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 rhythm as a therapeutic target

Abstract

The circadian clock evolved in diverse organisms to integrate external environmental changes and internal physiology. The clock endows the host with temporal precision and robust adaptation to the surrounding environment. When circadian rhythms are perturbed or misaligned, as a result of jet lag, shiftwork or other lifestyle factors, adverse health consequences arise, and the risks of diseases such as cancer, cardiovascular diseases or metabolic disorders increase. Although the negative impact of circadian rhythm disruption is now well established, it remains underappreciated how to take advantage of biological timing, or correct it, for health benefits. In this Review, we provide an updated account of the circadian system and highlight several key disease areas with altered circadian signalling. We discuss environmental and lifestyle modifications of circadian rhythm and clock-based therapeutic strategies, including chronotherapy, in which dosing time is deliberately optimized for maximum therapeutic index, and pharmacological agents that target core clock components and proximal regulators. Promising progress in research, disease models and clinical applications should encourage a concerted effort towards a new era of circadian medicine.

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: Hierarchical organization of the mammalian clock system.
Fig. 2: The cell-autonomous core components of the circadian oscillator govern the ~24-hour cycle of gene expression.
Fig. 3: Melatonin and orexin signalling in circadian rhythm.
Fig. 4: Pharmacological interventions for insomnia and jet lag.

Similar content being viewed by others

References

  1. Reddy, P. et al. Molecular analysis of the period locus in Drosophila melanogaster and identification of a transcript involved in biological rhythms. Cell 38, 701–710 (1984). This landmark article reports the discovery of the period (per) locus in Drosophila melanogaster by molecular mapping of chromosome aberrations.

    Article  CAS  PubMed  Google Scholar 

  2. Bargiello, T. A., Jackson, F. R. & Young, M. W. Restoration of circadian behavioural rhythms by gene transfer in Drosophila. Nature 312, 752–754 (1984). This article demonstrates that introduction of a DNA fragment from a per+ fly encoding a 4.5-kb poly(A)+ RNA (later known as per) in a per0 (arrhythmic) fly can restore its circadian rhythmicity.

    Article  CAS  PubMed  Google Scholar 

  3. Hardin, P. E., Hall, J. C. & Rosbash, M. Feedback of the Drosophila period gene product on circadian cycling of its messenger RNA levels. Nature 343, 536–540 (1990). This landmark article identifies a feedback loop through which the cyclic per RNA level is regulated by its own protein activity.

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  5. Schroeder, A. M. & Colwell, C. S. How to fix a broken clock. Trends Pharmacol. Sci. 34, 605–619 (2013).

    Article  CAS  PubMed  Google Scholar 

  6. Buhr, E. D., Yoo, S. H. & Takahashi, J. S. Temperature as a universal resetting cue for mammalian circadian oscillators. Science 330, 379–385 (2010).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  7. Hurd, M. W. & Ralph, M. R. The significance of circadian organization for longevity in the golden hamster. J. Biol. Rhythm. 13, 430–436 (1998).

    Article  CAS  Google Scholar 

  8. DeCoursey, P. J. Survival value of suprachiasmatic nuclei (SCN) in four wild sciurid rodents. Behav. Neurosci. 128, 240–249 (2014).

    Article  PubMed  Google Scholar 

  9. Bass, J. & Lazar, M. A. Circadian time signatures of fitness and disease. Science 354, 994–999 (2016).

    Article  CAS  PubMed  Google Scholar 

  10. Levi, F. & Schibler, U. Circadian rhythms: mechanisms and therapeutic implications. Annu. Rev. Pharmacol. Toxicol. 47, 593–628 (2007).

    Article  CAS  PubMed  Google Scholar 

  11. 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 

  12. Liu, A. C. et al. Intercellular coupling confers robustness against mutations in the SCN circadian clock network. Cell 129, 605–616 (2007).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  13. Yamazaki, S. & Takahashi, J. S. Real-time luminescence reporting of circadian gene expression in mammals. Methods Enzymol. 393, 288–301 (2005).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  14. Abe, M. et al. Circadian rhythms in isolated brain regions. J. Neurosci. 22, 350–356 (2002).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  15. Mohawk, J. A., Green, C. B. & Takahashi, J. S. Central and peripheral circadian clocks in mammals. Annu. Rev. Neurosci. 35, 445–462 (2012). This important review summarizes the hierarchy of circadian oscillators that function at the cellular, tissue and systems levels.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  16. Liu, A. C., Lewis, W. G. & Kay, S. A. Mammalian circadian signaling networks and therapeutic targets. Nat. Chem. Biol. 3, 630–639 (2007).

    Article  CAS  PubMed  Google Scholar 

  17. Paschos, G. K. & FitzGerald, G. A. Circadian clocks and vascular function. Circ. Res. 106, 833–841 (2010).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  18. Ko, C. H. & Takahashi, J. S. Molecular components of the mammalian circadian clock. Hum. Mol. Genet. 15 (Suppl 2), R271–R277 (2006).

    Article  CAS  PubMed  Google Scholar 

  19. Vitaterna, M. H. et al. Mutagenesis and mapping of a mouse gene, Clock, essential for circadian behavior. Science 264, 719–725 (1994). This landmark research work discovers Clock by studies of N-ethyl-N-nitrosourea mutation and map position. The combination of both methods provides a generally applicable approach to study complex behaviour in mammals.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  20. Bunger, M. K. et al. Mop3 is an essential component of the master circadian pacemaker in mammals. Cell 103, 1009–1017 (2000).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  21. Gallego, M. & Virshup, D. M. Post-translational modifications regulate the ticking of the circadian clock. Nat. Rev. Mol. Cell Biol. 8, 139–148 (2007).

    Article  CAS  PubMed  Google Scholar 

  22. Siepka, S. M. et al. Circadian mutant Overtime reveals F-box protein FBXL3 regulation of cryptochrome and period gene expression. Cell 129, 1011–1023 (2007).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  23. Yoo, S. H. et al. Competing E3 ubiquitin ligases govern circadian periodicity by degradation of CRY in nucleus and cytoplasm. Cell 152, 1091–1105 (2013). This important article finds that CRY ubiquitination is regulated by the balance and cellular compartmentalization of the competing E3 ligases encoded by Fbxl21 and Fbxl3.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  24. Preitner, N. et al. The orphan nuclear receptor REV-ERBalpha controls circadian transcription within the positive limb of the mammalian circadian oscillator. Cell 110, 251–260 (2002).

    Article  CAS  PubMed  Google Scholar 

  25. Ueda, H. R. et al. A transcription factor response element for gene expression during circadian night. Nature 418, 534–539 (2002).

    Article  CAS  PubMed  Google Scholar 

  26. Panda, S. et al. Coordinated transcription of key pathways in the mouse by the circadian clock. Cell 109, 307–320 (2002). This landmark article finds that most cycling transcripts are regulated in a tissue-specific manner in the SCN and the liver. Major biological processes and rate-limiting steps in these pathways are regulated by the core oscillator components.

