Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.
Advertisement
Nature Metabolism (2022)
17 Altmetric
Metrics details
Non-alcoholic fatty liver disease (NAFLD) and its inflammatory form, non-alcoholic steatohepatitis (NASH), have quickly risen to become the most prevalent chronic liver disease in the Western world and are risk factors for the development of hepatocellular carcinoma (HCC). HCC is not only one of the most common cancers but is also highly lethal. Nevertheless, there are currently no clinically approved drugs for NAFLD, and NASH-induced HCC poses a unique metabolic microenvironment that may influence responsiveness to certain treatments. Therefore, there is an urgent need to better understand the pathogenesis of this rampant disease to devise new therapies. In this line, preclinical mouse models are crucial tools to investigate mechanisms as well as novel treatment modalities during the pathogenesis of NASH and subsequent HCC in preparation for human clinical trials. Although, there are numerous genetically induced, diet-induced and toxin-induced models of NASH, not all of these models faithfully phenocopy and mirror the human pathology very well. In this Perspective, we shed some light onto the most widely used mouse models of NASH and highlight some of the key advantages and disadvantages of the various models with an emphasis on ‘Western diets’, which are increasingly recognized as some of the best models in recapitulating the human NASH pathology and comorbidities.
This is a preview of subscription content, access via your institution

Subscribe to Journal
Get full journal access for 1 year
92,52 €
only 7,71 € per issue
All prices are NET prices.
VAT will be added later in the checkout.
Tax calculation will be finalised during checkout.
Buy article
Get time limited or full article access on ReadCube.
$32.00
All prices are NET prices.
Oeppen, J. & Vaupel, J. W. Demography. Broken limits to life expectancy. Science 296, 1029–1031 (2002).
Article  Google Scholar 
Tchkonia, T., Zhu, Y., van Deursen, J., Campisi, J. & Kirkland, J. L. Cellular senescence and the senescent secretory phenotype: therapeutic opportunities. J. Clin. Invest. 123, 966–972 (2013).
Article  Google Scholar 
Bluher, M. Obesity: global epidemiology and pathogenesis. Nat. Rev. Endocrinol. 15, 288–298 (2019).
Article  Google Scholar 
Anstee, Q. M., Reeves, H. L., Kotsiliti, E., Govaere, O. & Heikenwalder, M. From NASH to HCC: current concepts and future challenges. Nat. Rev. Gastroenterol. Hepatol. 16, 411–428 (2019).
Article  Google Scholar 
Gallage, S. et al. The therapeutic landscape of hepatocellular carcinoma. Med 2, 505–552 (2021).
Article  Google Scholar 
Sung, H. et al. Global Cancer Statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 71, 209–249 (2021).
Article  Google Scholar 
Estes, C. et al. Modeling NAFLD disease burden in China, France, Germany, Italy, Japan, Spain, United Kingdom, and United States for the period 2016–2030. J. Hepatol. 69, 896–904 (2018).
Article  Google Scholar 
Younossi, Z. et al. Global perspectives on non-alcoholic fatty liver disease and non-alcoholic steatohepatitis. Hepatology 69, 2672–2682 (2019).
Article  Google Scholar 
Huang, D. Q., El-Serag, H. B. & Loomba, R. Global epidemiology of NAFLD-related HCC: trends, predictions, risk factors and prevention. Nat. Rev. Gastroenterol. Hepatol. 18, 223–238 (2021).
Article  Google Scholar 
Huang, D. Q. et al. Changing global epidemiology of liver cancer from 2010 to 2019: NASH is the fastest growing cause of liver cancer. Cell Metab. https://doi.org/10.1016/j.cmet.2022.05.003 (2022).
Dudek, M. et al. Auto-aggressive CXCR6+ CD8+ T cells cause liver immune pathology in NASH. Nature 592, 444–449 (2021).
Article  Google Scholar 
Leslie, J. et al. CXCR2 inhibition enables NASH–HCC immunotherapy. Gut https://doi.org/10.1136/gutjnl-2021-326259 (2022).
Pfister, D. et al. NASH limits anti-tumour surveillance in immunotherapy-treated HCC. Nature 592, 450–456 (2021). This paper demonstrates that anti-PD-1 immunotherapy is not effective against NASH-induced HCC in mouse NASH models.
Wabitsch, S. et al. Metformin treatment rescues CD8+ T cell response to immune checkpoint inhibitor therapy in mice with NAFLD. J. Hepatol. https://doi.org/10.1016/j.jhep.2022.03.010 (2022). This paper shows that metformin treatment cansensitize NASH-induced HCCs to anti-PD-1 immunotherapy in preclinical mouse models.
Anstee, Q. M. & Goldin, R. D. Mouse models in non-alcoholic fatty liver disease and steatohepatitis research. Int J. Exp. Pathol. 87, 1–16 (2006).
Article  Google Scholar 
Oligschlaeger, Y. & Shiri-Sverdlov, R. NAFLD Preclinical models: more than a handful, less of a concern? Biomedicines https://doi.org/10.3390/biomedicines8020028 (2020).
