Minireviews Open Access
Copyright ©The Author(s) 2022. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Clin Cases. Mar 26, 2022; 10(9): 2687-2699
Published online Mar 26, 2022. doi: 10.12998/wjcc.v10.i9.2687
Animal models applied to acute-on-chronic liver failure: Are new models required to understand the human condition?
Jaciara Fernanda Gomes Gama, Liana Monteiro da Fonseca Cardoso, Luiz Anastacio Alves, Laboratory of Cellular Communication, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Rio de Janeiro 21045900, Rio de Janeiro, Brazil
Jussara Machado Lagrota-Candido, Laboratory of Immunopathology, Department of Immunobiology, Fluminense Federal University, Niteroi 24210-200, Rio de Janeiro, Brazil
ORCID number: Jaciara Fernanda Gomes Gama (0000-0003-3945-8644); Liana Monteiro da Fonseca Cardoso (0000-0002-8567-3276); Jussara Machado Lagrota-Candido (0000-0002-6573-561X); Luiz Anastacio Alves (0000-0002-0785-7272).
Author contributions: Gama JFG general bibliographic review on the subject and main writing of the article; Cardoso LMDF manuscript preparation and final correction; Lagrota-Cândido JM critical revisions and final approval of the version; Alves LA proposal idealization and final approval of the version of the article for publication.
Supported by FIOCRUZ and FAPERJ Fundação Carlos Chagas Filho de Amparo à Pesquisa do Rio de Janeiro - "Redes de Pesquisa em Saúde no Estado do Rio de Janeiro", No. E-26/010.002422/2019.
Conflict-of-interest statement: The authors declare no conflict of interest.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Luiz Anastacio Alves, MD, PhD, Academic Research, Professor, Research Scientist, Laboratory of Cellular Communication, Oswaldo Cruz Institute, Oswaldo Cruz Foundation, Av. Brazil, 4365 Manguinhos, Rio de Janeiro 21045900, Rio de Janeiro, Brazil. alveslaa@gmail.com
Received: October 14, 2021
Peer-review started: October 14, 2021
First decision: December 12, 2021
Revised: December 20, 2021
Accepted: February 12, 2022
Article in press: February 12, 2022
Published online: March 26, 2022

Abstract

The liver is a multifaceted organ; its location and detoxifying function expose this organ to countless injuries. Acute-on-chronic failure liver (ACLF) is a severe syndrome that affects the liver due to acute decompensation in patients with chronic liver disease. An infection environment, ascites, increased liver enzymes and prothrombin time, encephalopathy and fast-evolving multiorgan failure, leading to death, usually accompany this. The pathophysiology remains poorly understand. In this context, animal models become a very useful tool in this regard, as understanding; the disease may be helpful in developing novel therapeutic methodologies for ACLF. However, although animal models display several similarities to the human condition, they do not represent all ACLF manifestations, resulting in significant challenges. An initial liver cirrhosis framework followed by the induction of an acute decompensation by administering lipopolysaccharide and D-GaIN, potentiating liver damage supports the methodologies applied to induce experimental ACLF. The entire methodology has been described mostly for rats. Nevertheless, a quick PubMed database search indicates about 30 studies concerning ACFL models and over 1000 regarding acute liver failure models. These findings demonstrate the clear need to establish easily reproducible ACFL models to elucidate questions about this quickly established and often fatal syndrome.

Key Words: Liver disease, Acute-on-chronic liver failure, Cirrhosis, Acute decompensate event, Translational study, Animal models

Core Tip: The liver is a multifaceted organ; its location and detoxifying function expose it to countless injuries. Acute-on-chronic failure liver (ACLF) is a severe syndrome that affects the liver due to acute decompensation in patients with chronic liver disease. Animal models become a very useful tool in this regard. However, although they display several similarities to the human condition, they do not represent all manifestations, resulting in significant challenges. A quick PubMed database search indicates about 30 studies concerning ACLF models. These findings demonstrate the need to establish easily reproducible models to elucidate questions about this quickly established and often fatal syndrome.



INTRODUCTION

The liver is a multifaceted organ that performs various functions, including protein and amino acid metabolism and plasma protein secretion, in addition to lipid oxidation and drug and xenobiotic blood detoxification[1]. Hepatic tissue is susceptible to countless injuries that may lead to liver cirrhosis. Most chronic liver lesions that culminate in cirrhosis processes are reversible, depending on their etiology, the amount of affected liver tissue and appropriate treatment, although this may lead to serious complication or be fatal in some patients[2]. Deaths caused by liver disease have increased by around 400% in the United Kingdom since the 1970s up to 2018[3]. About 2 million people die each year due to liver disease complications, 50% of which are due to liver cirrhosis. This is mostly due to alcohol consumption and non-alcoholic fatty liver diseases in the western world and hepatitis B in China and Asian countries[4]. Acute-on-chronic liver failure (ACLF) was diagnosed in 30% of all cases among 1343 hospitalized patients presenting liver cirrhosis according to a European study carried out between February and September 2011, with a mortality rate of 32.9% at 28 d and 51.2% at 90 d. Patients with liver cirrhosis may experience acute decompensating that leads to ACLF, characterized by ascites, hepatic encephalopathy, gastrointestinal bleeding or a combination of these symptoms[5,6]. The molecular, cellular, and immune mechanisms reported in patients with liver cirrhosis that develop ACLF, however, remain unclear. Therefore, effective therapeutic methodologies become a challenge, generating high costs, with liver transplants comprising the only effective treatment to date.

Animal models concerning hepatic disease have been very useful in preclinical research for decades, comprise an alternative in the understanding of ACLF pathophysiology, as they are reproducible, and are able to adequately mimic some ACLF events. However, many models do not faithfully reproduce human disease and require special management for understanding specific hypothesis. In this context, this review aims to provide an overview of the main animal models used in ACLF research, alongside their pros and cons. Furthermore, pathological events that do reproduce human ACLF are also discussed.

WHAT IS KNOWN ABOUT ACUTE-ON-CHRONIC LIVER FAILURE PATHOPHYSIOLOGY?

The vast majority of patients referred to specialist hepatological centers suffer from acute deterioration during chronic liver disease. Two important conditions are observed in patients with known chronic liver disease who exhibit acute decompensation, namely acutely decompensated cirrhosis and ACLF. The first is a widely accepted condition and refers to the development of ascites, encephalopathy, gastrointestinal hemorrhage, or any combination of these disorders in patients with cirrhosis[5,7]. The second, ACLF, identifies patients with known or unknown chronic liver disease who develop rapid liver function deterioration and high short-term mortality after an acute insult. The definitions of ACLF differ, with most addressing the role of both hepatic and extra-hepatic precipitating events and including extra-hepatic organ failures[8,9]. ACLF, a term suggested by Jalan and Williams in 2002[10], emerged from studies indicating the development of a syndrome associated with a high risk of short-term death (death < 28 d after hospital admission) in patients presenting acutely decompensated cirrhosis. Three major features characterize this syndrome, namely intense systemic inflammation, frequently displaying a close temporal relationship with pro-inflammatory precipitating events (e.g., infections or alcoholic hepatitis) associated with single- or multiple-organ failure. However, the first ACLF definition was only established by a consensus of the Asian Pacific Association for the study of the liver (APASL) in 2008. Unlike other definitions, this definition does not include extra-hepatic organ failures[7]. Thus, ACLF was defined as an acute hepatic insult in patients with chronic liver disease resulting in jaundice (total bilirubin ≥ 5 mg/dL) and coagulopathy (INR ≥ 1.5), complicated within four weeks by ascites and/or encephalopathy[7]. Thereafter, the North American Consortium for the Study of End-Stage Liver Disease associated ACLF with a 30-d mortality rate of 41% compared to 7% concerning acute decompensation without ACLF[11]. Meanwhile, the European Association Study Liver (EASL) defines ACLF as an acute decompensated liver insult in patients with cirrhosis or chronic liver disease that can often lead to sepsis due to bacterial infection, increasing the requirement for intensive care and resulting in a high 28-d mortality rate (≥ 15%)[12].

