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:

Hydrostatic pressure and the actomyosin cortex drive mitotic cell rounding

A Publisher Correction to this article was published on 26 June 2019

Abstract

During mitosis, adherent animal cells undergo a drastic shape change, from essentially flat to round1,2,3. Mitotic cell rounding is thought to facilitate organization within the mitotic cell and be necessary for the geometric requirements of division4,5,6,7. However, the forces that drive this shape change remain poorly understood in the presence of external impediments, such as a tissue environment2. Here we use cantilevers to track cell rounding force and volume. We show that cells have an outward rounding force, which increases as cells enter mitosis. We find that this mitotic rounding force depends both on the actomyosin cytoskeleton and the cells’ ability to regulate osmolarity. The rounding force itself is generated by an osmotic pressure. However, the actomyosin cortex is required to maintain this rounding force against external impediments. Instantaneous disruption of the actomyosin cortex leads to volume increase, and stimulation of actomyosin contraction leads to volume decrease. These results show that in cells, osmotic pressure is balanced by inwardly directed actomyosin cortex contraction. Thus, by locally modulating actomyosin-cortex-dependent surface tension and globally regulating osmotic pressure, cells can control their volume, shape and mechanical properties.

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

Figure 1: Cells exert an increased rounding pressure in mitosis.
Figure 2: Mitotic cells require a functional actin cytoskeleton to generate rounding pressure.
Figure 3: The actomyosin cortex contracts against an intracellular osmotic pressure.
Figure 4: Animal cells control shape in mitosis by modulating intracellular pressure in conjunction with actomyosin activity.

Similar content being viewed by others

References

  1. Cramer, L. P. & Mitchison, T. J. Investigation of the mechanism of retraction of the cell margin and rearward flow of nodules during mitotic cell rounding. Mol. Biol. Cell 8, 109–119 (1997)

    Article  CAS  Google Scholar 

  2. Gibson, M. C., Patel, A. B., Nagpal, R. & Perrimon, N. The emergence of geometric order in proliferating metazoan epithelia. Nature 442, 1038–1041 (2006)

    Article  ADS  CAS  Google Scholar 

  3. Harris, A. Location of cellular adhesions to solid substrata. Dev. Biol. 35, 97–114 (1973)

    Article  CAS  Google Scholar 

  4. Carreno, S. et al. Moesin and its activating kinase Slik are required for cortical stability and microtubule organization in mitotic cells. J. Cell Biol. 180, 739–746 (2008)

    Article  CAS  Google Scholar 

  5. Kunda, P. & Baum, B. The actin cytoskeleton in spindle assembly and positioning. Trends Cell Biol. 19, 174–179 (2009)

    Article  CAS  Google Scholar 

  6. Kunda, P., Pelling, A. E., Liu, T. & Baum, B. Moesin controls cortical rigidity, cell rounding, and spindle morphogenesis during mitosis. Curr. Biol. 18, 91–101 (2008)

    Article  CAS  Google Scholar 

  7. Théry, M. & Bornens, M. Cell shape and cell division. Curr. Opin. Cell Biol. 18, 648–657 (2006)

    Article  Google Scholar 

  8. Fujibuchi, T. et al. AIP1/WDR1 supports mitotic cell rounding. Biochem. Biophys. Res. Commun. 327, 268–275 (2005)

    Article  CAS  Google Scholar 

  9. Maddox, A. S. & Burridge, K. RhoA is required for cortical retraction and rigidity during mitotic cell rounding. J. Cell Biol. 160, 255–265 (2003)

    Article  CAS  Google Scholar 

  10. Matzke, R., Jacobson, K. & Radmacher, M. Direct, high-resolution measurement of furrow stiffening during division of adherent cells. Nature Cell Biol. 3, 607–610 (2001)

    Article  CAS  Google Scholar 

  11. Hiramoto, Y. Mechanical properties of sea urchin eggs. I. Surface force and elastic modulus of the cell membrane. Exp. Cell Res. 32, 59–75 (1963)

    Article  CAS  Google Scholar 

  12. Krendel, M., Zenke, F. T. & Bokoch, G. M. Nucleotide exchange factor GEF-H1 mediates cross-talk between microtubules and the actin cytoskeleton. Nature Cell Biol. 4, 294–301 (2002)

    Article  CAS  Google Scholar 

  13. Cunningham, C. C. Actin polymerization and intracellular solvent flow in cell surface blebbing. J. Cell Biol. 129, 1589–1599 (1995)

    Article  CAS  Google Scholar 

  14. Charras, G. T., Hu, C. K., Coughlin, M. & Mitchison, T. J. Reassembly of contractile actin cortex in cell blebs. J. Cell Biol. 175, 477–490 (2006)

    Article  CAS  Google Scholar 

  15. Charras, G. T., Coughlin, M., Mitchison, T. J. & Mahadevan, L. Life and times of a cellular bleb. Biophys. J. 94, 1836–1853 (2008)

    Article  CAS  Google Scholar 

  16. Tinevez, J. Y. et al. Role of cortical tension in bleb growth. Proc. Natl Acad. Sci. USA 106, 18581–18586 (2009)

    Article  ADS  CAS  Google Scholar 

  17. Lang, F. Mechanisms and Significance of Cell Volume Regulation (Karger, 2006)

    Book  Google Scholar 

  18. Wehner, F., Olsen, H., Tinel, H., Kinne-Saffran, E. & Kinne, R. K. Cell volume regulation: osmolytes, osmolyte transport, and signal transduction. Rev. Physiol. Biochem. Pharmacol. 148, 1–80 (2003)

