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:

Metabolic rivalry: circadian homeostasis and tumorigenesis

Abstract

Circadian rhythms govern a large array of physiological and metabolic functions. Perturbations of the daily cycle have been linked to elevated risk of developing cancer as well as poor prognosis in patients with cancer. Also, expression of core clock genes or proteins is remarkably attenuated particularly in tumours of a higher stage or that are more aggressive, possibly linking the circadian clock to cellular differentiation. Emerging evidence indicates that metabolic control by the circadian clock underpins specific hallmarks of cancer metabolism. Indeed, to support cell proliferation and biomass production, the clock may direct metabolic processes of cancer cells in concert with non-clock transcription factors to control how nutrients and metabolites are utilized in a time-specific manner. We hypothesize that the metabolic switch between differentiation or stemness of cancer may be coupled to the molecular clockwork. Moreover, circadian rhythms of host organisms appear to dictate tumour growth and proliferation. This Review outlines recent discoveries of the interplay between circadian rhythms, proliferative metabolism and cancer, highlighting potential opportunities in the development of future therapeutic strategies.

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: Organization of the circadian clock network.
Fig. 2: Molecular architecture of the circadian clock.
Fig. 3: Tumour-autonomous circadian clock.
Fig. 4: Regulation of proliferative metabolism by clock and non-clock TFs.
Fig. 5: Temporal communication between the circadian rhythm of host organisms and cancer-autonomous clocks.

Similar content being viewed by others

References

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

    CAS  PubMed  Google Scholar 

  2. Masri, S., Kinouchi, K. & Sassone-Corsi, P. Circadian clocks, epigenetics, and cancer. Curr. Opin. Oncol. 27, 50–56 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Masri, S. & Sassone-Corsi, P. The emerging link between cancer, metabolism, and circadian rhythms. Nat. Med. 24, 1795–1803 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  6. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Greco, C. M. & Sassone-Corsi, P. Circadian blueprint of metabolic pathways in the brain. Nat. Rev. Neurosci. 20, 71–82 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Gaucher, J., Montellier, E. & Sassone-Corsi, P. Molecular cogs: interplay between circadian clock and cell cycle. Trends Cell Biol. 28, 368–379 (2018).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  10. Yamazaki, S. et al. Resetting central and peripheral circadian oscillators in transgenic rats. Science 288, 682–685 (2000).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Reppert, S. M. & Weaver, D. R. Coordination of circadian timing in mammals. Nature 418, 935–941 (2002).

    CAS  PubMed  Google Scholar 

  13. Schibler, U. & Sassone-Corsi, P. A web of circadian pacemakers. Cell 111, 919–922 (2002). This review captures the original view of how circadian clocks are organized in a complex network that may be essential for their fundamental properties of timekeeping and entrainment to environmental signals.

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  15. Damiola, F. et al. Restricted feeding uncouples circadian oscillators in peripheral tissues from the central pacemaker in the suprachiasmatic nucleus. Genes Dev. 14, 2950–2961 (2000). This study illustrates that peripheral clocks are entrained by timed food intake and highlights the role of food as a zeitgeber.

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Stokkan, K. A., Yamazaki, S., Tei, H., Sakaki, Y. & Menaker, M. Entrainment of the circadian clock in the liver by feeding. Science 291, 490–493 (2001).

    CAS  PubMed  Google Scholar 

  17. Eckel-Mahan, K. L. et al. Reprogramming of the circadian clock by nutritional challenge. Cell 155, 1464–1478 (2013). This study uncovers that the circadian machinery can be reprogrammed transcriptionally and metabolically by food intake.

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Gaucher, J. et al. Distinct metabolic adaptation of liver circadian pathways to acute and chronic patterns of alcohol intake. Proc. Natl Acad. Sci. USA 116, 25250–25259 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  20. Guan, D. et al. Diet-induced circadian enhancer remodeling synchronizes opposing hepatic lipid metabolic processes. Cell 174, 831–842.e12 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  22. Dyar, K. A. et al. Atlas of circadian metabolism reveals system-wide coordination and communication between clocks. Cell 174, 1571–1585.e11 (2018). This study uses high-throughput metabolomics to build the first atlas of circadian metabolism and reveals the importance of temporal metabolic coordination across different tissues.

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Koronowski, K. B. et al. Defining the independence of the liver circadian clock. Cell 177, 1448–1462.e14 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Welz, P. S. et al. BMAL1-driven tissue clocks respond independently to light to maintain homeostasis. Cell 177, 1436–1447.e12 (2019). Together with Koronowski et al. (2019), this paper establishes the degree of autonomy of peripheral circadian clocks in otherwise clock-deficient mice.

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  26. Ukai-Tadenuma, M., Kasukawa, T. & Ueda, H. R. Proof-by-synthesis of the transcriptional logic of mammalian circadian clocks. Nat. Cell Biol. 10, 1154–1163 (2008).

    CAS  PubMed  Google Scholar 

  27. Koike, N. et al. Transcriptional architecture and chromatin landscape of the core circadian clock in mammals. Science 338, 349–354 (2012). This study identifies the parallel between circadian transcriptional regulation and temporal patterns of genome-wide occupancy of core circadian clock regulators.

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Ripperger, J. A. & Schibler, U. Rhythmic CLOCK–BMAL1 binding to multiple E-box motifs drives circadian Dbp transcription and chromatin transitions. Nat. Genet. 38, 369–374 (2006).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Fang, B. et al. Circadian enhancers coordinate multiple phases of rhythmic gene transcription in vivo. Cell 159, 1140–1152 (2014). This study investigates circadian enhancer RNAs and demonstrates the commitment of diverse TFs to temporal gene regulation.

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Kume, K. et al. mCRY1 and mCRY2 are essential components of the negative limb of the circadian clock feedback loop. Cell 98, 193–205 (1999).

    CAS  PubMed  Google Scholar 

  34. Ukai-Tadenuma, M. et al. Delay in feedback repression by cryptochrome 1 is required for circadian clock function. Cell 144, 268–281 (2011).

    CAS  PubMed  Google Scholar 

  35. van der Horst, G. T. et al. Mammalian Cry1 and Cry2 are essential for maintenance of circadian rhythms. Nature 398, 627–630 (1999).

    PubMed  Google Scholar 

  36. Shigeyoshi, Y. et al. Light-induced resetting of a mammalian circadian clock is associated with rapid induction of the mPer1 transcript. Cell 91, 1043–1053 (1997).

    CAS  PubMed  Google Scholar 

  37. Crosio, C., Cermakian, N., Allis, C. D. & Sassone-Corsi, P. Light induces chromatin modification in cells of the mammalian circadian clock. Nat. Neurosci. 3, 1241–1247 (2000).

    CAS  PubMed  Google Scholar 

  38. Ginty, D. D. et al. Regulation of CREB phosphorylation in the suprachiasmatic nucleus by light and a circadian clock. Science 260, 238–241 (1993).

    CAS  PubMed  Google Scholar 

  39. Sato, M., Murakami, M., Node, K., Matsumura, R. & Akashi, M. The role of the endocrine system in feeding-induced tissue-specific circadian entrainment. Cell Rep. 8, 393–401 (2014).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Adamovich, Y., Ladeuix, B., Golik, M., Koeners, M. P. & Asher, G. Rhythmic oxygen levels reset circadian clocks through HIF1α. Cell Metab. 25, 93–101 (2017).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Husse, J., Leliavski, A., Tsang, A. H., Oster, H. & Eichele, G. The light–dark cycle controls peripheral rhythmicity in mice with a genetically ablated suprachiasmatic nucleus clock. FASEB J. 28, 4950–4960 (2014).

