Abstract
Hepatic encephalopathy (HE) is a common, severe complication of advanced chronic liver disease (CLD) and has a devastating impact on the patient’s quality of life and prognosis. The neurotoxin ammonia and the presence of systemic and neurological inflammation are considered the key drivers of this neuropsychiatric syndrome. Treatment options available in routine clinical practice are limited, and the development of novel therapies is hampered owing to the complexity and heterogeneity of HE. This review article aims to outline the current understanding of the pathomechanisms of HE and the recent advances in the identification and development of novel therapeutic targets.
Keywords: Hepatic encephalopathy, hyperammonemia, inflammation, chronic liver disease
Introduction
In patients with advanced liver disease, liver insufficiency and/or portosystemic shunting may lead to the occurrence of a wide range of neuropsychiatric symptoms 1. This brain dysfunction, known as hepatic encephalopathy (HE), marks the end-stage of chronic liver disease (CLD) and has disastrous consequences for the quality of life of patients and their caregivers 2, 3. HE is a common feature in CLD, as it will develop in about 30–40% of patients at some point during the course of the disease. HE can be classified as overt (West Haven grade II–IV, diagnosed based on clinical symptoms ranging from disorientation to coma) or covert (minimal HE or West Haven grade I, diagnosis requiring specialist neuropsychologic testing [ Table 1]) 1. Overt HE development is unpredictable and rapid and often requires admission to the intensive care unit 4, 5. Prognosis of these patients is poor; unless there is access to liver transplantation, 1-year survival generally does not exceed 40% 6, 7. Also, minimal HE (mHE) is associated with a significant impact on quality of life and an increased risk of development of overt HE, hospital admission, and death. Despite advances in the understanding of HE and the development of novel therapies, recent data point out that it is still the leading cause for readmission and mortality in CLD 5.
Table 1. WHC and ISHEN classification (modified according to Vilstrup et al. 1).
WHC grade | ISHEN | Clinical features |
---|---|---|
Unimpaired | No present or previous HE | |
Minimal | Covert | Alterations of psychometric or
neuropsychological tests (i.e. PHES, CFF, EEG) without clinical manifestations |
Grade I | • Trivial lack of awareness
• Euphoria or anxiety • Shortened attention span • Impairment of addition or subtraction • Altered sleep rhythm |
|
Grade II | Overt | • Lethargy or apathy
• Disorientation for time • Obvious personality change • Inappropriate behavior • Dyspraxia • Asterixis |
Grade III | • Somnolence to semi-stupor
• Responsive to stimuli • Confused • Gross disorientation • Bizarre behavior |
|
Grade IV | Coma |
CFF, Critical Flicker Frequency; EEG, electroencephalography; HE, hepatic encephalopathy; ISHEN, International Society for Hepatic Encephalopathy and Nitrogen Metabolism; PHES, Psychometric Hepatic Encephalopathy Score; WHC, West Haven Criteria.
Although it is well established that the neurotoxin ammonia is key in the pathogenesis of HE, the neurochemical changes following ammonia metabolism are numerous and not all yet fully understood. Moreover, distinct pathomechanisms such as alterations in cerebral blood flow (CBF) and, more recently, inflammation have been shown to contribute. The heterogeneity of the clinical presentation as well as the complex and multifactorial pathogenesis of HE have hampered the optimization of its management and the development of effective therapies. This article aims to review the most recent advances in understanding of the disease, with a focus on ammonia and inflammation, and the translation into clinical management. Moreover, the future perspectives of the most promising therapeutic opportunities will be discussed.
Recent advances in understanding the complex pathophysiology
Ammonia: the traditional hypothesis
Of all neurotoxin candidates that have been studied over the last century to explain the neuropsychiatric phenotype in liver disease patients, ammonia has been investigated and discussed most extensively. Ammonia is a nitrogenous compound that is mainly derived from bacterial production and amino acid metabolism in the gut 8– 10. In healthy individuals, ammonia is metabolized in the liver by the urea cycle and glutamine synthetase (GS) and subsequently excreted by the kidneys. The very first link between ammonia and HE dates back almost a century (1922), when a causal relationship between ammonia and meat intoxication was reported in dogs with a portocaval shunt 11. Later (1954), the significance of portal-systemic shunting in cirrhosis in the pathogenesis of HE was reported by Sheila Sherlock et al. 12. They measured peripheral and hepatic venous blood levels of ammonia in CLD patients following oral ammonium chloride intake and showed that ammonia can enter the systemic circulation via the gut by passing through a cirrhotic liver and/or by bypassing it via portal-systemic collaterals. This was therefore defined by the term “portal-systemic HE”. This circulating ammonia is then able to cross the blood–brain barrier (BBB), where it induces a cascade of deleterious effects on the brain 13.
Ammonia: advances in understanding its cerebral effects
Once ammonia reaches the brain, its metabolism mainly relies on glutamine synthesis via GS, which is almost exclusively located in the astrocytes 14, 15. In physiological states, GS is already acting near its maximum rate, thereby efficiently converting ammonia into glutamine. However, in the setting of hyperammonemia, the brain becomes less efficient in ammonia removal because of an insufficient upregulation of GS activity and the absence of an alternative removal pathway 16. Nevertheless, glutamine concentrations are well known to be markedly increased in the brains of animals and patients with HE 16– 18. Inhibition of glutamine breakdown by ammonia has been suggested to be a contributing factor 19. Increased cytosolic glutamine creates an osmotic gradient and thereby contributes to the characteristic morphological changes and mild swelling of the astrocytes in chronic hyperammonemia, known as Alzheimer type II astrocytosis 20. Also, ammonia-related changes in the expression of key astrocytic proteins, such as glial fibrillary acidic protein 21, 22 and peripheral type benzodiazepine receptors 23– 25, contribute to the altered astrocyte morphology and dysfunction.
Besides low-grade brain edema and astrocyte dysfunction, HE in CLD seems to be characterized by a global (ammonia-induced) depression of the central nervous system’s function. This is reflected by 1) a net increase in inhibitory neurotransmission (mainly via impairment of the glutamate neurotransmitter system) 26– 28, 2) reduced CBF 29, 30, 3) reduced oxygen consumption and brain oxygenation 31, 32, and 4) reduced energy metabolism 33, 34. All of these factors seem to be closely interconnected, and imaging studies in cirrhotic patients with chronic hyperammonemia show similar redistribution patterns for CBF and the cerebral metabolic rate for glucose (CMR glucose), characterized by a decrease in the cortical and an increase in certain subcortical areas 35. This corresponds with regions of ammonia-induced suppression of brain metabolism and neurotransmission 36. The link between HE and impaired energy metabolism was first suggested in 1955 37. Thereafter, studies showed that key processes such as glycolysis 38– 40, the tricarboxylic acid cycle (TCA) 41, 42, and the electron transport chain (ETC) 43– 45 are affected by ammonia. An increased rate of glycolysis is a well-characterized phenomenon in HE and hyperammonemia. Although increased glycolysis would be anticipated to increase the operational rate of the TCA cycle, this is not the case in hyperammonemia. Instead of being used in the TCA cycle, pyruvate produced during glycolysis is converted into lactate 40, 46. Diminished availability of pyruvate in hyperammonemia may lead to a decreased operational rate of the TCA cycle, ETC (reduced oxaloacetate generation and availability of NAD/NADH), and ultimately ATP production 47. Reduced brain ATP levels have been reported in experimental models of both acute and chronic hyperammonemia 33, 48– 50. Whether reduced brain ATP levels reflect suppressed synthesis or increased consumption is not yet fully elucidated. More recent studies have been focusing on the role of ammonia-induced mitochondrial dysfunction as an underlying mechanism of impaired energy metabolism in HE. Particularly, the mitochondrial permeability transition (mPT) has been suggested to play a central role. It is characterized by a sudden increase in permeability of the inner mitochondrial membrane to small molecules by opening of the permeability transition pore (PTP) 51, 52. The most important triggers of PTP opening are increased mitochondrial Ca 2+ and (ammonia-/glutamine-induced) production of reactive oxygen species (ROS) 53. This then leads to depolarization of the mitochondrial membrane potential, osmotic swelling of the mitochondrial matrix, uncoupling of the ETC, and thus inhibition of ATP synthesis. Ammonia has been shown to induce an early increase in intracellular Ca 2+ in cultured astrocytes and subsequent induction of mPT 54– 57. Although results of various in vivo and in vitro studies of acute and chronic hyperammonemia support the role of ammonia in disturbed energy metabolism and mitochondrial dysfunction, it must be noted that conflicting results exist 47. This may be partly explained by differences in studied brain regions, cell types, and ammonia concentration and durations. The chronological order of events and interrelationships among changes in neurotransmission, CBF, and oxygen and energy metabolism are yet to be clarified and may guide us to better understand this complex condition and ultimately to develop novel therapeutic strategies.
