v
Search
Advanced Search

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

  • OPEN ACCESS

Mouse Models for the Study of Liver Fibrosis Regression In Vivo and Ex Vivo

  • Milena Schönke1,2,*  and
  • Patrick C.N. Rensen1,2
 Author information
Journal of Clinical and Translational Hepatology   2024

doi: 10.14218/JCTH.2024.00212

Abstract

This review discussed experimental mouse models used in the pre-clinical study of liver fibrosis regression, a pivotal process in preventing the progression of metabolic dysfunction-associated steatohepatitis to irreversible liver cirrhosis. These models provide a valuable resource for understanding the cellular and molecular processes underlying fibrosis regression in different contexts. The primary focus of this review is on the most commonly used models with diet- or hepatotoxin-induced fibrosis, but it also touches upon genetic models and mouse models with biliary atresia or parasite-induced fibrosis. In addition to emphasizing in vivo models, we briefly summarized current in vitro approaches designed for studying fibrosis regression and provided an outlook on evolving methodologies that aim to refine and reduce the number of experimental animals needed for these studies. Together, these models contribute significantly to unraveling the underlying mechanisms of liver fibrosis regression and offer insights into potential therapeutic interventions. By presenting a comprehensive overview of these models and highlighting their respective advantages and limitations, this review serves as a roadmap for future research.

Graphical Abstract

Keywords

Liver fibrosis, Liver steatosis, Metabolic dysfunction-associated steatotic liver disease, Metabolic dysfunction-associated steatohepatitis, Fibrosis regression, Mouse models of liver fibrosis

Introduction

Metabolic dysfunction-associated steatotic liver disease (MASLD), previously known as non-alcoholic fatty liver disease (NAFLD), is estimated to affect 25–30% of the world’s population and is strongly associated with other metabolic diseases, such as obesity, type 2 diabetes mellitus, and atherosclerotic cardiovascular disease, which are continuously on the rise.1 Over time, simple hepatic steatosis, characterized by excess lipid deposition in the liver, induces liver inflammation, marking the onset of metabolic dysfunction-associated steatohepatitis (MASH), which affects 25% of individuals with MASLD. Driven by steatohepatitis, scarring and thickening of the liver tissue, known as fibrosis, occurs. This process is reversible early on but may ultimately progress to the irreversible replacement of too much functioning tissue and lead to cirrhosis, a significant impairment of liver function. In addition to MASH, multiple other chronic conditions can cause liver fibrosis, including alcohol abuse, viral or parasitic hepatitis, biliary obstruction, and hemochromatosis.2,3 End-stage liver disease is currently only treatable with liver transplantation.4 Coincidentally, liver failure due to end-stage MASH has become the most frequent reason for liver transplantation, while the availability of donor livers has at the same time decreased due to the high rate of organ donors with liver steatosis, highlighting an imminent need for improvement of treatments.5 Accordingly, this review will primarily discuss fibrosis and fibrosis regression in the context of MASH, although many disease development and regression processes are shared with other chronic liver diseases.

The development of pharmacological treatments for MASH is making great strides, with the first compound, the liver-directed thyroid hormone receptor agonist resmetirom,6 recently receiving conditional approval from the U.S. Food and Drug Administration for the treatment of adults with noncirrhotic MASH with moderate to advanced fibrosis, to be used in conjunction with diet and exercise.7 Approval is also on the horizon for other candidates, such as FGF21,8 combined glucose-dependent insulinotropic polypeptide receptor (GIPR)/glucagon-like peptide-1 receptor (GLP1R) agonists such as tirzepatide,9 and dual glucagon receptor/GLP1R agonists such as survodutide.10

The prevention and reversal of liver fibrosis is an important clinical benchmark, and suitable pre-clinical research models are essential for the development of effective treatments for liver fibrosis. For this purpose, in vitro organoids that include fibrosis-mediating hepatic stellate cells (HSCs) are continuously being improved.11 However, to study the interaction with other metabolic tissues in the body and inflammatory processes, animal studies remain paramount for now. Interestingly, resmetirom, tirzepatide, and survodutide have all been shown to lower the fibrosis score in human MASH, seemingly without directly affecting HSCs.9,10,12 This suggests that the reduction of intrahepatic lipids and inflammation may be sufficient for the regression of liver fibrosis, underscoring the value of in vivo models where these processes can be studied in a whole-body context.

A large number of experimental mouse models for MASLD, ranging from diet-induced obesity models to genetic models, are available, though not all models develop all hallmarks of human fibrotic steatohepatitis. This review focuses on mouse models that develop hepatic fibrosis, particularly those in which fibrosis can be reversed, allowing for the study of fibrosis regression.

Liver fibrosis

Fibrogenesis occurs in response to repeated or continuous liver damage from processes such as lipid overload, toxin exposure (e.g., alcohol), infections, bile salt accumulation, or metal poisoning, and it results from insufficient degradation of the fibrillar extracellular matrix produced during the damage repair process. The molecular regulation of fibrogenesis is complex, but in MASH appears to be primarily driven by reactive oxygen species that form, for example, during lipid overload in cells. Reactive oxygen species activate fibroblasts and HSCs, which can transdifferentiate into profibrotic myofibroblasts.13 This transdifferentiation is also mediated by activated hepatic macrophages through the secretion of pro-inflammatory cytokines such as TGF-β, and clinical trials with compounds that inhibit monocyte migration to the liver have reported anti-fibrotic effects.14,15 A schematic overview of the development of liver fibrosis in MASLD is shown in Figure 1. Once the inflammation subsides, hepatic macrophages switch to a more fibrolytic phenotype and express matrix metalloproteases that degrade the extracellular matrix, leading to the regression of fibrosis, making this a dynamic process. This indicates that the best models for fibrotic MASH mimic not only the primary cellular contributors but also the inflammatory microenvironment and the metabolic (dys)regulation in the human liver that causes the onset of inflammation in the first place.

Schematic overview of the development of liver fibrosis in MASLD.
Fig. 1  Schematic overview of the development of liver fibrosis in MASLD.

MASLD, Metabolic dysfunction-associated steatotic liver disease; ROS, reactive oxygen species.

