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:

Regulation of Rho GTPase crosstalk, degradation and activity by RhoGDI1

Abstract

At steady state, most Rho GTPases are bound in the cytosol to Rho guanine nucleotide dissociation inhibitors (RhoGDIs)1. RhoGDIs have generally been considered to hold Rho proteins passively in an inactive state within the cytoplasm. Here we describe an evolutionarily conserved mechanism by which RhoGDI1 controls the homeostasis of Rho proteins in eukaryotic cells. We found that depletion of RhoGDI1 promotes misfolding and degradation of the cytosolic geranylgeranylated pool of Rho GTPases while activating the remaining membrane-bound fraction. Because RhoGDI1 levels are limiting, and Rho proteins compete for binding to RhoGDI1, overexpression of an exogenous Rho GTPase displaces endogenous Rho proteins bound to RhoGDI1, inducing their degradation and inactivation. These results raise important questions about the conclusions drawn from studies that manipulate Rho protein levels. In many cases the response observed may arise not simply from the overexpression itself but from additional effects on the levels and activity of other Rho GTPases as a result of competition for binding to RhoGDI1; this may require a re-evaluation of previously published studies that rely exclusively on these techniques.

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: RhoGDI1 depletion triggers degradation and activation of Rho proteins.
Figure 2: RhoGDI1 depletion impairs cell migration.
Figure 3: Rho-family GTPase degradation after RhoGDI1 depletion does not require activation of the Rho protein but depends on their geranylgeranylation and involves the molecular chaperone machinery.
Figure 4: Competitive interactions with RhoGDI1 regulate the levels and activities of Rho proteins.
Figure 5: RhoGDI regulates Rho protein homeostasis.

Similar content being viewed by others

References

  1. DerMardirossian, C. & Bokoch, G. M. GDIs: central regulatory molecules in Rho GTPase activation. Trends Cell Biol. 15, 356–363 (2005).

    Article  CAS  Google Scholar 

  2. Dransart, E., Morin, A., Cherfils, J. & Olofsson, B. Uncoupling of inhibitory and shuttling functions of rho GDP dissociation inhibitors. J. Biol. Chem. 280, 4674–4683 (2005).

    Article  CAS  Google Scholar 

  3. Gorovoy, M. et al. RhoGDI-1 modulation of the activity of monomeric RhoGTPase RhoA regulates endothelial barrier function in mouse lungs. Circ. Res. 101, 50–58 (2007).

    Article  CAS  Google Scholar 

  4. Abe, M., Qadota, H., Hirata, A. & Ohya, Y. Lack of GTP-bound Rho1p in secretory vesicles of Saccharomyces cerevisiae. J. Cell Biol. 162, 85–97 (2003).

    Article  CAS  Google Scholar 

  5. Tong, Z. et al. Adjacent positioning of cellular structures enabled by a Cdc42 GTPase-activating protein-mediated zone of inhibition. J. Cell Biol. 179, 1375–1384 (2007).

    Article  CAS  Google Scholar 

  6. Togawa, A. et al. Progressive impairment of kidneys and reproductive organs in mice lacking Rho GDIα. Oncogene 18, 5373–5380 (1999).

    Article  CAS  Google Scholar 

  7. Shibata, S. et al. Modification of mineralocorticoid receptor function by Rac1 GTPase: implication in proteinuric kidney disease. Nature Med. 14, 1370–1376 (2008).

    Article  CAS  Google Scholar 

  8. Bielek, H., Anselmo, A. & Dermardirossian, C. Morphological and proliferative abnormalities in renal mesangial cells lacking RhoGDI. Cell Signal. 21, 1974–1983 (2009).

    Article  CAS  Google Scholar 

  9. Tiedje, C., Sakwa, I., Just, U. & Hofken, T. The Rho GDI Rdi1 regulates Rho GTPases by distinct mechanisms. Mol. Biol. Cell 19, 2885–2896 (2008).

    Article  CAS  Google Scholar 

  10. Winter-Vann, A. M. & Casey, P. J. Post-prenylation-processing enzymes as new targets in oncogenesis. Nature Rev. Cancer 5, 405–412 (2005).

    Article  CAS  Google Scholar 

  11. Doye, A. et al. CNF1 exploits the ubiquitin-proteasome machinery to restrict Rho GTPase activation for bacterial host cell invasion. Cell 111, 553–564 (2002).

    Article  CAS  Google Scholar 

  12. Vega, F. M. & Ridley, A. J. SnapShot: Rho family GTPases. Cell 129, 1430 (2007).

    Article  Google Scholar 

  13. Shao, F. et al. Biochemical characterization of the Yersinia YopT protease: cleavage site and recognition elements in Rho GTPases. Proc. Natl Acad. Sci. USA 100, 904–909 (2003).

    Article  CAS  Google Scholar 

  14. Young, J. C., Agashe, V. R., Siegers, K. & Hartl, F. U. Pathways of chaperone-mediated protein folding in the cytosol. Nature Rev. Mol. Cell Biol. 5, 781–791 (2004).

