694
Views
21
CrossRef citations to date
0
Altmetric
Review

NAFLD in the Elderly

ORCID Icon & ORCID Icon
Pages 1633-1649 | Published online: 13 Sep 2021

References

  • FrithJ, DayCP, HendersonE, BurtAD, NewtonJL. Non-alcoholic fatty liver disease in older people. Gerontology. 2009;55(6):607–613. doi:10.1159/00023567719690397
  • MundayD, LeamanJ, O’MooreÉ, PluggeE. The prevalence of non-communicable disease in older people in prison: a systematic review and meta-analysis. Age Ageing. 2019;48(2):204–212. doi:10.1093/ageing/afy18630590404
  • López-OtínC, BlascoMA, PartridgeL, SerranoM, KroemerG. The Hallmarks of aging. Cell. 2013;153(6):1194–1217.23746838
  • AikataH, TakaishiH, KawakamiY, et al. Telomere reduction in human liver tissues with age and chronic inflammation. Exp Cell Res. 2000;256(2):578–582. doi:10.1006/excr.2000.486210772830
  • SinghR, KaushikS, WangY, et al. Autophagy regulates lipid metabolism. Nature. 2009;458(7242):1131–1135. doi:10.1038/nature0797619339967
  • ZhangC, CuervoAM. Restoration of chaperone-mediated autophagy in aging liver improves cellular maintenance and hepatic function. Nat Med. 2008;14(9):959–965. doi:10.1038/nm.185118690243
  • SchneiderJL, VillarroyaJ, Diaz‐CarreteroA, et al. Loss of hepatic chaperone-mediated autophagy accelerates proteostasis failure in aging. Aging Cell. 2015;14(2):249–264. doi:10.1111/acel.1231025620427
  • PassosJF, SaretzkiG, von ZglinickiT. DNA damage in telomeres and mitochondria during cellular senescence: is there a connection?Nucleic Acids Res. 2007;35(22):7505–7513. doi:10.1093/nar/gkm89317986462
  • VictorelliS, PassosJF. Telomeres and Cell senescence - size matters not. EBioMedicine. 2017;21:14–20. doi:10.1016/j.ebiom.2017.03.02728347656
  • CampisiJ, d’Adda Di FagagnaF. Cellular senescence: when bad things happen to good cells. Nat Rev Mol Cell Biol. 2007;8(9):729–740. doi:10.1038/nrm223317667954
  • RudolphKL, ChangS, MillardM, Schreiber-AgusN, DePinhoRA. Inhibition of experimental liver cirrhosis in mice by telomerase gene delivery. Science. 2000;287(5456):1253–1258. doi:10.1126/science.287.5456.125310678830
  • AravinthanA, MellsG, AllisonM, et al. Gene polymorphisms of cellular senescence marker p21 and disease progression in non-alcohol-related fatty liver disease. Cell Cycle. 2014;13(9):1489–1494. doi:10.4161/cc.2847124626178
  • AravinthanA, ScarpiniC, TachtatzisP, et al. Hepatocyte senescence predicts progression in non-alcohol-related fatty liver disease. J Hepatol. 2013;58(3):549–556. doi:10.1016/j.jhep.2012.10.03123142622
  • NakajimaT, NakashimaT, OkadaY, et al. Nuclear size measurement is a simple method for the assessment of hepatocellular aging in non-alcoholic fatty liver disease: comparison with telomere-specific quantitative FISH and p21 immunohistochemistry. Pathol Int. 2010;60(3):175–183. doi:10.1111/j.1440-1827.2009.02504.x20403043
  • VermaS, TachtatzisP, Penrhyn‐LoweS, et al. Sustained telomere length in hepatocytes and cholangiocytes with increasing age in normal liver. Hepatology. 2012;56(4):1510–1520. doi:10.1002/hep.2578722504828
  • WangM-J, ChenF, LiJ-X, et al. Reversal of hepatocyte senescence after continuous in vivo cell proliferation. Hepatology. 2014;60(1):349–361. doi:10.1002/hep.2709424711261
  • OgrodnikM, MiwaS, TchkoniaT, et al. Cellular senescence drives age-dependent hepatic steatosis. Nat Commun. 2017;8(1):15691.28608850
  • CavalliG, HeardE. Advances in epigenetics link genetics to the environment and disease. Nature. 2019;571(7766):489–499. doi:10.1038/s41586-019-1411-031341302
  • PogribnyIP, TryndyakVP, BagnyukovaTV, et al. Hepatic epigenetic phenotype predetermines individual susceptibility to hepatic steatosis in mice fed a lipogenic methyl-deficient diet. J Hepatol. 2009;51(1):176–186. doi:10.1016/j.jhep.2009.03.02119450891
  • ShockLS, ThakkarPV, PetersonEJ, MoranRG, TaylorSM. DNA methyltransferase 1, cytosine methylation, and cytosine hydroxymethylation in mammalian mitochondria. Proce Nat Acad Sci. 2011;108(9):3630–3635. doi:10.1073/pnas.1012311108
  • PirolaCJ, GianottiTF, BurgueñoAL, et al. Epigenetic modification of liver mitochondrial DNA is associated with histological severity of nonalcoholic fatty liver disease. Gut. 2013;62(9):1356–1363. doi:10.1136/gutjnl-2012-30296222879518
  • BacaliniMG, FranceschiC, GentiliniD, et al. Molecular Aging of human liver: an epigenetic/transcriptomic signature. J Gerontolo. 2019;74(1):1–8.
