Skip to main content

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.

  • Review Article
  • Published:

Metabolomics and lipidomics in NAFLD: biomarkers and non-invasive diagnostic tests

Abstract

Nonalcoholic fatty liver disease (NAFLD) is one of the most common liver diseases worldwide and is often associated with aspects of metabolic syndrome. Despite its prevalence and the importance of early diagnosis, there is a lack of robustly validated biomarkers for diagnosis, prognosis and monitoring of disease progression in response to a given treatment. In this Review, we provide an overview of the contribution of metabolomics and lipidomics in clinical studies to identify biomarkers associated with NAFLD and nonalcoholic steatohepatitis (NASH). In addition, we highlight the key metabolic pathways in NAFLD and NASH that have been identified by metabolomics and lipidomics approaches and could potentially be used as biomarkers for non-invasive diagnostic tests. Overall, the studies demonstrated alterations in amino acid metabolism and several aspects of lipid metabolism including circulating fatty acids, triglycerides, phospholipids and bile acids. Although we report several studies that identified potential biomarkers, few have been validated.

Key points

  • Nonalcoholic fatty liver disease (NAFLD) affects 25% of the adult world population; in about 20% of patients, it can progress to nonalcoholic steatohepatitis (NASH), which can lead to cirrhosis.

  • There is an urgent need for development of clinically relevant biomarkers and non-invasive diagnostic tests for NAFLD.

  • Metabolomics and lipidomics approaches have provided insightful evidence of altered metabolic pathways in NAFLD and NASH.

  • There is an association between circulating amino acids and steatohepatitis, and impairment in amino acid metabolism in NAFLD is strongly correlated with insulin resistance, particularly in the muscle.

  • An increase in oxidative stress results in a reduction in hepatic glutathione levels and is associated with liver damage and the progression of NAFLD to NASH.

  • NASH is strongly associated with alterations in circulating fatty acids and intact lipids, which is partially due to alterations in de novo liver lipogenesis, lipolysis rate and VLDL metabolism.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Overview of analytical approaches and workflows as commonly applied in metabolomic analysis, including lipidomics.
Fig. 2: Overview of the key metabolic pathways in fasting and postprandial states involved in the pathogenesis of NAFLD.
Fig. 3: Overview of the key bile acid pathways involved in NAFLD.
Fig. 4: Fluxomics in the insulin-resistant state in NAFLD.

Similar content being viewed by others

References

  1. Younossi, Z. M. et al. Global epidemiology of nonalcoholic fatty liver disease-meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology 64, 73–84 (2016).

    Article  PubMed  Google Scholar 

  2. Younossi, Z. et al. Global burden of NAFLD and NASH: trends, predictions, risk factors and prevention. Nat. Rev. Gastroenterol. Hepatol. 15, 11–20 (2018).

    Article  PubMed  Google Scholar 

  3. FDA-NIH Biomarker Working Group. BEST (Biomarkers, EndpointS, and other Tools) Resource [Internet] (FDA, 2016).

  4. Silverman, J. F. et al. Liver pathology in morbidly obese patients with and without diabetes. Am. J. Gastroenterol. 85, 1349–1355 (1990).

    CAS  PubMed  Google Scholar 

  5. Loguercio, C. et al. Non-alcoholic fatty liver disease in an area of southern Italy: main clinical, histological, and pathophysiological aspects. J. Hepatol. 35, 568–574 (2001).

    Article  CAS  PubMed  Google Scholar 

  6. Neuschwander-Tetri, B. A. & Bacon, B. R. Nonalcoholic steatohepatitis. Med. Clin. North Am. 80, 1147–1166 (1996).

    Article  CAS  PubMed  Google Scholar 

  7. Muriel, P. Role of free radicals in liver diseases. Hepatol. Int. 3, 526–536 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Rinella, M. E., Tacke, F., Sanyal, A. J., Anstee, Q. M. & Participants of the AASLD/EASL Workshop. Report on the AASLD/EASL joint workshop on clinical trial endpoints in NAFLD. J. Hepatol. 71, 823–833 (2019).

    Article  PubMed  Google Scholar 

  9. Ratziu, V. A critical review of endpoints for non-cirrhotic NASH therapeutic trials. J. Hepatol. 68, 353–361 (2018).

    Article  PubMed  Google Scholar 

  10. European Association for the Study of the Liver (EASL), European Association for the Study of Diabetes (EASD) & European Association for the Study of Obesity (EASO). EASL-EASD-EASO Clinical Practice Guidelines for the management of non-alcoholic fatty liver disease. J. Hepatol. 64, 1388–1402 (2016).

    Article  Google Scholar 

  11. Hoofnagle, J. H. et al. Vitamin E and changes in serum alanine aminotransferase levels in patients with non-alcoholic steatohepatitis. Aliment. Pharmacol. Ther. 38, 134–143 (2013).

    Article  CAS  PubMed  Google Scholar 

  12. Vilar-Gomez, E. et al. Development and validation of a noninvasive prediction model for nonalcoholic steatohepatitis resolution after lifestyle intervention. Hepatology 63, 1875–1887 (2016).

    Article  CAS  PubMed  Google Scholar 

  13. Shah, A. G. et al. Comparison of noninvasive markers of fibrosis in patients with nonalcoholic fatty liver disease. Clin. Gastroenterol. Hepatol. 7, 1104–1112 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Quehenberger, O. et al. Lipidomics reveals a remarkable diversity of lipids in human plasma. J. Lipid Res. 51, 3299–3305 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Hyotylainen, T. & Oresic, M. Systems biology strategies to study lipidomes in health and disease. Prog. Lipid Res. 55, 43–60 (2014).

    Article  CAS  PubMed  Google Scholar 

  16. Cajka, T. & Fiehn, O. Toward merging untargeted and targeted methods in mass spectrometry-based metabolomics and lipidomics. Anal. Chem. 88, 524–545 (2016).

    Article  CAS  PubMed  Google Scholar 

  17. Mardinoglu, A. et al. Genome-scale metabolic modelling of hepatocytes reveals serine deficiency in patients with non-alcoholic fatty liver disease. Nat. Commun. 5, 3083 (2014).

