Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Review Article
  • Published:

Circadian clocks in the digestive system

Abstract

Many molecular, physiological and behavioural processes display distinct 24-hour rhythms that are directed by the circadian system. The master clock, located in the suprachiasmatic nucleus region of the hypothalamus, is synchronized or entrained by the light–dark cycle and, in turn, synchronizes clocks present in peripheral tissues and organs. Other environmental cues, most importantly feeding time, also synchronize peripheral clocks. In this way, the circadian system can prepare the body for predictable environmental changes such as the availability of nutrients during the normal feeding period. This Review summarizes existing knowledge about the diurnal regulation of gastrointestinal processes by circadian clocks present in the digestive tract and its accessory organs. The circadian control of gastrointestinal digestion, motility, hormones and barrier function as well as of the gut microbiota are discussed. An overview is given of the interplay between different circadian clocks in the digestive system that regulate glucose homeostasis and lipid and bile acid metabolism. Additionally, the bidirectional interaction between the master clock and peripheral clocks in the digestive system, encompassing different entraining factors, is described. Finally, the possible behavioural adjustments or pharmacological strategies for the prevention and treatment of the adverse effects of chronodisruption are outlined.

Key points

  • The circadian system controls diurnal rhythms in gastrointestinal digestion, absorption, motility, hormones, barrier function and the gut microbiota.

  • Human studies disentangling the influence of behavioural cycles (for example, feeding–fasting or activity–sleep) and of the endogenous clock are sparse.

  • Feeding time is the most important synchronizer of peripheral clocks; gut hormones, most notably insulin and IGF1, are essential to communicate feeding time phase information to peripheral clocks.

  • Time-restricted eating, timed exercise and chronobiotics — agents that alter the phase, amplitude or period of the circadian time system — hold promise for preventing or treating chronodisruption and its associated diseases.

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: Diurnal processes in the digestive system and acrophases of gut hormone and leptin levels.
Fig. 2: Circadian regulation of glucose homeostasis.
Fig. 3: Circadian regulation of bile acid production.
Fig. 4: Overview of brain clocks that ensure the circadian control of food intake.
Fig. 5: Hormones that potentially feedback phase information to the circadian system.

Similar content being viewed by others

References

  1. Dunlap, J. C. Molecular bases for circadian clocks. Cell 96, 271–290 (1999).

    Article  CAS  PubMed  Google Scholar 

  2. Balsalobre, A., Damiola, F. & Schibler, U. A serum shock induces circadian gene expression in mammalian tissue culture cells. Cell 93, 929–937 (1998). This study identified the existence of circadian clocks in peripheral tissues.

    Article  CAS  PubMed  Google Scholar 

  3. Stephan, F. K. & Zucker, I. Circadian rhythms in drinking behavior and locomotor activity of rats are eliminated by hypothalamic lesions. Proc. Natl Acad. Sci. USA 69, 1583–1586 (1972).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Ralph, M. R., Foster, R. G., Davis, F. C. & Menaker, M. Transplanted suprachiasmatic nucleus determines circadian period. Science 247, 975–978 (1990).

    Article  CAS  PubMed  Google Scholar 

  5. Mohawk, J. A., Green, C. B. & Takahashi, J. S. Central and peripheral circadian clocks in mammals. Annu. Rev. Neurosci. 35, 445–462 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Challet, E. The circadian regulation of food intake. Nat. Rev. Endocrinol. 15, 393–405 (2019).

    Article  PubMed  Google Scholar 

  7. Laermans, J. & Depoortere, I. Chronobesity: role of the circadian system in the obesity epidemic. Obes. Rev. 17, 108–125 (2016).

    Article  CAS  PubMed  Google Scholar 

  8. Nojkov, B., Rubenstein, J. H., Chey, W. D. & Hoogerwerf, W. A. The impact of rotating shift work on the prevalence of irritable bowel syndrome in nurses. Am. J. Gastroenterol. 105, 842–847 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  9. Knutsson, A. & Boggild, H. Gastrointestinal disorders among shift workers. Scand. J. Work Environ. Health 36, 85–95 (2010).

    Article  PubMed  Google Scholar 

  10. Crosby, P. et al. Insulin/IGF-1 drives PERIOD synthesis to entrain circadian rhythms with feeding time. Cell 177, 896–909.e20 (2019). This study provides strong evidence that entrainment by feeding time is dependent on the upregulation of PER2 via the activation of insulin/IGF1 receptors.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Ribas-Latre, A. & Eckel-Mahan, K. Interdependence of nutrient metabolism and the circadian clock system: Importance for metabolic health. Mol. Metab. 5, 133–152 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Froy, O. Metabolism and circadian rhythms–implications for obesity. Endocr. Rev. 31, 1–24 (2010).

    Article  CAS  PubMed  Google Scholar 

  13. Mukherji, A., Kobiita, A., Ye, T. & Chambon, P. Homeostasis in intestinal epithelium is orchestrated by the circadian clock and microbiota cues transduced by TLRs. Cell 153, 812–827 (2013).

    Article  CAS  PubMed  Google Scholar 

  14. Weger, B. D. et al. The mouse microbiome is required for sex-specific diurnal rhythms of gene expression and metabolism. Cell Metab. 29, 362–382.e8 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Perelis, M. et al. Pancreatic beta cell enhancers regulate rhythmic transcription of genes controlling insulin secretion. Science 350, aac4250 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Dardente, H. et al. Daily and circadian expression of neuropeptides in the suprachiasmatic nuclei of nocturnal and diurnal rodents. Brain Res. Mol. Brain Res. 124, 143–151 (2004).

    Article  CAS  PubMed  Google Scholar 

  17. Kumar Jha, P., Challet, E. & Kalsbeek, A. Circadian rhythms in glucose and lipid metabolism in nocturnal and diurnal mammals. Mol. Cell. Endocrinol. 418 Pt 1, 74–88 (2015).

    Article  CAS  PubMed  Google Scholar 

  18. Dawes, C. Circadian rhythms in human salivary flow rate and composition. J. Physiol. 220, 529–545 (1972).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Zheng, L., Seon, Y. J., McHugh, J., Papagerakis, S. & Papagerakis, P. Clock genes show circadian rhythms in salivary glands. J. Dent. Res. 91, 783–788 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Satou, R. et al. Temporal expression patterns of clock genes and aquaporin 5/anoctamin 1 in rat submandibular gland cells. Front Physiol 8, 320 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  21. Furukawa, M. et al. Clock gene expression in the submandibular glands. J. Dent. Res. 84, 1193–1197 (2005).

