Discover the SciOpen Platform and Achieve Your Research Goals with Ease.
Search articles, authors, keywords, DOl and etc.
Aging is a contributor to liver disease. Hence, the concept of liver aging has become prominent and has attracted considerable interest, but its underlying mechanism remains poorly understood. In our study, the internal mechanism of liver aging was explored via multi-omics analysis and molecular experiments to support future targeted therapy. An aged rat liver model was established with d-galactose, and two other senescent hepatocyte models were established by treating HepG2 cells with d-galactose and H2O2. We then performed transcriptomic and metabolomic assays of the aged liver model and transcriptome analyses of the senescent hepatocyte models. In livers, genes related to peroxisomes, fatty acid elongation, and fatty acid degradation exhibited down-regulated expression with aging, and the hepatokine Fgf21 expression was positively correlated with the down-regulation of these genes. In senescent hepatocytes, similar to the results found in aged livers, FGF21 expression was also decreased. Moreover, the expressions of cell cycle-related genes were significantly down-regulated, and the down-regulated gene E2F8 was the key cell cycle-regulating transcription factor. We then validated that FGF21 overexpression can protect against liver aging and that FGF21 can attenuate the declines in the antioxidant and regenerative capacities in the aging liver. We successfully validated the results from cellular and animal experiments using human liver and blood samples. Our study indicated that FGF21 is an important target for inhibiting liver aging and suggested that pharmacological prevention of the reduction in FGF21 expression due to aging may be used to treat liver aging-related diseases.
Ramirez T, Li YM, Yin S, et al. Aging aggravates alcoholic liver injury and fibrosis in mice by downregulating sirtuin 1 expression. J Hepatol. 2017;66(3):601-609.
Aravinthan AD, Alexander GJM. Senescence in chronic liver disease: is the future in aging? J Hepatol. 2016;65(4):825-834.
Goikoetxea-Usandizaga N, Serrano-Maciá M, Delgado TC, et al. Mitochondrial bioenergetics boost macrophage activation, promoting liver regeneration in metabolically compromised animals. Hepatology. 2022;75(3):550-566.
Liu Z, Wang W, Zhuang L, et al. Clear mortality gap caused by graft macrosteatosis in Chinese patients after cadaveric liver transplantation. Hepatobiliary Surg Nutr. 2020;9(6):739-758.
Wang W, Liu Z, Qian J, et al. Systematic assessment of safety threshold for donor age in cadaveric liver transplantation. Front Med. 2021;8:596552.
Richardson MM, Jonsson JR, Powell EE, et al. Progressive fibrosis in nonalcoholic steatohepatitis: association with altered regeneration and a ductular reaction. Gastroenterology. 2007;133(1):80-90.
Nakajima T, Moriguchi M, Katagishi T, et al. Premature telomere shortening and impaired regenerative response in hepatocytes of individuals with NAFLD. Liver Int. 2006;26(1):23-31.
Aravinthan A, Scarpini C, Tachtatzis P, et al. Hepatocyte senescence predicts progression in non-alcohol-related fatty liver disease. J Hepatol. 2013;58(3):549-556.
McCabe LR, Irwin R, Tekalur A, et al. Exercise prevents high fat diet-induced bone loss, marrow adiposity and dysbiosis in male mice. Bone. 2019;118:20-31.
Byun S, Seok S, Kim YC, et al. Fasting-induced FGF21 signaling activates hepatic autophagy and lipid degradation via JMJD3 histone demethylase. Nat Commun. 2020;11(1):807.
Hill CM, Albarado DC, Coco LG, et al. FGF21 is required for protein restriction to extend lifespan and improve metabolic health in male mice. Nat Commun. 2022;13(1):1897.
Gao Y, Zhang W, Zeng LQ, et al. Exercise and dietary intervention ameliorate high-fat diet-induced NAFLD and liver aging by inducing lipophagy. Redox Biol. 2020;36:101635.