    Article  PubMed  Google Scholar 

  18. Mardinoglu, A. et al. Personal model-assisted identification of NAD(+) and glutathione metabolism as intervention target in NAFLD. Mol. Syst. Biol. 13, 916 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Hyotylainen, T. et al. Genome-scale study reveals reduced metabolic adaptability in patients with non-alcoholic fatty liver disease. Nat. Commun. 7, 8994 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Nielsen, J. Systems biology of metabolism: a driver for developing personalized and precision medicine. Cell Metab. 25, 572–579 (2017).

    Article  CAS  PubMed  Google Scholar 

  21. Kalhan, S. C. et al. Plasma metabolomic profile in nonalcoholic fatty liver disease. Metabolism 60, 404–413 (2011).

    Article  CAS  PubMed  Google Scholar 

  22. Lake, A. D. et al. Branched chain amino acid metabolism profiles in progressive human nonalcoholic fatty liver disease. Amino Acids 47, 603–615 (2015).

    Article  CAS  PubMed  Google Scholar 

  23. Gaggini, M. et al. Altered amino acid concentrations in NAFLD: impact of obesity and insulin resistance. Hepatology 67, 145–158 (2018).

    Article  CAS  PubMed  Google Scholar 

  24. Kakazu, E. et al. Branched chain amino acids are associated with the heterogeneity of the area of lipid droplets in hepatocytes of patients with non-alcoholic fatty liver disease. Hepatol. Res. 49, 860–871 (2019).

    Article  CAS  PubMed  Google Scholar 

  25. Barr, J. et al. Obesity-dependent metabolic signatures associated with nonalcoholic fatty liver disease progression. J. Proteome Res. 11, 2521–2532 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Biolo, G., Gastaldelli, A., Zhang, X. J. & Wolfe, R. R. Protein synthesis and breakdown in skin and muscle: a leg model of amino acid kinetics. Am. J. Physiol. 267, E467–E474 (1994).

    CAS  PubMed  Google Scholar 

  27. Lynch, C. J. & Adams, S. H. Branched-chain amino acids in metabolic signalling and insulin resistance. Nat. Rev. Endocrinol. 10, 723–736 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Fiehn, O. et al. Plasma metabolomic profiles reflective of glucose homeostasis in non-diabetic and type 2 diabetic obese African-American women. PLoS ONE 5, e15234 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Newgard, C. B. et al. A branched-chain amino acid-related metabolic signature that differentiates obese and lean humans and contributes to insulin resistance. Cell Metab. 9, 311–326 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Lee, Y. H. et al. Sarcopenia is associated with significant liver fibrosis independently of obesity and insulin resistance in nonalcoholic fatty liver disease: nationwide surveys (KNHANES 2008–2011). Hepatology 63, 776–786 (2016).

    Article  CAS  PubMed  Google Scholar 

  31. Adeva, M. M., Calvino, J., Souto, G. & Donapetry, C. Insulin resistance and the metabolism of branched-chain amino acids in humans. Amino Acids 43, 171–181 (2012).

    Article  CAS  PubMed  Google Scholar 

  32. Wang, T. J. et al. Metabolite profiles and the risk of developing diabetes. Nat. Med. 17, 448–453 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Luzi, L., Castellino, P. & DeFronzo, R. A. Insulin and hyperaminoacidemia regulate by a different mechanism leucine turnover and oxidation in obesity. Am. J. Physiol. 270, E273–E281 (1996).

    CAS  PubMed  Google Scholar 

  34. Qi, S. et al. Metabonomics screening of serum identifies pyroglutamate as a diagnostic biomarker for nonalcoholic steatohepatitis. Clin. Chim. Acta 473, 89–95 (2017).

    Article  CAS  PubMed  Google Scholar 

  35. Dong, S. et al. Urinary metabolomics analysis identifies key biomarkers of different stages of nonalcoholic fatty liver disease. World J. Gastroenterol. 23, 2771–2784 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Kitajima, Y. et al. Supplementation with branched-chain amino acids ameliorates hypoalbuminemia, prevents sarcopenia, and reduces fat accumulation in the skeletal muscles of patients with liver cirrhosis. J. Gastroenterol. 53, 427–437 (2018).

    Article  CAS  PubMed  Google Scholar 

  37. Kantartzis, K. et al. High cardiorespiratory fitness is an independent predictor of the reduction in liver fat during a lifestyle intervention in non-alcoholic fatty liver disease. Gut 58, 1281–1288 (2009).

    Article  CAS  PubMed  Google Scholar 

  38. Lehmann, R. et al. Circulating lysophosphatidylcholines are markers of a metabolically benign nonalcoholic fatty liver. Diabetes Care 36, 2331–2338 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Morgan, M. Y., Marshall, A. W., Milsom, J. P. & Sherlock, S. Plasma amino-acid patterns in liver disease. Gut 23, 362–370 (1982).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Morgan, M. Y., Milsom, J. P. & Sherlock, S. Plasma ratio of valine, leucine and isoleucine to phenylalanine and tyrosine in liver disease. Gut 19, 1068–1073 (1978).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Fischer, J. E. et al. The effect of normalization of plasma amino acids on hepatic encephalopathy in man. Surgery 80, 77–91 (1976).

    CAS  PubMed  Google Scholar 

  42. Michitaka, K. et al. Amino acid imbalance in patients with chronic liver diseases. Hepatol. Res. 40, 393–398 (2010).

    Article  CAS  PubMed  Google Scholar 

  43. Ishikawa, T. et al. Branched-chain amino acids to tyrosine ratio (BTR) predicts intrahepatic distant recurrence and survival for early hepatocellular carcinoma. Hepatogastroenterology 60, 2055–2059 (2013).

    CAS  PubMed  Google Scholar 

  44. Matthews, D. E. An overview of phenylalanine and tyrosine kinetics in humans. J. Nutr. 137, 1549S–1555S; discussion 1573S–1575S (2007).

    Article  CAS  PubMed  Google Scholar 

  45. Haufe, S. et al. Branched-chain and aromatic amino acids, insulin resistance and liver specific ectopic fat storage in overweight to obese subjects. Nutr. Metab. Cardiovasc. Dis. 26, 637–642 (2016).