  • Muller‐HockerJ, AustD, RohrbachH, et al. Defects of the respiratory chain in the normal human liver and in cirrhosis during aging. Hepatology. 1997;26(3):709–719. doi:10.1002/hep.5102603249303502
  • LazarouM, SmithSM, ThorburnDR, RyanMT, McKenzieM. Assembly of nuclear DNA-encoded subunits into mitochondrial complex IV, and their preferential integration into supercomplex forms in patient mitochondria. FEBS J. 2009;276(22):6701–6713. doi:10.1111/j.1742-4658.2009.07384.x19843159
  • BellantiF, RomanoAD, GiudettiAM, et al. Many faces of mitochondrial uncoupling during age: damage or defense?J Gerontolo. 2013;68(8):892–902. doi:10.1093/gerona/gls332
  • ModiHR, KatyareSS, PatelMA. Ageing-induced alterations in lipid/phospholipid profiles of rat brain and liver mitochondria: implications for mitochondrial energy-linked functions. J Membrane Biol. 2008;221(1):51–60. doi:10.1007/s00232-007-9086-018097631
  • SastreJ, PallardóFV, PláR, et al. Aging of the liver: age-associated mitochondrial damage in intact hepatocytes. Hepatology. 1996;24(5):1199–1205. doi:10.1002/hep.5102405368903398
  • WuJ-L, WuQ-P, PengY-P, ZhangJ-M. Effects of L-malate on mitochondrial oxidoreductases in liver of aged rats. Physiolo Res. 2011;329–336. doi:10.33549/physiolres.931986
  • ZorovDB, JuhaszovaM, SollottSJ. Mitochondrial Reactive Oxygen Species (ROS) and ROS-Induced ROS Release. Physiol Rev. 2014;94(3):909–950. doi:10.1152/physrev.00026.201324987008
  • ColavittiR, FinkelT. Reactive oxygen species as mediators of cellular senescence. IUBMB Life. 2005;57(4–5):277–281. doi:10.1080/1521654050009189016036611
  • Lozada-DelgadoJG, Torres-RamosCA, Ayala-PeñaS. Chapter 4 - Aging, oxidative stress, mitochondrial dysfunction, and the liver. In: PreedyVR, PatelVB, editors. Aging (Second Edition). Academic Press; 2020:37–46.
  • Pérez-CarrerasM, HoyoPD, MartínMA, et al. Defective hepatic mitochondrial respiratory chain in patients with nonalcoholic steatohepatitis. Hepatology. 2003;38(4):999–1007. doi:10.1002/hep.184038042614512887
  • TariqZ, GreenCJ, HodsonL. Are oxidative stress mechanisms the common denominator in the progression from hepatic steatosis towards non-alcoholic steatohepatitis (NASH)?Liver Int. 2014;34(7):e180–e190. doi:10.1111/liv.1252324621397
  • PolyzosSA, KountourasJ, MantzorosCS. Adipose tissue, obesity and non-alcoholic fatty liver disease. Minerva Endocrinol. 2017;42(2):92–108.27711029
  • VosB, MorenoC, NagyN, et al. Lean non-alcoholic fatty liver disease (Lean-NAFLD): a major cause of cryptogenic liver disease. Acta Gastroenterol Belg. 2011;74(3):389–394.22103042
  • EguchiY, HyogoH, OnoM, et al. Prevalence and associated metabolic factors of nonalcoholic fatty liver disease in the general population from 2009 to 2010 in Japan: a multicenter large retrospective study. J Gastroenterol. 2012;47(5):586–595. doi:10.1007/s00535-012-0533-z22328022
  • GolabiP, PaikJ, ReddyR, BugianesiE, TrimbleG, YounossiZM. Prevalence and long-term outcomes of non-alcoholic fatty liver disease among elderly individuals from the United States. BMC Gastroenterol. 2019;19(1):56. doi:10.1186/s12876-019-0972-630991959
  • YounossiZM, KoenigAB, AbdelatifD, FazelY, HenryL, WymerM. Global epidemiology of nonalcoholic fatty liver disease—Meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology. 2016;64(1):73–84. doi:10.1002/hep.2843126707365
  • SinghS, AllenAM, WangZ, ProkopLJ, MuradMH, LoombaR. Fibrosis Progression in Nonalcoholic Fatty Liver vs Nonalcoholic Steatohepatitis: a systematic review and meta-analysis of paired-biopsy studies. Clin Gastroenterol Hepatol. 2015;13(4):643–654.e649. doi:10.1016/j.cgh.2014.04.01424768810
  • LoombaR, WongR, FraysseJ, et al. Nonalcoholic fatty liver disease progression rates to cirrhosis and progression of cirrhosis to decompensation and mortality: a real world analysis of Medicare data. Aliment Pharmacol Ther. 2020;51(11):1149–1159. doi:10.1111/apt.1567932372515
  • DulaiPS, SinghS, PatelJ, et al. Increased risk of mortality by fibrosis stage in nonalcoholic fatty liver disease: systematic review and meta-analysis. Hepatology. 2017;65(5):1557–1565. doi:10.1002/hep.2908528130788
  • AnguloP, KleinerDE, Dam-LarsenS, et al. Liver Fibrosis, but no other histologic features, is associated with long-term outcomes of patients with nonalcoholic fatty liver disease. Gastroenterology. 2015;149(2):389–397.e310. doi:10.1053/j.gastro.2015.04.04325935633
