Skip to main content
Toxicology Reports logoLink to Toxicology Reports
. 2021 Apr 30;8:962–970. doi: 10.1016/j.toxrep.2021.04.011

Drug-induced-acute liver failure: A critical appraisal of the thioacetamide model for the study of hepatic encephalopathy

Ali Sepehrinezhad a,b, Ali Shahbazi c,a,**, Sajad Sahab Negah b,d,e,*, Mohammad Taghi Joghataei a,c, Fin Stolze Larsen f
PMCID: PMC8122178  PMID: 34026559

Highlights

  • To figure out the pathogenesis of HE, the improvement of the animal model is warranted.

  • TAA has been introduced as a valid model because it mimics many pathological features of patients with ALF and HE.

  • TAA as a chemical agent leads to liver injury by triggering the production of ROS and proinflammatory cytokines.

  • TAA causes impairment of CBF, gliopathy, neuroinflammation, and neurological impairments that are seen in ALF patients.

Abbreviations: ALT, alanine aminotransferase; AST, aspartate aminotransferase; AQP4, aquaporin 4 water channel; BBB, blood-brain barrier; B7, B7 molecules (CD80+CD86); CBF, cerebral blood flow; CCL2, chemokine ligand 2; CNS, central nervous system; CTLA4, Cytotoxic T-lymphocyte-associated Protein 4; CYP2E1, Cytochrome P450 family 2 subfamily E member 1; GFAP, glial fibrillary acidic protein; HE, hepatic encephalopathy; Iba1, ionized calcium-binding adaptor molecule 1; IL-β, interleukin 1 β; IL-6, interleukin 6; JNK, c-Jun N-terminal kinase; OA, L-ornithine-l-aspartate; NAC, N-acetylcysteine; NF-κB, nuclear factor κB; TAA, thioacetamide; ROS, reactive oxygen species; TASO, thioacetamide sulfoxide; TASO2, thioacetamide sulfdioxide; TLR-2, toll-like receptor 2; TLR-4, toll-like receptor 4; TNFα, tumor necrosis factor α

Keywords: Acute liver failure, Hepatic encephalopathy, Thioacetamide, Toxicity pathway, Animal model

Abstract

Hepatic encephalopathy (HE) following acute and chronic liver failure is defined as a complex of neuropsychiatric abnormalities, such as discrete personal changes, sleep disorder, forgetfulness, confusion, and decreasing the level of consciousness to coma. The use and design of suitable animal models that represent clinical features and pathological changes of HE are valuable to map the molecular mechanisms that result in HE. Among different types of animal models, thioacetamide (TAA) has been used extensively for the induction of acute liver injury and HE. This agent is not directly hepatotoxic but its metabolites induce liver injury through the induction of oxidative stress and produce systemic inflammation similar to that seen in acute HE patients. In this short review article, we shortly review the most important pathological findings in animal models of acute HE following the administration of TAA.

1. Introduction

Hepatic encephalopathy (HE) is a neuropsychiatric syndrome that is a consequence of acute and chronic liver failure (ACLF) or cirrhosis. Symptoms of HE become apparent depending on the primary cause of the existing liver injury and may include: anxiety, shortened attention span, sleep problems, lethargy, personality change, confusion, forgetfulness, and other serious complications until coma [1]. Patients with ALF have type A HE that can progress to a severe form that is life-threatening due to cerebral edema and intracranial hypertension [2]. As the pathogenesis of HE and cerebral edema is not fully understood an experimental model would be highly valuable to explore the pathological changes at a molecular level. The optimal animal model of HE should include (i) reversibility by appropriate treatment, (ii) reproducibility for induction of coma and for increasing the brain water content/ intracranial pressure, (iii) degree of liver failure, (iv) provide a therapeutic window [3] (Table 1).

Table 1.

Comparison of some chemical agents for inducing animal models of ALF according to Terblanche and Hickman criteria.

Agents Species Reversibility Reproducibility Death from liver failure Therapeutic window Animal size No hazard References
Acetaminophen Pig, dog, rabbit, rat, mouse × ✓× ✓× ✓✓ ✓✓ [18,19,20,21,22,23]
Azoxymethane Mouse ✓✓ × × [24,25,26,27]
Galactosamine Pig, dog, rabbit, rat, mouse × ✓✓ × ✓✓ × [23,28,29]
Lipopolysaccharide Rat, mouse × × × × [30,31,32]
Thioacetamide Guinea pig, rat, mouse ✓✓ ✓✓ × [26,33,34,35,36,37,38]

Several chemical agents have been utilized for the induction of HE following ALF (Table 1). Acetaminophen, galactosamine, and lipopolysaccharide are pharmacological agents that are used for the induction of HE but they are not reproducible because the pathological features of these components have not resembled those seen in HE patients and neuropathological changes are also very variable in different species [4]. For example, some neurological deficits in a low dose of acetaminophen have been reported [5]. Furthermore, there is a diversity in the induction of ALF between male and female mice after injection of acetaminophen. Female mice usually are more resistant to liver injury compared to male mice following the injection of acetaminophen [6]. Acetaminophen causes severe injury in liver sinusoidal endothelial cells and hypovolemic shock that these changes are different from ALF patients [7]. On the other hand, galactosamine and lipopolysaccharide have a short therapeutic window, and their pathological consequences, such as severe permeabilization of the blood-brain barrier (BBB) and cerebral tissue necrosis have not been seen in patients with ALF [4,8]. Galactosamine causes a variety of involvement of multiple organ diseases, such as renal failure and lung injury, and doesn't have specific ALF pathological features [9,10]. Also, azoxymethane and lipopolysaccharide are rarely used for animals larger than mice, due to their high cost and hazards [11,12]. Thioacetamide (TAA) has been widely used in the HE animal model because it is a reproducible model in many animal species, such as mice, rats, and guinea pigs that can induce liver injury that mimics ALF and HE as seen in patients. Furthermore, TAA provides a time window as in human ALF that makes it optimal to do experimental studies of HE.

TAA with molecular formula C2H5NS is an organosulfur compound that was recognized first as a hepatotoxic agent in rats by Fitzhugh and Nelson in 1948 [13]. To induce ALF and acute HE, high doses of TAA should be administrated in a short time according to the International Society for Hepatic Encephalopathy and Nitrogen Metabolism guidelines [4], while chronic low dose administration of TAA for 3–4 months may be valuable to induced cirrhosis and development of liver tumors [[14], [15], [16], [17]]. In this study, we review in more detail the pathological changes found after high doses of TAA administration in mice and rats.

The dose must have been delivered in a short time

2. Pathological consequences of TAA administration

2.1. Peripheral effects

2.1.1. Liver injury

TAA metabolites produce oxidative stress and cause liver injury [39,40] (Box 1). Injection of TAA intraperitoneally in mice causes a significant increase in aspartate aminotransferase (AST), alanine aminotransferase (ALT), and bilirubin [26,[41], [42], [43], [44]] indicates the destruction of the hepatocyte cell membrane [45]. Moreover, TAA may also induce histopathological changes, such as cell apoptosis, pre-portal inflammation, and periacinar hemorrhage [26,41,43,46]. Liver injuries induced by TAA are very predominant in rats and histological damages, such as infiltration of neutrophils and lymphocytes, sinusoidal congestion, sinusoidal hemorrhage, centrilobular necrosis, pigmentation, and hepatocyte vacuolation are observed followed by TAA injection in rats [36,[47], [48], [49], [50], [51], [52], [53]] (Box 2).

Box 1. Cytochrome P450 family 2 subfamily E member 1 mediates hepatotoxic effects of TAA.

Cytochrome P450 family 2 subfamily E member 1 (CYP2E1) is a member of the cytochrome P450 superfamily that plays an important role in the breakdown of many toxins that have entered the body. This protein is responsible for the oxidation of TAA to its metabolites in the liver. In the first step, CYP2E1 metabolizes TAA to thioacetamide sulfoxide (TASO) and then to its hepatotoxic and highly reactive metabolite thioacetamide sulfoxide (TASO2) [39]. Liver injury is induced by this reactive metabolite as a free radical when it covalently binds to the liver lipids and proteins [39,83].

CH3CSNH2TAACYP2E1CH3CSONH2TASO

CH3CSONH2CYP2E1CH3CSO2NH2TASO2Hepatotoxic metabolite

Alt-text: Box 1

Box 2. Strategies for successfully establishing acute hepatic encephalopathy by TAA.

Thioacetamide usually is packaged in bottles as a white or yellowish powder. This agent dissolves easily in water and/ or 0.9 % sodium chloride. To achieve the best results of the HE model using TAA, the concentration of TAA, the number of injections, and animal body weight should be mentioned. For instance, two and three consecutive injections of TAA at doses of 100, 300, and 350 mg/kg, could induce liver injury, hyperammonemia, systemic inflammation, BBB disruption, and CNS damage [26,75,116,142]. Also, a single injection with doses of 300, 600, or 1200 mg/kg are another widely used method for inducing HE [43,50]. Daily assessment of neurological scores and grades of encephalopathy can help to monitor the progression of the disease. To decrease the manifestations of hypoglycemia, hypovolemia, and dehydration, easy access to food on the cage floor, and injections of 5% dextrose and 0.45 % NaCl subcutaneously and daily control of body temperature are essential in animals [115,132].

Alt-text: Box 2

2.1.2. Hyperammonemia

Ammonia with the molecular formula NH3 is a nitrogenous compound that is produced by gut bacteria, enterocytes, and renal tubules via glutaminase enzyme. Under liver failure induced by TAA, ammonia cannot be converted into urea and is released from the hepatic veins into the systemic circulation causing hyperammonemia (Fig. 1). The exact mechanism underlying the neurotoxicity of HE remains unclear. Ammonia is a well-known neurotoxin and is a key molecule that is involved in the pathogenesis of HE [[54], [55], [56], [57]]. The persistence of hyperammonemia aggravates the degree of HE as results in the brain in human ALF while a reduction of the blood levels of NH3 alleviates HE [58]. Among the several toxins suggested, the case for ammonia is most convincing [57]. Events that lead to increased levels of blood or brain ammonia have been revealed to worsen HE, whereas reducing blood ammonia levels improves HE [[59], [60], [61]]. One of the great advantages of TAA is the increasing levels of ammonia in the systemic circulation [26,62,63], liver [50], and cerebral tissue [26,50,64] what has been seen in ALF patients [26]. Clinical, pathological, and biochemical alterations observed in HE can be mimicked by increasing blood or brain ammonia levels in experimental TAA models. The level of ammonia has been increased more than two-fold in animals treated with TAA [26]. Moreover, the severity of encephalopathy was also shown to correlate well with blood and brain ammonia levels in animal models of TAA [26,65,66]. This grading of severity is similar to that seen in HE patients [26,59].