    Article  CAS  PubMed  Google Scholar 

  46. Kawanaka, M. et al. Tyrosine levels are associated with insulin resistance in patients with nonalcoholic fatty liver disease. Hepat. Med. 7, 29–35 (2015).

    PubMed  PubMed Central  Google Scholar 

  47. Kahl, S. et al. Amino acid and fatty acid levels affect hepatic phosphorus metabolite content in metabolically healthy humans. J. Clin. Endocrinol. Metab. 103, 460–468 (2018).

    Article  PubMed  Google Scholar 

  48. Gastaldelli, A. et al. Relationship between hepatic/visceral fat and hepatic insulin resistance in nondiabetic and type 2 diabetic subjects. Gastroenterology 133, 496–506 (2007).

    Article  CAS  PubMed  Google Scholar 

  49. McCullough, A., Previs, S. & Kasumov, T. Stable isotope-based flux studies in nonalcoholic fatty liver disease. Pharmacol. Ther. 181, 22–33 (2018).

    Article  CAS  PubMed  Google Scholar 

  50. Zhu, L. et al. Upregulation of non-canonical Wnt ligands and oxidative glucose metabolism in NASH induced by methionine-choline deficient diet. Trends Cell Mol. Biol. 13, 47–56 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Zhou, Y. et al. Noninvasive detection of nonalcoholic steatohepatitis using clinical markers and circulating levels of lipids and metabolites. Clin. Gastroenterol. Hepatol. 14, 1463–1472.e6 (2016).

    Article  CAS  PubMed  Google Scholar 

  52. Dasarathy, S. et al. Glycine and urea kinetics in nonalcoholic steatohepatitis in human: effect of intralipid infusion. Am. J. Physiol. Gastrointest. Liver Physiol 297, G567–G575 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Kalhan, S. C. et al. Methionine and protein metabolism in non-alcoholic steatohepatitis: evidence for lower rate of transmethylation of methionine. Clin. Sci. 121, 179–189 (2011).

    Article  CAS  Google Scholar 

  54. Mato, J. M., Alonso, C., Noureddin, M. & Lu, S. C. Biomarkers and subtypes of deranged lipid metabolism in non-alcoholic fatty liver disease. World J. Gastroenterol. 25, 3009–3020 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Obeid, R. & Herrmann, W. Homocysteine and lipids: S-adenosyl methionine as a key intermediate. FEBS Lett. 583, 1215–1225 (2009).

    Article  CAS  PubMed  Google Scholar 

  56. Pastore, A. et al. Plasma levels of homocysteine and cysteine increased in pediatric NAFLD and strongly correlated with severity of liver damage. Int. J. Mol. Sci. 15, 21202–21214 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Lu, S. C. & Mato, J. M. S-adenosylmethionine in liver health, injury, and cancer. Physiol. Rev. 92, 1515–1542 (2012).

    Article  CAS  PubMed  Google Scholar 

  58. Alonso, C. et al. Metabolomic identification of subtypes of nonalcoholic steatohepatitis. Gastroenterology 152, 1449–1461.e7 (2017).

    Article  PubMed  Google Scholar 

  59. Sookoian, S. et al. Nonalcoholic steatohepatitis is associated with a state of betaine-insufficiency. Liver Int. 37, 611–619 (2017).

    Article  CAS  PubMed  Google Scholar 

  60. Zhang, W. et al. Betaine protects against high-fat-diet-induced liver injury by inhibition of high-mobility group Box 1 and Toll-like receptor 4 expression in rats. Dig. Dis. Sci. 58, 3198–3206 (2013).

    Article  CAS  PubMed  Google Scholar 

  61. Du, K. et al. Increased glutaminolysis marks active scarring in nonalcoholic steatohepatitis progression. Cell Mol. Gastroenterol. Hepatol. 10, 1–21 (2020).

    Article  PubMed  Google Scholar 

  62. Soga, T. et al. Serum metabolomics reveals γ-glutamyl dipeptides as biomarkers for discrimination among different forms of liver disease. J. Hepatol. 55, 896–905 (2011).

    Article  CAS  PubMed  Google Scholar 

  63. Koch, M. et al. Serum metabolomic profiling highlights pathways associated with liver fat content in a general population sample. Eur. J. Clin. Nutr. 71, 995–1001 (2017).

    Article  CAS  PubMed  Google Scholar 

  64. Cheng, J., Joyce, A., Yates, K., Aouizerat, B. & Sanyal, A. J. Metabolomic profiling to identify predictors of response to vitamin E for non-alcoholic steatohepatitis (NASH). PLoS ONE 7, e44106 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Leonetti, S., Herzog, R. I., Caprio, S., Santoro, N. & Tricò, D. Glutamate–serine–glycine index: a novel potential biomarker in pediatric non-alcoholic fatty liver disease. Children 7, 270 (2020).

    Article  PubMed Central  Google Scholar 

  66. Fabbrini, E. et al. Alterations in adipose tissue and hepatic lipid kinetics in obese men and women with nonalcoholic fatty liver disease. Gastroenterology 134, 424–431 (2008).

    Article  CAS  PubMed  Google Scholar 

  67. Cohen, J. C., Horton, J. D. & Hobbs, H. H. Human fatty liver disease: old questions and new insights. Science 332, 1519–1523 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. Westerbacka, J. et al. Splanchnic balance of free fatty acids, endocannabinoids, and lipids in subjects with nonalcoholic fatty liver disease. Gastroenterology 139, 1961–1971.e1 (2010).

    Article  CAS  PubMed  Google Scholar 

  69. Donnelly, K. L. et al. Sources of fatty acids stored in liver and secreted via lipoproteins in patients with nonalcoholic fatty liver disease. J. Clin. Invest. 115, 1343–1351 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Bugianesi, E. et al. Insulin resistance in non-diabetic patients with non-alcoholic fatty liver disease: sites and mechanisms. Diabetologia 48, 634–642 (2005).

    Article  CAS  PubMed  Google Scholar 

  71. Lomonaco, R. et al. Effect of adipose tissue insulin resistance on metabolic parameters and liver histology in obese patients with nonalcoholic fatty liver disease. Hepatology 55, 1389–1397 (2012).

