Skip to main content

Advertisement

Log in

Role of the Circadian Clock in the Metabolic Syndrome and Nonalcoholic Fatty Liver Disease

  • Review
  • Published:
Digestive Diseases and Sciences Aims and scope Submit manuscript

Abstract

Nonalcoholic fatty liver disease (NAFLD) is the most common chronic liver disease in industrialized nations and is strongly associated with the metabolic syndrome. The prevalence of NAFLD continues to rise along with the epidemic of the metabolic syndrome. Metabolic homeostasis is linked to the circadian clock (rhythm), with multiple signaling pathways in organs regulated by circadian clock genes, and recent studies of circadian clock gene functions suggest that disruption of the circadian rhythm is associated with significant morbidity and mortality, including the metabolic syndrome. In the industrialized world, various human behaviors and activities such as work and eating patterns, jet lag, and sleep deprivation interfere with the circadian rhythm, leading to perturbations in metabolism and development of the metabolic syndrome. In this review, we discuss how disruption of the circadian rhythm is associated with various metabolic conditions that comprise the metabolic syndrome and NAFLD.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Institutional subscriptions

Fig. 1
Fig. 2

Similar content being viewed by others

References

  1. Sayiner M, Koenig A, Henry L, Younossi ZM. Epidemiology of nonalcoholic fatty liver disease and nonalcoholic steatohepatitis in the United States and the rest of the world. Clin Liver Dis. 2016;20:205–214.

    PubMed  Google Scholar 

  2. Marchesini G, Marzocchi R. Metabolic syndrome and NASH. Clin Liver Dis. 2007;11:105–117, ix.

    PubMed  Google Scholar 

  3. Armstrong MJ, Adams LA, Canbay A, Syn WK. Extrahepatic complications of nonalcoholic fatty liver disease. Hepatology (Baltimore, MD). 2014;59:1174–1197.

    CAS  Google Scholar 

  4. Wong RJ, Ahmed A. Obesity and nonalcoholic fatty liver disease: disparate associations among Asian populations. World J Hepatol. 2014;6:263–273.

    PubMed  PubMed Central  Google Scholar 

  5. Kanwar P, Kowdley KV. The metabolic syndrome and its influence on nonalcoholic steatohepatitis. Clin Liver Dis. 2016;20:225–243.

    PubMed  Google Scholar 

  6. Day CP, James OF. Steatohepatitis: a tale of two “hits”? Gastroenterology. 1998;114:842–845.

    CAS  PubMed  Google Scholar 

  7. Peverill W, Powell LW, Skoien R. Evolving concepts in the pathogenesis of NASH: beyond steatosis and inflammation. Int J Mol Sci. 2014;15:8591–8638.

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Buzzetti E, Pinzani M, Tsochatzis EA. The multiple-hit pathogenesis of nonalcoholic fatty liver disease (NAFLD). Metab Clin Exp. 2016;65:1038–1048.

    CAS  PubMed  Google Scholar 

  9. Harper DG, Volicer L, Stopa EG, McKee AC, Nitta M, Satlin A. Disturbance of endogenous circadian rhythm in aging and Alzheimer disease. Am J Geriatr Psychiatry. 2005;13:359–368.

    PubMed  Google Scholar 

  10. Wang F, Yeung KL, Chan WC, et al. A meta-analysis on dose–response relationship between night shift work and the risk of breast cancer. Ann Oncol. 2013;24:2724–2732.

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Wang F, Zhang L, Zhang Y, et al. Meta-analysis on night shift work and risk of metabolic syndrome. Obes Rev. 2014;15:709–720.

    CAS  PubMed  Google Scholar 

  12. Reppert SM, Weaver DR. Coordination of circadian timing in mammals. Nature. 2002;418:935–941.

    CAS  PubMed  Google Scholar 

  13. Hirota T, Fukada Y. Resetting mechanism of central and peripheral circadian clocks in mammals. Zool Sci. 2004;21:359–368.

    Google Scholar 

  14. Berson DM, Dunn FA, Takao M. Phototransduction by retinal ganglion cells that set the circadian clock. Science (New York, N.Y.). 2002;295:1070–1073.

    CAS  Google Scholar 

  15. Yamazaki S, Numano R, Abe M, et al. Resetting central and peripheral circadian oscillators in transgenic rats. Science (New York, N.Y.). 2000;288:682–685.

    CAS  Google Scholar 

  16. Damiola F, Le Minh N, Preitner N, Kornmann B, Fleury-Olela F, Schibler U. Restricted feeding uncouples circadian oscillators in peripheral tissues from the central pacemaker in the suprachiasmatic nucleus. Genes Dev. 2000;14:2950–2961.

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Mendoza J, Graff C, Dardente H, Pevet P, Challet E. Feeding cues alter clock gene oscillations and photic responses in the suprachiasmatic nuclei of mice exposed to a light/dark cycle. J Neurosci. 2005;25:1514–1522.

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Ackermann K, Plomp R, Lao O, et al. Effect of sleep deprivation on rhythms of clock gene expression and melatonin in humans. Chronobiol Int. 2013;30:901–909.

    CAS  PubMed  Google Scholar 

  19. Cedernaes J, Osler ME, Voisin S, et al. Acute sleep loss induces tissue-specific epigenetic and transcriptional alterations to circadian clock genes in men. J Clin Endocrinol Metab. 2015;100:E1255–E1261.

    PubMed  Google Scholar 

  20. Potter GD, Skene DJ, Arendt J, Cade JE, Grant PJ, Hardie LJ. Circadian rhythm and sleep disruption: causes, metabolic consequences, and countermeasures. Endocr Rev. 2016;37:584–608.