Wu Y, Cao H, Baranova A, et al. Correction: multi-trait analysis for genome-wide association study of five psychiatric disorders. Transl Psychiatry. 2020;10:234.
Yu H, Pan R, Qi Y, et al. LEPR hypomethylation is significantly associated with gastric cancer in males. Exp Mol Pathol. 2020;116:104493.
Li H, Wang X, Lu X, et al. Co-expression network analysis identified hub genes critical to triglyceride and free fatty acid metabolism as key regulators of age-related vascular dysfunction in mice. Aging (Albany NY). 2019;11(18):7620-7638.
Azman KF, Safdar A, Zakaria R. D-galactose-induced liver aging model: its underlying mechanisms and potential therapeutic interventions. Exp Gerontol. 2021;150:111372.
Du Z, Yang Y, Hu Y, et al. A long-term high-fat diet increases oxidative stress, mitochondrial damage and apoptosis in the inner ear of D-galactose-induced aging rats. Hear Res. 2012;287(1–2):15-24.
Saleh DO, Mansour DF, Hashad IM, Bakeer RM. Effects of sulforaphane on D-galactose-induced liver aging in rats: role of keap-1/nrf-2 pathway. Eur J Pharmacol. 2019;855:40-49.
Oyebode OT, Giwa OD, Olorunsogo OO. Comparative effects of galactose-induced aging on mitochondrial permeability transition in rat liver and testis. Toxicol Mech Methods. 2020;30(5):388-396.
Hakimizadeh E, Hassanshahi J, Kaeidi A, et al. Ceftriaxone improves hepatorenal damages in mice subjected to D-galactose-induced aging. Life Sci. 2020;258:118119.
Xu Y, Li Y, Ma L, et al. D-galactose induces premature senescence of lens epithelial cells by disturbing autophagy flux and mitochondrial functions. Toxicol Lett. 2018;289:99-106.
Shen Y, Gao H, Shi X, et al. Glutamine synthetase plays a role in D-galactose-induced astrocyte aging in vitro and in vivo. Exp Gerontol. 2014;58:166-173.
Xu X, Shen X, Feng W, et al. D-galactose induces senescence of glioblastoma cells through YAP-CDK6 pathway. Aging (Albany NY). 2020;12(18):18501-18521.
Seo E, Kang H, Choi H, Choi W, Jun HS. Reactive oxygen species-induced changes in glucose and lipid metabolism contribute to the accumulation of cholesterol in the liver during aging. Aging Cell. 2019;18(2):e12895.
He ZH, Li M, Fang QJ, et al. FOXG1 promotes aging inner ear hair cell survival through activation of the autophagy pathway. Autophagy. 2021;17(12):4341-4362.
Facchin F, Bianconi E, Romano M, et al. Comparison of oxidative stress effects on senescence patterning of human adult and perinatal tissue-derived stem cells in short and long-term cultures. Int J Med Sci. 2018;15(13):1486-1501.
Xu X, Hueckstaedt LK, Ren J. Deficiency of insulin-like growth factor 1 attenuates aging-induced changes in hepatic function: role of autophagy. J Hepatol. 2013;59(2):308-317.
Sen B, Rastogi A, Nath R, et al. Senescent hepatocytes in decompensated liver show reduced UPRMT and its key player, CLPP, attenuates senescence in vitro. Cell Mol Gastroenterol Hepatol. 2019;8(1):73-94.
Seo E, Nam H, Jun HS. Reactive oxygen species induce HNF-4α expression via the ASK1-CREB pathway, promoting ChREBP expression and lipogenesis in hepatocytes. Life Sci. 2022(310):121042.
Chung KW, Lee EK, Kim DH, et al. Age-related sensitivity to endotoxin-induced liver inflammation: implication of inflammasome/IL-1β for steatohepatitis. Aging Cell. 2015;14(4):524-533.
Lee AK, Kim DH, Bang E, Choi YJ, Chung HY. β-hydroxybutyrate suppresses lipid accumulation in aged liver through GPR109A-mediated signaling. Aging Dis. 2020;11(4):777-790.