    Article  CAS  PubMed  Google Scholar 

  72. Rosso, C. et al. Crosstalk between adipose tissue insulin resistance and liver macrophages in non-alcoholic fatty liver disease. J. Hepatol. 71, 1012–1021 (2019).

    Article  CAS  PubMed  Google Scholar 

  73. Bell, L. N. et al. Relationship between adipose tissue insulin resistance and liver histology in nonalcoholic steatohepatitis: a pioglitazone versus vitamin E versus placebo for the treatment of nondiabetic patients with nonalcoholic steatohepatitis trial follow-up study. Hepatology 56, 1311–1318 (2012).

    Article  CAS  PubMed  Google Scholar 

  74. Gastaldelli, A. et al. Importance of changes in adipose tissue insulin resistance to histological response during thiazolidinedione treatment of patients with nonalcoholic steatohepatitis. Hepatology 50, 1087–1093 (2009).

    Article  CAS  PubMed  Google Scholar 

  75. Emken, E. A. Metabolism of dietary stearic acid relative to other fatty acids in human subjects. Am. J. Clin. Nutr. 60, 1023S–1028S (1994).

    Article  CAS  PubMed  Google Scholar 

  76. Christinat, N. & Masoodi, M. Comprehensive lipoprotein characterization using lipidomics analysis of human plasma. J. Proteome Res. 16, 2947–2953 (2017).

    Article  CAS  PubMed  Google Scholar 

  77. Kotronen, A. et al. Serum saturated fatty acids containing triacylglycerols are better markers of insulin resistance than total serum triacylglycerol concentrations. Diabetologia 52, 684–690 (2009).

    Article  CAS  PubMed  Google Scholar 

  78. Puri, P. et al. A lipidomic analysis of nonalcoholic fatty liver disease. Hepatology 46, 1081–1090 (2007).

    Article  CAS  PubMed  Google Scholar 

  79. Scorletti, E. & Byrne, C. D. Omega-3 fatty acids, hepatic lipid metabolism, and nonalcoholic fatty liver disease. Annu. Rev. Nutr. 33, 231–248 (2013).

    Article  CAS  PubMed  Google Scholar 

  80. Jeyapal, S. et al. Substitution of linoleic acid with α-linolenic acid or long chain n-3 polyunsaturated fatty acid prevents Western diet induced nonalcoholic steatohepatitis. Sci. Rep. 8, 10953 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  81. Valenzuela, R. et al. N-3 long-chain polyunsaturated fatty acid supplementation significantly reduces liver oxidative stress in high fat induced steatosis. PLoS ONE 7, e46400 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Schuster, S. et al. Oxidized linoleic acid metabolites induce liver mitochondrial dysfunction, apoptosis, and NLRP3 activation in mice. J. Lipid Res. 59, 1597–1609 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Ramsden, C. E. et al. Lowering dietary linoleic acid reduces bioactive oxidized linoleic acid metabolites in humans. Prostaglandins Leukot. Essent. Fat. Acids 87, 135–141 (2012).

    Article  CAS  Google Scholar 

  84. Puri, P. et al. The plasma lipidomic signature of nonalcoholic steatohepatitis. Hepatology 50, 1827–1838 (2009).

    Article  CAS  PubMed  Google Scholar 

  85. Loomba, R., Quehenberger, O., Armando, A. & Dennis, E. A. Polyunsaturated fatty acid metabolites as novel lipidomic biomarkers for noninvasive diagnosis of nonalcoholic steatohepatitis. J. Lipid Res. 56, 185–192 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  86. Caussy, C. et al. Plasma eicosanoids as noninvasive biomarkers of liver fibrosis in patients with nonalcoholic steatohepatitis. Ther. Adv. Gastroenterol. 13, 1756284820923904 (2020).

    Article  CAS  Google Scholar 

  87. Feldstein, A. E. et al. Mass spectrometric profiling of oxidized lipid products in human nonalcoholic fatty liver disease and nonalcoholic steatohepatitis. J. Lipid Res. 51, 3046–3054 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Santoro, N. et al. Oxidized fatty acids: a potential pathogenic link between fatty liver and type 2 diabetes in obese adolescents? Antioxid. Redox Signal. 20, 383–389 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Zein, C. O. et al. Pentoxifylline decreases oxidized lipid products in nonalcoholic steatohepatitis: new evidence on the potential therapeutic mechanism. Hepatology 56, 1291–1299 (2012).

    Article  CAS  PubMed  Google Scholar 

  90. Musso, G., Gambino, R., Cassader, M., Paschetta, E. & Sircana, A. Specialized proresolving mediators: enhancing nonalcoholic steatohepatitis and fibrosis resolution. Trends Pharmacol. Sci. 39, 387–401 (2018).

    Article  CAS  PubMed  Google Scholar 

  91. Kotronen, A. et al. Hepatic stearoyl-CoA desaturase (SCD)-1 activity and diacylglycerol but not ceramide concentrations are increased in the nonalcoholic human fatty liver. Diabetes 58, 203–208 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Gorden, D. L. et al. Biomarkers of NAFLD progression: a lipidomics approach to an epidemic. J. Lipid Res. 56, 722–736 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Valsesia, A., Saris, W. H., Astrup, A., Hager, J. & Masoodi, M. Distinct lipid profiles predict improved glycemic control in obese, nondiabetic patients after a low-caloric diet intervention: the diet, obesity and genes randomized trial. Am. J. Clin. Nutr. 104, 566–575 (2016).

    Article  CAS  PubMed  Google Scholar 

  94. Schwarz, J. M., Linfoot, P., Dare, D. & Aghajanian, K. Hepatic de novo lipogenesis in normoinsulinemic and hyperinsulinemic subjects consuming high-fat, low-carbohydrate and low-fat, high-carbohydrate isoenergetic diets. Am. J. Clin. Nutr. 77, 43–50 (2003).