  • EkstedtM, HagströmH, NasrP, et al. Fibrosis stage is the strongest predictor for disease-specific mortality in NAFLD after up to 33 years of follow-up. Hepatology. 2015;61(5):1547–1554. doi:10.1002/hep.2736825125077
  • YounossiZM, StepanovaM, RafiqN, et al. Pathologic criteria for nonalcoholic steatohepatitis: interprotocol agreement and ability to predict liver-related mortality. Hepatology. 2011;53(6):1874–1882. doi:10.1002/hep.2426821360720
  • KaganskyN, LevyS, KeterD, et al. Non-alcoholic fatty liver disease – a common and benign finding in octogenarian patients. Liver Int. 2004;24(6):588–594.15566509
  • MiyaakiH, IchikawaT, NakaoK, et al. Clinicopathological study of nonalcoholic fatty liver disease in Japan: the risk factors for fibrosis. Liver Int. 2008;28(4):519–524. doi:10.1111/j.1478-3231.2007.01614.x17976158
  • LonardoA, LombardiniS, ScaglioniF, et al. Fatty liver, carotid disease and gallstones: a study of age-related associations. World J Gastroenterol. 2006;12(36):5826–5833. doi:10.3748/wjg.v12.i36.582617007049
  • AllenAM, TherneauTM, LarsonJJ, CowardA, SomersVK, KamathPS. Nonalcoholic fatty liver disease incidence and impact on metabolic burden and death: a 20 year-community study. Hepatology. 2018;67(5):1726–1736. doi:10.1002/hep.2954628941364
  • LabenzC, HuberY, KalligaE, et al. Predictors of advanced fibrosis in non-cirrhotic non-alcoholic fatty liver disease in Germany. Aliment Pharmacol Ther. 2018;48(10):1109–1116. doi:10.1111/apt.1497630288767
  • KapsL, LabenzC, GallePR, Weinmann-MenkeJ, KostevK, SchattenbergJM. Non-alcoholic fatty liver disease increases the risk of incident chronic kidney disease. United Eur Gastroenterol J. 2020;8(8):942–948. doi:10.1177/2050640620944098
  • KimG-A, LeeHC, ChoeJ, et al. Association between non-alcoholic fatty liver disease and cancer incidence rate. J Hepatol. 2018;68(1):140–146. doi:10.1016/j.jhep.2017.09.012
  • WeinmannA, AltY, KochS, et al. Treatment and survival of non-alcoholic steatohepatitis associated hepatocellular carcinoma. BMC Cancer. 2015;15:210. doi:10.1186/s12885-015-1197-x25884354
  • KodamaK, TokushigeK, HashimotoE, TaniaiM, ShiratoriK. Hepatic and extrahepatic malignancies in cirrhosis caused by nonalcoholic steatohepatitis and alcoholic liver disease. Alcohol Clin Exp Res. 2013;37(s1):E247–E252. doi:10.1111/j.1530-0277.2012.01900.x23320802
  • BrunotA, SourdSL, PrachtM, EdelineJ. Hepatocellular carcinoma in elderly patients: challenges and solutions. J Hepatocellular Carcinoma. 2016;3:9–18. doi:10.2147/JHC.S10144827574587
  • NishikawaH, KimuraT, KitaR, OsakiY. Treatment for Hepatocellular Carcinoma in Elderly Patients: a Literature Review. J Cancer. 2013;4(8):635–643. doi:10.7150/jca.727924155775
  • TrevisaniF, CantariniMC, LabateAMM, et al. Surveillance for hepatocellular carcinoma in elderly Italian patients with cirrhosis: effects on cancer staging and patient survival. Am J Gastroenterol. 2004;99(8):1470–1476. doi:10.1111/j.1572-0241.2004.30137.x15307862
  • MarreroJA, KulikLM, SirlinCB, et al. Diagnosis, Staging, and Management of Hepatocellular Carcinoma: 2018 Practice Guidance by the American Association for the Study of Liver Diseases. Hepatology. 2018;68(2):723–750. doi:10.1002/hep.2991329624699
  • HungG-Y, HorngJ-L, YenH-J, LeeC-Y, LinL-Y. Changing incidence patterns of hepatocellular carcinoma among age groups in Taiwan. J Hepatol. 2015;63(6):1390–1396. doi:10.1016/j.jhep.2015.07.03226256438
  • WhiteDL, ThriftAP, KanwalF, DavilaJ, El-SeragHB. Incidence of Hepatocellular Carcinoma in All 50 United States, From 2000 Through 2012. Gastroenterology. 2017;152(4):812–820.e815. doi:10.1053/j.gastro.2016.11.02027889576
  • SinghAK, KumarR, PandeyAK. Hepatocellular Carcinoma: causes, Mechanism of Progression and Biomarkers. Curr Chem Genomics Translational Med. 2018;12:1. doi:10.2174/2213988501812010009
  • YounossiZM, GolabiP, de AvilaL, et al. The global epidemiology of NAFLD and NASH in patients with type 2 diabetes: a systematic review and meta-analysis. J Hepatol. 2019;71(4):793–801. doi:10.1016/j.jhep.2019.06.02131279902
  • MantovaniA, ByrneCD, BonoraE, TargherG. Nonalcoholic Fatty Liver Disease and Risk of Incident Type 2 Diabetes: a Meta-analysis. Diabetes Care. 2018;41(2):372–382. doi:10.2337/dc17-190229358469