    Article  CAS  Google Scholar 

  19. Putney, L. K. & Barber, D. L. Na-H exchange-dependent increase in intracellular pH times G2/M entry and transition. J. Biol. Chem. 278, 44645–44649 (2003)

    Article  CAS  Google Scholar 

  20. Valeva, A. et al. Staphylococcal alpha-toxin: repair of a calcium-impermeable pore in the target cell membrane. Mol. Microbiol. 36, 467–476 (2000)

    Article  CAS  Google Scholar 

  21. Koschinski, A. et al. Why Escherichia coli alpha-hemolysin induces calcium oscillations in mammalian cells–the pore is on its own. FASEB J. 20, 973–975 (2006)

    Article  CAS  Google Scholar 

  22. Kolega, J. Phototoxicity and photoinactivation of blebbistatin in UV and visible light. Biochem. Biophys. Res. Commun. 320, 1020–1025 (2004)

    Article  CAS  Google Scholar 

  23. Hochmuth, R. M. Micropipette aspiration of living cells. J. Biomech. 33, 15–22 (2000)

    Article  CAS  Google Scholar 

  24. Salbreux, G., Joanny, J. F., Prost, J. & Pullarkat, P. Shape oscillations of non-adhering fibroblast cells. Phys. Biol. 4, 268–284 (2007)

    Article  ADS  CAS  Google Scholar 

  25. Blaser, H. et al. Migration of zebrafish primordial germ cells: a role for myosin contraction and cytoplasmic flow. Dev. Cell 11, 613–627 (2006)

    Article  CAS  Google Scholar 

  26. Bereiter-Hahn, J. Mechanics of crawling cells. Med. Eng. Phys. 27, 743–753 (2005)

    Article  CAS  Google Scholar 

  27. Charras, G. & Paluch, E. Blebs lead the way: how to migrate without lamellipodia. Nature Rev. Mol. Cell Biol. 9, 730–736 (2008)

    Article  CAS  Google Scholar 

  28. Mitchison, T. J., Charras, G. T. & Mahadevan, L. Implications of a poroelastic cytoplasm for the dynamics of animal cell shape. Semin. Cell. Dev. Biol. 19, 215–223 (2008)

    Article  CAS  Google Scholar 

  29. Keren, K., Yam, P. T., Kinkhabwala, A., Mogilner, A. & Theriot, J. A. Intracellular fluid flow in rapidly moving cells. Nature Cell Biol. 11, 1219–1224 (2009)

    Article  CAS  Google Scholar 

  30. Harold, F. M. To shape a cell: an inquiry into the causes of morphogenesis of microorganisms. Microbiol. Rev. 54, 381–431 (1990)

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Neumann, B. et al. High-throughput RNAi screening by time-lapse imaging of live human cells. Nature Methods 3, 385–390 (2006)

    Article  CAS  Google Scholar 

  32. Ruta, M. et al. Nucleotide sequence of the two rat cellular rasH genes. Mol. Cell. Biol. 6, 1706–1710 (1986)

    Article  CAS  Google Scholar 

  33. Hutter, J. L. & Bechhoefer, J. Calibration of atomic-force microscope tips. Rev. Sci. Instrum. 64, 1868–1873 (1993)

    Article  ADS  CAS  Google Scholar 

  34. Hertz, H. Uber den Kontakt elastischer Korper. J. Reine Angew. Math. 92, 156–172 (1881)

    Google Scholar 

  35. Radmacher, M., Fritz, M. & Hansma, P. K. Imaging soft samples with the atomic force microscope: gelatin in water and propanol. Biophys. J. 69, 264–270 (1995)

    Article  ADS  CAS  Google Scholar 

Download references

Acknowledgements

DFG, BMBF and SNF supported this project. JSPS supported Y.T. A.A.H. is funded by the Max Planck Society. We thank S. Bhakdi for toxins and advice on their use, T. J. Mitchison for extensive discussions on osmotic pressure and critical reading of the manuscript, M. Krieg for valuable insights into cell blebbing, and B. Baum, C. Brangwynne, S. Grill, A. Helenius, J. Howard, F. Jülicher, Z. Maliga and E. Paluch for discussions and critical reading of the manuscript.

Author information

Authors and Affiliations

Authors

Contributions

M.P.S., D.J.M., Y.T. and A.A.H. designed and implemented the assay. M.P.S. performed the experiments except for the Young’s modulus measurements, which were made by J.H. S.P.R. contributed to Fig. 2 and Supplementary Fig. 6. Y.T. produced cell lines. J.H. designed the toxin experiments. M.P.S. analysed data and created the figures. M.P.S., J.H., D.J.M. and A.A.H. wrote the manuscript.

Corresponding authors

Correspondence to Daniel J. Muller or Anthony A. Hyman.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Information

This file contains Supplementary Figures 1-9 with legends, Supplementary Table 1, a Supplementary Discussion and additional references. (PDF 4704 kb)

PowerPoint slides

Rights and permissions

Reprints and permissions

About this article

Cite this article

Stewart, M., Helenius, J., Toyoda, Y. et al. Hydrostatic pressure and the actomyosin cortex drive mitotic cell rounding. Nature 469, 226–230 (2011). https://doi.org/10.1038/nature09642

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nature09642

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