The prevalence of ACLF ranges from 24% to 40% of hospitalized patients with liver cirrhosis. ACLF may develop in patients with previously compensated or decompensated cirrhosis, as well as in patients presenting the underlying chronic liver disease without cirrhosis[13]. Excessive alcohol consumption is also important in an ACLF context, as alcoholic hepatitis results in a high mortality rate and is one of the causes of acute-on-chronic liver injury[14]. Furthermore, over 50% of patients presenting alcoholic cirrhosis exhibit decompensated disease and, in the US, alcohol-associated liver diseases can lead to an indication for liver transplantation[15,16]. In contrast, the reactivation of hepatitis B virus (HBV) is the leading cause of ACLF in the Asian region, with high prevalence where HBV-related acute-on-chronic liver failure (HBV-ACLF) accounts for over 70% of ACLF[17,18]. However, early diagnosis and prevention measures through long-term HBV infection suppression with antiviral agents (such as lamivudine, tenofovir, entecavir or telbuvidine) or sustained eradication of hepatitis C virus (HCV) infection in patients with compensated or decompensated cirrhosis can decrease mortality and prevent the development of HBV-ACLF and HCV-related acute-on-chronic liver failure (HCV-ACLF) in this region[19,20]. Patients with chronic hepatitis B or HBV-related cirrhosis are at risk of developing ACLF, with multi-organ failure and high short-term mortality[21]. HBV reactivation could be either a spontaneous setting of treatment cessation[22] or due to intensive chemotherapy or immunosuppressive therapy[23,24], treatment related[25] or reactivation of the occult HBV infection by rituximab (anti-CD20)-based chemotherapy[26-28] or immune restoration after highly active antiretroviral therapy for HIV[29,30]. Similarly, HCV infection reactivation has also been reported, mainly following immune suppressive therapy[31,32]. Drugs such as anti-tuberculosis drugs, methotrexate and antiretroviral drugs in HIV/AIDS-infected individuals have been implicated in triggering liver injury, particularly in the setting of underlying chronic liver disease due to HBV or HCV[31,33-35]. Drugs are seen as a precipitating factor in ACLF, although databases on concerning drugs as an acute insult leading to ACLF are extremely scarce. This factor limits the study and knowledge of the effects of certain drugs and medications in ACLF development. This indicates the need for further data and assessments concerning models on hepatic injury caused by different herbal and medicinal preparations in cirrhosis patients[8].

ACLF pathogenesis is still poorly understood, and will depend on the origin of the condition, the number of organ failures and the patient's past medical history. It can, however, be characterized as a systemic inflammatory syndrome, whose evolution and pathophysiology are directly associated with the involved immune mechanisms, such as those in response to danger associated molecular patterns and pathogen associated molecular patterns (DAMPs and PAMPs, respectively), cytokine production and the inflammatory profile[36]. Patients with hepatitis B (HB) diagnosed with ACLF exhibit an exacerbated innate and adaptive inflammatory response evidenced by increased ROS (reactive oxygen species) production by macrophages and neutrophils, increased TLR4 expression and high blood cytokine levels. The induction of cytokine storms has been associated with interleukin 33 (IL-33) up-regulation, an important DAMP associated with disease severity[36,37]. The source of this inflammatory response exacerbation, however, remains unknown. Another event consists in the formation of the NLRP3 inflammasome, as reported in a study on 70 HB patients presenting ACLF. This is composed of a NOD-like receptor family, pyrin domain containing 3 (NLRP3), an intracellular PAMP receptor responsible for activating Caspase-1 that, consequently, cleaves pro-inflammatory IL-1β and IL-18 cytokines[38]. Immune system cells involved in the inflammatory response play a key role in the ACLF-caused mechanism, as the neutrophil-leucocyte ratio is an indicator of ACLF death severity[39]. In addition, the large liver cell death that occurs in ACLF may be closely related to mitochondrial damage, mainly in organ and multi-organ failure. In three CANONICAL studies in cirrhotic patients with ACLF was observed changes in mitochondrial markers associated with ACLF follow up[40-42]. The increase of fingerprints in metabolomics assays demonstrated the fatty acylcarnitines (FA) was raised, suggesting impairment in β-mitochondrial oxidation that can cause decreasing of the FA-derived energy in peripheral organs, and then, cell death and organ failure[40]. In addition, immunometabolism was required with the increased of GDF15 and FGF21, as wells as the up-regulation of inflammatory cytokines (e.g. MIP-1 α/β, MCP-1, IL-1ra and IL-6) and ultra-structural changes with cristae rarefication in mitochondrial morphology of the peripheral blood mononuclear cells[41,42]. These findings together indicate the impairment of mitochondrial function through the energy expenditure by systemic inflammation. Xue and coworkers demonstrated that mitofusin2 (Mtn-2) can regulated the autophagy and apoptosis levels, as well as decreasing reactive oxygen species (ROS)[43].

Another important issue concerning the knowledge and studies of the events that lead to ACLF development in humans is directly related to the current pandemic scenario caused by the new coronavirus disease 2019 (COVID-19). The hallmark of ACLF is excessive systemic inflammation, and patients with ACLF exhibit higher levels of inflammatory markers and pro-inflammatory cytokines—IL-6, IL-1β, and IL-8. Systemic inflammation inducers can be exogenous or endogenous and viruses have been described previously as triggering inflammation[13]. A cytokine storm has been reported in patients with COVID-19, characterized by increased IL-2, IL-7, G-SCF and TNF-α[44]. Thus, it is believed that the excessive inflammatory response associated with COVID-19 can serve as a trigger for ACLF in patients with underlying chronic liver disease, which could justify the increase in liver disease patient deaths[45]. However, other mechanisms may also contribute to ACLF development in COVID-19 patients, such as hypoxic changes and iatrogenic causes such as drugs and ventilation, exacerbating underlying liver disease[46,47]. A case study reported the development of ACLF precipitated by severe acute respiratory syndrome coronavirus-2 infection (SARS-CoV-2) in a patient with HBV-related cirrhosis without no previous anti-viral treatment. The authors suggest that the SARS-CoV-2 infection induced systemic inflammatory response syndrome, and the resultant immune dysregulation could have precipitated ACLF, in turn. Since the patient had not been on nucleoside analogs treatment for HBV prior to admission, it is possible that the ACLF was caused by HBV flare in a context of uncontrolled inflammation and dysbalance of innate and adaptive immune responses triggered by the SARS-CoV-2 infection[48]. This highlights the importance of the treatment in patients with HBV and other chronic liver disease in the current pandemic status worldwide. Nevertheless, long-term follow-up clinical studies are required to explore the potential relationship between ACLF development in COVID-19 patients[45].

Despite these relevant facts concerning ACLF pathophysiology, the low number of patients assessed to date, its poor diagnosis and the use of different methodological analyses are still a challenge in understanding this condition. Focused research on strategies to prevent and treat this potentially reversible syndrome are, thus, paramount. Therefore, animal models comprise a helpful tool, as they are reproducible, easy to manage and are able of mimicking several ACLF mechanisms.

ANIMAL MODELS IN LIVER RESEARCH

As described previously, acute and chronic liver diseases are frequent and potentially lethal conditions, displaying a high worldwide prevalence[49]. The development of new therapeutic strategies, drugs and the definition of effective potential biomarkers depends on understanding liver damage pathogenesis and progression, which can be investigated by making use of suitable animal models. Liver injury is highly complex and the absence of adequate animal models able to faithfully reproduce human liver disease characteristics limits the understanding of the mechanisms underlying this condition and treatment[50,51]. Rodent animal models are preferred in preclinical liver research, mainly in ACLF assessments, due to easy to maintain and breed in captivity, the genetic similarity to human, similar liver morphology and simple management concerning the use of genetic manipulation tools[52]. In contrast, large animal models, like pigs, previously used mostly in acute liver failure modeling, now play an important role in the assessment of various acute and chronic liver diseases[51]. Non-human primate models like baboons (Papio hamadryas) are also widely applied due proximity of the human condition, for example, as alcohol-caused liver disease (ALD) models. They displaying significant similarity to the pathogenesis of this disease, i.e., inducing fatty liver and fibrosis, and high annexin-2 Levels, directly associated to the final stage of the disease in humans, as well as fibrinolysis and high risk of bleeding[53]. Furthermore, Rhesus monkeys also significantly reproduce the pathology, biochemistry markers and genetic expression of human ALD[54]. However, after a deep and thorough data review, we observed that no primate non-human models have been reported in the literature concerning ACLF research, and that rodent (rats and mice) models are the most assessed (Table 1).