    Article  CAS  PubMed  Google Scholar 

  27. Storch, K. F. et al. Extensive and divergent circadian gene expression in liver and heart. Nature 417, 78–83 (2002).

    Article  CAS  PubMed  Google Scholar 

  28. Zhang, R., Lahens, N. F., Ballance, H. I., Hughes, M. E. & Hogenesch, J. B. A circadian gene expression atlas in mammals: implications for biology and medicine. Proc. Natl Acad. Sci. USA 111, 16219–16224 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed Central  PubMed  Google Scholar 

  30. Qu, M., Duffy, T., Hirota, T. & Kay, S. A. Nuclear receptor HNF4A transrepresses CLOCK:BMAL1 and modulates tissue-specific circadian networks. Proc. Natl Acad. Sci. USA 115, E12305–E12312 (2018). This article demonstrates that the transcriptional activity of the CLOCK–BMAL1 heterodimer can be transrepressed by nuclear receptor HNF4A, which defines a novel feedback loop in tissue-specific mammalian oscillators.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed Central  PubMed  Google Scholar 

  32. Papazyan, R., Zhang, Y. & Lazar, M. A. Genetic and epigenomic mechanisms of mammalian circadian transcription. Nat. Struct. Mol. Biol. 23, 1045–1052 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  33. Yoo, S. H. et al. Period2 3′-UTR and microRNA-24 regulate circadian rhythms by repressing PERIOD2 protein accumulation. Proc. Natl Acad. Sci. USA 114, E8855–E8864 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Green, C. B. Circadian posttranscriptional regulatory mechanisms in mammals. Cold Spring Harb. Perspect. Biol. 10, a030692 (2018).

    Article  PubMed Central  PubMed  Google Scholar 

  35. Saper, C. B., Scammell, T. E. & Lu, J. Hypothalamic regulation of sleep and circadian rhythms. Nature 437, 1257–1263 (2005).

    Article  CAS  PubMed  Google Scholar 

  36. Wurtman, R. J., Axelrod, J. & Fischer, J. E. Melatonin synthesis in the pineal gland: effect of light mediated by the sympathetic nervous system. Science 143, 1328–1329 (1964).

    Article  CAS  PubMed  Google Scholar 

  37. Fisher, S. P., Foster, R. G. & Peirson, S. N. The circadian control of sleep. Handb. Exp. Pharmacol. 217, 157–183 (2013).

    Article  CAS  Google Scholar 

  38. Liu, C. et al. Molecular dissection of two distinct actions of melatonin on the suprachiasmatic circadian clock. Neuron 19, 91–102 (1997). This study provides critically important insights into the molecular basis for two distinct, mechanistically separable effects of melatonin on SCN functions.

    Article  CAS  PubMed  Google Scholar 

  39. Pandi-Perumal, S. R. et al. Physiological effects of melatonin: role of melatonin receptors and signal transduction pathways. Prog. Neurobiol. 85, 335–353 (2008).

    Article  CAS  PubMed  Google Scholar 

  40. Hunt, A. E., Al-Ghoul, W. M., Gillette, M. U. & Dubocovich, M. L. Activation of MT2 melatonin receptors in rat suprachiasmatic nucleus phase advances the circadian clock. Am. J. Physiol. Cell Physiol. 280, C110–C118 (2001).

    Article  CAS  PubMed  Google Scholar 

  41. Dubocovich, M. L., Yun, K., Al-Ghoul, W. M., Benloucif, S. & Masana, M. I. Selective MT2 melatonin receptor antagonists block melatonin-mediated phase advances of circadian rhythms. FASEB J. 12, 1211–1220 (1998).

    Article  CAS  PubMed  Google Scholar 

  42. Poirel, V. J. et al. Contrary to other non-photic cues, acute melatonin injection does not induce immediate changes of clock gene mRNA expression in the rat suprachiasmatic nuclei. Neuroscience 120, 745–755 (2003).

    Article  CAS  PubMed  Google Scholar 

  43. Pfeffer, M., Rauch, A., Korf, H. W. & von Gall, C. The endogenous melatonin (MT) signal facilitates reentrainment of the circadian system to light-induced phase advances by acting upon MT2 receptors. Chronobiol. Int. 29, 415–429 (2012).

    Article  CAS  PubMed  Google Scholar 

  44. Agez, L., Laurent, V., Pevet, P., Masson-Pevet, M. & Gauer, F. Melatonin affects nuclear orphan receptors mRNA in the rat suprachiasmatic nuclei. Neuroscience 144, 522–530 (2007).

    Article  CAS  PubMed  Google Scholar 

  45. Sakurai, T. et al. Orexins and orexin receptors: a family of hypothalamic neuropeptides and G protein-coupled receptors that regulate feeding behavior. Cell 92, 573–585 (1998). This article demonstrates that by binding and activating the two orphan G-protein-coupled receptors in the rat brain, orexins could stimulate food consumption, suggesting its pivotal role in the feedback to the feeding behaviour.

    Article  CAS  PubMed  Google Scholar 

  46. Deboer, T. et al. Convergence of circadian and sleep regulatory mechanisms on hypocretin-1. Neuroscience 129, 727–732 (2004).

    Article  CAS  PubMed  Google Scholar 

  47. Xu, T. R., Yang, Y., Ward, R., Gao, L. & Liu, Y. Orexin receptors: multi-functional therapeutic targets for sleeping disorders, eating disorders, drug addiction, cancers and other physiological disorders. Cell. Signal. 25, 2413–2423 (2013).

    Article  CAS  PubMed  Google Scholar 

  48. Mieda, M. et al. Differential roles of orexin receptor-1 and -2 in the regulation of non-REM and REM sleep. J. Neurosci. 31, 6518–6526 (2011).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  49. Belle, M. D. et al. Acute suppressive and long-term phase modulation actions of orexin on the mammalian circadian clock. J. Neurosci. 34, 3607–3621 (2014).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  50. Tsimakouridze, E. V., Alibhai, F. J. & Martino, T. A. Therapeutic applications of circadian rhythms for the cardiovascular system. Front. Pharmacol. 6, 77 (2015).

    Article  PubMed Central  PubMed  Google Scholar 

  51. Sundar, I. K., Yao, H., Sellix, M. T. & Rahman, I. Circadian molecular clock in lung pathophysiology. Am. J. Physiol. Lung Cell. Mol. Physiol. 309, L1056–L1075 (2015).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  52. Burish, M. J., Chen, Z. & Yoo, S. H. Cluster headache is in part a disorder of the circadian system. JAMA Neurol. 75, 783–784 (2018).

    Article  PubMed Central  PubMed  Google Scholar 

  53. Bedrosian, T. A. & Nelson, R. J. Sundowning syndrome in aging and dementia: research in mouse models. Exp. Neurol. 243, 67–73 (2013).

    Article  PubMed  Google Scholar 

  54. Hood, S. & Amir, S. The aging clock: circadian rhythms and later life. J. Clin. Invest. 127, 437–446 (2017).

    Article  PubMed Central  PubMed  Google Scholar 

  55. Stubblefield, J. J. et al. Temporal control of metabolic amplitude by nocturnin. Cell Rep. 22, 1225–1235 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  56. Musiek, E. S., Xiong, D. D. & Holtzman, D. M. Sleep, circadian rhythms, and the pathogenesis of Alzheimer disease. Exp. Mol. Med. 47, e148 (2015).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  57. Karamitri, A., Renault, N., Clement, N., Guillaume, J. L. & Jockers, R. Minireview: toward the establishment of a link between melatonin and glucose homeostasis: association of melatonin MT2 receptor variants with type 2 diabetes. Mol. Endocrinol. 27, 1217–1233 (2013).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  58. Prokopenko, I. et al. Variants in MTNR1B influence fasting glucose levels. Nat. Genet. 41, 77–81 (2009).

    Article  CAS  PubMed  Google Scholar 

  59. Takahashi, J. S., Hong, H. K., Ko, C. H. & McDearmon, E. L. The genetics of mammalian circadian order and disorder: implications for physiology and disease. Nat. Rev. Genet. 9, 764–775 (2008).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  60. Jones, C. R., Huang, A. L., Ptacek, L. J. & Fu, Y. H. Genetic basis of human circadian rhythm disorders. Exp. Neurol. 243, 28–33 (2013).