    Article  CAS  PubMed  Google Scholar 

  22. Moore, J. G. & Halberg, F. Circadian rhythm of gastric acid secretion in men with active duodenal ulcer. Dig. Dis. Sci. 31, 1185–1191 (1986).

    Article  CAS  PubMed  Google Scholar 

  23. Rogers, M. J., Holmfield, J. H., Primrose, J. N., Gledhill, T. & Johnston, D. A prospective comparison of the effects of placebo, ranitidine and highly selective vagotomy on 24 h ambulatory intragastric pH in patients with duodenal ulcer. Br. J. Surg. 75, 961–965 (1988).

    Article  CAS  PubMed  Google Scholar 

  24. Barattini, P., Larsen, K. R., Moore, J. G. & Dayton, M. T. Circadian rhythm of pepsin efflux in the fasting rat stomach. Chronobiol. Int. 10, 403–409 (1993).

    Article  CAS  PubMed  Google Scholar 

  25. Semple, J., Newton, J., Westley, B. & May, F. Dramatic diurnal variation in the concentration of the human trefoil peptide TFF2 in gastric juice. Gut 48, 648–655 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Saito, M., Murakami, E. & Suda, M. Circadian rhythms in disaccharidases of rat small intestine and its relation to food intake. Biochim. Biophys. Acta 421, 177–179 (1976).

    Article  CAS  PubMed  Google Scholar 

  27. Hara, E. & Saito, M. Diurnal change in digestion and absorption of sucrose in vivo in rats. J. Nutr. Sci. Vitaminol. 35, 667–671 (1989).

    Article  CAS  PubMed  Google Scholar 

  28. Stevenson, N. R., Ferrigni, F., Parnicky, K., Day, S. & Fierstein, J. S. Effect of changes in feeding schedule on the diurnal rhythms and daily activity levels of intestinal brush border enzymes and transport systems. Biochim. Biophys. Acta 406, 131–145 (1975).

    Article  CAS  PubMed  Google Scholar 

  29. Stevenson, N. R., Sitren, H. S. & Furuya, S. Circadian rhythmicity in several small intestinal functions is independent of use of the intestine. Am. J. Physiol. 238, G203–G207 (1980).

    CAS  PubMed  Google Scholar 

  30. Keller, J. & Layer, P. Circadian pancreatic enzyme pattern and relationship between secretory and motor activity in fasting humans. J. Appl. Physiol. 93, 592–600 (2002).

    Article  CAS  PubMed  Google Scholar 

  31. Maouyo, D., Sarfati, P., Guan, D., Morisset, J. & Adelson, J. W. Circadian rhythm of exocrine pancreatic secretion in rats: major and minor cycles. Am. J. Physiol. 264, G792–G800 (1993).

    CAS  PubMed  Google Scholar 

  32. Oishi, K. et al. Disrupted fat absorption attenuates obesity induced by a high-fat diet in Clock mutant mice. FEBS Lett. 580, 127–130 (2006).

    Article  CAS  PubMed  Google Scholar 

  33. Iwashina, I., Mochizuki, K., Inamochi, Y. & Goda, T. Clock genes regulate the feeding schedule-dependent diurnal rhythm changes in hexose transporter gene expressions through the binding of BMAL1 to the promoter/enhancer and transcribed regions. J. Nutr. Biochem. 22, 334–343 (2011).

    Article  CAS  PubMed  Google Scholar 

  34. Pan, X. & Hussain, M. M. Clock is important for food and circadian regulation of macronutrient absorption in mice. J. Lipid Res. 50, 1800–1813 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Balakrishnan, A., Stearns, A. T., Ashley, S. W., Rhoads, D. B. & Tavakkolizadeh, A. PER1 modulates SGLT1 transcription in vitro independent of E-box status. Dig. Dis. Sci. 57, 1525–1536 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Saito, H., Terada, T., Shimakura, J., Katsura, T. & Inui, K. Regulatory mechanism governing the diurnal rhythm of intestinal H+/peptide cotransporter 1 (PEPT1). Am. J. Physiol. Gastrointest. Liver Physiol. 295, G395–G402 (2008).

    Article  CAS  PubMed  Google Scholar 

  37. Pan, X., Terada, T., Irie, M., Saito, H. & Inui, K. Diurnal rhythm of H+-peptide cotransporter in rat small intestine. Am. J. Physiol. Gastrointest. Liver Physiol. 283, G57–G64 (2002).

    Article  CAS  PubMed  Google Scholar 

  38. Pan, X., Terada, T., Okuda, M. & Inui, K. The diurnal rhythm of the intestinal transporters SGLT1 and PEPT1 is regulated by the feeding conditions in rats. J. Nutr. 134, 2211–2215 (2004).

    Article  CAS  PubMed  Google Scholar 

  39. Okamura, A. et al. Bile acid-regulated peroxisome proliferator-activated receptor-alpha (PPARα) activity underlies circadian expression of intestinal peptide absorption transporter PepT1/Slc15a1. J. Biol. Chem. 289, 25296–25305 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Lavery, D. J. & Schibler, U. Circadian transcription of the cholesterol 7 alpha hydroxylase gene may involve the liver-enriched bZIP protein DBP. Genes Dev. 7, 1871–1884 (1993).

    Article  CAS  PubMed  Google Scholar 

  41. Le Martelot, G. et al. REV-ERBalpha participates in circadian SREBP signaling and bile acid homeostasis. PLoS Biol. 7, e1000181 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  42. Han, S. et al. Circadian control of bile acid synthesis by a KLF15-Fgf15 axis. Nat. Commun. 6, 7231 (2015).

    Article  CAS  PubMed  Google Scholar 

  43. Duez, H. et al. Regulation of bile acid synthesis by the nuclear receptor REV-ERBα. Gastroenterology 135, 689–698 (2008).

    Article  CAS  PubMed  Google Scholar 

  44. Pan, X., Zhang, Y., Wang, L. & Hussain, M. M. Diurnal regulation of MTP and plasma triglyceride by CLOCK is mediated by SHP. Cell Metab. 12, 174–186 (2010). This study identifies the role of CLOCK–SHP in the diurnal regulation of MTP and plasma triglyceride and indicates that disruptions in circadian regulation might cause hyperlipidaemia.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Pan, X., Jiang, X. C. & Hussain, M. M. Impaired cholesterol metabolism and enhanced atherosclerosis in clock mutant mice. Circulation 128, 1758–1769 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Stein, H. J., Singh, S. & Demeester, T. R. ‘Efficacy’ of esophageal peristalsis: a manometric parameter to quantify esophageal body dysfunction. Dis. Esophagus. 17, 297–303 (2004).