  • MantovaniA, PetraccaG, BeatriceG. et al. Non-alcoholic fatty liver disease and risk of incident chronic kidney disease: an updated meta-analysis. Gut;2020. gutjnl-2020-323082. doi:10.1136/gutjnl-2020-323082
  • LiY, WangJ, TangY, et al. Bidirectional association between nonalcoholic fatty liver disease and type 2 diabetes in Chinese population: evidence from the Dongfeng-Tongji cohort study. PLoS One. 2017;12(3):e0174291. doi:10.1371/journal.pone.017429128350839
  • EkstedtM, FranzénLE, MathiesenUL, et al. Long-term follow-up of patients with NAFLD and elevated liver enzymes. Hepatology. 2006;44(4):865–873. doi:10.1002/hep.2132717006923
  • FrancqueSM, GraaffDVD, KwantenWJ. Non-alcoholic fatty liver disease and cardiovascular risk: pathophysiological mechanisms and implications. J Hepatol. 2016;65(2):425–443. doi:10.1016/j.jhep.2016.04.00527091791
  • HamaguchiM, KojimaT, TakedaN, et al. Nonalcoholic fatty liver disease is a novel predictor of cardiovascular disease. World J Gastroenterol. 2007;13(10):1579–1584. doi:10.3748/wjg.v13.i10.157917461452
  • KimD, KimWR, KimHJ, TherneauTM. Association between noninvasive fibrosis markers and mortality among adults with nonalcoholic fatty liver disease in the United States. Hepatology. 2013;57(4):1357–1365. doi:10.1002/hep.2615623175136
  • ParkJG, JungJ, VermaKK, et al. Liver stiffness by magnetic resonance elastography is associated with increased risk of cardiovascular disease in patients with non-alcoholic fatty liver disease. Aliment Pharmacol Ther. 2021;53(9):1030–1037.33764550
  • StepanovaM, YounossiZM. Independent Association Between Nonalcoholic Fatty Liver Disease and Cardiovascular Disease in the US Population. Clin Gastroenterol Hepatol. 2012;10(6):646–650. doi:10.1016/j.cgh.2011.12.03922245962
  • LazoM, HernaezR, BonekampS, et al. Non-alcoholic fatty liver disease and mortality among US adults: prospective cohort study. BMJ. 2011;343:d6891. doi:10.1136/bmj.d689122102439
  • AnsteeQM, MantovaniA, TilgH, TargherG. Risk of cardiomyopathy and cardiac arrhythmias in patients with nonalcoholic fatty liver disease. Nat Rev Gastroenterol Hepatol. 2018;15(7):425–439. doi:10.1038/s41575-018-0010-029713021
  • HattoriM, TaylorTD. The human intestinal microbiome: a new frontier of human biology. DNA Res. 2009;16(1):1–12. doi:10.1093/dnares/dsn03319147530
  • TurnbaughPJ, LeyRE, HamadyM, Fraser-LiggettCM, KnightR, GordonJI. The human microbiome project. Nature. 2007;449(7164):804–810. doi:10.1038/nature0624417943116
  • BiagiE, CandelaM, Fairweather-TaitS, FranceschiC, BrigidiP. Aging of the human metaorganism: the microbial counterpart. Age. 2012;34(1):247–267. doi:10.1007/s11357-011-9217-521347607
  • BiagiE, NylundL, CandelaM, et al. Through ageing, and beyond: gut microbiota and inflammatory status in seniors and centenarians. PLoS One. 2010;5(5):e10667. doi:10.1371/journal.pone.001066720498852
  • ClaessonMJ, JefferyIB, CondeS, et al. Gut microbiota composition correlates with diet and health in the elderly. Nature. 2012;488(7410):178–184. doi:10.1038/nature1131922797518
  • BoyceJM, ShoneGR. Effects of ageing on smell and taste. Postgrad Med J. 2006;82(966):239–241. doi:10.1136/pgmj.2005.03945316597809
  • LaurinD, BrodeurJM, BourdagesJ, ValléeR, LachapelleD. Fibre intake in elderly individuals with poor masticatory performance. J Can Dent Assoc. 1994;60(5):443–446.8004522
  • OudshoornC, van der CammenTJ, McMurdoME, van LeeuwenJP, ColinEM. Ageing and vitamin D deficiency: effects on calcium homeostasis and considerations for vitamin D supplementation. Br J Nutr. 2009;101(11):1597–1606. doi:10.1017/S000711450933884219393111
  • MaynardCL, WeaverCT. Intestinal effector T cells in health and disease. Immunity. 2009;31(3):389–400. doi:10.1016/j.immuni.2009.08.01219766082
  • LarbiA, FranceschiC, MazzattiD, SolanaR, WikbyA, PawelecG. Aging of the immune system as a prognostic factor for human longevity. Physiology. 2008;23:64–74.18400689
  • SchneiderKM, BieghsV, HeymannF, et al. CX3CR1 is a gatekeeper for intestinal barrier integrity in mice: limiting steatohepatitis by maintaining intestinal homeostasis. Hepatology. 2015;62(5):1405–1416. doi:10.1002/hep.2798226178698
