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.

  • Article
  • Published:

Aging promotes metabolic dysfunction-associated steatotic liver disease by inducing ferroptotic stress

Abstract

Susceptibility to the biological consequences of aging varies among organs and individuals. We analyzed hepatocyte transcriptomes of healthy young and aged male mice to generate an aging hepatocyte gene signature, used it to deconvolute transcriptomic data from humans and mice with metabolic dysfunction-associated liver disease, validated findings with functional studies in mice and applied the signature to transcriptomic data from other organs to determine whether aging-sensitive degenerative mechanisms are conserved. We discovered that the signature enriches in diseased livers in parallel with degeneration. It is also enriched in failing human hearts, diseased kidneys and pancreatic islets from individuals with diabetes. The signature includes genes that control ferroptosis. Aged mice develop more hepatocyte ferroptosis and liver degeneration than young mice when fed diets that induce metabolic stress. Inhibiting ferroptosis shifts the liver transcriptome of old mice toward that of young mice and reverses aging-exacerbated liver damage, identifying ferroptosis as a tractable, conserved mechanism for aging-related tissue degeneration.

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: Liver accumulation of biologically old HCs parallels MASLD severity in people.
Fig. 2: Aging HCs in MASLD lose metabolic functions and exhibit ferroptotic stress.
Fig. 3: Livers of patients with MASLD exhibit ferroptotic stress.
Fig. 4: Ferroptotic stress and lipotoxicity are increased in livers of old mice.
Fig. 5: Aging increases ferroptotic stress and promotes HC susceptibility to lipotoxicity.
Fig. 6: Fer1 protects HCs from aging-associated ferroptotic stress and senescence during diet-induced MASH.
Fig. 7: Fer1 prevents aging-related exacerbation of MASH.
Fig. 8: Liver aging correlates with dysfunction of multiple-organ systems.

Similar content being viewed by others

Data availability

All data generated by the present study are included in this article and the Supplementary Information. The bulk RNA-seq data from the 6-week CDA-HFD-fed young versus old mice have been deposited in the GEO with accession no. GSE262457. The snRNA-seq data from the 22-week-old, chow- versus CDA-HFD-fed mice have also been submitted to the GEO and can be accessed with accession no. GSE262939. Additional public transcriptomic datasets for the samples discussed in the present study are available in the GEO database under the following accession nos.: GSE181761 (comparison of old versus young mouse HCs), GSE132042 (old versus young mouse liver), GSE213623 (Duke MASLD cohort), GSE33814 (German MASLD cohort), GSE167523 (Japanese MASLD cohort), GSE135251 (European MASLD cohort), GSE174748 (snRNA-seq on liver of controls and patients with MASLD) and GSE50244 (pancreatic islets). Moreover, the RNA-seq dataset for patients with heart failure was obtained from Zenodo at https://doi.org/10.5281/zenodo.4114616 (ref. 106). The RNA-seq dataset for patients with kidney fibrosis was sourced from E-MTAB-2502 on the EMBL-EBI website at https://www.ebi.ac.uk/biostudies/arrayexpress/studies/E-MTAB-2502. In addition, the RNA-seq dataset for 226 healthy human livers was acquired from the GTEx Portal v.8 at https://gtexportal.org/home. Source data are provided with this paper.

Code availability

The present study did not generate any unique code or algorithm. The algorithms used for the analysis in the present study are all publicly available.

References

  1. Chang, A. Y., Skirbekk, V. F., Tyrovolas, S., Kassebaum, N. J. & Dieleman, J. L. Measuring population ageing: an analysis of the Global Burden of Disease Study 2017. Lancet Public Health 4, e159–e167 (2019).

    PubMed  PubMed Central  Google Scholar 

  2. Sieck, G. C. Physiology in perspective: aging and underlying pathophysiology. Physiology 32, 7–8 (2017).

    PubMed  Google Scholar 

  3. Tian, Y. E. et al. Heterogeneous aging across multiple organ systems and prediction of chronic disease and mortality. Nat. Med. 29, 1221–1231 (2023).

    CAS  PubMed  Google Scholar 

  4. Horvath, S. & Raj, K. DNA methylation-based biomarkers and the epigenetic clock theory of ageing. Nat. Rev. Genet. 19, 371–384 (2018).

    CAS  PubMed  Google Scholar 

  5. Maeso-Diaz, R. et al. Aging reduces liver resiliency by dysregulating Hedgehog signaling. Aging Cell 21, e13530 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Fitzgerald, K. N. et al. Potential reversal of epigenetic age using a diet and lifestyle intervention: a pilot randomized clinical trial. Aging 13, 9419–9432 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Poganik, J. R. et al. Biological age is increased by stress and restored upon recovery. Cell Metab. 35, 807–820 e805 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Raj, K. & Horvath, S. Current perspectives on the cellular and molecular features of epigenetic ageing. Exp. Biol. Med. 245, 1532–1542 (2020).