    Article  CAS  PubMed  Google Scholar 

  47. Chitkara, D. K., Fortunato, C. & Nurko, S. Prolonged monitoring of esophageal motor function in healthy children. J. Pediatr. Gastroenterol. Nutr. 38, 192–197 (2004).

    Article  PubMed  Google Scholar 

  48. Goo, R. H., Moore, J. G., Greenberg, E. & Alazraki, N. P. Circadian variation in gastric emptying of meals in humans. Gastroenterology 93, 515–518 (1987).

    Article  CAS  PubMed  Google Scholar 

  49. Grammaticos, P. C., Doumas, A. & Koliakos, G. Morning and night gastric emptying half-time differed more than 220% in two young healthy adults. Hell. J. Nucl. Med. 18, 60–62 (2015).

    PubMed  Google Scholar 

  50. Kumar, D., Wingate, D. & Ruckebusch, Y. Circadian variation in the propagation velocity of the migrating motor complex. Gastroenterology 91, 926–930 (1986).

    Article  CAS  PubMed  Google Scholar 

  51. Narducci, F., Bassotti, G., Gaburri, M. & Morelli, A. Twenty four hour manometric recording of colonic motor activity in healthy man. Gut 28, 17–25 (1987).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Rao, S. S., Sadeghi, P., Beaty, J., Kavlock, R. & Ackerson, K. Ambulatory 24-h colonic manometry in healthy humans. Am. J. Physiol. Gastrointest. Liver. Physiol. 280, G629–G639 (2001).

    Article  CAS  PubMed  Google Scholar 

  53. Auwerda, J. J., Bac, D. J. & Schouten, W. R. Circadian rhythm of rectal motor complexes. Dis. Colon. Rectum. 44, 1328–1332 (2001).

    Article  CAS  PubMed  Google Scholar 

  54. Hung, J.-S., Liu, T.-T., Yi, C.-H., Lei, W.-Y. & Chen, C.-L. Altered anorectal function in rotating shift workers: Association with autonomic dysfunction and sleep disturbance. Adv. Digest. Med. 3, 80–87 (2016).

    Article  Google Scholar 

  55. Yamaguchi, M. et al. Circadian rhythm genes CLOCK and PER3 polymorphisms and morning gastric motility in humans. PLoS ONE 10, e0120009 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Yamaguchi, M. et al. The CLOCK 3111T/C single nucleotide polymorphism and daytime fluctuations of gastric motility in healthy young women: a preliminary study. Chronobiol. Int. 34, 1478–1482 (2017).

    Article  CAS  PubMed  Google Scholar 

  57. Hoogerwerf, W. A. et al. Rhythmic changes in colonic motility are regulated by period genes. Am. J. Physiol. Gastrointest. Liver Physiol. 298, G143–G150 (2010).

    Article  CAS  PubMed  Google Scholar 

  58. Hoogerwerf, W. A. et al. Transcriptional profiling of mRNA expression in the mouse distal colon. Gastroenterology 135, 2019–2029 (2008).

    Article  CAS  PubMed  Google Scholar 

  59. Hoogerwerf, W. A. Role of clock genes in gastrointestinal motility. Am. J. Physiol. Gastrointest. Liver Physiol. 299, G549–G555 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Kentish, S. J., Frisby, C. L., Kennaway, D. J., Wittert, G. A. & Page, A. J. Circadian variation in gastric vagal afferent mechanosensitivity. J. Neurosci. 33, 19238–19242 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Laermans, J. et al. Shifting the circadian rhythm of feeding in mice induces gastrointestinal, metabolic and immune alterations which are influenced by ghrelin and the core clock gene Bmal1. PLoS ONE 9, e110176 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  62. Cummings, D. E. et al. A preprandial rise in plasma ghrelin levels suggests a role in meal initiation in humans. Diabetes 50, 1714–1719 (2001).

    Article  CAS  PubMed  Google Scholar 

  63. Schiavo-Cardozo, D., Lima, M. M., Pareja, J. C. & Geloneze, B. Appetite-regulating hormones from the upper gut: disrupted control of xenin and ghrelin in night workers. Clin. Endocrinol. 79, 807–811 (2013).

    Article  CAS  Google Scholar 

  64. Qian, J., Morris, C. J., Caputo, R., Garaulet, M. & Scheer, F. Ghrelin is impacted by the endogenous circadian system and by circadian misalignment in humans. Int. J. Obes. 43, 1644–1649 (2019).

    Article  CAS  Google Scholar 

  65. Yildiz, B. O., Suchard, M. A., Wong, M. L., McCann, S. M. & Licinio, J. Alterations in the dynamics of circulating ghrelin, adiponectin, and leptin in human obesity. Proc. Natl Acad. Sci. USA 101, 10434–10439 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Goel, N. et al. Circadian rhythm profiles in women with night eating syndrome. J. Biol. Rhythms 24, 85–94 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Shiiya, T. et al. Plasma ghrelin levels in lean and obese humans and the effect of glucose on ghrelin secretion. J. Clin. Endocrinol. Metab. 87, 240–244 (2002).

    Article  CAS  PubMed  Google Scholar 

  68. Laermans, J., Vancleef, L., Tack, J. & Depoortere, I. Role of the clock gene Bmal1 and the gastric ghrelin-secreting cell in the circadian regulation of the ghrelin-GOAT system. Sci. Rep. 5, 16748 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. LeSauter, J., Hoque, N., Weintraub, M., Pfaff, D. W. & Silver, R. Stomach ghrelin-secreting cells as food-entrainable circadian clocks. Proc. Natl Acad. Sci. USA 106, 13582–13587 (2009). This study demonstrates that ghrelin can modulate food-anticipatory activity and might act as an FEO.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Müller, T. D. et al. Glucagon-like peptide 1 (GLP-1). Mol. Metab. 30, 72–130 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  71. Elliott, R. M. et al. Glucagon-like peptide-1 (7-36) amide and glucose-dependent insulinotropic polypeptide secretion in response to nutrient ingestion in man: acute post-prandial and 24-h secretion patterns. J. Endocrinol. 138, 159–166 (1993).