Santhekadur, P. K., Kumar, D. P. & Sanyal, A. J. Preclinical models of non-alcoholic fatty liver disease. J. Hepatol. 68, 230–237 (2018).
Article  Google Scholar 
Febbraio, M. A. et al. Preclinical models for studying NASH-driven HCC: how useful are they. Cell Metab. 29, 18–26 (2019).
Article  Google Scholar 
Farrell, G. et al. Mouse models of non-alcoholic steatohepatitis: toward optimization of their relevance to human non-alcoholic steatohepatitis. Hepatology 69, 2241–2257 (2019).
Article  Google Scholar 
Haczeyni, F. et al. Mouse models of non-alcoholic steatohepatitis: a reflection on recent literature. J. Gastroenterol. Hepatol. 33, 1312–1320 (2018).
Article  Google Scholar 
Mann, J. P., Semple, R. K. & Armstrong, M. J. How useful are monogenic rodent models for the study of human non-alcoholic fatty liver disease? Front Endocrinol. 7, 145 (2016).
Article  Google Scholar 
Brunt, E. M. et al. Complexity of ballooned hepatocyte feature recognition: defining a training atlas for artificial intelligence-based imaging in NAFLD. J. Hepatol. 76, 1030–1041 (2022). This paper i llustrates the difficulty in identifying ballooned hepatocytes even by expert histopathologists and the potential of AI imaging.
Article  Google Scholar 
Ramos, M. J., Bandiera, L., Menolascina, F. & Fallowfield, J. A. In vitro models for non-alcoholic fatty liver disease: emerging platforms and their applications. iScience 25, 103549 (2022).
Article  Google Scholar 
Ströbel, S. et al. A 3D primary human cell-based in vitro model of non-alcoholic steatohepatitis for efficacy testing of clinical drug candidates. Sci. Rep. 11, 22765 (2021).
Article  Google Scholar 
Bissig-Choisat, B. et al. A human liver chimeric mouse model for non-alcoholic fatty liver disease. JHEP Rep. 3, 100281 (2021).
Article  Google Scholar 
Ma, J. et al. A novel humanized model of NASH and its treatment with META4, a potent agonist of MET. Cell Mol. Gastroenterol. Hepatol. 13, 565–582 (2022). The authors of this study developed a novel humanized model of NASH withhuman hepatocytes.
Article  Google Scholar 
Gallage, S. et al. Spontaneous cholemia in C57BL/6 mice predisposes to liver cancer in NASH. Cell Mol. Gastroenterol. Hepatol. 13, 875–878 (2022). This paper demonstrates that a subset of all C57BL/6 mice from either commercial breeders or inbred strains shows spontaneous cholaemia that can predispose them to liver cancer following high-caloric feeding.
Article  Google Scholar 
Glode, L. M. & Rosenstreich, D. L. Genetic control of B cell activation by bacterial lipopolysaccharide is mediated by multiple distinct genes or alleles. J. Immunol. 117, 2061–2066 (1976).
Google Scholar 
Fan, R., Wang, J. & Du, J. Association between body mass index and fatty liver risk: a dose-response analysis. Sci. Rep. 8, 15273 (2018).
Article  Google Scholar 
Im, Y. R. et al. A systematic review of animal models of NAFLD finds high-fat, high-fructose diets most closely resemble human NAFLD. Hepatology 74, 1884–1901 (2021).
Article  Google Scholar 
Trevaskis, J. L. et al. Glucagon-like peptide-1 receptor agonism improves metabolic, biochemical, and histopathological indices of non-alcoholic steatohepatitis in mice. Am. J. Physiol. Gastrointest. Liver Physiol. 302, G762–G772 (2012).
Article  Google Scholar 
Clapper, J. R. et al. Diet-induced mouse model of fatty liver disease and non-alcoholic steatohepatitis reflecting clinical disease progression and methods of assessment. Am. J. Physiol. Gastrointest. Liver Physiol. 305, G483–G495 (2013). The authors developed the AMLN Western-diet mouse model of NASH.
Radhakrishnan, S., Yeung, S. F., Ke, J. Y., Antunes, M. M. & Pellizzon, M. A. Considerations when choosing high-fat, high-fructose and high-cholesterol diets to induce experimental non-alcoholic fatty liver disease in laboratory animal models. Curr. Dev. Nutr. 5, nzab138 (2021).
Article  Google Scholar 
Boland, M. L. et al. Towards a standard diet-induced and biopsy-confirmed mouse model of non-alcoholic steatohepatitis: impact of dietary fat source. World J. Gastroenterol. 25, 4904–4920 (2019). The authors developed the GAN Western-diet mouse model of NASH.
Hansen, H. H. et al. Human translatability of the GAN diet-induced obese mouse model of non-alcoholic steatohepatitis. BMC Gastroenterol. 20, 210 (2020).
Article  Google Scholar 
Mollerhoj, M. B. 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. 15, 1167–1186 (2022).