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Young MW, Kay SA. Time zones: a comparative genetics of circadian clocks. Nat Rev Genet. 2001;2:702–715.

    CAS  PubMed  Google Scholar 

  22. Brown SA. Circadian rhythms. A new histone code for clocks? Science (New York, N.Y.). 2011;333:1833–1834.

    CAS  Google Scholar 

  23. Brown SA, Kowalska E, Dallmann R. (Re)inventing the circadian feedback loop. Dev Cell. 2012;22:477–487.

    CAS  PubMed  Google Scholar 

  24. Preitner N, Damiola F, Lopez-Molina L, et al. The orphan nuclear receptor REV-ERBalpha controls circadian transcription within the positive limb of the mammalian circadian oscillator. Cell. 2002;110:251–260.

    CAS  PubMed  Google Scholar 

  25. Sato TK, Panda S, Miraglia LJ, et al. A functional genomics strategy reveals Rora as a component of the mammalian circadian clock. Neuron. 2004;43:527–537.

    CAS  PubMed  Google Scholar 

  26. Canaple L, Rambaud J, Dkhissi-Benyahya O, et al. Reciprocal regulation of brain and muscle Arnt-like protein 1 and peroxisome proliferator-activated receptor alpha defines a novel positive feedback loop in the rodent liver circadian clock. Mol Endocrinol (Baltimore, MD). 2006;20:1715–1727.

    CAS  Google Scholar 

  27. Crumbley C, Wang Y, Banerjee S, Burris TP. Regulation of expression of citrate synthase by the retinoic acid receptor-related orphan receptor alpha (RORalpha). PLoS ONE. 2012;7:e33804.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Billon C, Sitaula S, Burris TP. Metabolic characterization of a novel RORalpha knockout mouse model without Ataxia. Front Endocrinol. 2017;8:141.

    Google Scholar 

  29. Akashi M, Tsuchiya Y, Yoshino T, Nishida E. Control of intracellular dynamics of mammalian period proteins by casein kinase I epsilon (CKIepsilon) and CKIdelta in cultured cells. Mol Cell Biol. 2002;22:1693–1703.

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Marcheva B, Ramsey KM, Affinati A, Bass J. Clock genes and metabolic disease. J Appl Physiol (Bethesda, MD, 1985). 2009;107:1638–1646.

    CAS  Google Scholar 

  31. Lamia KA, Sachdeva UM, DiTacchio L, et al. AMPK regulates the circadian clock by cryptochrome phosphorylation and degradation. Science (New York, NY). 2009;326:437–440.

    CAS  Google Scholar 

  32. Moller N, Jorgensen JO, Schmitz O, et al. Effects of a growth hormone pulse on total and forearm substrate fluxes in humans. Am J Physiol. 1990;258:E86–E91.

    CAS  PubMed  Google Scholar 

  33. Moller N, Jorgensen JO. Effects of growth hormone on glucose, lipid, and protein metabolism in human subjects. Endocr Rev. 2009;30:152–177.

    PubMed  Google Scholar 

  34. Linkowski P, Mendlewicz J, Kerkhofs M, et al. 24-Hour profiles of adrenocorticotropin, cortisol, and growth hormone in major depressive illness: effect of antidepressant treatment. J Clin Endocrinol Metab. 1987;65:141–152.

    CAS  PubMed  Google Scholar 

  35. Linkowski P, Kerkhofs M, Van Onderbergen A, et al. The 24-hour profiles of cortisol, prolactin, and growth hormone secretion in mania. Arch Gen Psychiatry. 1994;51:616–624.

    CAS  PubMed  Google Scholar 

  36. Ho KY, Veldhuis JD, Johnson ML, et al. Fasting enhances growth hormone secretion and amplifies the complex rhythms of growth hormone secretion in man. J Clin Investig. 1988;81:968–975.

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Vakili H, Jin Y, Cattini PA. Evidence for a circadian effect on the reduction of human growth hormone gene expression in response to excess caloric intake. J Biol Chem. 2016;291:13823–13833.

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Brown GM. Light, melatonin and the sleep–wake cycle. J Psychiatry Neurosci. 1994;19:345–353.

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Moore RY. Neural control of the pineal gland. Behav Brain Res. 1996;73:125–130.

    CAS  PubMed  Google Scholar 

  40. Klein DC. Arylalkylamine N-acetyltransferase: “the Timezyme”. J Biol Chem. 2007;282:4233–4237.

    CAS  PubMed  Google Scholar 

  41. Bouatia-Naji N, Bonnefond A, Cavalcanti-Proenca C, et al. A variant near MTNR1B is associated with increased fasting plasma glucose levels and type 2 diabetes risk. Nat Genet. 2009;41:89–94.

    CAS  PubMed  Google Scholar 

  42. Prokopenko I, Langenberg C, Florez JC, et al. Variants in MTNR1B influence fasting glucose levels. Nat Genet. 2009;41:77–81.

    CAS  PubMed  Google Scholar 

  43. Lane JM, Chang AM, Bjonnes AC, et al. Impact of common diabetes risk variant in MTNR1B on sleep, circadian, and melatonin physiology. Diabetes. 2016;65:1741–1751.

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Tuomi T, Nagorny CL, Singh P, et al. Increased melatonin signaling is a risk factor for type 2 diabetes. Cell Metab. 2016;23:1067–1077.

    CAS  PubMed  Google Scholar 

  45. Ahima RS, Prabakaran D, Mantzoros C, et al. Role of leptin in the neuroendocrine response to fasting. Nature. 1996;382:250–252.

    CAS  PubMed  Google Scholar 

  46. Bodosi B, Gardi J, Hajdu I, Szentirmai E, Obal F Jr, Krueger JM. Rhythms of ghrelin, leptin, and sleep in rats: effects of the normal diurnal cycle, restricted feeding, and sleep deprivation. Am J Physiol Regul Integr Comp Physiol. 2004;287:R1071–R1079.

    CAS  PubMed  Google Scholar 

  47. Ando H, Kumazaki M, Motosugi Y, et al. Impairment of peripheral circadian clocks precedes metabolic abnormalities in ob/ob mice. Endocrinology. 2011;152:1347–1354.

    CAS  PubMed  Google Scholar 

  48. Hwang CS, Mandrup S, MacDougald OA, Geiman DE, Lane MD. Transcriptional activation of the mouse obese (ob) gene by CCAAT/enhancer binding protein alpha. Proc Natl Acad Sci USA. 1996;93:873–877.

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Hollenberg AN, Susulic VS, Madura JP, et al. Functional antagonism between CCAAT/Enhancer binding protein-alpha and peroxisome proliferator-activated receptor-gamma on the leptin promoter. J Biol Chem. 1997;272:5283–5290.

    CAS  PubMed  Google Scholar 

  50. Kettner NM, Mayo SA, Hua J, Lee C, Moore DD, Fu L. Circadian dysfunction induces leptin resistance in mice. Cell Metab. 2015;22:448–459.

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Spiegel K, Tasali E, Penev P, Van Cauter E. Brief communication: sleep curtailment in healthy young men is associated with decreased leptin levels, elevated ghrelin levels, and increased hunger and appetite. Ann Intern Med. 2004;141:846–850.

    PubMed  Google Scholar 

  52. Scheer FA, Hilton MF, Mantzoros CS, Shea SA. Adverse metabolic and cardiovascular consequences of circadian misalignment. Proc Natl Acad Sci USA. 2009;106:4453–4458.

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Haus E. Chronobiology in the endocrine system. Adv Drug Deliv Rev. 2007;59:985–1014.

    CAS  PubMed  Google Scholar 

  54. Kalsbeek A, Fliers E. Daily regulation of hormone profiles. Handb Exp Pharmacol. 2013;217:185–226.

    CAS  Google Scholar 

  55. Sviridonova MA, Fadeyev VV, Sych YP, Melnichenko GA. Clinical significance of TSH circadian variability in patients with hypothyroidism. Endocr Res. 2013;38:24–31.

    CAS  PubMed  Google Scholar 

  56. Aninye IO, Matsumoto S, Sidhaye AR, Wondisford FE. Circadian regulation of Tshb gene expression by Rev-Erbalpha (NR1D1) and nuclear corepressor 1 (NCOR1). J Biol Chem. 2014;289:17070–17077.

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Chawla A, Lazar MA. Induction of Rev-ErbA alpha, an orphan receptor encoded on the opposite strand of the alpha-thyroid hormone receptor gene, during adipocyte differentiation. J Biol Chem. 1993;268:16265–16269.

    CAS  PubMed  Google Scholar 

  58. Rey G, Cesbron F, Rougemont J, Reinke H, Brunner M, Naef F. Genome-wide and phase-specific DNA-binding rhythms of BMAL1 control circadian output functions in mouse liver. PLoS Biol. 2011;9:e1000595.

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Lande-Diner L, Boyault C, Kim JY, Weitz CJ. A positive feedback loop links circadian clock factor CLOCK–BMAL1 to the basic transcriptional machinery. Proc Natl Acad Sci USA. 2013;110:16021–16026.

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Peliciari-Garcia RA, Previde RM, Nunes MT, Young ME. Interrelationship between 3,5,3-triiodothyronine and the circadian clock in the rodent heart. Chronobiol Int. 2016;33:1444–1454.

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Lightman SL, Conway-Campbell BL. The crucial role of pulsatile activity of the HPA axis for continuous dynamic equilibration. Nat Rev Neurosci. 2010;11:710–718.

    CAS  PubMed  Google Scholar 

  62. Droste SK, de Groote L, Atkinson HC, Lightman SL, Reul JM, Linthorst AC. Corticosterone levels in the brain show a distinct ultradian rhythm but a delayed response to forced swim stress. Endocrinology. 2008;149:3244–3253.

    CAS  PubMed  Google Scholar 

  63. Conway-Campbell BL, Pooley JR, Hager GL, Lightman SL. Molecular dynamics of ultradian glucocorticoid receptor action. Mol Cell Endocrinol. 2012;348:383–393.

    CAS  PubMed  Google Scholar 

  64. Surjit M, Ganti KP, Mukherji A, et al. Widespread negative response elements mediate direct repression by agonist-liganded glucocorticoid receptor. Cell. 2011;145:224–241.

    CAS  PubMed  Google Scholar 

  65. So AY, Bernal TU, Pillsbury ML, Yamamoto KR, Feldman BJ. Glucocorticoid regulation of the circadian clock modulates glucose homeostasis. Proc Natl Acad Sci USA. 2009;106:17582–17587.

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Conway-Campbell BL, Sarabdjitsingh RA, McKenna MA, et al. Glucocorticoid ultradian rhythmicity directs cyclical gene pulsing of the clock gene period 1 in rat hippocampus. J Neuroendocrinol. 2010;22:1093–1100.

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Torra IP, Tsibulsky V, Delaunay F, et al. Circadian and glucocorticoid regulation of Rev-erbalpha expression in liver. Endocrinology. 2000;141:3799–3806.

    CAS  PubMed  Google Scholar 

  68. de Guia RM, Rose AJ, Herzig S. Glucocorticoid hormones and energy homeostasis. Hormone Mol Biol Clin Investig. 2014;19:117–128.

    Google Scholar 

  69. Dourakis SP, Sevastianos VA, Kaliopi P. Acute severe steatohepatitis related to prednisolone therapy. Am J Gastroenterol. 2002;97:1074–1075.

    CAS  PubMed  Google Scholar 

  70. Matsumoto T, Yamasaki S, Arakawa A, et al. Exposure to a high total dosage of glucocorticoids produces nonalcoholic steatohepatitis. Pathol Int. 2007;57:388–389.

    PubMed  Google Scholar 

  71. Woods CP, Hazlehurst JM, Tomlinson JW. Glucocorticoids and nonalcoholic fatty liver disease. J Steroid Biochem Mol Biol. 2015;154:94–103.

    CAS  PubMed  Google Scholar 

  72. Dijk DJ, Czeisler CA. Paradoxical timing of the circadian rhythm of sleep propensity serves to consolidate sleep and wakefulness in humans. Neurosci Lett. 1994;166:63–68.

    CAS  PubMed  Google Scholar 

  73. Dijk DJ, Czeisler CA. Contribution of the circadian pacemaker and the sleep homeostat to sleep propensity, sleep structure, electroencephalographic slow waves, and sleep spindle activity in humans. J Neurosci. 1995;15:3526–3538.

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Stephan FK, Zucker I. Circadian rhythms in drinking behavior and locomotor activity of rats are eliminated by hypothalamic lesions. Proc Natl Acad Sci USA. 1972;69:1583–1586.

    CAS  PubMed  PubMed Central  Google Scholar 

  75. Nagai K, Nishio T, Nakagawa H, Nakamura S, Fukuda Y. Effect of bilateral lesions of the suprachiasmatic nuclei on the circadian rhythm of food-intake. Brain Res. 1978;142:384–389.

    CAS  PubMed  Google Scholar 

  76. Shimba S, Ishii N, Ohta Y, et al. Brain and muscle Arnt-like protein-1 (BMAL1), a component of the molecular clock, regulates adipogenesis. Proc Natl Acad Sci USA. 2005;102:12071–12076.

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Turek FW, Joshu C, Kohsaka A, et al. Obesity and metabolic syndrome in circadian clock mutant mice. Science (New York, NY). 2005;308:1043–1045.

    CAS  Google Scholar 

  78. Marcheva B, Ramsey KM, Buhr ED, et al. Disruption of the clock components CLOCK and BMAL1 leads to hypoinsulinaemia and diabetes. Nature. 2010;466:627–631.

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Shimba S, Ogawa T, Hitosugi S, et al. Deficient of a clock gene, brain and muscle Arnt-like protein-1 (BMAL1), induces dyslipidemia and ectopic fat formation. PLoS ONE. 2011;6:e25231.

    CAS  PubMed  PubMed Central  Google Scholar 

  80. Shi SQ, Ansari TS, McGuinness OP, Wasserman DH, Johnson CH. Circadian disruption leads to insulin resistance and obesity. Curr Biol CB. 2013;23:372–381.

    CAS  PubMed  Google Scholar 

  81. Husse J, Hintze SC, Eichele G, Lehnert H, Oster H. Circadian clock genes Per1 and Per2 regulate the response of metabolism-associated transcripts to sleep disruption. PLoS ONE. 2012;7:e52983.

    CAS  PubMed  PubMed Central  Google Scholar 

  82. Ford ES, Li C, Wheaton AG, Chapman DP, Perry GS, Croft JB. Sleep duration and body mass index and waist circumference among U.S. adults. Obesity (Silver Spring, MD). 2014;22:598–607.

    Google Scholar 

  83. Elder BL, Ammar EM, Pile D. Sleep duration, activity levels, and measures of obesity in adults. Public Health Nurs (Boston, MA). 2016;33:200–205.

    Google Scholar 

  84. Fatima Y, Doi SA, Mamun AA. Longitudinal impact of sleep on overweight and obesity in children and adolescents: a systematic review and bias-adjusted meta-analysis. Obes Rev. 2015;16:137–149.

    CAS  PubMed  Google Scholar 

  85. Rao MN, Neylan TC, Grunfeld C, Mulligan K, Schambelan M, Schwarz JM. Subchronic sleep restriction causes tissue-specific insulin resistance. J Clin Endocrinol Metabol. 2015;100:1664–1671.

    CAS  Google Scholar 

  86. Cappuccio FP, D’Elia L, Strazzullo P, Miller MA. Quantity and quality of sleep and incidence of type 2 diabetes: a systematic review and meta-analysis. Diabetes Care. 2010;33:414–420.

    PubMed  Google Scholar 

  87. Mullington JM, Simpson NS, Meier-Ewert HK, Haack M. Sleep loss and inflammation. Best Pract Res Clin Endocrinol Metab. 2010;24:775–784.

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Itani O, Kaneita Y, Tokiya M, et al. Short sleep duration, shift work, and actual days taken off work are predictive life-style risk factors for new-onset metabolic syndrome: a seven-year cohort study of 40,000 male workers. Sleep Med. 2017;39:87–94.

    PubMed  Google Scholar 

  89. Kim CW, Yun KE, Jung HS, et al. Sleep duration and quality in relation to nonalcoholic fatty liver disease in middle-aged workers and their spouses. J Hepatol. 2013;59:351–357.

    PubMed  Google Scholar 

  90. Miyake T, Kumagi T, Furukawa S, et al. Short sleep duration reduces the risk of nonalcoholic fatty liver disease onset in men: a community-based longitudinal cohort study. J Gastroenterol. 2015;50:583–589.

    PubMed  Google Scholar 

  91. Miyake T, Kumagi T, Hirooka M, et al. Significance of exercise in nonalcoholic fatty liver disease in men: a community-based large cross-sectional study. J Gastroenterol. 2015;50:230–237.

    PubMed  Google Scholar 

  92. Liu C, Zhong R, Lou J, et al. Nighttime sleep duration and risk of nonalcoholic fatty liver disease: the Dongfeng–Tongji prospective study. Ann Med. 2016;48:468–476.

    PubMed  Google Scholar 

  93. Rybnikova NA, Haim A, Portnov BA. Does artificial light-at-night exposure contribute to the worldwide obesity pandemic? Int J Obes. 2016;40:815–823.

    CAS  Google Scholar 

  94. Weingarten JA, Collop NA. Air travel: effects of sleep deprivation and jet lag. Chest. 2013;144:1394–1401.

    PubMed  Google Scholar 

  95. Fowler PM, McCall A, Jones M, Duffield R. Effects of long-haul transmeridian travel on player preparedness: case study of a national team at the 2014 FIFA World Cup. J Sci Med Sport. 2017;20:322–327.

    PubMed  Google Scholar 

  96. Roenneberg T, Allebrandt KV, Merrow M, Vetter C. Social jetlag and obesity. Curr Biol CB. 2012;22:939–943.

    CAS  PubMed  Google Scholar 

  97. Parsons MJ, Moffitt TE, Gregory AM, et al. Social jetlag, obesity and metabolic disorder: investigation in a cohort study. Int J Obes. 2015;39:842–848.

    CAS  Google Scholar 

  98. Hung HC, Yang YC, Ou HY, Wu JS, Lu FH, Chang CJ. The relationship between impaired fasting glucose and self-reported sleep quality in a Chinese population. Clin Endocrinol. 2013;78:518–524.

    CAS  Google Scholar 

  99. Hung HC, Yang YC, Ou HY, Wu JS, Lu FH, Chang CJ. The association between self-reported sleep quality and overweight in a Chinese population. Obesity (Silver Spring, MD). 2013;21:486–492.

    Google Scholar 

  100. Tasali E, Leproult R, Ehrmann DA, Van Cauter E. Slow-wave sleep and the risk of type 2 diabetes in humans. Proc Natl Acad Sci USA. 2008;105:1044–1049.

    CAS  PubMed  PubMed Central  Google Scholar 

  101. Della Monica C, Johnsen S, Atzori G, Groeger JA, Dijk DJ. Rapid eye movement sleep, sleep continuity and slow wave sleep as predictors of cognition, mood, and subjective sleep quality in healthy men and women, aged 20–84 years. Front Psychiatry. 2018;9:255.

    PubMed  PubMed Central  Google Scholar 

  102. Stamatakis KA, Punjabi NM. Effects of sleep fragmentation on glucose metabolism in normal subjects. Chest. 2010;137:95–101.

    CAS  PubMed  Google Scholar 

  103. Hara R, Wan K, Wakamatsu H, et al. Restricted feeding entrains liver clock without participation of the suprachiasmatic nucleus. Genes Cells Devot Mol Cell Mech. 2001;6:269–278.

    CAS  Google Scholar 

  104. Birketvedt GS, Sundsfjord J, Florholmen JR. Hypothalamic–pituitary–adrenal axis in the night eating syndrome. Am J Physiol Endocrinol Metab. 2002;282:E366–E369.

    CAS  PubMed  Google Scholar 

  105. Gallant AR, Lundgren J, Drapeau V. The night-eating syndrome and obesity. Obes Rev. 2012;13:528–536.

    CAS  PubMed  Google Scholar 

  106. Stunkard AJ, Grace WJ, Wolff HG. The night-eating syndrome: a pattern of food intake among certain obese patients. Am J Med. 1955;19:78–86.

    CAS  PubMed  Google Scholar 

  107. Bernsmeier C, Weisskopf DM, Pflueger MO, et al. Sleep disruption and daytime sleepiness correlating with disease severity and insulin resistance in nonalcoholic fatty liver disease: a comparison with healthy controls. PLoS ONE. 2015;10:e0143293.

    PubMed  PubMed Central  Google Scholar 

  108. Salgado-Delgado RC, Saderi N, Basualdo Mdel C, Guerrero-Vargas NN, Escobar C, Buijs RM. Shift work or food intake during the rest phase promotes metabolic disruption and desynchrony of liver genes in male rats. PLoS ONE. 2013;8:e60052.

    CAS  PubMed  PubMed Central  Google Scholar 

  109. Salgado-Delgado R, Angeles-Castellanos M, Saderi N, Buijs RM, Escobar C. Food intake during the normal activity phase prevents obesity and circadian desynchrony in a rat model of night work. Endocrinology. 2010;151:1019–1029.

    CAS  PubMed  Google Scholar 

  110. Donga E, van Dijk M, van Dijk JG, et al. A single night of partial sleep deprivation induces insulin resistance in multiple metabolic pathways in healthy subjects. J Clin Endocrinol Metabol. 2010;95:2963–2968.

    CAS  Google Scholar 

  111. Shan Z, Ma H, Xie M, et al. Sleep duration and risk of type 2 diabetes: a meta-analysis of prospective studies. Diabetes Care. 2015;38:529–537.

    PubMed  Google Scholar 

  112. Lin D, Sun K, Li F, et al. Association between habitual daytime napping and metabolic syndrome: a population-based study. Metab Clin Exp. 2014;63:1520–1527.

    CAS  PubMed  Google Scholar 

  113. Fang W, Li Z, Wu L, et al. Longer habitual afternoon napping is associated with a higher risk for impaired fasting plasma glucose and diabetes mellitus in older adults: results from the Dongfeng–Tongji cohort of retired workers. Sleep Med. 2013;14:950–954.

    PubMed  Google Scholar 

  114. Baoying H, Hongjie C, Changsheng Q, et al. Association of napping and night-time sleep with impaired glucose regulation, insulin resistance and glycated haemoglobin in Chinese middle-aged adults with no diabetes: a cross-sectional study. BMJ Open. 2014;4:e004419.

    PubMed  PubMed Central  Google Scholar 

  115. Leite NC, Villela-Nogueira CA, Pannain VL, et al. Histopathological stages of nonalcoholic fatty liver disease in type 2 diabetes: prevalences and correlated factors. Liver Int. 2011;31:700–706.

    PubMed  Google Scholar 

  116. Lamia KA, Storch KF, Weitz CJ. Physiological significance of a peripheral tissue circadian clock. Proc Natl Acad Sci USA. 2008;105:15172–15177.

    CAS  PubMed  PubMed Central  Google Scholar 

  117. Kalsbeek A, La Fleur S, Van Heijningen C, Buijs RM. Suprachiasmatic GABAergic inputs to the paraventricular nucleus control plasma glucose concentrations in the rat via sympathetic innervation of the liver. J Neurosci. 2004;24:7604–7613.

    CAS  PubMed  PubMed Central  Google Scholar 

  118. Cailotto C, La Fleur SE, Van Heijningen C, et al. The suprachiasmatic nucleus controls the daily variation of plasma glucose via the autonomic output to the liver: are the clock genes involved? Eur J Neurosci. 2005;22:2531–2540.

    PubMed  Google Scholar 

  119. Rudic RD, McNamara P, Curtis AM, et al. BMAL1 and CLOCK, two essential components of the circadian clock, are involved in glucose homeostasis. PLoS Biol. 2004;2:e377.

    PubMed  PubMed Central  Google Scholar 

  120. Zhang EE, Liu Y, Dentin R, et al. Cryptochrome mediates circadian regulation of cAMP signaling and hepatic gluconeogenesis. Nat Med. 2010;16:1152–1156.

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Organization WH. Obesity and Overweight. 2016; http://www.who.int/mediacentre/factsheets/fs311/en/. Accessed September 26, 2017.

  122. Younossi ZM, Koenig AB, Abdelatif D, Fazel Y, Henry L, Wymer M. Global epidemiology of nonalcoholic fatty liver disease—meta-analytic assessment of prevalence, incidence, and outcomes. Hepatology (Baltimore, MD). 2016;64:73–84.

    Google Scholar 

  123. Hasler G, Buysse DJ, Klaghofer R, et al. The association between short sleep duration and obesity in young adults: a 13-year prospective study. Sleep. 2004;27:661–666.

    PubMed  Google Scholar 

  124. Vioque J, Torres A, Quiles J. Time spent watching television, sleep duration and obesity in adults living in Valencia, Spain. Int J Obes Relat Metab Disorders. 2000;24:1683–1688.

    CAS  Google Scholar 

  125. Kripke DF, Garfinkel L, Wingard DL, Klauber MR, Marler MR. Mortality associated with sleep duration and insomnia. Arch Gen Psychiatry. 2002;59:131–136.

    PubMed  Google Scholar 

  126. Schmid SM, Hallschmid M, Jauch-Chara K, et al. Short-term sleep loss decreases physical activity under free-living conditions but does not increase food intake under time-deprived laboratory conditions in healthy men. Am J Clin Nutr. 2009;90:1476–1482.

    CAS  PubMed  Google Scholar 

  127. Capers PL, Fobian AD, Kaiser KA, Borah R, Allison DB. A systematic review and meta-analysis of randomized controlled trials of the impact of sleep duration on adiposity and components of energy balance. Obes Rev. 2015;16:771–782.

    CAS  PubMed  PubMed Central  Google Scholar 

  128. Calvin AD, Carter RE, Adachi T, et al. Effects of experimental sleep restriction on caloric intake and activity energy expenditure. Chest. 2013;144:79–86.

    PubMed  PubMed Central  Google Scholar 

  129. Spaeth AM, Dinges DF, Goel N. Resting metabolic rate varies by race and by sleep duration. Obesity (Silver Spring, MD). 2015;23:2349–2356.

    Google Scholar 

  130. Karlsson B, Knutsson A, Lindahl B. Is there an association between shift work and having a metabolic syndrome? Results from a population based study of 27,485 people. Occup Environ Med. 2001;58:747–752.

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Samblas M, Milagro FI, Mansego ML, Marti A, Martinez JA. PTPRS and PER3 methylation levels are associated with childhood obesity: results from a genome-wide methylation analysis. Pediatr Obes. 2017;13:149–158.

    PubMed  Google Scholar 

  132. Angulo P. Nonalcoholic fatty liver disease. N Engl J Med. 2002;346:1221–1231.

    CAS  PubMed  Google Scholar 

  133. Ahmed MH, Byrne CD. Obstructive sleep apnea syndrome and fatty liver: association or causal link? World J Gastroenterol. 2010;16:4243–4252.

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Li Y, Xu C, Yu C, Xu L, Miao M. Association of serum uric acid level with nonalcoholic fatty liver disease: a cross-sectional study. J Hepatol. 2009;50:1029–1034.

    CAS  PubMed  Google Scholar 

  135. Setji TL, Holland ND, Sanders LL, Pereira KC, Diehl AM, Brown AJ. Nonalcoholic steatohepatitis and nonalcoholic fatty liver disease in young women with polycystic ovary syndrome. J Clin Endocrinol Metab. 2006;91:1741–1747.

    CAS  PubMed  Google Scholar 

  136. Hsieh SD, Muto T, Murase T, Tsuji H, Arase Y. Association of short sleep duration with obesity, diabetes, fatty liver and behavioral factors in Japanese men. Intern Med (Tokyo, Japan). 2011;50:2499–2502.

    Google Scholar 

  137. Imaizumi H, Takahashi A, Tanji N, et al. The association between sleep duration and nonalcoholic fatty liver disease among Japanese men and women. Obes Facts. 2015;8:234–242.

    PubMed  PubMed Central  Google Scholar 

  138. Trovato FM, Martines GF, Brischetto D, Catalano D, Musumeci G, Trovato GM. Fatty liver disease and lifestyle in youngsters: diet, food intake frequency, exercise, sleep shortage and fashion. Liver Int. 2016;36:427–433.

    CAS  PubMed  Google Scholar 

  139. Katsagoni CN, Georgoulis M, Papatheodoridis GV, et al. Associations between lifestyle characteristics and the presence of nonalcoholic fatty liver disease: a case–control study. Metab Syndr Relat Disorders. 2017;15:72–79.

    CAS  Google Scholar 

  140. Katsagoni CN, Papatheodoridis GV, Papageorgiou MV, et al. A “healthy diet-optimal sleep” lifestyle pattern is inversely associated with liver stiffness and insulin resistance in patients with nonalcoholic fatty liver disease. Appl Physiol Nutr Metab. 2017;42:250–256.

    CAS  PubMed  Google Scholar 

  141. Balakrishnan M, El-Serag HB, Kanwal F, Thrift AP. Shiftwork is not associated with increased risk of NAFLD: findings from the national health and nutrition examination survey. Dig Dis Sci. 2017;62:526–533.

    PubMed  Google Scholar 

  142. Bedogni G, Bellentani S, Miglioli L, et al. The Fatty Liver Index: a simple and accurate predictor of hepatic steatosis in the general population. BMC Gastroenterol. 2006;6:33.

    PubMed  PubMed Central  Google Scholar 

  143. Qu H, Wang H, Deng M, Wei H, Deng H. Associations between longer habitual day napping and nonalcoholic fatty liver disease in an elderly Chinese population. PLoS ONE. 2014;9:e105583.

    PubMed  PubMed Central  Google Scholar 

  144. Mwangi SM, Nezami BG, Obukwelu B, et al. Glial cell line-derived neurotrophic factor protects against high-fat diet-induced obesity. Am J Physiol Gastrointest Liver Physiol. 2014;306:G515–G525.

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Satoh Y, Kawai H, Kudo N, Kawashima Y, Mitsumoto A. Time-restricted feeding entrains daily rhythms of energy metabolism in mice. Am J Physiol Regul Integr Comp Physiol. 2006;290:R1276–R1283.

    CAS  PubMed  Google Scholar 

  146. Yanagihara H, Ando H, Hayashi Y, Obi Y, Fujimura A. High-fat feeding exerts minimal effects on rhythmic mRNA expression of clock genes in mouse peripheral tissues. Chronobiol Int. 2006;23:905–914.

    CAS  PubMed  Google Scholar 

  147. Bray MS, Young ME. Diurnal variations in myocardial metabolism. Cardiovasc Res. 2008;79:228–237.

    CAS  PubMed  Google Scholar 

  148. Kohsaka A, Laposky AD, Ramsey KM, et al. High-fat diet disrupts behavioral and molecular circadian rhythms in mice. Cell Metab. 2007;6:414–421.

    CAS  PubMed  Google Scholar 

  149. Hsieh MC, Yang SC, Tseng HL, Hwang LL, Chen CT, Shieh KR. Abnormal expressions of circadian-clock and circadian clock-controlled genes in the livers and kidneys of long-term, high-fat-diet-treated mice. Int J Obes. 2010;34:227–239.

    CAS  Google Scholar 

  150. Djouadi F, Weinheimer CJ, Saffitz JE, et al. A gender-related defect in lipid metabolism and glucose homeostasis in peroxisome proliferator-activated receptor alpha-deficient mice. J Clin Investig. 1998;102:1083–1091.

    CAS  PubMed  PubMed Central  Google Scholar 

  151. Patel DD, Knight BL, Wiggins D, Humphreys SM, Gibbons GF. Disturbances in the normal regulation of SREBP-sensitive genes in PPAR alpha-deficient mice. J Lipid Res. 2001;42:328–337.

    CAS  PubMed  Google Scholar 

  152. Montagner A, Polizzi A, Fouche E, et al. Liver PPARalpha is crucial for whole-body fatty acid homeostasis and is protective against NAFLD. Gut. 2016;65:1202–1214.

    CAS  PubMed  Google Scholar 

  153. Stienstra R, Mandard S, Patsouris D, Maass C, Kersten S, Muller M. Peroxisome proliferator-activated receptor alpha protects against obesity-induced hepatic inflammation. Endocrinology. 2007;148:2753–2763.