Fig. 1.

Fig. 1

Schematic representation of cascades that may involve the induction of hepatic encephalopathy by thioacetamide. Acute injection of thioacetamide intraperitoneally leads to liver injury through an increase in the production of oxidative stress and inflammation. The dysfunctional liver cannot eliminate blood-borne toxins such as ammonia and lipopolysaccharides from circulation. These agents produce systemic inflammation through the activation of the circulatory monocytes and neutrophils. Ammonia and other toxic agents then reach the brain and cause pathological changes such as neuroinflammation, gliopathy, neuropathy, cerebral edema, intracranial hypertension, and brain herniation. Abbreviations: BBB: blood-brain barrier; B7: B7 molecules (CD80 + CD86); CCL2: C—C Motif Chemokine Ligand 2; CTLA4: Cytotoxic T-lymphocyte-associated Protein 4; CYP2E1: cytochrome P450 family 2 subfamily E member 1; DAMPs: damage-associated molecular patterns; HMGBs: high‑mobility group box proteins; HO-1: heme oxygenase 1; IFN-γ: interferon γ; IL-1β: interleukin 1β; IL-6: interleukin 6; iNOS: inducible nitric oxide synthetase; LPS: lipopolysaccharide; NH4+: ammonia; NF-kB: nuclear factor-kB; NO: nitric oxide; ROS: reactive oxygen species; S100: S100 proteins; TAA: thioacetamide; TASO2: thioacetamide sulfoxide; TNFα: tumor necrosis factor α.

2.1.3. Systemic inflammation

The issue of peripheral inflammation as an important contributor to HE has received considerable critical attention. A growing body of evidence now indicates that ammonia and inflammation synergistically regulate the onset and severity of HE [67,68]. Following hepatocyte injury by TAA, the production of reactive oxygen species (ROS) activates downstream signals, such as c-Jun N-terminal kinase (JNK) and caspase 3. These signaling pathways lead to mitochondrial dysfunctions and the induction of apoptosis in hepatocytes [69]. Injured hepatocytes release damage-associated molecular patterns (DAMPs), S100 proteins, and highmobility group box proteins (HMGBs) with a toll-like receptor 2 (TLR-2) and toll-like receptor 4 (TLR-4) dependent mechanism manner that activates hepatic macrophages called Kupffer cells [70,71] (Fig. 1). Activated Kupffer cells release pro-inflammatory cytokines and increase further liver injury [72]. Besides hyperammonemia, these pro-inflammatory cytokines are released into the blood circulation and activate the circulatory neutrophils. DAMPs are generated from the injured liver that activates the circulatory monocytes [73]. Circulatory activated monocytes and neutrophils also stimulate the expression of pro-inflammatory cytokines, such as tumor necrosis factor α (TNF-α) and interleukin 6 (IL-6) [74]. The severe cytokine storm in the TAA model was comprehensively investigated by Li-Qing Wang and colleagues. They showed that the expressions of IL-1β, IL-6, and TNF-α in plasma and brain significantly increased in TAA rats [75]. In addition to IL-6, TAA injection increases the concentration of serum cytokines, such as IL-1β and chemokine ligand 2 (CCL2) in mice [26,76] (Fig. 1). Likewise, TAA activates the nuclear factor κB (NF-κB) in the mice liver [77]. Similarly, TAA can increase the expression of pro-inflammatory cytokines in blood circulation and increase the level of NF-κB, heme oxygenase-1, TNF-α, and IL-6 in the rat liver [[78], [79], [80]]. Furthermore, it has been reported that the levels of soluble B7 molecules significantly increased in primary hepatocyte culture supernatants and serum of patients with ALF [81]. These molecules through interaction with cytotoxic T lymphocyte-associated protein 4 (CTLA4) modulate T cell activation [82]. In patients with ALF, B7 molecules are secreted from the failing liver that leads to the expression of CTLA4 on circulatory CD4+ cells and causes impairment of T cell function [81]. Overall, the most obvious findings to emerge from these studies are that TAA can induce ALF and causes systemic inflammation which is similar to that seen in HE patients.

2.2. Cerebral effects

2.2.1. Blood-brain barrier

The BBB is a selective structure that is indispensable for preventing the entry of neurotoxic substances into the central nervous system (CNS). BBB includes the basement membrane, endothelial monolayer, pericytes, and astrocyte end-feet. Any pathological alternation in each of these components can disrupt the integrity of BBB [84]. Kato and colleagues with an ultrastructural analysis of the cerebral cortex in nine ALF patients confirmed swelling of astrocytes end-feet and increasing of the number of vacuoles and vesicles in endothelial cells and pericytes. They also reported that the basement membrane was vacuolized while thigh junctions between endothelial cells were intact [85]. These findings indicate the disruption of BBB in ALF is physiochemical in nature compared to structural changes.

Another way of BBB permeabilization by TAA is the peripheral changes, such as the increased plasma level of ammonia, lipopolysaccharides, and pro-inflammatory cytokines that reach the BBB and cause the breakdown of this protective barrier against circulating toxins [26,86,87]. Experiential studies indicate that TAA exacerbates the permeability coefficient of an in vitro BBB model [26] and increases the level of Evans blue dye in the brain tissue of mice indicating BBB disruption [26,88]. However, other studies in rats with ALF induced by TAA have reported no significant changes in the level of Evans blue dye in the brain tissue [89]. Furthermore, the protein expression of claudin-5 and occludin, as two main proteins in the structure of BBB, were not significant between control and TAA rats [90].

2.2.2. Cerebral blood flow

Cerebral autoregulation is a homeostatic process that adjusts and keeps cerebral blood flow (CBF) constant during variations in arterial blood pressure [91]. Disrupted autoregulation causes metabolic impairment of the neural cells and induces cerebral ischemia [91,92]. Many studies have been shown that autoregulation of CBF is decreased or even lost under liver failure conditions and HE patients [[93], [94], [95], [96], [97], [98], [99]]. Importantly, neuropsychiatric symptoms that appear in HE are correlated with impairment of CBF [100,101]. Interestingly, these cerebral changes have been seen in TAA-induced HE rats. For instance, Larsen et al. reported that autoregulation of CBF in the TAA model disappears when the arterial blood pressure is manipulated as the results of injection of norepinephrine and venesection similar to that seen in HE patients [102]. Furthermore, a significant reduction in CBF and cerebral oxygen consumption was observed in TAA-induced HE rats [103].

2.2.3. Neuroinflammation

Ammonia can cross the BBB through ion transporters (in the form of NH4+) and by passive diffusion (in the form of NH3) [104]. Ammonia in the brain parenchyma triggers the activation of microglia in the CNS. Activated microglia releases IL-6 and TNFα which can result in gliopathy and neurodegeneration (Fig. 1). Furthermore, ammonia with activation of toll-like receptor 4 on endothelial cell membrane triggers the production of cytokines [105]. The use of TAA as a hyperammonemia model transforms the resting phenotype of cortical microglia to an activated phenotype with round cell bodies and long processes [26,76]. Furthermore, TAA significantly increases the concentration of CCL2 in the cerebral cortex [26]. Similarly, TAA enhances the concentration of pro-inflammatory cytokines, such as IL-6, IL-1β, TNFα, CCL2, and NF-κB in the brain of rats [65,75,106]. In addition, the expressions of cerebral proinflammatory cytokines were positively correlated with brain water content but negatively correlated with motor activity counts [75]. Also, the expression of ionized calcium-binding adaptor molecule 1 (Iba1) as a marker for microglia increases after acute injection of TAA in the cerebral cortex of rats [106,107]. It has been shown that the number of glial fibrillary acidic protein (GFAP+) cells as an index of neuroinflammation has increased two-fold in the TAA group compared to the control group [42].

2.2.4. Gliopathy

Glial cells are non-neuronal cells of the CNS which provide protection for neural cells and maintain homeostasis of the nervous system. The main glia in the CNS including microglia, astrocyte, and oligodendrocyte. Microglia with the production of inflammation and the ability for phagocytosis plays an important role in tissue repairing following brain injuries. Astrocytes are the supporting cells of CNS that prepare nutrients for neurons and preserve them from ammonia toxicity (the only cell type in CNS that contains glutamine synthetase) and other neurotoxic agents [108]. Evidence shows that TAA affects the function of glial cells in the CNS [42,109,110]. Injection of TAA increases the expression of the GFAP as a marker for astrogliosis in the hippocampus and cerebellum in mice [42,111]. Furthermore, the expression of Iba1 as a marker for microglia significantly increased in the cerebral cortex of rats when treated with TAA [26,106]. Likewise, TAA induces astrocyte swelling in the frontal cortex, cerebellum, hippocampus, and pons in rats [76,112,113] [107]. However, the effect of TAA on glial cell behavior is not fully understood; further studies on this topic are warranted.

2.2.5. Brain edema

One of the life-threatening complications of ALF is cerebral edema. This complication can lead to brain herniation followed by intracranial hypertension. Hyperammonemic conditions and neuroinflammation disturb the glia functions and cause neuronal injuries [114]. Mechanisms of brain edema in ALF remain fully understood. One widely used technique for the assessment of brain edema in animal models is the measurement of brain water content. In this protocol, some pieces of the brain cortex are weighed and dried in an oven overnight and then weighed again. The percentage of brain water content calculates the following formula: [(wet weight - dry weight)/ wet weight]×100 [115]. Acute injection of TAA increases the brain water content and induces brain edema in mice [26]. Furthermore, TAA enhances the brain water content in a neuroinflammation and ammonia-dependent manner in rats [36,75,116]. Aquaporin 4 water channel (AQP4) is the main water channel responsible for water efflux in CNS [117,118]. The expression of this channel in animal models of HE is controversial. For example, it has been reported that TAA-induced acute HE increased the expression of AQP4 and brain edema in the cerebral cortex [116], while in another study, even though the brain water content had been increased but the expression of AQP4 had not changed [119]. This discrepancy may be related to the stages of cerebral edema. In cytotoxic edema, cell swelling is mainly present in astrocytes and the BBB remains intact while in vasogenic edema, the permeability of BBB increases, and net flux of water and blood constituents occur into the extracellular space [120].

2.2.6. Neurological alternations

Patients with liver failure show degrees of motor changes, attention deficits, and cognitive impairment [121]. In mice with ALF following injection of TAA, locomotion score [36,111] and grip strength significantly decreased while ataxia coefficient [26] increased compared to the control group. Cognitive function was also impaired in these mice [111]. Furthermore, in the TAA group, the motor activity score [122], falling latency time in the rotarod test, and exploratory behavior in open field apparatus [65] decreased compared to the control group in rats. Also, all reflexes such as withdrawal, grasping, corneal, auditory startle, head shake, and righting reflex significantly decreased in these rats [107]. Surprisingly, TAA causes several key pathophysiological changes that are seen in HE patients (Table 2).