    Article  CAS  PubMed  Google Scholar 

  72. Galindo Munoz, J. S., Jimenez Rodriguez, D. & Hernandez Morante, J. J. Diurnal rhythms of plasma GLP-1 levels in normal and overweight/obese subjects: lack of effect of weight loss. J. Physiol. Biochem. 71, 17–28 (2015).

    Article  PubMed  Google Scholar 

  73. Mingrone, G. et al. Circadian rhythms of GIP and GLP1 in glucose-tolerant and in type 2 diabetic patients after biliopancreatic diversion. Diabetologia 52, 873–881 (2009).

    Article  CAS  PubMed  Google Scholar 

  74. Gil-Lozano, M. et al. Short-term sleep deprivation with nocturnal light exposure alters time-dependent glucagon-like peptide-1 and insulin secretion in male volunteers. Am. J. Physiol. Endocrinol. Metab. 310, E41–E50 (2016).

    Article  PubMed  Google Scholar 

  75. Hampton, S. M. et al. Postprandial hormone and metabolic responses in simulated shift work. J. Endocrinol. 151, 259–267 (1996).

    Article  CAS  PubMed  Google Scholar 

  76. Gil-Lozano, M., Wu, W. K., Martchenko, A. & Brubaker, P. L. High-fat diet and palmitate alter the rhythmic secretion of glucagon-like peptide-1 by the rodent L-cell. Endocrinology 157, 586–599 (2016).

    Article  CAS  PubMed  Google Scholar 

  77. Gil-Lozano, M., Mingomataj, E. L., Wu, W. K., Ridout, S. A. & Brubaker, P. L. Circadian secretion of the intestinal hormone GLP-1 by the rodent L cell. Diabetes 63, 3674–3685 (2014).

    Article  CAS  PubMed  Google Scholar 

  78. Hill, B. R., De Souza, M. J. & Williams, N. I. Characterization of the diurnal rhythm of peptide YY and its association with energy balance parameters in normal-weight premenopausal women. Am. J. Physiol. Endocrinol. Metab. 301, E409–E415 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Scheer, F. A., Hilton, M. F., Mantzoros, C. S. & Shea, S. A. Adverse metabolic and cardiovascular consequences of circadian misalignment. Proc. Natl Acad. Sci. USA 106, 4453–4458 (2009). This study showed that misalignment between the behavioural rhythm (feeding–fasting and sleep–wake cycles) and the endogenous circadian rhythm decreased leptin, increased glucose despite increased insulin, inverted the daily cortisol rhythm, increased mean arterial pressure and reduced sleep efficiency.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Schoeller, D. A., Cella, L. K., Sinha, M. K. & Caro, J. F. Entrainment of the diurnal rhythm of plasma leptin to meal timing. J. Clin. Invest. 100, 1882–1887 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Kalsbeek, A. et al. The suprachiasmatic nucleus generates the diurnal changes in plasma leptin levels. Endocrinology 142, 2677–2685 (2001).

    Article  CAS  PubMed  Google Scholar 

  82. Kettner, N. M. et al. Circadian dysfunction induces leptin resistance in mice. Cell. Metab. 22, 448–459 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Stenvers, D. J., Scheer, F., Schrauwen, P., la Fleur, S. E. & Kalsbeek, A. Circadian clocks and insulin resistance. Nat. Rev. Endocrinol. 15, 75–89 (2019).

    Article  PubMed  Google Scholar 

  84. Scheving, L. A. Biological clocks and the digestive system. Gastroenterology 119, 536–549 (2000).

    Article  CAS  PubMed  Google Scholar 

  85. Hoogerwerf, W. A. Role of biological rhythms in gastrointestinal health and disease. Rev. Endocr. Metab. Disord. 10, 293–300 (2009).

    Article  PubMed  Google Scholar 

  86. Gachon, F., Loizides-Mangold, U., Petrenko, V. & Dibner, C. Glucose homeostasis: regulation by peripheral circadian clocks in rodents and humans. Endocrinology 158, 1074–1084 (2017).

    Article  CAS  PubMed  Google Scholar 

  87. Petrenko, V. et al. Pancreatic alpha- and beta-cellular clocks have distinct molecular properties and impact on islet hormone secretion and gene expression. Genes Dev. 31, 383–398 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Boden, G., Ruiz, J., Urbain, J. L. & Chen, X. Evidence for a circadian rhythm of insulin secretion. Am. J. Physiol. 271, E246–E252 (1996).

    CAS  PubMed  Google Scholar 

  89. Ruiter, M. et al. The daily rhythm in plasma glucagon concentrations in the rat is modulated by the biological clock and by feeding behavior. Diabetes 52, 1709–1715 (2003).

    Article  CAS  PubMed  Google Scholar 

  90. Hodge, B. A. et al. The endogenous molecular clock orchestrates the temporal separation of substrate metabolism in skeletal muscle. Skelet. Muscle 5, 17 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  91. Doi, R., Oishi, K. & Ishida, N. CLOCK regulates circadian rhythms of hepatic glycogen synthesis through transcriptional activation of Gys2. J. Biol. Chem. 285, 22114–22121 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Reinke, H. & Asher, G. Circadian clock control of liver metabolic functions. Gastroenterology 150, 574–580 (2016).

    Article  PubMed  Google Scholar 

  93. Zhang, E. E. et al. Cryptochrome mediates circadian regulation of camp signaling and hepatic gluconeogenesis. Nat. Med. 16, 1152–1156 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Reinke, H. & Asher, G. Crosstalk between metabolism and circadian clocks. Nat. Rev. Mol. Cell Biol. 20, 227–241 (2019).

    Article  CAS  PubMed  Google Scholar 

  95. Gooley, J. J. Circadian regulation of lipid metabolism. Proc. Nutr. Soc. 75, 440–450 (2016).

    Article  CAS  PubMed  Google Scholar 

  96. Dallmann, R., Viola, A. U., Tarokh, L., Cajochen, C. & Brown, S. A. The human circadian metabolome. Proc. Natl Acad. Sci. USA 109, 2625–2629 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Chua, E. C. et al. Extensive diversity in circadian regulation of plasma lipids and evidence for different circadian metabolic phenotypes in humans. Proc. Natl Acad. Sci. USA 110, 14468–14473 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Loizides-Mangold, U. et al. Lipidomics reveals diurnal lipid oscillations in human skeletal muscle persisting in cellular myotubes cultured in vitro. Proc. Natl Acad. Sci. USA 114, E8565–E8574 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  99. Duane, W. C., Levitt, D. G., Mueller, S. M. & Behrens, J. C. Regulation of bile acid synthesis in man. Presence of a diurnal rhythm. J. Clin. Invest. 72, 1930–1936 (1983).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Kuipers, F., Bloks, V. W. & Groen, A. K. Beyond intestinal soap–bile acids in metabolic control. Nat. Rev. Endocrinol. 10, 488–498 (2014).

