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.

  • Letter
  • Published:

The reversibility of mitotic exit in vertebrate cells

Abstract

A guiding hypothesis for cell-cycle regulation asserts that regulated proteolysis constrains the directionality of certain cell-cycle transitions1,2. Here we test this hypothesis for mitotic exit, which is regulated by degradation of the cyclin-dependent kinase 1 (Cdk1) activator, cyclin B3,4,5. Application of chemical Cdk1 inhibitors to cells in mitosis induces cytokinesis and other normal aspects of mitotic exit, including cyclin B degradation. However, chromatid segregation fails, resulting in entrapment of chromatin in the midbody. If cyclin B degradation is blocked with a proteasome inhibitor or by expression of non-degradable cyclin B, Cdk inhibitors will nonetheless induce mitotic exit and cytokinesis. However, if after mitotic exit, the Cdk1 inhibitor is washed free from cells in which cyclin B degradation is blocked, the cells can revert back to M phase. This reversal is characterized by chromosome recondensation, nuclear envelope breakdown, assembly of microtubules into a mitotic spindle, and in most cases, dissolution of the midbody, reopening of the cleavage furrow, and realignment of chromosomes at the metaphase plate. These findings demonstrate that proteasome-dependent degradation of cyclin B provides directionality for the M phase to G1 transition.

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

Figure 1: The Cdk inhibitor flavopiridol induces reversible mitotic exit and cytokinesis if proteasome activity is inhibited.
Figure 2: Reversibility of mitotic exit requires preservation of cyclin B.
Figure 3: Cells expressing non-degradable cyclin B1 undergo mitotic exit reversal.
Figure 4: Primary cultures of human cells can undergo mitotic exit reversal.

Similar content being viewed by others

References

  1. Minshull, J. et al. The role of cyclin synthesis, modification and destruction in the control of cell division. J. Cell Sci. 12 (suppl.), 77–97 (1989)

    Article  CAS  Google Scholar 

  2. Reed, S. I. Ratchets and clocks: the cell cycle, ubiquitylation and protein turnover. Nature Rev. Mol. Cell Biol. 4, 855–864 (2003)

    Article  ADS  CAS  Google Scholar 

  3. Glotzer, M., Murray, A. W. & Kirschner, M. W. Cyclin is degraded by the ubiquitin pathway. Nature 349, 132–138 (1991)

    Article  ADS  CAS  Google Scholar 

  4. Murray, A. W. & Kirschner, M. W. Cyclin synthesis drives the early embryonic cell cycle. Nature 339, 275–280 (1989)

    Article  ADS  CAS  Google Scholar 

  5. Clute, P. & Pines, J. Temporal and spatial control of cyclin B1 destruction in metaphase. Nature Cell Biol. 1, 82–87 (1999)

    Article  CAS  Google Scholar 

  6. Geley, S. et al. Anaphase-promoting complex/cyclosome-dependent proteolysis of human cyclin A starts at the beginning of mitosis and is not subject to the spindle assembly checkpoint. J. Cell Biol. 153, 137–148 (2001)

    Article  CAS  Google Scholar 

  7. den Elzen, N. & Pines, J. Cyclin A is destroyed in prometaphase and can delay chromosome alignment and anaphase. J. Cell Biol. 153, 121–136 (2001)

    Article  CAS  Google Scholar 

  8. Cross, F. R., Schroeder, L., Kruse, M. & Chen, K. C. Quantitative characterization of a mitotic cyclin threshold regulating exit from mitosis. Mol. Biol. Cell 16, 2129–2138 (2005)

    Article  CAS  Google Scholar 

  9. Parry, D. H. & O'Farrell, P. H. The schedule of destruction of three mitotic cyclins can dictate the timing of events during exit from mitosis. Curr. Biol. 11, 671–683 (2001)

    Article  CAS  Google Scholar 

  10. Sigrist, S., Jacobs, H., Stratmann, R. & Lehner, C. F. Exit from mitosis is regulated by Drosophila fizzy and the sequential destruction of cyclins A, B and B3. EMBO J. 14, 4827–4838 (1995)

