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.

  • Article
  • Published:

Low-dimensional dynamics of two coupled biological oscillators

Abstract

The circadian clock and the cell cycle are two biological oscillatory processes that coexist within individual cells. These two oscillators were found to interact, which can lead to their synchronization. Here, we develop a method to identify a low-dimensional stochastic model of the coupled system directly from time-lapse imaging in single cells. In particular, we infer the coupling and nonlinear dynamics of the two oscillators from thousands of mouse and human single-cell fluorescence microscopy traces. This coupling predicts multiple phase-locked states showing different degrees of robustness against molecular fluctuations inherent to cellular-scale biological oscillators. For the 1:1 state, the predicted phase-shifts following period perturbations were validated experimentally. Moreover, the phase-locked states are temperature-independent and evolutionarily conserved from mouse to human, hinting at a common underlying dynamical mechanism. Finally, we detect a signature of the coupled dynamics in a physiological context, explaining why tissues with different proliferation states exhibited shifted circadian clock phases.

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: Reconstructing the phase dynamics and coupling of two biological oscillators.
Fig. 2: Influence of the cell cycle on the circadian phase enables 1:1 phase-locking.
Fig. 3: The coupling between the cell cycle and the circadian oscillator predicts phase-shifts and phase-locking attractors in perturbation experiments.
Fig. 4: Single-cell data and stochastic simulations reveal robust 1:1 and 1:2 phase-locked states.
Fig. 5: Conserved influence of the cell cycle on the circadian clock in human U2OS osteosarcoma cells.
Fig. 6: Temperature cycles do not entrain circadian oscillators in dividing cells and proliferation genes are associated with tissue-specific circadian phases.

Similar content being viewed by others

Data availability

The data supporting the figures and other findings of this study are available from the corresponding author on request.

Code availability

The code is available at https://c4science.ch/diffusion/9123/.

References

  1. 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).

    Article  Google Scholar 

  2. Mermet, J., Yeung, J. & Naef, F. Systems chronobiology: global analysis of gene regulation in a 24-hour periodic world. Cold Spring Harb. Perspect. Biol. 9, https://doi.org/10.1101/cshperspect.a028720 (2017).

  3. Hahn, A. T., Jones, J. T. & Meyer, T. Quantitative analysis of cell cycle phase durations and PC12 differentiation using fluorescent biosensors. Cell Cycle 8, 1044–1052 (2009).

    Article  Google Scholar 

  4. Spencer, S. L. et al. The proliferation–quiescence decision is controlled by a bifurcation in CDK2 activity at mitotic exit. Cell 155, 369–383 (2013).

    Article  Google Scholar 

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

    Article  Google Scholar 

  6. 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).

    Article  ADS  Google Scholar 

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

    Article  ADS  Google Scholar 

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

    Article  ADS  Google Scholar 

  9. Mori, T., Binder, B. & Johnson, C. H. Circadian gating of cell division in cyanobacteria growing with average doubling times of less than 24 hours. Proc. Natl Acad. Sci. USA 93, 10183–10188 (1996).

    Article  ADS  Google Scholar 

  10. Yang, Q., Pando, B. F., Dong, G., Golden, S. S. & van Oudenaarden, A. Circadian gating of the cell cycle revealed in single cyanobacterial cells. Science 327, 1522–1526 (2010).

    Article  ADS  Google Scholar 

  11. Matsu-Ura, T. et al. Intercellular coupling of the cell cycle and circadian clock in adult stem cell culture. Mol. Cell 64, 900–912 (2016).

    Article  Google Scholar 

  12. 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).

    Article  Google Scholar 

  13. Gerard, C. & Goldbeter, A. Entrainment of the mammalian cell cycle by the circadian clock: modeling two coupled cellular rhythms. PLoS Comput. Biol. 8, e1002516 (2012).

    Article  ADS  Google Scholar 

  14. Paijmans, J., Bosman, M., Ten Wolde, P. R. & Lubensky, D. K. Discrete gene replication events drive coupling between the cell cycle and circadian clocks. Proc. Natl Acad. Sci. USA 113, 4063–4068 (2016).

