v
Search
Advanced Search

Publications > Journals > Journal of Clinical and Translational Hepatology > Article Full Text

  • OPEN ACCESS

Prospect of Animal Models for Acute-on-chronic Liver Failure: A Mini-review

  • Hozeifa Mohamed Hassan1,2  and
  • Jun Li1,2,* 
 Author information
Journal of Clinical and Translational Hepatology   2022;10(5):995-1003

doi: 10.14218/JCTH.2022.00086

Abstract

Acute-on-chronic liver failure (ACLF) is a clinical syndrome that develops in patients with chronic liver diseases following a precipitating event and associated with a high mortality rate due to systemic multiorgan failure. Establishing a suitable and stable animal model to precisely elucidate the molecular basis of ACLF pathogenesis is essential for the development of effective early diagnostic and treatment strategies. In this context, this article provides a concise and inclusive review of breakthroughs in ACLF animal model development.

Graphical Abstract

Keywords

Liver failure, Acute-on-chronic liver failure, Animal model, Hepatotoxicants, Inflammation

Introduction

The liver is a unique, irreplaceable, and exceptional multifunctional organ in the vertebrate kingdom,1 as its vital functions vary with age, ranging from the performance, maintenance, and regulation of hematopoiesis in the fetal liver to the metabolism, detoxification and endo/exocrine secretion of hormones and enzymes in the fully developed liver.2,3 The mature liver is mainly composed of hepatocytes and cholangiocytes, which are generated from hepatoblast differentiation,4 as well as five distinct types of cellular components, including sinusoidal endothelial cells, macrophages, different types of lymphocytes, dendritic cells and stellate cells.5–7 Liver failure, among liver disease burdens, is the major health issue encountered worldwide and spectacularly contributes to increased mortality and morbidity, which has significant implications for universal health.8–10 Chronic liver disease (CLD) remains a global health challenge, with up to 2.1 million deaths, which constituted 2.3–2.6% of the global deaths rate.11

Acute-on-chronic liver failure (ACLF): the clinical dilemma

Standing as a distinct disease entity, ACLF is a catastrophic syndrome characterized by an acute deterioration of pre-existing CLD, usually related to a precipitating event and associated with an increased mortality rate due to multiple organ failure.12–15 Although ACLF lacks a universal standard definition and the detailed pathogenesis mechanism is still unclear, Moreau et al. first identified the ACLF diagnostic features and development criteria over the course of acute decompensation of cirrhosis among Western populations.16 However, the definition of ACLF based on cirrhosis, irrespective of etiology, remains controversial. Wu et al. developed a new ACLF definition based on the fact that hepatitis B virus (HBV)-related ACLF exhibits unique clinical characteristics, and their new definition bridges the gap in the criteria for HBV-related ACLF diagnosis.13 Recently, researchers highlighted immune-metabolism disorder as a key factor in deciphering ACLF pathophysiology, irrespective of the ACLF disease etiology and/or precipitating event.17–19 Currently, ACLF-specific treatments are deficient, and organ support and complication prevention are the only substitutes. If medical treatment fails, transplantation is the only option in eligible subjects,20 which is hindered by the shortage of organ donors, the scarcity of deceased organs and the inherent risk of living donor hepatectomy.21

Need of ACLF animal models

The use of animal models is vital in medical research and is mandatory to capture the whole picture of disease pathology to gain beneficial outcomes for both humans and animals, although social debates about the moral aspects of their applications, as well as animal welfare rules, might frequently upsurge.22 As science has evolved, researchers have taken advantage of genetic factors and engineering tools such as mutations, transgenesis, chromosomal rearrangements and conditional gene knock-in/out modifications to help pave the way for model genome manipulation, to generate animal models that are vulnerable or resilient to specific diseases in situations where natural models are not achievable. The utilization of animal models in liver failure is indispensable for pathogenesis clarification, the recognition of prospective targets and the development of innovative therapeutic strategies for liver failure;23 therefore, the availability of appropriate animal models is crucial in studying ACLF to improve our understanding of the dynamic nature of disease occurrence, development and severity progression and to develop novel diagnostic, prognostic, and therapeutic key tools that are dependent on the availability of robust and reproducible experimental animal models. Establishing such a suitable model is difficult due to the complex nature of ACLF prognosis, lack of a clear understanding of ACLF pathogenesis, and shortage of mechanistic studies related to sample unavailability, which escalates the difficulty of ACLF prognostic prediction. In this review, we summarize the current state of the development of ACLF models that potentially replicate clinical ACLF features, with credible prospective trends, a full list of the current ACLF animal models are highlighted in Supplementary Table 1.

Developing ACLF animal models

Distinct strategies have been adopted in vivo to induce ACLF in animals. The key elements in establishing such models are the initiation of CLD, mostly liver fibrosis and/or its advanced form cirrhosis, representing the mutual conclusive pathway of most types of chronic liver failure,24 followed by the administration of a second trigger that acts as precipitating acute insult boosting ACLF development. Agents frequently selected for the acute phase challenge are D-galactosamine (D-gal),25 bacterial lipopolysaccharide (LPS),26 ethyl alcohol,27 the combination of CCl4 and a bacterial load,28 acetaminophen and LPS,29 or D-gal and LPS, which prevail among acute phase triggers.30–33 Repeated administration of hepatotoxicants is considered the principal technique to generate a chronic state of liver failure, and the frequently applied models are listed below.