Table 2.

Symptoms of HE patients in comparison with TAA-induced HE.

Symptoms of HE patients TAA-induced HE model References
Hypovolemia [111,123,124]
Hypokalemia [111,123]
Hypoglycemia [111,119,123,125]
Hypoxia [126,127]
Lack of awareness ×
Euphoria ×
Lethargy or Apathy [75,125,128,129,130,131,132]
Dyspraxia ×
Asterixis ×
Reduced alertness [128,129,131]
Irritability ×
Cognitive impairment [111,133,134,135,136]
Forgetfulness and memory problems [133,137]
Motor function impairment [26,62,75,125,136]
Slurred speech ×
Anxiety [132,138]
Balance problems [26,75,123,125,128,129]
Personality changes ×
Shortened attention span ×
Myoclonus (muscle twitches) ×
Altered sleep rhythm and EEG abnormality [139,140,141]
Coma [26,75,119,128]

3. TAA-induced HE model and pharmacological studies

Pharmacological interventions are developing to relieve the main outcomes in HE patients. To figure out the underlying mechanisms in drug development, an appropriate animal model should be considered. To this point, the review of the most effective drugs used in clinical grades on TAA models can provide us a valuable platform for testing potential therapeutic strategies for HE. In this outline, we reviewed and summarized the results of some effective drugs that have been investigated on TAA models and applied in clinical grades.

L-ornithine-l-aspartate (OA) composes of ornithine and aspartic as natural amino acids. Data from experimental and clinical studies have shown that OA relieves some symptoms of HE by decreasing hyperammonemia [58,143,144]. Since TAA can cause alterations in cell permeability in liver cells [145], direct action of OA on liver cell leakage was investigated in the TAA model [146]. TAA-induced pathogenic changes in the levels of biochemical parameters (i.e., AST, ALT, and alkaline phosphatase levels) were notably improved by the OA treatment [146]. Furthermore, OA improved motor activity, grip strength, and severity of disease in TAA-induced HE in mice [147].

Lactulose and rifaximin are widely used for the treatment of HE patients [148]. Lactulose, a non-absorbable disaccharide, decreases the breakdown of nitrogenous compounds to ammonia with acidification of the intestinal tract [149]. It has been shown that TAA-induced pathogenic changes in the levels of blood AST, ALT, and ammonia were significantly improved by lactulose treatment [150,151]. Moreover, a significant improvement in survival rate, sensory behavior, and motor activity was observed by lactulose treatment in TAA-induced HE rats [150]. Furthermore, lactulose significantly reduced the level of total bilirubin and increased the concentration of albumin in TAA-induced HE [152]. Rifaximin, a broad-spectrum antibiotic, decreases the production and absorption of intestinal ammonia by affecting the population of gut flora; therefore, this intervention leads to a significant increase in the level of portal ammonia in HE [153]. Likewise, the administration of rifaximin significantly decreased the concentrations of AST and ALT as well as the level of serum ammonia in TAA-induced acute HE rats [154].

Indomethacin and N-acetylcysteine represent acceptable results in ALF patients [155,156]. Indomethacin is a nonsteroidal anti-inflammatory drug that has beneficial effects in uncontrolled intracranial pressure following acute liver failure [157,158]. N-acetylcysteine (NAC) is a first-line treatment for acetaminophen hepatotoxicity [159,160]. However, it is also suggested for non-acetaminophen-induced ALF patients [161]. Administration of NAC prevents the outcome of HE from worsening and decreases the duration of hospitalization in ALF patients [[162], [163], [164]]. The precise mechanisms of NAC in ALF patients remain unclear. However, antioxidant and anti-inflammatory properties have been previously reported [165,166]. NAC improves liver function tests, serum pro-inflammatory cytokines, and oxidative stress in TAA-induced ALF and HE [151,167,168]. Taken together, TAA imitates some critical aspects of HE pathogenesis; therefore, it is a reasonable model for drug development in the context of HE.

4. Conclusion

Administration of high doses of TAA intraperitoneally is an efficient model for studying HE pathogenesis. This agent acutely increases the level of liver, plasma, and brain ammonia that can induce systemic inflammation and neuroinflammation. Therefore, this model can be introduced as a valid model for studying the mechanisms of neuroinflammation and microglia activation under hyperammonemia conditions. Furthermore, the pathological processes that are involved in astrocyte swelling and brain edema can be investigated by the acute injection of TAA in mice and rats. However, the BBB seems less affected by TAA as seen in human ALF [85]. All of the pathophysiological alterations in the liver, circulation, and within the brain after TAA intoxication grossly mimic changes seen in human ALF. These make this model attractive for future studies of the devastating effects of severe HE.

Ethics approval and consent to participate

Not applicable.

Consent for publication

Not applicable

Availability of data and materials

No datasets were generated during the study.

Funding

Funding information is not applicable.

Declaration of Competing Interest

The authors declare that there is no conflict of interest.

Acknowledgments

Not applicable.

Edited by Dr. A.M Tsatsaka

Contributor Information

Ali Shahbazi, Email: shahbazi.a@iums.ac.ir.

Sajad Sahab Negah, Email: sahabnegahs@mums.ac.ir.