Table 1 Current experimental acute-on-chronic liver failure animal models.
Animal model
Liver chronic induction
Fibrosis establishment
AD
Immune response observed
Clear methodologyrandomized study
Ref.
Female Wistar ratsHSA subcutaneal and i.v. YesLPS/D-GaIN i.p.Inflammatory cytokines Unclear methodology regarding randomization1, n = 60Wang et al[56], 2012; Gao et al[83], 2016
Male NZ rabbitCCl4 in oil i.p.YesNot describedNot describedUnclear methodologyZhu et al[71], 2013
Female Wistar ratsHSA subcutaneal and i.v. YesLPS/D-GaIN i.p.Inflammatory pathway through cytokines increase Unclear methodology regarding randomization1, n = 15Yang et al[57], 2014
Female SD ratsCCl4 in oil i.p.YesLPS/D-GaIN i. p.Not describedUnclear methodologyZhang et al[67], 2014
Female Wistar ratsBD ligatureYesischemia 70% liver reductionInflammatory cytokines Unclear methodology regarding randomization, n = 10Hu et al[58], 2014
Male SD ratsCCl4 in oil i.p.YesLPS/D-GaIN i.p.Treg/Th17 imbalanceUnclear methodology regarding randomization1, n = 80Ni et al[59], 2017
Male Wistar ratsCholestasis induced by BD ligatureYesNot describedCD45+ up regulationUnclear methodology regarding randomization1, n = 20Gilsanz et al[60], 2017
Male Wistar ratsPS i.p. YesLPS/D-GaIN i.p.Inflammatory cytokines Unclear methodology regarding randomization1, n = 10 e 60Li et al[61], 2017
Wistar ratsBD ligatureYesLPS i.p.Not describedUnclear methodology regarding randomization1, n = 6/8Nataj, et al[80], 2018
Wistar or SD ratsCCl4 or TAA or BD ligatureYesLPS i.p.Neutrophil infiltration and NETUnclear methodology regarding randomization1, n = 6-9Tripathi et al [69],2018
Male C57BL/6JCCl4 in olive oil i.p.YesEthyl alcoholInflammatory cytokines and neutrophil infiltrationUnclear methodology regarding randomization1 Furuya, et al[14], 2019; Schwarzkopf et al[73], 2020
Male SD ratsCCl4 in oil i.p.YesLPS/D-GaIN i.p.Not describedUnclear methodology regarding randomization1, n = 10Xue et al[43], 2019
Male SD ratsCCl4 in oil i.p.YesLPS/D-GaIN i.p.Not described†§ Unclear methodology regarding randomization1, n = 10Xie et al[68], 2019
Male C57BL/6JCCl4 in oil i.p.YesKlebsiella pneumonia i.p.Increase in IL-6 and IFN-γ pathwayUnclear methodology regarding randomization1, n = 4-8Xiang, et al[70], 2020
SD rats BD ligationYesLPS i.p.Inflammatory cytokines Unclear methodologyMonteiro et al[62], 2021
Animal models as important contributors to the understanding of ACLF

Animal models are useful in understanding the mechanisms surrounding immune responses and multiorgan failure in inflammatory and systemic syndromes[55]. Many studies have attempted to comprehend this inflammatory scenario[56-62]. With the aim of mimicking the human disease, techniques able to induce ACLF, resulting in a chronic liver insult and, finally, a precipitating event, have been developed. It is important to note that the main precipitating events comprise the reactivation of hepatitis viral or acute hepatitis A virus infection, acute alcoholic hepatitis or acute bacterial infection in Asian patients and alcoholism and bacterial infection that aggravate chronic cirrhosis conditions in the western world[20]. In this context, most ACLF animal models induce chronic cirrhosis and a posterior acute insult, usually through the administration of Lipopolysaccharide (LPS), LPS/D-Galactosamine hydrochloride (D-GaIN) or ethyl alcohol (EtOH), in order to reproduce events of bacterial infection (Figure 1). However, few animal ACLF models have been established to date (described in the next section).

Figure 1
Figure 1 Acute-on-chronic liver failure animals models based on a literature review. Liver injury models applying chemical, biological and surgery induction leading to cirrhosis and subsequent acute decompensation with lipopolysaccharide or Galactosamine hydrochloride or both associated, Klebsiella pneumonia i.p. injection or Ethyl alcohol (EtOH), and CS surgery to Acute-on-chronic Liver Failure (ACLF) induction in rats or acute decompensation with EtOH in mice. Intense fibrosis and Aspartate aminotransferase, Alanine aminotransferase serum levels, an inflammatory response and impairment of mitochondrial function are observed. Unclear and non-reproducible data concerning the ACLF rabbit model induced by Carbon tetrachloride are described. SD: Sprague Dawley; HAS: Human albumin serum; PS: Porcine serum; TAA: Thioacetamide Administration; CS: Cecal slurry; BDL: Bile duct ligation; CCl4: Carbon tetrachloride; AST: Aspartate aminotransferase; ALT: Alanine aminotransferase; PT: Prothrombin time; ACLF: Acute-on-chronic liver failure.
Chemically induced ACLF animal models: Carbon tetrachloride in combination with LPS/GaIN

A widely applied model used to induce chronic lesions consist in the use of carbon tetrachloride (CCl4) as a hepatocellular damage inducer. This model leads to liver steatosis due to CCl4 covalent binding to cell metabolites, resulting in Ca2+ sequestration and K+/Na2+ channel imbalances in an anaerobic environment and cytochrome P450 (CYP450) action[63]. Liver CCl4 metabolism occurs through carbonyl chloride and free radicals formation (i.e., +CCl3), the latter binding to protein and lipids in the cell cytosol, even though no acid nucleic binding occurs[64]. Thus, toxicity due to CCl4 administrations is similar to alcohol-induced toxicity. Furthermore, fibrosis following significant liver destruction is also noted in this model, evidenced by increased hydroxiproline content and liver function failure, comprising increased alanine aminotransferase (ALT) and aspartate transaminase (AST) activities[63,65]. Aiming to mimic ACLF events, CCl4 in combination with the administration of D-GalN, with or without LPS, is the mostly used to induce chronic liver injury (Table 1). LPS is a bacterial endotoxin capable of activating Kupffer cells and stimulating TNF-α and an immune response by the NF-κB pathway, while D-galactosamine is able to potentiate this response by depleting the uridine nucleotides and interfering in protein synthesis, leading to acute insults as a precipitating event[66]. The major challenge verified in this model consists in determine differences between CCl4 and LPS/GaIN administration time and doses for ACLF induction. In one study, female Sprague-Dawley (SD) rats (180-200 g) were administered intraperitoneal (i.p.) CCl4 dissolved in olive oil (10%) twice a week for ten weeks. After cirrhosis confirmation, the animals received 0.70 g/kg body weight (BW) D-GaIN i.p.[67]. ACLF was confirmed due to increased AST and ALT activities and higher bilirrubin and plasma ammonia levels associated with changes consistent with necrosis as revealed by histological analyses. However, modified CCl4 doses were administered according to the liver function index and animal body weight[67]. In another study, ACLF was induced in male SD rats (150 - 170 g) by the i.p. injection of 1.5 mL/kg of CCl4 dissolved in vegetable oil (40%) twice a week for ten weeks. After cirrhosis establishment, LPS (100 µg/kg) and D-GaIN (0.5 g/kg) were i.p. administered[43]. In addition to the same biochemistry and histopathology events observed as in the first study, a macroscopic analysis also detected granules and severe liver surface adhesions, as well as increased B-cell lymphoma 2 (BCL-2) and BCL-2-associated X protein apoptosis regulator genes, which could probably be handled with the control of mitochondrial damage via Mtn-2[43,67]. Immune responses are also a target for investigation. Thus, another study evaluated male SD rats (160-180 g) received i.p. injections comprising CCl4 in peanut oil (1/1 w/v) once every three days for two months (1.5 mL/kg BW during the 1st month and 2 mL/kg BW during the 2nd month). Subsequently, 500 mg/Kg BW D-GalN and 80 μg/Kg BW LPS were i.p. administered, resulting in a Treg/Th17 ratio imbalance, necrosis and fibrotic tissue as revealed by histological assessments[59]. In addition to intense inflammatory response, the HIF-1α shows an important role in development of ACLF and the mitochondrial function was impairment. These findings was observed in a study with ACLF induced by i.p. CCl4 in vegetable oil (1.5 mL/kg) 3 days in 4 wk, in male SD rats (approximately 200 g), and then i.p. of LPS (100 μg/kg) in combination with D-GaIN (0.5 g/kg)[68]. These models show enhanced histopathological evidences; AST and ALT levels and prothrombin time (PT) was raised, as well as the liver mitochondrial ultrastructure was damage with nuclear fragmentation[68]. Its corroborated the energy expenditure due the oxidative stress and systemic inflammation[40,42]. Tripathi and collaborators reported that CCl4 is capable of inducing advanced chronic liver injury, as Wistar rats (50-75 g) subjected to CCl4 inhalation and receiving phenobarbital (0.3 g/L) in drinking water presented micronodular cirrhosis with ascites after 15 or 16 wk. Subsequently, LPS (from obtained from Escherichia coli, O111:B4 – 1 mg/kg) was i.p. injected to induce an acute insult in the decompensated cirrhosis group 4 h prior to a hemodynamic study and 24 h prior in the compensated cirrhosis group[69]. LPS administration was more effective in the decompensated cirrhosis animals, resulting in portal hypertension and increased neutrophil infiltration and neutrophil extracellular trap (NET), as well as a high inflammatory response and aggravated fibrosis[69]. Male C57BL/6J mice have also been reported as an ACLF model with disease induce by CCl4 i.p. (0.2 mL/Kg/twice a week, for 8 wk). Then, a double dose CCl4 injection (0.4 mL/kg) was administered. After that, klebsiella pneumonia (strain 43816- ATCC, Manassas, VA) was i.p. injected in order to mimic bacterial infection in ACLF[70]. This model was able to develop the main observed pathophysiological features of ACLF in humans, such as chronic/acute liver injury, bacterial infection and multiorgan failures. Rabbits have also been described as an ACLF model mimicking the condition of this disease. In one investigation, New Zealand Rabbits injected i.p. with CCl4 displayed changes in biochemical markers after 10 wk, including ALT, AST, albumin and protrombin levels, as well as altered liver morphology, reflecting fibrosis consistent with cirrhosis, revealed by HE- and Masson-stained analyses, following the administration of intravenously-injected D-GaIN[71]. It is important to note that the methodology applied in the model described in a previous study carried out with male New Zealand rabbits (2.73 ± 0.05 kg and 100 d old) that did not induce ACLF, only cirrhosis, by the intragastric administration of CCl4 once a week during 16 wk[71,72]. Therefore, rabbits have not yet been established as an ACLF model.