    Article  CAS  Google Scholar 

  11. Visintin, R. et al. The phosphatase Cdc14 triggers mitotic exit by reversal of Cdk-dependent phosphorylation. Mol. Cell 2, 709–718 (1998)

    Article  CAS  Google Scholar 

  12. Chang, D. C., Xu, N. & Luo, K. Q. Degradation of cyclin B is required for the onset of anaphase in mammalian cells. J. Biol. Chem. 278, 37865–37873 (2003)

    Article  CAS  Google Scholar 

  13. Carmena, M. & Earnshaw, W. C. The cellular geography of aurora kinases. Nature Rev. Mol. Cell Biol. 4, 842–854 (2003)

    Article  CAS  Google Scholar 

  14. Glotzer, M. The molecular requirements for cytokinesis. Science 307, 1735–1739 (2005)

    Article  ADS  CAS  Google Scholar 

  15. Mishima, M., Pavicic, V., Gruneberg, U., Nigg, E. A. & Glotzer, M. Cell cycle regulation of central spindle assembly. Nature 430, 908–913 (2004)

    Article  ADS  CAS  Google Scholar 

  16. Losiewicz, M. D., Carlson, B. A., Kaur, G., Sausville, E. A. & Worland, P. J. Potent inhibition of CDC2 kinase activity by the flavonoid L86–8275. Biochem. Biophys. Res. Commun. 201, 589–595 (1994)

    Article  CAS  Google Scholar 

  17. Niiya, F., Xie, X., Lee, K. S., Inoue, H. & Miki, T. Inhibition of cyclin-dependent kinase 1 induces cytokinesis without chromosome segregation in an ECT2 and MgcRacGAP-dependent manner. J. Biol. Chem. 280, 36502–36509 (2005)

    Article  CAS  Google Scholar 

  18. Dranovsky, A. et al. Cdc2 phosphorylation of nucleolin demarcates mitotic stages and Alzheimer's disease pathology. Neurobiol. Aging 22, 517–528 (2001)

    Article  CAS  Google Scholar 

  19. Kramer, E. R., Scheuringer, N., Podtelejnikov, A. V., Mann, M. & Peters, J. M. Mitotic regulation of the APC activator proteins CDC20 and CDH1. Mol. Biol. Cell 11, 1555–1569 (2000)

    Article  CAS  Google Scholar 

  20. Pomerening, J. R., Sontag, E. D. & Ferrell, J. E. Jr. Building a cell cycle oscillator: hysteresis and bistability in the activation of Cdc2. Nature Cell Biol. 5, 346–351 (2003)

    Article  CAS  Google Scholar 

  21. Sha, W. et al. Hysteresis drives cell-cycle transitions in Xenopus laevis egg extracts. Proc. Natl Acad. Sci. USA 100, 975–980 (2003)

    Article  ADS  CAS  Google Scholar 

  22. Evans, T., Rosenthal, E. T., Youngblom, J., Distel, D. & Hunt, T. Cyclin: a protein specified by maternal mRNA in sea urchin eggs that is destroyed at each cleavage division. Cell 33, 389–396 (1983)

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the staff at the OMRF Imaging Center and W. Martin for technical assistance. We thank Sanofi-Aventis Pharmaceuticals and the National Cancer Institute for providing flavopiridol. We thank J. Pines for providing cyclin B plasmids and P. Davies for TG-3 anti-phospho-nucleolin antibody. We thank J. Gannon and T. Hunt for antibodies against Cdk1 and cyclins. This work was supported by grants from the National Institutes of Health (to G.J.G. and P.T.S.), from the American Cancer Society (to P.T.S.) and from an NIH training grant (to D.L.S.).

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Gary J. Gorbsky.

Ethics declarations

Competing interests

Reprints and permissions information is available at npg.nature.com/reprintsandpermissions. The authors declare no competing financial interests.

Supplementary information

Supplementary Data

This file contains 3 supplementary figures with their legends. The first supplementary figure summarizes the main finding of the paper. The other two contain supplementary data. After the figures is a detailed description of the methods, a short discussion of the effectiveness of Cdk1 inhibitors other than Flavopiridol, and the Supplementary Video Legends. (PDF 5035 kb)

Supplementary Video 1

This video reveals the effects of the Cdk1 inhibitor Flavopiridol applied to a Xenopus S3 cell in prometaphase, causing premature mitotic exit and cytokinesis. This cell is depicted in Text Fig. 1a. (MOV 1237 kb)

Supplementary Video 2

This video shows that Flavopiridol induces mitotic exit and cytokinesis in a Xenopus S3 cell arrested at metaphase with the proteasome inhibitor MG132. This cell is depicted in Text Fig. 1b. (MOV 3640 kb)

Supplementary Video 3

This video shows that Flavopiridol-induced mitotic exit and cytokinesis in a Xenopus S3 cell cultured in the presence of proteasome inhibitor is reversible upon removal of the Flavopiridol. This cell is depicted in Text Fig. 1c. (QuickTime; 3.5MB) (MOV 3499 kb)

Supplementary Video 4

This video shows a second example of the reversal of mitotic exit and cytokinesis in a Xenopus S3 cell upon removal of Flavopiridol in the presence of proteasome inhibitor. (MOV 4466 kb)

Supplementary Video 5

This video shows a Xenopus S3 cultured in proteasome inhibitor and induced to go through reversal of mitotic exit by addition then removal of Flavopiridol. The proteasome inhibitor is then washed out and the cell subsequently undergoes normal anaphase and mitotic exit. (MOV 5474 kb)

Supplementary Video 6

This video shows a Xenopus S3 transiently arrested with the microtubule drug nocodazole then released to proteasome and subsequently induced to undergo mitotic exit and reversal by addition then removal of Flavopiridol. This cell is depicted in Text Fig. 2a. (MOV 5361 kb)

Supplementary Video 7

This video shows a Hela cell expressing non-degradable cyclin B1. Flavopiridol treatment causes it to exit mitosis and undergo cytokinesis without chromatid separation. Removal of Flavopiridol results in reversal of mitotic exit back to metaphase. This cell is depicted in Text Fig. 3a. (MOV 1948 kb)

Supplementary Video 8

This video shows a Hela cell expressing non-degradable cyclin B1. Flavopiridol treatment causes it to exit mitosis and undergo cytokinesis after chromatid separation. Removal of Flavopiridol results in reversal of mitotic exit back to an anaphase-like condition. This cell is depicted in Text Fig. 3b. (MOV 3054 kb)

Supplementary Video 9

This video shows a Hela cell expressing wild type cyclin B1. Flavopiridol treatment causes it to exit mitosis and undergo cytokinesis without chromatid separation. Removal of Flavopiridol does not result in reversal of mitotic exit. This cell is depicted in Text Fig. 3c. (MOV 2290 kb)

Supplementary Video 10

This video shows that Flavopiridol-induced mitotic exit and cytokinesis in a primary human keratinocyte cell cultured in the presence of proteasome inhibitor is reversible upon removal of the Flavopiridol. This cell is depicted in Text Fig. 4. (MOV 4863 kb)

Supplementary Video 11

This video shows that R0-31-8220 induces mitotic exit and cytokinesis in a Xenopus S3 cell arrested at metaphase with the proteasome inhibitor MG132. This cell is depicted in Supplementary Fig. 3a. (MOV 1298 kb)

Supplementary Video 12

This video shows the normal process of mitosis and cytokinesis in a control Xenopus S3 cell stably expressing GFP-α-tubulin. (MOV 3460 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Potapova, T., Daum, J., Pittman, B. et al. The reversibility of mitotic exit in vertebrate cells. Nature 440, 954–958 (2006). https://doi.org/10.1038/nature04652

Download citation

  • Received:

  • Accepted:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature04652

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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