References

  • 1.López-Franco Ó., Cortés-Sol A., Molina-Jiménez T., Del Moral D.I., Flores-Muñoz M., Roldán-Roldán G., Juárez Portilla C.J., Morin J.-P., Zepeda R.C. Cognitive impairment after resolution of hepatic encephalopathy: a systematic review and meta-analysis. Front. Neurosci. 2021;15:69. doi: 10.3389/fnins.2021.579263. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Scott T.R., Kronsten V.T., Hughes R.D., Shawcross D.L. Pathophysiology of cerebral oedema in acute liver failure. World J. Gastroenterol. 2013;19:9240–9255. doi: 10.3748/wjg.v19.i48.9240. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3.Terblanche J., Hickman R. Animal models of fulminant hepatic failure. Dig. Dis. Sci. 1991;36:770–774. doi: 10.1007/BF01311235. [DOI] [PubMed] [Google Scholar]
  • 4.Butterworth R.F., Norenberg M.D., Felipo V., Ferenci P., Albrecht J., Blei A.T. Experimental models of hepatic encephalopathy: ISHEN guidelines. Liver Int. 2009;29:783–788. doi: 10.1111/j.1478-3231.2009.02034.x. [DOI] [PubMed] [Google Scholar]
  • 5.McGill M.R., Williams C.D., Xie Y., Ramachandran A., Jaeschke H. Acetaminophen-induced liver injury in rats and mice: comparison of protein adducts, mitochondrial dysfunction, and oxidative stress in the mechanism of toxicity. Toxicol. Appl. Pharmacol. 2012;264:387–394. doi: 10.1016/j.taap.2012.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Du K., Williams C.D., McGill M.R., Jaeschke H. Lower susceptibility of female mice to acetaminophen hepatotoxicity: role of mitochondrial glutathione, oxidant stress and c-jun N-terminal kinase. Toxicol. Appl. Pharmacol. 2014;281:58–66. doi: 10.1016/j.taap.2014.09.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Yin H., Cheng L., Holt M., Hail N., Jr, Maclaren R., Ju C. Lactoferrin protects against acetaminophen-induced liver injury in mice. Hepatology (Baltimore, Md.) 2010;51:1007–1016. doi: 10.1002/hep.23476. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8.Rahman T.M., Hodgson H.J. Animal models of acute hepatic failure. Int. J. Exp. Pathol. 2000;81:145–157. doi: 10.1046/j.1365-2613.2000.00144.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Anand R., Harry D., Holt S., Milner P., Dashwood M., Goodier D., Jarmulowicz M., Moore K. Endothelin is an important determinant of renal function in a rat model of acute liver and renal failure. Gut. 2002;50:111–117. doi: 10.1136/gut.50.1.111. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 10.Oztay F., Gezginci-Oktayoglu S., Bayrak B.B., Yanardag R., Bolkent S. Cathepsin B inhibition improves lung injury associated to D-galactosamine/tumor necrosis factor-alpha-induced liver injury in mice. Mol. Cell. Biochem. 2010;333:65–72. doi: 10.1007/s11010-009-0205-3. [DOI] [PubMed] [Google Scholar]
  • 11.Tuñón M.J., Alvarez M., Culebras J.M., González-Gallego J. An overview of animal models for investigating the pathogenesis and therapeutic strategies in acute hepatic failure. World J. Gastroenterol. 2009;15:3086–3098. doi: 10.3748/wjg.15.3086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Kobaek-Larsen M., Fenger C., Ritskes-Hoitinga J. Secondary effects induced by the colon carcinogen azoxymethane in BDIX rats, APMIS : acta pathologica, microbiologica. et immunologica Scandinavica. 2004;112:319–329. doi: 10.1111/j.1600-0463.2004.apm1120601.x. [DOI] [PubMed] [Google Scholar]
  • 13.Fitzhugh O.G., Nelson A.A. Liver tumors in rats fed thiourea or thioacetamide. Science (New York, N.Y.) 1948;108:626–628. doi: 10.1126/science.108.2814.626. [DOI] [PubMed] [Google Scholar]
  • 14.David P., Alexandre E., Chenard-Neu M.P., Wolf P., Jaeck D., Richert L. Failure of liver cirrhosis induction by thioacetamide in Nagase analbuminaemic rats. Lab. Anim. 2002;36:158–164. doi: 10.1258/0023677021912442. [DOI] [PubMed] [Google Scholar]
  • 15.Cruz A., Padillo F.J., Torres E., Navarrete C.M., Muñoz-Castañeda J.R., Caballero F.J., Briceño J., Marchal T., Túnez I., Montilla P., Pera C., Muntané J. Melatonin prevents experimental liver cirrhosis induced by thioacetamide in rats. J. Pineal Res. 2005;39:143–150. doi: 10.1111/j.1600-079X.2005.00227.x. [DOI] [PubMed] [Google Scholar]
  • 16.Yang M.C., Chang C.P., Lei H.Y. Induction of liver fibrosis in a murine hepatoma model by thioacetamide is associated with enhanced tumor growth and suppressed antitumor immunity. Lab. Invest. 2010;90:1782–1793. doi: 10.1038/labinvest.2010.139. [DOI] [PubMed] [Google Scholar]
  • 17.Wallace M.C., Hamesch K., Lunova M., Kim Y., Weiskirchen R., Strnad P., Friedman S.L. Standard operating procedures in experimental liver research: thioacetamide model in mice and rats. Lab. Anim. 2015;49:21–29. doi: 10.1177/0023677215573040. [DOI] [PubMed] [Google Scholar]
  • 18.Doering C.B., Parker E.T., Nichols C.E., Lollar P. Decreased factor VIII levels during acetaminophen-induced murine fulminant hepatic failure. Blood. 2003;102:1743–1744. doi: 10.1182/blood-2003-03-0826. [DOI] [PubMed] [Google Scholar]
  • 19.Nguyen T.H., Mai G., Villiger P., Oberholzer J., Salmon P., Morel P., Bühler L., Trono D. Treatment of acetaminophen-induced acute liver failure in the mouse with conditionally immortalized human hepatocytes. J. Hepatol. 2005;43:1031–1037. doi: 10.1016/j.jhep.2005.05.036. [DOI] [PubMed] [Google Scholar]
  • 20.Jeong T.B., Kim J.-H., Kim S.H., Lee S., Son S.W., Lim Y., Cho J.-Y., Hwang D.Y., Kim K.S., Kwak J.-H., Jung Y.-S. Comparison of toxic responses to acetaminophen challenge in ICR mice originating from different sources. Lab. Anim. Res. 2019;35:16. doi: 10.1186/s42826-019-0017-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Eakins R., Walsh J., Randle L., Jenkins R.E., Schuppe-Koistinen I., Rowe C., Starkey Lewis P., Vasieva O., Prats N., Brillant N., Auli M., Bayliss M., Webb S., Rees J.A., Kitteringham N.R., Goldring C.E., Park B.K. Adaptation to acetaminophen exposure elicits major changes in expression and distribution of the hepatic proteome. Sci. Rep. 2015;5:16423. doi: 10.1038/srep16423. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Henne-Bruns D., Artwohl J., Broelsch C., Kremer B. Acetaminophen-induced acute hepatic failure in pigs: controversical results to other animal models. Res. Exp. Med. 1988;188:463–472. doi: 10.1007/BF01852004. [DOI] [PubMed] [Google Scholar]
  • 23.Newsome P.N., Plevris J.N., Nelson L.J., Hayes P.C. Animal models of fulminant hepatic failure: a critical evaluation. Liver Transpl. 2000;6:21–31. doi: 10.1002/lt.500060110. [DOI] [PubMed] [Google Scholar]
  • 24.Bélanger M., Côté J., Butterworth R.F. Neurobiological characterization of an azoxymethane mouse model of acute liver failure. Neurochem. Int. 2006;48:434–440. doi: 10.1016/j.neuint.2005.11.022. [DOI] [PubMed] [Google Scholar]
  • 25.Suzuki R., Kohno H., Sugie S., Nakagama H., Tanaka T. Strain differences in the susceptibility to azoxymethane and dextran sodium sulfate-induced colon carcinogenesis in mice. Carcinogenesis. 2005;27:162–169. doi: 10.1093/carcin/bgi205. [DOI] [PubMed] [Google Scholar]
  • 26.Grant S., McMillin M., Frampton G., Petrescu A.D., Williams E., Jaeger V., Kain J., DeMorrow S. Direct comparison of the thioacetamide and azoxymethane models of type a hepatic encephalopathy in mice. Gene Expr. 2018;18:171–185. doi: 10.3727/105221618X15287315176503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.McMillin M.A., Frampton G.A., Seiwell A.P., Patel N.S., Jacobs A.N., DeMorrow S. TGFβ1 exacerbates blood–brain barrier permeability in a mouse model of hepatic encephalopathy via upregulation of MMP9 and downregulation of claudin-5. Lab. Investig. 2015;95:903–913. doi: 10.1038/labinvest.2015.70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Le Minh K., Kuhla A., Abshagen K., Minor T., Stegemann J., Ibrahim S., Eipel C., Vollmar B. Uncoupling protein-2 deficiency provides protection in a murine model of endotoxemic acute liver failure. Crit. Care Med. 2009;37:215–222. doi: 10.1097/CCM.0b013e31819260ae. [DOI] [PubMed] [Google Scholar]
  • 29.Saracyn M., Zdanowski R., Brytan M., Kade G., Nowak Z., Patera J., Dyrla P., Gil J., Wańkowicz Z. D-Galactosamine intoxication in experimental animals: is it only an experimental model of acute liver failure? Med. Sci. Monit. 2015;21:1469–1477. doi: 10.12659/MSM.893291. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Ma K., Zhang Y., Zhu D., Lou Y. Protective effects of asiatic acid against D-galactosamine/lipopolysaccharide-induced hepatotoxicity in hepatocytes and kupffer cells co-cultured system via redox-regulated leukotriene C4 synthase expression pathway. Eur. J. Pharmacol. 2009;603:98–107. doi: 10.1016/j.ejphar.2008.11.054. [DOI] [PubMed] [Google Scholar]
  • 31.Liu Y., Li F., Zhang L., Wu J., Wang Y., Yu H. Taurine alleviates lipopolysaccharide‑induced liver injury by anti‑inflammation and antioxidants in rats. Mol. Med. Rep. 2017;16:6512–6517. doi: 10.3892/mmr.2017.7414. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Islam M.S., Yu H., Miao L., Liu Z., He Y., Sun H. Hepatoprotective effect of the ethanol extract of Illicium henryi against acute liver injury in mice induced by lipopolysaccharide. Antioxidants (Basel, Switzerland) 2019;8 doi: 10.3390/antiox8100446. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Chilakapati J., Shankar K., Korrapati M.C., Hill R.A., Mehendale H.M. Saturation toxicokinetics of thioacetamide: role in initiation of liver injury. Drug Metab. Dispos. 2005;33:1877–1885. doi: 10.1124/dmd.105.005520. [DOI] [PubMed] [Google Scholar]