  • BertolottiM, LonardoA, MussiC, et al. Nonalcoholic fatty liver disease and aging: epidemiology to management. World J Gastroenterol. 2014;20(39):14185–14204. doi:10.3748/wjg.v20.i39.1418525339806
  • CaussyC, TripathiA, HumphreyG, et al. A gut microbiome signature for cirrhosis due to nonalcoholic fatty liver disease. Nat Commun. 2019;10(1):1406. doi:10.1038/s41467-019-09455-930926798
  • KoliadaA, SyzenkoG, MoseikoV, et al. Association between body mass index and Firmicutes/Bacteroidetes ratio in an adult Ukrainian population. BMC Microbiol. 2017;17(1):120. doi:10.1186/s12866-017-1027-128532414
  • MuellerS, SaunierK, HanischC, et al. Differences in fecal microbiota in different European study populations in relation to age, gender, and country: a cross-sectional study. Appl Environ Microbiol. 2006;72(2):1027–1033. doi:10.1128/AEM.72.2.1027-1033.200616461645
  • JanssenI, HeymsfieldSB, RossR. Low relative skeletal muscle mass (sarcopenia) in older persons is associated with functional impairment and physical disability. J Am Geriatr Soc. 2002;50(5):889–896. doi:10.1046/j.1532-5415.2002.50216.x12028177
  • TilgH, MoschenAR. Insulin resistance, inflammation, and non-alcoholic fatty liver disease. Trends Endocrinol Metab. 2008;19(10):371–379. doi:10.1016/j.tem.2008.08.00518929493
  • AbbatecolaAM, PaolissoG, FattorettiP, et al. Discovering pathways of sarcopenia in older adults: a role for insulin resistance on mitochondria dysfunction. J Nutr Health Aging. 2011;15(10):890–895. doi:10.1007/s12603-011-0366-022159778
  • HongHC, HwangSY, ChoiHY, et al. Relationship between sarcopenia and nonalcoholic fatty liver disease: the Korean Sarcopenic Obesity Study. Hepatology. 2014;59(5):1772–1778. doi:10.1002/hep.2671623996808
  • BeyerI, MetsT, BautmansI. Chronic low-grade inflammation and age-related sarcopenia. Curr Opin Clin Nutr Metab Care. 2012;15(1):12–22. doi:10.1097/MCO.0b013e32834dd29722108098
  • ElectrophysiologyTFOTESOCTNA. Heart rate variability: standards of measurement, physiological interpretation and clinical use. Task Force of the European Society of Cardiology and the North American Society of Pacing and Electrophysiology. Circulation. 1996;93(5):1043–1065. doi:10.1161/01.CIR.93.5.10438598068
  • LiuYC, HungCS, WuYW, et al. Influence of non-alcoholic fatty liver disease on autonomic changes evaluated by the time domain, frequency domain, and symbolic dynamics of heart rate variability. PLoS One. 2013;8(4):e61803. doi:10.1371/journal.pone.006180323626730
  • NewtonJL. Systemic symptoms in non-alcoholic fatty liver disease. Dig Dis. 2010;28(1):214–219. doi:10.1159/00028208920460914
  • FrithJ, NewtonJL. Autonomic dysfunction in chronic liver disease. Liver Int. 2009;29(4):483–489. doi:10.1111/j.1478-3231.2009.01985.x19323779
  • NewtonJL, HollingsworthKG, TaylorR, et al. Cognitive impairment in primary biliary cirrhosis: symptom impact and potential etiology. Hepatology. 2008;48(2):541–549. doi:10.1002/hep.2237118563843
  • FortonDM, ThomasHC, MurphyCA, et al. Hepatitis C and cognitive impairment in a cohort of patients with mild liver disease. Hepatology. 2002;35(2):433–439. doi:10.1053/jhep.2002.3068811826420
  • YilmazY, OzdoganO. Liver disease as a risk factor for cognitive decline and dementia: an under-recognized issue. Hepatology. 2009;49(2):698. doi:10.1002/hep.2275219177568
  • BreaA, MosqueraD, MartínE, AriztiA, CorderoJL, RosE. Nonalcoholic fatty liver disease is associated with carotid atherosclerosis: a case-control study. Arterioscler Thromb Vasc Biol. 2005;25(5):1045–1050. doi:10.1161/01.ATV.0000160613.57985.1815731489
  • AbbatecolaAM, FerrucciL, MarfellaR, PaolissoG. Insulin resistance and cognitive decline may be common soil for frailty syndrome. Arch Intern Med. 2007;167(19):2145–2146. doi:10.1001/archinte.167.19.2145-b17954815
  • KoplayM, GulcanE, OzkanF. Association between serum vitamin B12 levels and the degree of steatosis in patients with nonalcoholic fatty liver disease. J Investig Med. 2011;59(7):1137–1140. doi:10.2310/JIM.0b013e31822a29f5
  • SepeA, TchkoniaT, ThomouT, ZamboniM, KirklandJL. Aging and regional differences in fat cell progenitors - a mini-review. Gerontology. 2011;57(1):66–75. doi:10.1159/00027975520110661
  • TchkoniaT, MorbeckDE, Von ZglinickiT, et al. Fat tissue, aging, and cellular senescence. Aging Cell. 2010;9(5):667–684. doi:10.1111/j.1474-9726.2010.00608.x20701600