    CAS  PubMed  Google Scholar 

  45. Ishida, A. et al. Light activates the adrenal gland: timing of gene expression and glucocorticoid release. Cell Metab. 2, 297–307 (2005). This paper illustrates a hypothalamus–pituitary–adrenal axis-independent, photic induction of glucocorticoid release from the adrenal gland.

    CAS  PubMed  Google Scholar 

  46. Kolbe, I., Leinweber, B., Brandenburger, M. & Oster, H. Circadian clock network desynchrony promotes weight gain and alters glucose homeostasis in mice. Mol. Metab. 30, 140–151 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Terazono, H. et al. Adrenergic regulation of clock gene expression in mouse liver. Proc. Natl Acad. Sci. USA 100, 6795–6800 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Balsalobre, A., Marcacci, L. & Schibler, U. Multiple signaling pathways elicit circadian gene expression in cultured Rat-1 fibroblasts. Curr. Biol. 10, 1291–1294 (2000).

    CAS  PubMed  Google Scholar 

  49. Robles, M. S., Humphrey, S. J. & Mann, M. Phosphorylation is a central mechanism for circadian control of metabolism and physiology. Cell Metab. 25, 118–127 (2017).

    CAS  PubMed  Google Scholar 

  50. Lamia, K. A. et al. AMPK regulates the circadian clock by cryptochrome phosphorylation and degradation. Science 326, 437–440 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Lipton, J. O. et al. The circadian protein BMAL1 regulates translation in response to S6K1-mediated phosphorylation. Cell 161, 1138–1151 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Sahar, S., Zocchi, L., Kinoshita, C., Borrelli, E. & Sassone-Corsi, P. Regulation of BMAL1 protein stability and circadian function by GSK3β-mediated phosphorylation. PLoS ONE 5, e8561 (2010).

    PubMed  PubMed Central  Google Scholar 

  53. Menzies, K. J., Zhang, H., Katsyuba, E. & Auwerx, J. Protein acetylation in metabolism—metabolites and cofactors. Nat. Rev. Endocrinol. 12, 43–60 (2016).

    CAS  PubMed  Google Scholar 

  54. Pietrocola, F., Galluzzi, L., Bravo-San Pedro, J. M., Madeo, F. & Kroemer, G. Acetyl coenzyme A: a central metabolite and second messenger. Cell Metab. 21, 805–821 (2015).

    CAS  PubMed  Google Scholar 

  55. Asher, G. et al. SIRT1 regulates circadian clock gene expression through PER2 deacetylation. Cell 134, 317–328 (2008).

    CAS  PubMed  Google Scholar 

  56. Hirayama, J. et al. CLOCK-mediated acetylation of BMAL1 controls circadian function. Nature 450, 1086–1090 (2007).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  58. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Ramsey, K. M. et al. Circadian clock feedback cycle through NAMPT-mediated NAD+ biosynthesis. Science 324, 651–654 (2009). Together with Nakahata et al. (2009), this paper uncovers the link between the core circadian clock and the cyclic production of NAD+ through a metabolic feedback loop.

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Fustin, J. M. et al. RNA-methylation-dependent RNA processing controls the speed of the circadian clock. Cell 155, 793–806 (2013).

    CAS  PubMed  Google Scholar 

  61. Hirano, A., Braas, D., Fu, Y. H. & Ptacek, L. J. FAD regulates CRYPTOCHROME protein stability and circadian clock in mice. Cell Rep. 19, 255–266 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  62. Kaasik, K. et al. Glucose sensor O-GlcNAcylation coordinates with phosphorylation to regulate circadian clock. Cell Metab. 17, 291–302 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  63. Li, M. D. et al. O-GlcNAc signaling entrains the circadian clock by inhibiting BMAL1/CLOCK ubiquitination. Cell Metab. 17, 303–310 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  64. Cao, Q. et al. A role for the clock gene per1 in prostate cancer. Cancer Res. 69, 7619–7625 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  65. de Assis, L. V. M. et al. Non-metastatic cutaneous melanoma induces chronodisruption in central and peripheral circadian clocks. Int. J. Mol. Sci. 19, 1065 (2018).

    PubMed Central  Google Scholar 

  66. Oh, E. Y. et al. Circadian transcription profile of mouse breast cancer under light–dark and dark–dark conditions. Cancer Genomics Proteomics 7, 311–322 (2010).

    CAS  PubMed  Google Scholar 

  67. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Shilts, J., Chen, G. & Hughey, J. J. Evidence for widespread dysregulation of circadian clock progression in human cancer. PeerJ 6, e4327 (2018).

    PubMed  PubMed Central  Google Scholar 

  69. Ye, Y. et al. The genomic landscape and pharmacogenomic interactions of clock genes in cancer chronotherapy. Cell Syst. 6, 314–328.e2 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Abreu, M., Basti, A., Genov, N., Mazzoccoli, G. & Relogio, A. The reciprocal interplay between TNFα and the circadian clock impacts on cell proliferation and migration in Hodgkin lymphoma cells. Sci. Rep. 8, 11474 (2018).

    PubMed  PubMed Central  Google Scholar 

  71. Rida, P., Syed, M. I. & Aneja, R. Time will tell: circadian clock dysregulation in triple negative breast cancer. Front. Biosci. 11, 178–192 (2019).

    Google Scholar 

  72. de The, H. Differentiation therapy revisited. Nat. Rev. Cancer 18, 117–127 (2018).

    PubMed  Google Scholar 

  73. Dierickx, P., Van Laake, L. W. & Geijsen, N. Circadian clocks: from stem cells to tissue homeostasis and regeneration. EMBO Rep. 19, 18–28 (2018).

    CAS  PubMed  Google Scholar 

  74. Sladek, M., Jindrakova, Z., Bendova, Z. & Sumova, A. Postnatal ontogenesis of the circadian clock within the rat liver. Am. J. Physiol. Regul. Integr. Comp. Physiol. 292, R1224–R1229 (2007).

    CAS  PubMed  Google Scholar 

  75. Umemura, Y. et al. Transcriptional program of Kpna2/Importin-α2 regulates cellular differentiation-coupled circadian clock development in mammalian cells. Proc. Natl Acad. Sci. USA 111, E5039–E5048 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  76. Umemura, Y. et al. Involvement of posttranscriptional regulation of Clock in the emergence of circadian clock oscillation during mouse development. Proc. Natl Acad. Sci. USA 114, E7479–E7488 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Yagita, K. et al. Development of the circadian oscillator during differentiation of mouse embryonic stem cells in vitro. Proc. Natl Acad. Sci. USA 107, 3846–3851 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. de Assis, L. V. M. et al. Expression of the circadian clock gene BMAL1 positively correlates with antitumor immunity and patient survival in metastatic melanoma. Front. Oncol. 8, 185 (2018).

    PubMed  PubMed Central  Google Scholar 

  79. Li, W. et al. Decreased circadian component Bmal1 predicts tumor progression and poor prognosis in human pancreatic ductal adenocarcinoma. Biochem. Biophys. Res. Commun. 472, 156–162 (2016).