Liu Z, Zhang C, Lee S, et al. Pyruvate kinase L/R is a regulator of lipid metabolism and mitochondrial function. Metab Eng. 2019;52:263-272.
Zhao H, Huang H, Alam A, et al. VEGF mitigates histone-induced pyroptosis in the remote liver injury associated with renal allograft ischemia-reperfusion injury in rats. Am J Transplant. 2018;18(8):1890-1903.
Schmittgen TD, Livak KJ. Analyzing real-time PCR data by the comparative C(T) method. Nat Protoc. 2008;3(6):1101-1108.
Robinson AD, Eich ML, Varambally S. Dysregulation of de novo nucleotide biosynthetic pathway enzymes in cancer and targeting opportunities. Cancer Lett. 2020;470:134-140.
Luo W, Pant G, Bhavnasi YK,, Brouwer C. Pathview Web: user friendly pathway visualization and data integration. Nucleic Acids Res. 2017;45(W1):W501-W508.
Shannon P, Markiel A, Ozier O, et al. Cytoscape: a software environment for integrated models of biomolecular interaction networks. Genome Res. 2003;13(11):2498-2504.
Azman KF, Zakaria R. D-Galactose-induced accelerated aging model: an overview. Biogerontology. 2019;20(6):763-782.
Weir HJ, Yao P, Huynh FK, et al. Dietary restriction and AMPK increase lifespan via mitochondrial network and peroxisome remodeling. Cell Metabol. 2017;26(6):884-896.e5.
Pascual-Ahuir A, Manzanares-Estreder S, Proft M. Pro- and antioxidant functions of the peroxisome-mitochondria connection and its impact on aging and disease. Oxid Med Cell Longev. 2017;2017:9860841.
Wei X, Guo J, Li Q, et al. Bach1 regulates self-renewal and impedes mesendodermal differentiation of human embryonic stem cells. Sci Adv. 2019;5(3):eaau7887.
Tian M, Huang Y, Song Y, et al. MYSM1 suppresses cellular senescence and the aging process to prolong lifespan. Adv Sci. 2020;7(22):2001950.
Nakagawa Y, Araki M, Han SI, Mizunoe Y, Shimano H. CREBH systemically regulates lipid metabolism by modulating and integrating cellular functions. Nutrients. 2021;13(9):3204.
Durand F, Levitsky J, Cauchy F, Gilgenkrantz H, Soubrane O, Francoz C. Age and liver transplantation. J Hepatol. 2019;70(4):745-758.
Timchenko NA. Aging and liver regeneration. Trends Endocrinol Metabol. 2009;20(4):171-176.
Tanaka H, Igata T, Etoh K, Koga T, Takebayashi SI, Nakao M. The NSD2/WHSC1/MMSET methyltransferase prevents cellular senescence-associated epigenomic remodeling. Aging Cell. 2020;19(7):e13173.
Macedo JC, Vaz S, Bakker B, et al. FoxM1 repression during human aging leads to mitotic decline and aneuploidy-driven full senescence. Nat Commun. 2018;9(1):2834.
Houben P, Döhler B, Weiß KH, Mieth M, Mehrabi A, Süsal C. Differential influence of donor age depending on the indication for liver transplantation - a collaborative transplant study report. Transplantation. 2020;104(4):779-787.
Di Micco R, Krizhanovsky V, Baker D, d'Adda di Fagagna F. Cellular senescence in ageing: from mechanisms to therapeutic opportunities. Nat Rev Mol Cell Biol. 2021;22(2):75-95.
Fisher FM, Maratos-Flier E. Understanding the physiology of FGF21. Annu Rev Physiol. 2016;78:223-241.
Wernicke C, Pohrt A, Pletsch-Borba L, et al. Effect of unsaturated fat and protein intake on liver fat in people at risk of unhealthy aging: 1-year results of a randomized controlled trial. Am J Clin Nutr. 2023;117(4):785-793.
Han Q, Li H, Jia M, et al. Age-related changes in metabolites in young donor livers and old recipient sera after liver transplantation from young to old rats. Aging Cell. 2021;20(7):e13425.
Katsumura S, Siddiqui N, Goldsmith MR, et al. Deadenylase-dependent mRNA decay of GDF15 and FGF21 orchestrates food intake and energy expenditure. Cell Metabol. 2022;34(4):564-580.e8.
Schmucker DL, Sanchez H. Liver regeneration and aging: a current perspective. Curr Gerontol Geriatr Res. 2011;2011:526379.
Dimri GP, Lee X, Basile G, et al. A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc Natl Acad Sci U S A. 1995;92(20):9363-9367.
He S, Sharpless NE. Senescence in health and disease. Cell. 2017;169(6):1000-1011.
Gonzales E, Julien B, Serrière-Lanneau V, et al. ATP release after partial hepatectomy regulates liver regeneration in the rat. J Hepatol. 2010;52(1):54-62.
Vajdová K, Graf R, Clavien PA. ATP-supplies in the cold-preserved liver: a long-neglected factor of organ viability. Hepatology. 2002;36(6):1543-1552.
Beauséjour CM, Krtolica A, Galimi F, et al. Reversal of human cellular senescence: roles of the p53 and p16 pathways. EMBO J. 2003;22(16):4212-4222.
Dulić V, Beney GE, Frebourg G, Drullinger LF, Stein GH. Uncoupling between phenotypic senescence and cell cycle arrest in aging p21-deficient fibroblasts. Mol Cell Biol. 2000;20(18):6741-6754.
Campisi J. Replicative senescence: an old lives' tale? Cell. 1996;84(4):497-500.
Coppé JP, Rodier F, Patil CK, Freund A, Desprez PY, Campisi J. Tumor suppressor and aging biomarker p16 (INK4a) induces cellular senescence without the associated inflammatory secretory phenotype. J Biol Chem. 2011;286(42):36396-36403.
Narita M, Nũnez S, Heard E, et al. Rb-mediated heterochromatin formation and silencing of E2F target genes during cellular senescence. Cell. 2003;113(6):703-716.
Aravinthan A. Cellular senescence: a hitchhiker's guide. Hum Cell. 2015;28(2):51-64.
Priyamvada S, Priyadarshini M, Arivarasu NA, et al. Studies on the protective effect of dietary fish oil on gentamicin-induced nephrotoxicity and oxidative damage in rat kidney. Prostaglandins Leukot Essent Fatty Acids. 2008;78(6):369-381.
Ghaznavi H, Fatemi I, Kalantari H, et al. Ameliorative effects of Gallic acid on gentamicin-induced nephrotoxicity in rats. J Asian Nat Prod Res. 2018;20(12):1182-1193.
Kliewer SA, Mangelsdorf DJ. A dozen years of discovery: insights into the physiology and pharmacology of FGF21. Cell Metabol. 2019;29(2):246-253.
Jensen-Cody SO, Potthoff MJ. Hepatokines and metabolism: deciphering communication from the liver. Mol Metabol. 2021;44:101138.
Stefan N, Schick F, Birkenfeld AL, Häring HU, White MF. The role of hepatokines in NAFLD. Cell Metabol. 2023;35(2):236-252.
Qu X, Wen Y, Jiao J, et al. PARK7 deficiency inhibits fatty acid β-oxidation via PTEN to delay liver regeneration after hepatectomy. Clin Transl Med. 2022;12(9):e1061.
Rudnick DA, Davidson NO. Functional relationships between lipid metabolism and liver regeneration. Int J Hepatol. 2012;2012:549241.
Gazit V, Weymann A, Hartman E, et al. Liver regeneration is impaired in lipodystrophic fatty liver dystrophy mice. Hepatology. 2010;52(6):2109-2117.
This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).