The severity of hepatic fibrosis is commonly scored histologically in four stages, which are assessed in addition to the MASLD activity score. This score evaluates the severity of steatosis (graded 0–3), liver cell injury such as ballooning (graded 0–2), and lobular inflammation (graded 0–3).16 The stages of fibrosis are typically defined as: 0 = no fibrosis, 1 = mild perisinusoidal/pericellular fibrosis (described as resembling chicken wire around the central vein of a liver lobule), 2 = perisinusoidal/pericellular and portal/periportal fibrosis (with dense collagen deposits around the portal vein), 3 = bridging fibrosis (where mature fibrous tissue forms a band connecting the portal area to the central vein), and 4 = cirrhosis (characterized by nodules of cells surrounded by thick, connecting fibrotic septa).17 In some scoring systems, such as the NASH Clinical Research Network score, stage 1 is further subdivided into 1a–c to distinguish the severity of perisinusoidal fibrosis. The MASLD activity scoring and fibrosis staging systems have been found to be applicable to rodent models and are widely used in mouse studies.18 During fibrosis regression, fibrous septa thin out and become fragmented, with hepatocytes pushing between the scar tissue. This is followed by vascular remodeling and the reestablishment of the normal architecture of the hepatic trabeculae.19

Mouse models for the study of liver fibrosis

Firstly, due to the highly comparable clinical profiles of NAFLD and MASLD,20 we regard mouse models previously described for the study of NAFLD as still applicable under the new nomenclature. The most commonly used mouse models in pre-clinical studies of MASLD are wild-type (C57BL/6) mice on a high-fat diet (HFD; often supplemented with lard, cholesterol, sucrose, or fructose to induce more liver damage21–23) and various genetically modified mice on similar diets. All these models have been reviewed extensively before, highlighting their advantages and disadvantages for studying certain disease hallmarks.24–27 Particularly when focusing on the development of fibrotic MASH, differences between the models become evident.28,29

For instance, C57BL/6 mice on HFD develop only minor hepatic fibrosis after 50 weeks of feeding, but more so after 80 weeks.30–32 At the other end of the spectrum, a methionine- and choline-deficient HFD, which lowers the antioxidant defense system of the liver and strongly accelerates hepatic lipid retention, induces severe steatosis and fibrosis in just six weeks.33,34 However, since animals lose weight on the methionine- and choline-deficient diet, the metabolic phenotype is not comparable to that of humans with MASLD. Overall, HFD with a high fructose content and the Gubra Amylin diet (an HFD with high fructose, sucrose, and cholesterol content) were found to most closely resemble human MASLD, with liver fibrosis detectable after 28 weeks of feeding.35–38

To study fibrotic MASH in the context of co-morbidities and whole-body metabolic dysfunction associated with the disease in humans, genetic mouse models can be useful. LDL receptor knockout mice and ApoE knockout mice, which are both commonly used to study hypercholesterolemia and atherosclerotic cardiovascular disease, develop periportal fibrosis after 6 to 12 weeks of high-fat, high-cholesterol feeding.39,40 Similarly, hyperlipidemic APOE*3-Leiden.CETP mice, which have a more humanized lipoprotein metabolism, on a C57BL/6J background, develop stage 3 fibrosis after 16 weeks of high-fat, high-cholesterol feeding, along with obesity and insulin resistance.41 Leptin-deficient hyperphagic ob/ob mice, commonly used in the study of obesity and hyperglycemia, develop fibrotic MASH when fed an HFD rich in cholesterol and trans-fatty acids for 20 weeks.42,43 Major urinary protein-urokinase-type plasminogen activator mice, a model of chronic ER stress in hepatocytes, develop MASH that closely resembles human pathology, with pericellular and bridging fibrosis and spontaneous progression to hepatocellular carcinoma over 24 weeks of HFD feeding.44,45 When studying the molecular processes of fibrosis development independently of metabolic dysfunction, organic hepatotoxins (such as ethanol, nitrosamines, tetrachloromethane (CCl4), or thioacetamide), surgical bile duct ligation to induce cholestasis, and infections (e.g., with the parasitic flatworm Schistosoma or hepatitis viruses) are also commonly used to generate animal models of liver fibrosis.46

Mouse models of reversible liver fibrosis

As murine livers typically require multiple insults to develop fibrotic MASH, removing these stimuli or restoring homeostasis through genetic tools can lead to fibrosis regression. This section focuses on mouse models in which liver fibrosis has been shown to be reversible. Table 1 lists models with reversible hepatic fibrosis induced by diet, hepatotoxins, genetic predisposition, bile duct atresia, and parasites.36,47–74