    Article  CAS  PubMed  Google Scholar 

  101. Campos, H., Khoo, C. & Sacks, F. M. Diurnal and acute patterns of postprandial apolipoprotein B-48 in VLDL, IDL, and LDL from normolipidemic humans. Atherosclerosis 181, 345–351 (2005).

    Article  CAS  PubMed  Google Scholar 

  102. Raspe, E. et al. Identification of Rev-Erbα as a physiological repressor of ApoC-III gene transcription. J. Lipid. Res. 43, 2172–2179 (2002).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Shostak, A., Meyer-Kovac, J. & Oster, H. Circadian regulation of lipid mobilization in white adipose tissues. Diabetes 62, 2195–2203 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Gombert, M., Carrasco-Luna, J., Pin-Arboledas, G. & Codoner-Franch, P. The connection of circadian rhythm to inflammatory bowel disease. Transl. Res. 206, 107–118 (2019).

    Article  PubMed  Google Scholar 

  106. Mazzoccoli, G. et al. Association study of a polymorphism in clock gene PERIOD3 and risk of inflammatory bowel disease. Chronobiol. Int. 29, 994–1003 (2012).

    Article  CAS  PubMed  Google Scholar 

  107. Palmieri, O. et al. Systematic analysis of circadian genes using genome-wide cDNA microarrays in the inflammatory bowel disease transcriptome. Chronobiol. Int. 32, 903–916 (2015).

    Article  PubMed  Google Scholar 

  108. Kyoko, O. O. et al. Expressions of tight junction proteins Occludin and Claudin-1 are under the circadian control in the mouse large intestine: implications in intestinal permeability and susceptibility to colitis. PLoS ONE 9, e98016 (2014).

    Article  PubMed  Google Scholar 

  109. Summa, K. C. et al. Disruption of the circadian clock in mice increases intestinal permeability and promotes alcohol-induced hepatic pathology and inflammation. PLoS ONE 8, e67102 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  110. Deaver, J. A., Eum, S. Y. & Toborek, M. Circadian disruption changes gut microbiome taxa and functional gene composition. Front. Microbiol. 9, 737 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  111. Pagel, R. et al. Circadian rhythm disruption impairs tissue homeostasis and exacerbates chronic inflammation in the intestine. FASEB J. 31, 4707–4719 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  112. Preuss, F. et al. Adverse effects of chronic circadian desynchronization in animals in a “challenging” environment. Am. J. Physiol. Regul. Integr. Comp. Physiol. 295, R2034–R2040 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Tang, Y., Preuss, F., Turek, F. W., Jakate, S. & Keshavarzian, A. Sleep deprivation worsens inflammation and delays recovery in a mouse model of colitis. Sleep Med. 10, 597–603 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  114. Tuganbaev, T. et al. Diet diurnally regulates small intestinal microbiome-epithelial-immune homeostasis and enteritis. Cell 182, 1441–1459.e21 (2020).

    Article  CAS  PubMed  Google Scholar 

  115. Thaiss, C. A. et al. Transkingdom control of microbiota diurnal oscillations promotes metabolic homeostasis. Cell 159, 514–529 (2014). This study demonstrates that clock disruption causes loss of diurnal fluctuations in microbiota and that this arrhythmic microbiota might contribute to the host’s metabolic disturbances.

    Article  CAS  PubMed  Google Scholar 

  116. Zarrinpar, A., Chaix, A., Yooseph, S. & Panda, S. Diet and feeding pattern affect the diurnal dynamics of the gut microbiome. Cell Metab. 20, 1006–1017 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Liang, X., Bushman, F. D. & FitzGerald, G. A. Rhythmicity of the intestinal microbiota is regulated by gender and the host circadian clock. Proc. Natl Acad. Sci. USA 112, 10479–10484 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  118. Paulose, J. K., Wright, J. M., Patel, A. G. & Cassone, V. M. Human gut bacteria are sensitive to melatonin and express endogenous circadian rhythmicity. PLoS ONE 11, e0146643 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  119. Leone, V. et al. Effects of diurnal variation of gut microbes and high-fat feeding on host circadian clock function and metabolism. Cell Host Microbe 17, 681–689 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Tahara, Y. et al. Gut microbiota-derived short chain fatty acids induce circadian clock entrainment in mouse peripheral tissue. Sci. Rep. 8, 1395 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  121. Parkar, S. G., Kalsbeek, A. & Cheeseman, J. F. Potential role for the gut microbiota in modulating host circadian rhythms and metabolic health. Microorganisms 7, 41 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  122. Segers, A. et al. The circadian clock regulates the diurnal levels of microbial short-chain fatty acids and their rhythmic effects on colon contractility in mice. Acta Physiol. 225, e13193 (2018).

    Google Scholar 

  123. Segers, A. et al. Night-time feeding of Bmal1-/- mice restores SCFA rhythms and their effect on ghrelin. J. Endocrinol. 245, 155–164 (2020).

    Article  CAS  PubMed  Google Scholar 

  124. Astiz, M., Heyde, I. & Oster, H. Mechanisms of communication in the mammalian circadian timing system. Int. J. Mol. Sci. 20, 343 (2019).

    Article  PubMed Central  Google Scholar 

  125. Kalsbeek, A., Teclemariam-Mesbah, R. & Pevet, P. Efferent projections of the suprachiasmatic nucleus in the golden hamster (Mesocricetus auratus). J. Comp. Neurol. 332, 293–314 (1993).

    Article  CAS  PubMed  Google Scholar 

  126. Buijs, R. M. et al. The suprachiasmatic nucleus balances sympathetic and parasympathetic output to peripheral organs through separate preautonomic neurons. J. Comp. Neurol. 464, 36–48 (2003).

    Article  PubMed  Google Scholar 

  127. Ishida, A. et al. Light activates the adrenal gland: timing of gene expression and glucocorticoid release. Cell Metab. 2, 297–307 (2005).