CCl4-based models

Being viewed as harmless chemical and introduced in clinical practice as anthelmintic in humans since 1920, CCl4 had extended documented history of intoxication in humans, either by ingestion or inhalation, causing serious liver injury.34 CCl4 is the principal prototypical hepatotoxicant, and CCl4 intoxication is the classic way to induce liver injuries in experimental animals. Although CCl4 can also be used in shorter protocols for the study of acute liver injury,35 chronic repeated administration of CCl4 has long been one of the most widely accepted models for inducing CLD; however, a great variety of protocols exist, making it sometimes challenging to compare outcomes from different research groups.36 Differences in the CCl4 protocols used include differences in the route of administration, dosage adjustment to match body weight alterations, frequency of dosing, nature of vehicle used, percentage of dilution, and eventual use of phenobarbitone in the drinking water as an enzyme inducer.37 As shown in Figure 1A, CCl4 is bioactivated via CYP2E1, expressed mainly by centrilobular hepatocytes, to the noxious trichloromethyl radical (CCl3), which mediates hepatocyte toxic effects through the induction of lipid peroxidation and oxidative stress and ultimately leads to hepatotoxic damage, inflammation, and fibrosis that might progress to hepatic cirrhosis.38,39

Hepatotoxicant induction of liver failure.
Fig. 1  Hepatotoxicant induction of liver failure.

(A) Mechanism of carbon tetrachloride (CCl4) induction of liver failure. The production of the trichloromethyl radical is the hallmark of oxidative damage. (B) Representative scheme of porcine serum (PS)-induced acute-on-chronic liver failure (ACLF), in which transcriptomic analysis of liver tissues reveals immune-metabolism disorder as the core axis in ACLF pathogenesis.

In general, CCl4 is administered to mice, rats (BALB/c mice have been described to be the most adequate for modeling, while Wistar rats have higher CCl4 susceptibility) or rabbits through intragastric administration, intraperitoneal injection, subcutaneous injection or inhalation. The protocol duration may vary depending on the dose and species chosen but is generally required for shorter periods of 6–8 weeks to reach a fibrotic state and longer than 8 weeks (8–12 weeks) to reach a stable cirrhotic state.40–42 Following CCl4 administration, the administration of suitable trigger as an acute insult results in the generation of ACLF. Zhu and others43 were the first to generate CCl4-based ACLF rabbits transplanted with xenogeneic porcine adipose-derived stem cells. Despite the advantages of CCl4-based models in the study and characterization of ACLF, multiple drawbacks are associated with CCl4-based models, as they are considered unstable and suffer from substantial death rates during development and inconsistencies in outcomes, which are attributed to animal tolerability. These serious limitations hinder the widespread use of CCl4-based animal models. Recently, Xiang et al.28 established an advanced CCl4-based mouse model consisting of a three-hit protocol (chronic CCl4 + acute CCl4 + bacterial load) to replicate multiorgan failure, a key phase in ACLF clinical prognosis, which has been described as substantial, novel breakthrough toward understanding and potentially targeting regeneration in ACLF.44 In the meantime, a new nonalcoholic-steatohepatitis (NASH)-induced cirrhosis-based ACLF, generated by combination of high-fat diet, CCl4, and repetitive LPS injections and transnasal stool inoculation, was developed to verify the association between advanced NASH and ACLF development in obese patients. Interestingly, the generated ACLF was accompanied by extra-hepatic multiorgan failure.45 Representatives of CCl4-dependent models are summarized in Table 1.28–30,32,45–56

Table 1

Representative examples of major ACLF animal models

HepatotoxicantAnimal typeModeling method
Duration requiredFundamental outcomesPossibilities of multiorgan failurePotential represented etiology in humansRef.
Chronic phaseAcute phase
Carbon tetrachloride (CCl4)C57BL6 miceIntraperitoneal injection of CCl4Intraperitoneal injection of APAP followed by LPS10 weeks for chronic phase followed by acute phase. ACLF state lasts for 11 daysThis model shows the clinical and histological features of human ACLF in terms of the presence of jaundice, ascites, acute tubular necrosis, and renal dysfunctionDevelopment of portal hypertension, renal dysfunction, and pulmonary dysfunctionInfection (endotoxemia)-induced ACLF29
C57BL6 miceIntraperitoneal injection of CCl4Intraperitoneal injection of double dose CCl4 followed by Klebsiella pneumoniae load8 weeks for chronic state followed by acute phase. ACLF duration last for up to 7 daysDeveloping an ACLF mice model with viable bacterial infection for studying liver regeneration and exploring the therapeutic potential of interleukin-22Fc by reprogramming impaired regenerative pathways and attenuating bacterial infectionkidney injury and impairment of blood circulation to the liver and kidneySepsis-induced ACLF28
SD ratsIntraperitoneal injection of CCl4Intraperitoneal injection of D-gal combined with LPS8–12 weeks for chronic phase followed by acute phase. ACLF duration is 24 h-7 daysThese different models were developed to investigates the protection capabilities against ACLF of certain natural products, as well as the influence of the immune system in ACLF pathogenesisNo genuine multiorgan failure has been reported in these modelsInflammation-induced ACLF30,32,4749
SD ratsHigh-fat Western diet combined with intraperitoneal CCl4 injections combined with phenobarbitalRepetitive intraperitoneal injections of LPS combined with transnasal stool inoculation7 weeks for chronic phase followed by acute phase. ACLF duration is unknownThis model develops ACLF in nonalcoholic-steatohepatitis-induced liver cirrhosis, which might be useful in testing pharmacological treatmentsSigns of liver failure, kidney failure, severe respiratory and cerebral failuresnonalcoholic-steatohepatitis-induced ACLF45
New Zealand
rabbit
Intraperitoneal injection of CCl4Intraperitoneal injection of either CCl4 or D-gal10 weeks for chronic phase followed by acute phase. ACLF state lasts for 12 hEstablishing a large animal model of ACLF in New Zealand white rabbits to search for suitable targets and biomarkersNo genuine multiorgan failure has been reportedInflammation-induced ACLF50
Porcine serum (PS)SD ratsIntraperitoneal injection of PSIntraperitoneal injection of D-gal combined with LPS12 weeks for chronic phase followed by acute phase. ACLF duration is 7 daysThis model mimics the clinical pathogenesis and indicates that immune-metabolism disorder is indispensable key core axis in ACLF pathogenesis. The utilization of transcriptomic and multiomic analysis of rat liver is considered a breakthrough in the search for ACLF key molecules and target biomarkersSigns of renal dysfunction as characterized by elevated levels of serum creatinine and blood urea nitrogenImmune-mediated/viral exacerbation-related HBV-ACLF46
Wistar ratsIntraperitoneal injection of PSIntravenous injection of LPS followed by intraperitoneal injection of D-gal11 weeks for chronic phase followed by acute phase. ACLF duration is 24 hIn this model, the transplantation of human-umbilical-cord-derived mesenchymal stem cells could be used to repair the hepatocellular damage following ACLF onset. This method might pave the way for the future application of hepatocyte-like cells in the management of ACLFNo genuine multiorgan failure has been reportedImmune-related/infection-induced ACLF51
Human serum albumin (HSA)Wistar ratsIntravenous injection of HSAInjection of D-gal and LPS combination6 weeks for chronic phase followed by acute phase. ACLF duration is 6–120 hThese different models were developed to analyze the usefulness of certain medicinal plant extracts in alleviating ACLF and the interaction of small molecules with the immune systemPossibilities of accompanying renal dysfunctionImmune-related/infection-induced ACLF5255
SurgerySD ratsSurgically with BDL operationIntraperitoneal injection LPS4 weeks for chronic state followed by acute phase. ACLF duration is 72 hThis model was developed to identify the risk of systemic inflammation in the ACLF prognosis and fatalityNo genuine multiorgan failure has been reportedInflammation-induced ACLF56

