 22% of patients, respectively256. Patients preventing recurrent episodes of HE265–267. Moreover, the
with cirrhosis frequently have zinc deficiency and oral combination of rifaximin and lactulose is effective and
zinc supplementation was reported to improve HE and even superior to lactulose monotherapy268,269.
HRQOL257. Insomnia can be a symptom of HE; if it does
not improve in response to HE therapy, cognition behav- Specific medical treatments
ioural therapy, hydroxyzine and chloralhydrate258 might The main specific targets for the treatment of HE are
be considered. An algorithm describing the screening ammonia and inflammation. Strategies to directly tar-
and selection of patients for screening and institution of get the underlying neurological mechanisms have lagged
primary prophylaxis is shown in Fig. 6a. behind generalized systemic approaches.

Treatment of an acute episode of HE Targeting ammonia: approved treatments. The mainstay


Supportive measurements and treatments are required of therapy for HE is the non-absorbable disaccharide lac-
in patients with liver cirrhosis and identified cognitive tulose. Meta-analytical reviews of trials of lactulose ver-
impairment and neurological symptoms depending sus no therapy demonstrated marked beneficial effects
on the severity of HE. In a patient hospitalized with of lactulose on HE severity, HE prevention, serious
high-grade HE (grade III–IV), prevention of airway liver-related adverse events and mortality270. However, a
obstruction and aspiration pneumonia, care of possi- major criticism of the evidence on lactulose in HE is the
ble harms caused by the patient’s disorientation, care lack of double-blind, multicentre studies. Nevertheless,
of intravenous lines, liquid balance, and monitoring of lactulose is very cheap and thus cost-effective and readily
vital signs, urine output, renal function, pH, blood gases, available from many sources. The proposed mechanism
electrolytes and glucose, are mandatory259. Moreover, an of action is increasing ammonia excretion in the gut
intensive search for and rigorous treatment of factors lumen by decreasing the faecal transit time and reducing
known to precipitate HE episodes are mandatory and absorption by acidification of stool. Adverse effects of
frequently the correction of such precipitating factors lactulose are mild and include nausea, vomiting, flatu-
will improve HE. Even in a patient with moderate lence and diarrhoea, which are easily controlled by dose
(grade II) HE, general care and monitoring should be reduction.
adopted according to clinical conditions259. Rifaximin, a gut-restricted antibiotic, has supporting
Consideration of nutritional status is appropriate evidence as an add-on therapy to lactulose to prevent
in all patients with HE, as protein-calorie malnutrition the recurrence of HE268. The use of rifaximin for this
and sarcopenia are associated with a lower capacity of indication is almost universal and recommended by the
ammonia detoxification260. In addition, chronic hyper- regulators. Moreover, single-centre studies have found
ammonaemia can decrease protein synthesis through that rifaximin can improve the survival of patients with
myostatin activation261, triggering further muscle deple- HE271. Rifaximin treatment is well tolerated. Although
tion. Accordingly, HE treatment guidelines include the mechanism of action is uncertain, rifaximin does
supporting patients with HE with adequate calorie and modestly reduce ammonia concentration but whether
protein requirements251. it reduces bacterial translocation is controversial271–273.
Adequate protein intake is recommended in patients Given the relatively low systemic absorption of rifaxi-
with overt HE to prevent muscle catabolism251. In min, the development of bacterial resistance is likely to
patients who cannot achieve adequate protein intake, be small.
oral branched-chain amino acid (BCAA) supple- In l-ornithine l-aspartate (LOLA), l-ornithine
ments, which increase muscle protein synthesis, may augments glutamine synthesis in perivenous scavenger
be of help262,263. In support of their use, one metanaly- cells by provision of glutamate. Additionally, aspar-
sis showed that BCAAs improved overt HE but had no tate, which like 2-oxoglutarate is preferentially taken
effect on mortality263. More, better-controlled studies are up by perivenous scavenger cells65, can provide gluta-
needed before BCAAs can be recommended for rou- mate for glutamine synthesis after transamination with
tine use. Enteral and parenteral nutrition can be used 2-oxoglutarate. Eight randomized controlled clinical
in patients who cannot consume an oral diet owing to trials of LOLA for the treatment of HE have been con-
high-grade HE for a period of more intensive nutritional ducted, each of variable quality274,275. Meta-analyses
support. Of note, compliance to dietary prescriptions have suggested beneficial effects of LOLA on HE in

14 | Article citation ID: (2022) 8:43 www.nature.com/nrdp

0123456789();:
Primer

the acute setting, but a lot of the existing data are from A pivotal phase III study is being considered based on
single-centre studies. The use of LOLA for the preven- the results of this post hoc analysis.
tion of HE is debated. Although LOLA seems to be safe, Glycerol phenylbutyrate is converted into phenyl­
it is available in only a few countries and is considered acetate and acts to remove glutamine, which is an
a second-line therapy. ammoniagenic amino acid, thereby reducing ammonia
Some patients with cirrhosis develop large spon- concentrations291,292. One large phase II study demon-
taneous portosystemic shunts for reasons that are not strated that glycerol phenylbutyrate reduced ammonia
clear, which can result in severe HE. In these patients, and prevented HE recurrence in patients with HE293.
radiological embolization of the shunt reduces ammonia However, the follow-up phase III trial was not performed
levels and is effective in reducing the severity of HE276–279. for unclear reasons.
No published randomized clinical trials on this treat- VS-01 involves the administration of specially engi-
ment are available, but data from case series provide neered, biocompatible microspheres that can adsorb
compelling evidence for its usefulness at least in the ammonia into the abdomen. Preliminary results of a
short term as most patients will ultimately need a liver phase Ib study confirmed the safety and proof of concept
transplant for long-term survival278,280. The procedure is for this approach294. Further clinical trials are planned.
also considered safe and efficacious only in patients with
well-compensated cirrhosis (MELD score <11), further Targeting inflammation: off-label and experimental
limiting its application280. approaches. In a regulated, multicentre, randomized,
controlled trial, albumin dialysis using the Molecular
Targeting ammonia: off-label and experimental Adsorbents Recirculating System (MARS) significantly
approaches. Polyethylene glycol is thought to work in a reduced time to HE resolution compared with stand-
similar way to lactulose, by increasing faecal transit, but ard of care295. Although not widely available, this treat-
it leads to profuse diarrhoea. A single dose was found to ment is used by some centres and should be considered
improve HE severity compared with control (lactulose a third-line treatment for HE. DIALIVE, a new device
treatment) but ammonia levels were unchanged281. More that exchanges albumin and removes damage-associated
confirmatory data are needed before widespread use. and pathogen-associated molecular patterns, was associ-
Early-phase trials have demonstrated that faecal ated with reduced severity of HE in preliminary studies
transplantation is safe in patients with HE and can lead in patients with ACLF, providing a rationale for future
to improvements in the severity of dysbiosis and mark- clinical trials296.
ers of MHE282,283. However, patients in these trials also One uncontrolled non-randomized study suggested
received lactulose and rifaximin, making interpretation possible benefits of albumin in patients with HE, but
of the data difficult282. In an extension of the concept, this finding was not confirmed in a randomized con-
bacteria were engineered to affect ammonia metabolism trolled trial297. However, in another single-centre study,
and inoculated into the gut. Although data from animal the combination of lactulose with albumin was more
models were impressive, this benefit did not translate to effective than lactulose alone in the complete reversal
humans, leading to discontinuation of the clinical devel- of HE298. Moreover, another randomized clinical trial299,
opment programme284,285. In single-centre, unregulated in which albumin was compared with placebo aiming
studies, probiotics have shown efficacy in patients with to reduce 90-day mortality in patients with grade II HE,
MHE; however, the wide range of available probiotics did not meet this primary end point.
and the lack of standardization and regulated stud- An early-phase trial of golexanolone (a GABA A
ies make it difficult to determine their usefulness and receptor antagonist) in patients with MHE161 demon-
further studies are required before widespread use286. strated the safety of the drug. However, no significant
Modified carbon microspheres (AST-120) or a effect of golexanolone on neuropsychological tests com-
combination of micro–macrospheres (CARBALIVE) pared with placebo was observed despite trends towards
adsorb toxins (including ammonia) in the gut lumen. improvement in the drug arm. Future trials are planned.
Clinical trials of AST-120 failed to show clinical benefit
in patients with MHE (ASTUTE study) and results of the Liver transplantation and reversibility of HE
early-phase clinical trials of CARBALIVE are awaited287. Liver transplantation is the only rescue therapy for
The basis for ornithine phenylacetate use in HE is the patients with HE, and assessment for transplantation
hypothesis of the synergistic action of l-ornithine as a should be considered in all patients with a first epi-
glutamate provider for glutamine synthesis, and pheny- sode of overt HE as these patients are at greater risk
lacetate, which aims to remove glutamine by formation of death300,301. Long-term resolution of HE is the most
of phenylacetylglutamine288. Although one phase IIb common outcome, even in patients with severe ACLF
study demonstrated that this drug was safe, it did not and coma if the brainstem remains intact. The severity
reach the primary end point (reduction in the time to of HE should not be considered a contraindication for
resolution of HE)289. An unplanned post hoc analysis liver transplantation; however, the widespread adoption
that excluded patients with confirmed hyperammonae- of organ allocation systems involving MELD302 disad-
mia at study entry did find a significant reduction in vantages patients with HE as patients with severe and
time to resolution of HE compared with placebo. A recurrent HE often have relatively low MELD scores303,
follow-up study has shown that ornithine phenylacetate which is not considered in the formulae used for prior-
reduced ammonia in a dose-dependent manner and its itization of organs. In fact, adding HE to the MELD score
reduction was associated with the resolution of HE290. improves its predictive ability247.