Article  Google Scholar 
Drescher, H. K. et al. The influence of different fat sources on steatohepatitis and fibrosis development in the Western diet mouse model of non-alcoholic steatohepatitis (NASH. Front Physiol 10, 770 (2019). This paper shows the validity of theWestern diet NTF mouse model of NASH.
The Jackson Laboratory. C57BL/6J mice https://www.jax.org/strain/000664 (2022).
Eng, J. M. & Estall, J. L. Diet-induced models of non-alcoholic fatty liver disease: food for thought on sugar, fat and cholesterol. Cells https://doi.org/10.3390/cells10071805 (2021).
Chen, H. et al. Consumption of sugar-sweetened beverages has a dose-dependent effect on the risk of non-alcoholic fatty liver disease: an updated systematic review and dose-response meta-analysis. Int. J. Environ. Res. Public Health https://doi.org/10.3390/ijerph16122192 (2019).
Zelber-Sagi, S. et al. Long term nutritional intake and the risk for non-alcoholic fatty liver disease (NAFLD): a population based study. J. Hepatol. 47, 711–717 (2007).
Article  Google Scholar 
Togo, J., Hu, S., Li, M., Niu, C. & Speakman, J. R. Impact of dietary sucrose on adiposity and glucose homeostasis in C57BL/6J mice depends on mode of ingestion: liquid or solid. Mol. Metab. 27, 22–32 (2019).
Article  Google Scholar 
Mastrocola, R. et al. Fructose liquid and solid formulations differently affect gut integrity, microbiota composition and related liver toxicity: a comparative in vivo study. J. Nutr. Biochem. 55, 185–199 (2018).
Article  Google Scholar 
Khitan, Z. & Kim, D. H. Fructose: a key factor in the development of metabolic syndrome and hypertension. J. Nutr. Metab. 2013, 682673 (2013).
Article  Google Scholar 
Febbraio, M. A. & Karin, M. ‘Sweet death’: fructose as a metabolic toxin that targets the gut–liver axis. Cell Metab. 33, 2316–2328 (2021).
Article  Google Scholar 
Jensen, T. et al. Fructose and sugar: a major mediator of non-alcoholic fatty liver disease. J. Hepatol. 68, 1063–1075 (2018).
Article  Google Scholar 
Todoric, J. et al. Fructose stimulated de novo lipogenesis is promoted by inflammation. Nat. Metab. 2, 1034–1045 (2020).
Article  Google Scholar 
Ioannou, G. N. et al. Cholesterol crystallization within hepatocyte lipid droplets and its role in murine NASH. J. Lipid Res. 58, 1067–1079 (2017).
Article  Google Scholar 
Van Rooyen, D. M. et al. Hepatic free cholesterol accumulates in obese, diabetic mice and causes non-alcoholic steatohepatitis. Gastroenterology 141, 1393–1403 (2011).
Article  Google Scholar 
Van Rooyen, D. M. et al. Pharmacological cholesterol lowering reverses fibrotic NASH in obese, diabetic mice with metabolic syndrome. J. Hepatol. 59, 144–152 (2013).
Article  Google Scholar 
Zhang, X. et al. Dietary cholesterol drives fatty liver-associated liver cancer by modulating gut microbiota and metabolites. Gut 70, 761–774 (2021). This study demonstrates that a low-cholesterol Western diet does not induce spontaneous NASH–HCC in C57BL/6 mice.
Article  Google Scholar 
Asgharpour, A. et al. A diet-induced animal model of non-alcoholic fatty liver disease and hepatocellular cancer. J. Hepatol. 65, 579–588 (2016). The authors developed the DIAMOND model of NASH–HCC in the mixed B6/129 strain.
Article  Google Scholar 
Wolf, M. J. et al. Metabolic activation of intrahepatic CD8+ T cells and NKT cells causes non-alcoholic steatohepatitis and liver cancer via cross-talk with hepatocytes. Cancer Cell 26, 549–564 (2014). The authors developed the CDHFD model of NASH and NASH–HCC in C57BL/6 mice.
Article  Google Scholar 
Grohmann, M. et al. Obesity drives STAT-1-dependent NASH and STAT-3-dependent HCC. Cell 175, 1289–1306 (2018).
Article  Google Scholar 
Kishida, N. et al. Development of a novel mouse model of hepatocellular carcinoma with non-alcoholic steatohepatitis using a high-fat, choline-deficient diet and intraperitoneal injection of diethylnitrosamine. BMC Gastroenterol. 16, 61 (2016).
Article  Google Scholar 
Malehmir, M. et al. Platelet GPIbα is a mediator and potential interventional target for NASH and subsequent liver cancer. Nat. Med. 25, 641–655 (2019).
Article  Google Scholar 
Smati, S. et al. Integrative study of diet-induced mouse models of NAFLD identifies PPARα as a sexually dimorphic drug target. Gut 71, 807–821 (2022).
Article  Google Scholar 
Yao, Z. M. & Vance, D. E. The active synthesis of phosphatidylcholine is required for very-low-density lipoprotein secretion from rat hepatocytes. J. Biol. Chem. 263, 2998–3004 (1988).