Heterologous serum-based models

The utilization of heterologous serum, such as swine or horse serum, egg albumin, human gamma globulin, or pig serum (the most commonly used), to induce immune-mediated CLD has been achievable since 1960, as Paronetto and Popper previously reported.57 These models are characterized by typical hepatic fibrosis/cirrhosis with minimal hepatocellular damage and animal death, which overcomes most limitations associated with CCl4-based models. Previous studies recommended the administration of human serum albumin (HSA) followed by D-gal/LPS administration to induce ACLF, but the use of HSA is hampered by substantial animal mortality, reaching up to 23%, even before ACLF model generation.58–60 Recently, porcine serum (PS) has replaced HSA to provoke the fibrosis/cirrhosis in ACLF preclinical models, as PS administration aggravates the immune response coupled with antibody generation that initiates hepatic fibrosis, which closely resembles human clinical manifestations.61–63 The contribution of the PS antigen immune-mediated response is considered the principal source and the decisive factor for hepatic fibrosis/cirrhosis development, as previously reported.64 Representative heterologous serum-based models are highlighted in Table 1.

PS-induced liver-cirrhosis-based ACLF models can be used to precisely clarify ACLF disease pathogenesis, and immune-metabolism disorder, characterized by immune dysregulation and metabolic disruption, has been identified as the fundamental axis responsible for ACLF development and progression using this model. We recently developed a liver-cirrhosis-based ACLF rat model with a two-hit protocol through the coadministration of PS with D-gal and LPS (Fig. 1B); the model rats show classic ACLF features characterized by cirrhotic nodules surrounded by fibrotic septa, liver dysfunction, cytokine storm, hepatocyte damage, massive necrosis, and cellular apoptosis. Transcriptomic profiling of the significantly differentially expressed genes in the liver tissues showed discernible differences during the process of ACLF rat model building, whereas functional synergy analysis revealed prominent immune dysregulation at the ACLF stage, whereas metabolic disruption was significantly downregulated. Relative proportions of innate-immune-related cells showed significant elevation of monocytes and macrophages, while adaptive-immune-related cells were reduced. Furthermore, validations of the underlying molecular pathogenesis in our generated stable rat model and ACLF patients confirmed that immune-metabolism disorder is indispensable in ACLF pathogenesis.46

Surrogate animal models

As alternatives to the abovementioned models, several other models have been used to replicate ACLF such as surgical-based models. Hu et al.65 described their two-step rat model established through surgical induction of obstructive jaundice, a method previously reported by Xiping et al.66 Furthermore, Tripathi and his group developed a bile duct ligation model that was irresponsive to LPS secondary insult, despite its merit in generating aggressive cirrhotic status.26 Nevertheless, preceding findings highlighted the success of sham operation and/or common bile ligation combined with LPS or CCl4 inhalation in establishing ACLF.67–69 Recently, Karus et al.70 highlighted the combination of bile duct ligation, ethanol binge drinking, repetitive LPS injections, transnasal stool inoculation and cecal ligation and puncture, a complicated approach to generate a new rat model of cholestatic liver cirrhosis-developing ACLF. The differences in final outcomes might impede the selection and rational application of these models to investigate ACLF pathophysiology. To evaluate target-oriented therapy, Schwarzkopf et al. developed a new ACLF model in mice through infection with adenovirus, which precipitates autoimmune hepatitis, resulting in liver fibrosis, followed by paracetamol overdose to trigger ACLF.71

The anchor behind the scene: HBV-based models

Infection with HBV and/or viral exacerbation has gained much attention because of the potential development of a series of CLDs, in which HBV-induced ACLF (HBV-ACLF) as the principal clinical entity culminated in a 50–90% death rate, as documented by the Chinese Group on the Study of Severe Hepatitis B (COSSH).13,17,72 Although HBV-ACLF disease progresses rapidly with limited treatment options, the pathogenesis is unclear, and there is a lack of effective biomarkers for the early diagnosis and prognosis of HBV-ACLF, necessitating the development of a specific and reliable HBV-ACLF animal model. Establishing such a robust model to investigate HBV-ACLF disease development and prognosis seems to be the greatest challenge for researchers, which is attributed to the extremely narrow host range of HBV, which primarily infects humans. While only humans and chimpanzees are sensitive to HBV infection, chimpanzees fail to develop CLD following HBV infection.73 Therefore, the dual humanized chimeric mouse stands as the most promising ideal HBV model.74,75 Yuan et al. previously developed HBV-related liver cirrhosis in a dual humanized mouse model transplanted with human bone marrow mesenchymal stem cells, which were sensitive to chronic HBV infection and generated liver cirrhosis.76 This model could better serve as a candidate model for HBV-ACLF following acute insult with appropriate precipitants, although the application of these approaches is questionable because of the very long time required to reach stable cirrhosis.

