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:

Cdc42–MRCK and Rho–ROCK signalling cooperate in myosin phosphorylation and cell invasion

Abstract

Actomyosin contractility is a mechanism by which cells exert locomotory force against their environment1. Signalling downstream of the small GTPase Rho increases contractility through Rho-kinase (ROCK)-mediated regulation of myosin-II light chain (MLC2) phosphorylation. Cdc42 signalling has been shown to control cell polarity2. Tumour cells can move through a three-dimensional matrix with either a rounded morphology3,4 characterized by Rho–ROCK dependence5 or with an elongated morphology3,4 characterized by Rho–ROCK independence5. Here we show that contractility necessary for elongated morphology and invasion can be generated by Cdc42–MRCK signalling. MRCK (myotonic dystrophy kinase-related Cdc42-binding kinase) cooperates with ROCK in the maintenance of elongated morphology and invasion and either MRCK or ROCK is sufficient for MLC2 phosphorylation, through the inhibitory phosphorylation of myosin phosphatase. By contrast, in rounded ROCK-dependent movement, where MLC2 phosphorylation is higher, MRCK has a smaller role. Our data show that a Cdc42–MRCK signal mediates myosin-dependent cell motility and highlight convergence between Rho and Cdc42 signalling.

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: BE cell invasion requires myosin contractility, MRCK or ROCK.
Figure 2: Elongated morphology requires myosin contractility, MRCK or ROCK.
Figure 4: Rounded morphology requires high MRCK- or ROCK-driven contractility.
Figure 3: MLC2 and MYPT1 phosphorylation require MRCK or ROCK.

Similar content being viewed by others

Accession codes

Accessions

GenBank/EMBL/DDBJ

References

  1. Lauffenburger, D. A. & Horwitz, A. F. Cell migration: a physically integrated molecular process. Cell 84, 359–369 (1996).

    Article  CAS  Google Scholar 

  2. Raftopoulou, M. & Hall, A. Cell migration: Rho GTPases lead the way. Dev. Biol. 265, 23–32 (2004).

    Article  CAS  Google Scholar 

  3. Wolf, K. et al. Compensation mechanism in tumor cell migration: mesenchymal-amoeboid transition after blocking of pericellular proteolysis. J. Cell Biol. 160, 267–277 (2003).

    Article  CAS  Google Scholar 

  4. Friedl, P. & Wolf, K. Tumour-cell invasion and migration: diversity and escape mechanisms. Nature Rev. Cancer 3, 362–374 (2003).

    Article  CAS  Google Scholar 

  5. Sahai, E. & Marshall, C. J. Differing modes of tumour cell invasion have distinct requirements for Rho/ROCK signalling and extracellular proteolysis. Nature Cell Biol. 5, 711–719 (2003).

    Article  CAS  Google Scholar 

  6. Ishizaki, T. et al. The small GTP-binding protein Rho binds to and activates a 160 kDa Ser/Thr protein kinase homologous to myotonic dystrophy kinase. EMBO J. 15, 1885–1893 (1996).

    Article  CAS  Google Scholar 

  7. Leung, T., Chen, X. Q., Manser, E. & Lim, L. The p160 RhoA-binding kinase ROK alpha is a member of a kinase family and is involved in the reorganization of the cytoskeleton. Mol. Cell. Biol. 16, 5313–5327 (1996).

    Article  CAS  Google Scholar 

  8. Matsui, T. et al. Rho-associated kinase, a novel serine/threonine kinase, as a putative target for small GTP binding protein Rho. EMBO J. 15, 2208–2216 (1996).

    Article  CAS  Google Scholar 

  9. Leung, T., Chen, X. Q., Tan, I., Manser, E. & Lim, L. Myotonic dystrophy kinase-related Cdc42-binding kinase acts as a Cdc42 effector in promoting cytoskeletal reorganization. Mol. Cell. Biol. 18, 130–140 (1998).

    Article  CAS  Google Scholar 

  10. Amano, M. et al. Phosphorylation and activation of myosin by Rho-associated kinase (Rho-kinase). J. Biol. Chem. 271, 20246–20249 (1996).

    Article  CAS  Google Scholar 

  11. Kimura, K. et al. Regulation of myosin phosphatase by Rho and Rho-associated kinase (Rho-kinase). Science 273, 245–248 (1996).

    Article  CAS  Google Scholar 

  12. Tan, I., Ng, C. H., Lim, L. & Leung, T. Phosphorylation of a novel myosin binding subunit of protein phosphatase 1 reveals a conserved mechanism in the regulation of actin cytoskeleton. J. Biol. Chem. 276, 21209–21216 (2001).

    Article  CAS  Google Scholar 

  13. Ikebe, M. & Hartshorne, D. J. Phosphorylation of smooth muscle myosin at two distinct sites by myosin light chain kinase. J. Biol. Chem. 260, 10027–10031 (1985).

    CAS  PubMed  Google Scholar 

  14. Komatsu, S. & Ikebe, M. ZIP kinase is responsible for the phosphorylation of myosin II and necessary for cell motility in mammalian fibroblasts. J. Cell Biol. 165, 243–254 (2004).