Article  Google Scholar 
Ashworth, C. T., Sanders, E. & Arnold, N. Hepatic lipids; fine structural changes in liver cells after high-fat, high-cholesterol, and choline-deficient diets in rats. Arch. Pathol. 72, 625–636 (1961).
Google Scholar 
Salaspuro, M. P. & Mäenpää, P. H. Influence of ethanol on the metabolism of perfused normal, fatty and cirrhotic rat livers. Biochem. J. 100, 768–778 (1966).
Article  Google Scholar 
Kalhan, S. C. et al. Plasma metabolomic profile in non-alcoholic fatty liver disease. Metabolism 60, 404–413 (2011).
Article  Google Scholar 
Ikawa-Yoshida, A. et al. Hepatocellular carcinoma in a mouse model fed a choline-deficient, l-amino acid-defined, high-fat diet. Int. J. Exp. Pathol. 98, 221–233 (2017). The authors developed the CDAHFD model of NASH–HCC in lean mice.
Article  Google Scholar 
Matsumoto, M. et al. An improved mouse model that rapidly develops fibrosis in non-alcoholic steatohepatitis. Int. J. Exp. Pathol. 94, 93–103 (2013). The authors developed the CDAHFD model of NASH in lean mice.
Lee, C. et al. Formyl peptide receptor 2 determines sex-specific differences in the progression of non-alcoholic fatty liver disease and steatohepatitis. Nat. Commun. 13, 578 (2022).
Article  Google Scholar 
Suzuki-Kemuriyama, N. et al. Nonobese mice with non-alcoholic steatohepatitis fed on a choline-deficient, l-amino acid-defined, high-fat diet exhibit alterations in signaling pathways. FEBS Open Bio 11, 2950–2965 (2021).
Article  Google Scholar 
Albhaisi, S., Chowdhury, A. & Sanyal, A. J. Non-alcoholic fatty liver disease in lean individuals. JHEP Rep. 1, 329–341 (2019).
Article  Google Scholar 
Tetri, L. H., Basaranoglu, M., Brunt, E. M., Yerian, L. M. & Neuschwander-Tetri, B. A. Severe NAFLD with hepatic necroinflammatory changes in mice fed trans fats and a high-fructose corn syrup equivalent. Am. J. Physiol. Gastrointest. Liver Physiol. 295, G987–G995 (2008). The authors developed the ALIOS model of NASH in mice.
Article  Google Scholar 
Harris, S. E. et al. The American lifestyle-induced obesity syndrome diet in male and female rodents recapitulates the clinical and transcriptomic features of no-nalcoholic fatty liver disease and non-alcoholic steatohepatitis. Am. J. Physiol. Gastrointest. Liver Physiol. 319, G345–G360 (2020).
Article  Google Scholar 
Itagaki, H., Shimizu, K., Morikawa, S., Ogawa, K. & Ezaki, T. Morphological and functional characterization of non-alcoholic fatty liver disease induced by a methionine-choline-deficient diet in C57BL/6 mice. Int. J. Clin. Exp. Pathol. 6, 2683–2696 (2013).
Google Scholar 
Gautheron, J. et al. A positive feedback loop between RIP3 and JNK controls non-alcoholic steatohepatitis. EMBO Mol. Med. 6, 1062–1074 (2014).
Article  Google Scholar 
Ma, C. et al. NAFLD causes selective CD4+ T lymphocyte loss and promotes hepatocarcinogenesis. Nature 531, 253–257 (2016).
Article  Google Scholar 
Jha, P. et al. Role of adipose tissue in methionine-choline-deficient model of non-alcoholic steatohepatitis (NASH). Biochim. Biophys. Acta 1842, 959–970 (2014).
Article  Google Scholar 
Rinella, M. E. & Green, R. M. The methionine-choline deficient dietary model of steatohepatitis does not exhibit insulin resistance. J. Hepatol. 40, 47–51 (2004).
Article  Google Scholar 
Leclercq, I. A., Farrell, G. C., Sempoux, C., dela Peña, A. & Horsmans, Y. Curcumin inhibits NF-κB activation and reduces the severity of experimental steatohepatitis in mice. J. Hepatol. 41, 926–934 (2004).
Article  Google Scholar 
Ramadori, P., Weiskirchen, R., Trebicka, J. & Streetz, K. Mouse models of metabolic liver injury. Lab. Anim. 49, 47–58 (2015).
Article  Google Scholar 
Fontaine, D. A. & Davis, D. B. Attention to background strain is essential for metabolic research: C57BL/6 and the International Knockout Mouse Consortium. Diabetes 65, 25–33 (2016).
Article  Google Scholar 
Champy, M. F. et al. Genetic background determines metabolic phenotypes in the mouse. Mamm. Genome 19, 318–331 (2008).
Article  Google Scholar 
Mekada, K. et al. Genetic differences among C57BL/6 substrains. Exp. Anim. 58, 141–149 (2009).
Article  Google Scholar 
Simon, M. M. et al. A comparative phenotypic and genomic analysis of C57BL/6J and C57BL/6N mouse strains. Genome Biol. 14, R82 (2013).