Table 1

Studies in mouse models of reversible liver fibrosis

Mouse strainFibrosis inductionFibrosis stageRegression methodConsiderationRef.
Diet models
C57BL/6HFD with 40% fat, 25% fructose, 2% cholesterol for eight months2Switch to a chow diet for 16 weeksRegression of portal but not perisinusoidal fibrosis (assessed via H&E staining) along with reduced steatosis and inflammation50
C57BL/6JGubra Amylin (GAN) diet (46% fat, 22% fructose, 10% sucrose, 2% cholesterol) for nine months≥1PPAR agonist lanifibranor (30 mg/kg orally per day) for another 12 weeks while still on the GAN dietFibrosis regression in only half of the lanifibranor-treated cohort (assessed via collagen I staining) along with reduced steatosis36
C57BL/6JCholine-deficient L-amino-defined (CDAA)-HFD for 8 weeks1Switch to a chow diet and a daily injection of MAIT cell inhibitor acetyl-6-formylpterin (Ac-6-FP) for eight days51
C57BL/6JHFD with 40% fat, 20% fructose, 10% sucrose, 2% cholesterol for 12 weeks1–2Nitro-oleic acid (OA-NO2) via minipump for another 12 weeks while still on HFD52
C57BL/6NMethionine-choline deficient (MCD) diet for seven weeks1PPARα agonist Wy-14,643 in the MCD diet for 12 daysMCD diet generally induces weight loss49
C57BL/6J with 129S1/SvImJHFD with 42% kcal from fat, 0.1% cholesterol, and drinking water with 23.1 g/L fructose and 18.9 g/L glucose for 40 to 52 weeks2–3Switch to a chow diet and normal drinking water for four weeksAlso known as diet-induced animal model of non-alcoholic fatty liver disease (DIAMOND mouse)53
Hepatotoxins
C57BL/6Jsingle s.c. injection of streptozotocin two days after birth followed by HFD feeding from four weeks of age1 (at nine weeks of age)Galectin-3 protein inhibitors (intravenously) for four weeks starting at nine weeks of age while still on HFDOnly modest fibrosis regression (assessed via Sirius Red staining) along with reduced steatosis54
C57BL/6CCl4 (0.5 mL/kg in olive oil, i.p. twice per week for 4 weeks)3Removal of CCl4 for one month55
C57BL/6JCCl4 (0.6 µL/g, 15% with olive oil i.p.) for six weeks3Removal of CCl4 for up to 14 days56
C57BL/6JEscalating doses of CCl4 (up to 1.25 ml/kg in corn oil, orally three times per week for eight weeks)3Removal of CCl4 for four weeks57
C57BL/6JCCl4 (1 mL/kg in olive oil, i.p. twice per week for four weeks)3Removal of CCl4 for six weeks58
C57BL/6JOlaHsdCCl4 (0.5 mg/kg in corn oil, i.p. twice per week for seven weeks)3Indoline derivative AN1284 (1 mg/kg/day for three weeks, four weeks after removal of CCl4)Reversal of more than 50% of fibrosis (assessed via Sirius Red and collagen IV staining)48
BALB/cCCl4 (diluted 1:8 v/v in corn oil, i.p. twice per week for 10 weeks)3Removal of CCl4 and treatment with macrophage-targeted PPARα agonist GW1929 (i.v. every three days for 10 days)Reversal of more than 70% of fibrosis (assessed via Sirius Red staining)59
BALB/cEscalating doses of CCl4 (up to 1 mL/kg in olive oil, i.p. twice per week for four weeks)3Removal of CCl4 and treatment with mIFNγ for two weeksNearly complete regression (assessed via collagen I gene expression and WNT-5A staining)60
BALB/cCCl4 (0.4 mL/kg in olive oil, i.p. twice per week for 6 weeks)2Removal of CCl4 and treatment with luteolin for two weeksNearly complete regression (assessed via Mallory trichrome staining)61
BALB/cTAA (200 mg/kg, i.p. every three days for four weeks)4Deactivating stellate cells using siRNA nanoparticle cocktail targeting Hedgehog and TGFβ1 (i.v. every three days, five times)Limited spontaneous regression (assessed via Mason’s Trichrome staining)47
MMP9-/- (FVB background)TAA (0.1 mg/g, i.p. every two days for eight weeks)n.d.Withdrawal of TAA for up to nine daysImproved regression after transplantation of wild-type Kupffer cells (assessed via Western blotting for collagen I, III, and IV)62
129/Svα-naphthylisothio-cyanate (ANIT; 75 mg/kg via the diet for 14 days)2PPARα activator fenofibrate (25 mg/kg via oral gavage, twice per day for 14 days while still on the ANIT diet)Cholestatic fibrosis model, stage 3 fibrosis after 28 days of ANIT diet (assessed via Sirius Red staining)63
Genetic models
NOD-Inflammation Fibrosis (N-IF, 24αβNOD. Rag2-/-)Spontaneous development of chronic inflammation and liver fibrosis3Anti-inflammatory Paquinimod (25 mg/kg body weight/day in the drinking water) for 10 weeksRag2-/- mice are severely immunodeficient and produce no mature B and T cells64
Conditional liver-specific expression of TGF-β1 (inhibited by doxycycline)10 cycles of TGF-β1 induction1TGF-β1 inhibition with doxycycline for 21 daysFluctuating body weight through doxycycline cycles65
Glycogen storage disease IIIa (Agl-/-)Progressive liver fibrosis associated with glycogen storage disease3AAV expressing the bacterial glycogen debranching enzyme pullulanase, reversal over 10 weeksOnly applicable to glycogen storage disease66
Peroxidasin knockout (Pxdn-/-)Choline-deficient L-amino-defined (CDAA)-HFD for 16 weeks1Switch to a chow diet for two weeksPeroxidasin deficiency results in atypical fibrosis formation (assessed via Sirius Red staining, collagen I staining, and electron microscopy)67
MUP-uPAHFD for six monthsHCCLorsatan (30 mg/kg in the drinking water) for another two months while still on HFD85% of MUP-uPA mice spontaneously progress to HCC when fed HFD68
Ldlr-/-.LeidenHFD with 45% fat from lard, 35% carbohydrates, 20% casein for 30 weeks1Running wheel access or switch to chow diet or a combination of both for 20 weeksLdlr-/- mice are also a model for atherosclerosis development69
Aryl hydrocarbon receptor knockout (AHR-/-)Fibrotic phenotype due to increased liver retinoid content1Vitamin A (retinol)-deficient diet for up to 18 weeks70
Mdr2-/- on FVB/NJ background (also called Abcb4 mice)Spontaneous sclerosing cholangitis2 (at 52 weeks of age)Hedgehog pathway inhibitor GDC-0449 (40 mg/kg, i.p., daily for nine days)Biliary fibrosis development (assessed via Sirius Red staining) from four weeks of age, spontaneous occurrence from six months on71
Biliary atresia
Collagen 1(α)1-GFPBile duct ligation for 14 days2Surgical gall bladder-jejunum shunt to bypass bile duct ligationRequires two surgical procedures72
Parasites
C57BL/6Schistosoma japonicum infectionFibro-cellular granulo-mas150–350 mg/kg praziquantel for five consecutive days seven weeks after the infectionNo steatosis development, mortality of 10-20% following infection73
Swiss albino CD-1Schistosoma mansoni infectionFibro-cellular granulo-masSingle intra-hepatic injection of Wharton’s jelly-derived mesenchymal stem cells combined with anti-helminth drug praziquantel (PZQ) eight to sixteen weeks after the infectionNo steatosis development, mortality of 10-20% following infection74

The most commonly used protocol to induce severe reversible hepatic fibrosis in mice involves the repeated intraperitoneal injection of CCl4, allowing for the study of fibrosis regression after the withdrawal of the hepatotoxin. Severe fibrosis can be achieved in a short timeframe (four to eight weeks) with this approach, and depending on the mouse strain, nearly complete fibrosis regression can be observed in a similar timeframe following CCl4 withdrawal. Fibrosis induced through thioacetamide treatment, on the other hand, appears less reversible. Lee et al. previously concluded that this model is less suitable for studying fibrosis regression.75 The general drawback of chemically induced liver fibrosis is that the metabolic phenotype, as well as the structure of the rapidly accumulating collagen deposits, may differ from human MASLD. Consequently, CCl4 exposure and withdrawal is a useful method to study the effects of gene therapy 47 or pharmacological treatments 48 targeting fibrogenic and fibrinolytic processes outside the context of the hepatic metabolic impairments seen in human MASLD. The choice of the genetic background of the mice hereby depends on prior data on the treatment in question and personal preference, as near-complete fibrosis regression can be achieved in both C57BL/6J and BALB/c mice with CCl4 withdrawal (see Table 1).