NATURE REVIEwS | DISEASE PRIMERS | Article citation ID: (2022) 8:43 15

0123456789();:
Primer

Long-term follow-up studies in patients with HE who patients with HE. Overall, 26–70% of patients with
received liver transplantation allows the exploration of cirrhosis can have sleep disturbances, which are more
the reversibility of HE. Lack of resolution of HE is more frequent in those with MHE, and can adversely affect
severe in patients with previous episodes of HE248,304. HRQOL3,311,315–318. These sleep disturbances are delayed
In fact, subsequent studies correlating neurocognitive initiation and frequent awakenings, which result in
changes with neuroimaging305 confirmed these findings reduced sleeping time and excessive daytime sleepiness.
and identified subgroups that continued to show evi- Moreover, patients with liver cirrhosis and MHE more
dence of reduction in brain neuronal mass. Further pro- frequently have falls and fall-related injuries that affect
spective studies are needed to better characterize these their HRQOL compared with patients with cirrhosis
data considering the effects of calcineurin neurotoxicity and no MHE319,320. MHE contributes to falls in cirrho-
and surgery (for a review, see ref.306). Nevertheless, the sis due to slowed reaction time, impaired attention and
idea of irreversibility of HE needs to be explored further visuomotor coordination, and psychomotor speed.
as emerging data start to point towards a loss of neurons Intake of psychoactive drugs, poor muscle strength
underlying the pathophysiology of HE. and sleep problems may also increase the risk of falls in
cirrhosis319. Thus, these risk factors should be assessed
Quality of life in this population and therapeutic interventions must
Commonly used tools to evaluate HRQOL assess- be designed for patients such as exercise to improve
ment include Short-Form survey-36, Sickness Impact strength and balance, medication assessment to limit
Profile and the liver-specific Chronic Liver Disease the use of benzodiazepines and antipsychotics, and
Questionnaire307. HRQOL is often impaired in patients home modifications to reduce fall hazards. Osteopenia
with chronic liver diseases8,308 and worsens with the and osteoporosis increase the risk of fractures that may
progression to advanced cirrhosis307. The development be associated with surgeries and decompensations,
of HE in patients with cirrhosis impairs their HRQOL which could adversely affect HRQOL in patients with
and that of their caregivers309. Among cirrhosis-specific cirrhosis321. An emerging area of interest is the inter-
decompensating events, HE is the sole event consistently action between MHE and mild cognitive impairment,
associated with impaired HRQOL310. especially in patients aged >65 years with cirrhosis, with
MHE impairs daily functioning, driving skills (Box 2) evidence suggesting that MHE is independently associ-
and HRQOL in patients with cirrhosis and is an impor- ated with poor HRQOL irrespective of mild cognitive
tant risk factor for the development of overt HE and impairment322.
mortality311–314. A recent study of patients with cirrho- Treatment-induced improvement in cognitive func-
sis and their caregivers10 demonstrated that caregiver tion can improve HRQOL in patients with cirrhosis311
burden scores increased significantly among patients such as with lactulose treatment 311 and rifaximin
with either previous overt HE or MHE compared with treatment314. Moreover, lactulose treatment can also
patients without HE and were positively correlated improve gut microbiota and recovery from MHE in
with liver disease severity scores and negatively with patients with cirrhosis323 and rifaximin can improve
socioeconomic status. Patients with MHE often have simulator-based driving performance in patients with
a preserved basic day-to-day functioning but complex MHE184. Rifaximin treatment may also improve objec-
activities, such as planning a trip or driving a car, are tive parameters of sleep architecture in patients with
often affected. Of note, all scales of the Sickness Impact cirrhosis and recurrent HE324.
Profile are impaired in patients with MHE311,313,314.
Comorbid conditions, such as sleep disturbances, Outlook
falls and osteopenia, can adversely affect HRQOL in The nomenclature and diagnosis of HE require unifica-
tion, with an objective, clinically and scientifically sound
approach. HE is a crude description of a neuropsycho-
Box 2 | Driving and HE logical syndrome and phenotype, which may encompass
Minimal hepatic encephalopathy (MHE) in patients with cirrhosis is associated with pathophysiologically different and heterogeneous enti-
poor driving skills190,335–337. Patients with MHE have greater impairment in several ties. To unravel these factors requires novel biomarkers
parameters, such as car handling, manoeuvring, adaptation and cautiousness, than and non-invasive methods of brain examination, and
patients without MHE337. Moreover, cognitive decline in patients with MHE is associated will have important consequences for personalized
with an increased risk of accidents190,338–340. However, a study assessing real on-road medicine.
driving showed that MHE or HE grade I did not necessarily predict the inability to drive Although considerable progress has been made in
a car in the individual patient190, although in the study group as a whole increasing
understanding the pathophysiology and biochemistry
HE severity paralleled significant performance deficits in traffic safety parameters190.
However, a couple of studies found no impairment in driving performance or increased
of HE, only a small amount of this knowledge has been
accident rate in patients with MHE341,342. translated to date into clinical practice and treatment.
No clear guidelines are available regarding restricting driving in patients with HE. Smartphone-based health monitoring may be applied
However, an ISHEN Consensus343 suggests that a short, objective and non-judgemental to patients with cirrhosis and HE and could be used for
driving history should be taken at each clinical visit. Cognitive testing is not useful to early detection of HE worsening325. Potential new treat-
determine who is a poor driver and is not recommended to restrict or resume driving. ment targets may focus on cerebral oxidative or nitro-
In patients with episodes of overt HE in <3 months, oral and written advice against sative stress and the oscillatory networks in the brain.
driving should be given to patients and caregivers and be documented. Patients who Investigation of treatment options may be facilitated
wish to resume driving should schedule a formal driving reassessment with the local by clinical trials, which require not only the definition
authorities based on local regulations343.
of study populations but also of appropriate study end

16 | Article citation ID: (2022) 8:43 www.nature.com/nrdp

0123456789();:
Primer

points and readouts for different forms of HE. This not be used in isolation to diagnose the presence of HE.
and the availability of novel biomarkers will also help Nevertheless, it is reasonable to assume that simple
to decide whether primary prophylaxis of HE should bedside tests may soon be developed that can be used
be considered245. Blood ammonia levels were shown in clinical practice. It is hoped that this Primer will
to be a biomarker of prognosis in patients with liver stimulate further research on this important disorder.
cirrhosis234; however, blood ammonia levels themselves
do not necessarily correlate with HE severity and should Published online xx xx xxxx