    Article  CAS  PubMed  Google Scholar 

  95. Mayo, R. et al. Metabolomic-based noninvasive serum test to diagnose nonalcoholic steatohepatitis: results from discovery and validation cohorts. Hepatol. Commun. 2, 807–820 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Bril, F. et al. Use of a metabolomic approach to non-invasively diagnose non-alcoholic fatty liver disease in patients with type 2 diabetes mellitus. Diabetes Obes. Metab. 20, 1702–1709 (2018).

    Article  CAS  PubMed  Google Scholar 

  97. Oresic, M. et al. Prediction of non-alcoholic fatty-liver disease and liver fat content by serum molecular lipids. Diabetologia 56, 2266–2274 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Rhee, E. P. et al. Lipid profiling identifies a triacylglycerol signature of insulin resistance and improves diabetes prediction in humans. J. Clin. Invest. 121, 1402–1411 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Suvitaival, T. et al. Lipidome as a predictive tool in progression to type 2 diabetes in Finnish men. Metabolism 78, 1–12 (2018).

    Article  CAS  PubMed  Google Scholar 

  100. Jorgenrud, B. et al. The influence of sample collection methodology and sample preprocessing on the blood metabolic profile. Bioanalysis 7, 991–1006 (2015).

    Article  CAS  PubMed  Google Scholar 

  101. Ramos-Molina, B. et al. A pilot study of serum sphingomyelin dynamics in subjects with severe obesity and non-alcoholic steatohepatitis after sleeve gastrectomy. Obes. Surg. 29, 983–989 (2019).

    Article  PubMed  Google Scholar 

  102. Petersen, M. C. & Shulman, G. I. Roles of diacylglycerols and ceramides in hepatic insulin resistance. Trends Pharmacol. Sci. 38, 649–665 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  103. Pagadala, M., Kasumov, T., McCullough, A. J., Zein, N. N. & Kirwan, J. P. Role of ceramides in nonalcoholic fatty liver disease. Trends Endocrinol. Metab. 23, 365–371 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Haus, J. M. et al. Plasma ceramides are elevated in obese subjects with type 2 diabetes and correlate with the severity of insulin resistance. Diabetes 58, 337–343 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Summers, S. A., Chaurasia, B. & Holland, W. L. Metabolic messengers: ceramides. Nat. Metab. 1, 1051–1058 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  106. Luukkonen, P. K. et al. Hepatic ceramides dissociate steatosis and insulin resistance in patients with non-alcoholic fatty liver disease. J. Hepatol. 64, 1167–1175 (2016).

    Article  CAS  PubMed  Google Scholar 

  107. Apostolopoulou, M. et al. Specific hepatic sphingolipids relate to insulin resistance, oxidative stress, and inflammation in nonalcoholic steatohepatitis. Diabetes Care 41, 1235–1243 (2018).

    Article  CAS  PubMed  Google Scholar 

  108. Chaurasia, B. et al. Targeting a ceramide double bond improves insulin resistance and hepatic steatosis. Science 365, 386–392 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  109. Xia, J. Y. et al. Targeted induction of ceramide degradation leads to improved systemic metabolism and reduced hepatic steatosis. Cell Metab. 22, 266–278 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Hyysalo, J. et al. Circulating triacylglycerol signatures in nonalcoholic fatty liver disease associated with the I148M variant in PNPLA3 and with obesity. Diabetes 63, 312–322 (2014).

    Article  CAS  PubMed  Google Scholar 

  111. Luukkonen, P. K. et al. Human PNPLA3-I148M variant increases hepatic retention of polyunsaturated fatty acids. JCI Insight 4, e127902 (2019).

    Article  PubMed Central  Google Scholar 

  112. Luukkonen, P. K. et al. Impaired hepatic lipid synthesis from polyunsaturated fatty acids in TM6SF2 E167K variant carriers with NAFLD. J. Hepatol. 67, 128–136 (2017).

    Article  CAS  PubMed  Google Scholar 

  113. Zhou, Y. et al. Circulating triacylglycerol signatures and insulin sensitivity in NAFLD associated with the E167K variant in TM6SF2. J. Hepatol. 62, 657–663 (2015).

    Article  CAS  PubMed  Google Scholar 

  114. Mancina, R. M. et al. The MBOAT7-TMC4 variant rs641738 increases risk of nonalcoholic fatty liver disease in individuals of European descent. Gastroenterology 150, 1219–1230.e6 (2016).

    Article  CAS  PubMed  Google Scholar 

  115. Lee, H. C. et al. Caenorhabditis elegans mboa-7, a member of the MBOAT family, is required for selective incorporation of polyunsaturated fatty acids into phosphatidylinositol. Mol. Biol. Cell 19, 1174–1184 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  116. Meroni, M. et al. Mboat7 down-regulation by hyper-insulinemia induces fat accumulation in hepatocytes. EBioMedicine 52, 102658 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  117. Helsley, R. N. et al. Obesity-linked suppression of membrane-bound O-acyltransferase 7 (MBOAT7) drives non-alcoholic fatty liver disease. Elife 8, e49882 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  118. Tanaka, Y. et al. LPIAT1/MBOAT7 depletion increases triglyceride synthesis fueled by high phosphatidylinositol turnover. Gut 70, 180–193 (2021).

    Article  CAS  PubMed  Google Scholar 

  119. Fondevila, M. F. et al. The L-α-lysophosphatidylinositol/G protein-coupled receptor 55 system induces the development of nonalcoholic steatosis and steatohepatitis. Hepatology 73, 606–624 (2021).

    Article  CAS  PubMed  Google Scholar 

  120. Luukkonen, P. K. et al. Hydroxysteroid 17-β dehydrogenase 13 variant increases phospholipids and protects against fibrosis in nonalcoholic fatty liver disease. JCI Insight 5, e132158 (2020).

    Article  PubMed Central  Google Scholar 

  121. Luukkonen, P. K. et al. MARC1 variant rs2642438 increases hepatic phosphatidylcholines and decreases severity of non-alcoholic fatty liver disease in humans. J. Hepatol. 73, 725–726 (2020).

    Article  CAS  PubMed  Google Scholar 

  122. Chiang, J. Y. Regulation of bile acid synthesis: pathways, nuclear receptors, and mechanisms. J. Hepatol. 40, 539–551 (2004).