A recently opened field in the exploration of the pathogenesis of HE is ammonia-induced cellular senescence of astrocytes. It has been described that ammonia can induce senescence via glutamine synthesis-dependent formation of ROS, p53 activation, and upregulation of cell cycle inhibitors (p21 and GADD45a) 58. Another study describes a role for heme oxygenase (HO)-1 in mediating ammonia-induced inhibition of astrocyte proliferation in cultures 59. Although it is currently unknown whether there is a role for astrocyte senescence in the development of cognitive impairment in HE, it seems to have exciting implications for explaining the increasing evidence that cognitive dysfunction does not fully reverse in all patients who experienced an acute episode of HE and may even persist after liver transplantation.
Ammonia: the refined hypothesis
The above-described selection of deleterious effects of ammonia on the brain form the basis of the traditional ammonia hypothesis. This is supported by the fact that ammonia-lowering therapies improve symptoms and outcome in HE, which are therefore the current cornerstones of therapy 1. However, this hypothesis is often criticized, mainly because the clinical value of ammonia measurements is, to date, still unclear, as plasma levels do not always correlate well with severity and outcome 60. This observation suggests that in different clinical situations the effect of ammonia on the brain may well be different. Features in cirrhosis such as inflammation, malnourishment, sodium levels, sarcopenia, co-morbidities, renal dysfunction/failure, and gastrointestinal bleeding (high intestinal protein load) may be some of the contributory factors.
Systemic inflammation in chronic liver disease: role in hepatic encephalopathy
The poor correlation between circulating ammonia levels and HE severity in CLD led to the hypothesis that other mechanisms are involved. Systemic inflammation, commonly referred to as systemic inflammatory response syndrome (SIRS), is a common phenomenon in CLD and can occur in the context of non-sterile (i.e. bacterial infection) as well as sterile inflammation 61. It is characterized by the systemic release of pro-inflammatory cytokines (“cytokine storm”), which may subsequently culminate in severe impairment of systemic hemodynamics and organ hypoperfusion, organ inflammation, cell death, microvascular damage, and eventually (multi-) organ failure. It is well described that sepsis without underlying liver disease can present similarly to HE with altered mental state and motor function, a condition also referred to as “septic encephalopathy” 62. This indicates that a pro-inflammatory state itself can precipitate an encephalopathic state. Previous studies have shown that the vast majority of patients admitted with severe HE indeed present with evidence of systemic inflammation 63. Moreover, patients with CLD are generally immunosuppressed and therefore prone to infections, which are well-recognized precipitants of overt HE 64. The presence of systemic inflammation has been found to significantly impact on mortality risk, and pro-inflammatory markers correlate well with the severity of HE. Also, in patients with mHE, serum levels of pro-inflammatory cytokines are increased (IL-6, IL-18) and correlate with the degree of neurocognitive dysfunction and driving ability 65, 66.
Peripheral inflammation can lead to neuroinflammation via several pathways, of which the humoral (circulating cytokines) and immune (activated immune cells) pathways are the most important 67. Firstly, translocation of Gram-negative bacteria across the intestinal barrier and the release of bacterial products (i.e. pathogen-associated molecular patterns [PAMPs]) play an important role in the development of systemic inflammation in CLD 68. PAMPs, such as lipopolysaccharide (LPS), bind to pattern recognition receptors resulting in the release of pro-inflammatory cytokines. Circulating cytokines can directly enter the brain by impacting on the permeability of the BBB or by binding to receptors of pro-inflammatory cytokines (TNFα, IL1β) expressed by endothelial cells in the BBB 69, 70. Subsequently, this leads to the release of secondary messenger molecules into the brain. These molecules (such as prostaglandins and nitric oxide [NO]) can induce the activation of microglia that can themselves produce inflammatory mediators. Secondly, activated immune cells can similarly bind to endothelial cells in the BBB, thereby inducing the release of secondary messenger molecules and microglia activation. There is a high number of studies providing evidence of microglia activation in the brains of both rodent models and patients with HE. This neuroinflammatory state can lead to changes in neurotransmission, oxidative stress, and neuronal cell death, as shown in both in vitro and in vivo studies 71– 73.
Besides the alterations in intestinal integrity and increased bacterial translocation in CLD, an upcoming field in exploring the role of the gut–brain axis in HE is the gut microbiome. Considering the disrupted intestinal barrier and suppressed immune system in CLD, it is not surprising that dysbiosis of gut microflora can contribute to inducing peripheral inflammation in CLD. Characteristic changes in microbiome associated with HE have been found to correlate with cognitive function and systemic inflammation and involve an abundance of non-autochthonous microorganisms such as Veillonellaceae, Alcaligenaceae, Enterococcus, Megasphaera, Burkholderia, Streptococcus salivarius, Staphylococcaceae, Porphyromonadaceae, and Lactobacillaceae 74– 76. Similar findings have been reported in both stool and salivary microbiota. This suggests that a global gastrointestinal dysbiosis strongly correlates with cognition and inflammation in CLD and therefore holds prognostic and therapeutic potential in HE 77, 78.
Systemic inflammation in chronic liver disease: synergy with ammonia
Increasing evidence points towards the fact that hyperammonemia and systemic inflammation are not two distinct mechanisms driving the severity of HE but that they are working synergistically by making the brain more susceptible to each other’s effects. An elegant clinical study by Shawcross et al. showed for the first time that scores of neuropsychological tests in stable cirrhotic patients are declining when hyperammonemia is induced in an inflammatory state but not after the infection has resolved 79. This synergism was later confirmed in animal models of CLD, showing that the administration of LPS results in hyperammonemia, brain swelling, and coma 80, 81. On the other hand, a reduction in blood ammonia was shown to protect the brain from a subsequent dosing of LPS, suggesting that not only does inflammation make the brain more susceptible to the effects of ammonia but also the reverse is true 82. Furthermore, hyperammonemia itself can directly induce microglia activation and neuroinflammation and appears to have a role in suppression of the immune system 83– 85. Induction of hyperammonemia in rats is associated with impaired neutrophil phagocytic activity leading to ROS production, thereby contributing to systemic inflammation and predisposing infections. Although the precise underlying mechanisms of the synergy between ammonia and inflammation in driving HE severity are not yet fully understood, it may provide essential novel therapeutic targets.
Future perspectives on novel therapeutic targets
Cornerstone pharmacotherapies: targeting the gut
The pharmacotherapies for the treatment of HE traditionally target the gut. The two main treatments in current routine clinical practice are non-absorbable disaccharides (i.e. lactulose) and the poorly absorbed antibiotic rifaximin 1. Lactulose is traditionally the first-line treatment in CLD-related HE. It reduces circulating ammonia levels by different mechanisms: 1) modulation of intestinal flora and therefore reduction in urease-producing bacteria and 2) its laxative effect reduces diffusion of ammonia and nitrogenous compounds into the bloodstream. Lactulose plays an important role as a first-line treatment of HE and in secondary prophylaxis of recurrent overt HE. A recent meta-analysis once again confirmed its beneficial effect regarding HE resolution, development of liver-related complications, and mortality 86, 87. These data, together with this treatment’s low cost and small spectrum of side effects, support the recommendation for lactulose as the initial therapy for HE in CLD. However, as a first-line treatment of an acute HE episode, there is no robust evidence that lactulose improves mortality and outcome. Rifaximin is derived from rifamycin and has a broad spectrum of action against Gram-positive, Gram-negative, aerobic, and anaerobic bacteria. Its mode of action is thought to be via modulation of the gut microbiota, thereby inducing a shift towards a less-pathogenic bacteria population and a reduction in ammonia, endotoxins, and pro-inflammatory cytokines. Addition of rifaximin to lactulose is recommended in patients with recurrent overt HE in CLD despite lactulose prophylaxis 1. In addition, several studies support the effectiveness of rifaximin in the setting of acute HE, but robust data showing improvement of survival in these patients are lacking 88, 89. Along with the globally increasing incidence of liver cirrhosis, the number of hospitalizations for HE has been continuing to grow over the last decade, despite the implementation of novel therapies, such as rifaximin 2, 5. Moreover, long-term antibiotic usage is associated with increased risk of infection with antibiotic-resistant strains of bacteria. Therefore, there is still a significant need for identifying and exploring novel therapeutic targets.