To mimic the slow development of fibrosis through hepatic metabolic dysfunction-induced inflammation seen in humans, modified HFDs are suitable in mice. However, the reversal of fibrosis through a switch from HFD to a regular chow diet is often incomplete and varies between individual animals, even after several months. Nevertheless, since incomplete and highly variable fibrosis regression is also observed clinically, this may not be a disadvantage of these mouse models and may instead mimic the clinical treatment response in addition to disease development.76,77 Accordingly, diets rich in fat, sucrose, fructose, and cholesterol, such as the Gubra Amylin diet—most commonly fed to mice on a C57BL/6J genetic background, are particularly useful for studying the effects of metabolic or inflammatory modulators on fibrosis regression. This may also include treatments that do not act directly on the liver but, for example, influence food intake.78 Unless particularly relevant to the research question, methionine-choline-deficient diets are less recommended for studying treatments in a human-like metabolic context of fibrotic MASLD, as they induce substantial weight loss due to adverse reactions.49 The applicability of genetic models, as well as biliary atresia and parasite-induced fibrosis models, highly depends on the research question, with the downside that advanced molecular gene-editing tools or surgical techniques are often required to induce and reverse fibrosis development. Among genetic models, major urinary protein-urokinase-type plasminogen activator mice are particularly versatile, as they allow for the study of liver fibrosis regression in the context of advanced liver disease and hepatocellular carcinoma, which is an important clinical outcome.26 The high mortality associated with parasitic infections makes these mouse models most useful for studying specific anti-parasitic treatments, where hepatic fibrosis regression may be a desirable side effect. Similarly, the effects of antiviral treatments on fibrosis may be best tested in models where hepatic fibrosis was induced by viral infections. In summary, the choice of the most appropriate mouse model for studying fibrosis regression depends heavily on the research question and whether the molecular processes of fibrosis regression are being studied in the context of inter-organ crosstalk, metabolic dysfunction, or other comorbidities.

Ex vivo and in vitro models to study fibrosis regression

To test anti-fibrotic compounds, the ex vivo treatment of human or murine precision-cut liver slices (PCLS) offers a well-controlled experimental setup.79,80 It was recently demonstrated that 250 µm thick PCLS from various diet-induced and hepatotoxin-induced liver fibrosis models remain stable in culture for up to 72 h, and gene expression responses to anti-fibrotic drugs matched those observed in liver tissue in vivo.81 Ongoing work focuses on extending the viability of these slices in vitro, as well as optimizing culturing conditions to induce and study the regression of fibrosis in PCLS.82 While this technique does not account for drug-induced changes in interorgan crosstalk or the liver’s physiological complexity—such as the influx of inflammatory cells or the modulation of the gut-liver axis through bacterial drug metabolism—it surpasses cell cultures of individual cell types, such as stellate cells, in terms of mechanistic insights. Similarly, organoids containing multiple or all cell types found in the liver are continuously being improved to accurately recapitulate human (patho)physiology on a miniature scale. Since these organoids can be generated from patient-derived induced pluripotent stem cells, they bypass the need for animal models and allow the development of personalized therapies. Organoids cultured using extracellular scaffolding matrices and self-assembling spheroids, both incorporating hepatic stellate cells, have demonstrated the development of fibrosis characterized by collagen deposits.83–85 However, as of now, there has been no documented regression of fibrosis in these models, possibly due to an altered response of stellate cells in vitro compared to in vivo, the limited representation of the liver’s immune environment, and the lack of vascularization, which provides essential oxygen and nutrient gradients in tissues. Lastly, profibrotic markers can be induced in MASH-on-chip models, where hepatocytes, HSCs, Kupffer cells, and endothelial cells are co-cultured under microfluidic dynamics.86,87 Modulating the stiffness of the hydrogel in which the cells are cultured has been shown to affect the activity of stellate cells, with a reduction in the expression of profibrotic markers such as α-smooth muscle actin as hydrogel stiffness decreases.88 This suggests that modeling fibrosis regression in organ-on-chip models may be achievable and cost-effective in setups where mechanical features are modifiable.89 In summary, ex vivo PCLS that preserve in vivo tissue structure are currently the most suitable models for studying the local response to anti-fibrotic treatments in the liver over short periods. However, the systemic interactions are not fully reflected in these models.

Outlook

Several technical advancements will allow for further refinement of in vivo studies on fibrosis regression in mice and potentially reduce the need for animal models through optimized in vitro setups. First, since MASLD and fibrosis grading in mice generally take place histologically in whole liver tissues collected postmortem, the non-invasive fibrosis quantification techniques used clinically, along with blood markers of liver fibrosis, need to be further adapted for mice. For this, the combination of magnetic resonance imaging (hereinafter referred to as MRI) and MRI-based elastography has been found to reliably predict hepatic collagen content in mice with CCl4-induced liver fibrosis, as well as fibrosis regression after CCl4 withdrawal.90 Additionally, the assessment of visco-elastic parameters using MRI-based elastography alone in a 5-minute scan in mice has shown good diagnostic performance for detecting substantial fibrosis, though it is less effective for diagnosing MASH.91,92 A serological miRNA-based scoring algorithm, developed for the clinical diagnosis of significant liver fibrosis in blood, was validated in mice and revealed a pronounced plasma enrichment of the miRNAs 451a, 142-5p, Let-7f-5p, and 378a-3p in mice with CCl4-induced liver fibrosis compared to mice with healthy livers. This suggests that the quantification of circulating miRNAs may aid in grading developing or regressing fibrosis.93 Lastly, while more invasive, repeated needle biopsies of the mouse liver for histological grading are possible, though they carry the downside of tissue damage at the biopsy site, which may locally accelerate fibrosis development.94 Together, these advancements are expected to enable reliable and repeated fibrosis assessments in the same mouse over time, thereby reducing the number of animals needed for these studies. Additionally, further refinement of mouse models is desirable, such as combining genetic and diet models to more closely mimic human liver fibrosis development and regression, particularly in the context of common comorbidities such as obesity, type 2 diabetes, and cardiovascular diseases in the case of MASH-induced liver fibrosis.

Declarations

Funding

MS is supported by the European Foundation for the Study of Diabetes and the Leiden University Fund. PCNR is supported by The Netherlands Cardiovascular Research Initiative CVON-GENIUS-2 supported by the Dutch Heart Foundation.

Conflict of interest

The authors have no conflict of interests related to this publication.

Authors’ contributions

MS wrote the original draft, and MS and PCNR revised and edited the manuscript. All authors have approved the final version and the publication of the manuscript.