    CAS  PubMed  Google Scholar 

  80. Mazzoccoli, G. et al. Clock gene expression levels and relationship with clinical and pathological features in colorectal cancer patients. Chronobiol. Int. 28, 841–851 (2011).

    CAS  PubMed  Google Scholar 

  81. Zeng, Z. L. et al. Overexpression of the circadian clock gene Bmal1 increases sensitivity to oxaliplatin in colorectal cancer. Clin. Cancer Res. 20, 1042–1052 (2014).

    CAS  PubMed  Google Scholar 

  82. Fekry, B. et al. Incompatibility of the circadian protein BMAL1 and HNF4α in hepatocellular carcinoma. Nat. Commun. 9, 4349 (2018).

    PubMed  PubMed Central  Google Scholar 

  83. Taniguchi, H. et al. Epigenetic inactivation of the circadian clock gene BMAL1 in hematologic malignancies. Cancer Res. 69, 8447–8454 (2009).

    CAS  PubMed  Google Scholar 

  84. Yeh, C. M. et al. Epigenetic silencing of ARNTL, a circadian gene and potential tumor suppressor in ovarian cancer. Int. J. Oncol. 45, 2101–2107 (2014).

    CAS  PubMed  Google Scholar 

  85. Zhang, S. et al. Circadian clock components RORα and Bmal1 mediate the anti-proliferative effect of MLN4924 in osteosarcoma cells. Oncotarget 7, 66087–66099 (2016).

    PubMed  PubMed Central  Google Scholar 

  86. Zeng, Z. L. et al. Effects of the biological clock gene Bmal1 on tumour growth and anti-cancer drug activity. J. Biochem. 148, 319–326 (2010).

    CAS  PubMed  Google Scholar 

  87. Ohashi, M. et al. Disruption of circadian clockwork in in vivo reprogramming-induced mouse kidney tumors. Genes Cell 23, 60–69 (2018).

    CAS  Google Scholar 

  88. Bu, Y. et al. A PERK–miR-211 axis suppresses circadian regulators and protein synthesis to promote cancer cell survival. Nat. Cell Biol. 20, 104–115 (2018).

    CAS  PubMed  Google Scholar 

  89. Yamada, Y., Haga, H. & Yamada, Y. Concise review: dedifferentiation meets cancer development: proof of concept for epigenetic cancer. Stem Cell Transl Med. 3, 1182–1187 (2014).

    CAS  Google Scholar 

  90. Dai, H. et al. The role of polymorphisms in circadian pathway genes in breast tumorigenesis. Breast Cancer Res. Treat. 127, 531–540 (2011).

    CAS  PubMed  Google Scholar 

  91. Alhopuro, P. et al. Mutations in the circadian gene CLOCK in colorectal cancer. Mol. Cancer Res. 8, 952–960 (2010).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  93. 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).

    CAS  PubMed  Google Scholar 

  94. Oshima, T. et al. Expression of circadian genes correlates with liver metastasis and outcomes in colorectal cancer. Oncol. Rep. 25, 1439–1446 (2011).

    CAS  PubMed  Google Scholar 

  95. Xia, H. C. et al. Deregulated expression of the Per1 and Per2 in human gliomas. Can. J. Neurol. Sci. 37, 365–370 (2010).

    PubMed  Google Scholar 

  96. Zhao, H. et al. Prognostic relevance of Period1 (Per1) and Period2 (Per2) expression in human gastric cancer. Int. J. Clin. Exp. Pathol. 7, 619–630 (2014).

    PubMed  PubMed Central  Google Scholar 

  97. Broadberry, E. et al. Disrupted circadian clocks and altered tissue mechanics in primary human breast tumours. Breast Cancer Res. 20, 125 (2018).

    PubMed  PubMed Central  Google Scholar 

  98. Gery, S. et al. Transcription profiling of C/EBP targets identifies Per2 as a gene implicated in myeloid leukemia. Blood 106, 2827–2836 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  99. Xiang, R. et al. Circadian clock gene Per2 downregulation in nonsmall cell lung cancer is associated with tumour progression and metastasis. Oncol. Rep. 40, 3040–3048 (2018).

    CAS  PubMed  Google Scholar 

  100. Gery, S., Virk, R. K., Chumakov, K., Yu, A. & Koeffler, H. P. The clock gene Per2 links the circadian system to the estrogen receptor. Oncogene 26, 7916–7920 (2007).

    CAS  PubMed  Google Scholar 

  101. Hua, H. et al. Circadian gene mPer2 overexpression induces cancer cell apoptosis. Cancer Sci. 97, 589–596 (2006).

    CAS  PubMed  Google Scholar 

  102. Miyazaki, K., Wakabayashi, M., Hara, Y. & Ishida, N. Tumor growth suppression in vivo by overexpression of the circadian component, PER2. Genes Cells 15, 351–358 (2010).

    CAS  PubMed  Google Scholar 

  103. Oda, A. et al. Clock gene mouse period2 overexpression inhibits growth of human pancreatic cancer cells and has synergistic effect with cisplatin. Anticancer Res. 29, 1201–1209 (2009).

    CAS  PubMed  Google Scholar 

  104. Cadenas, C. et al. Loss of circadian clock gene expression is associated with tumor progression in breast cancer. Cell Cycle 13, 3282–3291 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  105. 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 study demonstrates that loss of the Per2 gene leads to mice prone to cancer.

    CAS  PubMed  Google Scholar 

  106. Gu, X. et al. The circadian mutation PER2(S662G) is linked to cell cycle progression and tumorigenesis. Cell Death Differ. 19, 397–405 (2012).

    CAS  PubMed  Google Scholar 

  107. Wood, P. A. et al. Period 2 mutation accelerates ApcMin/+ tumorigenesis. Mol. Cancer Res. 6, 1786–1793 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  108. Hua, H. et al. Inhibition of tumorigenesis by intratumoral delivery of the circadian gene mPer2 in C57BL/6 mice. Cancer Gene Ther. 14, 815–818 (2007).

    CAS  PubMed  Google Scholar 

  109. Qin, T. et al. Effect of Period 2 on the proliferation, apoptosis and migration of osteosarcoma cells, and the corresponding mechanisms. Oncol. Lett. 16, 2668–2674 (2018).

    PubMed  PubMed Central  Google Scholar 

  110. Yang, X. et al. The circadian clock gene Per1 suppresses cancer cell proliferation and tumor growth at specific times of day. Chronobiol. Int. 26, 1323–1339 (2009).

    CAS  PubMed  Google Scholar 

  111. Yang, X. et al. Down regulation of circadian clock gene Period 2 accelerates breast cancer growth by altering its daily growth rhythm. Breast Cancer Res. Treat. 117, 423–431 (2009).

    PubMed  Google Scholar 

  112. Hwang-Verslues, W. W. et al. Loss of corepressor PER2 under hypoxia up-regulates OCT1-mediated EMT gene expression and enhances tumor malignancy. Proc. Natl Acad. Sci. USA 110, 12331–12336 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  113. Grimaldi, B. et al. PER2 controls lipid metabolism by direct regulation of PPARγ. Cell Metab. 12, 509–520 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  114. Gotoh, T. et al. Model-driven experimental approach reveals the complex regulatory distribution of p53 by the circadian factor Period 2. Proc. Natl Acad. Sci. USA 113, 13516–13521 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  115. Travnickova-Bendova, Z., Cermakian, N., Reppert, S. M. & Sassone-Corsi, P. Bimodal regulation of mPeriod promoters by CREB-dependent signaling and CLOCK/BMAL1 activity. Proc. Natl Acad. Sci. USA 99, 7728–7733 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  116. Yang, W. S. & Stockwell, B. R. Inhibition of casein kinase 1-ε induces cancer-cell-selective, PERIOD2-dependent growth arrest. Genome Biol. 9, R92 (2008).

    PubMed  PubMed Central  Google Scholar 

  117. Liu, J. et al. Distinct control of PERIOD2 degradation and circadian rhythms by the oncoprotein and ubiquitin ligase MDM2. Sci. Signal. 11, eaau0715 (2018).

    PubMed  Google Scholar 

  118. Lahav, G. et al. Dynamics of the p53–Mdm2 feedback loop in individual cells. Nat. Genet. 36, 147–150 (2004).

    CAS  PubMed  Google Scholar 

  119. Dyar, K. A. et al. Muscle insulin sensitivity and glucose metabolism are controlled by the intrinsic muscle clock. Mol. Metab. 3, 29–41 (2014).

    CAS  PubMed  Google Scholar 

  120. Yu, H. et al. Cryptochrome 1 overexpression correlates with tumor progression and poor prognosis in patients with colorectal cancer. PLoS ONE 8, e61679 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  121. Mannic, T. et al. Circadian clock characteristics are altered in human thyroid malignant nodules. J. Clin. Endocrinol. Metab. 98, 4446–4456 (2013).

    CAS  PubMed  Google Scholar 

  122. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  124. Chun, S. K. et al. A synthetic cryptochrome inhibitor induces anti-proliferative effects and increases chemosensitivity in human breast cancer cells. Biochem. Biophys. Res. Commun. 467, 441–446 (2015).

    CAS  PubMed  Google Scholar 

  125. Kriebs, A. et al. Circadian repressors CRY1 and CRY2 broadly interact with nuclear receptors and modulate transcriptional activity. Proc. Natl Acad. Sci. USA 114, 8776–8781 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  126. Busino, L. et al. SCFFbxl3 controls the oscillation of the circadian clock by directing the degradation of cryptochrome proteins. Science 316, 900–904 (2007).

    CAS  PubMed  Google Scholar 

  127. Godinho, S. I. et al. The after-hours mutant reveals a role for Fbxl3 in determining mammalian circadian period. Science 316, 897–900 (2007).

    CAS  PubMed  Google Scholar 

  128. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  129. Huber, A. L. et al. CRY2 and FBXL3 cooperatively degrade c-MYC. Mol. Cell 64, 774–789 (2016). This study identifies a possible mechanism through which the circadian clock might inhibit MYC.

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  131. Lee, S., Donehower, L. A., Herron, A. J., Moore, D. D. & Fu, L. Disrupting circadian homeostasis of sympathetic signaling promotes tumor development in mice. PLoS ONE 5, e10995 (2010).

    PubMed  PubMed Central  Google Scholar 

  132. Chen, S. T. et al. Deregulated expression of the PER1, PER2 and PER3 genes in breast cancers. Carcinogenesis 26, 1241–1246 (2005).

    CAS  PubMed  Google Scholar 

  133. Zhu, Y. et al. Epigenetic impact of long-term shiftwork: pilot evidence from circadian genes and whole-genome methylation analysis. Chronobiol. Int. 28, 852–861 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  134. Rao, R. C. & Dou, Y. Hijacked in cancer: the KMT2 (MLL) family of methyltransferases. Nat. Rev. Cancer 15, 334–346 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  135. Katada, S. & Sassone-Corsi, P. The histone methyltransferase MLL1 permits the oscillation of circadian gene expression. Nat. Struct. Mol. Biol. 17, 1414–1421 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  136. Valekunja, U. K. et al. Histone methyltransferase MLL3 contributes to genome-scale circadian transcription. Proc. Natl Acad. Sci. USA 110, 1554–1559 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  137. Miki, T. et al. PML regulates PER2 nuclear localization and circadian function. EMBO J. 31, 1427–1439 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  138. Vander Heiden, M. G. & DeBerardinis, R. J. Understanding the Intersections between metabolism and cancer biology. Cell 168, 657–669 (2017). This review article discusses limiting metabolic pathways in cancer cell proliferation.

    PubMed Central  Google Scholar 

  139. Tohyama, S. et al. Glutamine oxidation is indispensable for survival of human pluripotent stem cells. Cell Metab. 23, 663–674 (2016).

    CAS  PubMed  Google Scholar 

  140. Tohyama, S., Tanosaki, S., Someya, S., Fujita, J. & Fukuda, K. Manipulation of pluripotent stem cell metabolism for clinical application. Curr. Stem Cell Rep. 3, 28–34 (2017).

    PubMed  PubMed Central  Google Scholar 

  141. Peek, C. B. et al. Circadian clock NAD+ cycle drives mitochondrial oxidative metabolism in mice. Science 342, 1243417 (2013). This study demonstrates that loss of Bmal1 induces a metabolic switch from lipid oxidation to glycolysis.

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  144. Walton, Z. E. et al. Acid suspends the circadian clock in hypoxia through inhibition of mTOR. Cell 174, 72–87.e32 (2018). This study shows that the acid–base balance influences the circadian clock function.

    CAS  PubMed  PubMed Central  Google Scholar 

  145. Vander Heiden, M. G., Cantley, L. C. & Thompson, C. B. Understanding the Warburg effect: the metabolic requirements of cell proliferation. Science 324, 1029–1033 (2009).

    Google Scholar 

  146. Imai, S. Armstrong, C. M., Kaeberlein, M. & Guarente, L. Transcriptional silencing and longevity protein Sir2 is an NAD-dependent histone deacetylase. Nature 403 795–800 (2000).

    CAS  PubMed  Google Scholar 

  147. Elhanati, S. et al. Multiple regulatory layers of SREBP1/2 by SIRT6. Cell Rep. 4, 905–912 (2013).

    CAS  PubMed  Google Scholar 

  148. Masri, S. et al. Partitioning circadian transcription by SIRT6 leads to segregated control of cellular metabolism. Cell 158, 659–672 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  149. Ponugoti, B. et al. SIRT1 deacetylates and inhibits SREBP-1C activity in regulation of hepatic lipid metabolism. J. Biol. Chem. 285, 33959–33970 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  150. Currie, E., Schulze, A., Zechner, R., Walther, T. C. & Farese, R. V. Jr. Cellular fatty acid metabolism and cancer. Cell Metab. 18, 153–161 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  151. Svensson, R. U. et al. Inhibition of acetyl-CoA carboxylase suppresses fatty acid synthesis and tumor growth of non-small-cell lung cancer in preclinical models. Nat. Med. 22, 1108–1119 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  152. Bugge, A. et al. Rev-erbα and Rev-erbβ coordinately protect the circadian clock and normal metabolic function. Genes Dev. 26, 657–667 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  154. Dierickx, P. et al. SR9009 has REV-ERB-independent effects on cell proliferation and metabolism. Proc. Natl Acad. Sci. USA 116, 12147–12152 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  155. Dang, C. V. MYC, metabolism, cell growth, and tumorigenesis. Cold Spring Harb. Perspect. Med. 3, a014217 (2013).

    PubMed  PubMed Central  Google Scholar 

  156. Gu, W. et al. Glycolytic metabolism plays a functional role in regulating human pluripotent stem cell state. Cell Stem Cell 19, 476–490 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  157. Wang, R. et al. The transcription factor Myc controls metabolic reprogramming upon T lymphocyte activation. Immunity 35, 871–882 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  158. Altman, B. J. et al. MYC disrupts the circadian clock and metabolism in cancer cells. Cell Metab. 22, 1009–1019 (2015). This study provides a molecular underpinning by which MYC inhibits oscillation of the circadian clock.

    CAS  PubMed  PubMed Central  Google Scholar 

  159. Repouskou, A. & Prombona, A. c-MYC targets the central oscillator gene Per1 and is regulated by the circadian clock at the post-transcriptional level. Biochim. Biophys. Acta 1859, 541–552 (2016).

    CAS  PubMed  Google Scholar 

  160. Shostak, A. et al. MYC/MIZ1-dependent gene repression inversely coordinates the circadian clock with cell cycle and proliferation. Nat. Commun. 7, 11807 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  161. Hsieh, A. L. et al. Misregulation of Drosophila Myc disrupts circadian behavior and metabolism. Cell Rep. 29, 1778–1788.e4 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  162. Papagiannakopoulos, T. et al. Circadian rhythm disruption promotes lung tumorigenesis. Cell Metab. 24, 324–331 (2016). This study reveals that Per2 loss of function promotes metabolic reprogramming in mouse lung cancer cells.

    CAS  PubMed  PubMed Central  Google Scholar 

  163. Keith, B., Johnson, R. S. & Simon, M. C. HIF1α and HIF2α: sibling rivalry in hypoxic tumour growth and progression. Nat. Rev. Cancer 12, 9–22 (2011).

    PubMed  PubMed Central  Google Scholar 

  164. Hu, C. J., Wang, L. Y., Chodosh, L. A., Keith, B. & Simon, M. C. Differential roles of hypoxia-inducible factor 1α (HIF-1α) and HIF-2α in hypoxic gene regulation. Mol. Cell Biol. 23, 9361–9374 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  165. Iyer, N. V. et al. Cellular and developmental control of O2 homeostasis by hypoxia-inducible factor 1α. Genes Dev. 12, 149–162 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  166. Rankin, E. B. & Giaccia, A. J. The role of hypoxia-inducible factors in tumorigenesis. Cell Death Differ. 15, 678–685 (2008).

    CAS  PubMed  Google Scholar 

  167. Ryan, H. E., Lo, J. & Johnson, R. S. HIF-1α is required for solid tumor formation and embryonic vascularization. EMBO J. 17, 3005–3015 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  168. Peek, C. B. et al. Circadian clock interaction with HIF1α mediates oxygenic metabolism and anaerobic glycolysis in skeletal muscle. Cell Metab. 25, 86–92 (2017).

    CAS  PubMed  Google Scholar 

  169. 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).

    CAS  PubMed  Google Scholar 

  170. Kobayashi, M. et al. A circadian clock gene, PER2, activates HIF-1 as an effector molecule for recruitment of HIF-1α to promoter regions of its downstream genes. FEBS J. 284, 3804–3816 (2017).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  172. Manella, G. et al. Hypoxia induces a time- and tissue-specific response that elicits intertissue circadian clock misalignment. Proc. Natl Acad. Sci. USA 117, 779–786 (2020).

    CAS  PubMed  Google Scholar 

  173. Almendros, I. et al. Intermittent hypoxia increases melanoma metastasis to the lung in a mouse model of sleep apnea. Respir. Physiol. Neurobiol. 186, 303–307 (2013).

    CAS  PubMed  Google Scholar 

  174. Karoor, V. et al. Alveolar hypoxia promotes murine lung tumor growth through a VEGFR-2/EGFR-dependent mechanism. Cancer Prev. Res. 5, 1061–1071 (2012).

    CAS  Google Scholar 

  175. Bersten, D. C., Sullivan, A. E., Peet, D. J. & Whitelaw, M. L. bHLH–PAS proteins in cancer. Nat. Rev. Cancer 13, 827–841 (2013).

    CAS  PubMed  Google Scholar 

  176. King, A., Selak, M. A. & Gottlieb, E. Succinate dehydrogenase and fumarate hydratase: linking mitochondrial dysfunction and cancer. Oncogene 25, 4675–4682 (2006).

    CAS  PubMed  Google Scholar 

  177. Tabebi, M., Soderkvist, P. & Jensen, L. D. Hypoxia signaling and circadian disruption in and by pheochromocytoma. Front. Endocrinol. 9, 612 (2018).

    Google Scholar 

  178. Zhikrivetskaya, S. O. et al. Molecular markers of paragangliomas/pheochromocytomas. Oncotarget 8, 25756–25782 (2017).

    PubMed  PubMed Central  Google Scholar 

  179. Opitz, C. A. et al. An endogenous tumour-promoting ligand of the human aryl hydrocarbon receptor. Nature 478, 197–203 (2011).

    CAS  PubMed  Google Scholar 

  180. Horton, J. D., Goldstein, J. L. & Brown, M. S. SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver. J. Clin. Invest. 109, 1125–1131 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  181. Kuiper, R. P. et al. Upregulation of the transcription factor TFEB in t(6;11)(p21;q13)-positive renal cell carcinomas due to promoter substitution. Hum. Mol. Genet. 12, 1661–1669 (2003).

    CAS  PubMed  Google Scholar 

  182. Sardiello, M. et al. A gene network regulating lysosomal biogenesis and function. Science 325, 473–477 (2009).

    CAS  PubMed  Google Scholar 

  183. Sidhar, S. K. et al. The t(X;1)(p11.2;q21.2) translocation in papillary renal cell carcinoma fuses a novel gene PRCC to the TFE3 transcription factor gene. Hum. Mol. Genet. 5, 1333–1338 (1996).

    CAS  PubMed  Google Scholar 

  184. Weterman, M. A., Wilbrink, M. & Geurts van Kessel, A. Fusion of the transcription factor TFE3 gene to a novel gene, PRCC, in t(X;1)(p11;q21)-positive papillary renal cell carcinomas. Proc. Natl Acad. Sci. USA 93, 15294–15298 (1996).

    CAS  PubMed  PubMed Central  Google Scholar 

  185. Pastore, N. et al. Nutrient-sensitive transcription factors TFEB and TFE3 couple autophagy and metabolism to the peripheral clock. EMBO J. 38, e101347 (2019).

    PubMed  PubMed Central  Google Scholar 

  186. Michael, A. K. et al. Cancer/testis antigen PASD1 silences the circadian clock. Mol. Cell 58, 743–754 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  187. Hansen, J. Light at night, shiftwork, and breast cancer risk. J. Natl Cancer Inst. 93, 1513–1515 (2001).

    CAS  PubMed  Google Scholar 

  188. Knutsson, A. et al. Breast cancer among shift workers: results of the WOLF longitudinal cohort study. Scand. J. Work Environ. Health 39, 170–177 (2013).

    PubMed  Google Scholar 

  189. Megdal, S. P., Kroenke, C. H., Laden, F., Pukkala, E. & Schernhammer, E. S. Night work and breast cancer risk: a systematic review and meta-analysis. Eur. J. Cancer 41, 2023–2032 (2005).

    PubMed  Google Scholar 

  190. Rafnsson, V., Tulinius, H., Jonasson, J. G. & Hrafnkelsson, J. Risk of breast cancer in female flight attendants: a population-based study (Iceland). Cancer Causes Control 12, 95–101 (2001).

    CAS  PubMed  Google Scholar 

  191. Schernhammer, E. S. et al. Rotating night shifts and risk of breast cancer in women participating in the Nurses’ Health Study. J. Natl Cancer Inst. 93, 1563–1568 (2001).

    CAS  PubMed  Google Scholar 

  192. Srour, B. et al. Circadian nutritional behaviours and cancer risk: new insights from the NutriNet-Sante prospective cohort study: disclaimers. Int. J. Cancer 143, 2369–2379 (2018).

    CAS  PubMed  Google Scholar 

  193. Viswanathan, A. N. & Schernhammer, E. S. Circulating melatonin and the risk of breast and endometrial cancer in women. Cancer Lett. 281, 1–7 (2009).

    CAS  PubMed  Google Scholar 

  194. Viswanathan, A. N., Hankinson, S. E. & Schernhammer, E. S. Night shift work and the risk of endometrial cancer. Cancer Res. 67, 10618–10622 (2007).

    CAS  PubMed  Google Scholar 

  195. Conlon, M., Lightfoot, N. & Kreiger, N. Rotating shift work and risk of prostate cancer. Epidemiology 18, 182–183 (2007).

    PubMed  Google Scholar 

  196. Kloog, I., Haim, A., Stevens, R. G. & Portnov, B. A. Global co-distribution of light at night (LAN) and cancers of prostate, colon, and lung in men. Chronobiol. Int. 26, 108–125 (2009).

    PubMed  Google Scholar 

  197. Kubo, T. et al. Prospective cohort study of the risk of prostate cancer among rotating-shift workers: findings from the Japan collaborative cohort study. Am. J. Epidemiol. 164, 549–555 (2006).

    PubMed  Google Scholar 

  198. Schernhammer, E. S. et al. Night-shift work and risk of colorectal cancer in the Nurses’ Health Study. J. Natl Cancer Inst. 95, 825–828 (2003).

    PubMed  Google Scholar 

  199. Lahti, T. A., Partonen, T., Kyyronen, P., Kauppinen, T. & Pukkala, E. Night-time work predisposes to non-Hodgkin lymphoma. Int. J. Cancer 123, 2148–2151 (2008).

    CAS  PubMed  Google Scholar 

  200. Basen-Engquist, K. & Chang, M. Obesity and cancer risk: recent review and evidence. Curr. Oncol. Rep. 13, 71–76 (2011).

    PubMed  PubMed Central  Google Scholar 

  201. Hastings, M. H., Reddy, A. B. & Maywood, E. S. A clockwork web: circadian timing in brain and periphery, in health and disease. Nat. Rev. Neurosci. 4, 649–661 (2003).

    CAS  PubMed  Google Scholar 

  202. Filipski, E. et al. Disruption of circadian coordination accelerates malignant growth in mice. Pathol. Biol. 51, 216–219 (2003).

    PubMed  Google Scholar 

  203. Filipski, E. et al. Effects of light and food schedules on liver and tumor molecular clocks in mice. J. Natl Cancer Inst. 97, 507–517 (2005).

    CAS  PubMed  Google Scholar 

  204. van den Heiligenberg, S. et al. The tumor promoting effect of constant light exposure on diethylnitrosamine-induced hepatocarcinogenesis in rats. Life Sci. 64, 2523–2534 (1999).

    PubMed  Google Scholar 

  205. Van Dycke, K. C. et al. Chronically alternating light cycles increase breast cancer risk in mice. Curr. Biol. 25, 1932–1937 (2015).

    PubMed  Google Scholar 

  206. Blask, D. E. et al. Light exposure at night disrupts host/cancer circadian regulatory dynamics: impact on the Warburg effect, lipid signaling and tumor growth prevention. PLoS ONE 9, e102776 (2014).

    PubMed  PubMed Central  Google Scholar 

  207. Filipski, E., Li, X. M. & Levi, F. Disruption of circadian coordination and malignant growth. Cancer Causes Control 17, 509–514 (2006).

    PubMed  Google Scholar 

  208. Dauchy, R. T., Blask, D. E., Sauer, L. A., Brainard, G. C. & Krause, J. A. Dim light during darkness stimulates tumor progression by enhancing tumor fatty acid uptake and metabolism. Cancer Lett. 144, 131–136 (1999).

    CAS  PubMed  Google Scholar 

  209. Mhatre, M. C., Shah, P. N. & Juneja, H. S. Effect of varying photoperiods on mammary morphology, DNA synthesis, and hormone profile in female rats. J. Natl Cancer Inst. 72, 1411–1416 (1984).

    CAS  PubMed  Google Scholar 

  210. Proietti, S., Cucina, A., Minini, M. & Bizzarri, M. Melatonin, mitochondria, and the cancer cell. Cell. Mol. Life Sci. 74, 4015–4025 (2017).

    CAS  PubMed  Google Scholar 

  211. Pevet, P. & Challet, E. Melatonin: both master clock output and internal time-giver in the circadian clocks network. J. Physiol. Paris 105, 170–182 (2011).

    PubMed  Google Scholar 

  212. Talib, W. H. Melatonin and cancer hallmarks. Molecules 23, 518 (2018).

    PubMed Central  Google Scholar 

  213. Li, Y. et al. Melatonin for the prevention and treatment of cancer. Oncotarget 8, 39896–39921 (2017).

    PubMed  PubMed Central  Google Scholar 

  214. Li, J. C. & Xu, F. Influences of light–dark shifting on the immune system, tumor growth and life span of rats, mice and fruit flies as well as on the counteraction of melatonin. Biol. Signals 6, 77–89 (1997).

    PubMed  Google Scholar 

  215. Shah, P. N., Mhatre, M. C. & Kothari, L. S. Effect of melatonin on mammary carcinogenesis in intact and pinealectomized rats in varying photoperiods. Cancer Res. 44, 3403–3407 (1984).

    CAS  PubMed  Google Scholar 

  216. Reddy, A. B. et al. Glucocorticoid signaling synchronizes the liver circadian transcriptome. Hepatology 45, 1478–1488 (2007).

    CAS  PubMed  Google Scholar 

  217. So, A. Y., Bernal, T. U., Pillsbury, M. L., Yamamoto, K. R. & Feldman, B. J. Glucocorticoid regulation of the circadian clock modulates glucose homeostasis. Proc. Natl Acad. Sci. USA 106, 17582–17587 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  218. Kiessling, S., Sollars, P. J. & Pickard, G. E. Light stimulates the mouse adrenal through a retinohypothalamic pathway independent of an effect on the clock in the suprachiasmatic nucleus. PLoS ONE 9, e92959 (2014).

    PubMed  PubMed Central  Google Scholar 

  219. Oster, H. et al. The circadian rhythm of glucocorticoids is regulated by a gating mechanism residing in the adrenal cortical clock. Cell Metab. 4, 163–173 (2006).