    Article  CAS  PubMed  Google Scholar 

  128. Tordjman, S. et al. Melatonin: pharmacology, functions and therapeutic benefits. Curr. Neuropharmacol. 15, 434–443 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Herxheimer, A. & Petrie, K. J. Melatonin for the prevention and treatment of jet lag. Cochrane Database Syst. Rev. 2, CD001520 (2002).

    Google Scholar 

  130. Liira, J. et al. Pharmacological interventions for sleepiness and sleep disturbances caused by shift work. Cochrane Database Syst. Rev. 8, CD009776 (2014).

    Google Scholar 

  131. Zawilska, J. B., Skene, D. J. & Arendt, J. Physiology and pharmacology of melatonin in relation to biological rhythms. Pharmacol. Rep. 61, 383–410 (2009).

    Article  CAS  PubMed  Google Scholar 

  132. Chen, C. Q., Fichna, J., Bashashati, M., Li, Y. Y. & Storr, M. Distribution, function and physiological role of melatonin in the lower gut. World J. Gastroenterol. 17, 3888–3898 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  133. Söderquist, F., Hellström, P. M. & Cunningham, J. L. Human gastroenteropancreatic expression of melatonin and its receptors MT1 and MT2. PLoS ONE 10, e0120195 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  134. Campino, C. et al. Melatonin exerts direct inhibitory actions on ACTH responses in the human adrenal gland. Horm. Metab. Res. 43, 337–342 (2011).

    Article  CAS  PubMed  Google Scholar 

  135. Cuesta, M., Cermakian, N. & Boivin, D. B. Glucocorticoids entrain molecular clock components in human peripheral cells. FASEB J. 29, 1360–1370 (2015).

    Article  CAS  PubMed  Google Scholar 

  136. Moore, R. Y. & Eichler, V. B. Loss of a circadian adrenal corticosterone rhythm following suprachiasmatic lesions in the rat. Brain Res. 42, 201–206 (1972).

    Article  CAS  PubMed  Google Scholar 

  137. Oster, H. et al. The functional and clinical significance of the 24-hour rhythm of circulating glucocorticoids. Endocr. Rev. 38, 3–45 (2017).

    Article  PubMed  Google Scholar 

  138. Aschoff, J., von Goetz, C., Wildgruber, C. & Wever, R. A. Meal timing in humans during isolation without time cues. J. Biol. Rhythms 1, 151–162 (1986).

    Article  CAS  PubMed  Google Scholar 

  139. Bechtold, D. A. & Loudon, A. S. Hypothalamic clocks and rhythms in feeding behaviour. Trends Neurosci. 36, 74–82 (2013).

    Article  CAS  PubMed  Google Scholar 

  140. Mistlberger, R. E. Food-anticipatory circadian rhythms: concepts and methods. Eur. J. Neurosci. 30, 1718–1729 (2009).

    Article  PubMed  Google Scholar 

  141. Mistlberger, R. E. Neurobiology of food anticipatory circadian rhythms. Physiol. Behav. 104, 535–545 (2011).

    Article  CAS  PubMed  Google Scholar 

  142. Davidson, A. J. Lesion studies targeting food-anticipatory activity. Eur. J. Neurosci. 30, 1658–1664 (2009).

    Article  PubMed  Google Scholar 

  143. Guan, D. et al. The hepatocyte clock and feeding control chronophysiology of multiple liver cell types. Science 369, 1388–1394 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Wang, Q., Yin, Y. & Zhang, W. Ghrelin restores the disruption of the circadian clock in steatotic liver. Int. J. Mol. Sci. 19, 3134 (2018).

    Article  PubMed Central  Google Scholar 

  145. Yannielli, P. C., Molyneux, P. C., Harrington, M. E. & Golombek, D. A. Ghrelin effects on the circadian system of mice. J. Neurosci. 27, 2890–2895 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Landgraf, D. et al. Oxyntomodulin regulates resetting of the liver circadian clock by food. eLife 4, e06253 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  147. Ando, H., Ushijima, K. & Fujimura, A. Indirect effects of glucagon-like peptide-1 receptor agonist exendin-4 on the peripheral circadian clocks in mice. PLoS ONE 8, e81119 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  148. Gribkoff, V. K., Pieschl, R. L., Wisialowski, T. A., van den Pol, A. N. & Yocca, F. D. Phase shifting of circadian rhythms and depression of neuronal activity in the rat suprachiasmatic nucleus by neuropeptide Y: mediation by different receptor subtypes. J. Neurosci. 18, 3014–3022 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Edelsbrunner, M. E., Herzog, H. & Holzer, P. Evidence from knockout mice that peptide YY and neuropeptide Y enforce murine locomotion, exploration and ingestive behaviour in a circadian cycle- and gender-dependent manner. Behav. Brain Res. 203, 97–107 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  150. Ikeda, Y. et al. Glucagon and/or IGF-1 production regulates resetting of the liver circadian clock in response to a protein or amino acid-only diet. EBioMedicine 28, 210–224 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  151. Sun, X. et al. Glucagon-CREB/CRTC2 signaling cascade regulates hepatic BMAL1 protein. J. Biol. Chem. 290, 2189–2197 (2015).

    Article  CAS  PubMed  Google Scholar 

  152. Mukherji, A., Kobiita, A. & Chambon, P. Shifting the feeding of mice to the rest phase creates metabolic alterations, which, on their own, shift the peripheral circadian clocks by 12 hours. Proc. Natl Acad. Sci. USA 112, E6683–E6690 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  153. Mukherji, A. et al. Shifting eating to the circadian rest phase misaligns the peripheral clocks with the master SCN clock and leads to a metabolic syndrome. Proc. Natl Acad. Sci. USA 112, E6691–E6698 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  154. Mistlberger, R. E. & Marchant, E. G. Enhanced food-anticipatory circadian rhythms in the genetically obese Zucker rat. Physiol. Behav. 66, 329–335 (1999).

    Article  CAS  PubMed  Google Scholar 

  155. Ribeiro, A. C. et al. Contrasting effects of leptin on food anticipatory and total locomotor activity. PLoS ONE 6, e23364 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  156. Ando, H. et al. Impairment of peripheral circadian clocks precedes metabolic abnormalities in ob/ob mice. Endocrinology 152, 1347–1354 (2011).