  • SpaldingKL, ArnerE, WestermarkPO, et al. Dynamics of fat cell turnover in humans. Nature. 2008;453(7196):783–787. doi:10.1038/nature0690218454136
  • BaysHE, González-CampoyJM, BrayGA, et al. Pathogenic potential of adipose tissue and metabolic consequences of adipocyte hypertrophy and increased visceral adiposity. Expert Rev Cardiovasc Ther. 2008;6(3):343–368.18327995
  • GuoW, PirtskhalavaT, TchkoniaT, et al. Aging results in paradoxical susceptibility of fat cell progenitors to lipotoxicity. Am J Physiol Endocrinol Metab. 2007;292(4):E1041–1051. doi:10.1152/ajpendo.00557.200617148751
  • SuganamiT, Tanimoto-KoyamaK, NishidaJ, et al. Role of the Toll-like receptor 4/NF-kappaB pathway in saturated fatty acid-induced inflammatory changes in the interaction between adipocytes and macrophages. Arterioscler Thromb Vasc Biol. 2007;27(1):84–91. doi:10.1161/01.ATV.0000251608.09329.9a17082484
  • FinelliC, SommellaL, GioiaS, La SalaN, TarantinoG. Should visceral fat be reduced to increase longevity?Ageing Res Rev. 2013;12(4):996–1004. doi:10.1016/j.arr.2013.05.00723764746
  • ChengH-Y, WangH-Y, ChangW-H, et al. Nonalcoholic Fatty Liver Disease: prevalence, Influence on Age and Sex, and Relationship with Metabolic Syndrome and Insulin Resistance. Int J Gerontol. 2013;7(4):194–198. doi:10.1016/j.ijge.2013.03.008
  • American Diabetes Association. Comprehensive Medical Evaluation and Assessment of Comorbidities: standards of Medical Care in Diabetes-2020. Diabetes Care. 2020;43(Suppl1):S37–S47. doi:10.2337/dc20-S00431862747
  • BrowningJD, SzczepaniakLS, DobbinsR, et al. Prevalence of hepatic steatosis in an urban population in the United States: impact of ethnicity. Hepatology. 2004;40(6):1387–1395. doi:10.1002/hep.2046615565570
  • SorrentinoP, TarantinoG, ConcaP, et al. Silent non-alcoholic fatty liver disease—a clinical–histological study. J Hepatol. 2004;41(5):751–757. doi:10.1016/j.jhep.2004.07.01015519647
  • FracanzaniAL, ValentiL, BugianesiE, et al. Risk of severe liver disease in nonalcoholic fatty liver disease with normal aminotransferase levels: a role for insulin resistance and diabetes. Hepatology. 2008;48(3):792–798. doi:10.1002/hep.2242918752331
  • KotronenA, PeltonenM, HakkarainenA, et al. Prediction of Non-Alcoholic Fatty Liver Disease and Liver Fat Using Metabolic and Genetic Factors. Gastroenterology. 2009;137(3):865–872. doi:10.1053/j.gastro.2009.06.00519524579
  • HuangX, XuM, ChenY, et al. Validation of the Fatty Liver Index for Nonalcoholic Fatty Liver Disease in Middle-Aged and Elderly Chinese. Medicine. 2015;94(40):e1682. doi:10.1097/MD.000000000000168226448014
  • SviklāneL, OlmaneE, DzērveZ, KupčsK, PīrāgsV, SokolovskaJ. Fatty liver index and hepatic steatosis index for prediction of non-alcoholic fatty liver disease in type 1 diabetes. J Gastroenterol Hepatol. 2018;33(1):270–276. doi:10.1111/jgh.1381428464337
  • ByrneCD. EASL–EASD–EASO Clinical Practice Guidelines for the management of non-alcoholic fatty liver disease. J Hepatol. 2016;64(6):1388–1402. doi:10.1016/j.jhep.2015.11.00427062661
  • AdamsLA, GeorgeJ, BugianesiE, et al. Complex non-invasive fibrosis models are more accurate than simple models in non-alcoholic fatty liver disease. J Gastroenterol Hepatol. 2011;26(10):1536–1543. doi:10.1111/j.1440-1746.2011.06774.x21950746
  • AnguloP, HuiJM, MarchesiniG, et al. The NAFLD fibrosis score: a noninvasive system that identifies liver fibrosis in patients with NAFLD. Hepatology. 2007;45(4):846–854. doi:10.1002/hep.2149617393509
  • McPhersonS, HardyT, DufourJ-F, et al. Age as a Confounding Factor for the Accurate Non-Invasive Diagnosis of Advanced NAFLD Fibrosis. Official j Am College Gastroenterol. 2017;112(5):740–751. doi:10.1038/ajg.2016.453
  • PitisuttithumP, ChanWK, PiyachaturawatP, et al. Predictors of advanced fibrosis in elderly patients with biopsy-confirmed nonalcoholic fatty liver disease: the GOASIA study. BMC Gastroenterol. 2020;20(1):88. doi:10.1186/s12876-020-01240-z32252638
  • MichelM, SchattenbergJM. Liver-specific diagnostic for non-alcoholic fatty liver disease (NAFLD) - time to replace liver biopsy?. Z Gastroenterol. 2020;58(12):1233–1240.33291177
  • CasteraL, Friedrich-RustM, LoombaR. Noninvasive Assessment of Liver Disease in Patients With Nonalcoholic Fatty Liver Disease. Gastroenterology. 2019;156(5):1264–1281.e1264. doi:10.1053/j.gastro.2018.12.03630660725