References

  1. Caussy C, Aubin A, Loomba R. The Relationship Between Type 2 Diabetes, NAFLD, and Cardiovascular Risk. Curr Diab Rep 2021;21(5):15 View Article PubMed/NCBI
  2. Gines P, Krag A, Abraldes JG, Sola E, Fabrellas N, Kamath PS. Liver cirrhosis. Lancet 2021;398(10308):1359-1376 View Article PubMed/NCBI
  3. Mehta KJ, Farnaud SJ, Sharp PA. Iron and liver fibrosis: Mechanistic and clinical aspects. World J Gastroenterol 2019;25(5):521-538 View Article PubMed/NCBI
  4. Friedman SL, Neuschwander-Tetri BA, Rinella M, Sanyal AJ. Mechanisms of NAFLD development and therapeutic strategies. Nat Med 2018;24(7):908-922 View Article PubMed/NCBI
  5. Tien C, Remulla D, Kwon Y, Emamaullee J. Contemporary strategies to assess and manage liver donor steatosis: a review. Curr Opin Organ Transplant 2021;26(5):474-481 View Article PubMed/NCBI
  6. Harrison SA, Bedossa P, Guy CD, Schattenberg JM, Loomba R, Taub R, et al. A Phase 3, Randomized, Controlled Trial of Resmetirom in NASH with Liver Fibrosis. N Engl J Med 2024;390(6):497-509 View Article PubMed/NCBI
  7. Sookoian S, Pirola CJ. Resmetirom for treatment of MASH. Cell 2024;187(12):2897-2897.e2891 View Article PubMed/NCBI
  8. Loomba R, Sanyal AJ, Kowdley KV, Bhatt DL, Alkhouri N, Frias JP, et al. Randomized, Controlled Trial of the FGF21 Analogue Pegozafermin in NASH. N Engl J Med 2023;389(11):998-1008 View Article PubMed/NCBI
  9. Loomba R, Hartman ML, Lawitz EJ, Vuppalanchi R, Boursier J, Bugianesi E, et al. Tirzepatide for Metabolic Dysfunction-Associated Steatohepatitis with Liver Fibrosis. N Engl J Med 2024;391:299-310 View Article PubMed/NCBI
  10. Sanyal AJ, Bedossa P, Fraessdorf M, Neff GW, Lawitz E, Bugianesi E, et al. A Phase 2 Randomized Trial of Survodutide in MASH and Fibrosis. N Engl J Med 2024;391:311-319 View Article PubMed/NCBI
  11. Cools L, Kazemzadeh Dastjerd M, Smout A, Merens V, Yang Y, Reynaert H, et al. Human iPSC-derived liver co-culture spheroids to model liver fibrosis. Biofabrication 2024;16(3):035032 View Article PubMed/NCBI
  12. Wirth EK, Puengel T, Spranger J, Tacke F. Thyroid hormones as a disease modifier and therapeutic target in nonalcoholic steatohepatitis. Expert Rev Endocrinol Metab 2022;17(5):425-434 View Article PubMed/NCBI
  13. Pinzani M. Pathophysiology of Liver Fibrosis. Dig Dis 2015;33(4):492-497 View Article PubMed/NCBI
  14. Tacke F. Targeting hepatic macrophages to treat liver diseases. J Hepatol 2017;66(6):1300-1312 View Article PubMed/NCBI
  15. Wang Z, Du K, Jin N, Tang B, Zhang W. Macrophage in liver Fibrosis: Identities and mechanisms. Int Immunopharmacol 2023;120:110357 View Article PubMed/NCBI
  16. Younossi ZM, Loomba R, Anstee QM, Rinella ME, Bugianesi E, Marchesini G, et al. Diagnostic modalities for nonalcoholic fatty liver disease, nonalcoholic steatohepatitis, and associated fibrosis. Hepatology 2018;68(1):349-360 View Article PubMed/NCBI
  17. Heyens LJM, Busschots D, Koek GH, Robaeys G, Francque S. Liver Fibrosis in Non-alcoholic Fatty Liver Disease: From Liver Biopsy to Non-invasive Biomarkers in Diagnosis and Treatment. Front Med (Lausanne) 2021;8:615978 View Article PubMed/NCBI
  18. Liang W, Menke AL, Driessen A, Koek GH, Lindeman JH, Stoop R, et al. Establishment of a general NAFLD scoring system for rodent models and comparison to human liver pathology. PLoS One 2014;9(12):e115922 View Article PubMed/NCBI
  19. Lee MJ. A review of liver fibrosis and cirrhosis regression. J Pathol Transl Med 2023;57(4):189-195 View Article PubMed/NCBI
  20. Younossi ZM, Paik JM, Stepanova M, Ong J, Alqahtani S, Henry L. Clinical profiles and mortality rates are similar for metabolic dysfunction-associated steatotic liver disease and non-alcoholic fatty liver disease. J Hepatol 2024;80(5):694-701 View Article PubMed/NCBI
  21. Denk H, Abuja PM, Zatloukal K. Animal models of NAFLD from the pathologist’s point of view. Biochim Biophys Acta Mol Basis Dis 2019;1865(5):929-942 View Article PubMed/NCBI
  22. Cast A, Kumbaji M, D’Souza A, Rodriguez K, Gupta A, Karns R, et al. Liver Proliferation Is an Essential Driver of Fibrosis in Mouse Models of Nonalcoholic Fatty Liver Disease. Hepatol Commun 2019;3(8):1036-1049 View Article PubMed/NCBI
  23. Savard C, Tartaglione EV, Kuver R, Haigh WG, Farrell GC, Subramanian S, et al. Synergistic interaction of dietary cholesterol and dietary fat in inducing experimental steatohepatitis. Hepatology 2013;57(1):81-92 View Article PubMed/NCBI
  24. Gallage S, Avila JEB, Ramadori P, Focaccia E, Rahbari M, Ali A, et al. A researcher’s guide to preclinical mouse NASH models. Nat Metab 2022;4(12):1632-1649 View Article PubMed/NCBI
  25. Flessa CM, Nasiri-Ansari N, Kyrou I, Leca BM, Lianou M, Chatzigeorgiou A, et al. Genetic and Diet-Induced Animal Models for Non-Alcoholic Fatty Liver Disease (NAFLD) Research. Int J Mol Sci 2022;23(24):15791 View Article PubMed/NCBI
  26. Febbraio MA, Reibe S, Shalapour S, Ooi GJ, Watt MJ, Karin M. Preclinical Models for Studying NASH-Driven HCC: How Useful Are They?. Cell Metab 2019;29(1):18-26 View Article PubMed/NCBI
  27. Phung HH, Lee CH. Mouse models of nonalcoholic steatohepatitis and their application to new drug development. Arch Pharm Res 2022;45(11):761-794 View Article PubMed/NCBI