    Article  ADS  Google Scholar 

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

    Article  ADS  Google Scholar 

  16. Rougemont, J. & Naef, F. Collective synchronization in populations of globally coupled phase oscillators with drifting frequencies. Phys. Rev. E 73, 011104 (2006).

    Article  ADS  MathSciNet  Google Scholar 

  17. Rabiner, L. R. & Juang, B.-H. An introduction to hidden Markov models. IEEE ASSP Mag. 3, 4–16 (1986).

    Article  Google Scholar 

  18. Yeom, M., Pendergast, J. S., Ohmiya, Y. & Yamazaki, S. Circadian-independent cell mitosis in immortalized fibroblasts. Proc. Natl Acad. Sci. USA 107, 9665–9670 (2010).

    Article  ADS  Google Scholar 

  19. Vollmers, C., Panda, S. & DiTacchio, L. A high-throughput assay for siRNA-based circadian screens in human U2OS cells. PloS One 3, e3457 (2008).

    Article  ADS  Google Scholar 

  20. Maier, B. et al. A large-scale functional RNAi screen reveals a role for CK2 in the mammalian circadian clock. Genes Dev. 23, 708–718 (2009).

    Article  Google Scholar 

  21. Nicolas, D., Zoller, B., Suter, D. M. & Naef, F. Modulation of transcriptional burst frequency by histone acetylation. Proc. Natl Acad. Sci. USA 115, 7153–7158 (2018).

    Article  Google Scholar 

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

    Article  ADS  Google Scholar 

  23. Dibner, C., Schibler, U. & Albrecht, U. The mammalian circadian timing system: organization and coordination of central and peripheral clocks. Annu. Rev. Physiol. 72, 517–549 (2010).

    Article  Google Scholar 

  24. Saini, C., Morf, J., Stratmann, M., Gos, P. & Schibler, U. Simulated body temperature rhythms reveal the phase-shifting behavior and plasticity of mammalian circadian oscillators. Genes Dev. 26, 567–580 (2012).

    Article  Google Scholar 

  25. Hayes, O. et al. Cell confluency is as efficient as serum starvation for inducing arrest in the G0/G1 phase of the cell cycle in granulosa and fibroblast cells of cattle. Anim. Reprod. Sci. 87, 181–192 (2005).

    Article  Google Scholar 

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

    Article  ADS  Google Scholar 

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

    Article  Google Scholar 

  28. Cermakian, N., Monaco, L., Pando, M. P., Dierich, A. & Sassone-Corsi, P. Altered behavioral rhythms and clock gene expression in mice with a targeted mutation in the Period1 gene. EMBO J. 20, 3967–3974 (2001).

    Article  Google Scholar 

  29. Debruyne, J. P. et al. A clock shock: mouse CLOCK is not required for circadian oscillator function. Neuron 50, 465–477 (2006).

    Article  Google Scholar 

  30. Granada, A. E. & Herzel, H. How to achieve fast entrainment? The timescale to synchronization. PloS One 4, e7057 (2009).

    Article  ADS  Google Scholar 

  31. Heltberg, M., Kellogg, R. A., Krishna, S., Tay, S. & Jensen, M. H. Noise induces hopping between NF-κB entrainment modes. Cell Syst. 3, 532–539 e533 (2016).

    Article  Google Scholar 

  32. Aryal, R. P. et al. Macromolecular assemblies of the mammalian circadian clock. Mol. Cell 67, 770–782.e6 (2017).

    Article  Google Scholar 

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

    Article  Google Scholar 

  34. Mermet, J. et al. Clock-dependent chromatin topology modulates circadian transcription and behavior. Genes Dev. 32, 347–358 (2018).

    Article  Google Scholar 

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

    Article  Google Scholar 

  36. King, R. W., Deshaies, R. J., Peters, J. M. & Kirschner, M. W. How proteolysis drives the cell cycle. Science 274, 1652–1659 (1996).

    Article  ADS  Google Scholar 

  37. Hastings, M. H., Maywood, E. S. & Brancaccio, M. Generation of circadian rhythms in the suprachiasmatic nucleus. Nat. Rev. Neurosci. 19, 453–469 (2018).

    Article  Google Scholar 

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

    Article  Google Scholar 

  39. Pikovsky, A. R. M. K. J. Synchronization (Cambridge University Press, 2001).

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

    Article  Google Scholar 

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

    Article  Google Scholar 

  42. Brunton, S. L., Proctor, J. L. & Kutz, J. N. Discovering governing equations from data by sparse identification of nonlinear dynamical systems. Proc. Natl Acad. Sci. USA 113, 3932–3937 (2016).

    Article  ADS  MathSciNet  Google Scholar 

  43. Soroldoni, D. et al. Genetic oscillations. A Doppler effect in embryonic pattern formation. Science 345, 222–225 (2014).

    Article  ADS  Google Scholar 

  44. Dempster, A. P., Laird, N. M. & Rubin, D. B. Maximum likelihood from incomplete data via the EM algorithm. J. R. Stat. Soc. Ser. B 39, 1–38 (1977).

    MathSciNet  MATH  Google Scholar 

  45. Green, P. J. On use of the EM for penalized likelihood estimation. J. R. Stat. Soc. Ser. B 52, 443–452 (1990).

    MathSciNet  MATH  Google Scholar 

  46. Koksharov, M. I. & Ugarova, N. N. Approaches to engineer stability of beetle luciferases. Comput. Struct. Biotechnol. J. 2, e201209004 (2012).

    Article  Google Scholar 

  47. Huang da, W., Sherman, B. T. & Lempicki, R. A. Bioinformatics enrichment tools: paths toward the comprehensive functional analysis of large gene lists. Nucleic Acids Res. 37, 1–13 (2009).

    Article  Google Scholar 

Download references

Acknowledgements

We thank R. Cannavo for engineering the U2OS-Dual cell line and J. Bieler for initial analyses. We also thank F. Kuttler from the EPFL Biomolecular Screening Facility and L. Bozzo and J. Artacho from the EPFL Bioimaging and Optics Core Facility for assistance with the imaging. Fluorescence-activated cell sorting was performed at the EPFL Flow Cytometry Core Facility. This work was supported by the Swiss National Science Foundation grant 310030_173079 and the EPFL. E.R.P. was supported by a Canadian Institute of Health Research (CIHR 358808) and a SystemsX.ch Transition Postdoc Fellowship (51FSP0163584).

Author information

Authors and Affiliations

Authors

Contributions

C.D., E.R.P. and F.N. designed and participated in the study concept. C.D. and E.R.P. developed computational analysis tools. E.R.P. performed the experiments. C.D. and E.R.P. processed and analysed the experimental data. C.D., E.R.P. and F.N. interpreted the results. E.R.P. and F.N. acquired the funding. F.N. supervised the study. C.D., E.R.P. and F.N. wrote the manuscript.

Corresponding author

Correspondence to Felix Naef.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information: Nature Physics thanks Mogens Jensen, Joris Paijmans 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.

Supplementary information

Supplementary Information

Supplementary Figs. 1–5, methods and refs. 1–9.

Reporting Summary

Supplementary Table 1

Correlation analysis of tissue-specific phases and amplitudes with the mean transcript levels in mouse for every gene. Gene Ontology analyses for the top 200 positively and top 200 negatively correlated genes are provided for both phase and amplitude correlations.

Supplementary Video 1

Single-cell trajectories of circadian and cell-cycle phases cluster around a 1:1 phase-locked state.

Supplementary Video 2

Simulations of the deterministic model show phase-locking and quasiperiodicity.

Supplementary Video 3

Spectral analysis of the deterministic and stochastic simulations shows qualitative differences as a function of the cell-cycle period.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Droin, C., Paquet, E.R. & Naef, F. Low-dimensional dynamics of two coupled biological oscillators. Nat. Phys. 15, 1086–1094 (2019). https://doi.org/10.1038/s41567-019-0598-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41567-019-0598-1

This article is cited by

Search

Quick links

Nature Briefing AI and Robotics

Sign up for the Nature Briefing: AI and Robotics newsletter — what matters in AI and robotics research, free to your inbox weekly.

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