  • ImajoK, KessokuT, HondaY, et al. Magnetic Resonance Imaging More Accurately Classifies Steatosis and Fibrosis in Patients With Nonalcoholic Fatty Liver Disease Than Transient Elastography. Gastroenterology. 2016;150(3):626–637.e627. doi:10.1053/j.gastro.2015.11.04826677985
  • WongVW-S, VergniolJ, WongGL-H, et al. Diagnosis of fibrosis and cirrhosis using liver stiffness measurement in nonalcoholic fatty liver disease. Hepatology. 2010;51(2):454–462. doi:10.1002/hep.2331220101745
  • GilmoreIT, BurroughsA, Murray-LyonIM, WilliamsR, JenkinsD, HopkinsA. Indications, methods, and outcomes of percutaneous liver biopsy in England and Wales: an audit by the British Society of Gastroenterology and the Royal College of Physicians of London. Gut. 1995;36(3):437–441. doi:10.1136/gut.36.3.4377698705
  • EslamM, SanyalAJ, GeorgeJ. MAFLD: a Consensus-Driven Proposed Nomenclature for Metabolic Associated Fatty Liver Disease. Gastroenterology. 2020;158(7):1999–2014.e1991. doi:10.1053/j.gastro.2019.11.31232044314
  • FouadY, WakedI, BollipoS, GomaaA, AjlouniY, AttiaD. What’s in a name? Renaming ‘NAFLD’ to ‘MAFLD’. Liver Int. 2020;40(6):1254–1261. doi:10.1111/liv.1447832301554
  • PromratK, KleinerDE, NiemeierHM, et al. Randomized controlled trial testing the effects of weight loss on nonalcoholic steatohepatitis. Hepatology. 2010;51(1):121–129. doi:10.1002/hep.2327619827166
  • Vilar-GomezE, Martinez-PerezY, Calzadilla-BertotL, et al. Weight Loss Through Lifestyle Modification Significantly Reduces Features of Nonalcoholic Steatohepatitis. Gastroenterology. 2015;149(2):367–378.e365. doi:10.1053/j.gastro.2015.04.00525865049
  • WtA, WvL, McH, BbA. Lifestyle Modification for Obesity. Circulation. 2012;125(9):1157–1170. doi:10.1161/CIRCULATIONAHA.111.03945322392863
  • AggarwalB, LiaoM, AllegranteJP, MoscaL. Low Social Support Level is Associated with Non-Adherence to Diet at 1 Year in the Family Intervention Trial for Heart Health (FIT Heart). J Nutr Educ Behav. 2010;42(6):380–388. doi:10.1016/j.jneb.2009.08.00620696617
  • KrukowskiRA, Harvey-BerinoJ, BursacZ, AshikagaT, WestDS. Patterns of success: online self-monitoring in a web-based behavioral weight control program. Health Psychol. 2013;32(2):164–170. doi:10.1037/a002813522545978
  • RyuS, ChangY, JungH-S, et al. Relationship of sitting time and physical activity with non-alcoholic fatty liver disease. J Hepatol. 2015;63(5):1229–1237. doi:10.1016/j.jhep.2015.07.01026385766
  • Romero-GómezM, Zelber-SagiS, TrenellM. Treatment of NAFLD with diet, physical activity and exercise. J Hepatol. 2017;67(4):829–846. doi:10.1016/j.jhep.2017.05.01628545937
  • EvansC. Malnutrition in the elderly: a multifactorial failure to thrive. Perm J. 2005;9(3):38–41. doi:10.7812/TPP/05-05622811627
  • SieberCC. Malnutrition and sarcopenia. Aging Clin Exp Res. 2019;31(6):793–798. doi:10.1007/s40520-019-01170-131148100
  • YangX, XuZ, ZhangC, CaiZ, ZhangJ. Metformin, beyond an insulin sensitizer, targeting heart and pancreatic β cells. Biochimica et Biophysica Acta. 2017;1863(8):1984–1990. doi:10.1016/j.bbadis.2016.09.01927702625
  • LiY, LiuL, WangB, WangJ, ChenD. Metformin in non-alcoholic fatty liver disease: a systematic review and meta-analysis. Biomed Rep. 2013;1(1):57–64. doi:10.3892/br.2012.1824648894
  • RizosCV, KeiA, ElisafMS. The current role of thiazolidinediones in diabetes management. Arch Toxicol. 2016;90(8):1861–1881.27165418
  • BaileyCJ. Treating insulin resistance in type 2 diabetes with metformin and thiazolidinediones. Diabetes Obes Metab. 2005;7(6):675–691. doi:10.1111/j.1463-1326.2005.00497.x16219011
  • MussoG, CassaderM, PaschettaE, GambinoR. Thiazolidinediones and Advanced Liver Fibrosis in Nonalcoholic Steatohepatitis: a Meta-analysis. JAMA Intern Med. 2017;177(5):633. doi:10.1001/jamainternmed.2016.960728241279
  • BrilF, KalavalapalliS, ClarkVC, et al. Response to Pioglitazone in Patients With Nonalcoholic Steatohepatitis With vs Without Type 2 Diabetes. Clin Gastroenterol Hepatol. 2018;16(4):558–566.e552. doi:10.1016/j.cgh.2017.12.00129223443
  • BelfortR, HarrisonSA, BrownK, et al. A Placebo-Controlled Trial of Pioglitazone in Subjects with Nonalcoholic Steatohepatitis. N Eng J Med. 2006;355(22):2297–2307. doi:10.1056/NEJMoa060326