  • 34.Lin Y.-Y., Hu C.-T., Sun D.-S., Lien T.-S., Chang H.-H. Thioacetamide-induced liver damage and thrombocytopenia is associated with induction of antiplatelet autoantibody in mice. Sci. Rep. 2019;9:17497. doi: 10.1038/s41598-019-53977-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Czarnecka A., Aleksandrowicz M., Jasiński K., Jaźwiec R., Kalita K., Hilgier W., Zielińska M. Cerebrovascular reactivity and cerebral perfusion of rats with acute liver failure: role of L-glutamine and asymmetric dimethylarginine in L-arginine-induced response. J. Neurochem. 2018;147:692–704. doi: 10.1111/jnc.14578. [DOI] [PubMed] [Google Scholar]
  • 36.Heidari R., Jamshidzadeh A., Niknahad H., Mardani E., Ommati M.M., Azarpira N., Khodaei F., Zarei A., Ayarzadeh M., Mousavi S., Abdoli N., Yeganeh B.S., Saeedi A., Najibi A. Effect of taurine on chronic and acute liver injury: focus on blood and brain ammonia. Toxicol. Rep. 2016;3:870–879. doi: 10.1016/j.toxrep.2016.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Ra S.-H., Shin R.-H., Ri H.-C., Ri J.-H., Ri H.-C., Ri A.-J. Effect of lesimarin against thioacetamide-induced liver cirrhosis in rat. Braz. J. Pharm. Sci. 2019;55 [Google Scholar]
  • 38.Alalkam E. Hibiscus Sabdariffa aqueous extract Ameliorates thioacetamide-induced hepatic encephalopathy in Guinea pigs: role of ammonia extraction. Al-Azhar J. Pharm. Sci. 2018;58:19–36. [Google Scholar]
  • 39.Hunter A.L., Holscher M.A., Neal R.A. Thioacetamide-induced hepatic necrosis. I. Involvement of the mixed-function oxidase enzyme system. J. Pharmacol. Exp. Ther. 1977;200:439–448. [PubMed] [Google Scholar]
  • 40.Pallottini V., Martini C., Bassi A.M., Romano P., Nanni G., Trentalance A. Rat HMGCoA reductase activation in thioacetamide-induced liver injury is related to an increased reactive oxygen species content. J. Hepatol. 2006;44:368–374. doi: 10.1016/j.jhep.2005.06.011. [DOI] [PubMed] [Google Scholar]
  • 41.Avraham Y., Zolotarev O., Grigoriadis N.C., Poutahidis T., Magen I., Vorobiav L., Zimmer A., Ilan Y., Mechoulam R., Berry E.M. Cannabinoids and capsaicin improve liver function following thioacetamide-induced acute injury in mice. Am. J. Gastroenterol. 2008;103:3047–3056. doi: 10.1111/j.1572-0241.2008.02155.x. [DOI] [PubMed] [Google Scholar]
  • 42.Avraham Y., Grigoriadis N., Poutahidis T., Vorobiev L., Magen I., Ilan Y., Mechoulam R., Berry E. Cannabidiol improves brain and liver function in a fulminant hepatic failure-induced model of hepatic encephalopathy in mice. Br. J. Pharmacol. 2011;162:1650–1658. doi: 10.1111/j.1476-5381.2010.01179.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Miranda A.S., Rodrigues D.H., Vieira L.B., Lima C.X., Rachid M.A., Vidigal P.V., Gomez M.V., Reis H.J., Guatimosim C., Teixeira A.L. A thioacetamide-induced hepatic encephalopathy model in C57BL/6 mice: a behavioral and neurochemical study. Arquivos de neuro-psiquiatria. 2010;68:597–602. doi: 10.1590/s0004-282x2010000400022. [DOI] [PubMed] [Google Scholar]
  • 44.Fernández-Martínez A., Callejas N.A., Casado M., Boscá L., Martín-Sanz P. Thioacetamide-induced liver regeneration involves the expression of cyclooxygenase 2 and nitric oxide synthase 2 in hepatocytes. J. Hepatol. 2004;40:963–970. doi: 10.1016/j.jhep.2004.02.019. [DOI] [PubMed] [Google Scholar]
  • 45.Contreras-Zentella M.L., Hernández-Muñoz R. Is liver enzyme release really associated with cell necrosis induced by oxidant stress? Oxid. Med. Cell. Longev. 2016;2016 doi: 10.1155/2016/3529149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Honda H., Ikejima K., Hirose M., Yoshikawa M., Lang T., Enomoto N., Kitamura T., Takei Y., Sato N. Leptin is required for fibrogenic responses induced by thioacetamide in the murine liver. Hepatology (Baltimore, Md.) 2002;36:12–21. doi: 10.1053/jhep.2002.33684. [DOI] [PubMed] [Google Scholar]
  • 47.Mehul D., Varsha G. 2012. Effect of Polyherbal Preparation on Thioacetamide Induced Liver Damage and Hepatic Encephalopathy in Rats. [Google Scholar]
  • 48.Marciniak S., Wnorowski A., Smolińska K., Walczyna B., Turski W., Kocki T., Paluszkiewicz P., Parada-Turska J. Kynurenic acid protects against thioacetamide-induced liver injury in rats. Anal. Cell. Pathol. Amst. (Amst) 2018;2018 doi: 10.1155/2018/1270483. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.El-Latif El-Ghazaly M.A., Rashed E.R., Shafey G.M., Zaki H.F., Attia A.S. Amelioration of thioacetamide-induced hepatic encephalopathy in rats by low-dose gamma irradiation. Environ. Sci. Pollut. Res. Int. 2020;27:334–343. doi: 10.1007/s11356-019-06934-w. [DOI] [PubMed] [Google Scholar]
  • 50.Sathyasaikumar K.V., Swapna I., Reddy P.V., Murthy Ch R., Roy K.R., Dutta Gupta A., Senthilkumaran B., Reddanna P. Co-administration of C-Phycocyanin ameliorates thioacetamide-induced hepatic encephalopathy in Wistar rats. J. Neurol. Sci. 2007;252:67–75. doi: 10.1016/j.jns.2006.10.014. [DOI] [PubMed] [Google Scholar]
  • 51.Margeli A.P., Papadimitriou L., Ninos S., Manolis E., Mykoniatis M.G., Theocharis S.E. Hepatic stimulator substance administration ameliorates liver regeneration in an animal model of fulminant hepatic failure and encephalopathy. Liver Int. 2003;23:171–178. doi: 10.1034/j.1600-0676.2003.00828.x. [DOI] [PubMed] [Google Scholar]
  • 52.Özdemir-Kumral Z.N., Erkek B.E., Karakuş B., Almacı M., Fathi R., Yüksel M., Cumbul A., Alican İ. Potential effect of 1,25 dihydroxyvitamin d(3) on thioacetamide-induced hepatotoxicity in rats. J. Surg. Res. 2019;243:165–172. doi: 10.1016/j.jss.2019.05.020. [DOI] [PubMed] [Google Scholar]
  • 53.Mustafa H.N., El Awdan S.A., Hegazy G.A. Protective role of antioxidants on thioacetamide-induced acute hepatic encephalopathy: biochemical and ultrastructural study. Tissue Cell. 2013;45:350–362. doi: 10.1016/j.tice.2013.06.001. [DOI] [PubMed] [Google Scholar]
  • 54.Stewart C.A., Smith G.E. Minimal hepatic encephalopathy. Nat. Clin. Pract. Gastroenterol. Hepatol. 2007;4:677–685. doi: 10.1038/ncpgasthep0999. [DOI] [PubMed] [Google Scholar]
  • 55.Clemmesen J.O., Larsen F.S., Kondrup J., Hansen B.A., Ott P. Cerebral herniation in patients with acute E liver failure is correlated with arterial ammonia concentration. Hepatology (Baltimore, Md.) 1999;29:648–653. doi: 10.1002/hep.510290309. [DOI] [PubMed] [Google Scholar]
  • 56.Pozdeev V.I., Lang E., Görg B., Bidmon H.J., Shinde P.V., Kircheis G., Herebian D., Pfeffer K., Lang F., Häussinger D., Lang K.S., Lang P.A. TNFα induced up-regulation of Na+,K+,2Cl− cotransporter NKCC1 in hepatic ammonia clearance and cerebral ammonia toxicity. Sci. Rep. 2017;7:7938. doi: 10.1038/s41598-017-07640-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Liu J., Xu Y., Jiang B. Novel insights into pathogenesis and therapeutic strategies of hepatic encephalopathy, from the gut microbiota perspective. Front. Cell. Infect. Microbiol. 2021;11 doi: 10.3389/fcimb.2021.586427. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Butterworth R.F., Giguère J.F., Michaud J., Lavoie J., Layrargues G.P. Ammonia: key factor in the pathogenesis of hepatic encephalopathy. Neurochem. Pathol. 1987;6:1–12. doi: 10.1007/BF02833598. [DOI] [PubMed] [Google Scholar]
  • 59.Ong J.P., Aggarwal A., Krieger D., Easley K.A., Karafa M.T., Van Lente F., Arroliga A.C., Mullen K.D. Correlation between ammonia levels and the severity of hepatic encephalopathy. Am. J. Med. 2003;114:188–193. doi: 10.1016/s0002-9343(02)01477-8. [DOI] [PubMed] [Google Scholar]
  • 60.Holecek M. Ammonia and amino acid profiles in liver cirrhosis: effects of variables leading to hepatic encephalopathy. Nutrition. 2015;31:14–20. doi: 10.1016/j.nut.2014.03.016. [DOI] [PubMed] [Google Scholar]
  • 61.McKinney A.M., Lohman B., Sarikaya B., Uhlmann E., Spanbauer J., Singewald T., Brace J. Acute hepatic encephalopathy: diffusion-weighted and fluid-attenuated inversion recovery findings, and correlation with plasma ammonia level and clinical outcome. Am. J. Neuroradiol. 2010;31:1471–1479. doi: 10.3174/ajnr.A2112. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Mladenović D., Radosavljević T., Hrnčić D., Rašić-Marković A., Puškaš N., Maksić N., Djuric D., Stanojlović O. Behavioral and electroencephalographic manifestations of thioacetamide-induced encephalopathy in rats. Can. J. Physiol. Pharmacol. 2012;90:1219–1227. doi: 10.1139/y2012-088. [DOI] [PubMed] [Google Scholar]
  • 63.Nicaise C., Prozzi D., Viaene E., Moreno C., Gustot T., Quertinmont E., Demetter P., Suain V., Goffin P., Devière J., Hols P. Control of acute, chronic, and constitutive hyperammonemia by wild-type and genetically engineered Lactobacillus plantarum in rodents. Hepatology (Baltimore, Md.) 2008;48:1184–1192. doi: 10.1002/hep.22445. [DOI] [PubMed] [Google Scholar]
  • 64.Wysmyk U., Oja S.S., Saransaari P. Taurine release from brain slices in thioacetamide-induced hepatic encephalopathy in rats. Mol. Chem. Neuropathol. 1991;14:25–34. doi: 10.1007/BF03160995. [DOI] [PubMed] [Google Scholar]
  • 65.El-Marasy S.A., El Awdan S.A., Abd-Elsalam R.M. Protective role of chrysin on thioacetamide-induced hepatic encephalopathy in rats. Chem. Biol. Interact. 2019;299:111–119. doi: 10.1016/j.cbi.2018.11.021. [DOI] [PubMed] [Google Scholar]
  • 66.Mostafa R.E., Salama A.A., Abdel-Rahman R.F., Ogaly H.A. Hepato-and neuro-protective influences of biopropolis on thioacetamide-induced acute hepatic encephalopathy in rats. Can. J. Physiol. Pharmacol. 2017;95:539–547. doi: 10.1139/cjpp-2016-0433. [DOI] [PubMed] [Google Scholar]
  • 67.Aldridge D.R., Tranah E.J., Shawcross D.L. Pathogenesis of hepatic encephalopathy: role of ammonia and systemic inflammation. J. Clin. Exp. Hepatol. 2015;5:S7–s20. doi: 10.1016/j.jceh.2014.06.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Pedersen H.R., Ring-Larsen H., Olsen N.V., Larsen F.S. Hyperammonemia acts synergistically with lipopolysaccharide in inducing changes in cerebral hemodynamics in rats anaesthetised with pentobarbital. J. Hepatol. 2007;47:245–252. doi: 10.1016/j.jhep.2007.03.026. [DOI] [PubMed] [Google Scholar]