Another experimental model comprises the ACLF induction by chronic i.p. CCl4 injection and an acute insult with EtOH administered via an intragastric cannula, resulting in a condition resembling human alcoholic hepatitis[14,73]. Briefly, 12-wk-old C57BL/6J male mice were categorized into a control group receiving an olive oil i.p. injection and an experimental group receiving a CCl4 i.p. injection (0.2 mL/kg) twice a week during 6 wk to induce liver fibrosis. The experimental group was then submitted to surgical intragastric intubation, beginning at EtOH 16 g/kg/day, gradually increasing to 25 g/kg/day. The CCl4 associated with EtOH was able to mimic an acute-on-chronic injury, resulting in intense inflammation, hypoxia and opportunist infection by E. coli and Candida sp., as well as chromatin and DNA modifications with epigenetic gene dysregulation. This model is, however, difficult to reproduce, due to the applied surgery and an approximate mortality rate of 30%.

It is noteworthy that CCl4 has been widely applied to induce hepatotoxicity in experimental studies, although it is toxic. According to the National Institute of Health chemistry database (PubChem) CCl4 is a clear, colorless, volatile and very stable chlorinated hydrocarbon and its inhalation (> 200-250 ppm CCl4 for > 4 h) leads to accumulation mainly in fatty tissues and may result in nervous system depression, lung, kidney and liver damage and increased cancer susceptibility[74]. The intoxication level and affected tissue, however, may vary with exposure time and amount. Although no direct data concerning humans are available, animal experiments have indicated that toxic CCl4 metabolites are produced in reactions catalyzed by CYP450 oxigenases (e.g., CYP2E1; CYP3A)[74,75].

Chemically induced ACLF animal model: Thioacetamide in combination with LPS

Administration i.p of Thioacetamide (TAA) also has been used to induce ACLF In one assessment, male SD rats (150-200 g) received 250 mg/kg of TAA dissolved in saline twice a week for ten weeks to induce compensated cirrhosis without ascites development[69,76]. The animals subsequently received i.p. LPS injection obtained from Escherichia coli O111:B4 – 1 mg/kg) 24 h prior to a hemodynamic study. Increased liver enzymes, portal hypertension worsening and fibrosis were noted, similar to the human condition[69]. This is the only study to date applying TAA alongside LPS to induce ACLF in an animal model as an alternative methodology to CCL4 administration. However, TAA, a crystalline solid, is highly toxic following inhalation or dermal contact and is carcinogenic to humans[77], requiring care to avoid researcher, laboratory and environmental contamination.

Surgery protocol to reproduce the human ACLF condition

In addition to chemical ACLF induction, surgery can also be used to establish ACLF. An obstructive jaundice model was developed in female Wistar rats (200-250 g) through bile duct ligation (BDL) by the dissociation of the common bile duct of the hepatoduodenal ligament[58], through a double ligature using surgical threads at the proximal end of the common bile duct, followed by a layered suturing of the abdominal wall to close the abdominal cavity[58,78]. Reduced-size hepatic ischemia/reperfusion injury was performed, leading to cholestasis (0 min, 15 min, 30 min and 45 min of ischemia), followed by a choledochoduodenostomy to relieve this condition. A reduction of the 70% in the hepatic area was observed. This model was able to reproduce certain ACLF pathological features, such as increased AST, ALT and total bilirubin levels after the obstruction surgery[58]. Another study demonstrated that microsurgery applied to induce cholestasis also mimics certain pathological ACLF events. For example, male Wistar rats (200-400 g) have been submitted to an extra-hepatic biliary tract dissection through hepatic lobe and lobe ligament sectioning[60,79]. This technique is most applied in human secondary biliary hepatic cirrhosis and atresia assessments, but is also capable of inducing ACLF syndrome events, such as histological and biochemistry aspects and an international normalized ratio (INR) of up to 1.5. In addition, an increase in CD45+ leucocyte infiltration was also observed[60]. Recent studies have suggested the bile duct obstruction Wistar rat model followed by the i.p. injection of a single dose of LPS (1 mg/kg, extracted from Salmonella typhimurium)[80] or bile duct-obstructed SD rats followed by an i.p. injection of LPS (0.03 mg/kg BW). It was obtained from Klebsiella pneumoniae 28 d after cirrhosis as confirmed ACFL models presenting systemic disease. Alternatively, to induce acute decompensation, animals received a single intravenous LPS dose obtained from Escherichia coli O111:B4 - 6.25 mg/kg BW 3 wk after bile duct obstruction[62]. Using a similar methodology, common bile duct ligature was performed in SD rats for 28 d, with cirrhosis displaying ascites established on the 25th day. The animals were then injected with LPS 1 mg/kg obtained from Escherichia coli (O111:B4)[69]. An increase in plasma enzyme levels and splenomegaly were observed, mimicking pathological events and inducing the inflamassome (e.g. IL-1β; IL-18), as noted in the human syndrome[62,80]. Although these methodologies result in similar conditions to the human disease, the need for surgical procedures and animal survival maintenance make them more challenging and costly, requiring previous surgical knowledge.

Last but not least: Albumin serum-induced ACLF

Studies have demonstrated that the albumin, the most abundant protein in the human serum, metabolized in the liver and secreted to the plasma can be an alternative to treat decompensated cirrhosis, although it can become hepatotoxic when an imbalance in albumin levels occurs. Thus, albumin may induce cirrhosis events both when interacting with LPS or when undergoing irreversible alkalization by drug metabolization[81,82]. Therefore, human albumin serum (HSA) is a potential model for liver damage. In order to develop HSA-induced ACLF, female Wistar rats (180-220 g) were challenged with a subcutaneal HSA injection diluted in saline (8 g/L) and the same volume of incomplete Freund’s adjuvant for four times (0.5 mL containing 4 mg HSA). Next, 4 mg HSA were injected into the animal tail veins twice/week for six weeks[56]. After the establishment of immune cirrhosis, the animals received 400 mg/kg of D-GaIN and 100 μg/kg LPS, both i.p., inducing acute decompensation in chronic liver disease, leading to changes in liver histology and increased serum enzymes and several inflammatory markers, such as IL-6, IL-18 and HMGB-1. It is important to note that both NF-κB and TNF-α play an important role in HMGB-1-mediated responses[56,57,83,84], similar to human ACLF. Since the HSA ACLF model results in high mortality rates, it was developed a rat model using porcine serum (PS), resulting in better induction of pathophysiological events[56,61]. Male Wistar rats (120-150 g) received 0.5 mL i.p. PS twice/week for 11 wk and developed liver fibrosis as revealed by hepatic hydroxyproline levels. Then, following the induction of immune cirrhosis, LPS was i.p. injected at 50 μg/kg followed by 600 mg/kg i.p. of D-GaIN thirty minutes later, in order to induce acute liver insult on chronic liver damage[61]. This ACLF model presented several similarities with human ACLF, such as increased inflammatory marker (TNF-α, IL-6), plasma enzyme (AST, ALT) and ammonia levels, as well as higher prothrombin times consistent with coagulation function impairment[20,61]. Therefore, ACLF induced by serum albumin showed to be a good model and comprises an alternative to better understand ACLF physiopathology. However, the PS model may be more adequate, it induces a closer response to the human condition, easier to manipulate and results in lower mortality rates compared to the other mentioned models.

DISADVANTAGES AND CHALLENGES OF ACLF ANIMAL MODELS

Due to the complexity of liver injury, the understanding of underlying liver disease mechanisms and their treatment has been limited by the lack of satisfactory animal models. Currently, no model has been able to completely capture the corresponding human acute and chronic liver disorder[52,61,85].

The ideal ACLF model should combine bacterial infection and high short-term mortality. As described previously, several existing ACLF models have been developed by combining chronic and acute liver injury[70]. Chronic injury is most commonly induced by the injection of CCl4 or via BDL surgery, whereas acute injury is induced by the injection of D-GaIN/LPS. The principle of these models is to reproduce the bi-factorial disease profile comprising chronic liver injury, which leads to the development of progressive liver fibrosis, and a precipitating event inducing further organ injury, resulting in ACLF and considerable mortality. This is not, however, entirely consistent with ACLF pathogenesis, and the surgery required for the BDL model is difficult. The clinical situation is often more complex, and different modulating factors may occur concurrently or sequentially[86]. Typically, 50% of patients develop bacterial infection as an ACLF complication, although the (initial) precipitating event was non-inflammatory[87].