Transgenic ACLF animal models: beyond the harbor

The analysis of transgenic models provides insight into ACLF animal models in the future. Over the last decades, various strategies have been applied for the integration of exogenous genetic information within the mouse genome, which has become a common tactic in constructing models with variant physiological roles to provide virtual and detailed information about disease pathophysiological aspects.77,78 Numerous stable transgenic mouse models have been developed to mimic the clinical manifestations of different and intricate diseases, including neurodegenerative diseases,79 cancers,80 cardiovascular diseases,81 and hepatic diseases.82 Several transgenic mouse models were adopted as resources to study liver failure, which allowed investigators to obtain in-depth clues regarding the progression of CLD.83 Tsai et al.84 generated a mouse strain to investigate the role of high-fat diet and corticosterone in the pathogenesis of nonalcoholic fatty liver disease, and Fransén-Pettersson et al.85 developed a transgenic mouse that spontaneously developed inflammatory-derived fibrosis of multiple organs, including the liver. Remarkably, a promising approach is to model ethanol-induced acute-on-chronic liver injury in an Abcb4 knockout transgenic mice model with liver preinjury.86,87 These models could be adopted to clarify the complicated nature of ACLF and provide translational approaches for remarkable achievements in revealing substantial pathophysiology-guided therapeutic targets.

Challenges in selecting an ideal and clinically-relevant ACLF animal model

In fact, we fell short for an appropriate and universally accepted definition for ACLF so far because of the complexity and heterogenic nature of ACLF.88 Therefore, to assume the existence of an ideal, single ACLF model for interpreting disease pathogenesis, bridging the gap between molecular mechanisms and therapeutic strategies, is not logically possible. The models should distinctively reflect the clinical mechanism and prognosis of human ACLF pathogenesis. Despite those limitations, the above-mentioned models share most of the typical pathophysiological manifestations of ACLF such as severe deterioration of liver function and histology accompanied by single and/or multiorgan failure with concomitant high short-term mortality. The methods and models succeed in mimicking the reactivation of hepatitis viral infection, acute alcoholic hepatitis or acute bacterial infection in Asian populations, as well as alcohol abuse and bacterial infection that aggravate CLD conditions in the Western population. In this context, most ACLF animal models employ chemical induction (drugs/alcoholic/infection) or immune induction (heterologous serum), of CLD followed by an acute trigger to initiate ACLF. Selecting any method or model to imitate certain aspects in human ACLF should be supported by the purpose, strengths, flaws, and rationale of the study in mimicking explicit clinical scenario. Adoption of sophisticated techniques with advanced humanized mice models may represent major breakthroughs in ACLF animal model development. Furthermore, measurable histological and/or pathological endpoint markers should be adopted to ensure the successful recapitulation of the ACLF common disease features clinically seen in humans, such as presence of both chronic and acute liver injury indicators, ascites, encephalopathy, and secondary organ dysfunction.

Future insights in ACLF animal models

Although different models emulate and are complementary to ACLF development and prognosis in humans, the actual clinical manifestations of ACLF are intricate, and precise, stable, and novel ACLF animal models are required to achieve breakthroughs in the study of ACLF pathogenesis. Despite the existence of various models that aided in revealing ACLF pathogenesis and prognosis, a key challenge stands in identifying the utmost ACLF model that precisely replicates all aspects in human ACLF. Most models only can reflect certain features of disease pathophysiology as inflammatory, hepatic and/or extrahepatic organ failure that ultimately lead to high short-term mortality. In the meantime, establishing a rationale guidance for selecting distinct acute insult triggers as precipitating events in ACLF development, is of great importance. Regardless of the model chosen, adoption of standardized scoring systems for acute insult selection should be strongly encouraged. However, recent studies that focused on the potencies of secondary acute insults in ACLF are lacking. We hope that issue will be addressed in the near future by appropriate studies. The application of integrated investigational approaches with a suitable ACLF animal models using multiomic function correlation analysis and bioenergetic data should be performed in the near future to fully decipher ACLF pathogenesis and identify key molecular target biomarkers for early diagnosis and prediction of the occurrence, development, and prognosis of ACLF.

Supporting information

Supplementary Table 1

Animal models of ACLF.

(DOCX)

Abbreviations

ACLF: 

acute-on-chronic liver failure

CLD: 

chronic liver disease

COSSH: 

Chinese Group on the Study of Severe Hepatitis B

D-gal: 

D-galactosamine

HSA: 

human serum albumin

HBV: 

hepatitis B virus

HBV-ACLF: 

HBV-induced acute-on-chronic liver failure

LPS: 

lipopolysaccharide

NASH: 

nonalcoholic-steatohepatitis

PS: 

porcine serum

Declarations

Funding

This study was supported by the National S&T Major Project of China (2017ZX10304402 and 2017ZX10203201), the National Natural Science Foundation of China (81830073, 81771196, and 81901901), the State’s Key Project of Research and Development Plan of China (2016YFC1101303/4), and the National and Zhejiang Provincial special support program for high-level personnel recruitment (Ten-thousand Talents Program).