Potential future therapies targeting the gut
Probiotics have been studied in a small number of trials that reported positive results regarding primary prevention of HE, risk of HE-related hospitalization, and the severity of liver disease 90– 92. As for rifaximin, the mechanism of action is thought to be through modulation of the gut microbiome and metabolism. However, it should be noted that probiotics have been reported to be unsafe in some categories of patients, such as those with acute pancreatitis 93. A report released by the World Health Organization (WHO)/Food and Agriculture Organization (FA) ( https://www.who.int/foodsafety/fs_management/en/probiotic_guidelines.pdf) describes four types of potential side effects, namely systemic inflammation, deleterious metabolic activities, excessive immune stimulation in susceptible individuals, and gene transfer. A more recent report from the Agency for Healthcare Research and Quality (AHRQ) reviews the results of 622 studies and concludes that, based on the currently available literature, questions on the safety of probiotics cannot be entirely addressed 94. Another novel, upcoming approach to restore the gut dysbiosis is fecal microbiota transplantation (FMT). In pre-clinical studies, it has been proven that transplantation of fecal microbiota can effectively reduce ammonia levels 95, 96. A small number of clinical trials have shown promising results in terms of the safety/tolerability profile and efficacy (hospitalization, cognition, dysbiosis) of FMT 97, 98. However, a recent case report describes two patients included in two independent clinical trials (one of them had advanced CLD and refractory HE), who developed extended-spectrum beta-lactamase (ESBL)-producing Escherichia coli bacteremia after they had received FMT oral capsules derived from the same stool donor 99. Therefore, well-selected stool transplantation may be an attractive future target for the treatment of HE, but continuous research on the risks and benefits of FMT and the optimization of donor screening is needed.
Targeting ammonia and nitrogen metabolism
L-ornithine L-aspartate (LOLA) is a combination of two non-essential amino acids that can promote ammonia detoxification by acting as a substrate for the urea cycle and by activating GS 100. Intravenous administration has been reported to effectively reduce ammonia and improve mental state in overt HE, whereas the oral formulation seems effective in mHE by improving the outcome of psychometric tests 101. However, a recent Cochrane review states that the quality of evidence on the use of LOLA in HE is poor and further randomized controlled trials are needed 102. Therefore, administration of LOLA is currently restricted to countries in which it is approved for the treatment of HE. A potential risk of LOLA is the so-called “rebound” of ammonia production, as glutamine can be recycled into ammonia by glutaminases 103. The ammonia scavenger ornithine phenylacetate (OP) bypasses this issue by conjugating glutamine with phenylacetate, which forms a water-soluble molecule that can be excreted by the kidneys and hence prevents re-metabolism via glutaminase to glutamate and ammonia. OP was studied up to phase IIb trials and has been shown to safely and effectively reduce ammonia with a dose-related clinical improvement 104– 106. A planned phase III trial needs to confirm these findings.
Nitrogen scavengers that have been investigated in HE include sodium benzoate, glycerol phenylbutyrate, and sodium phenylbutyrate. These agents decrease ammonia by activating conjugation reactions, thereby promoting the elimination of waste nitrogen as amino acid conjugates instead of urea. Benzoate conjugates with glycine to form hippurate and phenylacetate with glutamine to form phenylacetylglutamine, which are both readily excreted by the kidneys 107, 108. Several trials have been showing a beneficial effect of these scavengers on ammonia levels and the risk of overt HE development 109. However, in a recent Cochrane review, it was concluded that the number of available studies is low and the quality of evidence poor at present 109. Furthermore, the high salt load required for sodium benzoate may imply a limited utility in the treatment of HE in CLD. Another approach that has been studied to reduce brain glutamine in HE is the inhibition of GS, for example by L-methionine-S,R-sulfoximine (MSO). Several pre-clinical studies have shown a beneficial effect of MSO on ammonia-induced astrocyte swelling and intracranial hypertension 110– 114. However, the translation of MSO to the clinical situation is hampered by the fact that it can cause convulsions, as observed in animal models 115, 116.
Finally, nutritional supplements, such as branch-chained amino acids (BCAAs) and zinc, have been studied. In the setting of impaired hepatic ammonia metabolism in CLD, skeletal muscle plays an important role in ammonia detoxification through glutamine synthesis by GS 117, 118. In cirrhosis, BCAAs are consumed in skeletal muscle to form α-ketoglutarate, which may be depleted due to enhanced amination to glutamate and subsequent glutamine synthesis 119– 121. Clinical trials assessing BCAA supplementation for the treatment of HE showed mixed results and further studies are required 122– 126. Nevertheless, BCAA supplementation is safe and seems to be useful in preventing the deterioration of liver failure and nutritional status of the cirrhotic patient 127, 128.
Zinc, often deficient in CLD, is considered a cofactor of urea cycle enzymes, and low levels are associated with hyperammonemia and HE 129– 132. The potential benefit of zinc supplementation has been studied in several clinical trials showing largely beneficial effects in HE 133– 135. A recent meta-analysis concluded that a combination of zinc with lactulose over 3–6 months may improve the outcome of psychometric tests in patients with covert HE as compared to lactulose alone 136.
Targeting systemic inflammation
The concept of the beneficial effect of anti-inflammatory agents on HE has initially been shown for ibuprofen (NSAID) and indomethacin (a potent inhibitor of cyclooxygenase-2) in pre-clinical models of HE 83, 137– 139. However, it needs to be considered that these non-steroidal anti-inflammatories are not indicated in the context of CLD because of their deleterious effects on kidney function and impact on risk of gastrointestinal bleeding. Currently, the majority of studies are focusing on reducing the degree of endotoxemia and systemic inflammation by gut-targeting therapies as described above. Also, albumin administration has been studied as a treatment for HE. Besides its property to promote the maintenance of systemic oncotic pressure, albumin is able to scavenge toxins, has anti-oxidant properties, and stabilizes endothelial function 140. However, albumin was found to be ineffective in improving HE severity, ammonia levels, and markers of oxidative stress and inflammation in clinical trials, but it prolonged survival 141, 142. However, although no survival benefit was reported when albumin was applied in extracorporeal liver assist devices (i.e. MARS), it was associated with a decline in HE severity 143. This dialysis system may have its indication in HE as a bridge to transplantation or spontaneous clinical improvement in specialized centers.
A few approaches have been studied to target neuroinflammation directly. N-methyl-D-aspartate (NMDA) receptors are known to play an important role in some types of learning. The function of these receptors is reduced in models of chronic hyperammonemia 144. Activation of these receptors increases calcium in postsynaptic neurons, thereby increasing NO and cyclic guanine monophosphate (cGMP). This so-called glutamate–NO–cGMP pathway plays a key role in inhibiting neuroinflammation and promoting neural cell survival. Sildenafil is a phosphodiesterase inhibitor that inhibits the degradation of cGMP and thereby improves the function of the glutamate–NO–cGMP pathway. Sildenafil has been shown to improve learning abilities in rodent models of minimal HE. In addition, it has been reported to reduce neuroinflammation 145– 147. Targeting the NMDA receptors directly is clinically difficult because in acute hyperammonemia (in contrast to its behavior during chronic hyperammonemia) the NMDA receptors are highly activated and account for ammonia-induced mortality 144. Other studied approaches to reduce neuroinflammation involve reducing microglial activation directly by inhibiting the p38 mitogen-activated protein kinase 148, 149. Agusti et al. showed that inhibiting p38 reduces neuroinflammation (microglial activation and inflammatory markers) and improves cognitive and motor function (learning ability, motor activity, and coordination) in rats with portal systemic shunt-induced minimal HE 150. Current and potential therapies for HE in CLD and their recommended or studied doses are summarized in Table 2.
Table 2. Overview of the discussed (potential) treatment options for HE in CLD and their recommended or studied doses.
Treatment | Drug | Recommended/studied dose |
---|---|---|
Non-absorbable disaccharides | Lactulose | Initial dose 25 mL. Dose titration
to maintain 2–3 loose bowel movements/day. |
Antibiotics | Rifaximin | 550 mg BD orally |
Ammonia/nitrogen scavengers | LOLA | 25–40 g/day (i.v.) |
OP | Up to 20 g/day (i.v.) | |
Sodium benzoate | 10 g/day (oral) | |
Sodium phenylbutyrate | 200 mg/kg/day (oral or via
nasogastric tube) |
|
Glycerol phenylbutyrate | 6 mL twice daily for 16 weeks (oral) | |
Albumin dialysis | MARS | various |
Probiotics | various | various |
FMT | n.a. | various |
BCAAs | n.a. | various (13.2–60 g/day) |
Zinc | n.a. | various (50–600 g/day) |
Selection of experimental
therapies targeting neuroinflammation |
Indomethacin | 0.5 mg/kg i.v. |
Sildenafil | unknown | |
SB239063
(MAP-kinase-p38 inhibitor) |
unknown |
BCAA, branched-chain amino acid; BD, twice daily; FMT, fecal microbiota transplantation; HE, hepatic encephalopathy; i.v., intravenously; LOLA, L-ornithine L-aspartate; OP, ornithine phenylacetate.
Conclusion
HE is a devastating complication of end-stage liver disease with an ever-persisting impact on morbidity, hospitalization, and mortality. Currently available therapies in clinical practice are limited, and the development of novel approaches has been hampered by the complexity and heterogeneity of the syndrome. Traditionally, elevated levels of the neurotoxin ammonia have been considered to be the key driver of “portal-systemic HE”. In the last few decades, the involvement of extrahepatic organs in ammonia metabolism and the role of systemic and neurological inflammation in the pathogenesis of HE have been more and more established and gained their role in clinical management ( Figure 1). The development of novel therapies is currently mainly focusing on scavenging ammonia and modulating the gut microbiome, which are attractive potential treatment approaches. The synergistic relationship between ammonia and inflammation in modulating the severity of HE needs to be further explored, as it might provide us with effective novel therapeutic targets, especially in the setting of an acute bout of HE in CLD.