1. Vilstrup, H. et al. Hepatic encephalopathy in chronic 20. Moreau, R. et al. Acute-on-chronic liver failure is a neuropsychological effects of induced hyperammonemia
liver disease: 2014 practice guideline by the American distinct syndrome that develops in patients with acute in cirrhosis. J. Hepatol. 40, 247–254 (2004).
Association for the Study of Liver Diseases and the decompensation of cirrhosis. Gastroenterology 144, 39. Merli, M. et al. Increased risk of cognitive impairment
European Association for the Study of the Liver. 1426–1437 (2013). in cirrhotic patients with bacterial infections. J. Hepatol.
Hepatology 60, 715–735 (2014). 21. Hirode, G., Vittinghoff, E. & Wong, R. J. Increasing 59, 243–250 (2013).
2. Riggio, O. et al. High prevalence of spontaneous burden of hepatic encephalopathy among hospitalized 40. Yuan, L.-T. et al. Multiple bacterial infections increase
portal-systemic shunts in persistent hepatic adults: an analysis of the 2010–2014 national the risk of hepatic encephalopathy in patients with
encephalopathy: a case-control study. Hepatology 42, inpatient sample. Dig. Dis. Sci. 64, 1448–1457 cirrhosis. PLoS ONE 13, e0197127 (2018).
1158–1165 (2005). (2019). 41. Solé, C. et al. Alterations in gut microbiome in cirrhosis
3. Cordoba, J. et al. Characteristics, risk factors, and 22. Di Pascoli, M. et al. Hospitalizations due to cirrhosis: as assessed by quantitative metagenomics: relationship
mortality of cirrhotic patients hospitalized for hepatic clinical aspects in a large cohort of italian patients and with acute-on-chronic liver failure and prognosis.
encephalopathy with and without acute-on-chronic cost analysis report. Dig. Dis. 35, 433–438 (2017). Gastroenterology 160, 206–218.e13 (2021).
liver failure (ACLF). J. Hepatol. 60, 275–281 (2014). 23. Riggio, O. et al. A Model for predicting development of 42. Bajaj, J. S. et al. Altered profile of human gut
4. Sawhney, R. et al. Role of ammonia, inflammation, overt hepatic encephalopathy in patients with cirrhosis. microbiome is associated with cirrhosis and its
and cerebral oxygenation in brain dysfunction of acute- Clin. Gastroenterol. Hepatol. 13, 1346–1352 (2015). complications. J. Hepatol. 60, 940–947 (2014).
on-chronic liver failure patients: Brain Dysfunction and 24. Guevara, M. et al. Hyponatremia is a risk factor of 43. Gupta, A. et al. Role of small intestinal bacterial
ACLF. Liver Transpl. 22, 732–742 (2016). hepatic encephalopathy in patients with cirrhosis: overgrowth and delayed gastrointestinal transit
5. Wright, G., Sharifi, Y., Jover-Cobos, M. & Jalan, R. The a prospective study with time-dependent analysis. time in cirrhotic patients with minimal hepatic
brain in acute on chronic liver failure. Metab. Brain Dis. Am. J. Gastroenterol. 104, 1382–1389 (2009). encephalopathy. J. Hepatol. 53, 849–855 (2010).
29, 965–973 (2014). 25. Gil-Gómez, A. et al. Development and validation 44. Dam, G., Vilstrup, H., Watson, H. & Jepsen, P.
6. Jalan, R., Moreau, R. & Arroyo, V. Acute-on-chronic of a clinical-genetic risk score to predict hepatic Proton pump inhibitors as a risk factor for hepatic
liver failure. Reply. N. Engl. J. Med. 383, 893–894 encephalopathy in patients with liver cirrhosis. encephalopathy and spontaneous bacterial peritonitis
(2020). Am. J. Gastroenterol. 116, 1238–1247 (2021). in patients with cirrhosis with ascites. Hepatology 64,
7. Lauridsen, M. M. et al. Validation of a simple quality- 26. Yoo, H. Y., Edwin, D. & Thuluvath, P. J. Relationship 1265–1272 (2016).
of-life score for identification of minimal and prediction of the model for end-stage liver disease (MELD) 45. Nardelli, S. et al. Proton pump inhibitors are
of overt hepatic encephalopathy. Hepatol. Commun. 4, scale to hepatic encephalopathy, as defined by associated with minimal and overt hepatic
1353–1361 (2020). electroencephalography and neuropsychometric encephalopathy and increased mortality in patients
8. Ladegaard Grønkjær, L., Hoppe Sehstedt, T., testing, and ascites. Am. J. Gastroenterol. 98, with cirrhosis. Hepatology 70, 640–649 (2019).
Norlyk, A. & Vilstrup, H. Overt hepatic encephalopathy 1395–1399 (2003). 46. Verma, N. et al. Dynamic assessments of hepatic
experienced by individuals with cirrhosis: a qualitative 27. Praktiknjo, M. et al. Total area of spontaneous encephalopathy and ammonia levels predict mortality
interview study. Gastroenterol. Nurs. 41, 468–476 portosystemic shunts independently predicts in acute-on-chronic liver failure. Hepatol. Int. 15,
(2018). hepatic encephalopathy and mortality in liver 970–982 (2021).
9. Fabrellas, N. et al. Psychological burden of hepatic cirrhosis. J. Hepatol. 72, 1140–1150 (2020). 47. Qvartskhava, N. et al. Hyperammonemia in gene-
encephalopathy on patients and caregivers. 28. Yin, X. et al. A nomogram to predict the risk of hepatic targeted mice lacking functional hepatic glutamine
Clin. Transl. Gastroenterol. 11, e00159 (2020). encephalopathy after transjugular intrahepatic synthetase. Proc. Natl Acad. Sci. 112, 5521–5526
10. Shrestha, D. et al. Factors affecting psychological portosystemic shunt in cirrhotic patients. Sci. Rep. (2015).
burden on the informal caregiver of patients with 10, 9381 (2020). 48. DeMorrow, S., Cudalbu, C., Davies, N., Jayakumar, A. R.
cirrhosis: looking beyond the patient. J. Clin. Exp. 29. Zhou, Y. et al. PTFE-covered TIPS is an effective & Rose, C. F. 2021 ISHEN guidelines on animal models
Hepatol. 10, 9–16 (2020). treatment for secondary preventing variceal of hepatic encephalopathy. Liver Int. 41, 1474–1488
11. Rose, C. F. et al. Hepatic encephalopathy: novel rebleeding in cirrhotic patients with high risks. Eur. (2021).
insights into classification, pathophysiology and J. Gastroenterol. Hepatol. 32, 1235–1243 (2020). 49. Häussinger, D., Kircheis, G., Fischer, R., Schliess, F. &
therapy. J. Hepatol. 73, 1526–1547 (2020). 30. Qi, X., Tian, Y., Zhang, W., Yang, Z. & Guo, X. Covered vom Dahl, S. Hepatic encephalopathy in chronic liver
12. Wendon, J. et al. EASL clinical practical guidelines versus bare stents for transjugular intrahepatic disease: a clinical manifestation of astrocyte swelling
on the management of acute (fulminant) liver failure. portosystemic shunt: an updated meta-analysis of and low-grade cerebral edema? J. Hepatol. 32,
J. Hepatol. 66, 1047–1081 (2017). randomized controlled trials. Ther. Adv. Gastroenterol. 1035–1038 (2000).
13. Nicoletti, V. et al. Hepatic encephalopathy in patients 10, 32–41 (2016). 50. Cudalbu, C. & Taylor-Robinson, S. D. Brain edema in
with non-cirrhotic portal hypertension: description, 31. Nicoară-Farcău, O. et al. Preemptive TIPS Individual chronic hepatic encephalopathy. J. Clin. Exp. Hepatol.
prevalence and risk factors. Dig. Liver Dis. 48, Data Metanalysis, International Variceal Bleeding 9, 362–382 (2019).
1072–1077 (2016). Study and Baveno Cooperation Study groups. Effects 51. Córdoba, J. et al. The development of low-grade
14. Kraglund, F., Jepsen, P., Amanavicius, N. & of early placement of transjugular portosystemic cerebral edema in cirrhosis is supported by the
Aagaard, N. K. Long-term effects and complications shunts in patients with high-risk acute variceal evolution of 1H-magnetic resonance abnormalities after
of the transjugular intrahepatic portosystemic shunt: bleeding: a meta-analysis of individual patient data. liver transplantation. J. Hepatol. 35, 598–604 (2001).
a single-centre experience. Scand. J. Gastroenterol. Gastroenterology 160, 193–205 (2021). 52. Llansola, M. et al. NMDA receptors in hyperammonemia
54, 899–904 (2019). 32. Jepsen, P., Watson, H., Andersen, P. K. & Vilstrup, H. and hepatic encephalopathy. Metab. Brain Dis. 22,
15. Sepanlou, S. G. et al. The global, regional, and Diabetes as a risk factor for hepatic encephalopathy 321–335 (2007).
national burden of cirrhosis by cause in 195 countries in cirrhosis patients. J. Hepatol. 63, 1133–1138 53. Ott, P. & Larsen, F. S. Blood-brain barrier permeability
and territories, 1990–2017: a systematic analysis (2015). to ammonia in liver failure: a critical reappraisal.
for the Global Burden of Disease Study 2017. 33. Ampuero, J. et al. Role of diabetes mellitus on hepatic Neurochem. Int. 44, 185–198 (2004).
Lancet Gastroenterol. Hepatol. 5, 245–266 encephalopathy. Metab. Brain Dis. 28, 277–279 54. Häussinger, D. & Blei, A. in Textbook of Hepatology
(2020). (2012). 728–760 (Wiley-Blackwell Oxford, 2007).
16. Lauridsen, M. M., Jepsen, P. & Vilstrup, H. Critical 34. Vilar-Gomez, E. et al. Type 2 diabetes and metformin 55. Palomero-Gallagher, N. & Zilles, K. Neurotransmitter
flicker frequency and continuous reaction times for use associate with outcomes of patients with receptor alterations in hepatic encephalopathy:
the diagnosis of minimal hepatic encephalopathy. nonalcoholic steatohepatitis-related, child-pugh a a review. Arch. Biochem. Biophys. 536, 109–121
A comparative study of 154 patients with liver cirrhosis. Clin. Gastroenterol. Hepatol. 19, 136–145.e6 (2013).
disease. Metab. Brain Dis. 26, 135–139 (2011). (2021). 56. Williams, E., Chu, C. & DeMorrow, S. A critical
17. Rathi, S. et al. Prevalence of minimal hepatic 35. Elkrief, L. et al. Diabetes mellitus is an independent review of bile acids and their receptors in hepatic
encephalopathy in patients with liver cirrhosis: prognostic factor for major liver-related outcomes encephalopathy. Anal. Biochem. 643, 114436
a cross-sectional, clinicoepidemiological, multicenter, in patients with cirrhosis and chronic hepatitis C. (2022).
nationwide study in India: the PREDICT study. Hepatology 60, 823–831 (2014). 57. Butterworth, R. F. Hepatic encephalopathy in cirrhosis:
J. Clin. Exp. Hepatol. 9, 476–483 (2019). 36. Jepsen, P., Christensen, J., Weissenborn, K., Watson, H. pathology and pathophysiology. Drugs 79, 17–21
18. Tapper, E. B., Henderson, J. B., Parikh, N. D., & Vilstrup, H. Epilepsy as a risk factor for hepatic (2019).
Ioannou, G. N. & Lok, A. S. Incidence of and risk encephalopathy in patients with cirrhosis: a cohort 58. Butterworth, R. F. Neurosteroids in hepatic
factors for hepatic encephalopathy in a population- study. BMC Gastroenterol. 16, 77–77 (2016). encephalopathy: novel insights and new therapeutic
based cohort of Americans with cirrhosis. Hepatol. 37. Nardelli, S. et al. Muscle alterations are associated opportunities. J. Steroid Biochem. Mol. Biol. 160,
Commun. 3, 1510–1519 (2019). with minimal and overt hepatic encephalopathy 94–97 (2016).
19. Jepsen, P., Ott, P., Andersen, P. K., Sørensen, H. T. in patients with liver cirrhosis. Hepatology 70, 59. Butterworth, R. F. Role of circulating neurotoxins in
& Vilstrup, H. Clinical course of alcoholic liver 1704–1713 (2019). the pathogenesis of hepatic encephalopathy: potential
cirrhosis: a Danish population-based cohort study. 38. Shawcross, D. L., Davies, N. A., Williams, R. & Jalan, R. for improvement following their removal by liver assist
Hepatology 51, 1675–1682 (2009). Systemic inflammatory response exacerbates the devices. Liver Int. 23 (Suppl. 3), 5–9 (2003).