Conflict of interest

JL has been an editorial board member of Journal of Clinical and Translational Hepatology since 2019. HMH has no conflict of interests related to this publication.

Authors’ contributions

Contributed equally to the main review conception and manuscript writing, organizing the presented models, performing the literature search and analysis, preparing figures and tables, and discussing and systematizing the presented literature data, and have approved the final manuscript (HMH, JL).

References

  1. Qin L, Crawford JM. . Zakim and Boyer’s hepatology: A textbook of liver disease. 7th edition. Elsevier, Inc.; 2018
  2. Vaja R, Rana M. Drugs and the liver. Anaesth Intensive Care Med 2020;21(10):517-523 View Article
  3. Trefts E, Gannon M, Wasserman DH. The liver. Curr Biol 2017;27(21):R1147-R1151 View Article PubMed/NCBI
  4. Gordillo M, Evans T, Gouon-Evans V. Orchestrating liver development. Development 2015;142(12):2094-2108 View Article PubMed/NCBI
  5. Guillot A, Tacke F. Liver Macrophages: Old Dogmas and New Insights. Hepatol Commun 2019;3(6):730-743 View Article PubMed/NCBI
  6. Ober EA, Lemaigre FP. Development of the liver: Insights into organ and tissue morphogenesis. J Hepatol 2018;68(5):1049-1062 View Article PubMed/NCBI
  7. Wong SW, Yeh SJ, Li CW, Wang LH, Chen BS. Investigation mechanisms between normal, developing and regenerating livers for regenerative liver drug design. Regen Med 2019;14(5):359-387 View Article PubMed/NCBI
  8. Xiao J, Wang F, Wong NK, He J, Zhang R, Sun R, et al. Global liver disease burdens and research trends: Analysis from a Chinese perspective. J Hepatol 2019;71(1):212-221 View Article PubMed/NCBI
  9. Horvatits T, Drolz A, Trauner M, Fuhrmann V. Liver Injury and Failure in Critical Illness. Hepatology 2019;70(6):2204-2215 View Article PubMed/NCBI
  10. Asrani SK, Devarbhavi H, Eaton J, Kamath PS. Burden of liver diseases in the world. J Hepatol 2019;70(1):151-171 View Article PubMed/NCBI
  11. GBD 2017 Cirrhosis Collaborators. The global, regional, and national burden of cirrhosis by cause in 195 countries and territories, 1990-2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet Gastroenterol Hepatol 2020;5(3):245-266 View Article PubMed/NCBI
  12. Kulkarni S, Sharma M, Rao PN, Gupta R, Reddy DN. Acute on Chronic Liver Failure-In-Hospital Predictors of Mortality in ICU. J Clin Exp Hepatol 2018;8(2):144-155 View Article PubMed/NCBI
  13. Wu T, Li J, Shao L, Xin J, Jiang L, Zhou Q, et al. Development of diagnostic criteria and a prognostic score for hepatitis B virus-related acute-on-chronic liver failure. Gut 2018;67(12):2181-2191 View Article PubMed/NCBI
  14. Arroyo V, Moreau R, Jalan R. Acute-on-Chronic Liver Failure. N Engl J Med 2020;382(22):2137-2145 View Article PubMed/NCBI
  15. Hernaez R, Solà E, Moreau R, Ginès P. Acute-on-chronic liver failure: an update. Gut 2017;66(3):541-553 View Article PubMed/NCBI
  16. Moreau R, Jalan R, Gines P, Pavesi M, Angeli P, Cordoba J, et al. Acute-on-chronic liver failure is a distinct syndrome that develops in patients with acute decompensation of cirrhosis. Gastroenterology 2013;144(7):1426-1437.E9 View Article PubMed/NCBI
  17. Li J, Liang X, Jiang J, Yang L, Xin J, Shi D, et al. PBMC transcriptomics identifies immune-metabolism disorder during the development of HBV-ACLF. Gut 2022;71(1):163-175 View Article PubMed/NCBI
  18. Moreau R, Clària J, Aguilar F, Fenaille F, Lozano JJ, Junot C, et al. Blood metabolomics uncovers inflammation-associated mitochondrial dysfunction as a potential mechanism underlying ACLF. J Hepatol 2020;72(4):688-701 View Article PubMed/NCBI
  19. Zhang IW, Curto A, López-Vicario C, Casulleras M, Duran-Güell M, Flores-Costa R, et al. Mitochondrial dysfunction governs immunometabolism in leukocytes of patients with acute-on-chronic liver failure. J Hepatol 2022;76(1):93-106 View Article PubMed/NCBI
  20. Vizzutti F, Arena U, Laffi G, Marra F. Acute on chronic liver failure: From pathophysiology to clinical management. Trends Anaesth. Crit. Care 2013;3(3):122-129 View Article
  21. Chan AC, Fan ST. Criteria for liver transplantation in ACLF and outcome. Hepatol Int 2015;9(3):355-359 View Article PubMed/NCBI
  22. Barré-Sinoussi F, Montagutelli X. Animal models are essential to biological research: issues and perspectives. Future Sci OA 2015;1(4):FSO63 View Article PubMed/NCBI
  23. Reza Khorramizadeh M, Saadat F. . Animal Biotechnology 2 nd edition. Academic Press; 2020
  24. Liu Y, Meyer C, Xu C, Weng H, Hellerbrand C, ten Dijke P, et al. Animal models of chronic liver diseases. Am J Physiol Gastrointest Liver Physiol 2013;304(5):G449-G468 View Article PubMed/NCBI