Abbreviations
BBB, blood–brain barrier; BCAA, branch-chained amino acid; CBF, cerebral blood flow; cGMP, cyclic guanine monophosphate; CLD, chronic liver disease; ETC, electron transport chain; FMT, fecal microbiota transplantation; GS, glutamine synthetase; HE, hepatic encephalopathy; LOLA, L-ornithine L-aspartate; LPS, lipopolysaccharide; mHE, minimal HE; MSO, L-methionine-S,R-sulfoximine; NMDA, N-methyl-D-aspartate; NO, nitric oxide; OP, ornithine phenylacetate; PAMP, pathogen-associated molecular pattern; PTP, permeability transition pore; ROS, reactive oxygen species; TCA, tricarboxylic acid.
Editorial Note on the Review Process
F1000 Faculty Reviews are commissioned from members of the prestigious F1000 Faculty and are edited as a service to readers. In order to make these reviews as comprehensive and accessible as possible, the referees provide input before publication and only the final, revised version is published. The referees who approved the final version are listed with their names and affiliations but without their reports on earlier versions (any comments will already have been addressed in the published version).
The referees who approved this article are:
Marcus-Alexander Wörns, Department of Internal Medicine I, University Medical Centre of the Johannes Gutenberg-University, Mainz, Germany
Christian Labenz, Department of Internal Medicine I, University Medical Centre of the Johannes Gutenberg-University, Mainz, Germany
Christopher Rose, Hepato-Neuro Laboratory, Department of Medicine, Université de Montréal, Montréal, Canada
Funding Statement
The author(s) declared that no grants were involved in supporting this work.
[version 1; peer review: 2 approved]
References
- 1. Vilstrup H, Amodio P, Bajaj J, et al. : Hepatic encephalopathy in chronic liver disease: 2014 Practice Guideline by the American Association for the Study of Liver Diseases and the European Association for the Study of the Liver. Hepatology. 2014;60(2):715–35. 10.1002/hep.27210 [DOI] [PubMed] [Google Scholar]
- 2. Stepanova M, Mishra A, Venkatesan C, et al. : In-hospital mortality and economic burden associated with hepatic encephalopathy in the United States from 2005 to 2009. Clin Gastroenterol Hepatol. 2012;10(9):1034–41.e1. 10.1016/j.cgh.2012.05.016 [DOI] [PubMed] [Google Scholar]
- 3. Poordad FF: Review article: the burden of hepatic encephalopathy. Aliment Pharmacol Ther. 2007;25(Suppl 1):3–9. 10.1111/j.1746-6342.2006.03215.x [DOI] [PubMed] [Google Scholar]
- 4. D'Amico G, Morabito A, Pagliaro L, et al. : Survival and prognostic indicators in compensated and decompensated cirrhosis. Dig Dis Sci. 1986;31(5):468–75. 10.1007/bf01320309 [DOI] [PubMed] [Google Scholar]
- 5. Bajaj JS, Reddy KR, Tandon P, et al. : The 3-month readmission rate remains unacceptably high in a large North American cohort of patients with cirrhosis. Hepatology. 2016;64(1):200–8. 10.1002/hep.28414 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6. Cordoba J, Ventura-Cots M, Simón-Talero M, et al. : Characteristics, risk factors, and mortality of cirrhotic patients hospitalized for hepatic encephalopathy with and without acute-on-chronic liver failure (ACLF). J Hepatol. 2014;60(2):275–81. 10.1016/j.jhep.2013.10.004 [DOI] [PubMed] [Google Scholar]
- 7. Romero-Gómez M, Montagnese S, Jalan R: Hepatic encephalopathy in patients with acute decompensation of cirrhosis and acute-on-chronic liver failure. J Hepatol. 2015;62(2):437–47. 10.1016/j.jhep.2014.09.005 [DOI] [PubMed] [Google Scholar]
- 8. Blei AT, Córdoba J, Practice Parameters Committee of the American College of Gastroenterology: Hepatic Encephalopathy. Am J Gastroenterol. 2001;96(7):1968–76. [DOI] [PubMed] [Google Scholar]
- 9. Albrecht J, Norenberg MD: Glutamine: a Trojan horse in ammonia neurotoxicity. Hepatology. 2006;44(4):788–94. 10.1002/hep.21357 [DOI] [PubMed] [Google Scholar]
- 10. Welbourne T, Weber M, Bank N: The effect of glutamine administration on urinary ammonium excretion in normal subjects and patients with renal disease. J Clin Invest. 1972;51(7):1852–60. 10.1172/JCI106987 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Matthews SA: Ammonia, a causative factor in meat poisoning in Eck fistula dogs. Am J Physiol. 1922;59:459–460. [Google Scholar]
- 12. Sherlock S, Summerskill WH, White L, Phear E: Portal-systemic encephalopathy; neurological complications of liver disease. Lancet. 1954;264(6836):453–7. 10.1016/S0140-6736(54)91874-7 [DOI] [PubMed] [Google Scholar]
- 13. Bosoi CR, Rose CF: Identifying the direct effects of ammonia on the brain. Metab Brain Dis. 2009;24(1):95–102. 10.1007/s11011-008-9112-7 [DOI] [PubMed] [Google Scholar]
- 14. Martinez-Hernandez A, Bell K, Norenberg M: Glutamine synthetase: glial localization in brain. Science. 1977;195(4284):1356–8. 10.1126/science.14400 [DOI] [PubMed] [Google Scholar]
- 15. Norenberg MD, Martinez-Hernandez A: Fine structural localization of glutamine synthetase in astrocytes of rat brain. Brain Res. 1979;161(2):303–10. 10.1016/0006-8993(79)90071-4 [DOI] [PubMed] [Google Scholar]
- 16. Cooper AJ, Mora SN, Cruz NF, et al. : Cerebral ammonia metabolism in hyperammonemic rats. J Neurochem. 1985;44(6):1716–23. 10.1111/j.1471-4159.1985.tb07159.x [DOI] [PubMed] [Google Scholar]
- 17. Lavoie J, Giguère JF, Layrargues GP, et al. : Amino acid changes in autopsied brain tissue from cirrhotic patients with hepatic encephalopathy. J Neurochem. 1987;49(3):692–7. 10.1111/j.1471-4159.1987.tb00949.x [DOI] [PubMed] [Google Scholar]
- 18. Cooper AJ, Plum F: Biochemistry and physiology of brain ammonia. Physiol Rev. 1987;67(2):440–519. 10.1152/physrev.1987.67.2.440 [DOI] [PubMed] [Google Scholar]
- 19. Matheson DF, van den Berg CJ: Ammonia and brain glutamine: inhibition of glutamine degradation by ammonia. Biochem Soc Trans. 1975;3(4):525–8. 10.1042/bst0030525 [DOI] [PubMed] [Google Scholar]
- 20. Norenberg MD: The astrocyte in liver disease. Adv Cell Neurobiol. 1981;2:303–352. 10.1016/B978-0-12-008302-2.50013-4 4109954 [DOI] [Google Scholar]
- 21. Bélanger M, Desjardins P, Chatauret N, et al. : Loss of expression of glial fibrillary acidic protein in acute hyperammonemia. Neurochem Int. 2002;41(2–3):155–60. 10.1016/s0197-0186(02)00037-2 [DOI] [PubMed] [Google Scholar]
- 22. Neary JT, Whittemore SR, Zhu Q, et al. : Destabilization of glial fibrillary acidic protein mRNA in astrocytes by ammonia and protection by extracellular ATP. J Neurochem. 1994;63(6):2021–7. 10.1046/j.1471-4159.1994.63062021.x [DOI] [PubMed] [Google Scholar]
- 23. Desjardins P, Bandeira P, Raghavendra Rao VL, et al. : Increased expression of the peripheral-type benzodiazepine receptor-isoquinoline carboxamide binding protein mRNA in brain following portacaval anastomosis. Brain Res. 1997;758(1–2):255–8. 10.1016/s0006-8993(97)00339-9 [DOI] [PubMed] [Google Scholar]
- 24. Giguère J-F, Hamel E, Butterworth RF: Increased densities of binding sites for the ‘peripheral-type’ benzodiazepine receptor ligand [ 3H]PK 11195 in rat brain following portacaval anastomosis. Brain Res. 1992;585(1–2):295–8. 10.1016/0006-8993(92)91222-z [DOI] [PubMed] [Google Scholar]
- 25. Lavoie J, Layrargues GP, Butterworth RF: Increased densities of peripheral-type benzodiazepine receptors in brain autopsy samples from cirrhotic patients with hepatic encephalopathy. Hepatology. 1990;11(5):874–8. 10.1002/hep.1840110524 [DOI] [PubMed] [Google Scholar]
- 26. Théorĕt Y, Bossu JL: Effects of ammonium salts on synaptic transmission to hippocampal CA1 and CA3 pyramidal cells in vivo. Neuroscience. 1985;14(3):807–21. 10.1016/0306-4522(85)90145-9 [DOI] [PubMed] [Google Scholar]
- 27. Rao VL, Agrawal AK, Murthy CR: Ammonia-induced alterations in glutamate and muscimol binding to cerebellar synaptic membranes. Neurosci Lett. 1991;130(2):251–4. 10.1016/0304-3940(91)90408-l [DOI] [PubMed] [Google Scholar]
- 28. Hermenegildo C, Montoliu C, Llansola M, et al. : Chronic hyperammonemia impairs the glutamate-nitric oxide-cyclic GMP pathway in cerebellar neurons in culture and in the rat in vivo. Eur J Neurosci. 1998;10(10):3201–9. 10.1046/j.1460-9568.1998.00329.x [DOI] [PubMed] [Google Scholar]
- 29. Posner JB, Plum F: The toxic effects of carbon dioxide and acetazolamide in hepatic encephalopathy. J Clin Invest. 1960;39:1246–58. 10.1172/JCI104140 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30. James IM, Garassini M: Effect of lactulose on cerebral metabolism in patients with chronic portosystemic encephalopathy. Gut. 1971;12(9):702–4. 10.1136/gut.12.9.702 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31. Iversen P, Sørensen M, Bak LK, et al. : Low cerebral oxygen consumption and blood flow in patients with cirrhosis and an acute episode of hepatic encephalopathy. Gastroenterology. 2009;136(3):863–71. 10.1053/j.gastro.2008.10.057 [DOI] [PubMed] [Google Scholar]
- 32. Dam G, Keiding S, Munk OL, et al. : Hepatic encephalopathy is associated with decreased cerebral oxygen metabolism and blood flow, not increased ammonia uptake. Hepatology. 2013;57(1):258–65. 10.1002/hep.25995 [DOI] [PubMed] [Google Scholar]
- 33. McCandless DW, Schenker S: Effect of acute ammonia intoxication on energy stores in the cerebral reticular activating system. Exp Brain Res. 1981;44(3):325–30. 10.1007/bf00236570 [DOI] [PubMed] [Google Scholar]
- 34. Hawkins RA, Miller AL, Nielsen RC, et al. : The acute action of ammonia on rat brain metabolism in vivo. Biochem J. 1973;134(4):1001–8. 10.1042/bj1341001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35. Lockwood AH, Murphy BW, Donnelly KZ, et al. : Positron-emission tomographic localization of abnormalities of brain metabolism in patients with minimal hepatic encephalopathy. Hepatology. 1993;18(5):1061–8. 10.1002/hep.1840180508 [DOI] [PubMed] [Google Scholar]
- 36. Felipo V, Butterworth RF: Neurobiology of ammonia. Prog Neurobiol. 2002;67(4):259–79. 10.1016/s0301-0082(02)00019-9 [DOI] [PubMed] [Google Scholar]
- 37. Bessman SP, Bessman AN: The cerebral and peripheral uptake of ammonia in liver disease with an hypothesis for the mechanism of hepatic coma. J Clin Invest. 1955;34(4):622–8. 10.1172/JCI103111 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. Muntz JA, Hurwitz J: Effect of potassium and ammonium ions upon glycolysis catalyzed by an extract of rat brain. Arch Biochem Biophys. 1951;32(1):124–36. 10.1016/0003-9861(51)90246-9 [DOI] [PubMed] [Google Scholar]
- 39. Abrahams SL, Younathan ES: Modulation of the kinetic properties of phosphofructokinase by ammonium ions. J Biol Chem. 1971;246(8):2464–7. [PubMed] [Google Scholar]
- 40. Ratnakumari L, Qureshi IA, Butterworth RF: Effects of congenital hyperammonemia on the cerebral and hepatic levels of the intermediates of energy metabolism in spf mice. Biochem Biophys Res Commun. 1992;184(2):746–51. 10.1016/0006-291x(92)90653-3 [DOI] [PubMed] [Google Scholar]
- 41. Shorey J, McCandless DW, Schenker S: Cerebral alpha-ketoglutarate in ammonia intoxication. Gastroenterology. 1967;53(5):706–11. 10.1016/S0016-5085(19)34154-X [DOI] [PubMed] [Google Scholar]
- 42. Lai JC, Cooper AJ: Neurotoxicity of ammonia and fatty acids: differential inhibition of mitochondrial dehydrogenases by ammonia and fatty acyl coenzyme A derivatives. Neurochem Res. 1991;16(7):795–803. 10.1007/bf00965689 [DOI] [PubMed] [Google Scholar]
- 43. Walshe JM, de Carli L, Davidson CS: Some factors influencing cerebral ammonia production in relation to hepatic coma. Clin Sci. 1958;17(1):27–36. [PubMed] [Google Scholar]
- 44. McKhann GM, Tower DB: Ammonia toxicity and cerebral oxidative metabolism. Am J Physiol. 1961;200:420–4. 10.1152/ajplegacy.1961.200.3.420 [DOI] [PubMed] [Google Scholar]
- 45. Kosenko E, Felipo V, Montoliu C, et al. : Effects of acute hyperammonemia in vivo on oxidative metabolism in nonsynaptic rat brain mitochondria. Metab Brain Dis. 1997;12(1):69–82. 10.1007/bf02676355 [DOI] [PubMed] [Google Scholar]
- 46. Kala G, Hertz L: Ammonia effects on pyruvate/lactate production in astrocytes--interaction with glutamate. Neurochem Int. 2005;47(4–12):4–12. 10.1016/j.neuint.2005.04.001 [DOI] [PubMed] [Google Scholar]
- 47. Rama Rao KV, Norenberg MD: Brain energy metabolism and mitochondrial dysfunction in acute and chronic hepatic encephalopathy. Neurochem Int. 2012;60(7):697–706. 10.1016/j.neuint.2011.09.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48. Hindfelt B, Plum F, Duffy TE: Effect of acute ammonia intoxication on cerebral metabolism in rats with portacaval shunts. J Clin Invest. 1977;59(3):386–96. 10.1172/JCI108651 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Astore D, Boicelli CA: Hyperammonemia and chronic hepatic encephalopathy: an in vivo PMRS study of the rat brain. MAGMA. 2000;10(3):160–6. 10.1007/bf02590641 [DOI] [PubMed] [Google Scholar]
- 50. Rao KV, Mawal YR, Qureshi IA: Progressive decrease of cerebral cytochrome C oxidase activity in sparse-fur mice: role of acetyl- L-carnitine in restoring the ammonia-induced cerebral energy depletion. Neurosci Lett. 1997;224(2):83–6. 10.1016/s0304-3940(97)13476-0 [DOI] [PubMed] [Google Scholar]
- 51. Bernardi P, Colonna R, Costantini P, et al. : The mitochondrial permeability transition. Biofactors. 1998;8(3–4):273–81. 10.1002/biof.5520080315 [DOI] [PubMed] [Google Scholar]
- 52. Norenberg M, Rao K: The mitochondrial permeability transition in neurologic disease. Neurochem Int. 2007;50(7–8):983–97. 10.1016/j.neuint.2007.02.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Kristal BS, Dubinsky JM: Mitochondrial Permeability Transition in the Central Nervous System: Induction by Calcium Cycling-Dependent and -Independent Pathways. J Neurochem. 1997;69(2):524–38. 10.1046/j.1471-4159.1997.69020524.x. [DOI] [PubMed] [Google Scholar]
- 54. Norenberg MD, Rama Rao KV, Jayakumar AR: The mitochondrial permeability transition in ammonia neurotoxicity.In: Jones E, Meijer AJ, Chamuleau AF, editors. Encephalopathy and Nitrogen Metabolism in Liver Failure Dordrecht: Kluwer;2003;267–286. Reference Source [Google Scholar]
- 55. Rose C: Effect of ammonia on astrocytic glutamate uptake/release mechanisms. J Neurochem. 2006;97(Suppl 1):11–5. 10.1111/j.1471-4159.2006.03796.x [DOI] [PubMed] [Google Scholar]
- 56. Jayakumar AR, Rama Rao KV, Tong XY, et al. : Calcium in the mechanism of ammonia-induced astrocyte swelling. J Neurochem. 2009;109(Suppl 1):252–7. 10.1111/j.1471-4159.2009.05842.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 57. Bai G, Rama Rao KV, Murthy CR, et al. : Ammonia induces the mitochondrial permeability transition in primary cultures of rat astrocytes. J Neurosci Res. 2001;66(5):981–91. 10.1002/jnr.10056 [DOI] [PubMed] [Google Scholar]
- 58. Görg B, Karababa A, Shafigullina A, et al. : Ammonia-induced senescence in cultured rat astrocytes and in human cerebral cortex in hepatic encephalopathy. Glia. 2015;63(1):37–50. 10.1002/glia.22731 [DOI] [PubMed] [Google Scholar]