    Article  CAS  PubMed  Google Scholar 

  123. Behr, A. C., Plinsch, C., Braeuning, A. & Buhrke, T. Activation of human nuclear receptors by perfluoroalkylated substances (PFAS). Toxicol. Vitro 62, 104700 (2020).

    Article  Google Scholar 

  124. Bjork, J. A., Butenhoff, J. L. & Wallace, K. B. Multiplicity of nuclear receptor activation by PFOA and PFOS in primary human and rodent hepatocytes. Toxicology 288, 8–17 (2011).

    Article  CAS  PubMed  Google Scholar 

  125. Zhang, L. et al. Persistent organic pollutants modify gut microbiota-host metabolic homeostasis in mice through aryl hydrocarbon receptor activation. Environ. Health Perspect. 123, 679–688 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  126. Chiang, J. Y. Recent advances in understanding bile acid homeostasis. F1000Res 6, 2029 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  127. Honda, A. et al. Regulation of bile acid metabolism in mouse models with hydrophobic bile acid composition. J. Lipid Res. 61, 54–69 (2020).

    Article  CAS  PubMed  Google Scholar 

  128. Ticho, A. L., Malhotra, P., Dudeja, P. K., Gill, R. K. & Alrefai, W. A. Bile acid receptors and gastrointestinal functions. Liver Res. 3, 31–39 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  129. Jiao, N. et al. Suppressed hepatic bile acid signalling despite elevated production of primary and secondary bile acids in NAFLD. Gut 67, 1881–1891 (2018).

    Article  CAS  PubMed  Google Scholar 

  130. Pineda Torra, I. et al. Bile acids induce the expression of the human peroxisome proliferator-activated receptor α gene via activation of the farnesoid X receptor. Mol. Endocrinol. 17, 259–272 (2003).

    Article  PubMed  Google Scholar 

  131. Kast, H. R. et al. Farnesoid X-activated receptor induces apolipoprotein C-II transcription: a molecular mechanism linking plasma triglyceride levels to bile acids. Mol. Endocrinol. 15, 1720–1728 (2001).

    Article  CAS  PubMed  Google Scholar 

  132. Dasarathy, S. et al. Elevated hepatic fatty acid oxidation, high plasma fibroblast growth factor 21, and fasting bile acids in nonalcoholic steatohepatitis. Eur. J. Gastroenterol. Hepatol. 23, 382–388 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Aranha, M. M. et al. Bile acid levels are increased in the liver of patients with steatohepatitis. Eur. J. Gastroenterol. Hepatol. 20, 519–525 (2008).

    Article  CAS  PubMed  Google Scholar 

  134. Mouzaki, M. et al. Bile acids and dysbiosis in non-alcoholic fatty liver disease. PLoS ONE 11, e0151829 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  135. Puri, P. et al. The presence and severity of nonalcoholic steatohepatitis is associated with specific changes in circulating bile acids. Hepatology 67, 534–548 (2018).

    Article  CAS  PubMed  Google Scholar 

  136. Christinat, N., Valsesia, A. & Masoodi, M. Untargeted profiling of bile acids and lysophospholipids identifies the lipid signature associated with glycemic outcome in an obese non-diabetic clinical cohort. Biomolecules 10, 1049 (2020).

    Article  CAS  PubMed Central  Google Scholar 

  137. Wruck, W. & Adjaye, J. Meta-analysis reveals up-regulation of cholesterol processes in non-alcoholic and down-regulation in alcoholic fatty liver disease. World J. Hepatol. 9, 443–454 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  138. Legry, V. et al. Bile acid alterations are associated with insulin resistance, but not with NASH, in obese subjects. J. Clin. Endocrinol. Metab. 102, 3783–3794 (2017).

    Article  PubMed  Google Scholar 

  139. Bechmann, L. P. et al. Free fatty acids repress small heterodimer partner (SHP) activation and adiponectin counteracts bile acid-induced liver injury in superobese patients with nonalcoholic steatohepatitis. Hepatology 57, 1394–1406 (2013).

    Article  CAS  PubMed  Google Scholar 

  140. Lake, A. D. et al. Decreased hepatotoxic bile acid composition and altered synthesis in progressive human nonalcoholic fatty liver disease. Toxicol. Appl. Pharmacol. 268, 132–140 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  141. Tanaka, N., Matsubara, T., Krausz, K. W., Patterson, A. D. & Gonzalez, F. J. Disruption of phospholipid and bile acid homeostasis in mice with nonalcoholic steatohepatitis. Hepatology 56, 118–129 (2012).

    Article  CAS  PubMed  Google Scholar 

  142. Valanejad, L. et al. Dysregulation of Δ(4)-3-oxosteroid 5β-reductase in diabetic patients: implications and mechanisms. Mol. Cell Endocrinol. 470, 127–141 (2018).

    Article  CAS  PubMed  Google Scholar 

  143. Chen, J. et al. Ratio of conjugated chenodeoxycholic to muricholic acids is associated with severity of nonalcoholic steatohepatitis. Obesity 27, 2055–2066 (2019).

    Article  CAS  PubMed  Google Scholar 

  144. Chavez-Talavera, O., Tailleux, A., Lefebvre, P. & Staels, B. Bile acid control of metabolism and inflammation in obesity, type 2 diabetes, dyslipidemia, and nonalcoholic fatty liver disease. Gastroenterology 152, 1679–1694.e3 (2017).

    Article  CAS  PubMed  Google Scholar 

  145. 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  CAS  PubMed  PubMed Central  Google Scholar 

  146. Aragones, G. et al. Circulating microbiota-derived metabolites: a “liquid biopsy? Int. J. Obes. 44, 875–885 (2020).

    Article  CAS  Google Scholar 

  147. Ferslew, B. C. et al. Altered bile acid metabolome in patients with nonalcoholic steatohepatitis. Dig. Dis. Sci. 60, 3318–3328 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  148. Aron-Wisnewsky, J. et al. Gut microbiota and human NAFLD: disentangling microbial signatures from metabolic disorders. Nat. Rev. Gastroenterol. Hepatol. 17, 279–297 (2020).