  28. St Rose K, Yan J, Xu F, Williams J, Dweck V, Saxena D, et al. Mouse model of NASH that replicates key features of the human disease and progresses to fibrosis stage 3. Hepatol Commun 2022;6(10):2676-2688 View Article PubMed/NCBI
  29. Delire B, Starkel P, Leclercq I. Animal Models for Fibrotic Liver Diseases: What We Have, What We Need, and What Is under Development. J Clin Transl Hepatol 2015;3(1):53-66 View Article PubMed/NCBI
  30. Oligschlaeger Y, Shiri-Sverdlov R. NAFLD Preclinical Models: More than a Handful, Less of a Concern?. Biomedicines 2020;8(2):28 View Article PubMed/NCBI
  31. Ito M, Suzuki J, Tsujioka S, Sasaki M, Gomori A, Shirakura T, et al. Longitudinal analysis of murine steatohepatitis model induced by chronic exposure to high-fat diet. Hepatol Res 2007;37(1):50-57 View Article PubMed/NCBI
  32. Velazquez KT, Enos RT, Bader JE, Sougiannis AT, Carson MS, Chatzistamou I, et al. Prolonged high-fat-diet feeding promotes non-alcoholic fatty liver disease and alters gut microbiota in mice. World J Hepatol 2019;11(8):619-637 View Article PubMed/NCBI
  33. Nakano Y, Kamiya A, Sumiyoshi H, Tsuruya K, Kagawa T, Inagaki Y. A Deactivation Factor of Fibrogenic Hepatic Stellate Cells Induces Regression of Liver Fibrosis in Mice. Hepatology 2020;71(4):1437-1452 View Article PubMed/NCBI
  34. Rangnekar AS, Lammert F, Igolnikov A, Green RM. Quantitative trait loci analysis of mice administered the methionine-choline deficient dietary model of experimental steatohepatitis. Liver Int 2006;26(8):1000-1005 View Article PubMed/NCBI
  35. Im YR, Hunter H, de Gracia Hahn D, Duret A, Cheah Q, Dong J, et al. A Systematic Review of Animal Models of NAFLD Finds High-Fat, High-Fructose Diets Most Closely Resemble Human NAFLD. Hepatology 2021;74(4):1884-1901 View Article PubMed/NCBI
  36. Mollerhoj MB, Veidal SS, Thrane KT, Oro D, Overgaard A, Salinas CG, et al. Hepatoprotective effects of semaglutide, lanifibranor and dietary intervention in the GAN diet-induced obese and biopsy-confirmed mouse model of NASH. Clin Transl Sci 2022;15(5):1167-1186 View Article PubMed/NCBI
  37. Boland ML, Oro D, Tolbol KS, Thrane ST, Nielsen JC, Cohen TS, et al. Towards a standard diet-induced and biopsy-confirmed mouse model of non-alcoholic steatohepatitis: Impact of dietary fat source. World J Gastroenterol 2019;25(33):4904-4920 View Article PubMed/NCBI
  38. Hansen HH, HM AE, Oro D, Evers SS, Heeboll S, Eriksen PL, et al. Human translatability of the GAN diet-induced obese mouse model of non-alcoholic steatohepatitis. BMC Gastroenterol 2020;20(1):210 View Article PubMed/NCBI
  39. Bieghs V, Van Gorp PJ, Wouters K, Hendrikx T, Gijbels MJ, van Bilsen M, et al. LDL receptor knock-out mice are a physiological model particularly vulnerable to study the onset of inflammation in non-alcoholic fatty liver disease. PLoS One 2012;7(1):e30668 View Article PubMed/NCBI
  40. Schierwagen R, Maybuchen L, Zimmer S, Hittatiya K, Back C, Klein S, et al. Seven weeks of Western diet in apolipoprotein-E-deficient mice induce metabolic syndrome and non-alcoholic steatohepatitis with liver fibrosis. Sci Rep 2015;5:12931 View Article PubMed/NCBI
  41. Hui ST, Kurt Z, Tuominen I, Norheim F, Davis RC, Pan C, et al. The Genetic Architecture of Diet-Induced Hepatic Fibrosis in Mice. Hepatology 2018;68(6):2182-2196 View Article PubMed/NCBI
  42. Hansen HH, Feigh M, Veidal SS, Rigbolt KT, Vrang N, Fosgerau K. Mouse models of nonalcoholic steatohepatitis in preclinical drug development. Drug Discov Today 2017;22(11):1707-1718 View Article PubMed/NCBI
  43. Kristiansen MN, Veidal SS, Rigbolt KT, Tolbol KS, Roth JD, Jelsing J, et al. Obese diet-induced mouse models of nonalcoholic steatohepatitis-tracking disease by liver biopsy. World J Hepatol 2016;8(16):673-684 View Article PubMed/NCBI
  44. Nakagawa H. Recent advances in mouse models of obesity- and nonalcoholic steatohepatitis-associated hepatocarcinogenesis. World J Hepatol 2015;7(17):2110-2118 View Article PubMed/NCBI
  45. Nakagawa H, Umemura A, Taniguchi K, Font-Burgada J, Dhar D, Ogata H, et al. ER stress cooperates with hypernutrition to trigger TNF-dependent spontaneous HCC development. Cancer Cell 2014;26(3):331-343 View Article PubMed/NCBI
  46. Wu S, Wang X, Xing W, Li F, Liang M, Li K, et al. An update on animal models of liver fibrosis. Front Med (Lausanne) 2023;10:1160053 View Article PubMed/NCBI
  47. Younis MA, Sato Y, Elewa YHA, Harashima H. Reprogramming activated hepatic stellate cells by siRNA-loaded nanocarriers reverses liver fibrosis in mice. J Control Release 2023;361:592-603 View Article PubMed/NCBI
  48. Yehezkel AS, Abudi N, Nevo Y, Benyamini H, Elgavish S, Weinstock M, et al. AN1284 attenuates steatosis, lipogenesis, and fibrosis in mice with pre-existing non-alcoholic steatohepatitis and directly affects aryl hydrocarbon receptor in a hepatic cell line. Front Endocrinol (Lausanne) 2023;14:1226808 View Article PubMed/NCBI
  49. Ip E, Farrell G, Hall P, Robertson G, Leclercq I. Administration of the potent PPARalpha agonist, Wy-14,643, reverses nutritional fibrosis and steatohepatitis in mice. Hepatology 2004;39(5):1286-1296 View Article PubMed/NCBI