    CAS  PubMed  Google Scholar 

  220. Son, G. H. et al. Adrenal peripheral clock controls the autonomous circadian rhythm of glucocorticoid by causing rhythmic steroid production. Proc. Natl Acad. Sci. USA 105, 20970–20975 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  221. Charmandari, E. et al. Peripheral CLOCK regulates target-tissue glucocorticoid receptor transcriptional activity in a circadian fashion in man. PLoS ONE 6, e25612 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  222. Kino, T., Charmandari, E. & Chrousos, G. P. Glucocorticoid receptor: implications for rheumatic diseases. Clin. Exp. Rheumatol. 29, S32–S41 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  223. Moreno-Smith, M., Lutgendorf, S. K. & Sood, A. K. Impact of stress on cancer metastasis. Future Oncol. 6, 1863–1881 (2010).

    PubMed  Google Scholar 

  224. Yang, H. et al. Stress–glucocorticoid–TSC22D3 axis compromises therapy-induced antitumor immunity. Nat. Med. 25, 1428–1441 (2019).

    CAS  PubMed  Google Scholar 

  225. 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).

    CAS  PubMed  Google Scholar 

  226. Chen, D. S. & Mellman, I. Oncology meets immunology: the cancer–immunity cycle. Immunity 39, 1–10 (2013).

    PubMed  Google Scholar 

  227. Suzuki, S. et al. Circadian rhythm of leucocytes and lymphocytes subsets and its possible correlation with the function of the autonomic nervous system. Clin. Exp. Immunol. 110, 500–508 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  228. Maestroni, G. J. et al. Neural and endogenous catecholamines in the bone marrow. Circadian association of norepinephrine with hematopoiesis? Exp. Hematol. 26, 1172–1177 (1998).

    CAS  PubMed  Google Scholar 

  229. Okamoto, S., Ibaraki, K., Hayashi, S. & Saito, M. Ventromedial hypothalamus suppresses splenic lymphocyte activity through sympathetic innervation. Brain Res. 739, 308–313 (1996).

    CAS  PubMed  Google Scholar 

  230. 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).

    CAS  PubMed  Google Scholar 

  231. Creed, S. J. et al. β2-Adrenoceptor signaling regulates invadopodia formation to enhance tumor cell invasion. Breast Cancer Res. 17, 145 (2015).

    PubMed  PubMed Central  Google Scholar 

  232. Magnon, C. et al. Autonomic nerve development contributes to prostate cancer progression. Science 341, 1236361 (2013).

    PubMed  Google Scholar 

  233. Sloan, E. K. et al. The sympathetic nervous system induces a metastatic switch in primary breast cancer. Cancer Res. 70, 7042–7052 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  234. Mormont, M. C. et al. Marker rhythms of circadian system function: a study of patients with metastatic colorectal cancer and good performance status. Chronobiol. Int. 19, 141–155 (2002).

    PubMed  Google Scholar 

  235. Mormont, M. C. & Waterhouse, J. Contribution of the rest–activity circadian rhythm to quality of life in cancer patients. Chronobiol. Int. 19, 313–323 (2002).

    PubMed  Google Scholar 

  236. Innominato, P. F. et al. Circadian rhythm in rest and activity: a biological correlate of quality of life and a predictor of survival in patients with metastatic colorectal cancer. Cancer Res. 69, 4700–4707 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  237. Sephton, S. E., Sapolsky, R. M., Kraemer, H. C. & Spiegel, D. Diurnal cortisol rhythm as a predictor of breast cancer survival. J. Natl Cancer Inst. 92, 994–1000 (2000).

    CAS  PubMed  Google Scholar 

  238. Hojo, H. et al. Remote reprogramming of hepatic circadian transcriptome by breast cancer. Oncotarget 8, 34128–34140 (2017).

    PubMed  PubMed Central  Google Scholar 

  239. Masri, S. et al. Lung adenocarcinoma distally rewires hepatic circadian homeostasis. Cell 165, 896–909 (2016). This study reveals that lung tumours influence host circadian metabolism and illustrates the importance of temporal communications between the host and the tumour.

    CAS  PubMed  PubMed Central  Google Scholar 

  240. Flint, T. R. et al. Tumor-induced IL-6 reprograms host metabolism to suppress anti-tumor immunity. Cell Metab. 24, 672–684 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  241. Ocana, M. C., Martinez-Poveda, B., Quesada, A. R. & Medina, M. A. Metabolism within the tumor microenvironment and its implication on cancer progression: an ongoing therapeutic target. Med. Res. Rev. 39, 70–113 (2019).

    PubMed  Google Scholar 

  242. Faubert, B. et al. Lactate metabolism in human lung tumors. Cell 171, 358–371.e9 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  243. Hui, S. et al. Glucose feeds the TCA cycle via circulating lactate. Nature 551, 115–118 (2017).

    PubMed  PubMed Central  Google Scholar 

  244. Borniger, J. C. et al. A role for hypocretin/orexin in metabolic and sleep abnormalities in a mouse model of non-metastatic breast cancer. Cell Metab. 28, 118–129 e115 (2018). This study identifies neuroendocrine pathways through which breast cancer modulates sleep and glucose homeostasis in mice.

    CAS  PubMed  PubMed Central  Google Scholar 

  245. Honma, K., Honma, S., Kohsaka, M. & Fukuda, N. Seasonal variation in the human circadian rhythm: dissociation between sleep and temperature rhythm. Am. J. Physiol. 262, R885–R891 (1992).

    CAS  PubMed  Google Scholar 

  246. Noya, S. B. et al. The forebrain synaptic transcriptome is organized by clocks but its proteome is driven by sleep. Science 366, eaav2642 (2019).

    CAS  PubMed  Google Scholar 

  247. Hakim, F. et al. Fragmented sleep accelerates tumor growth and progression through recruitment of tumor-associated macrophages and TLR4 signaling. Cancer Res. 74, 1329–1337 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  248. Liang, J. A. et al. Non-apnea sleep disorders will increase subsequent liver cancer risk—a nationwide population-based cohort study. Sleep Med. 13, 869–874 (2012).

    PubMed  Google Scholar 

  249. Li, X. M., Vincenti, M. & Levi, F. Pharmacological effects of vinorelbine on body temperature and locomotor activity circadian rhythms in mice. Chronobiol. Int. 19, 43–55 (2002).

    PubMed  Google Scholar 

  250. Lyall, M. J. et al. Diurnal profile of interstitial glucose following dexamethasone prophylaxis for chemotherapy treatment of gynaecological cancer. Diabet. Med. 35, 1508–1514 (2018).

    CAS  PubMed  Google Scholar 

  251. Palesh, O. et al. Prevalence, putative mechanisms, and current management of sleep problems during chemotherapy for cancer. Nat. Sci. Sleep 4, 151–162 (2012).

    PubMed  PubMed Central  Google Scholar 

  252. Wu, M. W., Li, X. M., Xian, L. J. & Levi, F. Effects of meal timing on tumor progression in mice. Life Sci. 75, 1181–1193 (2004).

    CAS  PubMed  Google Scholar 

  253. Li, X. M. et al. Cancer inhibition through circadian reprogramming of tumor transcriptome with meal timing. Cancer Res. 70, 3351–3360 (2010).