  25. 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(2):133-141 View Article PubMed/NCBI
  26. Tripathi DM, Vilaseca M, Lafoz E, Garcia-Calderó H, Viegas Haute G, Fernández-Iglesias A, et al. Simvastatin Prevents Progression of Acute on Chronic Liver Failure in Rats With Cirrhosis and Portal Hypertension. Gastroenterology 2018;155(5):1564-1577 View Article PubMed/NCBI
  27. Furuya S, Cichocki JA, Konganti K, Dreval K, Uehara T, Katou Y, et al. 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(2):427-437 View Article PubMed/NCBI
  28. Xiang X, Feng D, Hwang S, Ren T, Wang X, Trojnar E, et al. Interleukin-22 ameliorates acute-on-chronic liver failure by reprogramming impaired regeneration pathways in mice. J Hepatol 2020;72(4):736-745 View Article PubMed/NCBI
  29. Nautiyal N, Maheshwari D, Tripathi DM, Kumar D, Kumari R, Gupta S, et al. Establishment of a murine model of acute-on-chronic liver failure with multi-organ dysfunction. Hepatol Int 2021;15(6):1389-1401 View Article PubMed/NCBI
  30. Ni S, Li S, Yang N, Tang X, Zhang S, Hu D, et al. Deregulation of Regulatory T Cells in Acute-on-Chronic Liver Failure: A Rat Model. Mediators Inflamm 2017;2017:1390458 View Article PubMed/NCBI
  31. Xie F, Dong J, Zhu Y, Wang K, Liu X, Chen D, et al. HIF1a Inhibitor Rescues Acute-on-Chronic Liver Failure. Ann Hepatol 2019;18(5):757-764 View Article PubMed/NCBI
  32. Xue R, Zhu X, Jia L, Wu J, Yang J, Zhu Y, et al. Mitofusin2, a rising star in acute-on-chronic liver failure, triggers macroautophagy via the mTOR signalling pathway. J Cell Mol Med 2019;23(11):7810-7818 View Article PubMed/NCBI
  33. Xue R, Yang J, Jia L, Zhu X, Wu J, Zhu Y, et al. Mitofusin2, as a Protective Target in the Liver, Controls the Balance of Apoptosis and Autophagy in Acute-on-Chronic Liver Failure. Front Pharmacol 2019;10:601 View Article PubMed/NCBI
  34. Teschke R. Liver Injury by Carbon Tetrachloride Intoxication in 16 Patients Treated with Forced Ventilation to Accelerate Toxin Removal via the Lungs: A Clinical Report. Toxics 2018;6(2):E25 View Article PubMed/NCBI
  35. Zhang X, Kuang G, Wan J, Jiang R, Ma L, Gong X, et al. Salidroside protects mice against CCl4-induced acute liver injury via down-regulating CYP2E1 expression and inhibiting NLRP3 inflammasome activation. Int Immunopharmacol 2020;85:106662 View Article PubMed/NCBI
  36. Weber LW, Boll M, Stampfl A. Hepatotoxicity and mechanism of action of haloalkanes: carbon tetrachloride as a toxicological model. Crit Rev Toxicol 2003;33(2):105-136 View Article PubMed/NCBI
  37. Regimbeau JM, Fuks D, Kohneh-Shahri N, Terris B, Soubrane O. Restrictive model of compensated carbon tetrachloride-induced cirrhosis in rats. World J Gastroenterol 2008;14(45):6943-6947 View Article PubMed/NCBI
  38. Tabet E, Genet V, Tiaho F, Lucas-Clerc C, Gelu-Simeon M, Piquet-Pellorce C, et al. Chlordecone potentiates hepatic fibrosis in chronic liver injury induced by carbon tetrachloride in mice. Toxicol Lett 2016;255:1-10 View Article PubMed/NCBI
  39. Khalil MR, El-Demerdash RS, Elminshawy HH, Mehanna ET, Mesbah NM, Abo-Elmatty DM. Therapeutic effect of bone marrow mesenchymal stem cells in a rat model of carbon tetrachloride induced liver fibrosis. Biomed J 2021;44(5):598-610 View Article PubMed/NCBI
  40. Fortea JI, Fernández-Mena C, Puerto M, Ripoll C, Almagro J, Bañares J, et al. Comparison of Two Protocols of Carbon Tetrachloride-Induced Cirrhosis in Rats - Improving Yield and Reproducibility. Sci Rep 2018;8(1):9163 View Article PubMed/NCBI
  41. Jin B, Alter HJ, Zhang ZC, Shih JW, Esteban JM, Sun T, et al. Reversibility of experimental rabbit liver cirrhosis by portal collagenase administration. Lab Invest 2005;85(8):992-1002 View Article PubMed/NCBI
  42. Brandão CG, Ferreira HH, Piovesana H, Polimeno NC, Ferraz JG, de Nucci G, et al. Development of an experimental model of liver cirrhosis in rabbits. Clin Exp Pharmacol Physiol 2000;27(12):987-990 View Article PubMed/NCBI
  43. 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(1):60-67 View Article PubMed/NCBI
  44. Engelmann C, Mehta G, Tacke F. Regeneration in acute-on-chronic liver failure - the phantom lost its camouflage. J Hepatol 2020;72(4):610-612 View Article PubMed/NCBI
  45. Kraus N, Moeslein M, Schierwagen R, Ortiz C, Torres S, Tyc O, et al. New Rat Model of Advanced Non-Alcoholic-Steatohepatitis-Cirrhosis Developing Acute-on-Chronic-Liver failure. Z Gastroenterol 2022;60(01):e10 View Article
  46. Hassan HM, Cai Q, Liang X, Xin J, Ren K, Jiang J, et al. Transcriptomics reveals immune-metabolism disorder in acute-on-chronic liver failure in rats. Life Sci Alliance 2022;5(3):e202101189 View Article PubMed/NCBI