The significant challenge to develop an ACLF model is the ability to unite all the clinical characteristics observed in humans, as this is a multifactorial disease with multiple precipitators and complications and, therefore, varying disease phenotypes and organ failures, making it almost impossible to develop a single experimental model capable of triggering all of the most important clinical features[86]. In a recent study by Xiang and coworkers (2020), the authors developed a new ACLF model that could sequentially reproduce three important clinical ACLF disease factors. To this end, a severe liver injury model was prepared by combining chronic injury (CCl4 injection), acute hepatic insult (injection of a CCl4 double dose), and systemic bacterial infection (i.p. injection of bacteria Klebsiella pneumonia). The findings indicate that this severe liver injury model developed acute-on-chronic liver injury, bacterial infection, multi-organ injury, and high mortality, some of the features of clinical ACLF. The authors highlight that the single bacterial infection step is crucial in inducing multi-organ failure in this model, as chronic-plus-acute liver injury did not drive the full course of ACLF in mice without bacterial infection[70]. In contrast, Schwarzkopf and coworkers developed a model combining chronic liver disease (CCl4/EtOH or CYP2D6-linked adenovirus (ADV)-induced autoimmune hepatitis) with different precipitating events [two EtOH binges or i.p. polymicrobial infection by cecal slurry (CS)]. After 7 wk of CCl4/EtOH, ACLF was induced with two alcohol binges (alcohol gavage with 31.5% Vol.) with an interval of 3 days between binges. Mice mortality was observed, as well as systemic inflammation and significant elevation of serum ALT levels alongside other ACLF features[73]. According to the authors, polymicrobial sepsis by CS is closer to human infection-triggered ACLF than the K. pneumonia injection employed by Xiang[70,73]. These variabilities in current data also significantly interfere with the development of a standardized ACLF animal model. Furthermore, it is not yet possible to identify all ACLF precipitants, as over 40% of patients who develop ACLF exhibit no known precipitant, requiring further knowledge of ACLF activation events[22].

CONCLUSION

Animal models are helpful in understanding human diseases and play a relevant role in preclinical research, as they are capable of reproducing major pathophysiological events that occur in humans. ACLF mechanisms remain unclear, displaying a poor diagnosis and high mortality rates worldwide, it becomes relevant to highlight current animal models and how they can elucidate ACLF mechanisms. These models are also crucial in testing new drugs and novel bioengineering and genetic therapies, as the only effective therapy to date is liver transplantation. In this context, few viable livers are available and the quick clinical evolution of this condition was observed. Currently, rats are the most widely applied models (Figure 1), as their maintenance and management is simple compared to other models (Table 1). Furthermore, ACLF induced by PS combined with LPS/D-GaIN seems to be the most adequate methodology in the establishment of an ACLF animal model. However, further studies are required, as controversies concerning model designs, reproducibility and safety are still noted. In addition, the animal models are incapable to reproduce all manifestation of ACLF condition due to different causes of pathology. For example, the HBV infection model remains a challenge, because of use of transgenic mice that were capable to support virus replication or virus products. Thus, understanding current models in order to establish a safe and easily reproducible model to study complementary alternatives to liver transplantation is paramount.

Footnotes

Provenance and peer review: Unsolicited article; Externally peer reviewed.

Peer-review model: Single blind

Specialty type: Gastroenterology and hepatology

Country/Territory of origin: Brazil

Peer-review report’s scientific quality classification

Grade A (Excellent): 0

Grade B (Very good): B

Grade C (Good): C, C

Grade D (Fair): D

Grade E (Poor): 0

P-Reviewer: Çalışkan ARR, Li Z, Pham TTT S-Editor: Liu JH L-Editor: A P-Editor: Liu JH