NATURE REVIEwS | DISEASE PRIMERS | Article citation ID: (2022) 8:43 17

0123456789();:
Primer

60. Rivera-Mancía, S., Ríos, C. & Montes, S. Manganese 86. Schliess, F. et al. Ammonia induces MK-801-sensitive 108. Görg, B. et al. O-GlcNAcylation-dependent
accumulation in the CNS and associated pathologies. nitration and phosphorylation of protein tyrosine upregulation of HO1 triggers ammonia-induced
Biometals 24, 811–825 (2011). residues in rat astrocytes. FASEB J. 16, 739–741 oxidative stress and senescence in hepatic
61. Dejong, C. H. C., van de Poll, M. C. G., Soeters, P. B., (2002). encephalopathy. J. Hepatol. 71, 930–941 (2019).
Jalan, R. & Olde Damink, S. W. M. Aromatic amino 87. Mayer, M. L., Westbrook, G. L. & Guthrie, P. B. 109. Jayakumar, A. R. et al. Na-K-Cl Cotransporter-1
acid metabolism during liver failure. J. Nutr. 137, Voltage-dependent block by Mg2+ of NMDA in the mechanism of ammonia-induced astrocyte
1579S–1585S (2007). responses in spinal cord neurones. Nature 309, swelling. J. Biol. Chem. 283, 33874–33882
62. Bernardini, P. & Fischer, J. E. Amino acid imbalance 261–263 (1984). (2008).
and hepatic encephalopathy. Annu. Rev. Nutr. 2, 88. Kloda, A., Lua, L., Hall, R., Adams, D. J. & 110. Vaquero, J., Chung, C., Cahill, M. & Blei, A.
419–454 (1982). Martinac, B. Liposome reconstitution and modulation Pathogenesis of hepatic encephalopathy in
63. Frieg, B., Görg, B., Gohlke, H. & Häussinger, D. of recombinant N-methyl-D-aspartate receptor acute liver failure. Semin. Liver Dis. 23, 259–270
Glutamine synthetase as a central element in hepatic channels by membrane stretch. Proc. Natl Acad. (2003).
glutamine and ammonia metabolism: novel aspects. Sci. USA 104, 1540–1545 (2007). 111. Carbonero-Aguilar, P. et al. Brain biomolecules
Biol. Chem. 402, 1063–1072 (2021). 89. Görg, B. et al. Ammonia triggers exocytotic release oxidation in portacaval-shunted rats. Liver Int. 31,
64. Häussinger, D. Nitrogen metabolism in liver: structural of L-glutamate from cultured rat astrocytes. Glia 6, 964–969 (2011).
and functional organization and physiological 691–705 (2010). 112. Bai, Y., Wang, Y. & Yang, Y. Hepatic encephalopathy
relevance. Biochem. J. 267, 281–290 (1990). 90. Reinehr, R. et al. Hypoosmotic swelling and ammonia changes mitochondrial dynamics and autophagy in the
65. Stoll, B., McNelly, S., Buscher, H. P. & Häussinger, D. increase oxidative stress by NADPH oxidase in substantia nigra. Metab. Brain Dis. 33, 1669–1678
Functional hepatocyte heterogeneity in glutamate, cultured astrocytes and vital brain slices. Glia 55, (2018).
aspartate and alpha-ketoglutarate uptake: 758–771 (2007). 113. Lu, K. et al. Hepatic encephalopathy is linked
a histoautoradiographical study. Hepatology 13, 91. Kruczek, C. et al. Hypoosmotic swelling affects zinc to alterations of autophagic flux in astrocytes.
247–253 (1991). homeostasis in cultured rat astrocytes. Glia 57, EBioMedicine 48, 539–553 (2019).
66. Norenberg, M. D. The role of astrocytes in hepatic 79–92 (2009). 114. Luo, S., Au Yeung, S. L., Zhao, J. V., Burgess, S. &
encephalopathy. Neurochem. Pathol. 6, 13–33 92. Chastre, A., Jiang, W., Desjardins, P. & Schooling, C. M. Association of genetically predicted
(1987). Butterworth, R. F. Ammonia and proinflammatory testosterone with thromboembolism, heart failure,
67. Häussinger, D. et al. Proton magnetic resonance cytokines modify expression of genes coding for and myocardial infarction: mendelian randomisation
spectroscopy studies on human brain Myo-inositol astrocytic proteins implicated in brain edema in acute study in UK Biobank. BMJ https://doi.org/10.1136/
in hypo-osmolarity and hepatic encephalopathy. liver failure. Metab. Brain Dis. 25, 17–21 (2010). bmj.l476 (2019).
Gastroenterology 107, 1475–1480 (1994). 93. Sinke, A. P. et al. NFkappaB in the mechanism 115. Gelpi, E. et al. The autophagic marker p62 highlights
68. Shah, N. J. et al. Quantitative cerebral water content of ammonia-induced astrocyte swelling in culture. Alzheimer type II astrocytes in metabolic/hepatic
mapping in hepatic encephalopathy. NeuroImage 41, J. Neurochem. 106, 2302–2311 (2008). encephalopathy. Neuropathology 40, 358–366
706–717 (2008). 94. Brück, J. et al. Locomotor impairment and (2020).
69. Winterdahl, M. et al. Cerebral water content mapping cerebrocortical oxidative stress in portal vein ligated 116. Frank, M. et al. Mitophagy is triggered by mild
in cirrhosis patients with and without manifest HE. rats in vivo. J. Hepatol. 54, 251–257 (2011). oxidative stress in a mitochondrial fission dependent
Metab. Brain Dis. 34, 1071–1076 (2019). 95. Suárez, I., Bodega, G., Rubio, M., Felipo, V. & manner. Biochim. Biophys. Acta 1823, 2297–2310
70. Görg, B. et al. Oxidative stress markers in the brain Fernández, B. Neuronal and inducible nitric oxide (2012).
of patients with cirrhosis and hepatic encephalopathy. synthase expression in the rat cerebellum following 117. Görg, B., Karababa, A., Shafigullina, A., Bidmon, H. J.
Hepatology 52, 256–265 (2010). portacaval anastomosis. Brain Res. 1047, 205–213 & Häussinger, D. Ammonia-induced senescence in
71. Häussinger, D. & Görg, B. in Oxidative Stress: (2005). cultured rat astrocytes and in human cerebral
Eustress and Distress 669–693 (Elsevier, 2019). 96. Suárez, I., Bodega, G., Arilla, E., Felipo, V. & cortex in hepatic encephalopathy. Glia 63, 37–50
72. Häussinger, D., Butz, M., Schnitzler, A. & Görg, B. Fernández, B. The expression of nNOS, iNOS and (2014).
Pathomechanisms in hepatic encephalopathy. nitrotyrosine is increased in the rat cerebral cortex 118. Drews, L. et al. Ammonia inhibits energy metabolism
Biol. Chem. 402, 1087–1102 (2021). in experimental hepatic encephalopathy. Neuropathol. in astrocytes in a rapid and glutamate dehydrogenase
73. Lachmann, V., Görg, B., Bidmon, H. J., Keitel, V. & Appl. Neurobiol. 32, 594–604 (2006). 2-dependent manner. Dis. Model. Mech. 13,
Häussinger, D. Precipitants of hepatic encephalopathy 97. Görg, B., Bidmon, H.-J. & Häussinger, D. Gene dmm047134 (2020).
induce rapid astrocyte swelling in an oxidative stress expression profiling in the cerebral cortex of 119. Rama Rao, K. V. & Norenberg, M. D. Brain energy
dependent manner. Arch. Biochem. Biophys. 536, patients with cirrhosis with and without hepatic metabolism and mitochondrial dysfunction in acute
143–151 (2013). encephalopathy. Hepatology 57, 2436–2447 (2013). and chronic hepatic encephalopathy. Neurochem. Int.
74. Schliess, F., Görg, B. & Häussinger, D. Pathogenetic 98. Kosenko, E. A. et al. Portacaval shunting causes 60, 697–706 (2012).
interplay between osmotic and oxidative stress: the differential mitochondrial superoxide production in 120. Zimmermann, M. & Reichert, A. S. Rapid metabolic
hepatic encephalopathy paradigm. Biol. Chem. 387, brain regions. Free Radic. Biol. Med. 113, 109–118 and bioenergetic adaptations of astrocytes under
1363–1370 (2006). (2017). hyperammonemia – a novel perspective on hepatic
75. Görg, B. et al. Inflammatory cytokines induce protein 99. Widmer, R., Kaiser, B., Engels, M., Jung, T. & encephalopathy. Biol. Chem. 402, 1103–1113
tyrosine nitration in rat astrocytes. Arch. Biochem. Grune, T. Hyperammonemia causes protein oxidation (2021).
Biophys. 449, 104–114 (2006). and enhanced proteasomal activity in response to 121. Görg, B. et al. Ammonia induces RNA oxidation in
76. Görg, B. Benzodiazepine-induced protein tyrosine mitochondria-mediated oxidative stress in rat primary cultured astrocytes and brain in vivo. Hepatology 48,
nitration in rat astrocytes. Hepatology 37, 334–342 astrocytes. Arch. Biochem. Biophys. 464, 1–11 567–579 (2008).
(2003). (2007). 122. Chan, H. Effects of ammonia on glutamate
77. Schliess, F., Foster, N., Görg, B., Reinehr, R. & 100. Cardona, C. et al. Expression of Gls and Gls2 transporter (GLAST) protein and mRNA in cultured
Häussinger, D. Hypoosmotic swelling increases protein glutaminase isoforms in astrocytes. Glia 63, 365–382 rat cortical astrocytes. Neurochem. Int. 37, 243–248
tyrosine nitration in cultured rat astrocytes. Glia 47, (2014). (2000).
21–29 (2004). 101. Jördens, M. S. et al. Multidrug resistance-associated 123. Zhou, B.-G. & Norenberg, M. D. Ammonia
78. Häussinger, D. & Schliess, F. Pathogenetic mechanisms protein 4 expression in ammonia-treated cultured downregulates GLAST mRNA glutamate transporter in
of hepatic encephalopathy. Gut 57, 1156–1165 (2008). rat astrocytes and cerebral cortex of cirrhotic patients rat astrocyte cultures. Neurosci. Lett. 276,
79. Rama Rao, K. V., Jayakumar, A. R., Tong, X., with hepatic encephalopathy. Glia 63, 2092–2105 145–148 (1999).
Alvarez, V. M. & Norenberg, M. D. Marked potentiation (2015). 124. Limbad, C. et al. Astrocyte senescence promotes
of cell swelling by cytokines in ammonia-sensitized 102. Kruczek, C. et al. Ammonia increases nitric oxide, glutamate toxicity in cortical neurons. PLoS ONE
cultured astrocytes. J. Neuroinflammation 7, 66 free Zn2+, and metallothionein mRNA expression in 15, e0227887–e0227887 (2020).
(2010). cultured rat astrocytes. Biol. Chem. 392, 1155–1165 125. Bajaj, J. S. et al. Persistence of cognitive impairment
80. Jayakumar, A. R., Tong, X. Y., Ospel, J. & (2011). after resolution of overt hepatic encephalopathy.
Norenberg, M. D. Role of cerebral endothelial cells in 103. Lavoie, J., Layrargues, G. P. & Butterworth, R. F. Gastroenterology 138, 2332–2340 (2010).
the astrocyte swelling and brain edema associated Increased densities of peripheral-type benzodiazepine 126. Riggio, O. et al. Evidence of persistent cognitive
with acute hepatic encephalopathy. Neuroscience receptors in brain autopsy samples from cirrhotic impairment after resolution of overt hepatic
218, 305–316 (2012). patients with hepatic encephalopathy. Hepatology 11, encephalopathy. Clin. Gastroenterol. Hepatol. 9,
81. Rama Rao, K. V. & Norenberg, M. D. Glutamine 874–878 (1990). 181–183 (2011).
in the pathogenesis of hepatic encephalopathy: 104. Ahboucha, S. et al. Indomethacin improves 127. Sobczyk, K., Jördens, M. S., Karababa, A., Görg, B.
the trojan horse hypothesis revisited. Neurochem. locomotor deficit and reduces brain concentrations & Häussinger, D. Ephrin/ephrin receptor expression
Res. 39, 593–598 (2013). of neuroinhibitory steroids in rats following portacaval in ammonia-treated rat astrocytes and in human
82. Zemtsova, I. et al. Microglia activation in hepatic anastomosis. Neurogastroenterol. Motil. 20, cerebral cortex in hepatic encephalopathy.
encephalopathy in rats and humans. Hepatology 54, 949–957 (2008). Neurochem. Res. 40, 274–283 (2014).
204–215 (2011). 105. Ahboucha, S., Pomier-Layrargues, G., Mamer, O. 128. Schrimpf, A. et al. Hyperammonemia-induced
83. Bosoi, C. R. et al. Systemic oxidative stress is & Butterworth, R. F. Increased levels of pregnenolone changes in the cerebral transcriptome and proteome.
implicated in the pathogenesis of brain edema in and its neuroactive metabolite allopregnanolone in Anal. Biochem. 641, 114548 (2022).
rats with chronic liver failure. Free Radic. Biol. Med. autopsied brain tissue from cirrhotic patients who 129. Cabrera-Pastor, A. et al. Peripheral inflammation
52, 1228–1235 (2012). died in hepatic coma. Neurochem. Int. 49, 372–378 induces neuroinflammation that alters
84. Nikolov, P. et al. Altered motor cortical plasticity (2006). neurotransmission and cognitive and motor function
in patients with hepatic encephalopathy: a paired 106. Keitel, V. et al. The bile acid receptor TGR5 (Gpbar-1) in hepatic encephalopathy: underlying mechanisms
associative stimulation study. Clin. Neurophysiol. acts as a neurosteroid receptor in brain. Glia 58, and therapeutic implications. Acta Physiol. 226,
132, 2332–2341 (2021). 1794–1805 (2010). e13270 (2019).
85. Butz, M. et al. Motor impairment in liver cirrhosis 107. Oenarto, J. et al. Ammonia-induced miRNA 130. Coltart, I., Tranah, T. H. & Shawcross, D. L.
without and with minimal hepatic encephalopathy. expression changes in cultured rat astrocytes. Inflammation and hepatic encephalopathy.
Acta Neurol. Scand. 122, 27–35 (2009). Sci. Rep. 6, 18493–18493 (2016). Arch. Biochem. Biophys. 536, 189–196 (2013).