Article  Google Scholar 
Green, C. D. et al. A new preclinical model of western diet-induced progression of non-alcoholic steatohepatitis to hepatocellular carcinoma. FASEB J. https://doi.org/10.1096/fj.202200346R (2022). This paper shows that a Western diet with sugar water can induce NASH and NASH–HCC in C57BL/6N mice.
Arsov, T. et al. Adaptive failure to high-fat diet characterizes steatohepatitis in Alms1 mutant mice. Biochem. Biophys. Res. Commun. 342, 1152–1159 (2006).
Article  Google Scholar 
Arsov, T. et al. Fat aussie–a new Alström syndrome mouse showing a critical role for ALMS1 in obesity, diabetes, and spermatogenesis. Mol. Endocrinol. 20, 1610–1622 (2006).
Article  Google Scholar 
Larter, C. Z. et al. Roles of adipose restriction and metabolic factors in progression of steatosis to steatohepatitis in obese, diabetic mice. J. Gastroenterol. Hepatol. 24, 1658–1668 (2009).
Article  Google Scholar 
Ganguly, S. et al. non-alcoholic steatohepatitis and HCC in a hyperphagic mouse accelerated by Western diet. Cell Mol. Gastroenterol. Hepatol. 12, 891–920 (2021). Foz/Foz mice were fed a Western diet to induce NASH and subsequent NASH–HCC.
Article  Google Scholar 
Nakagawa, H. et al. ER stress cooperates with hypernutrition to trigger TNF-dependent spontaneous HCC development. Cancer Cell 26, 331–343 (2014). This study developed the MUP-uPA model of NASH and NASH–HCC.
Article  Google Scholar 
Shalapour, S. et al. Inflammation-induced IgA+ cells dismantle anti-liver cancer immunity. Nature 551, 340–345 (2017).
Article  Google Scholar 
Fujii, M. et al. A murine model for non-alcoholic steatohepatitis showing evidence of association between diabetes and hepatocellular carcinoma. Med. Mol. Morphol. 46, 141–152 (2013).
Article  Google Scholar 
Park, E. J. et al. Dietary and genetic obesity promote liver inflammation and tumorigenesis by enhancing IL-6 and TNF expression. Cell 140, 197–208 (2010). This paper shows that obesity promotes liver carcinogenesis.
Liu, F. et al. Long non-coding RNA SNHG6 couples cholesterol sensing with mTORC1 activation in hepatocellular carcinoma. Nat. Metab. 4, 1022–1040 (2022).
Article  Google Scholar 
Ge, C. et al. Hepatocyte phosphatase DUSP22 mitigates NASH–HCC progression by targeting FAK. Nat. Commun. 13, 5945 (2022).
Article  Google Scholar 
Tsuchida, T. et al. A simple diet- and chemical-induced murine NASH model with rapid progression of steatohepatitis, fibrosis and liver cancer. J. Hepatol. 69, 385–395 (2018). The authors developed an accelerated model of NASH and NASH–HCC with profound fibrosis by combining a Western diet with weekly CCl4 injections.
Article  Google Scholar 
Horie, Y. et al. Hepatocyte-specific Pten deficiency results in steatohepatitis and hepatocellular carcinomas. J. Clin. Invest. 113, 1774–1783 (2004). This paper shows that hepatocyte-specific deletion of Pten results in NASH and subsequent liver cancer.
Stiles, B. et al. Liver-specific deletion of negative regulator Pten results in fatty liver and insulin hypersensitivity. Proc. Natl Acad. Sci. USA 101, 2082–2087 (2004).
Article  Google Scholar 
Ortega-Molina, A. & Serrano, M. PTEN in cancer, metabolism, and aging. Trends Endocrinol. Metab. 24, 184–189 (2013).
Article  Google Scholar 
Panasyuk, G. et al. PPARγ contributes to PKM2 and HK2 expression in fatty liver. Nat. Commun. 3, 672 (2012).
Article  Google Scholar 
Peyrou, M. et al. Hepatic PTEN deficiency improves muscle insulin sensitivity and decreases adiposity in mice. J. Hepatol. 62, 421–429 (2015).
Article  Google Scholar 
Langford, C. R. et al. Improved pathology reporting in NAFLD/NASH for clinical trials. J. Clin. Pathol. 75, 73–75 (2022).
Article  Google Scholar 
Kleiner, D. E. et al. Design and validation of a histological scoring system for non-alcoholic fatty liver disease. Hepatology 41, 1313–1321 (2005). The authors designed the NAS to assess the histopathology of liver tissue.
Article  Google Scholar 
Brunt, E. M. et al. NAFLD: reporting histologic findings in clinical practice. Hepatology 73, 2028–2038 (2021).
Article  Google Scholar 
Ahn, J. C., Connell, A., Simonetto, D. A., Hughes, C. & Shah, V. H. Application of artificial intelligence for the diagnosis and treatment of liver diseases. Hepatology 73, 2546–2563 (2021).