    Article  CAS  Google Scholar 

  15. Michaelson, D. et al. Differential localization of Rho GTPases in live cells: regulation by hypervariable regions and RhoGDI binding. J. Cell Biol. 152, 111–126 (2001).

    Article  CAS  Google Scholar 

  16. Hart, M. J. et al. A GDP dissociation inhibitor that serves as a GTPase inhibitor for the Ras-like protein CDC42Hs. Science 258, 812–815 (1992).

    Article  CAS  Google Scholar 

  17. Ueda, T., Kikuchi, A., Ohga, N., Yamamoto, J. & Takai, Y. Purification and characterization from bovine brain cytosol of a novel regulatory protein inhibiting the dissociation of GDP from and the subsequent binding of GTP to rhoB p20, a ras p21-like GTP-binding protein. J. Biol. Chem. 265, 9373–9380 (1990).

    CAS  PubMed  Google Scholar 

  18. Clark, E. A., Golub, T. R., Lander, E. S. & Hynes, R. O. Genomic analysis of metastasis reveals an essential role for RhoC. Nature 406, 532–535 (2000).

    Article  CAS  Google Scholar 

  19. Guthrie, C., Fink, G., Simon, M. I. & Abelson, J. N. Guide to yeast genetics and molecular biology. Methods Enzymol. 194, 1–863 (1991).

    Google Scholar 

  20. Adamo, J. E. et al. Yeast Cdc42 functions at a late step in exocytosis, specifically during polarized growth of the emerging bud. J. Cell Biol. 155, 581–592 (2001).

    Article  CAS  Google Scholar 

  21. Garcia-Mata, R. et al. Analysis of activated GAPs and GEFs in cell lysates. Methods Enzymol. 406, 425–437 (2006).

    Article  CAS  Google Scholar 

  22. Wennerberg, K. et al. RhoG signals in parallel with Rac1 and Cdc42. J. Biol. Chem. 277, 47810–47817 (2002).

    Article  CAS  Google Scholar 

  23. Arthur, W. T., Noren, N. K. & Burridge, K. Regulation of Rho family GTPases by cell–cell and cell–matrix adhesion. Biol. Res. 35, 239–246 (2002).

    Article  CAS  Google Scholar 

  24. van Buul, J. D. et al. RhoG regulates endothelial apical cup assembly downstream from ICAM1 engagement and is involved in leukocyte trans-endothelial migration. J. Cell Biol. 178, 1279–1293 (2007).

    Article  CAS  Google Scholar 

  25. Huesken, D. et al. Design of a genome-wide siRNA library using an artificial neural network. Nature Biotechnol. 23, 995–1001 (2005).

    Article  CAS  Google Scholar 

  26. Vouret-Craviari, V., Boulter, E., Grall, D., Matthews, C. & Van Obberghen-Schilling, E. ILK is required for the assembly of matrix-forming adhesions and capillary morphogenesis in endothelial cells. J. Cell Sci. 117, 4559–4569 (2004).

    Article  CAS  Google Scholar 

  27. Bagrodia, S., Taylor, S. J., Jordon, K. A., Van Aelst, L. & Cerione, R. A. A novel regulator of p21-activated kinases. J. Biol. Chem. 273, 23633–23636 (1998).

    Article  CAS  Google Scholar 

  28. Ren, X. D., Kiosses, W. B. & Schwartz, M. A. Regulation of the small GTP-binding protein Rho by cell adhesion and the cytoskeleton. EMBO J. 18, 578–585 (1999).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank Lisa Sharek for her technical support, and Channing Der, Jim Bear and Chloé Féral for comments. This study was supported by National Institutes of Health Grants GM029860 (to K.B.) and GM054712 (to P.B.), a Department of Defense Breast Cancer Predoctoral Fellowship (BC051092 to A.D.), a Susan Komen Foundation Postdoctoral Fellowship and a AHA Beginning Grant in Aid (5-40078 to R.G.M.) and a Fondation pour la Recherche Médicale Fellowship (to E.B.), an AHA Postdoctoral Fellowship (0825333E to E.B.) and an Allocation INSERM InCa/AVENIR (R08227AS to E.B.).

Author information

Authors and Affiliations

Authors

Contributions

E.B. and R.G.M. designed, performed experiments and wrote the manuscript. C.G. and A.D. helped with experimental design and procedures. G.R. and P.B. designed and performed the experiments in S. cerevisiae. K.B. directed the project and revised the manuscript. All authors provided detailed comments.

Corresponding author

Correspondence to Keith Burridge.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Information

Supplementary Information (PDF 1262 kb)

Supplementary Information

Supplementary Movie 1 (AVI 11723 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Boulter, E., Garcia-Mata, R., Guilluy, C. et al. Regulation of Rho GTPase crosstalk, degradation and activity by RhoGDI1. Nat Cell Biol 12, 477–483 (2010). https://doi.org/10.1038/ncb2049

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb2049

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