  • 69.Marderstein E.L., Bucher B., Guo Z., Feng X., Reid K., Geller D.A. Protection of rat hepatocytes from apoptosis by inhibition of c-Jun N-terminal kinase. Surgery. 2003;134:280–284. doi: 10.1067/msy.2003.237. [DOI] [PubMed] [Google Scholar]
  • 70.Kuramochi M., Izawa T., Pervin M., Bondoc A., Kuwamura M., Yamate J. The kinetics of damage-associated molecular patterns (DAMPs) and toll-like receptors during thioacetamide-induced acute liver injury in rats. Exp. Toxicol. Pathol. 2016;68:471–477. doi: 10.1016/j.etp.2016.06.005. [DOI] [PubMed] [Google Scholar]
  • 71.Erridge C. Endogenous ligands of TLR2 and TLR4: agonists or assistants? J. Leukoc. Biol. 2010;87:989–999. doi: 10.1189/jlb.1209775. [DOI] [PubMed] [Google Scholar]
  • 72.Possamai L.A., Antoniades C.G., Anstee Q.M., Quaglia A., Vergani D., Thursz M., Wendon J. Role of monocytes and macrophages in experimental and human acute liver failure. World J. Gastroenterol. 2010;16:1811–1819. doi: 10.3748/wjg.v16.i15.1811. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 73.Larsen F.S., Schmidt L.E., Bernsmeier C., Rasmussen A., Isoniemi H., Patel V.C., Triantafyllou E., Bernal W., Auzinger G., Shawcross D., Eefsen M., Bjerring P.N., Clemmesen J.O., Hockerstedt K., Frederiksen H.J., Hansen B.A., Antoniades C.G., Wendon J. High-volume plasma exchange in patients with acute liver failure: an open randomised controlled trial. J. Hepatol. 2016;64:69–78. doi: 10.1016/j.jhep.2015.08.018. [DOI] [PubMed] [Google Scholar]
  • 74.Tranah T.H., Vijay G.K.M., Ryan J.M., Shawcross D.L. Systemic inflammation and ammonia in hepatic encephalopathy. Metab. Brain Dis. 2013;28:1–5. doi: 10.1007/s11011-012-9370-2. [DOI] [PubMed] [Google Scholar]
  • 75.Wang L.-Q., Zhou H.-J., Pan C.-F., Zhu S.-M., Xu L.-M. Expression of IL-1β, IL-6 and TNF-α in rats with thioacetamide-induced acute liver failure and encephalopathy: correlation with brain edema. Asian Biomed. 2011;5:205. [Google Scholar]
  • 76.Faleiros B.E., Miranda A.S., Campos A.C., Gomides L.F., Kangussu L.M., Guatimosim C., Camargos E.R., Menezes G.B., Rachid M.A., Teixeira A.L. Up-regulation of brain cytokines and chemokines mediates neurotoxicity in early acute liver failure by a mechanism independent of microglial activation. Brain Res. 2014;1578:49–59. doi: 10.1016/j.brainres.2014.07.001. [DOI] [PubMed] [Google Scholar]
  • 77.Lin X., Wei J., Nie J., Bai F., Zhu X., Zhuo L., Lu Z., Huang Q. Inhibition of RKIP aggravates thioacetamide-induced acute liver failure in mice. Exp. Ther. Med. 2018;16:2992–2998. doi: 10.3892/etm.2018.6542. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 78.Chu C.J., Hsiao C.C., Wang T.F., Chan C.Y., Lee F.Y., Chang F.Y., Chen Y.C., Huang H.C., Wang S.S., Lee S.D. Prostacyclin inhibition by indomethacin aggravates hepatic damage and encephalopathy in rats with thioacetamide-induced fulminant hepatic failure. World J. Gastroenterol. 2005;11:232–236. doi: 10.3748/wjg.v11.i2.232. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Demirel U., Yalniz M., Aygün C., Orhan C., Tuzcu M., Sahin K., Ozercan I.H., Bahçecioğlu I.H. Allopurinol ameliorates thioacetamide-induced acute liver failure by regulating cellular redox-sensitive transcription factors in rats. Inflammation. 2012;35:1549–1557. doi: 10.1007/s10753-012-9470-5. [DOI] [PubMed] [Google Scholar]
  • 80.Luo M., Dong L., Li J., Wang Y., Shang B. Protective effects of pentoxifylline on acute liver injury induced by thioacetamide in rats. Int. J. Clin. Exp. Pathol. 2015;8:8990–8996. [PMC free article] [PubMed] [Google Scholar]
  • 81.Khamri W., Abeles R.D., Hou T.Z., Anderson A.E., El-Masry A., Triantafyllou E., Bernsmeier C., Larsen F.S., Singanayagam A., Kudo N., Possamai L.A., Lebosse F., Auzinger G., Bernal W., Willars C., Weston C.J., Lombardi G., Wendon J., Thursz M., Antoniades C.G. Increased Expression of Cytotoxic T-Lymphocyte-Associated Protein 4 by T Cells, Induced by B7 in Sera, Reduces Adaptive Immunity in Patients With Acute Liver Failure. Gastroenterology. 2017;153 doi: 10.1053/j.gastro.2017.03.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82.Sansom D.M. CD28, CTLA-4 and their ligands: who does what and to whom? Immunology. 2000;101:169–177. doi: 10.1046/j.1365-2567.2000.00121.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Hajovsky H., Hu G., Koen Y., Sarma D., Cui W., Moore D.S., Staudinger J.L., Hanzlik R.P. Metabolism and toxicity of thioacetamide and thioacetamide S-oxide in rat hepatocytes. Chem. Res. Toxicol. 2012;25:1955–1963. doi: 10.1021/tx3002719. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Banks W.A. The blood–brain barrier as an endocrine tissue. Nat. Rev. Endocrinol. 2019;15:444–455. doi: 10.1038/s41574-019-0213-7. [DOI] [PubMed] [Google Scholar]
  • 85.Kato M., Hughes R.D., Keays R.T., Williams R. Electron microscopic study of brain capillaries in cerebral edema from fulminant hepatic failure. Hepatology (Baltimore, Md.) 1992;15:1060–1066. doi: 10.1002/hep.1840150615. [DOI] [PubMed] [Google Scholar]
  • 86.Northrop N.A., Halpin L.E., Yamamoto B.K. Peripheral ammonia and blood brain barrier structure and function after methamphetamine. Neuropharmacology. 2016;107:18–26. doi: 10.1016/j.neuropharm.2016.03.018. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Shmarakov I.O., Borschovetska V.L., Marchenko M.M., Blaner W.S. Retinoids modulate thioacetamide-induced acute hepatotoxicity. Toxicol. Sci. 2014;139:284–292. doi: 10.1093/toxsci/kfu045. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Liu L., Miao M., Chen Y., Wang Z., Sun B., Liu X. Altered function and expression of ABC transporters at the blood-brain barrier and increased brain distribution of phenobarbital in acute liver failure mice. Front. Pharmacol. 2018;9:190. doi: 10.3389/fphar.2018.00190. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Jin S., Wang X.T., Liu L., Yao D., Liu C., Zhang M., Guo H.F., Liu X.D. P-glycoprotein and multidrug resistance-associated protein 2 are oppositely altered in brain of rats with thioacetamide-induced acute liver failure. Liver Int. 2013;33:274–282. doi: 10.1111/j.1478-3231.2012.02862.x. [DOI] [PubMed] [Google Scholar]
  • 90.Li Y., Zhang J., Xu P., Sun B., Zhong Z., Liu C., Ling Z., Chen Y., Shu N., Zhao K., Liu L., Liu X. Acute liver failure impairs function and expression of breast cancer-resistant protein (BCRP) at rat blood-brain barrier partly via ammonia-ROS-ERK1/2 activation. J. Neurochem. 2016;138:282–294. doi: 10.1111/jnc.13666. [DOI] [PubMed] [Google Scholar]
  • 91.Armstead W.M. Cerebral blood flow autoregulation and dysautoregulation. Anesthesiol. Clin. 2016;34:465–477. doi: 10.1016/j.anclin.2016.04.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 92.Hossmann K.A. Viability thresholds and the penumbra of focal ischemia. Ann. Neurol. 1994;36:557–565. doi: 10.1002/ana.410360404. [DOI] [PubMed] [Google Scholar]
  • 93.Dam G., Keiding S., Munk O.L., Ott P., Vilstrup H., Bak L.K., Waagepetersen H.S., Schousboe A., Sørensen M. Hepatic encephalopathy is associated with decreased cerebral oxygen metabolism and blood flow, not increased ammonia uptake. Hepatology (Baltimore, Md.) 2013;57:258–265. doi: 10.1002/hep.25995. [DOI] [PubMed] [Google Scholar]
  • 94.Zheng G., Lu H., Yu W., Luo S., Liu Y., Liu W., Liu H., Wu L., Zheng L., Kong X., Zhang L.J., Lu G.M. Severity-specific alterations in CBF, OEF and CMRO2 in cirrhotic patients with hepatic encephalopathy. Eur. Radiol. 2017;27:4699–4709. doi: 10.1007/s00330-017-4809-9. [DOI] [PubMed] [Google Scholar]
  • 95.Larsen F.S., Strauss G., Møller K., Hansen B.A. Regional cerebral blood flow autoregulation in patients with fulminant hepatic failure. Liver Transplant. 2000;6:795–800. doi: 10.1053/jlts.2000.18705. [DOI] [PubMed] [Google Scholar]
  • 96.Larsen F.S., Ejlersen E., Strauss G., Rasmussen A., Kirkegaard P., Hansen B.A., Secher N. Cerebrovascular metabolic autoregulation is impaired during liver transplantation. Transplantation. 1999;68:1472–1476. doi: 10.1097/00007890-199911270-00007. [DOI] [PubMed] [Google Scholar]
  • 97.Larsen F.S., Olsen K.S., Ejlersen E., Hansen B.A., Paulson O.B., Knudsen G.M. Cerebral blood flow autoregulation and transcranial doppler sonography in patients with cirrhosis. Hepatology (Baltimore, Md.) 1995;22:730–736. [PubMed] [Google Scholar]
  • 98.Jalan R., Olde Damink S.W.M., Hayes P.C., Deutz N.E.P., Lee A. Pathogenesis of intracranial hypertension in acute liver failure: inflammation, ammonia and cerebral blood flow. J. Hepatol. 2004;41:613–620. doi: 10.1016/j.jhep.2004.06.011. [DOI] [PubMed] [Google Scholar]
  • 99.Bjerring P.N., Gluud L.L., Larsen F.S. Cerebral blood flow and metabolism in hepatic Encephalopathy-A meta-analysis. J. Clin. Exp. Hepatol. 2018;8:286–293. doi: 10.1016/j.jceh.2018.06.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Iversen P., Sørensen M., Bak L.K., Waagepetersen H.S., Vafaee M.S., Borghammer P., Mouridsen K., Jensen S.B., Vilstrup H., Schousboe A., Ott P., Gjedde A., Keiding S. Low cerebral oxygen consumption and blood flow in patients with cirrhosis and an acute episode of hepatic encephalopathy. Gastroenterology. 2009;136:863–871. doi: 10.1053/j.gastro.2008.10.057. [DOI] [PubMed] [Google Scholar]
  • 101.Zheng G., Zhang L.J., Wang Z., Qi R.F., Shi D., Wang L., Fan X., Lu G.M. Changes in cerebral blood flow after transjugular intrahepatic portosystemic shunt can help predict the development of hepatic encephalopathy: an arterial spin labeling MR study. Eur. J. Radiol. 2012;81:3851–3856. doi: 10.1016/j.ejrad.2012.07.003. [DOI] [PubMed] [Google Scholar]