    Article  PubMed  Google Scholar 

  149. Lefere, S. & Tacke, F. Macrophages in obesity and non-alcoholic fatty liver disease: crosstalk with metabolism. JHEP Rep. 1, 30–43 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  150. Szabo, G., Bala, S., Petrasek, J. & Gattu, A. Gut-liver axis and sensing microbes. Dig. Dis. 28, 737–744 (2010).

    Article  PubMed  Google Scholar 

  151. Ji, Y., Yin, Y., Li, Z. & Zhang, W. Gut microbiota-derived components and metabolites in the progression of non-alcoholic fatty liver disease (NAFLD). Nutrients 11, 1712 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  152. Jasirwan, C. O. M., Lesmana, C. R. A., Hasan, I., Sulaiman, A. S. & Gani, R. A. The role of gut microbiota in non-alcoholic fatty liver disease: pathways of mechanisms. Biosci. Microbiota Food Health 38, 81–88 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Liu, Q. et al. Role and effective therapeutic target of gut microbiota in NAFLD/NASH. Exp. Ther. Med. 18, 1935–1944 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Pan, X., Wen, S. W., Kaminga, A. C. & Liu, A. Gut metabolites and inflammation factors in non-alcoholic fatty liver disease: a systematic review and meta-analysis. Sci. Rep. 10, 8848 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  155. Jiang, W. et al. Dysbiosis gut microbiota associated with inflammation and impaired mucosal immune function in intestine of humans with non-alcoholic fatty liver disease. Sci. Rep. 5, 8096 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Mouzaki, M. et al. Intestinal microbiota in patients with nonalcoholic fatty liver disease. Hepatology 58, 120–127 (2013).

    Article  CAS  PubMed  Google Scholar 

  157. Rau, M. et al. Fecal SCFAs and SCFA-producing bacteria in gut microbiome of human NAFLD as a putative link to systemic T-cell activation and advanced disease. United Eur. Gastroenterol. J. 6, 1496–1507 (2018).

    Article  CAS  Google Scholar 

  158. Ramirez-Perez, O., Cruz-Ramon, V., Chinchilla-Lopez, P. & Mendez-Sanchez, N. The role of the gut microbiota in bile acid metabolism. Ann. Hepatol. 16, s15–s20 (2017).

    Article  PubMed  Google Scholar 

  159. van Best, N. et al. Bile acids drive the newborn’s gut microbiota maturation. Nat Commun 11, 3692 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  160. Tremblay, S. et al. Bile acid administration elicits an intestinal antimicrobial program and reduces the bacterial burden in two mouse models of enteric infection. Infect. Immun. 85, e00942 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  161. Ridlon, J. M., Kang, D. J. & Hylemon, P. B. Bile salt biotransformations by human intestinal bacteria. J. Lipid Res. 47, 241–259 (2006).

    Article  CAS  PubMed  Google Scholar 

  162. Sarathy, J. et al. The Yin and Yang of bile acid action on tight junctions in a model colonic epithelium. Physiol. Rep. 5, e13294 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  163. Corbin, K. D. & Zeisel, S. H. Choline metabolism provides novel insights into nonalcoholic fatty liver disease and its progression. Curr. Opin. Gastroenterol. 28, 159–165 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  164. Chen, Y. M. et al. Associations of gut-flora-dependent metabolite trimethylamine-N-oxide, betaine and choline with non-alcoholic fatty liver disease in adults. Sci. Rep. 6, 19076 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Hoyles, L. et al. Molecular phenomics and metagenomics of hepatic steatosis in non-diabetic obese women. Nat. Med. 24, 1070–1080 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  166. Caussy, C. & Loomba, R. Gut microbiome, microbial metabolites and the development of NAFLD. Nat. Rev. Gastroenterol. Hepatol. 15, 719–720 (2018).

    Article  PubMed  Google Scholar 

  167. Krishnan, S. et al. Gut microbiota-derived tryptophan metabolites modulate inflammatory response in hepatocytes and macrophages. Cell Rep. 23, 1099–1111 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  168. Gastaldelli, A. & Cusi, K. From NASH to diabetes and from diabetes to NASH: mechanisms and treatment options. JHEP Rep. 1, 312–328 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  169. Wolfe, R. R. & Chinkes, D. L. Isotope Tracers in Metabolic Research: Principles and Practice of Kinetic Analysis (Wiley, 2004).

  170. Cusi, K., Kashyap, S., Gastaldelli, A., Bajaj, M. & Cersosimo, E. Effects on insulin secretion and insulin action of a 48-h reduction of plasma free fatty acids with acipimox in nondiabetic subjects genetically predisposed to type 2 diabetes. Am. J. Physiol. Endocrinol. Metab. 292, E1775–E1781 (2007).

    Article  CAS  PubMed  Google Scholar 

  171. Smith, G. I. et al. Insulin resistance drives hepatic de novo lipogenesis in nonalcoholic fatty liver disease. J. Clin. Invest. 130, 1453–1460 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  172. Kalhan, S. C., Bugianesi, E., McCullough, A. J., Hanson, R. W. & Kelley, D. E. Estimates of hepatic glyceroneogenesis in type 2 diabetes mellitus in humans. Metabolism 57, 305–312 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  173. Jin, E. S., Browning, J. D., Murphy, R. E. & Malloy, C. R. Fatty liver disrupts glycerol metabolism in gluconeogenic and lipogenic pathways in humans. J. Lipid Res. 59, 1685–1694 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  174. Lytle, K. A. et al. Hepatic fatty acid balance and hepatic fat content in humans with severe obesity. J. Clin. Endocrinol. Metab. 104, 6171–6181 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  175. Satapati, S. et al. Mitochondrial metabolism mediates oxidative stress and inflammation in fatty liver. J. Clin. Invest. 125, 4447–4462 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  176. Sunny, N. E. et al. Cross-talk between branched-chain amino acids and hepatic mitochondria is compromised in nonalcoholic fatty liver disease. Am. J. Physiol. Endocrinol. Metab. 309, E311–E319 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  177. Burla, B. et al. MS-based lipidomics of human blood plasma: a community-initiated position paper to develop accepted guidelines. J. Lipid Res. 59, 2001–2017 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Bowden, J. A. et al. Harmonizing lipidomics: NIST interlaboratory comparison exercise for lipidomics using SRM 1950-metabolites in frozen human plasma. J. Lipid Res. 58, 2275–2288 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  179. Feldman, A. et al. Clinical and metabolic characterization of lean Caucasian subjects with non-alcoholic fatty liver. Am. J. Gastroenterol. 112, 102–110 (2017).