  • SanyalAJ, ChalasaniN, KowdleyKV, et al. Pioglitazone, Vitamin E, or Placebo for Nonalcoholic Steatohepatitis. N Eng J Med. 2010;362(18):1675–1685. doi:10.1056/NEJMoa0907929
  • HernandezAV, UsmaniA, RajamanickamA, MoheetA. Thiazolidinediones and Risk of Heart Failure in Patients with or at High Risk of Type 2 Diabetes Mellitus. Am J Cardiovascular Drugs. 2011;11(2):115–128. doi:10.2165/11587580-000000000-00000
  • KakuK, HashiramotoM. Thiazolidinediones and bone fractures. J Diabetes Investig. 2011;2(5):354–355. doi:10.1111/j.2040-1124.2011.00142.x
  • EnsrudKE. Epidemiology of Fracture Risk With Advancing Age. J Gerontolo. 2013;68(10):1236–1242. doi:10.1093/gerona/glt092
  • LuY, HajifathalianK, EzzatiM, WoodwardM, RimmEB, DanaeiG. Metabolic mediators of the effects of body-mass index, overweight, and obesity on coronary heart disease and stroke: a pooled analysis of 97 prospective cohorts with 1·8 million participants. Lancet. 2014;383(9921):970–983.24269108
  • WangXX, JiangT, ShenY, et al. The farnesoid X receptor modulates renal lipid metabolism and diet-induced renal inflammation, fibrosis, and proteinuria. Am J Physiol Renal Physiol. 2009;297(6):F1587–F1596. doi:10.1152/ajprenal.00404.200919776172
  • MudaliarS, HenryRR, SanyalAJ, et al. Efficacy and Safety of the Farnesoid X Receptor Agonist Obeticholic Acid in Patients With Type 2 Diabetes and Nonalcoholic Fatty Liver Disease. Gastroenterology. 2013;145(3):574–582.e571. doi:10.1053/j.gastro.2013.05.04223727264
  • Neuschwander-TetriBA, LoombaR, SanyalAJ, et al. Farnesoid X nuclear receptor ligand obeticholic acid for non-cirrhotic, non-alcoholic steatohepatitis (FLINT): a multicentre, randomised, placebo-controlled trial. Lancet. 2015;385(9972):956–965. doi:10.1016/S0140-6736(14)61933-425468160
  • YounossiZM, RatziuV, LoombaR, et al. Obeticholic acid for the treatment of non-alcoholic steatohepatitis: interim analysis from a multicentre, randomised, placebo-controlled phase 3 trial. Lancet. 2019;394(10215):2184–2196. doi:10.1016/S0140-6736(19)33041-731813633
  • KatoutM, ZhuH, RutskyJ, et al. Effect of GLP-1 Mimetics on Blood Pressure and Relationship to Weight Loss and Glycemia Lowering: results of a Systematic Meta-Analysis and Meta-Regression. Am J Hypertens. 2014;27(1):130–139. doi:10.1093/ajh/hpt19624263424
  • SunF, WuS, WangJ, et al. Effect of Glucagon-like Peptide-1 Receptor Agonists on Lipid Profiles Among Type 2 Diabetes: a Systematic Review and Network Meta-analysis. Clin Ther. 2015;37(1):225–241.e228. doi:10.1016/j.clinthera.2014.11.00825554560
  • ArmstrongMJ, GauntP, AithalGP, et al. Liraglutide safety and efficacy in patients with non-alcoholic steatohepatitis (LEAN): a multicentre, double-blind, randomised, placebo-controlled phase 2 study. Lancet. 2016;387(10019):679–690. doi:10.1016/S0140-6736(15)00803-X26608256
  • NewsomePN, BuchholtzK, CusiK, et al. A Placebo-Controlled Trial of Subcutaneous Semaglutide in Nonalcoholic Steatohepatitis. N Eng J Med. 2020;2:4.
  • StaigerH, KeuperM, BertiL. Hrabě de Angelis M, Häring H-U. Fibroblast Growth Factor 21—Metabolic Role in Mice and Men. Endocr Rev. 2017;38(5):468–488.28938407
  • VerzijlCRC, PeppelIPVD, StruikD, JonkerJW. Pegbelfermin (BMS-986036): an investigational PEGylated fibroblast growth factor 21 analogue for the treatment of nonalcoholic steatohepatitis. Expert Opin Investig Drugs. 2020;29(2):125–133.
  • SquibbB-M. A Phase 2B Randomized, Double-Blind, Placebo-Controlled Study Evaluating the Safety and Efficacy of BMS-986036 (PEG-FGF21) in Adults with Nonalcoholic Steatohepatitis (NASH) and Compensated Liver Cirrhosis. clinicaltrials.gov; 2020.
  • SquibbB-M. A Phase 2B Randomized Double-Blind, Placebo-Controlled Study Evaluating the Safety and Efficacy of BMS-986036 (PEG-FGF21) in Adults with Nonalcoholic Steatohepatitis (NASH) and Stage 3 Liver Fibrosis. clinicaltrials.gov; 2021.
  • EmergingYZ. Research on MGL-3196 for the Treatment of Nonalcoholic Steatohepatitis. Gastroenterol Hepatol (N Y). 2019;15(6):317–319.31391799
  • Madrigal PharmaceuticalsI. A Phase 3, Multinational, Double-Blind, Randomized, Placebo-Controlled Study of MGL-3196 (Resmetirom) in Patients with Non-Alcoholic Steatohepatitis (NASH) and Fibrosis to Resolve NASH and Reduce Progression to Cirrhosis And/Or Hepatic Decompensation. clinicaltrials.gov; 2021.