    Article  PubMed  Google Scholar 

  61. Xu, Y. et al. Modeling of a human circadian mutation yields insights into clock regulation by PER2. Cell 128, 59–70 (2007).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  62. Hirano, A. et al. A cryptochrome 2 mutation yields advanced sleep phase in humans. eLife 5, e16695 (2016).

    Article  PubMed Central  PubMed  Google Scholar 

  63. Xing, W. et al. SCF(FBXL3) ubiquitin ligase targets cryptochromes at their cofactor pocket. Nature 496, 64–68 (2013).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  64. Sack, R. L. et al. Circadian rhythm sleep disorders: part II, advanced sleep phase disorder, delayed sleep phase disorder, free-running disorder, and irregular sleep-wake rhythm. An American Academy of Sleep Medicine review. Sleep 30, 1484–1501 (2007).

    Article  PubMed Central  PubMed  Google Scholar 

  65. Archer, S. N. et al. A length polymorphism in the circadian clock gene Per3 is linked to delayed sleep phase syndrome and extreme diurnal preference. Sleep 26, 413–415 (2003).

    Article  PubMed  Google Scholar 

  66. Patke, A. et al. Mutation of the human circadian clock gene CRY1 in familial delayed sleep phase disorder. Cell 169, 203–215 e13 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  67. Zarbock, A. et al. Effect of remote ischemic preconditioning on kidney injury among high-risk patients undergoing cardiac surgery: a randomized clinical trial. JAMA 313, 2133–2141 (2015). This multicentre randomized clinical trial demonstrates that remote ischaemic preconditioning significantly reduced acute kidney injury and uses of renal replacement therapy in high-risk patients undergoing cardiac surgery, representing a simple and promising strategy to improve postoperative outcomes.

    Article  CAS  PubMed  Google Scholar 

  68. Eltzschig, H. K. & Eckle, T. Ischemia and reperfusion — from mechanism to translation. Nat. Med. 17, 1391–1401 (2011). This review discusses the molecular and immunological consequences of ischaemia and reperfusion and potential strategies that could be used as innovative therapies for treating patients with tissue inflammation due to this condition.

    Article  CAS  PubMed  Google Scholar 

  69. Koeppen, M. et al. Hypoxia-inducible factor 2-alpha-dependent induction of amphiregulin dampens myocardial ischemia-reperfusion injury. Nat. Commun. 9, 816 (2018).

    Article  PubMed Central  PubMed  Google Scholar 

  70. Eltzschig, H. K. & Carmeliet, P. Hypoxia and inflammation. N. Engl. J. Med. 364, 656–665 (2011).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  71. Eltzschig, H. K., Bratton, D. L. & Colgan, S. P. Targeting hypoxia signalling for the treatment of ischaemic and inflammatory diseases. Nat. Rev. Drug Discov. 13, 852–869 (2014).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  72. Peek, C. B. et al. Circadian clock interaction with HIF1alpha mediates oxygenic metabolism and anaerobic glycolysis in skeletal muscle. Cell Metab. 25, 86–92 (2017).

    Article  CAS  PubMed  Google Scholar 

  73. Wu, Y. et al. Reciprocal regulation between the circadian clock and hypoxia signaling at the genome level in mammals. Cell Metab. 25, 73–85 (2017).

    Article  CAS  PubMed  Google Scholar 

  74. Eckle, T. et al. Adora2b-elicited Per2 stabilization promotes a HIF-dependent metabolic switch crucial for myocardial adaptation to ischemia. Nat. Med. 18, 774–782 (2012). This important study identifies a functional role for extracellular adenosine signalling via adenosine receptor A2B to promote Per2 stabilization, causing a metabolic switch providing cardioprotection against ischaemia. This study is among the first reports to demonstrate a functional interaction between hypoxia-inducible factor and circadian rhythm signalling, and the potential use of light therapy as a mechanism to dampen myocardial ischaemia and reperfusion injury.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  75. Eckle, T., Kohler, D., Lehmann, R., El Kasmi, K. C. & Eltzschig, H. K. Hypoxia-inducible factor-1 is central to cardioprotection: a new paradigm for ischemic preconditioning. Circulation 118, 166–175 (2008).

    Article  CAS  PubMed  Google Scholar 

  76. Correa-Costa, M. et al. Carbon monoxide protects the kidney through the central circadian clock and CD39. Proc. Natl Acad. Sci. USA 115, E2302–E2310 (2018). This important study indicates a cellular signalling mechanism whereby carbon monoxide modulates purinergic responses involving increased CD39 ectonucleotidase expression and Per2 to protect against kidney ischaemia and reperfusion injury. These findings support therapeutic use of carbon monoxide to treat ischaemia and reperfusion injury in association with organ transplantation, stroke or myocardial infarction.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Yuan, D. et al. Blue light reduces organ injury from ischemia and reperfusion. Proc. Natl Acad. Sci. USA 113, 5239–5244 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Oyama, Y. et al. Intense light-mediated circadian cardioprotection via transcriptional reprogramming of the endothelium. Cell Rep. 28, 1471–1484 e11 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  79. Crnko, S., Du Pre, B. C., Sluijter, J. P. G. & Van Laake, L. W. Circadian rhythms and the molecular clock in cardiovascular biology and disease. Nat. Rev. Cardiol. 16, 437–447 (2019).

    Article  PubMed  Google Scholar 

  80. Muller, J. E. et al. Circadian variation in the frequency of onset of acute myocardial infarction. N. Engl. J. Med. 313, 1315–1322 (1985).

    Article  CAS  PubMed  Google Scholar 

  81. Bulluck, H. et al. Circadian variation in acute myocardial infarct size assessed by cardiovascular magnetic resonance in reperfused STEMI patients. Int. J. Cardiol. 230, 149–154 (2017).

    Article  PubMed Central  PubMed  Google Scholar 

  82. Montaigne, D. et al. Daytime variation of perioperative myocardial injury in cardiac surgery and its prevention by Rev-Erbalpha antagonism: a single-centre propensity-matched cohort study and a randomised study. Lancet 391, 59–69 (2018). This landmark article demonstrates that perioperative myocardial injury is transcriptionally orchestrated by the circadian clock in patients undergoing aortic valve replacement with improved patient outcomes in the afternoon.

    Article  PubMed  Google Scholar 

  83. Fortuyn-van Leijden, C. E. Some observations on periodic nuclear division in the cat. KNAB 19, 38–44 (1917).

    Google Scholar 

  84. Canaple, L., Kakizawa, T. & Laudet, V. The days and nights of cancer cells. Cancer Res. 63, 7545–7552 (2003).

    CAS  PubMed  Google Scholar 

  85. Matsuo, T. et al. Control mechanism of the circadian clock for timing of cell division in vivo. Science 302, 255–259 (2003).

    Article  CAS  PubMed  Google Scholar 

  86. Gery, S. et al. The circadian gene per1 plays an important role in cell growth and DNA damage control in human cancer cells. Mol. Cell 22, 375–382 (2006).

    Article  CAS  PubMed  Google Scholar 

  87. Fu, L., Pelicano, H., Liu, J., Huang, P. & Lee, C. The circadian gene Period2 plays an important role in tumor suppression and DNA damage response in vivo. Cell 111, 41–50 (2002). This important article finds that Per2 acts as a tumour suppressor by regulating DNA damage-responsive pathways, indicating that the circadian clock could also play an important part in the response to unpredicted hazards that are detrimental to genomic material.