18 | Article citation ID: (2022) 8:43 www.nature.com/nrdp

0123456789();:
Primer

131. Felipo, V. et al. Contribution of hyperammonemia 154. Arenas, Y. M., Cabrera-Pastor, A., Juciute, N., 179. Jindal, A. & Jagdish, R. K. Sarcopenia: ammonia
and inflammatory factors to cognitive impairment Mora-Navarro, E. & Felipo, V. Blocking glycine metabolism and hepatic encephalopathy. Clin. Mol.
in minimal hepatic encephalopathy. Metab. Brain Dis. receptors reduces neuroinflammation and Hepatol. 25, 270–279 (2019).
27, 51–58 (2011). restores neurotransmission in cerebellum 180. García, P. S., Cabbabe, A., Kambadur, R., Nicholas, G.
132. Pozdeev, V. I. et al. TNFα induced up-regulation through ADAM17-TNFR1-NF-κβ pathway. & Csete, M. Brief-reports: elevated myostatin levels
of Na+,K+,2Cl− cotransporter NKCC1 in hepatic J. Neuroinflammation 17, 269 (2020). in patients with liver disease: a potential contributor
ammonia clearance and cerebral ammonia toxicity. 155. Yurdaydin, C. et al. Increased serotoninergic and to skeletal muscle wasting. Anesth. Analg. 111,
Sci. Rep. 7, 7938 (2017). noradrenergic activity in hepatic encephalopathy 707–709 (2010).
133. Rodrigo, R. et al. Hyperammonemia induces in rats with thioacetamide-induced acute liver failure. 181. Conn, H. O. et al. Comparison of lactulose and
neuroinflammation that contributes to cognitive Hepatology 12, 695–700 (1990). neomycin in the treatment of chronic portal-systemic
impairment in rats with hepatic encephalopathy. 156. García-Ayllón, M.-S. et al. Brain cholinergic encephalopathy. Gastroenterology 72, 573–583
Gastroenterology 139, 675–684 (2010). impairment in liver failure. Brain J. Neurol. 131, (1977).
134. Balzano, T. et al. Chronic hyperammonemia induces 2946–2956 (2008). 182. Conn, H. & Lieberthal, M. in The Hepatic Coma
peripheral inflammation that leads to cognitive 157. Chen, B. et al. The critical role of hippocampal Syndromes and Lactulose 5–8 (Williams & Wilkins,
impairment in rats: reversed by anti-TNF-α treatment. dopamine in the pathogenesis of hepatic 1979).
J. Hepatol. 73, 582–592 (2020). encephalopathy. Physiol. Res. 70, 101–110 183. Reuter, B. et al. Assessment of the spectrum of hepatic
135. Görg, B., Wettstein, M., Metzger, S., Schliess, F. & (2021). encephalopathy: a multicenter study. Liver Transpl.
Häussinger, D. Lipopolysaccharide-induced tyrosine 158. Hassan, S. S. et al. Cerebellar inhibition in 24, 587–594 (2018).
nitration and inactivation of hepatic glutamine hepatic encephalopathy. Clin. Neurophysiol. 130, 184. Bajaj, J. S. et al. Review article: the design of clinical
synthetase in the rat. Hepatology 41, 1065–1073 886–892 (2019). trials in hepatic encephalopathy-an International
(2005). 159. Groiss, S. J. et al. GABA-ergic tone hypothesis Society for Hepatic Encephalopathy and Nitrogen
136. Bajaj, J. S. et al. Colonic mucosal microbiome differs in hepatic encephalopathy–revisited. Metabolism (ISHEN) consensus statement. Aliment.
from stool microbiome in cirrhosis and hepatic Clin. Neurophysiol. 130, 911–916 (2019). Pharmacol. Ther. 33, 739–747 (2011).
encephalopathy and is linked to cognition and 160. Nardone, R. et al. Intracortical inhibitory and 185. Montagnese, S. et al. Hepatic encephalopathy 2018:
inflammation. Am. J. Physiol. Gastrointest. Liver excitatory circuits in subjects with minimal hepatic a clinical practice guideline by the Italian Association
Physiol. 303, G675–G685 (2012). encephalopathy: a TMS study. Metab. Brain Dis. 31, for the Study of the Liver (AISF). Dig. Liver Dis. 51,
137. Dhiman, R. K. Gut microbiota, inflammation and 1065–1070 (2016). 190–205 (2019).
hepatic encephalopathy: a puzzle with a solution 161. Montagnese, S. et al. A pilot study of golexanolone, 186. Montagnese, S. et al. Covert hepatic encephalopathy:
in sight. J. Clin. Exp. Hepatol. 2, 207–210 (2012). a new GABA-A receptor-modulating steroid antagonist, agreement and predictive validity of different
138. Acharya, C. & Bajaj, J. S. Altered microbiome in in patients with covert hepatic encephalopathy. indices. World J. Gastroenterol. 20, 15756–15762
patients with cirrhosis and complications. Clin. J. Hepatol. 75, 98–107 (2021). (2014).
Gastroenterol. Hepatol. 17, 307–321 (2019). 162. Taoro-González, L. et al. Differential role of interleukin- 187. Thomsen, K. L., Macnaughtan, J., Tritto, G.,
139. Iebba, V. et al. Combining amplicon sequencing 1β in neuroinflammation-induced impairment of Mookerjee, R. P. & Jalan, R. Clinical and
and metabolomics in cirrhotic patients highlights spatial and nonspatial memory in hyperammonemic pathophysiological characteristics of cirrhotic patients
distinctive microbiota features involved in bacterial rats. FASEB J. 33, 9913–9928 (2019). with grade 1 and minimal hepatic encephalopathy.
translocation, systemic inflammation and hepatic 163. Schnitzler, A. & Gross, J. Normal and pathological PLoS ONE 11, e0146076 (2016).
encephalopathy. Sci. Rep. 8, 8210–8210 (2018). oscillatory communication in the brain. Nat. Rev. 188. Romero-Gómez, M., Boza, F., García-Valdecasas, M. S.,
140. Montoliu, C. et al. IL-6 and IL-18 in blood may Neurosci. 6, 285–296 (2005). García, E. & Aguilar-Reina, J. Subclinical hepatic
discriminate cirrhotic patients with and without 164. Butz, M., May, E. S., Häussinger, D. & Schnitzler, A. encephalopathy predicts the development of overt
minimal hepatic encephalopathy. J. Clin. The slowed brain: cortical oscillatory activity in hepatic encephalopathy. Am. J. Gastroenterol. 96,
Gastroenterol. 43, 272–279 (2009). hepatic encephalopathy. Arch. Biochem. Biophys. 2718–2723 (2001).
141. Azhari, H. & Swain, M. G. Role of peripheral 536, 197–203 (2013). 189. Schomerus, H. & Hamster, W. Quality of life in
inflammation in hepatic encephalopathy. 165. Timmermann, L., Gross, J., Kircheis, G., cirrhotics with minimal hepatic encephalopathy.
J. Clin. Exp. Hepatol. 8, 281–285 (2018). Häussinger, D. & Schnitzler, A. Cortical origin Metab. Brain Dis. 16, 37–41 (2001).
142. Dadsetan, S. et al. Reducing peripheral inflammation of mini-asterixis in hepatic encephalopathy. Neurology 190. Kircheis, G. et al. Hepatic encephalopathy and fitness
with infliximab reduces neuroinflammation and 58, 295–298 (2002). to drive. Gastroenterology 137, 1706–1715.e9
improves cognition in rats with hepatic encephalopathy. 166. Timmermann, L. et al. Mini-asterixis in hepatic (2009).
Front. Mol. Neurosci. 9, 106–106 (2016). encephalopathy induced by pathologic thalamo- 191. Kircheis, G., Hilger, N. & Häussinger, D. Value of
143. Mangas-Losada, A. et al. Minimal hepatic motor-cortical coupling. Neurology 61, 689–692 critical flicker frequency and psychometric hepatic
encephalopathy is associated with expansion and (2003). encephalopathy score in diagnosis of low-grade
activation of CD4+CD28−, Th22 and Tfh and B 167. Timmermann, L. et al. Impaired cerebral hepatic encephalopathy. Gastroenterology 146,
lymphocytes. Sci. Rep. 7, 6683–6683 (2017). oscillatory processing in hepatic encephalopathy. 961–969.e11 (2014).
144. Shen, J. et al. The role of exosomes in hepatitis, Clin. Neurophysiol. 119, 265–272 (2008). 192. Häussinger, D. et al. in Hepatic Encephalopathy and
liver cirrhosis and hepatocellular carcinoma. 168. Kahlbrock, N. et al. Lowered frequency and Nitrogen Metabolism 423–432 (Springer Dordrecht,
J. Cell. Mol. Med. 21, 986–992 (2017). impaired modulation of gamma band oscillations 2006).
145. Thietart, S. & Rautou, P.-E. Extracellular vesicles in a bimodal attention task are associated with 193. Weissenborn, K., Heidenreich, S., Ennen, J., Rückert, N.
as biomarkers in liver diseases: a clinician’s point reduced critical flicker frequency. NeuroImage 61, & Hecker, H. Attention deficits in minimal hepatic
of view. J. Hepatol. 73, 1507–1525 (2020). 216–227 (2012). encephalopathy. Metab. Brain Dis. 16, 13–19
146. Eguchi, A. & Feldstein, A. E. Extracellular vesicles 169. Fries, P. Neuronal gamma-band synchronization (2001).
in non-alcoholic and alcoholic fatty liver diseases. as a fundamental process in cortical computation. 194. Campagna, F. et al. The animal naming test: an easy
Liver Res. 2, 30–34 (2018). Annu. Rev. Neurosci. 32, 209–224 (2009). tool for the assessment of hepatic encephalopathy.
147. Izquierdo-Altarejos, P., Cabrera-Pastor, A., 170. May, E. S. et al. Hepatic encephalopathy is associated Hepatology 66, 198–208 (2017).
Gonzalez-King, H., Montoliu, C. & Felipo, V. with slowed and delayed stimulus-associated 195. Lauridsen, M. M., Grønbæk, H., Næser, E. B.,
Extracellular vesicles from hyperammonemic rats somatosensory alpha activity. Clin. Neurophysiol. Leth, S. T. & Vilstrup, H. Gender and age effects
induce neuroinflammation and motor incoordination 125, 2427–2435 (2014). on the continuous reaction times method in volunteers
in control rats. Cells 9, 572 (2020). 171. Lazar, M. et al. Impaired tactile temporal discrimination and patients with cirrhosis. Metab. Brain Dis. 27,
148. McMillin, M. et al. Bile acid-mediated sphingosine-1- in patients with hepatic encephalopathy. Front. Psychol. 559–565 (2012).
phosphate receptor 2 signaling promotes 9, 2059 (2018). 196. Lauridsen, M. M., Thiele, M., Kimer, N. & Vilstrup, H.
neuroinflammation during hepatic encephalopathy 172. Baumgarten, T. J., Schnitzler, A. & Lange, J. Beta The continuous reaction times method for diagnosing,
in mice. Front. Cell. Neurosci. 11, 191–191 oscillations define discrete perceptual cycles in the grading, and monitoring minimal/covert hepatic
(2017). somatosensory domain. Proc. Natl Acad. Sci. USA encephalopathy. Metab. Brain Dis. 28, 231–234
149. D’Mello, C., Le, T. & Swain, M. G. Cerebral microglia 112, 12187–12192 (2015). (2013).
recruit monocytes into the brain in response to 173. Kullmann, F. et al. Brain electrical activity mapping 197. Bajaj, J. S. et al. Inhibitory control test is
tumor necrosis factoralpha signaling during peripheral of EEG for the diagnosis of (sub)clinical hepatic a simple method to diagnose minimal hepatic
organ inflammation. J. Neurosci. 29, 2089–2102 encephalopathy in chronic liver disease. Eur. J. encephalopathy and predict development of overt
(2009). Gastroenterol. Hepatol. 13, 513–522 (2001). hepatic encephalopathy. Am. J. Gastroenterol. 102,
150. Balzano, T. et al. Rifaximin prevents T-lymphocytes 174. Baumgarten, T. J. et al. Connecting occipital alpha 754–760 (2007).
and macrophages infiltration in cerebellum and band peak frequency, visual temporal resolution, 198. Amodio, P. et al. Improving the inhibitory control
restores motor incoordination in rats with mild and occipital GABA levels in healthy participants and task to detect minimal hepatic encephalopathy.
liver damage. Biomedicines 9, 1002 (2021). hepatic encephalopathy patients. NeuroImage Clin. Gastroenterology 139, 510–518.e2 (2010).
151. Dennis, C. V. et al. Microglial proliferation 20, 347–356 (2018). 199. Bajaj, J. S. et al. The Stroop smartphone application
in the brain of chronic alcoholics with hepatic 175. VanRullen, R. Perceptual cycles. Trends Cogn. Sci. 20, is a short and valid method to screen for minimal
encephalopathy. Metab. Brain Dis. 29, 1027–1039 723–735 (2016). hepatic encephalopathy. Hepatology 58, 1122–1132
(2014). 176. Hadjihambi, A. et al. Impaired brain glymphatic flow in (2013).
152. Balzano, T. et al. The cerebellum of patients with experimental hepatic encephalopathy. J. Hepatol. 70, 200. Amodio, P. et al. Clinical features and survivial
steatohepatitis shows lymphocyte infiltration, 40–49 (2019). of cirrhotic patients with subclinical cognitive
microglial activation and loss of purkinje and granular 177. Claeys, W. et al. The neurogliovascular unit in alterations detected by the number connection test
neurons. Sci. Rep. 8, 3004–3004 (2018). hepatic encephalopathy. JHEP Rep. Innov. Hepatol. and computerized psychometric tests. Hepatology 29,
153. Cauli, O., Mansouri, M. T., Agusti, A. & Felipo, V. 3, 100352 (2021). 1662–1667 (1999).
Hyperammonemia increases GABAergic tone in 178. Costa, R. & Montagnese, S. The role of astrocytes 201. Amodio, P. et al. Spectral versus visual EEG analysis
the cerebellum but decreases it in the rat cortex. in generating circadian rhythmicity in health and in mild hepatic encephalopathy. Clin. Neurophysiol.
Gastroenterology 136, 1359–1367.e2 (2009). disease. J. Neurochem. 157, 42–52 (2021). 110, 1334–1344 (1999).