Article  Google Scholar 
Soon, G. S. T. et al. Artificial intelligence improves pathologist agreement for fibrosis scores in non-alcoholic steatohepatitis patients. Clin. Gastroenterol. Hepatol. https://doi.org/10.1016/j.cgh.2022.05.027 (2022).
Nam, D., Chapiro, J., Paradis, V., Seraphin, T. P. & Kather, J. N. Artificial intelligence in liver diseases: improving diagnostics, prognostics and response prediction. JHEP Rep. 4, 100443 (2022).
Article  Google Scholar 
Liu, F. et al. qFIBS: an automated technique for quantitative evaluation of fibrosis, inflammation, ballooning, and steatosis in patients with non-alcoholic steatohepatitis. Hepatology 71, 1953–1966 (2020).
Article  Google Scholar 
Taylor-Weiner, A. et al. A machine learning approach enables quantitative measurement of liver histology and disease monitoring in NASH. Hepatology 74, 133–147 (2021).
Article  Google Scholar 
Angulo, P. et al. Liver fibrosis, but no other histologic features, is associated with long-term outcomes of patients with non-alcoholic fatty liver disease. Gastroenterology 149, 389–397 (2015). This study demonstrates the importance of liver fibrosis for outcomes in NASH patients.
Article  Google Scholar 
Dulai, P. S. et al. Increased risk of mortality by fibrosis stage in non-alcoholic fatty liver disease: systematic review and meta-analysis. Hepatology 65, 1557–1565 (2017).
Article  Google Scholar 
Kingma, B. R., Frijns, A. J., Schellen, L. & van Marken Lichtenbelt, W. D. Beyond the classic thermoneutral zone: Including thermal comfort. Temp. 1, 142–149 (2014).
Google Scholar 
Škop, V. et al. Mouse thermoregulation: introducing the concept of the thermoneutral point. Cell Rep. 31, 107501 (2020).
Article  Google Scholar 
Giles, D. A. et al. Thermoneutral housing exacerbates non-alcoholic fatty liver disease in mice and allows for sex-independent disease modeling. Nat. Med. 23, 829–838 (2017). This paper shows that thermoneutral housing can exacerbate NASH pathogenesis.
Article  Google Scholar 
Bray, F. et al. Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J. Clin. 68, 394–424 (2018).
Article  Google Scholar 
Burra, P. et al. Clinical impact of sexual dimorphism in non-alcoholic fatty liver disease (NAFLD) and non-alcoholic steatohepatitis (NASH). Liver Int 41, 1713–1733 (2021).
Article  Google Scholar 
Lonardo, A. et al. Sex differences in non-alcoholic fatty liver disease: state of the art and identification of research gaps. Hepatology 70, 1457–1469 (2019).
Article  Google Scholar 
Vandel, J. et al. Hepatic molecular signatures highlight the sexual dimorphism of non-alcoholic steatohepatitis (NASH). Hepatology 73, 920–936 (2021).
Article  Google Scholar 
Iyer, J. K., Kalra, M., Kaul, A., Payton, M. E. & Kaul, R. Estrogen receptor expression in chronic hepatitis C and hepatocellular carcinoma pathogenesis. World J. Gastroenterol. 23, 6802–6816 (2017).
Article  Google Scholar 
Louet, J. F., LeMay, C. & Mauvais-Jarvis, F. Antidiabetic actions of estrogen: insight from human and genetic mouse models. Curr. Atheroscler. Rep. 6, 180–185 (2004).
Article  Google Scholar 
Espeland, M. A. et al. Effect of postmenopausal hormone therapy on glucose and insulin concentrations. PEPI Investigators. Postmenopausal Estrogen/Progestin Interventions. Diabetes Care 21, 1589–1595 (1998).
Article  Google Scholar 
Hassan, M. M. et al. Estrogen replacement reduces risk and increases survival times of women with hepatocellular carcinoma. Clin. Gastroenterol. Hepatol. 15, 1791–1799 (2017).
Article  Google Scholar 
McGlynn, K. A. et al. Menopausal hormone therapy use and risk of primary liver cancer in the clinical practice research datalink. Int J. Cancer 138, 2146–2153 (2016).
Article  Google Scholar 
McGlynn, K. A. et al. Reproductive factors, exogenous hormone use and risk of hepatocellular carcinoma among US women: results from the Liver Cancer Pooling Project. Br. J. Cancer 112, 1266–1272 (2015).
Article  Google Scholar 
McKenzie, J. et al. Effects of HRT on liver enzyme levels in women with type 2 diabetes: a randomized placebo-controlled trial. Clin. Endocrinol. 65, 40–44 (2006).
Article  Google Scholar 
Nakatani, T., Roy, G., Fujimoto, N., Asahara, T. & Ito, A. Sex hormone dependency of diethylnitrosamine-induced liver tumors in mice and chemoprevention by leuprorelin. Jpn. J. Cancer Res. 92, 249–256 (2001).
Article  Google Scholar 
Le Roy, T. et al. Intestinal microbiota determines development of non-alcoholic fatty liver disease in mice. Gut 62, 1787–1794 (2013).