    Article  CAS  PubMed  Google Scholar 

  88. Kettner, N. M. et al. Circadian homeostasis of liver metabolism suppresses hepatocarcinogenesis. Cancer Cell 30, 909–924 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  89. Anafi, R. C., Francey, L. J., Hogenesch, J. B. & Kim, J. CYCLOPS reveals human transcriptional rhythms in health and disease. Proc. Natl Acad. Sci. USA 114, 5312–5317 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Papagiannakopoulos, T. et al. Circadian rhythm disruption promotes lung tumorigenesis. Cell Metab. 24, 324–331 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  91. Huber, A. L. et al. CRY2 and FBXL3 cooperatively degrade c-MYC. Mol. Cell 64, 774–789 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  92. Ozturk, N., Lee, J. H., Gaddameedhi, S. & Sancar, A. Loss of cryptochrome reduces cancer risk in p53 mutant mice. Proc. Natl Acad. Sci. USA 106, 2841–2846 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  93. Puram, R. V. et al. Core circadian clock genes regulate leukemia stem cells in AML. Cell 165, 303–316 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  94. Dong, Z. et al. Targeting glioblastoma stem cells through disruption of the circadian clock. Cancer Discov. 9, 1556–1573 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  95. Morales-Santana, S. et al. An overview of the polymorphisms of circadian genes associated with endocrine cancer. Front. Endocrinol. 10, 104 (2019).

    Article  Google Scholar 

  96. Oklejewicz, M. et al. Phase resetting of the mammalian circadian clock by DNA damage. Curr. Biol. 18, 286–291 (2008).

    Article  CAS  PubMed  Google Scholar 

  97. Masri, S. et al. Lung adenocarcinoma distally rewires hepatic circadian homeostasis. Cell 165, 896–909 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  98. Altman, B. J. et al. MYC disrupts the circadian clock and metabolism in cancer cells. Cell Metab. 22, 1009–1019 (2015).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  99. Knutson, K. L., Spiegel, K., Penev, P. & Van Cauter, E. The metabolic consequences of sleep deprivation. Sleep Med. Rev. 11, 163–178 (2007).

    Article  PubMed Central  PubMed  Google Scholar 

  100. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Buxton, O. M. et al. Adverse metabolic consequences in humans of prolonged sleep restriction combined with circadian disruption. Sci. Transl. Med. 4, 129ra43 (2012).

    Article  PubMed Central  PubMed  Google Scholar 

  102. Bass, J. & Takahashi, J. S. Circadian integration of metabolism and energetics. Science 330, 1349–1354 (2010).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  103. Turek, F. W. et al. Obesity and metabolic syndrome in circadian Clock mutant mice. Science 308, 1043–1045 (2005). This study identifies the important role of the circadian gene Clock in mammalian energy balance.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  104. Shi, S. Q., Ansari, T. S., McGuinness, O. P., Wasserman, D. H. & Johnson, C. H. Circadian disruption leads to insulin resistance and obesity. Curr. Biol. 23, 372–381 (2013).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  105. Fonken, L. K. et al. Light at night increases body mass by shifting the time of food intake. Proc. Natl Acad. Sci. USA 107, 18664–18669 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  107. Ramsey, K. M. et al. Circadian clock feedback cycle through NAMPT-mediated NAD+ biosynthesis. Science 324, 651–654 (2009).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  108. Nakahata, Y., Sahar, S., Astarita, G., Kaluzova, M. & Sassone-Corsi, P. Circadian control of the NAD+ salvage pathway by CLOCK-SIRT1. Science 324, 654–657 (2009).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  109. Nakahata, Y. et al. The NAD+-dependent deacetylase SIRT1 modulates CLOCK-mediated chromatin remodeling and circadian control. Cell 134, 329–340 (2008).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  110. Peek, C. B. et al. Circadian clock NAD+ cycle drives mitochondrial oxidative metabolism in mice. Science 342, 1243417 (2013).

    Article  PubMed Central  PubMed  Google Scholar 

  111. Yeung, J. et al. Transcription factor activity rhythms and tissue-specific chromatin interactions explain circadian gene expression across organs. Genome Res. 28, 182–191 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  112. Marcheva, B. et al. Disruption of the clock components CLOCK and BMAL1 leads to hypoinsulinaemia and diabetes. Nature 466, 627–631 (2010).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  113. Sinturel, F. et al. Diurnal oscillations in liver mass and cell size accompany ribosome assembly cycles. Cell 169, 651–663 e14 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  114. Sanchez-Lasheras, C., Konner, A. C. & Bruning, J. C. Integrative neurobiology of energy homeostasis-neurocircuits, signals and mediators. Front. Neuroendocrinol. 31, 4–15 (2010).

    Article  CAS  PubMed  Google Scholar 

  115. Huang, W., Moynihan Ramsey, K., Marcheva, B. & Bass, J. Circadian rhythms, sleep, and metabolism. J. Clin. Invest. 121, 2133–2141 (2011).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  116. Yi, C. X. et al. Ventromedial arcuate nucleus communicates peripheral metabolic information to the suprachiasmatic nucleus. Endocrinology 147, 283–294 (2006).

    Article  CAS  PubMed  Google Scholar 

  117. Hill, J. W. Gene expression and the control of food intake by hypothalamic POMC/CART neurons. Open Neuroendocrinol. J. 3, 21–27 (2010).

    CAS  PubMed Central  PubMed  Google Scholar 

  118. Mieda, M. & Yanagisawa, M. Sleep, feeding, and neuropeptides: roles of orexins and orexin receptors. Curr. Opin. Neurobiol. 12, 339–345 (2002).

    Article  CAS  PubMed  Google Scholar 

  119. Hara, J. et al. Genetic ablation of orexin neurons in mice results in narcolepsy, hypophagia, and obesity. Neuron 30, 345–354 (2001).

    Article  CAS  PubMed  Google Scholar 

  120. Mahoney, C. E., Cogswell, A., Koralnik, I. J. & Scammell, T. E. The neurobiological basis of narcolepsy. Nat. Rev. Neurosci. 20, 83–93 (2019).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  121. Iwasaki, A. & Medzhitov, R. Toll-like receptor control of the adaptive immune responses. Nat. Immunol. 5, 987–995 (2004).

    Article  CAS  PubMed  Google Scholar 

  122. Serhan, C. N. & Savill, J. Resolution of inflammation: the beginning programs the end. Nat. Immunol. 6, 1191–1197 (2005). This comprehensive review summarizes the mechanisms of an active, coordinated program for inflammation resolution which may bring profound advances in therapies aimed at terminating persistent inflammation.

    Article  CAS  PubMed  Google Scholar 

  123. Ehrentraut, H. et al. CD73+ regulatory T cells contribute to adenosine-mediated resolution of acute lung injury. FASEB J. 27, 2207–2219 (2013).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  124. Serhan, C. N. et al. Resolvins: a family of bioactive products of omega-3 fatty acid transformation circuits initiated by aspirin treatment that counter proinflammation signals. J. Exp. Med. 196, 1025–1037 (2002).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  125. Man, K., Loudon, A. & Chawla, A. Immunity around the clock. Science 354, 999–1003 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  126. Adrover, J. M. et al. A neutrophil timer coordinates immune defense and vascular protection. Immunity 51, 966–967 (2019).

    Article  CAS  PubMed  Google Scholar 

  127. Halberg, F., Johnson, E. A., Brown, B. W. & Bittner, J. J. Susceptibility rhythm to E. coli endotoxin and bioassay. Proc. Soc. Exp. Biol. Med. 103, 142–144 (1960).

    Article  CAS  PubMed  Google Scholar 

  128. Scheiermann, C. et al. Adrenergic nerves govern circadian leukocyte recruitment to tissues. Immunity 37, 290–301 (2012).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  129. Gibbs, J. E. et al. The nuclear receptor REV-ERBalpha mediates circadian regulation of innate immunity through selective regulation of inflammatory cytokines. Proc. Natl Acad. Sci. USA 109, 582–587 (2012).