NATURE REVIEwS | DISEASE PRIMERS | Article citation ID: (2022) 8:43 19

0123456789();:
Primer

202. Schiff, S. et al. A low-cost, user-friendly 225. Zhang, L. J., Yang, G., Yin, J., Liu, Y. & Qi, J. review and meta-analysis. Front. Neurosci. 15,
electroencephalographic recording system for the Neural mechanism of cognitive control impairment in 579263 (2021).
assessment of hepatic encephalopathy. Hepatology patients with hepatic cirrhosis: a functional magnetic 249. Matsumoto, S. et al. Urea cycle disorders-update.
63, 1651–1659 (2016). resonance imaging study. Acta Radiol. 48, 577–587 J. Hum. Genet. 64, 833–847 (2019).
203. Kircheis, G., Wettstein, M., Timmermann, L., (2007). 250. Häberle, J. et al. Suggested guidelines for the
Schnitzler, A. & Häussinger, D. Critical flicker 226. Broyd, S. J. et al. Default-mode brain dysfunction diagnosis and management of urea cycle disorders:
frequency for quantification of low-grade hepatic in mental disorders: a systematic review. Neurosci. First revision. J. Inherit. Metab. Dis. 42, 1192–1230
encephalopathy. Hepatology 35, 357–366 (2002). Biobehav. Rev. 33, 279–296 (2009). (2019).
204. Romero-Gómez, M. Critical flicker frequency: 227. Rovira, A., Grivé, E., Pedraza, S., Rovira, A. & 251. Plauth, M. et al. ESPEN guideline on clinical
it is time to break down barriers surrounding minimal Alonso, J. Magnetization transfer ratio values and nutrition in liver disease. Clin. Nutr. 38, 485–521
hepatic encephalopathy. J. Hepatol. 47, 10–11 proton MR spectroscopy of normal-appearing cerebral (2019).
(2007). white matter in patients with liver cirrhosis. AJNR Am. 252. Amodio, P. et al. The nutritional management of
205. Kircheis, G. et al. Diagnostic and prognostic values J. Neuroradiol. 22, 1137–1142 (2001). hepatic encephalopathy in patients with cirrhosis:
of critical flicker frequency determination as new 228. Laubenberger, J. et al. Proton magnetic resonance International society for hepatic encephalopathy
diagnostic tool for objective HE evaluation in patients spectroscopy of the brain in symptomatic and and nitrogen metabolism consensus. Hepatology 58,
undergoing TIPS implantation. Eur. J. Gastroenterol. asymptomatic patients with liver cirrhosis. 325–336 (2013).
Hepatol. 21, 1383–1394 (2009). Gastroenterology 112, 1610–1616 (1997). 253. Yoshiji, H. et al. Evidence-based clinical practice
206. Berlioux, P. et al. Pre-transjugular intrahepatic 229. McPhail, M. J. W., Patel, N. R. & Taylor-Robinson, S. D. guidelines for liver cirrhosis 2020. J. Gastroenterol.
portosystemic shunts (TIPS) prediction of post-TIPS Brain imaging and hepatic encephalopathy. Clin. Liver 56, 593–619 (2021).
overt hepatic encephalopathy: the critical flicker Dis. 16, 57–72 (2012). 254. European Association for the Study of the Liver. EASL
frequency is more accurate than psychometric tests. 230. Grover, V. P. B. et al. A longitudinal study of clinical practice guidelines on nutrition in chronic liver
Hepatology 59, 622–629 (2013). patients with cirrhosis treated with L-ornithine disease. J. Hepatol. 70, 172–193 (2019).
207. Romero-Gómez, M. et al. Value of the critical L-aspartate, examined with magnetization transfer, 255. Tsien, C. D., McCullough, A. J. & Dasarathy, S.
flicker frequency in patients with minimal hepatic diffusion-weighted imaging and magnetic resonance Late evening snack: exploiting a period of anabolic
encephalopathy. Hepatology 45, 879–885 spectroscopy. Metab. Brain Dis. 32, 77–86 (2016). opportunity in cirrhosis. J. Gastroenterol. Hepatol. 27,
(2007). 231. Taylor-Robinson, S. D., Buckley, C., Changani, K. K., 430–441 (2012).
208. Ampuero, J. et al. Minimal hepatic encephalopathy Hodgson, H. J. F. & Bell, J. D. Cerebral proton and 256. Maharshi, S., Sharma, B. C., Sachdeva, S.,
and critical flicker frequency are associated with phosphorus-31 magnetic resonance spectroscopy Srivastava, S. & Sharma, P. Efficacy of nutritional
survival of patients with cirrhosis. Gastroenterology in patients with subclinical hepatic encephalopathy. therapy for patients with cirrhosis and minimal
149, 1483–1489 (2015). Liver Int. 19, 389–398 (1999). hepatic encephalopathy in a randomized trial.
209. Bandookwala, M. & Sengupta, P. 3-Nitrotyrosine: 232. Patwardhan, V. R. et al. Serum ammonia is associated Clin. Gastroenterol. Hepatol. 14, 454–460.e3
a versatile oxidative stress biomarker for major with transplant-free survival in hospitalized patients (2016).
neurodegenerative diseases. Int. J. Neurosci. 130, with acutely decompensated cirrhosis [corrected]. 257. Takuma, Y., Nouso, K., Makino, Y., Hayashi, M.
1047–1062 (2020). J. Clin. Gastroenterol. 50, 345–350 (2016). & Takahashi, H. Clinical trial: oral zinc in hepatic
210. Montoliu, C. et al. 3-nitro-tyrosine as a peripheral 233. Vierling, J. M. et al. Fasting blood ammonia predicts encephalopathy. Aliment. Pharmacol. Ther. 32,
biomarker of minimal hepatic encephalopathy in risk and frequency of hepatic encephalopathy episodes 1080–1090 (2010).
patients with liver cirrhosis. Am. J. Gastroenterol. in patients with cirrhosis. Clin. Gastroenterol. Hepatol. 258. Bruyneel, M. & Sersté, T. Sleep disturbances in
106, 1629–1637 (2011). 14, 903–906.e1 (2016). patients with liver cirrhosis: prevalence, impact,
211. Gairing, S. J. et al. Evaluation of IL-6 for stepwise 234. Shalimar et al. Prognostic role of ammonia in patients and management challenges. Nat. Sci. Sleep 10,
diagnosis of minimal hepatic encephalopathy with cirrhosis. Hepatology 70, 982–994 (2019). 369–375 (2018).
in patients with liver cirrhosis. Hepatol. Commun. 235. Clemmesen, J. O. et al. Hepatic blood flow and 259. Riggio, O., Nardelli, S., Gioia, S., Lucidi, C. &
https://doi.org/10.1002/hep4.1883 (2022). splanchnic oxygen consumption in patients with Merli, M. Management of hepatic encephalopathy
212. Labenz, C. et al. Raised serum interleukin-6 identifies liver failure. Effect of high-volume plasmapheresis. as an inpatient. Clin. Liver Dis. 5, 79–82 (2015).
patients with liver cirrhosis at high risk for overt Hepatology 29, 347–355 (1999). 260. Davuluri, G. et al. Hyperammonaemia-induced
hepatic encephalopathy. Aliment. Pharmacol. Ther. 236. Bhatia, V., Singh, R. & Acharya, S. K. Predictive skeletal muscle mitochondrial dysfunction results
50, 1112–1119 (2019). value of arterial ammonia for complications and in cataplerosis and oxidative stress. J. Physiol. 594,
213. Quero Guillén, J. C. & Herrerías Gutiérrez, J. M. outcome in acute liver failure. Gut 55, 98–104 7341–7360 (2016).
Diagnostic methods in hepatic encephalopathy. (2006). 261. Dasarathy, S. & Hatzoglou, M. Hyperammonemia and
Clin. Chim. Acta 365, 1–8 (2006). 237. Rose, C. F., Jalan, R. & Shawcross, D. L. Erroneous proteostasis in cirrhosis. Curr. Opin. Clin. Nutr. Metab.
214. Grover, V. B. Current and future applications of magnetic ammonia measurement is not synonymous with a lack Care 21, 30–36 (2018).
resonance imaging and spectroscopy of the brain in of efficacy of ammonia-lowering therapies in hepatic 262. Dam, G., Aamann, L., Vistrup, H. & Gluud, L. L.
hepatic encephalopathy. World J. Gastroenterol. 12, encephalopathy. Clin. Gastroenterol. Hepatol. 19, The role of branched chain amino acids in the
2969 (2006). 2456–2457 (2021). treatment of hepatic encephalopathy. J. Clin.
215. Pagani, E., Bizzi, A., Di Salle, F., De Stefano, N. 238. Kramer, L. et al. Partial pressure of ammonia versus Exp. Hepatol. 8, 448–451 (2018).
& Filippi, M. Basic concepts of advanced MRI ammonia in hepatic encephalopathy. Hepatology 31, 263. Gluud, L. L. et al. in Cochrane Database of Systematic
techniques. Neurol. Sci. 29, 290–295 (2008). 30–34 (2000). Reviews (John Wiley & Sons, Ltd., 2015).
216. Berding, G. et al. Radiotracer imaging studies in 239. Drolz, A. et al. Clinical impact of arterial ammonia 264. Lai, J. C. & Tandon, P. Improving nutritional status
hepatic encephalopathy: ISHEN practice guidelines. levels in ICU patients with different liver diseases. in patients with cirrhosis. Am. J. Gastroenterol. 113,
Liver Int. 29, 621–628 (2009). Intensive Care Med. 39, 1227–1237 (2013). 1574–1576 (2018).
217. Smith, S. M. et al. Accurate, robust, and automated 240. Huizenga, J. R., Gips, C. H., Conn, H. O. & Jansen, P. L. 265. Sharma, B. C., Sharma, P., Agrawal, A. & Sarin, S. K.
longitudinal and cross-sectional brain change analysis. Determination of ammonia in ear-lobe capillary blood Secondary prophylaxis of hepatic encephalopathy:
NeuroImage 17, 479–489 (2002). is an alternative to arterial blood ammonia. Clin. Chim. an open-label randomized controlled trial of lactulose
218. Patel, N., White, S., Dhanjal, N. S., Oatridge, A. Acta Int. J. Clin. Chem. 239, 65–70 (1995). versus placebo. Gastroenterology 137, 885–891,
& Taylor-Robinson, S. D. Changes in brain size in 241. Bersagliere, A. et al. Ammonia-related changes 891.e1 (2009).
hepatic encephalopathy: a coregistered MRI study. in cerebral electrogenesis in healthy subjects and 266. Agrawal, A., Sharma, B. C., Sharma, P. &
Metab. Brain Dis. 19, 431–445 (2004). patients with cirrhosis. Clin. Neurophysiol. 124, Sarin, S. K. Secondary prophylaxis of hepatic
219. Taylor-Robinson, S. D. et al. MR imaging of the 492–496 (2013). encephalopathy in cirrhosis: an open-label,
basal ganglia in chronic liver disease: correlation 242. Hartmann, I. J. C. et al. The prognostic significance randomized controlled trial of lactulose, probiotics,
of T1-weighted and magnetisation transfer of subclinical hepatic encephalopathy. Am. J. and no therapy. Am. J. Gastroenterol. 107,
contrast measurements with liver dysfunction Gastroenterol. 95, 2029–2034 (2000). 1043–1050 (2012).
and neuropsychiatric status. Metab. Brain Dis. 10, 243. Patidar, K. R. et al. Covert hepatic encephalopathy 267. Dhiman, R. K. et al. Probiotic VSL#3 reduces liver
175–188 (1995). is independently associated with poor survival and disease severity and hospitalization in patients
220. Kumar, R. et al. Voxel-based diffusion tensor magnetic increased risk of hospitalization. Am. J. Gastroenterol. with cirrhosis: a randomized, controlled trial.
resonance imaging evaluation of low-grade hepatic 109, 1757–1763 (2014). Gastroenterology 147, 1327–1337.e3 (2014).
encephalopathy. J. Magn. Reson. Imaging 27, 244. Flud, C. R. & Duarte-Rojo, A. Prognostic implications 268. Bass, N. M. et al. Rifaximin treatment in hepatic
1061–1068 (2008). of minimal/covert hepatic encephalopathy: large-scale encephalopathy. N. Engl. J. Med. 362, 1071–1081
221. Miese, F. et al. 1H-MR spectroscopy, magnetization validation cohort studies. J. Clin. Exp. Hepatol. 9, (2010).
transfer, and diffusion-weighted imaging in alcoholic 112–116 (2019). 269. Mullen, K. D. et al. Rifaximin is safe and well tolerated
and nonalcoholic patients with cirrhosis with hepatic 245. Bustamante, J. et al. Prognostic significance of hepatic for long-term maintenance of remission from overt
encephalopathy. AJNR Am. J. Neuroradiol. 27, encephalopathy in patients with cirrhosis. J. Hepatol. hepatic encephalopathy. Clin. Gastroenterol. Hepatol.
1019–1026 (2006). 30, 890–895 (1999). 12, 1390–1397.e2 (2014).
222. McPhail, M. J. W. & Taylor-Robinson, S. D. The role 246. D’Amico, G., Garcia-Tsao, G. & Pagliaro, L. Natural 270. Gluud, L. L., Vilstrup, H. & Morgan, M. Y.
of magnetic resonance imaging and spectroscopy in history and prognostic indicators of survival in Non-absorbable disaccharides versus placebo/no
hepatic encephalopathy. Metab. Brain Dis. 25, 65–72 cirrhosis: a systematic review of 118 studies. intervention and lactulose versus lactitol for the
(2010). J. Hepatol. 44, 217–231 (2006). prevention and treatment of hepatic encephalopathy
223. Zafiris, O. et al. Neural mechanism underlying 247. Lucidi, C. et al. Hepatic encephalopathy expands in people with cirrhosis.Cochrane Database Syst. Rev.
impaired visual judgement in the dysmetabolic the predictivity of model for end-stage liver disease https://doi.org/10.1002/14651858.cd003044.pub3
brain: an fMRI study. NeuroImage 22, 541–552 in liver transplant setting: evidence by means of (2016).
(2004). 2 independent cohorts. Liver Transpl. 22, 1333–1342 271. Kimer, N., Krag, A., Møller, S., Bendtsen, F. &
224. Cox, I. J. Development and applications of (2016). Gluud, L. L. Systematic review with meta-analysis:
in vivo clinical magnetic resonance spectroscopy. 248. López-Franco, Ó. et al. Cognitive impairment after the effects of rifaximin in hepatic encephalopathy.
Prog. Biophys. Mol. Biol. 65, 45–81 (1996). resolution of hepatic encephalopathy: a systematic Aliment. Pharmacol. Ther. 40, 123–132 (2014).