References
1.  Trefts E, Gannon M, Wasserman DH. The liver. Curr Biol. 2017;27:R1147-R1151.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 340]  [Cited by in F6Publishing: 746]  [Article Influence: 124.3]  [Reference Citation Analysis (0)]
2.  Tapper EB, Parikh ND. Mortality due to cirrhosis and liver cancer in the United States, 1999-2016: observational study. BMJ. 2018;362:k2817.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 337]  [Cited by in F6Publishing: 497]  [Article Influence: 82.8]  [Reference Citation Analysis (0)]
3.   British Liver Trust. The alarming impact of liver disease in the UK [Internet]. Cited 23 June 2021. Available from: https://britishlivertrust.org.uk/about-us/media-centre/statistics/.  [PubMed]  [DOI]  [Cited in This Article: ]
4.  Asrani SK, Devarbhavi H, Eaton J, Kamath PS. Burden of liver diseases in the world. J Hepatol. 2019;70:151-171.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1382]  [Cited by in F6Publishing: 1789]  [Article Influence: 357.8]  [Reference Citation Analysis (0)]
5.  Moreau R, Jalan R, Gines P, Pavesi M, Angeli P, Cordoba J, Durand F, Gustot T, Saliba F, Domenicali M, Gerbes A, Wendon J, Alessandria C, Laleman W, Zeuzem S, Trebicka J, Bernardi M, Arroyo V; CANONIC Study Investigators of the EASL–CLIF Consortium. Acute-on-chronic liver failure is a distinct syndrome that develops in patients with acute decompensation of cirrhosis. Gastroenterology. 2013;144:1426-1437, 1437.e1.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1720]  [Cited by in F6Publishing: 1889]  [Article Influence: 171.7]  [Reference Citation Analysis (5)]
6.  Arroyo V, Moreau R, Jalan R. Acute-on-Chronic Liver Failure. N Engl J Med. 2020;382:2137-2145.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 219]  [Cited by in F6Publishing: 319]  [Article Influence: 79.8]  [Reference Citation Analysis (2)]
7.  Sarin SK, Kumar A, Almeida JA, Chawla YK, Fan ST, Garg H, de Silva HJ, Hamid SS, Jalan R, Komolmit P, Lau GK, Liu Q, Madan K, Mohamed R, Ning Q, Rahman S, Rastogi A, Riordan SM, Sakhuja P, Samuel D, Shah S, Sharma BC, Sharma P, Takikawa Y, Thapa BR, Wai CT, Yuen MF. Acute-on-chronic liver failure: consensus recommendations of the Asian Pacific Association for the study of the liver (APASL). Hepatol Int. 2009;3:269-282.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 630]  [Cited by in F6Publishing: 610]  [Article Influence: 40.7]  [Reference Citation Analysis (0)]
8.  Sarin SK, Choudhury A, Sharma MK, Maiwall R, Al Mahtab M, Rahman S, Saigal S, Saraf N, Soin AS, Devarbhavi H, Kim DJ, Dhiman RK, Duseja A, Taneja S, Eapen CE, Goel A, Ning Q, Chen T, Ma K, Duan Z, Yu C, Treeprasertsuk S, Hamid SS, Butt AS, Jafri W, Shukla A, Saraswat V, Tan SS, Sood A, Midha V, Goyal O, Ghazinyan H, Arora A, Hu J, Sahu M, Rao PN, Lee GH, Lim SG, Lesmana LA, Lesmana CR, Shah S, Prasad VGM, Payawal DA, Abbas Z, Dokmeci AK, Sollano JD, Carpio G, Shresta A, Lau GK, Fazal Karim M, Shiha G, Gani R, Kalista KF, Yuen MF, Alam S, Khanna R, Sood V, Lal BB, Pamecha V, Jindal A, Rajan V, Arora V, Yokosuka O, Niriella MA, Li H, Qi X, Tanaka A, Mochida S, Chaudhuri DR, Gane E, Win KM, Chen WT, Rela M, Kapoor D, Rastogi A, Kale P, Sharma CB, Bajpai M, Singh V, Premkumar M, Maharashi S, Olithselvan A, Philips CA, Srivastava A, Yachha SK, Wani ZA, Thapa BR, Saraya A, Shalimar, Kumar A, Wadhawan M, Gupta S, Madan K, Sakhuja P, Vij V, Sharma BC, Garg H, Garg V, Kalal C, Anand L, Vyas T, Mathur RP, Kumar G, Jain P, Pasupuleti SSR, Chawla YK, Chowdhury A, Song DS, Yang JM, Yoon EL; APASL ACLF Research Consortium (AARC) for APASL ACLF working Party. Acute-on-chronic liver failure: consensus recommendations of the Asian Pacific association for the study of the liver (APASL): an update. Hepatol Int. 2019;13:353-390.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 459]  [Cited by in F6Publishing: 431]  [Article Influence: 86.2]  [Reference Citation Analysis (0)]
9.  Mahmud N, Kaplan DE, Taddei TH, Goldberg DS. Incidence and Mortality of Acute-on-Chronic Liver Failure Using Two Definitions in Patients with Compensated Cirrhosis. Hepatology. 2019;69:2150-2163.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 98]  [Cited by in F6Publishing: 127]  [Article Influence: 25.4]  [Reference Citation Analysis (0)]
10.  Jalan R, Williams R. Acute-on-chronic liver failure: pathophysiological basis of therapeutic options. Blood Purif. 2002;20:252-261.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 209]  [Cited by in F6Publishing: 215]  [Article Influence: 10.2]  [Reference Citation Analysis (1)]
11.  Gustot T, Moreau R. Acute-on-chronic liver failure vs. traditional acute decompensation of cirrhosis. J Hepatol. 2018;69:1384-1393.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 33]  [Cited by in F6Publishing: 34]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
12.  Jalan R, Perricone G, Moreau R, Arroyo V, Williams R. Acute-on-Chronic Liver Failure: A New Disease or an Old One Hiding in Plain Sight? Clin Liver Dis (Hoboken). 2020;15:S45-S51.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 11]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
13.  Hernaez R, Solà E, Moreau R, Ginès P. Acute-on-chronic liver failure: an update. Gut. 2017;66:541-553.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 360]  [Cited by in F6Publishing: 397]  [Article Influence: 56.7]  [Reference Citation Analysis (0)]
14.  Furuya S, Cichocki JA, Konganti K, Dreval K, Uehara T, Katou Y, Fukushima H, Kono H, Pogribny IP, Argemi J, Bataller R, Rusyn I. Histopathological and Molecular Signatures of a Mouse Model of Acute-on-Chronic Alcoholic Liver Injury Demonstrate Concordance With Human Alcoholic Hepatitis. Toxicol Sci. 2019;170:427-437.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
15.  Morgan MY. The prognosis and outcome of alcoholic liver disease. Alcohol Alcohol Suppl. 1994;2:335-343.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Julien J, Ayer T, Bethea ED, Tapper EB, Chhatwal J. Projected prevalence and mortality associated with alcohol-related liver disease in the USA, 2019-40: a modelling study. Lancet Public Health. 2020;5:e316-e323.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 79]  [Article Influence: 19.8]  [Reference Citation Analysis (0)]
17.  Chen T, Yang Z, Choudhury AK, Al Mahtab M, Li J, Chen Y, Tan SS, Han T, Hu J, Hamid SS, Huei LG, Ghazinian H, Nan Y, Chawla YK, Yuen MF, Devarbhavi H, Shukla A, Abbas Z, Sahu M, Dokmeci AK, Lesmana LA, Lesmana CRA, Xin S, Duan Z, Guo W, Ma K, Zhang Z, Cheng Q, Jia J, Sharma BC, Sarin SK, Ning Q. Complications constitute a major risk factor for mortality in hepatitis B virus-related acute-on-chronic liver failure patients: a multi-national study from the Asia-Pacific region. Hepatol Int. 2019;13:695-705.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 33]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
18.  Cui YL, Yan F, Wang YB, Song XQ, Liu L, Lei XZ, Zheng MH, Tang H, Feng P. Nucleoside analogue can improve the long-term prognosis of patients with hepatitis B virus infection-associated acute on chronic liver failure. Dig Dis Sci. 2010;55:2373-2380.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 65]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
19.  Yang J, Chen G, Chen X, Zhang H, Jiang D, Yang G. Initial combination anti-viral therapy with lamivudine and adefovir dipivoxil decreases short-term fatality rate of hepatitis-B-virus-related acute-on-chronic liver failure. Virol J. 2015;12:97.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 10]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
20.  Arroyo V, Moreau R, Kamath PS, Jalan R, Ginès P, Nevens F, Fernández J, To U, García-Tsao G, Schnabl B. Acute-on-chronic liver failure in cirrhosis. Nat Rev Dis Primers. 2016;2:16041.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 236]  [Cited by in F6Publishing: 269]  [Article Influence: 33.6]  [Reference Citation Analysis (0)]
21.  Zhao RH, Shi Y, Zhao H, Wu W, Sheng JF. Acute-on-chronic liver failure in chronic hepatitis B: an update. Expert Rev Gastroenterol Hepatol. 2018;12:341-350.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 61]  [Cited by in F6Publishing: 63]  [Article Influence: 10.5]  [Reference Citation Analysis (0)]
22.  Cullaro G, Sharma R, Trebicka J, Cárdenas A, Verna EC. Precipitants of Acute-on-Chronic Liver Failure: An Opportunity for Preventative Measures to Improve Outcomes. Liver Transpl. 2020;26:283-293.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
23.  Xu L, Tu Z, Xu G, Wang Y, Pan W, Zhan X, Luo Q, Huang Y, Chen J, Huang A. Epirubicin directly promotes hepatitis B virus (HBV) replication in stable HBV-expressing cell lines: a novel mechanism of HBV reactivation following anticancer chemotherapy. Mol Med Rep. 2014;9:1345-1350.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 35]  [Article Influence: 3.5]  [Reference Citation Analysis (0)]
24.  Mikulska M, Nicolini L, Signori A, Rivoli G, Del Bono V, Raiola AM, Di Grazia C, Dominietto A, Varaldo R, Ghiso A, Bacigalupo A, Viscoli C. Hepatitis B reactivation in HBsAg-negative/HBcAb-positive allogeneic haematopoietic stem cell transplant recipients: risk factors and outcome. Clin Microbiol Infect. 2014;20:O694-O701.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 60]  [Cited by in F6Publishing: 66]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
25.  Lim SG, Wai CT, Rajnakova A, Kajiji T, Guan R. Fatal hepatitis B reactivation following discontinuation of nucleoside analogues for chronic hepatitis B. Gut. 2002;51:597-599.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 90]  [Cited by in F6Publishing: 96]  [Article Influence: 4.4]  [Reference Citation Analysis (0)]
26.  Tsutsumi Y, Yamamoto Y, Shimono J, Ohhigashi H, Teshima T. Hepatitis B virus reactivation with rituximab-containing regimen. World J Hepatol. 2013;5:612-620.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 20]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
27.  Kusumoto S, Tanaka Y, Mizokami M, Ueda R. Strategy for preventing hepatitis B reactivation in patients with resolved hepatitis B virus infection after rituximab-containing chemotherapy. Hepatology. 2014;60:765-766.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