    Article  CAS  PubMed  Google Scholar 

  130. Curtis, A. M. et al. Circadian control of innate immunity in macrophages by miR-155 targeting Bmal1. Proc. Natl Acad. Sci. USA 112, 7231–7236 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  131. Nguyen, K. D. et al. Circadian gene Bmal1 regulates diurnal oscillations of Ly6Chi inflammatory monocytes. Science 341, 1483–1488 (2013).

    Article  CAS  PubMed  Google Scholar 

  132. Gibbs, J. et al. An epithelial circadian clock controls pulmonary inflammation and glucocorticoid action. Nat. Med. 20, 919–926 (2014).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  133. Jetten, A. M. Retinoid-related orphan receptors (RORs): critical roles in development, immunity, circadian rhythm, and cellular metabolism. Nucl. Recept. Signal. 7, e003 (2009).

    Article  PubMed Central  PubMed  Google Scholar 

  134. Yu, X. et al. TH17 cell differentiation is regulated by the circadian clock. Science 342, 727–730 (2013). This landmark study reports how the molecular circadian clock directly regulates the differentiation of TH17 cells in the intestine via the transcription factor NFIL3, which suggest that both nutrition and light are important environmental factors that directly regulate the immune response.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  135. Scheiermann, C., Gibbs, J., Ince, L. & Loudon, A. Clocking in to immunity. Nat. Rev. Immunol. 18, 423–437 (2018).

    Article  CAS  PubMed  Google Scholar 

  136. Shimba, A. et al. Glucocorticoids drive diurnal oscillations in T cell distribution and responses by inducing interleukin-7 receptor and CXCR4. Immunity 48, 286–298 e6 (2018).

    Article  CAS  PubMed  Google Scholar 

  137. Silver, A. C., Arjona, A., Walker, W. E. & Fikrig, E. The circadian clock controls toll-like receptor 9-mediated innate and adaptive immunity. Immunity 36, 251–261 (2012).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  138. McKenna, H., van der Horst, G. T. J., Reiss, I. & Martin, D. Clinical chronobiology: a timely consideration in critical care medicine. Crit. Care 22, 124 (2018).

    Article  PubMed Central  PubMed  Google Scholar 

  139. Oldham, M. A., Lee, H. B. & Desan, P. H. Circadian rhythm disruption in the critically ill: an opportunity for improving outcomes. Crit. Care Med. 44, 207–217 (2016).

    Article  PubMed  Google Scholar 

  140. Solanas, G. et al. Aged stem cells reprogram their daily rhythmic functions to adapt to stress. Cell 170, 678–692 e20 (2017).

    Article  CAS  PubMed  Google Scholar 

  141. Sato, S. et al. Circadian reprogramming in the liver identifies metabolic pathways of aging. Cell 170, 664–677 e11 (2017).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  142. Levine, D. C. et al. NAD+ controls circadian reprogramming through PER2 nuclear translocation to counter aging. Mol. Cell 78, 835–849 e7 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  143. Kondratov, R. V., Kondratova, A. A., Gorbacheva, V. Y., Vykhovanets, O. V. & Antoch, M. P. Early aging and age-related pathologies in mice deficient in BMAL1, the core componentof the circadian clock. Genes Dev. 20, 1868–1873 (2006).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  144. Wang, R. H. et al. Negative reciprocal regulation between Sirt1 and Per2 modulates the circadian clock and aging. Sci. Rep. 6, 28633 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  145. Wu, H., Dunnett, S., Ho, Y. S. & Chang, R. C. The role of sleep deprivation and circadian rhythm disruption as risk factors of Alzheimer’s disease. Front. Neuroendocrinol. 54, 100764 (2019).

    Article  PubMed  Google Scholar 

  146. Hermida, R. C. et al. Chronotherapy improves blood pressure control and reduces vascular risk in CKD. Nat. Rev. Nephrol. 9, 358–368 (2013).

    Article  CAS  PubMed  Google Scholar 

  147. Kobayashi, M., Wood, P. A. & Hrushesky, W. J. Circadian chemotherapy for gynecological and genitourinary cancers. Chronobiol. Int. 19, 237–251 (2002).

    Article  CAS  PubMed  Google Scholar 

  148. Gorbacheva, V. Y. et al. Circadian sensitivity to the chemotherapeutic agent cyclophosphamide depends on the functional status of the CLOCK/BMAL1 transactivation complex. Proc. Natl Acad. Sci. USA 102, 3407–3412 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  149. Griffett, K. & Burris, T. P. The mammalian clock and chronopharmacology. Bioorg. Med. Chem. Lett. 23, 1929–1934 (2013).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  150. Medalie, L. & Cifu, A. S. Management of chronic insomnia disorder in adults. JAMA 317, 762–763 (2017).

    Article  PubMed  Google Scholar 

  151. Wade, A. G. et al. Nightly treatment of primary insomnia with prolonged release melatonin for 6 months: a randomized placebo controlled trial on age and endogenous melatonin as predictors of efficacy and safety. BMC Med. 8, 51 (2010).

    Article  PubMed Central  PubMed  Google Scholar 

  152. Wilson, S. et al. British Association for Psychopharmacology consensus statement on evidence-based treatment of insomnia, parasomnias and circadian rhythm disorders: an update. J. Psychopharmacol. 33, 923–947 (2019).

    Article  PubMed  Google Scholar 

  153. Williams, W. P. 3rd, McLin, D. E. 3rd, Dressman, M. A. & Neubauer, D. N. Comparative review of approved melatonin agonists for the treatment of circadian rhythm sleep-wake disorders. Pharmacotherapy 36, 1028–1041 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  154. Kuriyama, A., Honda, M. & Hayashino, Y. Ramelteon for the treatment of insomnia in adults: a systematic review and meta-analysis. Sleep Med. 15, 385–392 (2014).

    Article  PubMed  Google Scholar 

  155. Comai, S., Ochoa-Sanchez, R. & Gobbi, G. Sleep-wake characterization of double MT1/MT2 receptor knockout mice and comparison with MT1 and MT2 receptor knockout mice. Behav. Brain Res. 243, 231–238 (2013).

    Article  CAS  PubMed  Google Scholar 

  156. Gotter, A. L. et al. Orexin 2 receptor antagonism is sufficient to promote NREM and REM sleep from mouse to man. Sci. Rep. 6, 27147 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  157. Morairty, S. R. et al. Dual hypocretin receptor antagonism is more effective for sleep promotion than antagonism of either receptor alone. PLoS ONE 7, e39131 (2012).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  158. Coleman, P. J., Gotter, A. L., Herring, W. J., Winrow, C. J. & Renger, J. J. The discovery of suvorexant, the first orexin receptor drug for insomnia. Annu. Rev. Pharmacol. Toxicol. 57, 509–533 (2017).

    Article  CAS  PubMed  Google Scholar 

  159. Tannenbaum, P. L. et al. Inhibition of orexin signaling promotes sleep yet preserves salient arousability in monkeys. Sleep 39, 603–612 (2016).

    Article  PubMed Central  PubMed  Google Scholar 

  160. Uslaner, J. M. et al. Orexin receptor antagonists differ from standard sleep drugs by promoting sleep at doses that do not disrupt cognition. Sci. Transl. Med. 5, 179ra44 (2013).

    Article  PubMed  Google Scholar 

  161. Arendt, J. Approaches to the pharmacological management of jet lag. Drugs 78, 1419–1431 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  162. Arendt, J. & Skene, D. J. Melatonin as a chronobiotic. Sleep Med. Rev. 9, 25–39 (2005).