Article  Google Scholar 
Moschen, A. R., Kaser, S. & Tilg, H. Non-alcoholic steatohepatitis: a microbiota-driven disease. Trends Endocrinol. Metab. 24, 537–545 (2013).
Article  Google Scholar 
Yoshimoto, S. et al. Obesity-induced gut microbial metabolite promotes liver cancer through senescence secretome. Nature 499, 97–101 (2013). This study highlights that the secondary bile acid deoxycholic acid from gut microbiota can driveobesity-dependent liver carcinogenesis.
Article  Google Scholar 
Yang, J. & Chun, J. Taxonomic composition and variation in the gut microbiota of laboratory mice. Mamm. Genome 32, 297–310 (2021).
Article  Google Scholar 
Weiskirchen, S., Weiper, K., Tolba, R. H. & Weiskirchen, R. All you can feed: some comments on production of mouse diets used in biomedical research with special emphasis on non-alcoholic fatty liver disease research. Nutrients https://doi.org/10.3390/nu12010163 (2020).
Ericsson, A. C. & Franklin, C. L. The gut microbiome of laboratory mice: considerations and best practices for translational research. Mamm. Genome 32, 239–250 (2021).
Article  Google Scholar 
Chiang, J. Y. Bile acid metabolism and signaling. Compr. Physiol. 3, 1191–1212 (2013).
Article  Google Scholar 
Haeusler, R. A., Astiarraga, B., Camastra, S., Accili, D. & Ferrannini, E. Human insulin resistance is associated with increased plasma levels of 12α-hydroxylated bile acids. Diabetes 62, 4184–4191 (2013).
Article  Google Scholar 
Prinz, P. et al. Plasma bile acids show a positive correlation with body mass index and are negatively associated with cognitive restraint of eating in obese patients. Front Neurosci. 9, 199 (2015).
Article  Google Scholar 
Sun, W. et al. Insulin resistance is associated with total bile acid level in type 2 diabetic and nondiabetic population: a cross-sectional study. Med. 95, e2778 (2016).
Article  Google Scholar 
Sydor, S. et al. Altered microbiota diversity and bile acid signaling in cirrhotic and noncirrhotic NASH–HCC. Clin. Transl. Gastroenterol. 11, e00131 (2020).
Article  Google Scholar 
Chávez-Talavera, O., Tailleux, A., Lefebvre, P. & Staels, B. Bile acid control of metabolism and inflammation in obesity, type 2 diabetes, dyslipidemia, and non-alcoholic fatty liver disease. Gastroenterology 152, 1679–1694 (2017).
Article  Google Scholar 
Cudalbu, C. et al. The C57BL/6J mouse exhibits sporadic congenital portosystemic shunts. PLoS ONE 8, e69782 (2013). This paper shows that a subset of C57BL/6 mice display portosystemic shunts that may predispose to liver pathologies.
d’Anjou, M. A., Penninck, D., Cornejo, L. & Pibarot, P. Ultrasonographic diagnosis of portosystemic shunting in dogs and cats. Vet. Radio. Ultrasound 45, 424–437 (2004).
Article  Google Scholar 
Lemoine, C. et al. Liver histopathology in patients with hepatic masses and the Abernethy malformation. J. Pediatr. Surg. 54, 266–271 (2019).
Article  Google Scholar 
Ronda, O. et al. Spontaneous liver disease in wild-type C57BL/6JOlaHsd mice fed semisynthetic diet. PLoS ONE 15, e0232069 (2020). This paper demonstrates that wild-type C57BL/6OlaHsd mice also show spontaneous cholaemia and can predispose to liver pathology.
Article  Google Scholar 
Sharma, R. et al. Congenital extrahepatic portosystemic shunt complicated by the development of hepatocellular carcinoma. Hepatobiliary Pancreat. Dis. Int. 14, 552–557 (2015).
Article  Google Scholar 
Gastaldelli, A. & Cusi, K. From NASH to diabetes and from diabetes to NASH: Mechanisms and treatment options. JHEP Rep. 1, 312–328 (2019).
Article  Google Scholar 
Korenblat, K. M., Fabbrini, E., Mohammed, B. S. & Klein, S. Liver, muscle, and adipose tissue insulin action is directly related to intrahepatic triglyceride content in obese subjects. Gastroenterology 134, 1369–1375 (2008).
Article  Google Scholar 
Xu, X. et al. Targeted therapeutics and novel signaling pathways in non-alcohol-associated fatty liver/steatohepatitis (NAFL/NASH). Signal Transduct. Target Ther. 7, 287 (2022).
DeFronzo, R. A., Tobin, J. D. & Andres, R. Glucose clamp technique: a method for quantifying insulin secretion and resistance. Am. J. Physiol. 237, E214–E223 (1979).
Google Scholar 
Ayala, J. E., Lantier, L., McGuinness, O. P. & Wasserman, D. H. Peeling back the layers of the glucose clamp. Nat. Metab. 4, 496–498 (2022).