    Article  PubMed  Google Scholar 

  180. Pirola, C. J. & Sookoian, S. Multiomics biomarkers for the prediction of nonalcoholic fatty liver disease severity. World J. Gastroenterol. 24, 1601–1615 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Sookoian, S. & Pirola, C. J. Liver enzymes, metabolomics and genome-wide association studies: from systems biology to the personalized medicine. World J. Gastroenterol. 21, 711–725 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  182. Valsesia, A. et al. Integrative phenotyping of glycemic responders upon clinical weight loss using multi-omics. Sci. Rep. 10, 9236 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Magnusdottir, S. et al. Generation of genome-scale metabolic reconstructions for 773 members of the human gut microbiota. Nat. Biotechnol. 35, 81–89 (2017).

    Article  CAS  PubMed  Google Scholar 

  184. Sen, P. & Oresic, M. Metabolic modeling of human gut microbiota on a genome scale: an overview. Metabolites 9, 22 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  185. Gastaldelli A. in Non-Alcoholic Fatty Liver Disease (ed. Bugianesi, E.) 49–71 (Springer, 2020)

  186. Kleiner, D. E. et al. Design and validation of a histological scoring system for nonalcoholic fatty liver disease. Hepatology 41, 1313–1321 (2005).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

All authors of this manuscript are members of the Liver Investigation: Testing Marker Utility in Steatohepatitis (LITMUS) Consortium. The authors are supported by the EU’s Horizon 2020 Programme under the EPoS project (grant no. 634413) and the Innovative Medicines Initiative 2 Joint Undertaking under the LITMUS project (grant no. 777377) and Swiss National Science Foundation (SNSF) (grant no. 190686). This Joint Undertaking receives support from the EU’s Horizon 2020 programme and EFPIA.

Author information

Authors and Affiliations

Authors

Contributions

M.M. researched data for the article, made a substantial contribution to discussion of content, wrote the first draft, and reviewed/edited the manuscript before submission. A.G. and T.H. made a substantial contribution to discussion of content, wrote the article, and reviewed/edited the manuscript before submission. E.A. researched data for the article and wrote the article. C.A. and M.G. researched data for the article, and reviewed/edited the manuscript before submission. J.B., Q.M.A., O.M., P.O. and J.M.M. reviewed/edited the manuscript before submission. J.-F.D. made a substantial contribution to discussion of content, and reviewed/edited the manuscript before submission. M.O. researched data for the article, made a substantial contribution to discussion of content, contributed to writing and reviewed/edited the manuscript before submission.

Corresponding authors

Correspondence to Mojgan Masoodi or Matej Orešič.

Ethics declarations

Competing interests

J.B. was an employee and shareholder of Pfizer. E.A., C.A. and P.O. are employees of OWL Metabolomics. A.G. is a consultant for Eli Lilly, Inventiva, Genentech, Menarini, Gilead, Novo Nordisk, AstraZeneca and Boehringer outside the submitted work. J.-F.D. is on advisory committees for Abbvie, Bayer, BMS, Falk, Genfit, Genkyotex, Gilead Science, HepaRegenix, Intercept, Lilly, Merck and Novartis; speaking and teaching at Bayer, BMS, Intercept, Genfit, Gilead Science and Novartis. Q.M.A. has active research collaborations (including research collaborations supported through the EU IMI2 LITMUS Consortium*) with Abbvie, Antaros Medical*, Allergan/Tobira*, AstraZeneca*, BMS*, Boehringer Ingelheim International GMBH*, Echosens*, Ellegaard Gottingen Minipigs AS*, Eli Lilly & Company Ltd.*, Exalenz Bioscience Ltd.*, Genfit SA*, Glympse Bio, GlaxoSmithKline, HistoIndex*, Intercept Pharma Europe Ltd.*, iXscient Ltd.*, Nordic Bioscience*, Novartis Pharma AG*, Novo Nordisk A/S*, One Way Liver SL*, Perspectum Diagnostics*, Pfizer Ltd.*, Resoundant*, Sanofi-Aventis Deutschland GMBH*, SomaLogic Inc.* and Takeda Pharmaceuticals International SA*, acts as a consultant for 89Bio, Abbott Laboratories, Acuitas Medical, Allergan/Tobira, Altimmune, AstraZeneca, Axcella, Blade, BMS, BNN Cardio, Celgene, Cirius, CymaBay, EcoR1, E3Bio, Eli Lilly & Company Ltd., Galmed, Genentech, Genfit SA, Gilead, Grunthal, HistoIndex, Indalo, Imperial Innovations, Intercept Pharma Europe Ltd., Inventiva, IQVIA, Janssen, Madrigal, MedImmune, Metacrine, NewGene, NGMBio, North Sea Therapeutics, Novartis, Novo Nordisk A/S, Pfizer Ltd., Poxel, ProSciento, Raptor Pharma, Servier, Terns, Viking Therapeutics and Speaker for Abbott Laboratories, Allergan/Tobira, BMS, Clinical Care Options, Falk, Fishawack, Genfit SA, Gilead, Integritas Communications, Kenes and MedScape, and has received royalties from Elsevier Ltd (Davidson’s Principles & Practice of Medicine textbook). All other authors declare no competing interests.

Additional information

Peer review information

Nature Reviews Gastroenterology & Hepatology thanks C. Pirola, F. Bril and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Related links

IMI2 LITMUS: https://litmus-project.eu/

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Masoodi, M., Gastaldelli, A., Hyötyläinen, T. et al. Metabolomics and lipidomics in NAFLD: biomarkers and non-invasive diagnostic tests. Nat Rev Gastroenterol Hepatol 18, 835–856 (2021). https://doi.org/10.1038/s41575-021-00502-9

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41575-021-00502-9

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing