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:

Clocking in to immunity

Abstract

Circadian rhythms are a ubiquitous feature of virtually all living organisms, regulating a wide diversity of physiological systems. It has long been established that the circadian clockwork plays a key role in innate immune responses, and recent studies reveal that several aspects of adaptive immunity are also under circadian control. We discuss the latest insights into the genetic and biochemical mechanisms linking immunity to the core circadian clock of the cell and hypothesize as to why the immune system is so tightly controlled by circadian oscillations. Finally, we consider implications for human health, including vaccination strategies and the emerging field of chrono-immunotherapy.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Operation and disruption of the molecular clock in immune cells.
Fig. 2: Regulation of innate immunity in the macrophage and the lung.
Fig. 3: Regulation of adaptive immunity in the lymph node.
Fig. 4: Interactions of the clock, microbiome and immune function.
Fig. 5: Chronotherapy and the clock as a therapeutic target.

Similar content being viewed by others

References

  1. Edgar, R. S. et al. Peroxiredoxins are conserved markers of circadian rhythms. Nature 485, 459–464 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  2. Pittendrigh, C. S. Temporal organization: reflections of a Darwinian clock-watcher. Annu. Rev. Physiol. 55, 16–54 (1993).

    Article  PubMed  CAS  Google Scholar 

  3. Brown, T. M. & Piggins, H. D. Electrophysiology of the suprachiasmatic circadian clock. Prog. Neurobiol. 82, 229–255 (2007).

    Article  PubMed  CAS  Google Scholar 

  4. Brancaccio, M. et al. Network-mediated encoding of circadian time: the suprachiasmatic nucleus (SCN) from genes to neurons to circuits, and back. J. Neurosci. 34, 15192–15199 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  5. Herzog, E. D., Hermanstyne, T., Smyllie, N. J. & Hastings, M. H. Regulating the suprachiasmatic nucleus (SCN) circadian clockwork: interplay between cell-autonomous and circuit-level mechanisms. Cold Spring Harb. Perspect. Biol. 9, a027706 (2017).

    Article  PubMed  CAS  Google Scholar 

  6. Buijs, F. N. et al. The circadian system: a regulatory feedback network of periphery and brain. Physiology 31, 170–181 (2016).

    Article  PubMed  Google Scholar 

  7. Dumbell, R., Matveeva, O. & Oster, H. Circadian clocks, stress, and immunity. Front. Endocrinol. 7, 37 (2016).

    Article  Google Scholar 

  8. Thaiss, C. A. et al. Microbiota diurnal rhythmicity programs host transcriptome oscillations. Cell 167, 1495–1510 (2016).

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  11. Spoelstra, K., Wikelski, M., Daan, S., Loudon, A. S. & Hau, M. Natural selection against a circadian clock gene mutation in mice. Proc. Natl Acad. Sci. USA 113, 686–691 (2016).

    Article  PubMed  CAS  Google Scholar 

  12. McHill, A. W. & Wright, K. P. Jr. Role of sleep and circadian disruption on energy expenditure and in metabolic predisposition to human obesity and metabolic disease. Obes. Rev. 18 (Suppl. 1), 15–24 (2017).

    Article  PubMed  Google Scholar 

  13. Qian, J. & Scheer, F. A. Circadian system and glucose metabolism: implications for physiology and disease. Trends Endocrinol. Metab. 27, 282–293 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Reitz, C. J. & Martino, T. A. Disruption of circadian rhythms and sleep on critical illness and the impact on cardiovascular events. Curr. Pharm. Des. 21, 3505–3511 (2015).

    Article  PubMed  CAS  Google Scholar 

  15. Yuan, X. et al. Night shift work increases the risks of multiple primary cancers in women: a systematic review and meta-analysis of 61 articles. Cancer Epidemiol. Biomarkers Prev. 27, 25–40 (2018).

    Article  PubMed  Google Scholar 

  16. Berenbaum, F. & Meng, Q. J. The brain-joint axis in osteoarthritis: nerves, circadian clocks and beyond. Nat. Rev. Rheumatol. 12, 508–516 (2016).

    Article  PubMed  Google Scholar 

  17. Cutolo, M. Rheumatoid arthritis: circadian and circannual rhythms in RA. Nat. Rev. Rheumatol. 7, 500–502 (2011).

    Article  PubMed  Google Scholar 

  18. 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  PubMed  CAS  Google Scholar 

  19. Curtis, A. M., Bellet, M. M., Sassone-Corsi, P. & O’Neill, L. A. Circadian clock proteins and immunity. Immunity 40, 178–186 (2014).

    Article  PubMed  CAS  Google Scholar 

  20. Labrecque, N. & Cermakian, N. Circadian clocks in the immune system. J. Biol. Rhythms 30, 277–290 (2015).

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  22. Scheiermann, C., Kunisaki, Y. & Frenette, P. S. Circadian control of the immune system. Nat. Rev. Immunol. 13, 190–198 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Nguyen, K. D. et al. Circadian gene Bmal1 regulates diurnal oscillations of Ly6C(hi) inflammatory monocytes. Science 341, 1483–1488 (2013). This paper shows that oscillations in the number of inflammatory monocytes in tissues are driven by the myeloid cell clock.

    Article  PubMed  CAS  Google Scholar 

  24. Keller, M. et al. A circadian clock in macrophages controls inflammatory immune responses. Proc. Natl Acad. Sci. USA 106, 21407–21412 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Silver, A. C., Arjona, A., Hughes, M. E., Nitabach, M. N. & Fikrig, E. Circadian expression of clock genes in mouse macrophages, dendritic cells, and B cells. Brain Behav. Immun. 26, 407–413 (2012).

    Article  PubMed  CAS  Google Scholar 

  26. Wang, X., Reece, S. P., Van Scott, M. R. & Brown, J. M. A circadian clock in murine bone marrow-derived mast cells modulates IgE-dependent activation in vitro. Brain Behav. Immun. 25, 127–134 (2011).

    Article  PubMed  CAS  Google Scholar 

  27. Baumann, A. et al. The circadian clock is functional in eosinophils and mast cells. Immunology 140, 465–474 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  28. Ella, K., Csepanyi-Komi, R. & Kaldi, K. Circadian regulation of human peripheral neutrophils. Brain Behav. Immun. 57, 209–221 (2016).

    Article  PubMed  CAS  Google Scholar 

  29. Arjona, A. & Sarkar, D. K. Circadian oscillations of clock genes, cytolytic factors, and cytokines in rat NK cells. J. Immunol. 174, 7618–7624 (2005).

    Article  PubMed  CAS  Google Scholar 

  30. Oliva-Ramirez, J., Moreno-Altamirano, M. M., Pineda-Olvera, B., Cauich-Sanchez, P. & Sanchez-Garcia, F. J. Crosstalk between circadian rhythmicity, mitochondrial dynamics and macrophage bactericidal activity. Immunology 143, 490–497 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  31. 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). This article presents the first demonstration that synthetic ligands targeting a circadian clock protein modify inflammatory responses.

    Article  PubMed  Google Scholar 

  32. Nakamura, Y., Ishimaru, K., Shibata, S. & Nakao, A. Regulation of plasma histamine levels by the mast cell clock and its modulation by stress. Sci. Rep. 7, 39934 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  33. Nakamura, Y. et al. Circadian regulation of allergic reactions by the mast cell clock in mice. J. Allergy Clin. Immunol. 133, 568–575 (2014).

    Article  PubMed  CAS  Google Scholar 

  34. Logan, R. W., Wynne, O., Levitt, D., Price, D. & Sarkar, D. K. Altered circadian expression of cytokines and cytolytic factors in splenic natural killer cells of Per1(−/−) mutant mice. J. Interferon Cytokine Res. 33, 108–114 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Logan, R. W. et al. Chronic shift-lag alters the circadian clock of NK cells and promotes lung cancer growth in rats. J. Immunol. 188, 2583–2591 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Lam, M. T. et al. Rev-Erbs repress macrophage gene expression by inhibiting enhancer-directed transcription. Nature 498, 511–515 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Sato, S. et al. A circadian clock gene, Rev-erbα, modulates the inflammatory function of macrophages through the negative regulation of Ccl2 expression. J. Immunol. 192, 407–417 (2014).

    Article  PubMed  CAS  Google Scholar 

  38. Narasimamurthy, R. et al. Circadian clock protein cryptochrome regulates the expression of proinflammatory cytokines. Proc. Natl Acad. Sci. USA 109, 12662–12667 (2012).

    Article  PubMed  PubMed Central  Google Scholar 

  39. Born, J., Lange, T., Hansen, K., Molle, M. & Fehm, H. L. Effects of sleep and circadian rhythm on human circulating immune cells. J. Immunol. 158, 4454–4464 (1997).

    PubMed  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Prendergast, B. J. et al. Impaired leukocyte trafficking and skin inflammatory responses in hamsters lacking a functional circadian system. Brain Behav. Immun. 32, 94–104 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  42. Haspel, J. A. et al. Circadian rhythm reprogramming during lung inflammation. Nat. Commun. 5, 4753 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Gibbs, J. et al. An epithelial circadian clock controls pulmonary inflammation and glucocorticoid action. Nat. Med. 20, 919–926 (2014). This paper shows circadian regulation of neutrophil influx via rhythmic inhibition of chemoattractant production by bronchial epithelial cells in the lung.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Kiessling, S. et al. The circadian clock in immune cells controls the magnitude of Leishmania parasite infection. Sci. Rep. 7, 10892 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Casanova-Acebes, M. et al. Rhythmic modulation of the hematopoietic niche through neutrophil clearance. Cell 153, 1025–1035 (2013). This study describes how rhythmic homing of aged neutrophils generates homeostatic cues that trigger release of haematopoietic stem cells from the bone marrow.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Nussbaum, J. C. et al. Type 2 innate lymphoid cells control eosinophil homeostasis. Nature 502, 245–248 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Chong, S. Z. et al. CXCR4 identifies transitional bone marrow premonocytes that replenish the mature monocyte pool for peripheral responses. J. Exp. Med. 213, 2293–2314 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  48. Bellet, M. M. et al. Circadian clock regulates the host response to Salmonella. Proc. Natl Acad. Sci. USA 110, 9897–9902 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Rijo-Ferreira, F., Pinto-Neves, D., Barbosa-Morais, N. L., Takahashi, J. S. & Figueiredo, L. M. Trypanosoma brucei metabolism is under circadian control. Nat. Microbiol. 2, 17032 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Majumdar, T., Dhar, J., Patel, S., Kondratov, R. & Barik, S. Circadian transcription factor BMAL1 regulates innate immunity against select RNA viruses. Innate Immun. 23, 147–154 (2017).

    Article  PubMed  CAS  Google Scholar 

  51. Edgar, R. S. et al. Cell autonomous regulation of herpes and influenza virus infection by the circadian clock. Proc. Natl Acad. Sci. USA 113, 10085–10090 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  52. Fernandes, G., Halberg, F., Yunis, E. J. & Good, R. A. Circadian rhythmic plaque-forming cell response of spleens from mice immunized with SRBC. J. Immunol. 117, 962–966 (1976).

    PubMed  CAS  Google Scholar 

  53. Kaplan, M. S. et al. Circadian rhythm of stimulated lymphocyte blastogenesis. A 24 h cycle in the mixed leukocyte culture reaction and with SKSD stimulation. J. Allergy Clin. Immunol. 58, 180–189 (1976).

    Article  CAS  Google Scholar 

  54. Druzd, D. et al. Lymphocyte circadian clocks control lymph node trafficking and adaptive immune responses. Immunity 46, 120–132 (2017). This study shows dynamic regulation of lymphocyte numbers in blood, lymph node and lymph, driven by rhythmic expression of migration and egress factors.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Hemmers, S. & Rudensky, A. Y. The cell-intrinsic circadian clock is dispensable for lymphocyte differentiation and function. Cell Rep. 11, 1339–1349 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  56. Bollinger, T. et al. Circadian clocks in mouse and human CD4 + T cells. PLoS ONE 6, e29801 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Sun, Y. et al. MOP3, a component of the molecular clock, regulates the development of B cells. Immunology 119, 451–460 (2006).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Yu, X. et al. TH17 cell differentiation is regulated by the circadian clock. Science 342, 727–730 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Yu, X. et al. The basic leucine zipper transcription factor NFIL3 directs the development of a common innate lymphoid cell precursor. eLife 3, e04406 (2014).

    Article  PubMed Central  CAS  Google Scholar 

  60. Qiu, J. et al. Group 3 innate lymphoid cells inhibit T-cell-mediated intestinal inflammation through aryl hydrocarbon receptor signaling and regulation of microflora. Immunity 39, 386–399 (2013).

    Article  PubMed  CAS  Google Scholar 

  61. Farez, M. F. et al. Melatonin contributes to the seasonality of multiple sclerosis relapses. Cell 162, 1338–1352 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  62. Martinez-Llordella, M. et al. CD28-inducible transcription factor DEC1 is required for efficient autoreactive CD4 + T cell response. J. Exp. Med. 210, 1603–1619 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  63. Kreslavsky, T. et al. Essential role for the transcription factor Bhlhe41 in regulating the development, self-renewal and BCR repertoire of B-1a cells. Nat. Immunol. 18, 442–455 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  64. Besedovsky, L., Born, J. & Lange, T. Endogenous glucocorticoid receptor signaling drives rhythmic changes in human T-cell subset numbers and the expression of the chemokine receptor CXCR4. FASEB J. 28, 67–75 (2014).

    Article  PubMed  CAS  Google Scholar 

  65. Dimitrov, S. et al. Cortisol and epinephrine control opposing circadian rhythms in T cell subsets. Blood 113, 5134–5143 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Suzuki, K., Hayano, Y., Nakai, A., Furuta, F. & Noda, M. Adrenergic control of the adaptive immune response by diurnal lymphocyte recirculation through lymph nodes. J. Exp. Med. 213, 2567–2574 (2016). This study shows how adrenergic tone modulates lymphocyte trafficking rhythms and humoral immune responses through β2-adrenergic receptors expressed by lymphocytes.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Zhao, Y. et al. Uncovering the mystery of opposite circadian rhythms between mouse and human leukocytes in humanized mice. Blood 130, 1995–2005 (2017). This is a very interesting report of cell-intrinsic phase encoding of leukocytes, driven by opposite effects of p38MAPK–MK2 signalling upon HIF1α induction and CXCR4 expression.

    Article  PubMed  CAS  Google Scholar 

  68. 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). This work illustrates an immune-enhancing role of glucocorticoids via upregulation of homing receptors, promoting rhythmic T cell accumulation and heightened responses to systemic infection.

    Article  PubMed  CAS  Google Scholar 

  69. Forster, R., Davalos-Misslitz, A. C. & Rot, A. CCR7 and its ligands: balancing immunity and tolerance. Nat. Rev. Immunol. 8, 362–371 (2008).

    Article  PubMed  CAS  Google Scholar 

  70. Stein, J. V. & Nombela-Arrieta, C. Chemokine control of lymphocyte trafficking: a general overview. Immunology 116, 1–12 (2005).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Cyster, J. G. & Schwab, S. R. Sphingosine-1-phosphate and lymphocyte egress from lymphoid organs. Annu. Rev. Immunol. 30, 69–94 (2012).

    Article  PubMed  CAS  Google Scholar 

  72. Nakai, A., Hayano, Y., Furuta, F., Noda, M. & Suzuki, K. Control of lymphocyte egress from lymph nodes through β2-adrenergic receptors. J. Exp. Med. 211, 2583–2598 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  73. Esquifino, A. I., Selgas, L., Arce, A., Maggiore, V. D. & Cardinali, D. P. Twenty-four-hour rhythms in immune responses in rat submaxillary lymph nodes and spleen: effect of cyclosporine. Brain Behav. Immun. 10, 92–102 (1996).

    Article  PubMed  CAS  Google Scholar 

  74. Fortier, E. E. et al. Circadian variation of the response of T cells to antigen. J. Immunol. 187, 6291–6300 (2011).

    Article  PubMed  CAS  Google Scholar 

  75. 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  PubMed  PubMed Central  CAS  Google Scholar 

  76. Sutton, C. E. et al. Loss of the molecular clock in myeloid cells exacerbates T cell-mediated CNS autoimmune disease. Nat. Commun. 8, 1923 (2017). This report shows the importance of appropriate immune cell crosstalk in an EAE model, as disruption of the myeloid clock increases T H 1 and T H 17 cell responses in the central nervous system.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  77. Long, J. E. et al. Morning vaccination enhances antibody response over afternoon vaccination: a cluster-randomised trial. Vaccine 34, 2679–2685 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  78. 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  PubMed  PubMed Central  CAS  Google Scholar 

  79. Huo, M. et al. Myeloid Bmal1 deletion increases monocyte recruitment and worsens atherosclerosis. FASEB J. 31, 1097–1106 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  80. 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  PubMed  CAS  Google Scholar 

  81. Wang, Y. et al. The intestinal microbiota regulates body composition through NFIL3 and the circadian clock. Science 357, 912–916 (2017). Along with previous work from the same authors (reference 58), this study links the clock protein REV-ERBα with rhythmic expression of NFIL3, a critical regulator of both T H 17 cell differentiation and enterocyte lipid metabolism.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  82. Zhang, D. et al. Neutrophil ageing is regulated by the microbiome. Nature 525, 528–532 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  83. Martin, C. et al. Chemokines acting via CXCR2 and CXCR4 control the release of neutrophils from the bone marrow and their return following senescence. Immunity 19, 583–593 (2003).

    Article  PubMed  CAS  Google Scholar 

  84. Tanji-Matsuba, K. et al. Functional changes in aging polymorphonuclear leukocytes. Circulation 97, 91–98 (1998).

    Article  PubMed  CAS  Google Scholar 

  85. 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  PubMed  PubMed Central  CAS  Google Scholar 

  86. Thaiss, C. A. et al. Transkingdom control of microbiota diurnal oscillations promotes metabolic homeostasis. Cell 159, 514–529 (2014).

    Article  PubMed  CAS  Google Scholar 

  87. Balsalobre, A., Damiola, F. & Schibler, U. A serum shock induces circadian gene expression in mammalian tissue culture cells. Cell 93, 929–937 (1998).

    Article  PubMed  CAS  Google Scholar 

  88. Balsalobre, A. et al. Resetting of circadian time in peripheral tissues by glucocorticoid signaling. Science 289, 2344–2347 (2000).

    Article  PubMed  CAS  Google Scholar 

  89. Shackelford, P. G. & Feigin, R. D. Periodicity of susceptibility to pneumococcal infection: influence of light and adrenocortical secretions. Science 182, 285–287 (1973).

    Article  PubMed  CAS  Google Scholar 

  90. Early, J. O. & Curtis, A. M. Immunometabolism: Is it under the eye of the clock? Semin. Immunol. 28, 478–490 (2016).

    Article  PubMed  CAS  Google Scholar 

  91. Ando, N. et al. Circadian gene clock regulates psoriasis-like skin inflammation in mice. J. Invest. Dermatol. 135, 3001–3008 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  92. Castanon-Cervantes, O. et al. Dysregulation of inflammatory responses by chronic circadian disruption. J. Immunol. 185, 5796–5805 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  94. 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  PubMed  PubMed Central  CAS  Google Scholar 

  95. Li, W. Q., Qureshi, A. A., Schernhammer, E. S. & Han, J. Rotating night-shift work and risk of psoriasis in US women. J. Invest. Dermatol. 133, 565–567 (2013).

    Article  PubMed  CAS  Google Scholar 

  96. 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 

  97. Cuesta, M., Boudreau, P., Dubeau-Laramee, G., Cermakian, N. & Boivin, D. B. Simulated night shift disrupts circadian rhythms of immune functions in humans. J. Immunol. 196, 2466–2475 (2016).

    Article  PubMed  CAS  Google Scholar 

  98. Durrington, H. J., Farrow, S. N., Loudon, A. S. & Ray, D. W. The circadian clock and asthma. Thorax 69, 90–92 (2014).