    CAS  Google Scholar 

  9. Jensen-Cody, S. O. & Potthoff, M. J. Hepatokines and metabolism: deciphering communication from the liver. Mol. Metab. 44, 101138 (2021).

    CAS  PubMed  Google Scholar 

  10. Rui, L. Energy metabolism in the liver. Compr. Physiol. 4, 177–197 (2014).

    PubMed  PubMed Central  Google Scholar 

  11. Timchenko, N. A. Aging and liver regeneration. Trends Endocrinol. Metab. 20, 171–176 (2009).

    CAS  PubMed  Google Scholar 

  12. Kim, I. H., Kisseleva, T. & Brenner, D. A. Aging and liver disease. Curr. Opin. Gastroenterol. 31, 184–191 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Alqahtani, S. A. & Schattenberg, J. M. NAFLD in the elderly. Clin. Interv. Aging 16, 1633–1649 (2021).

    PubMed  PubMed Central  Google Scholar 

  14. Loomba, R. et al. DNA methylation signatures reflect aging in patients with non-alcoholic steatohepatitis. JCI Insight https://doi.org/10.1172/jci.insight.96685 (2018).

  15. Palmer, A. K. & Jensen, M. D. Metabolic changes in aging humans: current evidence and therapeutic strategies. J. Clin. Invest. https://doi.org/10.1172/JCI158451 (2022).

  16. Chen, T. et al. Hepatocyte smoothened activity controls susceptibility to insulin resistance and non-alcoholic fatty liver disease. Cell. Mol. Gastroenterol. Hepatol. 15, 949–970 (2023).

  17. Maeso-Diaz, R. et al. Targeting senescent hepatocytes using the thrombomodulin-PAR1 inhibitor vorapaxar ameliorates NAFLD progression. Hepatology 78, 1209–1222 (2023).

    PubMed  Google Scholar 

  18. Tam, B. T., Morais, J. A. & Santosa, S. Obesity and ageing: two sides of the same coin. Obes. Rev. 21, e12991 (2020).

    PubMed  Google Scholar 

  19. Fabbrini, E., Sullivan, S. & Klein, S. Obesity and non-alcoholic fatty liver disease: biochemical, metabolic, and clinical implications. Hepatology 51, 679–689 (2010).

  20. Consortium, G. T. The Genotype-Tissue Expression (GTEx) project. Nat. Genet. 45, 580–585 (2013).

    Google Scholar 

  21. Carithers, L. J. et al. A novel approach to high-quality postmortem tissue procurement: the GTEx Project. Biopreserv. Biobank 13, 311–319 (2015).

    PubMed  PubMed Central  Google Scholar 

  22. 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).

  23. Filliol, A. et al. Opposing roles of hepatic stellate cell subpopulations in hepatocarcinogenesis. Nature 610, 356–365 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

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

  25. Stockwell, B. R. Ferroptosis turns 10: emerging mechanisms, physiological functions, and therapeutic applications. Cell 185, 2401–2421 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Miao, R. et al. Iron metabolism and ferroptosis in type 2 diabetes mellitus and complications: mechanisms and therapeutic opportunities. Cell Death Dis. 14, 186 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Zhang, S. et al. Ferroptosis increases obesity: crosstalk between adipocytes and the neuroimmune system. Front. Immunol. 13, 1049936 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Zhang, H., Zhang, E. & Hu, H. Role of ferroptosis in non-alcoholic fatty liver disease and its implications for therapeutic strategies. Biomedicines https://doi.org/10.3390/biomedicines9111660 (2021).

  29. Zheng, J. & Conrad, M. The metabolic underpinnings of ferroptosis. Cell Metab. 32, 920–937 (2020).

    CAS  PubMed  Google Scholar 

  30. Duan, J. Y. et al. Ferroptosis and its potential role in metabolic diseases: a curse or revitalization? Front. Cell Dev. Biol. 9, 701788 (2021).

    PubMed  PubMed Central  Google Scholar 

  31. George, D. K. et al. Increased hepatic iron concentration in non-alcoholic steatohepatitis is associated with increased fibrosis. Gastroenterology 114, 311–318 (1998).

  32. Sumida, Y. et al. Serum thioredoxin levels as a predictor of steatohepatitis in patients with non-alcoholic fatty liver disease. J. Hepatol. 38, 32–38 (2003).

  33. Nelson, J. E. et al. Relationship between the pattern of hepatic iron deposition and histological severity in non-alcoholic fatty liver disease. Hepatology 53, 448–457 (2011).