    Article  CAS  PubMed  Google Scholar 

  157. Fu, L., Patel, M. S., Bradley, A., Wagner, E. F. & Karsenty, G. The molecular clock mediates leptin-regulated bone formation. Cell 122, 803–815 (2005).

    Article  CAS  PubMed  Google Scholar 

  158. Vieira, E. et al. The clock gene Rev-Erbα regulates pancreatic beta-cell function: modulation by leptin and high-fat diet. Endocrinology 153, 592–601 (2012).

    Article  CAS  PubMed  Google Scholar 

  159. Prosser, R. A. & Bergeron, H. E. Leptin phase-advances the rat suprachiasmatic circadian clock in vitro. Neurosci. Lett. 336, 139–142 (2003).

    Article  CAS  PubMed  Google Scholar 

  160. Mendoza, J. et al. Dimorphic effects of leptin on the circadian and hypocretinergic systems of mice. J. Neuroendocrinol. 23, 28–38 (2011).

    Article  CAS  PubMed  Google Scholar 

  161. Grosbellet, E., Gourmelen, S., Pevet, P., Criscuolo, F. & Challet, E. Leptin normalizes photic synchronization in male ob/ob mice, via indirect effects on the suprachiasmatic nucleus. Endocrinology 156, 1080–1090 (2015).

    Article  CAS  PubMed  Google Scholar 

  162. Pivovarova, O. et al. Changes of dietary fat and carbohydrate content alter central and peripheral clock in humans. J. Clin. Endocrinol. Metab. 100, 2291–2302 (2015).

    Article  CAS  PubMed  Google Scholar 

  163. Kohsaka, A. et al. High-fat diet disrupts behavioral and molecular circadian rhythms in mice. Cell Metab. 6, 414–421 (2007).

    Article  CAS  PubMed  Google Scholar 

  164. Chaix, A., Zarrinpar, A., Miu, P. & Panda, S. Time-restricted feeding is a preventative and therapeutic intervention against diverse nutritional challenges. Cell Metab. 20, 991–1005 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  165. Tognini, P. et al. Distinct circadian signatures in liver and gut clocks revealed by ketogenic diet. Cell Metab. 26, 523–538.e5 (2017).

    Article  CAS  PubMed  Google Scholar 

  166. Wang, Y. et al. The intestinal microbiota regulates body composition through NFIL3 and the circadian clock. Science 357, 912–916 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  167. Govindarajan, K. et al. Unconjugated bile acids influence expression of circadian genes: a potential mechanism for microbe-host crosstalk. PLoS ONE 11, e0167319 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  168. Li, T. et al. H. pylori infection induced BMAL1 expression and rhythm disorder aggravate gastric inflammation. EBioMedicine 39, 301–314 (2019).

    Article  PubMed  Google Scholar 

  169. Kervezee, L., Kosmadopoulos, A. & Boivin, D. B. Metabolic and cardiovascular consequences of shift work: the role of circadian disruption and sleep disturbances. Eur. J. Neurosci. 51, 396–412 (2020).

    Article  PubMed  Google Scholar 

  170. Cheng, P. & Drake, C. Shift work disorder. Neurol. Clin. 37, 563–577 (2019).

    Article  PubMed  Google Scholar 

  171. Roenneberg, T., Allebrandt, K. V., Merrow, M. & Vetter, C. Social jetlag and obesity. Curr. Biol. 22, 939–943 (2012).

    Article  CAS  PubMed  Google Scholar 

  172. Dibner, C. The importance of being rhythmic: Living in harmony with your body clocks. Acta Physiol. 228, e13281 (2020).

    Article  CAS  Google Scholar 

  173. Vieira, E. et al. Altered clock gene expression in obese visceral adipose tissue is associated with metabolic syndrome. PLoS ONE 9, e111678 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  174. Lessan, N. & Ali, T. Energy metabolism and intermittent fasting: the Ramadan perspective. Nutrients 11, 1192 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  175. Kul, S., Savas, E., Ozturk, Z. A. & Karadag, G. Does Ramadan fasting alter body weight and blood lipids and fasting blood glucose in a healthy population? A meta-analysis. J. Relig. Health 53, 929–942 (2014).

    Article  PubMed  Google Scholar 

  176. Sadeghirad, B., Motaghipisheh, S., Kolahdooz, F., Zahedi, M. J. & Haghdoost, A. A. Islamic fasting and weight loss: a systematic review and meta-analysis. Public Health Nutr. 17, 396–406 (2014).

    Article  PubMed  Google Scholar 

  177. Eckel-Mahan, K. et al. Reprogramming of the circadian clock by nutritional challenge. Cell 155, 1464–1478 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Chaix, A., Lin, T., Le, H. D., Chang, M. W. & Panda, S. Time-restricted feeding prevents obesity and metabolic syndrome in mice lacking a circadian clock. Cell Metab. 29, 303–319.e4 (2019).

    Article  CAS  PubMed  Google Scholar 

  179. Hatori, M. et al. Time-restricted feeding without reducing caloric intake prevents metabolic diseases in mice fed a high-fat diet. Cell Metab. 15, 848–860 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  180. Chaix, A., Manoogian, E. N. C., Melkani, G. C. & Panda, S. Time-restricted eating to prevent and manage chronic metabolic diseases. Annu. Rev. Nutr. 39, 291–315 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  181. Paoli, A., Tinsley, G., Bianco, A. & Moro, T. The influence of meal frequency and timing on health in humans: the role of fasting. Nutrients 11, 719 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  182. Sutton, E. F. et al. Early time-restricted feeding improves insulin sensitivity, blood pressure, and oxidative stress even without weight loss in men with prediabetes. Cell Metab. 27, 1212–1221.e3 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  183. Cienfuegos, S. et al. Effects of 4- and 6-h time-restricted feeding on weight and cardiometabolic health: a randomized controlled trial in adults with obesity. Cell Metab. 32, 366–378.e3 (2020). This is the first randomized control trial in humans that studied the effects of TRF (4 and 6 hours) on bodyweight and cardiometabolic risk factors in adults with obesity. TRF of 4 and 6 hours induced mild reductions in energy intake, bodyweight, insulin resistance and oxidative stress over 8 weeks.