  • 102.Larsen F.S., Knudsen G.M., Paulson O.B., Vilstrup H. Cerebral blood flow autoregulation is absent in rats with thioacetamide-induced hepatic failure. J. Hepatol. 1994;21:491–495. doi: 10.1016/s0168-8278(94)80091-x. [DOI] [PubMed] [Google Scholar]
  • 103.Pluta R., Albrecht J. Changes in arterial and cerebral venous blood gases, cerebral blood flow and cerebral oxygen consumption at different stages of thioacetamide-induced hepatogenic encephalopathy in rat. Resuscitation. 1986;14:135–139. doi: 10.1016/0300-9572(86)90117-6. [DOI] [PubMed] [Google Scholar]
  • 104.Bosoi C.R., Rose C.F. Identifying the direct effects of ammonia on the brain. Metab. Brain Dis. 2009;24:95–102. doi: 10.1007/s11011-008-9112-7. [DOI] [PubMed] [Google Scholar]
  • 105.Jayakumar A.R., Rama Rao K.V., Norenberg M.D. Neuroinflammation in hepatic encephalopathy: mechanistic aspects. J. Clin. Exp. Hepatol. 2015;5:S21–28. doi: 10.1016/j.jceh.2014.07.006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106.Zhang L., Tan J., Jiang X., Qian W., Yang T., Sun X., Chen Z., Zhu Q. Neuron-derived CCL2 contributes to microglia activation and neurological decline in hepatic encephalopathy. Biol. Res. 2017;50:26. doi: 10.1186/s40659-017-0130-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Mladenović D., Hrnčić D., Petronijević N., Jevtić G., Radosavljević T., Rašić-Marković A., Puškaš N., Maksić N., Stanojlović O. Finasteride improves motor, EEG, and cellular changes in rat brain in thioacetamide-induced hepatic encephalopathy, American journal of physiology. Gastrointest. Liver Physiol. 2014;307:G931–940. doi: 10.1152/ajpgi.00463.2013. [DOI] [PubMed] [Google Scholar]
  • 108.Jäkel S., Dimou L. Glial cells and their function in the adult brain: a journey through the history of their ablation. Front. Cell. Neurosci. 2017;11:24. doi: 10.3389/fncel.2017.00024. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Albrecht J., Gadamski R., Kuhrt H., Walski M., Reichenbach A. Retinal gliopathy accompanying thioacetamide-induced liver insufficiency: light and electron microscopic observations. Acta Neuropathol. 1998;96:57–66. doi: 10.1007/s004010050860. [DOI] [PubMed] [Google Scholar]
  • 110.Obara-Michlewska M., Pannicke T., Karl A., Bringmann A., Reichenbach A., Szeliga M., Hilgier W., Wrzosek A., Szewczyk A., Albrecht J. Down-regulation of Kir4.1 in the cerebral cortex of rats with liver failure and in cultured astrocytes treated with glutamine: implications for astrocytic dysfunction in hepatic encephalopathy. J. Neurosci. Res. 2011;89:2018–2027. doi: 10.1002/jnr.22656. [DOI] [PubMed] [Google Scholar]
  • 111.Avraham Y., Grigoriadis N.C., Magen I., Poutahidis T., Vorobiav L., Zolotarev O., Ilan Y., Mechoulam R., Berry E.M. Capsaicin affects brain function in a model of hepatic encephalopathy associated with fulminant hepatic failure in mice. Br. J. Pharmacol. 2009;158:896–906. doi: 10.1111/j.1476-5381.2009.00368.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 112.Peeling J., Shoemaker L., Gauthier T., Benarroch A., Sutherland G.R., Minuk G.Y. Cerebral metabolic and histological effects of thioacetamide-induced liver failure. Am. J. Physiol. 1993;265:G572–578. doi: 10.1152/ajpgi.1993.265.3.G572. [DOI] [PubMed] [Google Scholar]
  • 113.Szumanska G., Albrecht J. Lectin histochemistry of the rat brain following thioacetamide-induced hepatic failure. Mol. Chem. Neuropathol. 1997;32:163–177. doi: 10.1007/BF02815174. [DOI] [PubMed] [Google Scholar]
  • 114.Sepehrinezhad A., Zarifkar A., Namvar G., Shahbazi A., Williams R. Astrocyte swelling in hepatic encephalopathy: molecular perspective of cytotoxic edema. Metab. Brain Dis. 2020;35:559–578. doi: 10.1007/s11011-020-00549-8. [DOI] [PubMed] [Google Scholar]
  • 115.Jayakumar A.R., Valdes V., Tong X.Y., Shamaladevi N., Gonzalez W., Norenberg M.D. Sulfonylurea receptor 1 contributes to the astrocyte swelling and brain edema in acute liver failure. Transl. Stroke Res. 2014;5:28–37. doi: 10.1007/s12975-014-0328-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 116.Jia W., Liu J., Hu R., Hu A., Tang W., Li L., Li J. Xiaochaihutang improves the cortical astrocyte edema in thioacetamide-induced rat acute hepatic encephalopathy by activating NRF2 pathway. Front. Pharmacol. 2020;11:382. doi: 10.3389/fphar.2020.00382. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.Rash J.E., Yasumura T., Hudson C.S., Agre P., Nielsen S. Direct immunogold labeling of aquaporin-4 in square arrays of astrocyte and ependymocyte plasma membranes in rat brain and spinal cord. Proc. Natl. Acad. Sci. U.S.A. 1998;95:11981–11986. doi: 10.1073/pnas.95.20.11981. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Mader S., Brimberg L. Aquaporin-4 water channel in the brain and its implication for health and disease. Cells. 2019;8 doi: 10.3390/cells8020090. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Rama Rao K.V., Jayakumar A.R., Tong X., Curtis K.M., Norenberg M.D. Brain aquaporin-4 in experimental acute liver failure. J. Neuropathol. Exp. Neurol. 2010;69:869–879. doi: 10.1097/NEN.0b013e3181ebe581. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Klatzo I. Neuropathological aspects of brain edema*. J. Neuropathol. Exp. Neurol. 1967;26:1–14. doi: 10.1097/00005072-196701000-00001. [DOI] [PubMed] [Google Scholar]
  • 121.Montoliu C., Llansola M., Felipo V. Neuroinflammation and neurological alterations in chronic liver diseases. Neuroimmunol. Neuroinflamm. 2015;2:138–144. [Google Scholar]
  • 122.Huang H.C., Chang C.C., Wang S.S., Chan C.Y., Lee F.Y., Chuang C.L., Hsin I.F., Teng T.H., Lin H.C., Lee S.D. Pravastatin for thioacetamide-induced hepatic failure and encephalopathy. Eur. J. Clin. Invest. 2012;42:139–145. doi: 10.1111/j.1365-2362.2011.02566.x. [DOI] [PubMed] [Google Scholar]
  • 123.Wei D.-D., Wang J.-S., Li M.-H., Guo P.-P., Dong G., Yang M.-H., Kong L.-Y. A pilot study of the onset of hepatic encephalopathy (OHE) in mice induced by thioacetamide and the protective effect of taurine by holistic metabolic characterization. Metabolomics. 2015;11:559–570. [Google Scholar]
  • 124.Hosseini S., Ebrahimi A., Bagheri F., Emami Y., Esmaeilzadeh E., Azarpira N., Ebrahimi S., Ashkani-Esfahani S. Effect of resveratrol on thioacetamide-induced liver damage in rat models. Hepat. Mon. 2020;20 [Google Scholar]
  • 125.Gomides L.F., Marques P.E., Faleiros B.E., Pereira R.V., Amaral S.S., Lage T.R., Resende G.H., Guidine P.A., Foureaux G., Ribeiro F.M., Martins F.P., Fontes M.A., Ferreira A.J., Russo R.C., Teixeira M.M., Moraes M.F., Teixeira A.L., Menezes G.B. Murine model to study brain, behavior and immunity during hepatic encephalopathy. World J. Hepatol. 2014;6:243–250. doi: 10.4254/wjh.v6.i4.243. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126.Tak E., Jung D.-H., Kim S.-H., Park G.-C., Jun D.Y., Lee J., Jung B.-h., Kirchner V.A., Hwang S., Song G.-W., Lee S.-G. Protective role of hypoxia-inducible factor-1α-dependent CD39 and CD73 in fulminant acute liver failure. Toxicol. Appl. Pharmacol. 2017;314:72–81. doi: 10.1016/j.taap.2016.11.016. [DOI] [PubMed] [Google Scholar]
  • 127.Ahmad S., Cameron S., Naz N., Moriconi F. Mediators of hypoxia in a rat model of sterile-induced acute liver injury. Int. J. Clin. Exp. Pathol. 2017;10:11471–11479. [PMC free article] [PubMed] [Google Scholar]
  • 128.Rahman T.M., Hodgson H.J.F. The effects of early and late administration of inhibitors of inducible nitric oxide synthase in a thioacetamide-induced model of acute hepatic failure in the rat. J. Hepatol. 2003;38:583–590. doi: 10.1016/s0168-8278(03)00050-3. [DOI] [PubMed] [Google Scholar]
  • 129.Abo El gheit R.E., Atef M.M., Badawi G.A., Elwan W.M., Alshenawy H.A., Emam M.N. Role of serine protease inhibitor, ulinastatin, in rat model of hepatic encephalopathy: aquaporin 4 molecular targeting and therapeutic implication. J. Physiol. Biochem. 2020;76:573–586. doi: 10.1007/s13105-020-00762-0. [DOI] [PubMed] [Google Scholar]
  • 130.Osada J., Aylagas H., Mirõ-Obradors M.J., Arce C., Palacios-Alaiz E., Cascales M. Effect of acute thioacetamide administration on rat brain phospholipid metabolism. Neurochem. Res. 1990;15:927–931. doi: 10.1007/BF00965914. [DOI] [PubMed] [Google Scholar]
  • 131.Püspök A., Herneth A., Steindl P., Peter F. Hepatic encephalopathy in rats with thioacetamide-induced acute liver failure is not mediated by endogenous benzodiazepines. Gastroenterology. 1993;105:851–857. doi: 10.1016/0016-5085(93)90904-q. [DOI] [PubMed] [Google Scholar]
  • 132.Ashkani-Esfahani S., Bagheri F., Emami Y., Esmaeilzadeh E., Azarpira N., Hassanabadi N., Keshtkar M., Farjam M., Koohi-Hosseinabadi O., Noorafshan A. Protective effects of Co-enzyme Q10 on thioacetamide-induced acute liver damage and its correlation with behavioral, biochemical, and pathological factors. Iran. Red Crescent Med. J. 2016;18 doi: 10.5812/ircmj.29166. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Hajipour S., Sarkaki A., Dianat M., Rashno M., Khorsandi L.S., Farbood Y. The effects of thymoquinone on memory impairment and inflammation in rats with hepatic encephalopathy induced by thioacetamide. Metab. Brain Dis. 2021 doi: 10.1007/s11011-021-00688-6. [DOI] [PubMed] [Google Scholar]
  • 134.Baraka S.M., Saleh D.O., Ghaly N.S., Melek F.R., Gamal el Din A.A., Khalil W.K.B., Said M.M., Medhat A.M. Flavonoids from Barnebydendron riedelii leaf extract mitigate thioacetamide-induced hepatic encephalopathy in rats: the interplay of NF-κB/IL-6 and Nrf2/HO-1 signaling pathways. Bioorg. Chem. 2020;105 doi: 10.1016/j.bioorg.2020.104444. [DOI] [PubMed] [Google Scholar]