  34. Buzzetti, E. et al. Evaluating the association of serum ferritin and hepatic iron with disease severity in non-alcoholic fatty liver disease. Liver Int. 39, 1325–1334 (2019).

    CAS  PubMed  Google Scholar 

  35. Eder, S. K. et al. Mesenchymal iron deposition is associated with adverse long-term outcome in non-alcoholic fatty liver disease. Liver Int. 40, 1872–1882 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Nemeth, E. et al. Hepcidin regulates cellular iron efflux by binding to ferroportin and inducing its internalization. Science 306, 2090–2093 (2004).

    CAS  PubMed  Google Scholar 

  37. Maus, M. et al. Iron accumulation drives fibrosis, senescence and the senescence-associated secretory phenotype. Nat. Metab. 5, 2111–2130 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Ferrucci, L. & Kuchel, G. A. Heterogeneity of aging: individual risk factors, mechanisms, patient priorities, and outcomes. J. Am. Geriatr. Soc. 69, 610–612 (2021).

    PubMed  PubMed Central  Google Scholar 

  39. Masaldan, S. et al. Iron accumulation in senescent cells is coupled with impaired ferritinophagy and inhibition of ferroptosis. Redox Biol. 14, 100–115 (2018).

    CAS  PubMed  Google Scholar 

  40. Pirpamer, L. et al. Determinants of iron accumulation in the normal aging brain. Neurobiol. Aging 43, 149–155 (2016).

    CAS  PubMed  Google Scholar 

  41. Jiang, X., Stockwell, B. R. & Conrad, M. Ferroptosis: mechanisms, biology and role in disease. Nat. Rev. Mol. Cell Biol. 22, 266–282 (2021).

    PubMed  PubMed Central  Google Scholar 

  42. Hardy, T., Oakley, F., Anstee, Q. M. & Day, C. P. Non-alcoholic fatty liver disease: pathogenesis and disease spectrum. Annu. Rev. Pathol. 11, 451–496 (2016).

  43. Lopez-Otin, C., Blasco, M. A., Partridge, L., Serrano, M. & Kroemer, G. Hallmarks of aging: an expanding universe. Cell 186, 243–278 (2023).

    CAS  PubMed  Google Scholar 

  44. 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).

    PubMed  Google Scholar 

  45. Yu, D. et al. Higher dietary choline intake is associated with lower risk of non-alcoholic fatty liver in normal-weight Chinese women. J. Nutr. 144, 2034–2040 (2014).

  46. Chai, C. et al. Dietary choline intake and non-alcoholic fatty liver disease (NAFLD) in U.S. adults: National Health and Nutrition Examination Survey (NHANES) 2017–2018. Eur. J. Clin. Nutr. 77, 1160–1166 (2023).

    PubMed  Google Scholar 

  47. He, S. & Sharpless, N. E. Senescence in health and disease. Cell 169, 1000–1011 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Coradduzza, D. et al. Ferroptosis and senescence: a systematic review. Int. J. Mol. Sci. https://doi.org/10.3390/ijms24043658 (2023).

  49. Papatheodoridi, A. M., Chrysavgis, L., Koutsilieris, M. & Chatzigeorgiou, A. The role of senescence in the development of non-alcoholic fatty liver disease and progression to non-alcoholic steatohepatitis. Hepatology 71, 363–374 (2020).

  50. Saul, D. et al. A new gene set identifies senescent cells and predicts senescence-associated pathways across tissues. Nat. Commun. 13, 4827 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Xiong, H. et al. Suppressed farnesoid X receptor by iron overload in mice and humans potentiates iron-induced hepatotoxicity. Hepatology 76, 387–403 (2022).

    CAS  PubMed  Google Scholar 

  52. Tschuck, J. et al. Farnesoid X receptor activation by bile acids suppresses lipid peroxidation and ferroptosis. Nat. Commun. 14, 6908 (2023).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Kim, D. H. et al. Farnesoid X receptor protects against cisplatin-induced acute kidney injury by regulating the transcription of ferroptosis-related genes. Redox Biol. 54, 102382 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Tang, J. et al. Farnesoid X receptor plays a key role in ochratoxin A-induced nephrotoxicity by targeting ferroptosis in vivo and in vitro. J. Agric. Food Chem. 71, 14365–14378 (2023).

    CAS  PubMed  Google Scholar 

  55. Chiang, J. Y. L. & Ferrell, J. M. Bile acid metabolism in liver pathobiology. Gene Expr. 18, 71–87 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Wang, C. Y. & Babitt, J. L. Liver iron sensing and body iron homeostasis. Blood 133, 18–29 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Sanyal, A. J. et al. Prospective study of outcomes in adults with non-alcoholic fatty liver disease. N. Engl. J. Med. 385, 1559–1569 (2021).