- 59. Görg B, Karababa A, Schütz E, et al. : O-GlcNAcylation-dependent upregulation of HO1 triggers ammonia-induced oxidative stress and senescence in hepatic encephalopathy. J Hepatol. 2019;71(5):930–41. 10.1016/j.jhep.2019.06.020 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
- 60. Lockwood AH: Blood ammonia levels and hepatic encephalopathy. Metab Brain Dis. 2004;19(3–4):345–9. 10.1023/b:mebr.0000043980.74574.eb [DOI] [PubMed] [Google Scholar]
- 61. American College of Chest Physicians/Society of Critical Care Medicine Consensus Conference: definitions for sepsis and organ failure and guidelines for the use of innovative therapies in sepsis. Crit Care Med. 1992;20(6):864–74. 10.1097/00003246-199206000-00025 [DOI] [PubMed] [Google Scholar]
- 62. Papadopoulos MC, Davies DC, Moss RF, et al. : Pathophysiology of septic encephalopathy: a review. Crit Care Med. 2000;28(8):3019–24. 10.1097/00003246-200008000-00057 [DOI] [PubMed] [Google Scholar]
- 63. Shawcross DL, Sharifi Y, Canavan JB, et al. : Infection and systemic inflammation, not ammonia, are associated with Grade 3/4 hepatic encephalopathy, but not mortality in cirrhosis. J Hepatol. 2011;54(4):640–9. 10.1016/j.jhep.2010.07.045 [DOI] [PubMed] [Google Scholar]
- 64. Irvine KM, Ratnasekera I, Powell EE, et al. : Causes and Consequences of Innate Immune Dysfunction in Cirrhosis. Front Immunol. 2019;10:293. 10.3389/fimmu.2019.00293 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
- 65. Wein C, Koch H, Popp B, et al. : Minimal hepatic encephalopathy impairs fitness to drive. Hepatology. 2004;39(3):739–45. 10.1002/hep.20095 [DOI] [PubMed] [Google Scholar]
- 66. Felipo V, Urios A, Valero P, et al. : Serum nitrotyrosine and psychometric tests as indicators of impaired fitness to drive in cirrhotic patients with minimal hepatic encephalopathy. Liver Int. 2013;33(10):1478–89. 10.1111/liv.12206 [DOI] [PubMed] [Google Scholar]
- 67. Azhari H, Swain MG: Role of Peripheral Inflammation in Hepatic Encephalopathy. J Clin Exp Hepatol. 2018;8(3):281–285. 10.1016/j.jceh.2018.06.008 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
- 68. Bernardi M, Moreau R, Angeli P, et al. : Mechanisms of decompensation and organ failure in cirrhosis: From peripheral arterial vasodilation to systemic inflammation hypothesis. J Hepatol. 2015;63(5):1272–84. 10.1016/j.jhep.2015.07.004 [DOI] [PubMed] [Google Scholar]
- 69. D'Mello C, Swain MG: Immune-to-Brain Communication Pathways in Inflammation-Associated Sickness and Depression. Curr Top Behav Neurosci. 2017;31:73–94. 10.1007/7854_2016_37 [DOI] [PubMed] [Google Scholar]
- 70. Capuron L, Miller AH: Immune system to brain signaling: neuropsychopharmacological implications. Pharmacol Ther. 2011;130(2):226–38. 10.1016/j.pharmthera.2011.01.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71. DiSabato DJ, Quan N, Godbout JP: Neuroinflammation: the devil is in the details. J Neurochem. 2016;139(Suppl 2):136–53. 10.1111/jnc.13607 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72. Glass CK, Saijo K, Winner B, et al. : Mechanisms underlying inflammation in neurodegeneration. Cell. 2010;140(6):918–34. 10.1016/j.cell.2010.02.016 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73. McCoy MK, Tansey MG: TNF signaling inhibition in the CNS: implications for normal brain function and neurodegenerative disease. J Neuroinflammation. 2008;5:45. 10.1186/1742-2094-5-45 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Bajaj JS, Heuman DM, Hylemon PB, et al. : Altered profile of human gut microbiome is associated with cirrhosis and its complications. J Hepatol. 2014;60(5):940–7. 10.1016/j.jhep.2013.12.019 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Chen Y, Yang F, Lu H, et al. : Characterization of fecal microbial communities in patients with liver cirrhosis. Hepatology. 2011;54(2):562–72. 10.1002/hep.24423 [DOI] [PubMed] [Google Scholar]
- 76. Bajaj JS, Ridlon JM, Hylemon PB, et al. : Linkage of gut microbiome with cognition in hepatic encephalopathy. Am J Physiol Gastrointest Liver Physiol. 2012;302(1):G168–75. 10.1152/ajpgi.00190.2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Bajaj JS, Betrapally NS, Hylemon PB, et al. : Salivary microbiota reflects changes in gut microbiota in cirrhosis with hepatic encephalopathy. Hepatology. 2015;62(4):1260–71. 10.1002/hep.27819 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78. Bajaj JS, Fagan A, White MB, et al. : Specific Gut and Salivary Microbiota Patterns Are Linked With Different Cognitive Testing Strategies in Minimal Hepatic Encephalopathy. Am J Gastroenterol. 2019;114(7):1080–90. 10.14309/ajg.0000000000000102 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
- 79. Shawcross DL, Davies NA, Williams R, et al. : Systemic inflammatory response exacerbates the neuropsychological effects of induced hyperammonemia in cirrhosis. J Hepatol. 2004;40(2):247–54. 10.1016/j.jhep.2003.10.016 [DOI] [PubMed] [Google Scholar]
- 80. Wright G, Davies NA, Shawcross DL, et al. : Endotoxemia produces coma and brain swelling in bile duct ligated rats. Hepatology. 2007;45(6):1517–26. 10.1002/hep.21599 [DOI] [PubMed] [Google Scholar]
- 81. Pedersen HR, Ring-Larsen H, Olsen NV, et al. : Hyperammonemia acts synergistically with lipopolysaccharide in inducing changes in cerebral hemodynamics in rats anaesthetised with pentobarbital. J Hepatol. 2007;47(2):245–52. 10.1016/j.jhep.2007.03.026 [DOI] [PubMed] [Google Scholar]
- 82. Marini JC, Broussard SR: Hyperammonemia increases sensitivity to LPS. Mol Genet Metab. 2006;88(2):131–7. 10.1016/j.ymgme.2005.12.013 [DOI] [PubMed] [Google Scholar]
- 83. Rodrigo R, Cauli O, Gomez-Pinedo U, et al. : Hyperammonemia induces neuroinflammation that contributes to cognitive impairment in rats with hepatic encephalopathy. Gastroenterology. 2010;139(2):675–84. 10.1053/j.gastro.2010.03.040 [DOI] [PubMed] [Google Scholar]
- 84. Bezzi P, Domercq M, Vesce S, et al. : Neuron-astrocyte cross-talk during synaptic transmission: Physiological and neuropathological implications. Prog Brain Res. 2001;132:255–65. 10.1016/S0079-6123(01)32081-2 [DOI] [PubMed] [Google Scholar]
- 85. Shawcross DL, Wright GA, Stadlbauer V, et al. : Ammonia impairs neutrophil phagocytic function in liver disease. Hepatology. 2008;48(4):1202–12. 10.1002/hep.22474 [DOI] [PubMed] [Google Scholar]
- 86. Als-Nielsen B, Gluud LL, Gluud C: Non-absorbable disaccharides for hepatic encephalopathy: Systematic review of randomised trials. BMJ. 2004;328(7447):1046. 10.1136/bmj.38048.506134.EE [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87. Gluud LL, Vilstrup H, Morgan MY: Nonabsorbable disaccharides for hepatic encephalopathy: A systematic review and meta-analysis. Hepatology. 2016;64(3):908–22. 10.1002/hep.28598 [DOI] [PubMed] [Google Scholar]
- 88. Courson A, Jones GM, Twilla JD: Treatment of Acute Hepatic Encephalopathy: Comparing the Effects of Adding Rifaximin to Lactulose on Patient Outcomes. J Pharm Pract. 2016;29(3):212–7. 10.1177/0897190014566312 [DOI] [PubMed] [Google Scholar]
- 89. Kang SH, Lee YB, Lee JH, et al. : Rifaximin treatment is associated with reduced risk of cirrhotic complications and prolonged overall survival in patients experiencing hepatic encephalopathy. Aliment Pharmacol Ther. 2017;46(9):845–855. 10.1111/apt.14275 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
- 90. Liu Q, Duan ZP, Ha DK, et al. : Synbiotic modulation of gut flora: Effect on minimal hepatic encephalopathy in patients with cirrhosis. Hepatology. 2004;39(5):1441–9. 10.1002/hep.20194 [DOI] [PubMed] [Google Scholar]