    Article  PubMed  Google Scholar 

  163. McArthur, A. J., Hunt, A. E. & Gillette, M. U. Melatonin action and signal transduction in the rat suprachiasmatic circadian clock: activation of protein kinase C at dusk and dawn. Endocrinology 138, 627–634 (1997).

    Article  CAS  PubMed  Google Scholar 

  164. McArthur, A. J., Gillette, M. U. & Prosser, R. A. Melatonin directly resets the rat suprachiasmatic circadian clock in vitro. Brain Res. 565, 158–161 (1991).

    Article  CAS  PubMed  Google Scholar 

  165. Pfeffer, M., Korf, H. W. & Wicht, H. Synchronizing effects of melatonin on diurnal and circadian rhythms. Gen. Comp. Endocrinol. 258, 215–221 (2018).

    Article  CAS  PubMed  Google Scholar 

  166. Rajaratnam, S. M. et al. Melatonin agonist tasimelteon (VEC-162) for transient insomnia after sleep-time shift: two randomised controlled multicentre trials. Lancet 373, 482–491 (2009).

    Article  CAS  PubMed  Google Scholar 

  167. Stauch, B. et al. Structural basis of ligand recognition at the human MT1 melatonin receptor. Nature 569, 284–288 (2019).

    Article  CAS  PubMed  Google Scholar 

  168. Stein, R. M. et al. Virtual discovery of melatonin receptor ligands to modulate circadian rhythms. Nature 579, 609–614 (2020). This important article identifies several MT1 receptor-selective inverse agonists from structure-based screens of diverse, ultralarge libraries. These chemotypes could phase-advance the mouse circadian clock, therefore illuminating new in vivo pharmacology.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  169. Riha, R. L. The use and misuse of exogenous melatonin in the treatment of sleep disorders. Curr. Opin. Pulm. Med. 24, 543–548 (2018).

    Article  CAS  PubMed  Google Scholar 

  170. Lewy, A. J., Ahmed, S., Jackson, J. M. & Sack, R. L. Melatonin shifts human circadian rhythms according to a phase-response curve. Chronobiol. Int. 9, 380–392 (1992).

    Article  CAS  PubMed  Google Scholar 

  171. Burgess, H. J., Revell, V. L. & Eastman, C. I. A three pulse phase response curve to three milligrams of melatonin in humans. J. Physiol. 586, 639–647 (2008).

    Article  CAS  PubMed  Google Scholar 

  172. Roach, G. D. & Sargent, C. Interventions to minimize jet lag after westward and eastward flight. Front. Physiol. 10, 927 (2019).

    Article  PubMed Central  PubMed  Google Scholar 

  173. Wickwire, E. M., Geiger-Brown, J., Scharf, S. M. & Drake, C. L. Shift work and shift work sleep disorder: clinical and organizational perspectives. Chest 151, 1156–1172 (2017).

    Article  PubMed  Google Scholar 

  174. Green, C. B., Takahashi, J. S. & Bass, J. The meter of metabolism. Cell 134, 728–742 (2008).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  175. Wegrzyn, L. R. et al. Rotating night-shift work and the risk of breast cancer in the nurses’ health studies. Am. J. Epidemiol. 186, 532–540 (2017).

    Article  PubMed Central  PubMed  Google Scholar 

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

  177. Agostino, P. V., Plano, S. A. & Golombek, D. A. Sildenafil accelerates reentrainment of circadian rhythms after advancing light schedules. Proc. Natl Acad. Sci. USA 104, 9834–9839 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  178. Ju, D. et al. Chemical perturbations reveal that RUVBL2 regulates the circadian phase in mammals. Sci. Transl. Med. 12, eaba0769 (2020). This important study demonstrates that pharmacological perturbation of RUVBL2 by the adenosine derivative cordycepin shifts the circadian phase both in human cells and in mouse tissue and rapidly entrains the circadian phase in a mouse jet lag model. This phase-shifting effect is the result of disassembly of interaction between RUVBL2 and BMAL1, thereby leading to the release of repression of clock gene transcription.

    Article  CAS  PubMed  Google Scholar 

  179. Grant, D. et al. GSK4112, a small molecule chemical probe for the cell biology of the nuclear heme receptor Rev-erbalpha. ACS Chem. Biol. 5, 925–932 (2010).

    Article  CAS  PubMed  Google Scholar 

  180. Trump, R. P. et al. Optimized chemical probes for REV-ERBalpha. J. Med. Chem. 56, 4729–4737 (2013).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  181. Solt, L. A. et al. Regulation of circadian behaviour and metabolism by synthetic REV-ERB agonists. Nature 485, 62–68 (2012).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  182. Sulli, G. et al. Pharmacological activation of REV-ERBs is lethal in cancer and oncogene-induced senescence. Nature 553, 351–355 (2018).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  183. Banerjee, S. et al. Pharmacological targeting of the mammalian clock regulates sleep architecture and emotional behaviour. Nat. Commun. 5, 5759 (2014).

    Article  CAS  PubMed  Google Scholar 

  184. Woldt, E. et al. Rev-erb-alpha modulates skeletal muscle oxidative capacity by regulating mitochondrial biogenesis and autophagy. Nat. Med. 19, 1039–1046 (2013).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  185. Thomes, P. G., Brandon-Warner, E., Li, T., Donohue, T. M. Jr. & Schrum, L. W. Rev-erb agonist and TGF-beta similarly affect autophagy but differentially regulate hepatic stellate cell fibrogenic phenotype. Int. J. Biochem. Cell Biol. 81, 137–147 (2016).

    Article  CAS  PubMed  Google Scholar 

  186. Chung, S. et al. Impact of circadian nuclear receptor REV-ERBalpha on midbrain dopamine production and mood regulation. Cell 157, 858–868 (2014).

    Article  CAS  PubMed  Google Scholar 

  187. Chen, Z., Yoo, S. H. & Takahashi, J. S. Small molecule modifiers of circadian clocks. Cell. Mol. Life Sci. 70, 2985–2998 (2013).

    Article  CAS  PubMed  Google Scholar 

  188. 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 Central  PubMed  Google Scholar 

  189. Kojetin, D. J. & Burris, T. P. REV-ERB and ROR nuclear receptors as drug targets. Nat. Rev. Drug Discov. 13, 197–216 (2014).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  190. Chang, M. R. et al. Antiobesity effect of a small molecule repressor of RORgamma. Mol. Pharmacol. 88, 48–56 (2015).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  191. Huh, J. R. et al. Digoxin and its derivatives suppress TH17 cell differentiation by antagonizing RORγt activity. Nature 472, 486–490 (2011).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  192. 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 

  193. Solt, L. A. et al. Suppression of TH17 differentiation and autoimmunity by a synthetic ROR ligand. Nature 472, 491–494 (2011).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  194. Hirota, T. et al. Identification of small molecule activators of cryptochrome. Science 337, 1094–1097 (2012).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  195. Nangle, S., Xing, W. & Zheng, N. Crystal structure of mammalian cryptochrome in complex with a small molecule competitor of its ubiquitin ligase. Cell Res. 23, 1417–1419 (2013).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  196. Humphries, P. S. et al. Carbazole-containing sulfonamides and sulfamides: discovery of cryptochrome modulators as antidiabetic agents. Bioorg. Med. Chem. Lett. 26, 757–760 (2016).

    Article  CAS  PubMed  Google Scholar 

  197. Miller, S. et al. Isoform-selective regulation of mammalian cryptochromes. Nat. Chem. Biol. 16, 676–685 (2020).

    Article  CAS  PubMed  Google Scholar 

  198. Nangle, S. N. et al. Molecular assembly of the period-cryptochrome circadian transcriptional repressor complex. eLife 3, e03674 (2014).