    CAS  PubMed  Google Scholar 

  254. Kinouchi, K. et al. Fasting imparts a switch to alternative daily pathways in liver and muscle. Cell Rep. 25, 3299–3314.e6 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  255. Nencioni, A., Caffa, I., Cortellino, S. & Longo, V. D. Fasting and cancer: molecular mechanisms and clinical application. Nat. Rev. Cancer 18, 707–719 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  257. Yoshino, J., Mills, K. F., Yoon, M. J. & Imai, S. Nicotinamide mononucleotide, a key NAD+ intermediate, treats the pathophysiology of diet- and age-induced diabetes in mice. Cell Metab. 14, 528–536 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  258. Gui, D. Y. et al. Environment dictates dependence on mitochondrial complex I for NAD+ and aspartate production and determines cancer cell sensitivity to metformin. Cell Metab. 24, 716–727 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  259. Yang, G. et al. Timing of expression of the core clock gene Bmal1 influences its effects on aging and survival. Sci. Transl Med. 8, 324ra316 (2016).

    Google Scholar 

  260. Antoch, M. P. et al. Disruption of the circadian clock due to the Clock mutation has discrete effects on aging and carcinogenesis. Cell Cycle 7, 1197–1204 (2008).

    CAS  PubMed  Google Scholar 

  261. Dang, C. V. MYC on the path to cancer. Cell 149, 22–35 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  262. Semenza, G. L. Targeting HIF-1 for cancer therapy. Nat. Rev. Cancer 3, 721–732 (2003).

    CAS  PubMed  Google Scholar 

  263. Xu, C. X., Wang, C., Krager, S. L., Bottum, K. M. & Tischkau, S. A. Aryl hydrocarbon receptor activation attenuates Per1 gene induction and influences circadian clock resetting. Toxicol. Sci. 132, 368–378 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  264. Tischkau, S. A., Jaeger, C. D. & Krager, S. L. Circadian clock disruption in the mouse ovary in response to 2,3,7,8-tetrachlorodibenzo-p-dioxin. Toxicol. Lett. 201, 116–122 (2011).

    CAS  PubMed  Google Scholar 

  265. Murray, I. A., Patterson, A. D. & Perdew, G. H. Aryl hydrocarbon receptor ligands in cancer: friend and foe. Nat. Rev. Cancer 14, 801–814 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  266. Hatori, M. et al. Light-dependent and circadian clock-regulated activation of sterol regulatory element-binding protein, X-box-binding protein 1, and heat shock factor pathways. Proc. Natl Acad. Sci. USA 108, 4864–4869 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  267. Gilardi, F. et al. Genome-wide analysis of SREBP1 activity around the clock reveals its combined dependency on nutrient and circadian signals. PLoS Genet. 10, e1004155 (2014).

    PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  269. Chen, M. et al. An aberrant SREBP-dependent lipogenic program promotes metastatic prostate cancer. Nat. Genet. 50, 206–218 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  270. Kauffman, E. C. et al. Molecular genetics and cellular features of TFE3 and TFEB fusion kidney cancers. Nat. Rev. Urol. 11, 465–475 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  271. Hochegger, H., Takeda, S. & Hunt, T. Cyclin-dependent kinases and cell-cycle transitions: does one fit all? Nat. Rev. Mol. Cell Biol. 9, 910–916 (2008).

    CAS  PubMed  Google Scholar 

  272. Grechez-Cassiau, A., Rayet, B., Guillaumond, F., Teboul, M. & Delaunay, F. The circadian clock component BMAL1 is a critical regulator of p21WAF1/CIP1 expression and hepatocyte proliferation. J. Biol. Chem. 283, 4535–4542 (2008).

    CAS  PubMed  Google Scholar 

  273. Kowalska, E. et al. NONO couples the circadian clock to the cell cycle. Proc. Natl Acad. Sci. USA 110, 1592–1599 (2013).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  275. Plikus, M. V. et al. Local circadian clock gates cell cycle progression of transient amplifying cells during regenerative hair cycling. Proc. Natl Acad. Sci. USA 110, E2106–E2115 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  276. Geyfman, M. et al. Brain and muscle Arnt-like protein-1 (BMAL1) controls circadian cell proliferation and susceptibility to UVB-induced DNA damage in the epidermis. Proc. Natl Acad. Sci. USA 109, 11758–11763 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  277. Feillet, C. et al. Phase locking and multiple oscillating attractors for the coupled mammalian clock and cell cycle. Proc. Natl Acad. Sci. USA 111, 9828–9833 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  278. Nagoshi, E. et al. Circadian gene expression in individual fibroblasts: cell-autonomous and self-sustained oscillators pass time to daughter cells. Cell 119, 693–705 (2004).

    CAS  PubMed  Google Scholar 

  279. Bieler, J. et al. Robust synchronization of coupled circadian and cell cycle oscillators in single mammalian cells. Mol. Syst. Biol. 10, 739 (2014).

    PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank S. Masri for insight and advice. They thank all members of the Sassone-Corsi laboratory for discussions and support. K.K. was supported by a JSPS fellowship. The authors’ research is supported by the National Institutes of Health (NIH), a Novo Nordisk Challenge Grant and INSERM (France).

Author information

Authors and Affiliations

Authors

Contributions

P.S.-C. and K.K. researched data for the article, made substantial contributions to discussion of content and wrote, reviewed and edited the manuscript.

Corresponding authors

Correspondence to Kenichiro Kinouchi or Paolo Sassone-Corsi.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Dedication

Kenichiro Kinouchi dedicates this article to the memory of Paolo Sassone-Corsi, for his outstanding contributions and scientific advancements in the fields of circadian rhythms, epigenetics and metabolism, and expresses the deepest appreciation for his mentorship and generosity.

Peer review information

Nature Reviews Cancer thanks J. Borniger, C. Dang and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Publisher’s note

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

Glossary

Diurnal feeding–fasting behaviour

The daily behaviour of feeding and fasting under day–night cycles.

Hypothalamic suprachiasmatic nucleus

A group of oscillator neurons that govern circadian rhythms of physiology and behaviour.

Autonomic nervous system

A network of peripheral nerves that govern fundamental organ functions, often unconsciously.

Sympathetic nervous system

A stimulatory branch of the autonomic nervous system that is activated in fight or flight response.

Lipogenesis

A reductive metabolic process of synthesizing lipids by gaining electrons frequently activated in proliferating cells.

Lysosomal dispersion

The peripheral redistribution of normally perinuclear lysosomes.

Cancer/testis antigen

A class of antigenic proteins present on some human cancers but not on adult normal tissues except for testes.

Polysynaptic pathways

Neural pathways comprising multiple neural connections in which signals are transmitted.

Ventromedial hypothalamus

A specialized hypothalamic nucleus that controls appetite and thermogenesis.

Ketogenesis

A metabolic process that produces ketone bodies from fatty acids or amino acids.

Hypocretin/orexin neurons

A group of neurons in the lateral hypothalamus that regulate wakefulness and appetite.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kinouchi, K., Sassone-Corsi, P. Metabolic rivalry: circadian homeostasis and tumorigenesis. Nat Rev Cancer 20, 645–661 (2020). https://doi.org/10.1038/s41568-020-0291-9

Download citation

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41568-020-0291-9

This article is cited by

Search

Quick links

Nature Briefing: Cancer

Sign up for the Nature Briefing: Cancer newsletter — what matters in cancer research, free to your inbox weekly.

Get what matters in cancer research, free to your inbox weekly. Sign up for Nature Briefing: Cancer