    Article  PubMed  Google Scholar 

  99. Olsen, N. J., Brooks, R. H. & Furst, D. Variability of immunologic and clinical features in patients with rheumatoid arthritis studied over 24 h. J. Rheumatol. 20, 940–943 (1993).

    PubMed  CAS  Google Scholar 

  100. Panzer, S. E., Dodge, A. M., Kelly, E. A. & Jarjour, N. N. Circadian variation of sputum inflammatory cells in mild asthma. J. Allergy Clin. Immunol. 111, 308–312 (2003).

    Article  PubMed  Google Scholar 

  101. Perry, M. G., Kirwan, J. R., Jessop, D. S. & Hunt, L. P. Overnight variations in cortisol, interleukin 6, tumour necrosis factor alpha and other cytokines in people with rheumatoid arthritis. Ann. Rheum. Dis. 68, 63–68 (2009).

    Article  PubMed  CAS  Google Scholar 

  102. Takeda, N. & Maemura, K. Circadian clock and the onset of cardiovascular events. Hypertens. Res. 39, 383–390 (2016).

    Article  PubMed  CAS  Google Scholar 

  103. Culic, V. Daylight saving time transitions and acute myocardial infarction. Chronobiol. Int. 30, 662–668 (2013).

    Article  PubMed  Google Scholar 

  104. Dopico, X. C. et al. Widespread seasonal gene expression reveals annual differences in human immunity and physiology. Nat. Commun. 6, 7000 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  105. Donaldson, G. C. & Wedzicha, J. A. The causes and consequences of seasonal variation in COPD exacerbations. Int. J. Chron. Obstruct Pulmon. Dis. 9, 1101–1110 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  106. Spelman, T. et al. Seasonal variation of relapse rate in multiple sclerosis is latitude dependent. Ann. Neurol. 76, 880–890 (2014).

    Article  PubMed  Google Scholar 

  107. 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). This article presents a comprehensive analysis of oscillating genes and non-coding RNAs in murine organs, highlighting the relationships between rhythmic genes, disease-associated genes and current drug targets.

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  108. Buttgereit, F. et al. Efficacy of modified-release versus standard prednisone to reduce duration of morning stiffness of the joints in rheumatoid arthritis (CAPRA-1): a double-blind, randomised controlled trial. Lancet 371, 205–214 (2008).

    Article  PubMed  CAS  Google Scholar 

  109. Lamia, K. A. et al. Cryptochromes mediate rhythmic repression of the glucocorticoid receptor. Nature 480, 552–556 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  110. Okabe, T. et al. REV-ERBalpha influences the stability and nuclear localization of the glucocorticoid receptor. J. Cell Sci. 129, 4143–4154 (2016).

    PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  112. Sitaula, S., Billon, C., Kamenecka, T. M., Solt, L. A. & Burris, T. P. Suppression of atherosclerosis by synthetic REV-ERB agonist. Biochem. Biophys. Res. Commun. 460, 566–571 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  113. Kojetin, D., Wang, Y., Kamenecka, T. M. & Burris, T. P. Identification of SR8278, a synthetic antagonist of the nuclear heme receptor REV-ERB. ACS Chem. Biol. 6, 131–134 (2011).

    Article  PubMed  CAS  Google Scholar 

  114. Gagnidze, K. et al. Nuclear receptor REV-ERBα mediates circadian sensitivity to mortality in murine vesicular stomatitis virus-induced encephalitis. Proc. Natl Acad. Sci. USA 113, 5730–5735 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  116. Hand, L. E. et al. The circadian clock regulates inflammatory arthritis. FASEB J. 30, 3759–3770 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  117. Partch, C. L., Green, C. B. & Takahashi, J. S. Molecular architecture of the mammalian circadian clock. Trends Cell Biol. 24, 90–99 (2014).

    Article  PubMed  CAS  Google Scholar 

  118. Guilding, C. et al. Suppressed cellular oscillations in after-hours mutant mice are associated with enhanced circadian phase-resetting. J. Physiol. 591, 1063–1080 (2013).