    Article  PubMed Central  PubMed  Google Scholar 

  199. Walton, K. M. et al. Selective inhibition of casein kinase 1ϵ minimally alters circadian clock period. J. Pharmacol. Exp. Ther. 330, 430–439 (2009).

    Article  CAS  PubMed  Google Scholar 

  200. Meng, Q. J. et al. Entrainment of disrupted circadian behavior through inhibition of casein kinase 1 (CK1) enzymes. Proc. Natl Acad. Sci. USA 107, 15240–15245 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  201. Hirota, T. et al. High-throughput chemical screen identifies a novel potent modulator of cellular circadian rhythms and reveals CKIalpha as a clock regulatory kinase. PLoS Biol. 8, e1000559 (2010).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  202. Barion, A. & Zee, P. C. A clinical approach to circadian rhythm sleep disorders. Sleep Med. 8, 566–577 (2007).

    Article  PubMed Central  PubMed  Google Scholar 

  203. LeGates, T. A., Fernandez, D. C. & Hattar, S. Light as a central modulator of circadian rhythms, sleep and affect. Nat. Rev. Neurosci. 15, 443–454 (2014).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  204. Asher, G. & Sassone-Corsi, P. Time for food: the intimate interplay between nutrition, metabolism, and the circadian clock. Cell 161, 84–92 (2015).

    Article  CAS  PubMed  Google Scholar 

  205. 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 Central  PubMed  Google Scholar 

  206. Jakubowicz, D., Barnea, M., Wainstein, J. & Froy, O. High caloric intake at breakfast vs. dinner differentially influences weight loss of overweight and obese women. Obesity 21, 2504–2512 (2013).

    Article  CAS  PubMed  Google Scholar 

  207. 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 Central  PubMed  Google Scholar 

  208. Colles, S. L., Dixon, J. B. & O’Brien, P. E. Night eating syndrome and nocturnal snacking: association with obesity, binge eating and psychological distress. Int. J. Obes. 31, 1722–1730 (2007).

    Article  CAS  Google Scholar 

  209. Markwald, R. R. et al. Impact of insufficient sleep on total daily energy expenditure, food intake, and weight gain. Proc. Natl Acad. Sci. USA 110, 5695–5700 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  210. Mrosovsky, N. & Salmon, P. A. A behavioural method for accelerating re-entrainment of rhythms to new light-dark cycles. Nature 330, 372–373 (1987).

    Article  CAS  PubMed  Google Scholar 

  211. Maywood, E. S., Mrosovsky, N., Field, M. D. & Hastings, M. H. Rapid down-regulation of mammalian period genes during behavioral resetting of the circadian clock. Proc. Natl Acad. Sci. USA 96, 15211–15216 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  212. Welsh, D., Richardson, G. S. & Dement, W. C. Effect of running wheel availability on circadian patterns of sleep and wakefulness in mice. Physiol. Behav. 43, 771–777 (1988).

    Article  CAS  PubMed  Google Scholar 

  213. Buxton, O. M., Lee, C. W., L’Hermite-Baleriaux, M., Turek, F. W. & Van Cauter, E. Exercise elicits phase shifts and acute alterations of melatonin that vary with circadian phase. Am. J. Physiol. Regul. Integr. Comp. Physiol. 284, R714–R724 (2003).

    Article  CAS  PubMed  Google Scholar 

  214. Reid, K. J. et al. Aerobic exercise improves self-reported sleep and quality of life in older adults with insomnia. Sleep Med. 11, 934–940 (2010).

    Article  PubMed Central  PubMed  Google Scholar 

  215. Pallier, P. N. et al. Pharmacological imposition of sleep slows cognitive decline and reverses dysregulation of circadian gene expression in a transgenic mouse model of Huntington’s disease. J. Neurosci. 27, 7869–7878 (2007).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  216. Hofstra, W. A. & de Weerd, A. W. How to assess circadian rhythm in humans: a review of literature. Epilepsy Behav. 13, 438–444 (2008).

    Article  PubMed  Google Scholar 

  217. Horne, J. A. & Ostberg, O. A self-assessment questionnaire to determine morningness-eveningness in human circadian rhythms. Int. J. Chronobiol. 4, 97–110 (1976).

    CAS  PubMed  Google Scholar 

  218. Roenneberg, T. et al. Epidemiology of the human circadian clock. Sleep Med. Rev. 11, 429–438 (2007).

    Article  PubMed  Google Scholar 

  219. Ancoli-Israel, S. et al. The role of actigraphy in the study of sleep and circadian rhythms. Sleep 26, 342–392 (2003).

    Article  PubMed  Google Scholar 

  220. Gill, S. & Panda, S. A smartphone app reveals erratic diurnal eating patterns in humans that can be modulated for health benefits. Cell Metab. 22, 789–798 (2015).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  221. Pagani, L. et al. The physiological period length of the human circadian clock in vivo is directly proportional to period in human fibroblasts. PLoS ONE 5, e13376 (2010).

    Article  PubMed Central  PubMed  Google Scholar 

  222. Akashi, M. et al. Noninvasive method for assessing the human circadian clock using hair follicle cells. Proc. Natl Acad. Sci. USA 107, 15643–15648 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  223. Kasukawa, T. et al. Human blood metabolite timetable indicates internal body time. Proc. Natl Acad. Sci. USA 109, 15036–15041 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  224. Chen, C. Y. et al. Effects of aging on circadian patterns of gene expression in the human prefrontal cortex. Proc. Natl Acad. Sci. USA 113, 206–211 (2016).

    Article  CAS  PubMed  Google Scholar 

  225. van Maanen, A., Meijer, A. M., van der Heijden, K. B. & Oort, F. J. The effects of light therapy on sleep problems: a systematic review and meta-analysis. Sleep Med. Rev. 29, 52–62 (2016).

    Article  PubMed  Google Scholar 

  226. Johnston, J. D. Physiological responses to food intake throughout the day. Nutr. Res. Rev. 27, 107–118 (2014).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  227. Liu, J. et al. MT1 and MT2 melatonin receptors: a therapeutic perspective. Annu. Rev. Pharmacol. Toxicol. 56, 361–383 (2016).

    Article  CAS  PubMed  Google Scholar 

  228. Seki, T. et al. Nobiletin-rich citrus reticulata peels, a kampo medicine for Alzheimer’s disease: a case series. Geriatr. Gerontol. Int. 13, 236–238 (2013).

    Article  PubMed  Google Scholar 

  229. Kondratova, A. A. & Kondratov, R. V. The circadian clock and pathology of the ageing brain. Nat. Rev. Neurosci. 13, 325–335 (2012).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  230. Telias, I. & Wilcox, M. E. Sleep and circadian rhythm in critical illness. Crit. Care 23, 82 (2019).

    Article  PubMed Central  PubMed  Google Scholar 

  231. Jang, J. et al. The cryptochrome inhibitor KS15 enhances E-box-mediated transcription by disrupting the feedback action of a circadian transcription-repressor complex. Life Sci. 200, 49–55 (2018).

    Article  CAS  PubMed  Google Scholar 

  232. Shinozaki, A. et al. Potent effects of flavonoid nobiletin on amplitude, period, and phase of the circadian clock rhythm in PER2::LUCIFERASE mouse embryonic fibroblasts. PLoS ONE 12, e0170904 (2017).

    Article  PubMed Central  PubMed  Google Scholar 

  233. Mulvihill, E. E., Burke, A. C. & Huff, M. W. Citrus flavonoids as regulators of lipoprotein metabolism and atherosclerosis. Annu. Rev. Nutr. 36, 275–299 (2016).