- 91. Viramontes Hörner D, Avery A, Stow R: The Effects of Probiotics and Symbiotics on Risk Factors for Hepatic Encephalopathy: A Systematic Review. J Clin Gastroenterol. 2017;51(4):312–23. 10.1097/MCG.0000000000000789 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
- 92. Marlicz W, Wunsch E, Mydlowska M, et al. : The Effect of Short Term Treatment With Probiotic VSL#3 on Various Clinical and Biochemical Parameters in Patients With Liver Cirrhosis. J Physiol Pharmacol. 2016;67(6):867–77. [PubMed] [Google Scholar]
- 93. Doron S, Snydman DR: Risk and Safety of Probiotics. Clin Infect Dis. 2015;60 Suppl 2:S129–S134. 10.1093/cid/civ085 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94. Hempel S, Newberry S, Ruelaz A, et al. : Safety of Probiotics Used to Reduce Risk and Prevent or Treat Disease. Evid Rep Technol Assess (Full Rep). 2011; (200):1–645. [PMC free article] [PubMed] [Google Scholar]
- 95. Shen TCD, Albenberg L, Bittinger K, et al. : Engineering the Gut Microbiota to Treat Hyperammonemia. J Clin Invest. 2015;125(7):2841–50. 10.1172/JCI79214 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Wang WW, Zhang Y, Huang XB, et al. : Fecal Microbiota Transplantation Prevents Hepatic Encephalopathy in Rats With Carbon Tetrachloride-Induced Acute Hepatic Dysfunction. World J Gastroenterol. 2017;23(38):6983–94. 10.3748/wjg.v23.i38.6983 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
- 97. Bajaj JS, Kassam Z, Fagan A, et al. : Fecal Microbiota Transplant From a Rational Stool Donor Improves Hepatic Encephalopathy: A Randomized Clinical Trial. Hepatology. 2017;66:1727–38. 10.1002/hep.29306 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
- 98. Bajaj JS, Fagan A, Gavis EA, et al. : Long-term Outcomes of Fecal Microbiota Transplantation in Patients With Cirrhosis. Gastroenterology. 2019;156(6):1921–1923.e3. 10.1053/j.gastro.2019.01.033 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
- 99. DeFilipp Z, Bloom PP, Torres Soto M, et al. : Drug-Resistant E. coli Bacteremia Transmitted by Fecal Microbiota Transplant. N Engl J Med. 2019;381(21):2043–50. 10.1056/NEJMoa1910437 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
- 100. Kircheis G, Lüth S: Pharmacokinetic and Pharmacodynamic Properties of L-Ornithine L-Aspartate (LOLA) in Hepatic Encephalopathy. Drugs. 2019;79(Suppl 1):23–9. 10.1007/s40265-018-1023-2 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
- 101. Bai M, Yang Z, Qi X, et al. : l-ornithine-l-aspartate for hepatic encephalopathy in patients with cirrhosis: A meta-analysis of randomized controlled trials. J Gastroenterol Hepatol. 2013;28(5):783–92. 10.1111/jgh.12142 [DOI] [PubMed] [Google Scholar]
- 102. Goh ET, Stokes CS, Sidhu SS, et al. : L-ornithine L-aspartate for prevention and treatment of hepatic encephalopathy in people with cirrhosis. Cochrane Database Syst Rev. 2018;5:CD012410. 10.1002/14651858.CD012410.pub2 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
- 103. Bai M, He C, Yin Z, et al. : Randomised clinical trial: L-ornithine-l-aspartate reduces significantly the increase of venous ammonia concentration after TIPSS. Aliment Pharmacol Ther. 2014;40(1):63–71. 10.1111/apt.12795 [DOI] [PubMed] [Google Scholar]
- 104. Stravitz RT, Gottfried M, Durkalski V, et al. : Safety, tolerability, and pharmacokinetics of l-ornithine phenylacetate in patients with acute liver injury/failure and hyperammonemia. Hepatology. 2018;67(3):1003–1013. 10.1002/hep.29621 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
- 105. Ventura-Cots M, Concepción M, Arranz JA, et al. : Impact of ornithine phenylacetate (OCR-002) in lowering plasma ammonia after upper gastrointestinal bleeding in cirrhotic patients. Therap Adv Gastroenterol. 2016;9(6):823–835. 10.1177/1756283X16658252 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Ventura-Cots M, Arranz JA, Simón-Talero M, et al. : Safety of ornithine phenylacetate in cirrhotic decompensated patients: an open-label, dose-escalating, single-cohort study. J Clin Gastroenterol. 2013;47(10):881–7. 10.1097/MCG.0b013e318299c789 [DOI] [PubMed] [Google Scholar]
- 107. Batshaw ML, Brusilow S, Waber L, et al. : Treatment of inborn errors of urea synthesis: Activation of alternative pathways of waste nitrogen synthesis and excretion. N Engl J Med. 1982;306(23):1387–92. 10.1056/NEJM198206103062303 [DOI] [PubMed] [Google Scholar]
- 108. Brusilow S, Tinker J, Batshaw ML: Amino acid acylation: A mechanism of nitrogen excretion in inborn errors of urea synthesis. Science. 1980;207(4431):659–61. 10.1126/science.6243418 [DOI] [PubMed] [Google Scholar]
- 109. Zacharias HD, Zacharias AP, Gluud LL, et al. : Pharmacotherapies that specifically target ammonia for the prevention and treatment of hepatic encephalopathy in adults with cirrhosis. Cochrane Database Syst Rev. 2019;6:CD012334. 10.1002/14651858.CD012334.pub2 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
- 110. Warren KS, Schenker S: Effect of an inhibitor of glutamine synthesis (methionine sulfoximine) on ammonia toxicity and metabolism. J Lab Clin Med. 1964;64:442–9. [PubMed] [Google Scholar]
- 111. Takahashi H, Koehler RC, Brusilow SW, et al. : Inhibition of brain glutamine accumulation prevents cerebral edema in hyperammonemic rats. Am J Physiol. 1991;261(3 Pt 2):H825–9. 10.1152/ajpheart.1991.261.3.H825 [DOI] [PubMed] [Google Scholar]
- 112. Blei AT, Olafsson S, Therrien G, et al. : Ammonia-induced brain edema and intracranial hypertension in rats after portacaval anastomosis. Hepatology. 1994;19(6):1437–44. 10.1002/hep.1840190619 [DOI] [PubMed] [Google Scholar]
- 113. Willard-Mack CL, Koehler RC, Hirata T, et al. : Inhibition of glutamine synthetase reduces ammonia-induced astrocyte swelling in rat. Neuroscience. 1996;71(2):589–99. 10.1016/0306-4522(95)00462-9 [DOI] [PubMed] [Google Scholar]
- 114. Tanigami H, Rebel A, Martin LJ, et al. : Effect of glutamine synthetase inhibition on astrocyte swelling and altered astroglial protein expression during hyperammonemia in rats. Neuroscience. 2005;131(2):437–49. 10.1016/j.neuroscience.2004.10.045 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 115. Brumback RA, Lapham LW: DNA synthesis in Alzheimer type II astrocytosis. The question of astrocytic proliferation and mitosis in experimentally induced hepatic encephalopathy. Arch Neurol. 1989;46(8):845–8. 10.1001/archneur.1989.00520440027016 [DOI] [PubMed] [Google Scholar]
- 116. Gershoff SN, Elvehjem CA: The relative effect of methionine sulfoximine on different animal species. J Nutr. 1951;45(3):451–8. 10.1093/jn/45.3.451 [DOI] [PubMed] [Google Scholar]
- 117. Olde Damink SW, Deutz NE, Dejong CH, et al. : Interorgan ammonia metabolism in liver failure. Neurochem Int. 2002;41(2–3):177–88. 10.1016/s0197-0186(02)00040-2 [DOI] [PubMed] [Google Scholar]
- 118. Ganda OP, Ruderman NB: Muscle nitrogen metabolism in chronic hepatic insufficiency. Metab Clin Exp. 1976;25(4):427–35. 10.1016/0026-0495(76)90075-5 [DOI] [PubMed] [Google Scholar]
- 119. Uribe M, Márquez MA, Garcia Ramos G, et al. : Treatment of chronic portal--systemic encephalopathy with vegetable and animal protein diets. A controlled crossover study. Dig Dis Sci. 1982;27(12):1109–16. 10.1007/BF01391449 [DOI] [PubMed] [Google Scholar]
- 120. Holecek M: Evidence of a vicious cycle in glutamine synthesis and breakdown in pathogenesis of hepatic encephalopathy-therapeutic perspectives. Metab Brain Dis. 2014;29(1):9–17. 10.1007/s11011-013-9428-9 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121. Holecek M, Kandar R, Sispera L, et al. : Acute hyperammonemia activates branched-chain amino acid catabolism and decreases their extracellular concentrations: different sensitivity of red and white muscle. Amino Acids. 2011;40(2):575–84. 10.1007/s00726-010-0679-z [DOI] [PubMed] [Google Scholar]