    Article  PubMed  CAS  Google Scholar 

  119. Meng, Q. J. et al. Setting clock speed in mammals: the CK1 epsilon tau mutation in mice accelerates circadian pacemakers by selectively destabilizing PERIOD proteins. Neuron 58, 78–88 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  120. Yamaguchi, S. et al. Role of DBP in the circadian oscillatory mechanism. Mol. Cell. Biol. 20, 4773–4781 (2000).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  121. Ueda, H. R. et al. System-level identification of transcriptional circuits underlying mammalian circadian clocks. Nat. Genet. 37, 187–192 (2005).

    Article  PubMed  CAS  Google Scholar 

  122. Fu, L. & Lee, C. C. The circadian clock: pacemaker and tumour suppressor. Nat. Rev. Cancer 3, 350–361 (2003).

    Article  PubMed  CAS  Google Scholar 

  123. Yu, E. A. & Weaver, D. R. Disrupting the circadian clock: gene-specific effects on aging, cancer, and other phenotypes. Aging 3, 479–493 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

  125. Cho, H. et al. Regulation of circadian behaviour and metabolism by REV-ERB-alpha and REV-ERB-beta. Nature 485, 123–127 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  126. Zhang, Y. et al. Discrete functions of nuclear receptor Rev-erbα couple metabolism to the clock. Science 348, 1488–1492 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  128. Sulli, G. et al. Pharmacological activation of REV-ERBs is lethal in cancer and oncogene-induced senescence. Nature 553, 351–355 (2018). This study shows that agonism of REV-ERBs is specifically lethal to cancer cells via inhibition of autophagy and lipogenesis

  129. Fonken, L. K. et al. Microglia inflammatory responses are controlled by an intrinsic circadian clock. Brain Behav. Immun. 45, 171–179 (2015).

    Article  PubMed  CAS  Google Scholar 

  130. Rudic, R. D. et al. Bioinformatic analysis of circadian gene oscillation in mouse aorta. Circulation 112, 2716–2724 (2005).

    Article  PubMed  CAS  Google Scholar 

  131. Nakazato, R. et al. The intrinsic microglial clock system regulates interleukin-6 expression. Glia 65, 198–208 (2017).

    Article  PubMed  Google Scholar 

  132. Alvarez-Sanchez, N. et al. Melatonin controls experimental autoimmune encephalomyelitis by altering the T effector/regulatory balance. Brain Behav. Immun. 50, 101–114 (2015).

    Article  PubMed  CAS  Google Scholar 

  133. Borniger, J. C. et al. Time-of-day dictates transcriptional inflammatory responses to cytotoxic chemotherapy. Sci. Rep 7, 41220 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  134. Durrington, H. J., Farrow, S. N. & Ray, D. Recent advances in chronotherapy for the management of asthma. ChronoPhysiology Ther. 4, 125–135 (2014).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors thank V. Lavilla for creating the video and M. F. Loudon for providing the voice-over to it. C.S. is funded by the German Research Foundation (DFG) (Emmy-Noether grant (SCHE 1645/2-1) and SFB914 projects B09 and Z03), the European Research Council (ERC; starting grant 635872, CIRCODE), the DZHK (German Centre for Cardiovascular Research) and the BMBF (German Ministry of Education and Research). J.G. is an Arthritis Research UK Career Development Fellow (Ref. 20629). A.L. acknowledges the support of the Wellcome Trust (grant 107851/Z/15/Z).

Author information

Authors and Affiliations

Authors

Contributions

All authors contributed to the research, discussion of content, writing and review of this manuscript.

Corresponding authors

Correspondence to Christoph Scheiermann or Andrew Loudon.

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.