28.  Martin ST, Cardwell SM, Nailor MD, Gabardi S. Hepatitis B reactivation and rituximab: a new boxed warning and considerations for solid organ transplantation. Am J Transplant. 2014;14:788-796.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 26]  [Cited by in F6Publishing: 29]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
29.  Magalhães RK, Ferreira D, Salgado M, Pedroto I. Hepatitis B virus reactivation after chemotherapy in an HIV-positive patient. J Clin Gastroenterol. 2013;47:565-566.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 6]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
30.  Pei R, Grund S, Verheyen J, Esser S, Chen X, Lu M. Spontaneous reactivation of hepatitis B virus replication in an HIV coinfected patient with isolated anti-Hepatitis B core antibodies. Virol J. 2014;11:9.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
31.  Huang M, Huang L. Reactivation of hepatitis C viral infection after treatment with infliximab. J Clin Gastroenterol. 2014;48:189-190.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
32.  Sagnelli E, Pisaturo M, Stanzione M, Messina V, Alessio L, Sagnelli C, Starace M, Pasquale G, Coppola N. Clinical presentation, outcome, and response to therapy among patients with acute exacerbation of chronic hepatitis C. Clin Gastroenterol Hepatol. 2013;11:1174-1180.e11.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 43]  [Article Influence: 3.9]  [Reference Citation Analysis (0)]
33.  Chalasani N, Bonkovsky HL, Fontana R, Lee W, Stolz A, Talwalkar J, Reddy KR, Watkins PB, Navarro V, Barnhart H, Gu J, Serrano J; United States Drug Induced Liver Injury Network. Features and Outcomes of 899 Patients With Drug-Induced Liver Injury: The DILIN Prospective Study. Gastroenterology. 2015;148:1340-52.e7.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 495]  [Cited by in F6Publishing: 550]  [Article Influence: 61.1]  [Reference Citation Analysis (0)]
34.  Lo Re V 3rd, Carbonari DM, Lewis JD, Forde KA, Goldberg DS, Reddy KR, Haynes K, Roy JA, Sha D, Marks AR, Schneider JL, Strom BL, Corley DA. Oral Azole Antifungal Medications and Risk of Acute Liver Injury, Overall and by Chronic Liver Disease Status. Am J Med. 2016;129:283-91.e5.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 52]  [Article Influence: 6.5]  [Reference Citation Analysis (0)]
35.  Hoffmann CJ, Charalambous S, Thio CL, Martin DJ, Pemba L, Fielding KL, Churchyard GJ, Chaisson RE, Grant AD. Hepatotoxicity in an African antiretroviral therapy cohort: the effect of tuberculosis and hepatitis B. AIDS. 2007;21:1301-1308.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 126]  [Article Influence: 7.4]  [Reference Citation Analysis (0)]
36.  Wang Y, Chen C, Qi J, Wu F, Guan J, Chen Z, Zhu H. Altered PGE2-EP2 is associated with an excessive immune response in HBV-related acute-on-chronic liver failure. J Transl Med. 2019;17:93.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
37.  Du XX, Shi Y, Yang Y, Yu Y, Lou HG, Lv FF, Chen Z, Yang Q. DAMP molecular IL-33 augments monocytic inflammatory storm in hepatitis B-precipitated acute-on-chronic liver failure. Liver Int. 2018;38:229-238.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 27]  [Article Influence: 4.5]  [Reference Citation Analysis (0)]
38.  Zhao Q, Wu CS, Fang Y, Qian Y, Wang H, Fan YC, Wang K. Glucocorticoid Regulates NLRP3 in Acute-On-Chronic Hepatitis B Liver Failure. Int J Med Sci. 2019;16:461-469.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 8]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
39.  Bernsmeier C, Cavazza A, Fatourou EM, Theocharidou E, Akintimehin A, Baumgartner B, Dhar A, Auzinger G, Thursz M, Bernal W, Wendon JA, Karvellas CJ, Antoniades CG, McPhail MJW. Leucocyte ratios are biomarkers of mortality in patients with acute decompensation of cirrhosis and acute-on-chronic liver failure. Aliment Pharmacol Ther. 2020;52:855-865.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 23]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
40.  Moreau R, Clària J, Aguilar F, Fenaille F, Lozano JJ, Junot C, Colsch B, Caraceni P, Trebicka J, Pavesi M, Alessandria C, Nevens F, Saliba F, Welzel TM, Albillos A, Gustot T, Fernández J, Moreno C, Baldassarre M, Zaccherini G, Piano S, Montagnese S, Vargas V, Genescà J, Solà E, Bernal W, Butin N, Hautbergue T, Cholet S, Castelli F, Jansen C, Steib C, Campion D, Mookerjee R, Rodríguez-Gandía M, Soriano G, Durand F, Benten D, Bañares R, Stauber RE, Gronbaek H, Coenraad MJ, Ginès P, Gerbes A, Jalan R, Bernardi M, Arroyo V, Angeli P; CANONIC Study Investigators of the EASL Clif Consortium;  Grifols Chair;  European Foundation for the Study of Chronic Liver Failure (EF Clif). Blood metabolomics uncovers inflammation-associated mitochondrial dysfunction as a potential mechanism underlying ACLF. J Hepatol. 2020;72:688-701.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 197]  [Article Influence: 49.3]  [Reference Citation Analysis (1)]
41.  Ruiz-Margáin A, Pohlmann A, Ryan P, Schierwagen R, Chi-Cervera LA, Jansen C, Mendez-Guerrero O, Flores-García NC, Lehmann J, Torre A, Macías-Rodríguez RU, Trebicka J. Fibroblast growth factor 21 is an early predictor of acute-on-chronic liver failure in critically ill patients with cirrhosis. Liver Transpl. 2018;24:595-605.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 23]  [Cited by in F6Publishing: 25]  [Article Influence: 4.2]  [Reference Citation Analysis (0)]
42.  Zhang IW, Curto A, López-Vicario C, Casulleras M, Duran-Güell M, Flores-Costa R, Colsch B, Aguilar F, Aransay AM, Lozano JJ, Hernández-Tejero M, Toapanta D, Fernández J, Arroyo V, Clària J. Mitochondrial dysfunction governs immunometabolism in leukocytes of patients with acute-on-chronic liver failure. J Hepatol. 2021;.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 49]  [Article Influence: 24.5]  [Reference Citation Analysis (0)]
43.  Xue R, Zhu X, Jia L, Wu J, Yang J, Zhu Y, Meng Q. Mitofusin2, a rising star in acute-on-chronic liver failure, triggers macroautophagy via the mTOR signalling pathway. J Cell Mol Med. 2019;23:7810-7818.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
44.  Godeau D, Petit A, Richard I, Roquelaure Y, Descatha A. Return-to-work, disabilities and occupational health in the age of COVID-19. Scand J Work Environ Health. 2021;47:408-409.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 97]  [Article Influence: 32.3]  [Reference Citation Analysis (0)]
45.  Khan MU, Mushtaq K, Alkaabi SR. Acute-on-Chronic Liver Failure: Possibly the Main Culprit of Increased Mortality in COVID-19 Patients with Liver Disease. Gastroenterology. 2021;160:1894-1895.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 8]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
46.  Muhović D, Bojović J, Bulatović A, Vukčević B, Ratković M, Lazović R, Smolović B. First case of drug-induced liver injury associated with the use of tocilizumab in a patient with COVID-19. Liver Int. 2020;40:1901-1905.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 68]  [Cited by in F6Publishing: 73]  [Article Influence: 18.3]  [Reference Citation Analysis (0)]
47.  Nardo AD, Schneeweiss-Gleixner M, Bakail M, Dixon ED, Lax SF, Trauner M. Pathophysiological mechanisms of liver injury in COVID-19. Liver Int. 2021;41:20-32.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 165]  [Cited by in F6Publishing: 222]  [Article Influence: 74.0]  [Reference Citation Analysis (2)]
48.  Chen W, Zhang Q, Li S, Liang R, Liang H, Yu Y, Qiu C, Lv J, Zhang W.   Acute-on-Chronic Liver Failure Precipitated by Severe Acute Respiratory Syndrome Coronavirus 2 Infection in a Patient with Hepatitis B Virus-Related Cirrhosis: A Case Report. Infectious Diseases & Immunity 2021; 1: 169–173.  [PubMed]  [DOI]  [Cited in This Article: ]
49.  Marcellin P, Kutala BK. Liver diseases: A major, neglected global public health problem requiring urgent actions and large-scale screening. Liver Int. 2018;38 Suppl 1:2-6.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 264]  [Cited by in F6Publishing: 287]  [Article Influence: 47.8]  [Reference Citation Analysis (0)]
50.  Nevzorova YA, Boyer-Diaz Z, Cubero FJ, Gracia-Sancho J. Animal models for liver disease - A practical approach for translational research. J Hepatol. 2020;73:423-440.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 94]  [Cited by in F6Publishing: 122]  [Article Influence: 30.5]  [Reference Citation Analysis (0)]
51.  Malečková A, Tonar Z, Mik P, Michalová K, Liška V, Pálek R, Rosendorf J, Králíčková M, Třeška V. Animal models of liver diseases and their application in experimental surgery. Rozhl Chir. 2019;98:100-109.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Liu Y, Meyer C, Xu C, Weng H, Hellerbrand C, ten Dijke P, Dooley S. Animal models of chronic liver diseases. Am J Physiol Gastrointest Liver Physiol. 2013;304:G449-G468.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 146]  [Cited by in F6Publishing: 139]  [Article Influence: 12.6]  [Reference Citation Analysis (0)]
53.  Seth D, Leo MA, McGuinness PH, Lieber CS, Brennan Y, Williams R, Wang XM, McCaughan GW, Gorrell MD, Haber PS. Gene expression profiling of alcoholic liver disease in the baboon (Papio hamadryas) and human liver. Am J Pathol. 2003;163:2303-2317.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 62]  [Cited by in F6Publishing: 65]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
54.  Wang H, Tan T, Wang J, Niu Y, Yan Y, Guo X, Kang Y, Duan Y, Chang S, Liao J, Si C, Ji W, Si W. Rhesus monkey model of liver disease reflecting clinical disease progression and hepatic gene expression analysis. Sci Rep. 2015;5:15019.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 12]  [Cited by in F6Publishing: 9]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
55.  Moreau R. The Pathogenesis of ACLF: The Inflammatory Response and Immune Function. Semin Liver Dis. 2016;36:133-140.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 67]  [Article Influence: 8.4]  [Reference Citation Analysis (0)]
56.  Wang LW, Wang LK, Chen H, Fan C, Li X, He CM, Gong ZJ. Ethyl pyruvate protects against experimental acute-on-chronic liver failure in rats. World J Gastroenterol. 2012;18:5709-5718.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 25]  [Cited by in F6Publishing: 23]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