20 | Article citation ID: (2022) 8:43 www.nature.com/nrdp

0123456789();:
Primer

272. Bajaj, J. S. et al. Modulation of the metabiome by 294. Uschner, F. et al. Safety and preliminary efficacy and 317. Montagnese, S. et al. Sleep-wake abnormalities in
rifaximin in patients with cirrhosis and minimal hepatic pharmocokinetics of intraperitoneal VS-01 infusions patients with cirrhosis. Hepatology 59, 705–712
encephalopathy. PLoS ONE 8, e60042–e60042 in patients with decompensated liver cirrhosis: (2013).
(2013). a first in human open label phase 1b clinical trial. 318. Samanta, J. et al. Correlation between degree
273. DuPont, H. L. Review article: the antimicrobial Hepatology 74, 139A (2021). and quality of sleep disturbance and the level
effects of rifaximin on the gut microbiota. 295. Hassanein, T. I. et al. Randomized controlled of neuropsychiatric impairment in patients with
Aliment. Pharmacol. Ther. 43, 3–10 (2015). study of extracorporeal albumin dialysis for hepatic liver cirrhosis. Metab. Brain Dis. 28, 249–259
274. Goh, E. T. et al. L-ornithine L-aspartate for prevention encephalopathy in advanced cirrhosis. Hepatology 46, (2013).
and treatment of hepatic encephalopathy in people 1853–1862 (2007). 319. Román, E. et al. Minimal hepatic encephalopathy
with cirrhosis. Cochrane Database Syst. Rev. 5, 296. Agarwal, B., Saliba, F. & Tomescu, D. A multi-centre, is associated with falls. Am. J. Gastroenterol. 106,
CD012410 (2018). randomized controlled study, to evaluate the safety 476–482 (2011).
275. Butterworth, R. F. & McPhail, M. J. W. L-Ornithine and performance of the DIALIVE liver dialysis device 320. Soriano, G. et al. Cognitive dysfunction in cirrhosis is
L-aspartate (LOLA) for hepatic encephalopathy in in patients with acute on chronic liver failure (ACLF) associated with falls: a prospective study. Hepatology
cirrhosis: results of randomized controlled trials versus standard of care (SOC) (ALIVER Consortium). 55, 1922–1930 (2012).
and meta-analyses. Drugs 79, 31–37 (2019). Gut 70 (Suppl. 3), A54 (2021). 321. Diamond, T. et al. Osteoporosis and skeletal
276. Naeshiro, N. et al. Percutaneous transvenous 297. Simón-Talero, M. et al. Effects of intravenous fractures in chronic liver disease. Gut 31, 82–87
embolization for portosystemic shunts associated with albumin in patients with cirrhosis and episodic (1990).
encephalopathy: long-term outcomes in 14 patients. hepatic encephalopathy: a randomized double-blind 322. Bajaj, J. S. et al. Minimal hepatic encephalopathy and
Hepatol. Res. 44, 740–749 (2014). study. J. Hepatol. 59, 1184–1192 (2013). mild cognitive impairment worsen quality of life in
277. Lv, Y. et al. Concurrent large spontaneous portosystemic 298. Sharma, B. C. et al. Randomized controlled trial elderly patients with cirrhosis. Clin. Gastroenterol.
shunt embolization for the prevention of overt hepatic comparing lactulose plus albuminversuslactulose Hepatol. 18, 3008–3016.e2 (2020).
encephalopathy after TIPS: a randomized controlled alone for treatment of hepatic encephalopathy. 323. Wang, J. Y. et al. Lactulose improves cognition,
trial. Hepatology https://doi.org/10.1002/hep.32453 J. Gastroenterol. Hepatol. 32, 1234–1239 quality of life, and gut microbiota in minimal hepatic
(2022). (2017). encephalopathy: a multicenter, randomized controlled
278. Álvarez-López, P. et al. Spontaneous portosystemic 299. Ventura-Cots, M. et al. Effects of albumin on survival trial. J. Dig. Dis. 20, 547–556 (2019).
shunt embolization in liver transplant recipients with after a hepatic encephalopathy episode: randomized 324. Bruyneel, M. et al. Improvement of sleep architecture
recurrent hepatic encephalopathy. Ann. Hepatol. 27, double-blind trial and meta-analysis. J. Clin. Med. 10, parameters in cirrhotic patients with recurrent
100687 (2022). 4885 (2021). hepatic encephalopathy with the use of rifaximin.
279. Temmerman, F., Laleman, W., Maleux, G. & Nevens, F. 300. Sundaram, V. et al. Longterm outcomes of patients Eur. J. Gastroenterol. Hepatol. 29, 302–308
Treatment of recurrent severe hepatic encephalopathy undergoing liver transplantation for acute-on-chronic (2017).
in patients with large porto-collaterals shunts or liver failure. Liver Transplant. 26, 1594–1602 325. Sack, J. & Hashemi, N. Smartphone-based remote
transjugular portosystemic shunt. Acta Gastroenterol. (2020). health monitoring-implications for healthcare delivery
Belg. 83, 67–71 (2020). 301. European Association for the Study of the Liver. in patients with cirrhosis. J. Gen. Intern. Med. 34,
280. Laleman, W. et al. Embolization of large spontaneous EASL clinical practice guidelines: liver transplantation. 2726–2727 (2019).
portosystemic shunts for refractory hepatic J. Hepatol. 64, 433–485 (2016). 326. Romero-Gómez, M. Variations in the promoter region
encephalopathy: a multicenter survey on safety 302. Kamath, P. S. et al. A model to predict survival in of the glutaminase gene and the development of
and efficacy. Hepatology 57, 2448–2457 (2013). patients with end-stage liver disease. Hepatology 33, hepatic encephalopathy in patients with cirrhosis.
281. Rahimi, R. S., Singal, A. G., Cuthbert, J. A. & 464–470 (2001). Ann. Intern. Med. 153, 281 (2010).
Rockey, D. C. Lactulose vs polyethylene Glycol 303. Sundaram, V. et al. Patients with severe acute-on- 327. Yokoyama, K. et al. Hydrogen-producing small
3350-electrolyte solution for treatment of overt chronic liver failure are disadvantaged by model intestinal bacterial overgrowth is associated with
hepatic encephalopathy. JAMA Intern. Med. 174, for end-stage liver disease-based organ allocation hepatic encephalopathy and liver function. PLoS ONE
1727 (2014). policy. Aliment. Pharmacol. Ther. 52, 1204–1213 17, e0264459 (2022).
282. Bajaj, J. S. et al. Fecal microbiota transplant from a (2020). 328. Rush, B. et al. Lower 90-day hospital readmission
rational stool donor improves hepatic encephalopathy: 304. Sotil, E. U., Gottstein, J., Ayala, E., Randolph, C. rates for esophageal variceal bleeding after TIPS:
a randomized clinical trial. Hepatology 66, & Blei, A. T. Impact of preoperative overt hepatic a nationwide linked analysis. J. Clin. Gastroenterol.
1727–1738 (2017). encephalopathy on neurocognitive function after 54, 90–95 (2020).
283. Mullish, B. H., McDonald, J. A. K., Thursz, M. R. liver transplantation. Liver Transplant. 15, 184–192 329. Labenz, C. et al. Association between diabetes
& Marchesi, J. R. Fecal microbiota transplant (2009). mellitus and hepatic encephalopathy in patients with
from a rational stool donor improves hepatic 305. Garcia-Martinez, R. et al. Hepatic encephalopathy cirrhosis. Aliment. Pharmacol. Ther. 52, 527–536
encephalopathy: a randomized clinical trial. is associated with posttransplant cognitive function (2020).
Hepatology 66, 1354–1355 (2017). and brain volume. Liver Transplant. 17, 38–46 330. Häussinger, D., Görg, B., Reinehr, R. & Schliess, F.
284. Kurtz, C. B. et al. An engineered E. coli Nissle (2011). Protein tyrosine nitration in hyperammonemia
improves hyperammonemia and survival in mice and 306. Ochoa-Sanchez, R., Tamnanloo, F. & Rose, C. F. and hepatic encephalopathy. Metab. Brain Dis. 20,
shows dose-dependent exposure in healthy humans. Hepatic encephalopathy: from metabolic to 285–294 (2005).