  50. Ding ZM, Xiao Y, Wu X, Zou H, Yang S, Shen Y, et al. Progression and Regression of Hepatic Lesions in a Mouse Model of NASH Induced by Dietary Intervention and Its Implications in Pharmacotherapy. Front Pharmacol 2018;9:410 View Article PubMed/NCBI
  51. Mabire M, Hegde P, Hammoutene A, Wan J, Caer C, Sayegh RA, et al. MAIT cell inhibition promotes liver fibrosis regression via macrophage phenotype reprogramming. Nat Commun 2023;14(1):1830 View Article PubMed/NCBI
  52. Rom O, Xu G, Guo Y, Zhu Y, Wang H, Zhang J, et al. Nitro-fatty acids protect against steatosis and fibrosis during development of nonalcoholic fatty liver disease in mice. EBioMedicine 2019;41:62-72 View Article PubMed/NCBI
  53. Ng N, Tai D, Ren Y, Chng E, Seneshaw M, Mirshahi F, et al. Second-Harmonic Generated Quantifiable Fibrosis Parameters Provide Signatures for Disease Progression and Regression in Nonalcoholic Fatty Liver Disease. Clin Pathol 2023;16:2632010X231162317 View Article PubMed/NCBI
  54. Traber PG, Zomer E. Therapy of experimental NASH and fibrosis with galectin inhibitors. PLoS One 2013;8(12):e83481 View Article PubMed/NCBI
  55. Arroyo N, Villamayor L, Diaz I, Carmona R, Ramos-Rodriguez M, Munoz-Chapuli R, et al. GATA4 induces liver fibrosis regression by deactivating hepatic stellate cells. JCI Insight 2021;6(23):e150059 View Article PubMed/NCBI
  56. Song P, Duan JL, Ding J, Liu JJ, Fang ZQ, Xu H, et al. Cellular senescence primes liver fibrosis regression through Notch-EZH2. MedComm (2020) 2023;4(5):e346 View Article PubMed/NCBI
  57. Yong H, Shan S, Wang S, Liu Z, Liu Z, Zhang C, et al. Activation of mitophagy by rapamycin eliminated the accumulation of TDP-43 on mitochondrial and promoted the resolution of carbon tetrachloride-induced liver fibrosis in mice. Toxicology 2022;471:153176 View Article PubMed/NCBI
  58. Zhao H, Zhu H, Zhang Y, Ding Y, Feng R, Li J, et al. Lymphocyte-Specific Protein Tyrosine Kinase Contributes to Spontaneous Regression of Liver Fibrosis may by Interacting with Suppressor of Cytokine Signaling 1. Inflammation 2023;46(5):1653-1669 View Article PubMed/NCBI
  59. Moreno-Lanceta A, Medrano-Bosch M, Simon-Codina B, Barber-Gonzalez M, Jimenez W, Melgar-Lesmes P. PPAR-gamma Agonist GW1929 Targeted to Macrophages with Dendrimer-Graphene Nanostars Reduces Liver Fibrosis and Inflammation. Pharmaceutics 2023;15(5):1452 View Article PubMed/NCBI
  60. Beljaars L, Daliri S, Dijkhuizen C, Poelstra K, Gosens R. WNT-5A regulates TGF-beta-related activities in liver fibrosis. Am J Physiol Gastrointest Liver Physiol 2017;312(3):G219-G227 View Article PubMed/NCBI
  61. Domitrovic R, Jakovac H, Tomac J, Sain I. Liver fibrosis in mice induced by carbon tetrachloride and its reversion by luteolin. Toxicol Appl Pharmacol 2009;241(3):311-321 View Article PubMed/NCBI
  62. Feng M, Ding J, Wang M, Zhang J, Zhu X, Guan W. Kupffer-derived matrix metalloproteinase-9 contributes to liver fibrosis resolution. Int J Biol Sci 2018;14(9):1033-1040 View Article PubMed/NCBI
  63. Lu Z, Li S, Luo J, Luo Y, Dai M, Zheng X, et al. Fenofibrate reverses liver fibrosis in cholestatic mice induced by alpha-naphthylisothiocyanate. Pharmazie 2021;76(2):103-108 View Article PubMed/NCBI
  64. Fransen Pettersson N, Deronic A, Nilsson J, Hannibal TD, Hansen L, Schmidt-Christensen A, et al. The immunomodulatory quinoline-3-carboxamide paquinimod reverses established fibrosis in a novel mouse model for liver fibrosis. PLoS One 2018;13(9):e0203228 View Article PubMed/NCBI
  65. Ueberham E, Low R, Ueberham U, Schonig K, Bujard H, Gebhardt R. Conditional tetracycline-regulated expression of TGF-beta1 in liver of transgenic mice leads to reversible intermediary fibrosis. Hepatology 2003;37(5):1067-1078 View Article PubMed/NCBI
  66. Lim JA, Kishnani PS, Sun B. Suppression of pullulanase-induced cytotoxic T cell response with a dual promoter in GSD IIIa mice. JCI Insight 2022;7:23 View Article PubMed/NCBI
  67. Sojoodi M, Erstad DJ, Barrett SC, Salloum S, Zhu S, Qian T, et al. Peroxidasin Deficiency Re-programs Macrophages Toward Pro-fibrolysis Function and Promotes Collagen Resolution in Liver. Cell Mol Gastroenterol Hepatol 2022;13(5):1483-1509 View Article PubMed/NCBI
  68. Gu L, Zhu Y, Lee M, Nguyen A, Ryujin NT, Huang J, et al. Angiotensin II receptor inhibition ameliorates liver fibrosis and enhances hepatocellular carcinoma infiltration by effector T cells. bioRxiv 2023 View Article PubMed/NCBI
  69. van den Hoek AM, de Jong J, Worms N, van Nieuwkoop A, Voskuilen M, Menke AL, et al. Diet and exercise reduce pre-existing NASH and fibrosis and have additional beneficial effects on the vasculature, adipose tissue and skeletal muscle via organ-crosstalk. Metabolism 2021;124:154873 View Article PubMed/NCBI
  70. Andreola F, Calvisi DF, Elizondo G, Jakowlew SB, Mariano J, Gonzalez FJ, et al. Reversal of liver fibrosis in aryl hydrocarbon receptor null mice by dietary vitamin A depletion. Hepatology 2004;39(1):157-166 View Article PubMed/NCBI
  71. Philips GM, Chan IS, Swiderska M, Schroder VT, Guy C, Karaca GF, et al. Hedgehog signaling antagonist promotes regression of both liver fibrosis and hepatocellular carcinoma in a murine model of primary liver cancer. PLoS One 2011;6(9):e23943 View Article PubMed/NCBI