  58. Pei, Z. et al. FUNDC1 insufficiency sensitizes high fat diet intake-induced cardiac remodeling and contractile anomaly through ACSL4-mediated ferroptosis. Metabolism 122, 154840 (2021).

    CAS  PubMed  Google Scholar 

  59. Zhao, X. et al. Adipose tissue macrophage-derived exosomes induce ferroptosis via glutathione synthesis inhibition by targeting SLC7A11 in obesity-induced cardiac injury. Free Radic. Biol. Med. 182, 232–245 (2022).

    CAS  PubMed  Google Scholar 

  60. Balzer, M. S. et al. Single-cell analysis highlights differences in druggable pathways underlying adaptive or fibrotic kidney regeneration. Nat. Commun. 13, 4018 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Targher, G., Corey, K. E., Byrne, C. D. & Roden, M. The complex link between NAFLD and type 2 diabetes mellitus—mechanisms and treatments. Nat. Rev. Gastroenterol. Hepatol. 18, 599–612 (2021).

    PubMed  Google Scholar 

  62. Elumalai, S., Karunakaran, U., Moon, J. S. & Won, K. C. Ferroptosis signaling in pancreatic beta-cells: novel insights & therapeutic targeting. Int. J. Mol. Sci. https://doi.org/10.3390/ijms232213679 (2022).

  63. Vitalakumar, D., Sharma, A. & Flora, S. J. S. Ferroptosis: a potential therapeutic target for neurodegenerative diseases. J. Biochem. Mol. Toxicol. 35, e22830 (2021).

    CAS  PubMed  Google Scholar 

  64. Hahn, V. S. et al. Myocardial gene expression signatures in human heart failure with preserved ejection fraction. Circulation 143, 120–134 (2021).

    CAS  PubMed  Google Scholar 

  65. Salah, H. M. et al. Relationship of non-alcoholic fatty liver disease and heart failure with preserved ejection fraction. JACC Basic Transl. Sci. 6, 918–932 (2021).

  66. Kang, H. M. et al. Defective fatty acid oxidation in renal tubular epithelial cells has a key role in kidney fibrosis development. Nat. Med. 21, 37–46 (2015).

    CAS  PubMed  Google Scholar 

  67. Fadista, J. et al. Global genomic and transcriptomic analysis of human pancreatic islets reveals novel genes influencing glucose metabolism. Proc. Natl Acad. Sci. USA 111, 13924–13929 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Bugianesi, E., McCullough, A. J. & Marchesini, G. Insulin resistance: a metabolic pathway to chronic liver disease. Hepatology 42, 987–1000 (2005).

    CAS  PubMed  Google Scholar 

  69. Rowe, J. W., Minaker, K. L., Pallotta, J. A. & Flier, J. S. Characterization of the insulin resistance of aging. J. Clin. Invest. 71, 1581–1587 (1983).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Miotto, G. et al. Insight into the mechanism of ferroptosis inhibition by ferrostatin-1. Redox Biol. 28, 101328 (2020).

    CAS  PubMed  Google Scholar 

  71. Semmler, G., Datz, C., Reiberger, T. & Trauner, M. Diet and exercise in NAFLD/NASH: beyond the obvious. Liver Int. 41, 2249–2268 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Shu, Y. Y. et al. Attenuation by time-restricted feeding of high-fat and high-fructose diet-Induced NASH in mice is related to Per2 and ferroptosis. Oxid. Med. Cell Longev. 2022, 8063897 (2022).

    PubMed  PubMed Central  Google Scholar 

  73. Liu, T. et al. Treadmill training reduces cerebral ischemia-reperfusion injury by inhibiting ferroptosis through activation of SLC7A11/GPX4. Oxid. Med. Cell Longev. 2022, 8693664 (2022).

    PubMed  PubMed Central  Google Scholar 

  74. Violi, F. & Cangemi, R. Pioglitazone, vitamin E, or placebo for non-alcoholic steatohepatitis. N. Engl. J. Med. 363, 1185–1186 (2010).

  75. Doll, S. et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat. Chem. Biol. 13, 91–98 (2017).

    CAS  PubMed  Google Scholar 

  76. Sumida, Y. & Yoneda, M. Current and future pharmacological therapies for NAFLD/NASH. J. Gastroenterol. 53, 362–376 (2018).

    CAS  PubMed  Google Scholar 

  77. Han, J. X. et al. SGLT2 inhibitor empagliflozin promotes revascularization in diabetic mouse hindlimb ischemia by inhibiting ferroptosis. Acta Pharmacol. Sin. 44, 1161–1174 (2023).