57.  Yang F, Li X, Wang LK, Wang LW, Han XQ, Zhang H, Gong ZJ. Inhibitions of NF-κB and TNF-α result in differential effects in rats with acute on chronic liver failure induced by d-Gal and LPS. Inflammation. 2014;37:848-857.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 37]  [Cited by in F6Publishing: 41]  [Article Influence: 4.6]  [Reference Citation Analysis (0)]
58.  Hu C, Shen S, Zhang A, Ren B, Lin F. The liver protective effect of methylprednisolone on a new experimental acute-on-chronic liver failure model in rats. Dig Liver Dis. 2014;46:928-935.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 6]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
59.  Ni S, Li S, Yang N, Tang X, Zhang S, Hu D, Lu M. Deregulation of Regulatory T Cells in Acute-on-Chronic Liver Failure: A Rat Model. Mediators Inflamm. 2017;2017:1390458.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 2.0]  [Reference Citation Analysis (0)]
60.  Gilsanz C, Aller MA, Fuentes-Julian S, Prieto I, Blázquez-Martinez A, Argudo S, Fernández-Delgado J, Beleña J, Arias J, De Miguel MP. Adipose-derived mesenchymal stem cells slow disease progression of acute-on-chronic liver failure. Biomed Pharmacother. 2017;91:776-787.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 11]  [Cited by in F6Publishing: 13]  [Article Influence: 1.9]  [Reference Citation Analysis (0)]
61.  Li F, Miao L, Sun H, Zhang Y, Bao X, Zhang D. Establishment of a new acute-on-chronic liver failure model. Acta Pharm Sin B. 2017;7:326-333.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
62.  Monteiro S, Grandt J, Uschner FE, Kimer N, Madsen JL, Schierwagen R, Klein S, Welsch C, Schäfer L, Jansen C, Claria J, Alcaraz-Quiles J, Arroyo V, Moreau R, Fernandez J, Bendtsen F, Mehta G, Gluud LL, Møller S, Praktiknjo M, Trebicka J. Differential inflammasome activation predisposes to acute-on-chronic liver failure in human and experimental cirrhosis with and without previous decompensation. Gut. 2021;70:379-387.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 30]  [Cited by in F6Publishing: 48]  [Article Influence: 16.0]  [Reference Citation Analysis (0)]
63.  Boll M, Weber LW, Becker E, Stampfl A. Mechanism of carbon tetrachloride-induced hepatotoxicity. Hepatocellular damage by reactive carbon tetrachloride metabolites. Z Naturforsch C J Biosci. 2001;56:649-659.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 167]  [Cited by in F6Publishing: 155]  [Article Influence: 6.7]  [Reference Citation Analysis (0)]
64.  Shah H, Hartman SP, Weinhouse S. Formation of carbonyl chloride in carbon tetrachloride metabolism by rat liver in vitro. Cancer Res. 1979;39:3942-3947.  [PubMed]  [DOI]  [Cited in This Article: ]
65.  Dong S, Chen QL, Song YN, Sun Y, Wei B, Li XY, Hu YY, Liu P, Su SB. Mechanisms of CCl4-induced liver fibrosis with combined transcriptomic and proteomic analysis. J Toxicol Sci. 2016;41:561-572.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 84]  [Cited by in F6Publishing: 126]  [Article Influence: 15.8]  [Reference Citation Analysis (0)]
66.  Kemelo MK, Wojnarová L, Kutinová Canová N, Farghali H. D-galactosamine/Lipopolysaccharide-induced hepatotoxicity downregulates sirtuin 1 in rat liver: role of sirtuin 1 modulation in hepatoprotection. Physiol Res. 2014;63:615-623.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 28]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
67.  Zhang Y, Chen XM, Sun DL. Effects of coencapsulation of hepatocytes with adipose-derived stem cells in the treatment of rats with acute-on-chronic liver failure. Int J Artif Organs. 2014;37:133-141.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 1.6]  [Reference Citation Analysis (0)]
68.  Xie F, Dong J, Zhu Y, Wang K, Liu X, Chen D, Meng Q. HIF1a Inhibitor Rescues Acute-on-Chronic Liver Failure. Ann Hepatol. 2019;18:757-764.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
69.  Tripathi DM, Vilaseca M, Lafoz E, Garcia-Calderó H, Viegas Haute G, Fernández-Iglesias A, Rodrigues de Oliveira J, García-Pagán JC, Bosch J, Gracia-Sancho J. Simvastatin Prevents Progression of Acute on Chronic Liver Failure in Rats With Cirrhosis and Portal Hypertension. Gastroenterology. 2018;155:1564-1577.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 72]  [Cited by in F6Publishing: 80]  [Article Influence: 13.3]  [Reference Citation Analysis (0)]
70.  Xiang X, Feng D, Hwang S, Ren T, Wang X, Trojnar E, Matyas C, Mo R, Shang D, He Y, Seo W, Shah VH, Pacher P, Xie Q, Gao B. Interleukin-22 ameliorates acute-on-chronic liver failure by reprogramming impaired regeneration pathways in mice. J Hepatol. 2020;72:736-745.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 97]  [Cited by in F6Publishing: 96]  [Article Influence: 24.0]  [Reference Citation Analysis (0)]
71.  Zhu W, Shi XL, Xiao JQ, Gu GX, Ding YT, Ma ZL. Effects of xenogeneic adipose-derived stem cell transplantation on acute-on-chronic liver failure. Hepatobiliary Pancreat Dis Int. 2013;12:60-67.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 20]  [Cited by in F6Publishing: 24]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
72.  Brandão CG, Ferreira HH, Piovesana H, Polimeno NC, Ferraz JG, de Nucci G, Pedrazzoli J Jr. Development of an experimental model of liver cirrhosis in rabbits. Clin Exp Pharmacol Physiol. 2000;27:987-990.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 19]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
73.  Schwarzkopf KM, Eberle L, Uschner FE, Klein S, Schierwagen R, Mücke MM, Schaefer L, Clària J, Zeuzem S, Hintermann E, Christen U, Lange CM, Trebicka J, Welsch C. Interleukin-22 in acute-on-chronic liver failure: A matter of ineffective levels, receptor dysregulation or defective signalling? J Hepatol. 2020;73:980-982.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 5]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
74.   PubChem. Carbon tetrachloride [Internet]. Available from: https://pubchem.ncbi.nlm.nih.gov/compound/5943 Cited 20 June 2021.  [PubMed]  [DOI]  [Cited in This Article: ]
75.   Dianyi Yu. Carbon Tetrachloride Toxicity: What Are the Toxicological Effects of Carbon Tetrachloride Exposure? | Environmental Medicine | ATSDR [Internet]. Available from: https://www.atsdr.cdc.gov/csem/carbon-tetrachloride/toxicological_effects.html.  [PubMed]  [DOI]  [Cited in This Article: ]
76.  de Mesquita FC, Guixé-Muntet S, Fernández-Iglesias A, Maeso-Díaz R, Vila S, Hide D, Ortega-Ribera M, Rosa JL, García-Pagán JC, Bosch J, de Oliveira JR, Gracia-Sancho J. Liraglutide improves liver microvascular dysfunction in cirrhosis: Evidence from translational studies. Sci Rep. 2017;7:3255.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 44]  [Cited by in F6Publishing: 45]  [Article Influence: 6.4]  [Reference Citation Analysis (0)]
77.   PubChem. Thioacetamide [Internet]. Available from: https://pubchem.ncbi.nlm.nih.gov/compound/2723949 Cited 20 June 2021.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Xiping Z, Ke W, Yaping Y, Hongchan Z, Qihui C. Protective effect and mechanisms of radix astragali injection on the intestinal mucosa of rats with obstructive jaundice. Mediators Inflamm. 2010;2010:757191.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 6]  [Cited by in F6Publishing: 10]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
79.  Aller MA, Lorente L, Alonso S, Arias J. A model of cholestasis in the rat, using a microsurgical technique. Scand J Gastroenterol. 1993;28:10-14.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 28]  [Cited by in F6Publishing: 31]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
80.  Nataj A, Eftekhari G, Raoufy MR, Mani AR. The effect of fractal-like mechanical ventilation on vital signs in a rat model of acute-on-chronic liver failure. Physiol Meas. 2018;39:114008.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 3]  [Cited by in F6Publishing: 3]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
81.  Arroyo V, García-Martinez R, Salvatella X. Human serum albumin, systemic inflammation, and cirrhosis. J Hepatol. 2014;61:396-407.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 294]  [Cited by in F6Publishing: 329]  [Article Influence: 32.9]  [Reference Citation Analysis (0)]
82.  Li F, Chordia MD, Woodling KA, Macdonald TL. Irreversible alkylation of human serum albumin by zileuton metabolite 2-acetylbenzothiophene-S-oxide: a potential model for hepatotoxicity. Chem Res Toxicol. 2007;20:1854-1861.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 13]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
83.  Gao DD, Fu J, Qin B, Huang WX, Yang C, Jia B. Recombinant adenovirus containing hyper-interleukin-6 and hepatocyte growth factor ameliorates acute-on-chronic liver failure in rats. World J Gastroenterol. 2016;22:4136-4148.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in CrossRef: 9]  [Cited by in F6Publishing: 9]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
84.  Liu XH, Chen Y, Wang TL, Lu J, Zhang LJ, Song CZ, Zhang J, Duan ZP. [Establishment of a D-galactosamine/Lipopolysaccharide induced acute-on-chronic liver failure model in rats]. Zhonghua Gan Zang Bing Za Zhi. 2007;15:771-775.  [PubMed]  [DOI]  [Cited in This Article: ]
85.  Kopec AK, Joshi N, Luyendyk JP. Role of hemostatic factors in hepatic injury and disease: animal models de-liver. J Thromb Haemost. 2016;14:1337-1349.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 32]  [Cited by in F6Publishing: 29]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
86.  Engelmann C, Mehta G, Tacke F. Regeneration in acute-on-chronic liver failure - the phantom lost its camouflage. J Hepatol. 2020;72:610-612.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 7]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
87.  Jalan R, Fernandez J, Wiest R, Schnabl B, Moreau R, Angeli P, Stadlbauer V, Gustot T, Bernardi M, Canton R, Albillos A, Lammert F, Wilmer A, Mookerjee R, Vila J, Garcia-Martinez R, Wendon J, Such J, Cordoba J, Sanyal A, Garcia-Tsao G, Arroyo V, Burroughs A, Ginès P. Bacterial infections in cirrhosis: a position statement based on the EASL Special Conference 2013. J Hepatol. 2014;60:1310-1324.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 568]  [Cited by in F6Publishing: 592]  [Article Influence: 59.2]  [Reference Citation Analysis (0)]