- 122. Amodio P, Bemeur C, Butterworth R, et al. : The nutritional management of hepatic encephalopathy in patients with cirrhosis: International Society for Hepatic Encephalopathy and Nitrogen Metabolism Consensus. Hepatology. 2013;58(1):325–36. 10.1002/hep.26370 [DOI] [PubMed] [Google Scholar]
- 123. Als-Nielsen B, Koretz RL, Kjaergard LL, et al. : Branched-chain amino acids for hepatic encephalopathy. Cochrane Database Syst Rev. 2003; (2):CD001939. 10.1002/14651858.CD001939 [DOI] [PubMed] [Google Scholar]
- 124. de Bruijn KM, Blendis LM, Zilm DH, et al. : Effect of dietary protein manipulation in subclinical portal-systemic encephalopathy. Gut. 1983;24(1):53–60. 10.1136/gut.24.1.53 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Horst D, Grace ND, Conn HO, et al. : Comparison of dietary protein with an oral, branched chain-enriched amino acid supplement in chronic portal-systemic encephalopathy: a randomized controlled trial. Hepatology. 1984;4(2):279–87. 10.1002/hep.1840040218 [DOI] [PubMed] [Google Scholar]
- 126. Les I, Doval E, García-Martínez R, et al. : Effects of branched-chain amino acids supplementation in patients with cirrhosis and a previous episode of hepatic encephalopathy: a randomized study. Am J Gastroenterol. 2011;106(6):1081–8. 10.1038/ajg.2011.9 [DOI] [PubMed] [Google Scholar]
- 127. Marchesini G, Bianchi G, Merli M, et al. : Nutritional supplementation with branched-chain amino acids in advanced cirrhosis: a double-blind, randomized trial. Gastroenterology. 2003;124(7):1792–801. 10.1016/s0016-5085(03)00323-8 [DOI] [PubMed] [Google Scholar]
- 128. Muto Y, Sato S, Watanabe A, et al. : Effects of oral branched-chain amino acid granules on event-free survival in patients with liver cirrhosis. Clin Gastroenterol Hepatol. 2005;3(7):705–13. 10.1016/s1542-3565(05)00017-0 [DOI] [PubMed] [Google Scholar]
- 129. Loomba V, Pawar G, Dhar KL, et al. : Serum zinc levels in hepatic encephalopathy. Indian J Gastroenterol. 1995;14(2):51–3. [PubMed] [Google Scholar]
- 130. Bresci G, Parisi G, Banti S: Management of hepatic encephalopathy with oral zinc supplementation: a long-term treatment. Eur J Med. 1993;2(7):414–6. [PubMed] [Google Scholar]
- 131. Reding P, Duchateau J, Bataille C: Oral zinc supplementation improves hepatic encephalopathy. Results of a randomised controlled trial. Lancet. 1984;324(8401):493–5. 10.1016/s0140-6736(84)92567-4 [DOI] [PubMed] [Google Scholar]
- 132. Riggio O, Ariosto F, Merli M, et al. : Short-term oral zinc supplementation does not improve chronic hepatic encephalopathy. Results of a double-blind crossover trial. Dig Dis Sci. 1991;36(9):1204–8. 10.1007/bf01307509 [DOI] [PubMed] [Google Scholar]
- 133. Van der Rijt CC, Schalm SW, Schat H, et al. : Overt hepatic encephalopathy precipitated by zinc deficiency. Gastroenterology. 1991;100(4):1114–8. 10.1016/0016-5085(91)90290-2 [DOI] [PubMed] [Google Scholar]
- 134. Marchesini G, Fabbri A, Bianchi G, et al. : Zinc supplementation and amino acid-nitrogen metabolism in patients with advanced cirrhosis. Hepatology. 1996;23(5):1084–92. 10.1053/jhep.1996.v23.pm0008621138 [DOI] [PubMed] [Google Scholar]
- 135. Takuma Y, Nouso K, Makino Y, et al. : Clinical trial: oral zinc in hepatic encephalopathy. Aliment Pharmacol Ther. 2010;32(9):1080–90. 10.1111/j.1365-2036.2010.04448.x [DOI] [PubMed] [Google Scholar]
- 136. Shen YC, Chang YH, Fang CJ, et al. : Zinc supplementation in patients with cirrhosis and hepatic encephalopathy: a systematic review and meta-analysis. Nutr J. 2019;18(1):34. 10.1186/s12937-019-0461-3 [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
- 137. Cauli O, Rodrigo R, Piedrafita B, et al. : Inflammation and hepatic encephalopathy: ibuprofen restores learning ability in rats with portacaval shunts. Hepatology. 2007;46(2):514–9. 10.1002/hep.21734 [DOI] [PubMed] [Google Scholar]
- 138. Cauli O, Rodrigo R, Piedrafita B, et al. : Neuroinflammation contributes to hypokinesia in rats with hepatic encephalopathy: ibuprofen restores its motor activity. J Neurosci Res. 2009;87(6):1369–74. 10.1002/jnr.21947 [DOI] [PubMed] [Google Scholar]
- 139. Ahboucha S, Layrargues GP, Mamer O, et al. : Increased brain concentrations of a neuroinhibitory steroid in human hepatic encephalopathy. Ann Neurol. 2005;58(1):169–70. 10.1002/ana.20534 [DOI] [PubMed] [Google Scholar]
- 140. Garcia-Martinez R, Caraceni P, Bernardi M, et al. : Albumin: pathophysiologic basis of its role in the treatment of cirrhosis and its complications. Hepatology. 2013;58(5):1836–46. 10.1002/hep.26338 [DOI] [PubMed] [Google Scholar]
- 141. Jalan R, Kapoor D: Reversal of diuretic-induced hepatic encephalopathy with infusion of albumin but not colloid. Clin Sci (Lond). 2004;106(5):467–74. | 10.1042/CS20030357 [DOI] [PubMed] [Google Scholar]
- 142. Simón-Talero M, García-Martínez R, Torrens M, et al. : Effects of intravenous albumin in patients with cirrhosis and episodic hepatic encephalopathy: a randomized double-blind study. J Hepatol. 2013;59(6):1184–92. | 10.1016/j.jhep.2013.07.020 [DOI] [PubMed] [Google Scholar]
- 143. Bañares R, Nevens F, Larsen FS, et al. : Extracorporeal albumin dialysis with the molecular adsorbent recirculating system in acute-on-chronic liver failure: the RELIEF trial. Hepatology. 2013;57(3):1153–62. | 10.1002/hep.26185 [DOI] [PubMed] [Google Scholar]; F1000 Recommendation
- 144. Llansola M, Rodrigo R, Monfort P, et al. : NMDA receptors in hyperammonemia and hepatic encephalopathy. Metab Brain Dis. 2007;22(3–4):321–35. | 10.1007/s11011-007-9067-0 [DOI] [PubMed] [Google Scholar]
- 145. Erceg S, Monfort P, Hernández-Viadel M, et al. : Oral administration of sildenafil restores learning ability in rats with hyperammonemia and with portacaval shunts. Hepatology. 2005;41(2):299–306. | 10.1002/hep.20565 [DOI] [PubMed] [Google Scholar]
- 146. Agusti A, Hernández-Rabaza V, Balzano T, et al. : Sildenafil reduces neuroinflammation in cerebellum, restores GABAergic tone, and improves motor in-coordination in rats with hepatic encephalopathy. CNS Neurosci Ther. 2017;23(5):386–94. | 10.1111/cns.12688| [DOI] [PMC free article] [PubMed] [Google Scholar]; F1000 Recommendation
- 147. Hernandez-Rabaza V, Agusti A, Cabrera-Pastor A, et al. : Sildenafil reduces neuroinflammation and restores spatial learning in rats with hepatic encephalopathy: underlying mechanisms. J Neuroinflammation. 2015;12:195. | 10.1186/s12974-015-0420-7| [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Strassburger M, Braun H, Reymann KG: Anti-inflammatory treatment with the p38 mitogen-activated protein kinase inhibitor SB239063 is neuroprotective, decreases the number of activated microglia and facilitates neurogenesis in oxygen-glucose-deprived hippocampal slice cultures. Eur J Pharmacol. 2008;592(1–3):55–61. | 10.1016/j.ejphar.2008.06.099 [DOI] [PubMed] [Google Scholar]
- 149. Schindler JF, Monahan JB, Smith WG: p38 pathway kinases as anti-inflammatory drug targets. J Dent Res. 2016;86(9):800–11. | 10.1177/154405910708600902 [DOI] [PubMed] [Google Scholar]
- 150. Agusti A, Cauli O, Rodrigo R, et al. : p38 MAP kinase is a therapeutic target for hepatic encephalopathy in rats with portacaval shunts. Gut. 2011;60(11):1572–9. | 10.1136/gut.2010.236083 [DOI] [PubMed] [Google Scholar]