    Article  CAS  Google Scholar 

  15. Kawano, Y. et al. Phosphorylation of myosin-binding subunit (MBS) of myosin phosphatase by Rho-kinase in vivo. J. Cell Biol. 147, 1023–1038 (1999).

    Article  CAS  Google Scholar 

  16. Feng, J. et al. Inhibitory phosphorylation site for Rho-associated kinase on smooth muscle myosin phosphatase. J. Biol. Chem. 274, 37385–37390 (1999).

    Article  CAS  Google Scholar 

  17. Mills, J.C., Stone, N. L., Erhardt, J. & Pittman, R. N. Apoptotic membrane blebbing is regulated by myosin light chain phosphorylation. J. Cell Biol. 140, 627–636 (1998).

    Article  CAS  Google Scholar 

  18. Straight, A. F. et al. Dissecting temporal and spatial control of cytokinesis with a myosin II inhibitor. Science 299, 1743–1747 (2003).

    Article  CAS  Google Scholar 

  19. Friedl, P. & Brocker, E. B. Reconstructing leukocyte migration in 3D extracellular matrix by time-lapse videomicroscopy and computer-assisted tracking. Methods Mol. Biol. 239, 77–90 (2004).

    PubMed  Google Scholar 

  20. Mukai, H. The structure and function of PKN, a protein kinase having a catalytic domain homologous to that of PKC. J. Biochem. (Tokyo) 133, 17–27 (2003).

    Article  CAS  Google Scholar 

  21. Tan, I., Seow, K. T., Lim, L. & Leung, T. Intermolecular and intramolecular interactions regulate catalytic activity of myotonic dystrophy kinase-related Cdc42-binding kinase alpha. Mol. Cell. Biol. 21, 2767–2778 (2001).

    Article  CAS  Google Scholar 

  22. Nakamura, N. et al. Phosphorylation of ERM proteins at filopodia induced by Cdc42. Genes Cells 5, 571–581 (2000).

    Article  CAS  Google Scholar 

  23. Dong, J. M., Leung, T., Manser, E. & Lim, L. Cdc42 antagonizes inductive action of cAMP on cell shape, via effects of the myotonic dystrophy kinase-related Cdc42-binding kinase (MRCK) on myosin light chain phosphorylation. Eur. J. Cell Biol. 81, 231–242 (2002).

    Article  CAS  Google Scholar 

  24. Ichikawa, K., Ito, M. & Hartshorne, D. J. Phosphorylation of the large subunit of myosin phosphatase and inhibition of phosphatase activity. J. Biol. Chem. 271, 4733–4740 (1996).

    Article  CAS  Google Scholar 

  25. Kiosses, W. B., Daniels, R. H., Otey, C., Bokoch, G. M. & Schwartz, M. A. A role for p21-activated kinase in endothelial cell migration. J. Cell Biol. 147, 831–844 (1999).

    Article  CAS  Google Scholar 

  26. Sells, M. A., Boyd, J. T. & Chernoff, J. p21-activated kinase 1 (Pak1) regulates cell motility in mammalian fibroblasts. J. Cell Biol. 145, 837–849 (1999).

    Article  CAS  Google Scholar 

  27. Sanders, L. C., Matsumura, F., Bokoch, G. M. & de Lanerolle, P. Inhibition of myosin light chain kinase by p21-activated kinase. Science 283, 2083–2085 (1999).

    Article  CAS  Google Scholar 

  28. De Wever, O. et al. Tenascin-C and SF/HGF produced by myofibroblasts in vitro provide convergent pro-invasive signals to human colon cancer cells through RhoA and Rac. FASEB J. 18, 1016–1018 (2004).

    Article  CAS  Google Scholar 

  29. Itoh, K. et al. An essential part for Rho-associated kinase in the transcellular invasion of tumor cells. Nature Med. 5, 221–225 (1999).

    Article  CAS  Google Scholar 

  30. Malliri, A. et al. The transcription factor AP-1 is required for EGF-induced activation of rho-like GTPases, cytoskeletal rearrangements, motility, and in vitro invasion of A431 cells. J. Cell Biol. 143, 1087–1099 (1998).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank T. Leung for generous gifts of reagents, A. Klippel for the gift of anti-PKN3 antiserum, C. MacKintosh for the gift of microcystin-LR, J. Metcalfe for peptide synthesis, and A. Hall, J. Rosenblatt and E. Sahai for helpful discussion. S.W. is funded by an Institute of Cancer Research studentship. C.J.M. is a Gibb Life Fellow of Cancer Research UK. Work in C.J.M.'s laboratory is supported by Cancer Research UK.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Christopher J. Marshall.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Cite this article

Wilkinson, S., Paterson, H. & Marshall, C. Cdc42–MRCK and Rho–ROCK signalling cooperate in myosin phosphorylation and cell invasion. Nat Cell Biol 7, 255–261 (2005). https://doi.org/10.1038/ncb1230

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb1230

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

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