    Article  CAS  PubMed  Google Scholar 

  184. Leidy, H. J., Gwin, J. A., Roenfeldt, C. A., Zino, A. Z. & Shafer, R. S. Evaluating the intervention-based evidence surrounding the causal role of breakfast on markers of weight management, with specific focus on breakfast composition and size. Adv. Nutr. 7, 563s–575s (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  185. Sharafi, M., Alamdari, N., Wilson, M., Leidy, H. J. & Glynn, E. L. Effect of a high-protein, high-fiber beverage preload on subjective appetite ratings and subsequent ad libitum energy intake in overweight men and women: a randomized, double-blind placebo-controlled, crossover study. Curr. Dev. Nutr. 2, nzy022 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  186. Douglas, S. M., Byers, A. W. & Leidy, H. J. Habitual breakfast patterns do not influence appetite and satiety responses in normal vs. high-protein breakfasts in overweight adolescent girls. Nutrients 11, 1223 (2019).

    Article  CAS  PubMed Central  Google Scholar 

  187. Sofer, S., Stark, A. H. & Madar, Z. Nutrition targeting by food timing: time-related dietary approaches to combat obesity and metabolic syndrome. Adv. Nutr. 6, 214–223 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  188. Ezagouri, S. et al. Physiological and molecular dissection of daily variance in exercise capacity. Cell Metab. 30, 78–91.e74 (2019).

    Article  CAS  PubMed  Google Scholar 

  189. Sato, S. et al. Time of exercise specifies the impact on muscle metabolic pathways and systemic energy homeostasis. Cell Metab. 30, 92–110.e4 (2019).

    Article  CAS  PubMed  Google Scholar 

  190. Savikj, M. et al. Afternoon exercise is more efficacious than morning exercise at improving blood glucose levels in individuals with type 2 diabetes: a randomised crossover trial. Diabetologia 62, 233–237 (2019).

    Article  CAS  PubMed  Google Scholar 

  191. Banno, M. et al. Exercise can improve sleep quality: a systematic review and meta-analysis. PeerJ 6, e5172 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  192. Cederroth, C. R. et al. Medicine in the fourth dimension. Cell Metab. 30, 238–250 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  193. Mure, L. S. et al. Diurnal transcriptome atlas of a primate across major neural and peripheral tissues. Science 359, eaao0318 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  194. Reinberg, A. & Smolensky, M. H. Circadian changes of drug disposition in man. Clin. Pharmacokinet 7, 401–420 (1982).

    Article  CAS  PubMed  Google Scholar 

  195. Scherholz, M. L., Schlesinger, N. & Androulakis, I. P. Chronopharmacology of glucocorticoids. Adv. Drug Deliv. Rev. 151-152, 245–261 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  196. Yagita, K., Yamanaka, I., Koinuma, S., Shigeyoshi, Y. & Uchiyama, Y. Mini screening of kinase inhibitors affecting period-length of mammalian cellular circadian clock. Acta Histochem. Cytochem. 42, 89–93 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  197. Chen, Z., Yoo, S. H. & Takahashi, J. S. Development and therapeutic potential of small-molecule modulators of circadian systems. Annu. Rev. Pharmacol. Toxicol. 58, 231–252 (2018).

    Article  CAS  PubMed  Google Scholar 

  198. Nohara, K. et al. Nobiletin fortifies mitochondrial respiration in skeletal muscle to promote healthy aging against metabolic challenge. Nat. Commun. 10, 3923 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  199. He, B. et al. The small molecule nobiletin targets the molecular oscillator to enhance circadian rhythms and protect against metabolic syndrome. Cell Metab. 23, 610–621 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  200. Petrenko, V. et al. In pancreatic islets from type 2 diabetes patients, the dampened circadian oscillators lead to reduced insulin and glucagon exocytosis. Proc. Natl Acad. Sci. USA 117, 2484–2495 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Synchronicity Pharma. Therapeutic strategy: focus on cryptochrome modulation Synchronicity Pharma http://www.synchronicitypharma.com/therapeutic-strategy/ (2019).

  202. Humphries, P. S. et al. Carbazole-containing amides and ureas: discovery of cryptochrome modulators as antihyperglycemic agents. Bioorg. Med. Chem. Lett. 28, 293–297 (2018).

    Article  CAS  PubMed  Google Scholar 

  203. Takahashi, S. M. Transcriptional architecture of the mammalian circadian clock. Nat. Rev. Genet. 18, 164–179 (2017).

    Article  CAS  PubMed  Google Scholar 

Download references

Author information

Authors and Affiliations

Authors

Contributions

The authors contributed equally to all aspects of the article.

Corresponding author

Correspondence to Inge Depoortere.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information

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

Publisher’s note

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

Glossary

Diurnal rhythms

Any biological rhythm that shows an oscillation of about 24 hours.

Free-running period

The duration of the circadian period in the absence of external time cues.

Peripheral clocks

All circadian clocks present in tissues or organs outside of the suprachiasmatic nucleus.

Zeitgeber

An external cue that entrains or synchronizes an organism’s circadian clock.

Chronodisruption

Chronic disruption of circadian rhythms by misalignment between the light–dark cycle and the behavioural cycle resulting in adverse health effects; examples are rotating shift work and frequent flying across different time zones.

Circadian rhythms

Biological rhythms that show an endogenous, entrainable oscillation of about 24 hours that remains over a range of physiological temperatures.

Dark-phase feeding period

Mice are nocturnal animals so they consume most of their food at night.

E-box

Enhancer box, DNA motifs with the consensus sequence CANNTG that appear in a broad variety of promotors and enhancers and serve as protein binding sites; E-boxes can be bound by proteins such as the CLOCK–BMAL1 heterodimer to initiate transcription.

Ileal brake

A primary inhibitory feedback mechanism that is initiated when nutrients arrive in the ileum to promote nutrient digestion and absorption; it inhibits gastric emptying, intestinal transit, food intake, gastric acid secretion, exocrine pancreatic secretion and gallbladder emptying but stimulates postprandial absorption of fluids and electrolytes.

Constant routine protocol

A method used in human circadian rhythm research to study endogenous circadian rhythms in the absence of external cues. Individuals are kept in constant conditions for at least 24 hours under constant temperature, dim light, semi-recumbent posture and awake. Food intake is evenly distributed throughout the protocol.

Chronobiotics

Compounds that can affect the phase, amplitude and/or period of a circadian rhythm.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Segers, A., Depoortere, I. Circadian clocks in the digestive system. Nat Rev Gastroenterol Hepatol 18, 239–251 (2021). https://doi.org/10.1038/s41575-020-00401-5

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41575-020-00401-5

This article is cited by

Search

Quick links

Nature Briefing

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

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