Related links

Manchester Biological Timing: https://www.bmh.manchester.ac.uk/research/biological-timing/

Loudon Laboratory: http://www.manchester.ac.uk/research/Andrew.loudon/personaldetails

Scheiermann Laboratory: http://scheiermannlab.de/

Gibbs Laboratory: https://www.research.manchester.ac.uk/portal/Julie.Gibbs.html

Supplementary information

41577_2018_8_MOESM1_ESM.mp4

Movie 1: Rhythmic leukocyte activity throughout the body. Migration of leukocytes from blood to tissues (and back) is regulated at multiple levels by the circadian clock. In mice, blood leukocyte content is high during the day (rest phase) and lower at night (active phase). The factors, which generate these oscillations vary between cells and tissues. For example, neutrophils in the blood express higher levels of the chemokine receptor CXCR4 during the late day. This receptor drives neutrophil homing to bone marrow, which is therefore elevated at this time point. During the night, CXCR4 expression is reduced and less homing occurs to this organ. In the lung, resident stromal cells rhythmically produce the neutrophil chemoattractant CXCL5. Inflammatory challenge by lipopolysaccharide (LPS) inhalation during the day stimulates greater production of CXCL5 than challenge at night. The differential production of chemoattractant, along with greater numbers of neutrophils in blood, therefore leads to increased neutrophil influx to the lung during the day. By contrast, cells largely home to lymph nodes at night. During the day, T cell and B cell expression of the lymph node-homing receptor CCR7 is low, and few cells migrate into the lymph node. In addition, expression of S1PR1, the receptor which mediates lymphocyte egress, is high and cells are more prone to leave the lymph node during the day. At night, the inverse occurs and T cells and B cells are retained in the lymph node for longer. Differentiation of cells is also regulated by time-of-day, as in the case of TH17 cell development in the gut. During the day, levels of the differentiation factor RORγt are high and increased differentiation is observed relative to the dark phase. At night, RORγt activity is repressed by NFIL3 and so the differentiation stimulus is reduced. In this way, the body is primed to respond differently to inflammatory challenge at different times of day, and disruption to the circadian rhythm can have severe consequences for immune function

Glossary

Circadian

A free-running rhythm with a period of approximately 24 h that persists in the absence of external entrainment, such as in constant darkness.

Suprachiasmatic nuclei

(SCN). A bilateral structure in the anterior hypothalamus, home to the central pacemaker, which processes light input and conveys timing information to the rest of the body.

Diurnal

A pattern that occurs over the course of a day in which external entrainment (such as light–dark cycles) is used; the onset of the light cycle is defined as Zeitgeber time 0 (ZT0).

Period circadian protein homologue 1

(PER1). PER1, PER2 and PER3 are PAS (PER–ARNT–SIM) domain-containing proteins that associate with CRY proteins to inhibit BMAL1–CLOCK-mediated gene expression.

REV-ERB

REV-ERBα (encoded by NR1D1) and REV-ERBβ (encoded by NR1D2) are transcriptional repressors that bind to ROR response element (RORE) motifs in the BMAL1 promoter to regulate the rhythmic expression of BMAL1.

Cryptochromes

(CRYs). CRY1 and CRY2 are transcriptional repressors that associate with PER proteins to inhibit BMAL1–CLOCK-mediated gene transcription.

Brain and muscle ARNT-like 1

(BMAL1). A basic helix–loop–helix PER–ARNT–SIM (bHLH–PAS) domain transcription factor that dimerizes with CLOCK to bind E-boxes in gene promoters to induce circadian gene expression.

CLOCK

(Circadian locomoter output cycles kaput). A basic helix–loop–helix PER–ARNT–SIM (bHLH–PAS) domain transcription factor that can dimerize with BMAL1 to regulate circadian gene expression.

Zeitgeber time

Zeitgeber, literally ‘time giver’, is a cue (such as light) that entrains the circadian clock. Zeitgeber time (ZT) is the time after light onset; for example, lights on is ZT0 and lights off is ZT12 in a 12 h light-12 h dark cycle.

Circadian time

(CT). A measure of subjective time used when organisms are isolated from Zeitgebers (for example, constant darkness). CT0 represents the start of subjective day and CT12 represents the start of subjective night.

RORα

The nuclear receptors RORα, RORβ and RORγ are transcriptional activators that bind to ROR response element (RORE) sites in target gene promoters.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Scheiermann, C., Gibbs, J., Ince, L. et al. Clocking in to immunity. Nat Rev Immunol 18, 423–437 (2018). https://doi.org/10.1038/s41577-018-0008-4

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41577-018-0008-4

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