  72. Yoshino K, Taura K, Iwaisako K, Masano Y, Uemoto Y, Kimura Y, et al. Novel mouse model for cholestasis-induced liver fibrosis resolution by cholecystojejunostomy. J Gastroenterol Hepatol 2021;36(9):2493-2500 View Article PubMed/NCBI
  73. Chuah C, Jones MK, McManus DP, Nawaratna SK, Burke ML, Owen HC, et al. Characterising granuloma regression and liver recovery in a murine model of schistosomiasis japonica. Int J Parasitol 2016;46(4):239-252 View Article PubMed/NCBI
  74. Hammam OA, Elkhafif N, Attia YM, Mansour MT, Elmazar MM, Abdelsalam RM, et al. Wharton’s jelly-derived mesenchymal stem cells combined with praziquantel as a potential therapy for Schistosoma mansoni-induced liver fibrosis. Sci Rep 2016;6:21005 View Article PubMed/NCBI
  75. Lee YS, Seki E. In Vivo and In Vitro Models to Study Liver Fibrosis: Mechanisms and Limitations. Cell Mol Gastroenterol Hepatol 2023;16(3):355-367 View Article PubMed/NCBI
  76. Ellis EL, Mann DA. Clinical evidence for the regression of liver fibrosis. J Hepatol 2012;56(5):1171-1180 View Article PubMed/NCBI
  77. Rowe IA, Parker R. The Placebo Response in Randomized Trials in Nonalcoholic Steatohepatitis Simply Explained. Clin Gastroenterol Hepatol 2022;20(3):e564-e572 View Article PubMed/NCBI
  78. Nestor JJ, Parkes D, Feigh M, Suschak JJ, Harris MS. Effects of ALT-801, a GLP-1 and glucagon receptor dual agonist, in a translational mouse model of non-alcoholic steatohepatitis. Sci Rep 2022;12(1):6666 View Article PubMed/NCBI
  79. Westra IM, Pham BT, Groothuis GM, Olinga P. Evaluation of fibrosis in precision-cut tissue slices. Xenobiotica 2013;43(1):98-112 View Article PubMed/NCBI
  80. Olinga P, Schuppan D. Precision-cut liver slices: a tool to model the liver ex vivo. J Hepatol 2013;58(6):1252-1253 View Article PubMed/NCBI
  81. Wang Y, Leaker B, Qiao G, Sojoodi M, Eissa IR, Epstein ET, et al. Precision-Cut Liver Slices as an ex vivo model to evaluate antifibrotic therapies for liver fibrosis and cirrhosis. bioRxiv 2023 View Article PubMed/NCBI
  82. Paish HL, Reed LH, Brown H, Bryan MC, Govaere O, Leslie J, et al. A Bioreactor Technology for Modeling Fibrosis in Human and Rodent Precision-Cut Liver Slices. Hepatology 2019;70(4):1377-1391 View Article PubMed/NCBI
  83. Tsang HY, Lo PHY, Lee KKH. Generation of liver organoids from human induced pluripotent stem cells as liver fibrosis and steatosis models. bioRxiv 2021 View Article
  84. Pingitore P, Sasidharan K, Ekstrand M, Prill S, Linden D, Romeo S. Human Multilineage 3D Spheroids as a Model of Liver Steatosis and Fibrosis. Int J Mol Sci 2019;20(7):1629 View Article PubMed/NCBI
  85. Leite SB, Roosens T, El Taghdouini A, Mannaerts I, Smout AJ, Najimi M, et al. Novel human hepatic organoid model enables testing of drug-induced liver fibrosis in vitro. Biomaterials 2016;78:1-10 View Article PubMed/NCBI
  86. Deguchi S, Takayama K. State-of-the-art liver disease research using liver-on-a-chip. Inflamm Regen 2022;42(1):62 View Article PubMed/NCBI
  87. Freag MS, Namgung B, Reyna Fernandez ME, Gherardi E, Sengupta S, Jang HL. Human Nonalcoholic Steatohepatitis on a Chip. Hepatol Commun 2021;5(2):217-233 View Article PubMed/NCBI
  88. Caliari SR, Perepelyuk M, Soulas EM, Lee GY, Wells RG, Burdick JA. Gradually softening hydrogels for modeling hepatic stellate cell behavior during fibrosis regression. Integr Biol (Camb) 2016;8(6):720-728 View Article PubMed/NCBI
  89. Carvalho AM, Bansal R, Barrias CC, Sarmento B. The Material World of 3D-Bioprinted and Microfluidic-Chip Models of Human Liver Fibrosis. Adv Mater 2024;36(2):e2307673 View Article PubMed/NCBI
  90. Chen J, Martin-Mateos R, Li J, Yin Z, Chen J, Lu X, et al. Multiparametric magnetic resonance imaging/magnetic resonance elastography assesses progression and regression of steatosis, inflammation, and fibrosis in alcohol-associated liver disease. Alcohol Clin Exp Res 2021;45(10):2103-2117 View Article PubMed/NCBI
  91. Khalfallah M, Doblas S, Hammoutene A, Julea F, Postic C, Valla D, et al. Visco-Elastic Parameters at Three-Dimensional MR Elastography for Diagnosing Non-Alcoholic Steatohepatitis and Substantial Fibrosis in Mice. J Magn Reson Imaging 2024;59(1):97-107 View Article PubMed/NCBI
  92. Yin M, Woollard J, Wang X, Torres VE, Harris PC, Ward CJ, et al. Quantitative assessment of hepatic fibrosis in an animal model with magnetic resonance elastography. Magn Reson Med 2007;58(2):346-353 View Article PubMed/NCBI
  93. Lambrecht J, Verhulst S, Reynaert H, van Grunsven LA. The miRFIB-Score: A Serological miRNA-Based Scoring Algorithm for the Diagnosis of Significant Liver Fibrosis. Cells 2019;8(9):1003 View Article PubMed/NCBI
  94. Shao W, Ichimura-Shimizu M, Ogawa H, Jin S, Sutoh M, Nakamura S, et al. Establishment of repeated liver biopsy technique in experimental mice. Heliyon 2023;9(6):e16978 View Article PubMed/NCBI
  • Journal of Clinical and Translational Hepatology
  • pISSN 2225-0719
  • eISSN 2310-8819
Back to Top

Mouse Models for the Study of Liver Fibrosis Regression In Vivo and Ex Vivo

Milena Schönke, Patrick C.N. Rensen
  • Reset Zoom
  • Download TIFF