    CAS  PubMed  Google Scholar 

  78. Gu, Y. et al. Comparative efficacy of glucagon-like peptide 1 (GLP-1) receptor agonists, pioglitazone and vitamin E for liver histology among patients with non-alcoholic fatty liver disease: systematic review and pilot network meta-analysis of randomized controlled trials. Expert Rev. Gastroenterol. Hepatol. 17, 273–282 (2023).

  79. An, J. R. et al. Liraglutide alleviates cognitive deficit in db/db mice: involvement in oxidative stress, iron overload, and ferroptosis. Neurochem. Res. 47, 279–294 (2022).

    CAS  PubMed  Google Scholar 

  80. Li, Q. et al. Ferroptosis: the potential target in heart failure with preserved ejection fraction. Cells https://doi.org/10.3390/cells11182842 (2022).

  81. Zhou, Y. et al. The role of ferroptosis in the development of acute and chronic kidney diseases. J. Cell. Physiol. 237, 4412–4427 (2022).

    CAS  PubMed  Google Scholar 

  82. Wang, T. W. et al. Blocking PD-L1-PD-1 improves senescence surveillance and ageing phenotypes. Nature 611, 358–364 (2022).

    CAS  PubMed  Google Scholar 

  83. Luukkonen, P. K. et al. Inhibition of HSD17B13 protects against liver fibrosis by inhibition of pyrimidine catabolism in non-alcoholic steatohepatitis. Proc. Natl Acad. Sci. USA 120, e2217543120 (2023).

  84. 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).

  85. Fujinuma, S. et al. FOXK1 promotes non-alcoholic fatty liver disease by mediating mTORC1-dependent inhibition of hepatic fatty acid oxidation. Cell Rep. 42, 112530 (2023).

  86. Wang, Y. G. et al. Ferrostatin-1 inhibits toll-like receptor 4/NF-kappaB signaling to alleviate intervertebral disc degeneration in rats. Am. J. Pathol. 193, 430–441 (2023).

    CAS  PubMed  Google Scholar 

  87. Chen, K. N. et al. Ferrostatin-1 obviates seizures and associated cognitive deficits in ferric chloride-induced posttraumatic epilepsy via suppressing ferroptosis. Free Radic. Biol. Med. 179, 109–118 (2022).

    CAS  PubMed  Google Scholar 

  88. Xiao, Z. et al. Ferrostatin-1 alleviates lipopolysaccharide-induced cardiac dysfunction. Bioengineered 12, 9367–9376 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  89. Wang, H. et al. Characterization of ferroptosis in murine models of hemochromatosis. Hepatology 66, 449–465 (2017).

    CAS  PubMed  Google Scholar 

  90. Yu, Y. et al. Hepatic transferrin plays a role in systemic iron homeostasis and liver ferroptosis. Blood 136, 726–739 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  91. Yamada, N. et al. Iron overload as a risk factor for hepatic ischemia-reperfusion injury in liver transplantation: potential role of ferroptosis. Am. J. Transplant. 20, 1606–1618 (2020).

    CAS  PubMed  Google Scholar 

  92. Jiang, H. et al. Ferrostatin-1 ameliorates liver dysfunction via reducing iron in thioacetamide-induced acute liver injury in mice. Front. Pharmacol. 13, 869794 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Liu, C. Y. et al. Ferroptosis is involved in alcohol-induced cell death in vivo and in vitro. Biosci. Biotechnol. Biochem. 84, 1621–1628 (2020).

  94. Liu, B., Yi, W., Mao, X., Yang, L. & Rao, C. Enoyl coenzyme A hydratase 1 alleviates non-alcoholic steatohepatitis in mice by suppressing hepatic ferroptosis. Am. J. Physiol. Endocrinol. Metab. 320, E925–E937 (2021).

  95. Li, S. et al. Obeticholic acid and ferrostatin-1 differentially ameliorate non-alcoholic steatohepatitis in AMLN diet-fed ob/ob mice. Front. Pharmacol. 13, 1081553 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  96. Luo, Y. et al. Protective effects of ferroptosis inhibition on high fat diet-induced liver and renal injury in mice. Int. J. Clin. Exp. Pathol. 13, 2041–2049 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  97. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    CAS  PubMed  Google Scholar 

  98. Liao, Y., Smyth, G. K. & Shi, W. featureCounts: an efficient general purpose program for assigning sequence reads to genomic features. Bioinformatics 30, 923–930 (2014).