  47. Tang L, Wang F, Xiao L, Shen M, Xia S, Zhang Z, et al. Yi-Qi-Jian-Pi formula modulates the PI3K/AKT signaling pathway to attenuate acute-on-chronic liver failure by suppressing hypoxic injury and apoptosis in vivo and in vitro. J Ethnopharmacol 2021;280:114411 View Article PubMed/NCBI
  48. Wang F, Tang L, Liang B, Jin C, Gao L, Li Y, et al. Yi-Qi-Jian-Pi Formula Suppresses RIPK1/RIPK3-Complex-Dependent Necroptosis of Hepatocytes Through ROS Signaling and Attenuates Liver Injury in Vivo and in Vitro. Front Pharmacol 2021;12:658811 View Article PubMed/NCBI
  49. Diao J, Li H, Huang W, Ma W, Dai H, Liu Y, et al. SHYCD induces APE1/Ref-1 subcellular localization to regulate the p53-apoptosis signaling pathway in the prevention and treatment of acute on chronic liver failure. Oncotarget 2017;8(49):84782-84797 View Article PubMed/NCBI
  50. Guangxiang GU, Xiaolei SHI, Hualian H, Yitao D. Animal models of acute-on-chronic liver failure in rabbits. Chin J Exp Surg 2011;28(5):809-812
  51. He Y, Guo X, Lan T, Xia J, Wang J, Li B, et al. Human umbilical cord-derived mesenchymal stem cells improve the function of liver in rats with acute-on-chronic liver failure via downregulating Notch and Stat1/Stat3 signaling. Stem Cell Res Ther 2021;12(1):396 View Article PubMed/NCBI
  52. Hou W, Hao Y, Yang W, Tian T, Fang P, Du Y, et al. The Jieduan-Niwan (JDNW) Formula Ameliorates Hepatocyte Apoptosis: A Study of the Inhibition of E2F1-Mediated Apoptosis Signaling Pathways in Acute-on-Chronic Liver Failure (ACLF) Using Rats. Drug Des Devel Ther 2021;15:3845-3862 View Article PubMed/NCBI
  53. Xu Y, Wang H, Bao S, Tabassam F, Cai W, Xiang X, et al. Amelioration of liver injury by continuously targeted intervention against TNFRp55 in rats with acute-on-chronic liver failure. PLoS One 2013;8(7):e68757 View Article PubMed/NCBI
  54. Pfingstgraf IO, Taulescu M, Pop RM, Orăsan R, Vlase L, Uifalean A, et al. Protective Effects of Taraxacum officinale L. (Dandelion) Root Extract in Experimental Acute on Chronic Liver Failure. Antioxidants (Basel) 2021;10(4):504 View Article PubMed/NCBI
  55. Zhang Q, Yang F, Li X, Wang LW, Chu XG, Zhang H, et al. Trichostatin A Protects Against Experimental Acute-on-Chronic Liver Failure in Rats Through Regulating the Acetylation of Nuclear Factor-κB. Inflammation 2015;38(3):1364-1373 View Article PubMed/NCBI
  56. Monteiro S, Grandt J, Uschner FE, Kimer N, Madsen JL, Schierwagen R, et al. Differential inflammasome activation predisposes to acute-on-chronic liver failure in human and experimental cirrhosis with and without previous decompensation. Gut 2021;70(2):379-387 View Article PubMed/NCBI
  57. Paronetto F, Popper H. Chronic liver injury induced by immunologic reactions. Cirrhosis following immunization with heterologous sera. Am J Pathol 1966;49(6):1087-1101 PubMed/NCBI
  58. Wang LW, Wang LK, Chen H, Fan C, Li X, He CM, et al. Ethyl pyruvate protects against experimental acute-on-chronic liver failure in rats. World J Gastroenterol 2012;18(40):5709-5718 View Article PubMed/NCBI
  59. Liu XH, Chen Y, Wang TL, Lu J, Zhang LJ, Song CZ, et al. Establishment of a D-galactosamine/lipopolysaccharide induced acute-on-chronic liver failure model in rats. Zhonghua Gan Zang Bing Za Zhi 2007;15(10):771-775 PubMed/NCBI
  60. Yang W, Hao Y, Hou W, Fang X, Fang P, Jiang T, et al. Jieduan-Niwan Formula Reduces Liver Apoptosis in a Rat Model of Acute-on-Chronic Liver Failure by Regulating the E2F1-Mediated Intrinsic Apoptosis Pathway. Evid Based Complement Alternat Med 2019;2019:8108503 View Article PubMed/NCBI
  61. Liu H, Wei W, Sun WY, Li X. Protective effects of astragaloside IV on porcine-serum-induced hepatic fibrosis in rats and in vitro effects on hepatic stellate cells. J Ethnopharmacol 2009;122(3):502-508 View Article PubMed/NCBI
  62. Liu H, Wei W, Li X. Celecoxib exacerbates hepatic fibrosis and induces hepatocellular necrosis in rats treated with porcine serum. Prostaglandins Other Lipid Mediat 2009;88(3-4):63-67 View Article PubMed/NCBI
  63. Li J, Zhang Q, Gao L, Du Y, Chen Y. Efficacy of decoction from Jieduan Niwan formula on rat model of acute-on-chronic liver failure induced by porcine serum. J Tradit Chin Med 2020;40(4):602-612 View Article PubMed/NCBI
  64. Baba Y, Uetsuka K, Nakayama H, Dot K. Rat strain differences in the early stage of porcine-serum-induced hepatic fibrosis. Exp Toxicol Pathol 2004;55(5):325-330 View Article PubMed/NCBI
  65. 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(10):928-935 View Article PubMed/NCBI
  66. 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 View Article PubMed/NCBI
  67. Harry D, Anand R, Holt S, Davies S, Marley R, Fernando B, et al. Increased sensitivity to endotoxemia in the bile duct-ligated cirrhotic Rat. Hepatology 1999;30(5):1198-1205 View Article PubMed/NCBI
  68. Engelmann C, Adebayo D, Oria M, De Chiara F, Novelli S, Habtesion A, et al. Recombinant Alkaline Phosphatase Prevents Acute on Chronic Liver Failure. Sci Rep 2020;10(1):389 View Article PubMed/NCBI
  69. Chang J, Meinke J, Geck M, Hebest M, Böhling N, Dolscheid-Pommerich R, et al. Extrahepatic Surgery in Cirrhosis Significantly Increases Portal Pressure in Preclinical Animal Models. Front Physiol 2021;12:720898 View Article PubMed/NCBI
  70. Kraus N, Moeslein M, Schierwagen R, Ortiz C, Torres S, Tyc O, et al. New Rat Model of Cholestatic Liver Cirrhosis Developing Acute-on-Chronic-Liver failure. Z Gastroenterol 2022;60(01):e9-e10 View Article
  71. Schwarzkopf K, Rüschenbaum S, Muecke MM, Fuchs S, Messmer M, Bayer M, et al. THU-051-Establishment of a new animal model for the acute-on-chronic liver failure. J Hepatol 2019;70(1):e183 View Article
  72. Li J, Liang X, You S, Feng T, Zhou X, Zhu B, et al. Development and validation of a new prognostic score for hepatitis B virus-related acute-on-chronic liver failure. J Hepatol 2021;75(5):1104-1115 View Article PubMed/NCBI
  73. Liu Y, Meyer C, Xu C, Weng H, Hellerbrand C, ten Dijke P, et al. Animal models of chronic liver diseases. Am J Physiol Gastrointest Liver Physiol 2013;304(5):G449-468 View Article PubMed/NCBI
  74. Sun S, Li J. Humanized chimeric mouse models of hepatitis B virus infection. Int J Infect Dis 2017;59:131-136 View Article PubMed/NCBI
  75. Zhang X, Wang X, Wu M, Ghildyal R, Yuan Z. Animal Models for the Study of Hepatitis B Virus Pathobiology and Immunity: Past, Present, and Future. Front Microbiol 2021;12:715450 View Article PubMed/NCBI
  76. Yuan L, Jiang J, Liu X, Zhang Y, Zhang L, Xin J, et al. HBV infection-induced liver cirrhosis development in dual-humanised mice with human bone mesenchymal stem cell transplantation. Gut 2019;68(11):2044-2056 View Article PubMed/NCBI
  77. Babinet C, Morello D, Renard JP. Transgenic mice. Genome 1989;31(2):938-949 View Article PubMed/NCBI
  78. Pu XA, Young AP, Kubisch HM. Production of Transgenic Mice by Pronuclear Microinjection. Methods Mol Biol 2019;1874:17-41 View Article PubMed/NCBI
  79. Lithner CU, Hedberg MM, Nordberg A. Transgenic mice as a model for Alzheimer’s disease. Curr Alzheimer Res 2011;8(8):818-831 View Article PubMed/NCBI
  80. Pérez-Guijarro E, Day CP, Merlino G, Zaidi MR. Genetically engineered mouse models of melanoma. Cancer 2017;123(S11):2089-2103 View Article PubMed/NCBI
  81. Lavoie JL, Bianco RA, Sakai K, Keen HL, Ryan MJ, Sigmund CD. Transgenic mice for studies of the renin-angiotensin system in hypertension. Acta Physiol Scand 2004;181(4):571-577 View Article PubMed/NCBI
  82. Larkin J, Clayton M, Sun B, Perchonock CE, Morgan JL, Siracusa LD, et al. Hepatitis B virus transgenic mouse model of chronic liver disease. Nat Med 1999;5(8):907-912 View Article PubMed/NCBI
  83. Hayashi H, Sakai T. Animal models for the study of liver fibrosis: new insights from knockout mouse models. Am J Physiol Gastrointest Liver Physiol 2011;300(5):G729-G738 View Article PubMed/NCBI
  84. Tsai SF, Hung HC, Shih MM, Chang FC, Chung BC, Wang CY, et al. High-fat diet-induced increases in glucocorticoids contribute to the development of non-alcoholic fatty liver disease in mice. FASEB J 2022;36(1):e22130 View Article PubMed/NCBI
  85. Fransén-Pettersson N, Duarte N, Nilsson J, Lundholm M, Mayans S, Larefalk Å, et al. A New Mouse Model That Spontaneously Develops Chronic Liver Inflammation and Fibrosis. PLoS One 2016;11(7):e0159850 View Article PubMed/NCBI
  86. Karatayli E, Hall RA, Weber SN, Dooley S, Lammert F. Effect of alcohol on the interleukin 6-mediated inflammatory response in a new mouse model of acute-on-chronic liver injury. Biochim Biophys Acta Mol Basis Dis 2019;1865(2):298-307 View Article PubMed/NCBI
  87. Christidis G, Karatayli E, Hall RA, Weber SN, Reichert MC, Hohl M, et al. Fibroblast Growth Factor 21 Response in a Preclinical Alcohol Model of Acute-on-Chronic Liver Injury. Int J Mol Sci 2021;22(15):7898 View Article PubMed/NCBI
  88. Schulz M, Trebicka J. Acute-on-chronic liver failure: a global disease. Gut 2022;71(1):5-6 View Article PubMed/NCBI
  • Journal of Clinical and Translational Hepatology
  • pISSN 2225-0719
  • eISSN 2310-8819
Back to Top

Prospect of Animal Models for Acute-on-chronic Liver Failure: A Mini-review

Hozeifa Mohamed Hassan, Jun Li
  • Reset Zoom
  • Download TIFF