  • 135.Yuan D.S., Huang Y.Q., Fu Y.J., Xie J., Huang Y.L., Zhou S.S., Sun P.Y., Tang X.Q. Hydrogen sulfide alleviates cognitive deficiency and hepatic dysfunction in a mouse model of acute liver failure. Exp. Ther. Med. 2020;20:671–677. doi: 10.3892/etm.2020.8680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136.Avraham Y., Israeli E., Gabbay E., Okun A., Zolotarev O., Silberman I., Ganzburg V., Dagon Y., Magen I., Vorobia L., Pappo O., Mechoulam R., Ilan Y., Berry E.M. Endocannabinoids affect neurological and cognitive function in thioacetamide-induced hepatic encephalopathy in mice. Neurobiol. Dis. 2006;21:237–245. doi: 10.1016/j.nbd.2005.07.008. [DOI] [PubMed] [Google Scholar]
  • 137.El Khiat A., El Hiba O., Aitihya M., Tamegart L., Draoui A., El Fari R., Gamrani H. P: 22 Deficit of short working memory in rat with thioacetamide-induced progressive acute hepatic encephalopathy involving serotonin innervation and astroglia dysfunctions. Off. J. Am. Coll. Gastroenterol. ACG. 2019;114:S12. [Google Scholar]
  • 138.Hajipour S., Farbood Y., Dianat M., Rashno M., Khorsandi L.S., Sarkaki A. Thymoquinone improves behavioral and biochemical deficits in hepatic encephalopathy induced by thioacetamide in rats. Neurosci. Lett. 2021;745 doi: 10.1016/j.neulet.2020.135617. [DOI] [PubMed] [Google Scholar]
  • 139.Fujisawa K., Takami T., Matsumoto T., Yamamoto N., Sakaida I. Profiling of the circadian metabolome in thioacetamide-induced liver cirrhosis in mice. Hepatol. Commun. 2017;1:704–718. doi: 10.1002/hep4.1075. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Mladenović D., Hrnčić D., Rašić-Marković A., Macut D., Stanojlović O. The influence of finasteride on mean and relative spectral density of EEG bands in rat model of thioacetamide-induced hepatic encephalopathy. Neurotox. Res. 2016;30:150–158. doi: 10.1007/s12640-016-9610-z. [DOI] [PubMed] [Google Scholar]
  • 141.Mladenović D., Hrnčić D., Rašić-Marković A., Puškaš N., Petrovich S., Stanojlović O. Spectral analysis of thioacetamide-induced electroencephalographic changes in rats. Hum. Exp. Toxicol. 2013;32:90–100. doi: 10.1177/0960327112456312. [DOI] [PubMed] [Google Scholar]
  • 142.Abdelsalam R.M., Rizk H.A., Masoud M.A., Mansour H.A.E. 2015. Lactulose and Donepezil Ameliorate Thioacetamide-Induced Hepatic Encephalopathy in Rats. [Google Scholar]
  • 143.Kircheis G., Wettstein M., Vom Dahl S., Häussinger D. Clinical efficacy of L-ornithine–L-aspartate in the management of hepatic encephalopathy. Metab. Brain Dis. 2002;17:453–462. doi: 10.1023/a:1021934607762. [DOI] [PubMed] [Google Scholar]
  • 144.Jiang Q., Jiang X.H., Zheng M.H., Chen Y.P. l‐Ornithine‐l‐aspartate in the management of hepatic encephalopathy: a meta‐analysis. J. Gastroenterol. Hepatol. 2009;24:9–14. doi: 10.1111/j.1440-1746.2008.05582.x. [DOI] [PubMed] [Google Scholar]
  • 145.Neal R.A., Halpert J. Toxicology of thiono-sulfur compounds. Annu. Rev. Pharmacol. Toxicol. 1982;22:321–339. doi: 10.1146/annurev.pa.22.040182.001541. [DOI] [PubMed] [Google Scholar]
  • 146.Najmi A.K., Pillai K.K., Pal S.N., Akhtar M., Aqil M., Sharma M. Effect of l-ornithine l-aspartate against thioacetamide-induced hepatic damage in rats. Indian J. Pharmacol. 2010;42:384–387. doi: 10.4103/0253-7613.71926. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 147.Najmi A.K., Pillai K.K., Pal S.N., Ahmad A., Nazmi A.S., Akhtar M. Ornithine aspartate attenuates thioacetamide induced hepatic encephalopathy through GABA-benzodiazepine receptors. Int. J. Pharmacol. Res. 2014;4:134–137. [Google Scholar]
  • 148.Volk M.L., Burne R., Guérin A., Shi S., Joseph G.J., Heimanson Z., Ahmad M. Hospitalizations and healthcare costs associated with rifaximin versus lactulose treatment among commercially insured patients with hepatic encephalopathy in the United States. J. Med. Econ. 2021;24:202–211. doi: 10.1080/13696998.2021.1877148. [DOI] [PubMed] [Google Scholar]
  • 149.Sharma B.C., Sharma P., Lunia M.K., Srivastava S., Goyal R., Sarin S. A randomized, double-blind, controlled trial comparing rifaximin plus lactulose with lactulose alone in treatment of overt hepatic encephalopathy. Am. J. Gastroenterol. 2013;108:1458–1463. doi: 10.1038/ajg.2013.219. [DOI] [PubMed] [Google Scholar]
  • 150.Fadillioglu E., Gursul C., Iraz M. Effects of caffeic acid phenethyl ester on thioacetamide-induced hepatic encephalopathy in rats. Prog. Neuropsychopharmacol. Biol. Psychiatry. 2010;34:1440–1445. doi: 10.1016/j.pnpbp.2010.07.034. [DOI] [PubMed] [Google Scholar]
  • 151.Heibashy M., Mazen G., Ibrahim M. The levels of cytokines and S 100β which associated with the pathogenesis of hepatic encephalopathy in rats: role of lactulose and N-Acetylcysteine in its treatment. Isotope Radiat. Res. 2012;44:771–788. [Google Scholar]
  • 152.Saeed R.M.A., Ahmed H.H., Saleh A.A.S., Ahmed Y.S. Curative role of lactulose, L-carnitine, alpha-lipoic acid and combination of L-carnitine and alpha-lipoic acid in a rat model of acute hepatic encephalopathy: biochemical observations. Trop. J. Pharm. Res. 2017;16:2161–2168. [Google Scholar]
  • 153.Bass N.M., Mullen K.D., Sanyal A., Poordad F., Neff G., Leevy C.B., Sigal S., Sheikh M.Y., Beavers K., Frederick T. Rifaximin treatment in hepatic encephalopathy. N. Engl. J. Med. 2010;362:1071–1081. doi: 10.1056/NEJMoa0907893. [DOI] [PubMed] [Google Scholar]
  • 154.Harputluoglu M.M., Demirel U., Gul M., Temel I., Gursoy S., Selcuk E.B., Aladag M., Bilgic Y., Gunduz E., Seckin Y. Effects of rifaximin on bacterial translocation in thioacetamide-induced liver injury in rats. Inflammation. 2012;35:1512–1517. doi: 10.1007/s10753-012-9465-2. [DOI] [PubMed] [Google Scholar]
  • 155.Jones M.C., Lasak-Myall T., Abdelhak T.M., Varelas P.N. Indomethacin for treatment of refractory intracranial hypertension secondary to acute liver failure. Am. J. Health-system Pharm. 2015;72:1020–1025. doi: 10.2146/ajhp140539. [DOI] [PubMed] [Google Scholar]
  • 156.Lee W.M., Hynan L.S., Rossaro L., Fontana R.J., Stravitz R.T., Larson A.M., Davern T.J., 2nd, Murray N.G., McCashland T., Reisch J.S., Robuck P.R. Intravenous N-acetylcysteine improves transplant-free survival in early stage non-acetaminophen acute liver failure. Gastroenterology. 2009;137:856–864. doi: 10.1053/j.gastro.2009.06.006. 864.e851. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 157.Tofteng F., Larsen F.S. The effect of indomethacin on intracranial pressure, cerebral perfusion and extracellular lactate and glutamate concentrations in patients with fulminant hepatic failure. J. Cerebral Blood Flow Metabol. 2004;24:798–804. doi: 10.1097/01.WCB.0000125648.03213.1D. [DOI] [PubMed] [Google Scholar]
  • 158.Clemmesen J.O., Hansen B.A., Larsen F.S. Indomethacin normalizes intracranial pressure in acute liver failure: a twenty‐three‐year‐old woman treated with indomethacin. Hepatology (Baltimore, Md.) 1997;26:1423–1425. doi: 10.1002/hep.510260608. [DOI] [PubMed] [Google Scholar]
  • 159.Heard K.J. Acetylcysteine for acetaminophen poisoning. New Engl. J. Med. 2008;359:285–292. doi: 10.1056/NEJMct0708278. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 160.Downs J.W., Cumpston K.L., Kershner E.K., Troendle M.M., Rose S.R., Wills B.K. Clinical outcome of massive acetaminophen overdose treated with standard-dose N-acetylcysteine. Clin. Toxicol. 2021:1–8. doi: 10.1080/15563650.2021.1887493. [DOI] [PubMed] [Google Scholar]
  • 161.Nabi T., Nabi S., Rafiq N., Shah A. Role of N-acetylcysteine treatment in non-acetaminophen-induced acute liver failure: a prospective study. Saudi J. gastroenterol. 2017;23:169–175. doi: 10.4103/1319-3767.207711. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 162.Sotelo N., de los Ángeles Durazo M., Gonzalez A., Dhanakotti N. Early treatment with N-acetylcysteine in children with acute liver failure secondary to hepatitis A. Ann. Hepatol. 2009;8:353–358. [PubMed] [Google Scholar]
  • 163.Saleem A.F., Abbas Q., Haque A. Use of N-acetylcysteine in children with fulminant hepatic failure caused by acute viral hepatitis. JCPSP. 2015;25:354. [PubMed] [Google Scholar]
  • 164.Kortsalioudaki C., Taylor R.M., Cheeseman P., Bansal S., Mieli-Vergani G., Dhawan A. Safety and efficacy of N-acetylcysteine in children with non-acetaminophen-induced acute liver failure. Liver Transplant. 2008;14:25–30. doi: 10.1002/lt.21246. [DOI] [PubMed] [Google Scholar]
  • 165.James L.P., Simpson P.M., Farrar H.C., Kearns G.L., Wasserman G.S., Blumer J.L., Reed M.D., Sullivan J.E., Hinson J.A. Cytokines and toxicity in acetaminophen overdose. J. Clin. Pharmacol. 2005;45:1165–1171. doi: 10.1177/0091270005280296. [DOI] [PubMed] [Google Scholar]
  • 166.Bémeur C., Vaquero J., Desjardins P., Butterworth R.F. N-Acetylcysteine attenuates cerebral complications of non-acetaminophen-induced acute liver failure in mice: antioxidant and anti-inflammatory mechanisms. Metabol. Brain Dis. 2010;25:241–249. doi: 10.1007/s11011-010-9201-2. [DOI] [PubMed] [Google Scholar]
  • 167.Akbay A., Cinar K., Uzunalimoğlu O., Eranil S., Yurdaydin C., Bozkaya H., Bozdayi M. Serum cytotoxin and oxidant stress markers in N-acetylcysteine treated thioacetamide hepatotoxicity of rats. Hum Exp Toxicol. 1999;18:669–676. doi: 10.1191/096032799678839518. [DOI] [PubMed] [Google Scholar]
  • 168.Harputluoglu M.M., Demirel U., Ciralik H., Temel I., Firat S., Ara C., Aladag M., Karincaoglu M., Hilmioglu F. Protective effects of Gingko biloba on thioacetamide-induced fulminant hepatic failure in rats. Hum. Exp. Toxicol. 2006;25:705–713. doi: 10.1177/0960327106073827. [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

No datasets were generated during the study.


Articles from Toxicology Reports are provided here courtesy of Elsevier

RESOURCES