Article  Google Scholar 
Virtue, S. & Vidal-Puig, A. GTTs and ITTs in mice: simple tests, complex answers. Nat. Metab. 3, 883–886 (2021). This paper describes the intracies of metabolic phenotyping of mice with glucose and insulin tolerance tests.
Article  Google Scholar 
Matthews, D. R. et al. Homeostasis model assessment: insulin resistance and beta-cell function from fasting plasma glucose and insulin concentrations in man. Diabetologia 28, 412–419 (1985).
Article  Google Scholar 
Wallace, T. M., Levy, J. C. & Matthews, D. R. Use and abuse of HOMA modeling. Diabetes Care 27, 1487–1495 (2004).
Article  Google Scholar 
Download references
M. Heikenwalder was supported by a European Research Council (ERC) Consolidator grant (HepatoMetaboPath), the ERC POC (Faith), the Helmholtz Future topic Inflammation and Immunology, Zukunftssthema ‘Immunology and Inflammation’ (ZT-0027), SFB/TR 209 project ID 314905040, SFB 1479 (Project ID: 441891347), Project ID 272983813-SFBTR 179, the Rainer Hoenig Stiftung, Research Foundation Flanders (FWO)under grant 30826052 (EOS Convention MODEL-IDI) and a seed funding from HI-TRON. Funded by the German Research Foundation (DFG) as part of the Excellence Strategy of the federal and state governments — EXC 2180 — 390900677.
Division of Chronic Inflammation and Cancer, German Cancer Research Center (DKFZ), Heidelberg, Germany
Suchira Gallage, Jose Efren Barragan Avila, Pierluigi Ramadori, Enrico Focaccia, Mohammad Rahbari, Adnan Ali & Mathias Heikenwalder
The M3 Research Institute, Eberhard Karls University Tübingen, Tuebingen, Germany
Suchira Gallage, Nisar P. Malek & Mathias Heikenwalder
Department Internal Medicine I, Eberhard-Karls University, Tuebingen, Germany
Nisar P. Malek
Cluster of Excellence iFIT (EXC 2180) “Image-Guided and Functionally Instructed Tumor Therapies”, University of Tuebingen, Tuebingen, Germany
Nisar P. Malek & Mathias Heikenwalder
Newcastle NIHR Biomedical Research Centre, Newcastle upon Tyne Hospitals, NHS Foundation Trust, Newcastle upon Tyne, UK
Quentin M. Anstee
Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, UK
Quentin M. Anstee
You can also search for this author in PubMed Google Scholar
You can also search for this author in PubMed Google Scholar
You can also search for this author in PubMed Google Scholar
You can also search for this author in PubMed Google Scholar
You can also search for this author in PubMed Google Scholar
You can also search for this author in PubMed Google Scholar
You can also search for this author in PubMed Google Scholar
You can also search for this author in PubMed Google Scholar
You can also search for this author in PubMed Google Scholar
Correspondence to Suchira Gallage or Mathias Heikenwalder.
Q.M.A. is coordinator of the LITMUS (Liver Investigation: Testing Marker Utility in Steatohepatitis) consortium funded by the Innovative Medicines Initiative (IMI2) Program of the European Union under grant agreement 777377. This multi-stakeholder consortium includes industry partners and received funding from EFPIA. Q.M.A. received funding from AstraZeneca, Boehringer Ingelheim, Intercept and royalties from Elsevier. Q.M.A. received consulting fees on behalf of Newcastle University from Alimentiv, Akero, AstraZeneca, Axcella, 89Bio, Boehringer Ingelheim, Bristol Myers Squibb, Galmed, Genfit, Genentech, Gilead, GlaxoSmithKline, Hanmi, HistoIndex, Intercept, Inventiva, Ionis, IQVIA, Janssen, Madrigal, Medpace, Merck, NGMBio, Novartis, Novo Nordisk, PathAI, Pfizer, Poxel, Resolution Therapeutics, Roche, Ridgeline Therapeutics, RTI, Shionogi and Terns. Q.M.A. received fees for lectures from Fishawack, Integritas Communications, Kenes, Novo Nordisk, Madrigal, Medscape and Springer Healthcare. The other authors declare no competing interests.
Nature Metabolism thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editor: Isabella Samuelson, in collaboration with the Nature Metabolism team.
Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.
Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.
Reprints and Permissions
Gallage, S., Avila, J.E.B., Ramadori, P. et al. A researcher’s guide to preclinical mouse NASH models. Nat Metab (2022). https://doi.org/10.1038/s42255-022-00700-y
Download citation
Received: 10 May 2022
Accepted: 26 October 2022
Published: 20 December 2022
DOI: https://doi.org/10.1038/s42255-022-00700-y
Anyone you share the following link with will be able to read this content:
Sorry, a shareable link is not currently available for this article.

Provided by the Springer Nature SharedIt content-sharing initiative

Advertisement
Nature Metabolism (Nat Metab) ISSN 2522-5812 (online)
© 2022 Springer Nature Limited
Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

source

By admin

Leave a Reply

Your email address will not be published. Required fields are marked *