Sci. Transl. Med. 11, eaau7975 (2019). neurodegenerative. Neurochem. Res. 46, 2612–2625 331. Brenner, M. et al. Patients with manifest hepatic
285. Synlogic. Synlogic discontinues development of (2021). encephalopathy can reveal impaired thermal
SYNB1020 to treat hyperammonemia. Synlogic 307. Younossi, Z. M. et al. Health-related quality of life perception. Acta Neurol. Scand. 132, 156–163
https://investor.synlogictx.com/news-releases/news- in chronic liver disease: the impact of type and (2015).
release-details/synlogic-discontinues-development- severity of disease. Am. J. Gastroenterol. 96, 332. Götz, T. et al. Impaired evoked and resting-state brain
synb1020-treat-hyperammonemia (2019). 2199–2205 (2001). oscillations in patients with liver cirrhosis as revealed
286. Dalal, R., McGee, R. G., Riordan, S. M. & Webster, A. C. 308. Afendy, A. et al. Predictors of health-related by magnetoencephalography. NeuroImage Clin. 2,
Probiotics for people with hepatic encephalopathy. quality of life in patients with chronic liver disease. 873–882 (2013).
Cochrane Database Syst. Rev. 2, CD008716 (2017). Aliment. Pharmacol. Ther. 30, 469–476 (2009). 333. Oeltzschner, G. et al. Low visual cortex GABA levels in
287. Wiest, R., Albillos, A., Trauner, M., Bajaj, J. S. 309. Montagnese, S. & Bajaj, J. S. Impact of hepatic hepatic encephalopathy: links to blood ammonia,
& Jalan, R. Targeting the gut-liver axis in liver disease. encephalopathy in cirrhosis on quality-of-life issues. critical flicker frequency, and brain osmolytes. Metab.
J. Hepatol. 67, 1084–1103 (2017). Drugs 79, 11–16 (2019). Brain Dis. 30, 1429–1438 (2015).
288. Jalan, R., Wright, G., Davies, N. A. & Hodges, S. J. 310. Rabiee, A. et al. Factors associated with health-related 334. Zöllner, H. J. et al. Chemical exchange saturation
l-Ornithine phenylacetate (OP): a novel treatment quality of life in patients with cirrhosis: a systematic transfer imaging in hepatic encephalopathy.
for hyperammonemia and hepatic encephalopathy. review. Liver Int. 41, 6–15 (2020). NeuroImage Clin. 22, 101743 (2019).
Med. Hypotheses 69, 1064–1069 (2007). 311. Prasad, S. et al. Lactulose improves cognitive 335. Schomerus, H. et al. Latent portasystemic
289. Rahimi, R. S. et al. Efficacy and safety of ornithine functions and health-related quality of life in encephalopathy. Dig. Dis. Sci. 26, 622–630
phenylacetate for treating overt hepatic encephalopathy patients with cirrhosis who have minimal hepatic (1981).
in a randomized trial. Clin. Gastroenterol. Hepatol. 19, encephalopathy. Hepatology 45, 549–559 (2007). 336. Watanabe, A., Tuchida, T., Yata, Y. & Kuwabara, Y.
2626–2635.e7 (2021). 312. Agrawal, S., Umapathy, S. & Dhiman, R. K. Minimal Evaluation of neuropsychological function in patients
290. Safadi, R. et al. Pharmacokinetics/pharmacodynamics hepatic encephalopathy impairs quality of life. with liver cirrhosis with special reference to their
of L-ornithine phenylacetate in overt hepatic J. Clin. Exp. Hepatol. 5, S42–S48 (2015). driving ability. Metab. Brain Dis. 10, 239–248
encephalopathy and the effect of plasma ammonia 313. Groeneweg, M. et al. Subclinical hepatic (1995).
concentration reduction on clinical outcomes. encephalopathy impairs daily functioning. Hepatology 337. Wein, C., Koch, H., Popp, B., Oehler, G. & Schauder, P.
Clin. Transl. Sci. https://doi.org/10.1111/cts.13257 28, 45–49 (1998). Minimal hepatic encephalopathy impairs fitness to
(2022). 314. Sidhu, S. S. et al. Rifaximin improves psychometric drive. Hepatology 39, 739–745 (2004).
291. Maestri, N. E., Brusilow, S. W., Clissold, D. B. & performance and health-related quality of life 338. Marottoli, R. A. Predictors of automobile crashes
Bassett, S. S. Long-term treatment of girls with in patients with minimal hepatic encephalopathy and moving violations among elderly drivers.
ornithine transcarbamylase deficiency. N. Engl. (The RIME Trial). Am. J. Gastroenterol. 106, 307–316 Ann. Intern. Med. 121, 842 (1994).
J. Med. 335, 855–859 (1996). (2011). 339. Bajaj, J. S., Hafeezullah, M., Hoffmann, R. G.
292. Butterworth, R. F. Ammonia removal by metabolic 315. De Rui, M. et al. Excessive daytime sleepiness & Saeian, K. Minimal hepatic encephalopathy:
scavengers for the prevention and treatment of and hepatic encephalopathy: it is worth asking. a vehicle for accidents and traffic violations.
hepatic encephalopathy in cirrhosis. Drugs R. D. 21, Metab. Brain Dis. 28, 245–248 (2012). Am. J. Gastroenterol. 102, 1903–1909 (2007).
123–132 (2021). 316. Montagnese, S., Middleton, B., Skene, D. J. & 340. Bajaj, J. S., Saeian, K., Hafeezullah, M., Hoffmann, R. G.
293. Rockey, D. C. et al. Randomized, double-blind, Morgan, M. Y. Night-time sleep disturbance does & Hammeke, T. A. Patients with minimal hepatic
controlled study of glycerol phenylbutyrate in hepatic not correlate with neuropsychiatric impairment in encephalopathy have poor insight into their driving
encephalopathy. Hepatology 59, 1073–1083 patients with cirrhosis. Liver Int. 29, 1372–1382 skills. Clin. Gastroenterol. Hepatol. 6, 1135–1139
(2014). (2009). (2008).


NATURE REVIEwS | DISEASE PRIMERS | Article citation ID: (2022) 8:43 21

0123456789();:
Primer

341. Srivastava, A., Mehta, R., Rothke, S. P., Author contributions licensed to Mallinckrodt Pharma. He is also the founder of
Rademaker, A. W. & Blei, A. T. Fitness to Introduction (D.H.); Epidemiology (D.H. and H.V.); Yaqrit Ltd., a spin-out company from University College London,
drive in patients with cirrhosis and portal- Mechanisms/pathophysiology (D.H., B.G., V.F. and A.S.); Cyberliver Ltd. and Hepyx Ltd. S.D.T.-R., B.G., M.R.-G., M.M.,
systemic shunting: a pilot study evaluating Diagnosis, screening and prevention (D.H., S.M., G.K., R.K.D., H.V., A.S. and V.F. declare no competing interests.
driving performance. J. Hepatol. 21, 1023–1028 S.D.T.-R. and M.R.-G.); Management (D.H., H.V., S.M., M.M.
(1994). and R.J.); Quality of life (D.H., H.V., G.K. and R.K.D.); Outlook Peer review information
342. Subasinghe, S. K. C. E. et al. Association (D.H. and R.J.); Overview of Primer (D.H.). Nature Reviews Disease Primers thanks Patrick Kamath,
between road accidents and low-grade hepatic Naveen Nasser-Ghodsi and the other, anonymous, reviewer(s)
encephalopathy among Sri Lankan drivers with Competing interests for their contribution to the peer review of this work.
cirrhosis: a prospective case control study. BMC Res. D.H. and G.K. are members of the “Flicker Diagnostics GbR”.
Notes 9, 303 (2016). The S.M. group has received research funding from AlfaSigma Publisher’s note
343. Bajaj, J. S. et al. Important unresolved questions S.p.a., Norgine Ltd., Merz Pharmaceuticals GmbH, Umecrine Springer Nature remains neutral with regard to jurisdictional
in the management of hepatic encephalopathy: Cognition AB and Versantis AG. R.J. has research collabora- claims in published maps and institutional affiliations.
an ISHEN consensus. Am. J. Gastroenterol. 115, tions with Yaqrit Ltd. and Takeda. R.J. is the inventor of OPA,
989–1002 (2020). which has been patented by University College London and © Springer Nature Limited 2022

22 | Article citation ID: (2022) 8:43 www.nature.com/nrdp

0123456789();:

You might also like