    CAS  PubMed  Google Scholar 

  99. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    PubMed  PubMed Central  Google Scholar 

  100. Hanzelmann, S., Castelo, R. & Guinney, J. GSVA: gene set variation analysis for microarray and RNA-seq data. BMC Bioinf. 14, 7 (2013).

    Google Scholar 

  101. Starmann, J. et al. Gene expression profiling unravels cancer-related hepatic molecular signatures in steatohepatitis but not in steatosis. PLoS ONE 7, e46584 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  102. Kozumi, K. et al. Transcriptomics identify thrombospondin-2 as a biomarker for NASH and advanced liver fibrosis. Hepatology 74, 2452–2466 (2021).

    CAS  PubMed  Google Scholar 

  103. Govaere, O. et al. Transcriptomic profiling across the non-alcoholic fatty liver disease spectrum reveals gene signatures for steatohepatitis and fibrosis. Sci. Transl. Med. https://doi.org/10.1126/scitranslmed.aba4448 (2020).

  104. Aibar, S. et al. SCENIC: single-cell regulatory network inference and clustering. Nat. Methods 14, 1083–1086 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. Tabula Muris, C. et al. Single-cell transcriptomics of 20 mouse organs creates a Tabula Muris. Nature 562, 367–372 (2018).

    Google Scholar 

  106. Knutsdottir, H. baderzone/HFpEF_2020: Data update. Zenodo https://doi.org/10.5281/zenodo.4114616 (2020).

Download references

Acknowledgements

This work was supported by the 2021 AASLD Pinnacle Award to K.D. and NIH grants (nos. R01 AA010154, R01 DK077794 and R56 DK134334) and Sponsored Research Study Agreement (no. 337521) supported by Boehringer Ingelheim Pharmaceuticals, awarded to A.M.D. We thank the patients who donated liver tissue for analysis, M. Abdelmalek and C. Guy and the clinical staff and coordinators who created and maintain the Duke NAFLD Biorepository, S. Gregory and his team at Duke Molecular Physiology Institute for snRNA-seq of control and MASLD liver tissues, and S. Pullen and his team at Boehringer Ingelheim Pharmaceuticals and Z. Man (Duke Department of Neurology) for their assistance in the bioinformatics analysis.

Author information

Authors and Affiliations

Authors

Contributions

K.D., L.W. and A.M.D. conceived the experiments. K.D., L.W. and J.H.J. performed the experiments. K.D., L.W., J.H.J., R.K.D., R.M.D., S.H.O., D.C.K. and A.M.D. analyzed the data. K.D., L.W., J.H.J. and A.M.D. wrote the paper. K.D. and A.M.D. secured the funding for the study. All authors reviewed and approved the paper.

Corresponding author

Correspondence to Anna Mae Diehl.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Aging thanks Philippe Lefebvre and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

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

Extended data

Extended Data Fig. 1 Top 10 upregulated/downregulated KEGG pathways in old versus young mouse hepatocytes.

Red arrows point to the upregulated pathways of interest, and blue arrows point to the downregulated pathways of interest. p values in GSEA plot was calculated using permutation test, then adjusted for multiple comparison testing using the Benjamini-Hochberg method.

Extended Data Fig. 2 Identification and characterization of the aging hepatocyte gene signature (AHGS).

(A) Number of DEGs in each gene set after applying different adjusted p-value cutoffs (p = 0.05 or 0.01) and log2 fold changes (1 - 5) for DEGs. (B) Jaccard index, examining similarity of overlapping DEGs of primary hepatocytes from old vs young mice fed with chow diet, and liver tissues from old vs young mice fed with CDA-HFD diet, revealed that AHGS with p-value < 0.01 and Log2 FC > 3 resulted in the best gene set by showing a reduced gene number and higher Jaccard similarity. (C) GO:MF (Molecular Function) clustering of the AHGS. Gene Set Enrichment Analysis (GSEA) of AHGS further identified (D) upregulated GO:BP (GO Biological Process) pathways and (E) upregulated HPO (Human Phenotype Ontology) pathways. p values were calculated using: unpaired two-tailed t.test in A, B; permutation test, then adjusted for multiple comparison testing using the Benjamini-Hochberg method in C, D, E.

Extended Data Fig. 3 Age is a major risk factor of MASLD development.