    Article  CAS  PubMed  Google Scholar 

  234. Helleboid, S. et al. The identification of naturally occurring neoruscogenin as a bioavailable, potent, and high-affinity agonist of the nuclear receptor RORalpha (NR1F1). J. Biomol. Screen. 19, 399–406 (2014).

    Article  CAS  PubMed  Google Scholar 

  235. Kumar, N. et al. Identification of SR3335 (ML-176): a synthetic RORalpha selective inverse agonist. ACS Chem. Biol. 6, 218–222 (2011).

    Article  CAS  PubMed  Google Scholar 

  236. Byun, J. K. et al. Retinoic acid-related orphan receptor alpha reprograms glucose metabolism in glutamine-deficient hepatoma cells. Hepatology 61, 953–964 (2015).

    Article  CAS  PubMed  Google Scholar 

  237. He, B. & Chen, Z. Molecular targets for small-molecule modulators of circadian clocks. Curr. Drug Metab. 17, 503–512 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  238. Eltzschig, H. K., Bonney, S. K. & Eckle, T. Attenuating myocardial ischemia by targeting A2B adenosine receptors. Trends Mol. Med. 19, 345–354 (2013).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  239. Li, J., Lu, W. Q., Beesley, S., Loudon, A. S. & Meng, Q. J. Lithium impacts on the amplitude and period of the molecular circadian clockwork. PLoS ONE 7, e33292 (2012).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  240. Welsh, D. K., Takahashi, J. S. & Kay, S. A. Suprachiasmatic nucleus: cell autonomy and network properties. Annu. Rev. Physiol. 72, 551–577 (2010).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  241. Sonoda, T. et al. A noncanonical inhibitory circuit dampens behavioral sensitivity to light. Science 368, 527–531 (2020). This important article indicates that the inhibitory neurotransmitter GABA released by a subpopulation of intrinsically photosensitive retinal ganglion cells decreases the sensitivity of non-image-forming behaviours at low light levels.

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  242. Shimomura, K. et al. Genetic suppression of the circadian Clock mutation by the melatonin biosynthesis pathway. Proc. Natl Acad. Sci. USA 107, 8399–8403 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  243. Knutsson, A., Jonsson, B. G., Akerstedt, T. & Orth-Gomer, K. Increased risk of ischaemic heart disease in shift workers. Lancet 328, 89–92 (1986).

    Article  Google Scholar 

  244. Panda, S. Circadian physiology of metabolism. Science 354, 1008–1015 (2016).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  245. Savvidis, C. & Koutsilieris, M. Circadian rhythm disruption in cancer biology. Mol. Med. 18, 1249–1260 (2012).

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  246. Kelleher, F. C., Rao, A. & Maguire, A. Circadian molecular clocks and cancer. Cancer Lett. 342, 9–18 (2014).

    Article  CAS  PubMed  Google Scholar 

  247. Vgontzas, A. N. & Chrousos, G. P. Sleep, the hypothalamic-pituitary-adrenal axis, and cytokines: multiple interactions and disturbances in sleep disorders. Endocrinol. Metab. Clin. North Am. 31, 15–36 (2002).

    Article  CAS  PubMed  Google Scholar 

  248. Sephton, S. & Spiegel, D. Circadian disruption in cancer: a neuroendocrine-immune pathway from stress to disease? Brain Behav. Immun. 17, 321–328 (2003).

    Article  CAS  PubMed  Google Scholar 

  249. Leng, Y., Musiek, E. S., Hu, K., Cappuccio, F. P. & Yaffe, K. Association between circadian rhythms and neurodegenerative diseases. Lancet Neurol. 18, 307–318 (2019).

    Article  PubMed Central  PubMed  Google Scholar 

  250. Chen, Z. et al. Identification of diverse modulators of central and peripheral circadian clocks by high-throughput chemical screening. Proc. Natl Acad. Sci. USA 109, 101–106 (2012).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

This work was in part supported by US National Institute of Health grants R01HL154720, R01DK122796, R01DK109574, R01HL133900 and Department of Defense Grant W81XWH2110032 to H.K.E., an American Thoracic Society unrestricted grant, American Heart Association grant 19CDA34660279, American Lung Association grant CA-622265, the Center for Clinical and Translational Sciences pilot project award 1UL1TR003167–01 and a Parker B. Francis Fellowship to X.Y., and National Natural Science Foundation of China grants 81201448 and 81972312 and Natural Science Foundation of Hunan Province grant 2018JJ3736 to W.R. The authors thank S.-H. Yoo and Z. Chen for their help with the initial draft of the manuscript. The authors acknowledge Y. Wang and K. Wallen for assisting with manuscript editing.

Author information

Authors and Affiliations

Authors

Contributions

W.R. And X.Y. contributed equally and were involved in all aspects of this article. H.K.E. contributed substantially to discussion of the content and reviewed and edited the manuscript before submission.

Corresponding author

Correspondence to Holger K. Eltzschig.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note

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

Glossary

Zeitgebers

A German word (meaning ‘time giver’), from Zeit (meaning ‘time’) and Geber (meaning ‘giver’). It refers to any external or intracellular cues that can reset or entrain an organism’s biological rhythms to the 24-hour day–night cycle of Earth.

Phase advance

A phase shift in the circadian rhythm whereby the bedtime and wake-up time will move earlier in the day.

Non-rapid-eye-movement sleep

Also known as quiescent sleep, with little or no eye movement, rare dreaming and less muscle paralysation than in rapid eye movement sleep.

Rapid eye movement sleep

Also known as paradoxical sleep, a unique phase of sleep in mammals and birds characterized by rapid eye movement, low muscle tone throughout the body and vivid dreaming sometimes.

Phase dissociation

In the context of this Review, the dissociation of intrinsic circadian rhythm and the environmental light–dark cycle, as in jet lag and sleep disorders.

Delayed sleep phase syndrome

A circadian sleep disorder in which a person’s sleep is delayed by 2 hours or more beyond what is considered a conventional bedtime, thus causing difficulty in waking up in the morning.

Ischaemic preconditioning

An experimental technique (usually via repeated short episodes of ischaemia via coronary artery occlusion) to increase tolerance to the loss of blood supply, and thus oxygen, if there is a subsequent prolonged insult.

Circadian wheel-running rhythms

When rodents have free access to a running wheel, voluntary use of this wheel is active during the night and inactive during the day to generate a specific circadian rhythm, which serves as a particularly reliable and convenient measure of the output of the master circadian clock.

Phase delay

A phase shift in the circadian rhythm whereby the bedtime and wake-up time will move later in the day.

Lipopolysaccharide challenge

An experimental technique to elicit inflammation by administering lipopolysaccharide via intraperitoneal injection to mice.

Caecal ligation and puncture

A commonly used preclinical rodent model to study sepsis via ligation and perforation of the caecum, resulting in polymicrobial infection and systemic inflammation.

Zeitgeber time

A standardized 24-hour notation for the phase in an entrained circadian cycle with reference to environmental regularities or zeitgebers.

Chronobiotic

An agent that is able to influence, directly or indirectly, the phase and/or the period of the body clock.

Chronotypes

Characterized by individual differences in the timing of sleep–wake schedules, biological parameters (such as core body temperature, melatonin and cortisol) and cognitive performance (such as attention).

Circadian time

A notation for the phase in a circadian cycle that represents an organism’s endogenous circadian clock, without reference to any environmental regularities or zeitgebers.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ruan, W., Yuan, X. & Eltzschig, H.K. Circadian rhythm as a therapeutic target. Nat Rev Drug Discov 20, 287–307 (2021). https://doi.org/10.1038/s41573-020-00109-w

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41573-020-00109-w

This article is cited by

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research