    CAS  PubMed  Google Scholar 

  154. Ip E, Farrell GC, Robertson G, Hall P, Kirsch R, Leclercq I. Central role of PPARalpha-dependent hepatic lipid turnover in dietary steatohepatitis in mice. Hepatology (Baltimore, MD). 2003;38:123–132.

    CAS  Google Scholar 

  155. Ip E, Farrell G, Hall P, Robertson G, Leclercq I. Administration of the potent PPARalpha agonist, Wy-14,643, reverses nutritional fibrosis and steatohepatitis in mice. Hepatology (Baltimore, MD). 2004;39:1286–1296.

    CAS  Google Scholar 

  156. Kettner NM, Voicu H, Finegold MJ, et al. Circadian homeostasis of liver metabolism suppresses hepatocarcinogenesis. Cancer Cell. 2016;30:909–924.

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Sateia MJ, Buysse DJ, Krystal AD, Neubauer DN, Heald JL. Clinical practice guideline for the pharmacologic treatment of chronic insomnia in adults: an american academy of sleep medicine clinical practice guideline. J Clin Sleep Med. 2017;13:307–349.

    PubMed  PubMed Central  Google Scholar 

  158. Irwin MR, Cole JC, Nicassio PM. Comparative meta-analysis of behavioral interventions for insomnia and their efficacy in middle-aged adults and in older adults 55+ years of age. Health Psychol. 2006;25:3–14.

    PubMed  Google Scholar 

  159. Srinivasan V, Singh J, Pandi-Perumal SR, Brown GM, Spence DW, Cardinali DP. Jet lag, circadian rhythm sleep disturbances, and depression: the role of melatonin and its analogs. Adv Therapy. 2010;27:796–813.

    CAS  Google Scholar 

  160. Grossman E, Laudon M, Zisapel N. Effect of melatonin on nocturnal blood pressure: meta-analysis of randomized controlled trials. Vasc Health Risk Manag. 2011;7:577–584.

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Goyal A, Terry PD, Superak HM, et al. Melatonin supplementation to treat the metabolic syndrome: a randomized controlled trial. Diabetol Metab Syndr. 2014;6:124.

    PubMed  PubMed Central  Google Scholar 

  162. Raygan F, Ostadmohammadi V, Bahmani F, Reiter RJ, Asemi Z. Melatonin administration lowers biomarkers of oxidative stress and cardio-metabolic risk in type 2 diabetic patients with coronary heart disease: a randomized, double-blind, placebo-controlled trial. Clin Nutr (Edinburgh, Scotland). 2017. https://doi.org/10.1016/j.clnu.2017.12.004.

    Article  Google Scholar 

  163. Gonciarz M, Gonciarz Z, Bielanski W, et al. The effects of long-term melatonin treatment on plasma liver enzymes levels and plasma concentrations of lipids and melatonin in patients with nonalcoholic steatohepatitis: a pilot study. J Physiol Pharmacol. 2012;63:35–40.

    CAS  PubMed  Google Scholar 

  164. Gonciarz M, Gonciarz Z, Bielanski W, et al. The pilot study of 3-month course of melatonin treatment of patients with nonalcoholic steatohepatitis: effect on plasma levels of liver enzymes, lipids and melatonin. J Physiol Pharmacol. 2010;61:705–710.

    CAS  PubMed  Google Scholar 

  165. Hirota T, Lee JW, St John PC, et al. Identification of small molecule activators of cryptochrome. Science (New York, NY). 2012;337:1094–1097.

    CAS  Google Scholar 

  166. Lee JW, Hirota T, Kumar A, Kim NJ, Irle S, Kay SA. Development of small-molecule cryptochrome stabilizer derivatives as modulators of the circadian clock. ChemMedChem. 2015;10:1489–1497.

    CAS  PubMed  PubMed Central  Google Scholar 

  167. Shah YM, Morimura K, Yang Q, Tanabe T, Takagi M, Gonzalez FJ. Peroxisome proliferator-activated receptor alpha regulates a microRNA-mediated signaling cascade responsible for hepatocellular proliferation. Mol Cell Biol. 2007;27:4238–4247.

    CAS  PubMed  PubMed Central  Google Scholar 

  168. Cariou B, Staels B. GFT505 for the treatment of nonalcoholic steatohepatitis and type 2 diabetes. Expert Opin Investig Drugs. 2014;23:1441–1448.

    CAS  PubMed  Google Scholar 

  169. Sahebkar A, Chew GT, Watts GF. New peroxisome proliferator-activated receptor agonists: potential treatments for atherogenic dyslipidemia and nonalcoholic fatty liver disease. Expert Opin Pharmacother. 2014;15:493–503.

    CAS  PubMed  Google Scholar 

Download references

Acknowledgments

Figures were created with “Biological illustration” (http://smart.servier.com) by Servier, used under Creative Commons Attribution 3.0 Unported License, and modified by Akshay Shetty and Paul Manka.

Funding

Resources were provided by the Ralph H Johnson VAMC, Charleston, South Carolina.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Wing-Kin Syn.

Ethics declarations

Conflict of interest

None.

Additional information

Disclaimer: Contents do not represent the views of the U.S. Department of Veterans Affairs or the United States Government.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shetty, A., Hsu, J.W., Manka, P.P. et al. Role of the Circadian Clock in the Metabolic Syndrome and Nonalcoholic Fatty Liver Disease. Dig Dis Sci 63, 3187–3206 (2018). https://doi.org/10.1007/s10620-018-5242-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s10620-018-5242-x

Keywords

Navigation