(A) Re-analysis of the Duke MASLD cohort (GSE213623) revealed that the chronological age positively correlates with MASLD histological markers including hepatocyte ballooning, portal inflammation and fibrosis in MASLD patients (control n = 69; MASLD patients n = 299; ballooning score 0, n = 32, score 1, n = 118, score 2, n = 149; portal_inf score 0, n = 146, score 1, n = 142; fibrosis F0F1, n = 97; F2, n = 107; F3F4, n = 95). **p < 0.01; ***p < 0.001; ****p < 0.0001. (B, C) GSEA using KEGG revealed that genes related to longevity and its associated mechanisms (for example nicotinamide; FoxO signaling) are depleted (B), while genes related to programmed cell death (C) are highly enriched in the transcriptomics of MASLD patients. Red arrows point to the pathways of interest. (D) AHGS enrichment increases with chronological age in MASLD patients but not control subjects. (E) AHGS enrichment score with chronological age in RNA-seq data of 226 normal liver samples from GTEx v8 database (https://gtexportal.org/home/). p-values were calculated using Wilcoxon Rank Sum test. Boxplot shows the upper quantile (75%), median (50%) and lower quantile (25%) of overall data distribution (A, D, E). p values in A, D, E were calculated using Wilcoxon Rank Sum test; p values in GSEA plots in B and C were calculated using permutation test, then adjusted for multiple comparison testing using the Benjamini-Hochberg method.

Extended Data Fig. 4 Aging-associated mechanisms are altered during MASLD development.

GSEA of bulk liver RNA seq data from Duke MASLD patients (GSE213623) revealed that (A) genes related to longevity and its associated mechanisms (for example NAD metabolism, sirtuins) are depleted, (B) while genes related to senescence are highly enriched in the transcriptomics of MASLD patients. (C) AHGS was applied to deconvolute bulk liver RNA seq data (GSE135251, heathy control n = 13; MASH n = 12). AHGS is enriched in transcriptomes of MASH patients and distinguishes MASH patients from healthy controls. Boxplot shows the upper quantile (75%), median (50%) and lower quantile (25%) of overall data distribution. p-values were calculated using: permutation test, then adjusted for multiple comparison testing using the Benjamini-Hochberg method in A, B; Wilcoxon Rank Sum test in C.

Extended Data Fig. 5 snRNA-seq analysis and bulk RNA-seq GSEA of MASLD liver.

(A, C) Uniform manifold approximation and projection (UMAP) visualization of liver cells in single-nucleus RNA-seq dataset generated from two MASLD patients or two healthy controls (GSE174748) (A), or mice fed with CDA-HFD diet or chow diet for 22 weeks (C). (B, D) We focused our analysis on the clusters that are most enriched for hepatocyte markers and depleted for non-hepatocyte markers in both human (B) and mouse dataset (D). GSEA of bulk liver RNA-seq data from Duke MASLD patients (GSE213623) revealed that genes related to (E) apoptosis, (F) pyroptosis and (G) necroptosis are all enriched in the liver transcriptomics of MASLD patients. p values in GSEA plots were calculated using permutation test, then adjusted for multiple comparison testing using the Benjamini-Hochberg method.

Extended Data Fig. 6 Livers of MASLD patients exhibit ferroptotic stress.

(A) Duke MASLD cohort patients (GSE213623) with the same fibrosis stage were stratified into young (age ≤ 35), middle age (35 <age < 55) and old groups (age ≥ 55). Expression of ferroptotic-associated genes was compared among the different age groups (age 20-35, fibrosis F0F1, n = 2; F2, n = 9; F3F4, n = 4; age 36-55, fibrosis F0F1, n = 55; F2, n = 61; F3F4, n = 39; age 56-80, fibrosis F0F1, n = 19; F2, n = 37; F3F4, n = 52). Box plot showed the upper quantile (75%), median (50%) and lower quantile (25%) (A) of overall data distribution in MASLD patients with different fibrosis stages. (B) Expression of genes related to iron homeostasis and (C) ferroptotic stress was compared between MASLD patients versus controls (healthy n = 2; MASLD n = 2). The average expression of in each single cell was calculated by the AddModuleScore function of Seurat. Feature plot showed single cells on dimensional reduction plot, with cell color indicating their relative expression levels. Violin plot showed the maxima, upper quantile (75%), median (50%), lower quantile (25%) and minima of overall data distribution in the healthy and MASLD group. p-values were calculated using: Wilcoxon Rank Sum test in A; unpaired two-tailed t.test in B, C.

Extended Data Fig. 7 Ferroptotic stress, lipotoxicity, inflammation and collagen metabolism are increased in livers of old mice.

(A) MDA levels were measured by western blots in livers and primary hepatocytes from chow-fed young (3 months old, n = 4) and old (2 years old, n = 5 for liver lysates, n = 4 for hepatocyte lysates). Data are graphed as mean ± SEM. (B) A published dataset (GSE132042) was re-analyzed to compare the transcriptomes of very old mouse liver ( ≥ 24 months old, n = 7) to young mouse liver ( ≤ 3 months old, n = 12). AHGS was enriched in the transcriptome of liver of aged mice. Boxplot shows the upper quantile (75%), median (50%) and lower quantile (25%) of overall data distribution. GSEA analysis further revealed that the transcriptomes of aged mouse liver are enriched with genes associated with (C) hepatocyte aging, (D) lipid metabolism, (E) inflammation and (F) collagen activity. p-values were calculated using: unpaired two-tailed t.test in A; Wilcoxon Rank Sum test in B; permutation test, then adjusted for multiple comparison testing using the Benjamini-Hochberg method in C, D, E, F.

Source data

Extended Data Fig. 8 Ferrostatin-1 protects hepatocytes from aging-related ferroptotic stress and senescence during diet-induced MASH.

Young and old mice were fed with a chow diet or CDA-HFD diet for 6 weeks. Old mice were intraperitoneally injected with Fer1 or its vehicle every other day during the last 8 days of feeding. (A) Body weights and liver weights were measured on the day of sacrifice (young+veh, n = 9; old+veh, n = 6; old+Fer-1, n = 6). (B) Expression of ferroptosis-related proteins detected by western blotting. (C) Representative Tfrc staining and quantification of the positively-stained areas (young+veh, n = 9; old+veh, n = 6; old+Fer-1, n = 6). Scale bars = 100μm (D) Expression of senescent marker p21 detected by western blotting. (E) GSEA demonstrated that transcriptome of old mice treated with Fer1 is depleted with genes involved in response to oxidative stress and related mortality. Protein expression was quantified by densitomeric analyses of western blots. Data are graphed as mean ± SEM. #p < 0.05 versus young mice + veh; $p < 0.05 versus old mice + veh. p-values were calcualted using: one-way ANOVA in A, B, C, D; permutation test, then adjusted for multiple comparison testing using the Benjamini-Hochberg method in E.

Source data

Extended Data Fig. 9 GSEA of the DEGs that reversed by ferrostatin1 in aged mice.

(A) Venn diagram identified 520 DEGs that were upregulated in old mice (vs young mice + veh) but reversed by Fer1 (vs old mice + veh). GSEA further identified 14 significantly enriched hallmark pathways in these DEGs. (B) Venn diagram identified 658 DEGs that were downregulated in old mice (vs young mice + veh) but reversed by Fer1 (vs old mice + veh). GSEA further identified 7 significantly enriched hallmark pathways in these DEGs. p values were calculated using permutation test, then adjusted for multiple comparison testing using the Benjamini-Hochberg method.

Extended Data Fig. 10 FXR and ferroptotic stress interact to modulate age-dependent susceptibility to MASLD.

Young and old mice were fed with CDA-HFD diet for 6 weeks. These mice were intraperitoneally injected with Fer1 or its vehicle every other day during the last 8 days of feeding. Transcriptomes of liver tissues were analysed by RNA-seq (n = 4 mice/group). (A) GSEA demonstrated that liver transcriptome of old mice is depleted of genes involved in bile acid metabolism and this is reversed by Fer1 treatment in the old mice. (B) Expression of FXR/Nr1h4. (C) GSEA demonstrated that liver transcriptome of old mice is depleted of genes involved in FXR pathway activity, and this is reversed by Fer1 treatment in the old mcie. (D) Expression of FXR/Nr1h4 in liver of Duke NAFLD cohorts (GSE213623). (E) Expression of FXR/Nr1h4 in hepatocytes subpopulations from single-nucleus RNA-seq dataset of two MASLD patients or two healthy controls (GSE174748). The average expression of in each single cell was calculated by the AddModuleScore function of Seurat. Feature plot showed single cells on dimensional reduction plot, with cell color indicating their relative expression levels. Box plot showed the upper quantile (75%), median (50%) and lower quantile (25%) (B, D), while violin plot showed the maxima, upper quantile (75%), median (50%), lower quantile (25%) and minima (E) of overall data distribution. p-values were calcualted using: permutation test, then adjusted for multiple comparison testing using the Benjamini-Hochberg method in A, C; Wilcoxon Rank Sum test in B, D; unpaired two-tailed t.test in E.

Supplementary information

Supplementary Information

Supplementary Tables 2–5.

Reporting Summary

Supplementary Table 1

AHGS Aging_geneSig_l2fc.

Source data

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 4

Unprocessed western blots.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Fig. 7

Unprocessed western blots.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 7

Unprocessed western blots.

Source Data Extended Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 8

Unprocessed western blots.

Rights and permissions

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

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Du, K., Wang, L., Jun, J.H. et al. Aging promotes metabolic dysfunction-associated steatotic liver disease by inducing ferroptotic stress. Nat Aging (2024). https://doi.org/10.1038/s43587-024-00652-w

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s43587-024-00652-w

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