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:

Feedback regulation through myosin II confers robustness on RhoA signalling at E-cadherin junctions

Abstract

Actomyosin at the epithelial zonula adherens (ZA) generates junctional tension for tissue integrity and morphogenesis. This requires the RhoA GTPase, which establishes a strikingly stable active zone at the ZA. Mechanisms must then exist to confer robustness on junctional RhoA signalling at the population level. We now identify a feedback network that generates a stable mesoscopic RhoA zone out of dynamic elements. The key is scaffolding of ROCK1 to the ZA by myosin II. ROCK1 protects junctional RhoA by phosphorylating Rnd3 to prevent the cortical recruitment of the Rho suppressor, p190B RhoGAP. Combining predictive modelling and experimentation, we show that this network constitutes a bistable dynamical system that is realized at the population level of the ZA. Thus, stability of the RhoA zone is an emergent consequence of the network of interactions that allow myosin II to feedback to RhoA.

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: Myosin II supports a stable Rho zone at the ZA.
Figure 2: p190B RhoGAP disrupts the junctional Rho zone when NMII is inactivated.
Figure 3: Myosin antagonizes the junctional localization of Rnd3.
Figure 4: Rnd3 recruits p190B to the ZA when NMII is inhibited.
Figure 5: ROCK1 phosphorylates Rnd3 to support junctional RhoA signalling.
Figure 6: Myosin II scaffolds ROCK1 at the epithelial ZA.
Figure 7: Bistable properties of junctional RhoA.

Similar content being viewed by others

References

  1. Fernandez-Gonzalez, R., Simoes Sde, M., Roper, J. C., Eaton, S. & Zallen, J. A. Myosin II dynamics are regulated by tension in intercalating cells. Dev. Cell 17, 736–743 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Smutny, M. et al. Myosin II isoforms identify distinct functional modules that support integrity of the epithelial zonula adherens. Nat. Cell Biol. 12, 696–702 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Martin, A. C., Kaschube, M. & Wieschaus, E. F. Pulsed contractions of an actin-myosin network drive apical constriction. Nature 457, 495–499 (2009).

    Article  CAS  PubMed  Google Scholar 

  4. Rauzi, M., Lenne, P. F. & Lecuit, T. Planar polarized actomyosin contractile flows control epithelial junction remodelling. Nature 468, 1110–1114 (2010).

    Article  CAS  PubMed  Google Scholar 

  5. Priya, R., Yap, A. S. & Gomez, G. A. E-cadherin supports steady-state Rho signaling at the epithelial zonula adherens. Differentiation 86, 133–140 (2013).

    Article  CAS  PubMed  Google Scholar 

  6. Wu, S. K. et al. Cortical F-actin stabilization generates apical-lateral patterns of junctional contractility that integrate cells into epithelia. Nat. Cell Biol. 16, 167–178 (2014).

    Article  CAS  PubMed  Google Scholar 

  7. Ratheesh, A. et al. Centralspindlin and α-catenin regulate Rho signalling at the epithelial zonula adherens. Nat. Cell Biol. 14, 818–828 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Martin, A. C., Gelbart, M., Fernandez-Gonzalez, R., Kaschube, M. & Wieschaus, E. Integration of contractile forces during tissue invagination. J. Cell Biol. 188, 735–749 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Carramusa, L., Ballestrem, C., Zilberman, Y. & Bershadsky, A. D. Mammalian diaphanous-related formin Dia1 controls the organization of E-cadherin-mediated cell–cell junctions. J. Cell Sci. 120, 3870–3882 (2007).

    Article  CAS  PubMed  Google Scholar 

  10. Yamada, S. & Nelson, W. J. Localized zones of Rho and Rac activities drive initiation and expansion of epithelial cell–cell adhesion. J. Cell Biol. 178, 517–527 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Bos, J. L., Rehmann, H. & Wittinghofer, A. GEFs and GAPs: critical elements in the control of small G proteins. Cell 129, 865–877 (2007).

    Article  CAS  PubMed  Google Scholar 

  12. Bement, W. M., Miller, A. L. & von Dassow, G. Rho GTPase activity zones and transient contractile arrays. Bioessays 28, 983–993 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Jaffe, A. B. & Hall, A. Rho GTPases: biochemistry and biology. Annu. Rev. Cell Dev. Biol. 21, 247–269 (2005).

    Article  CAS  PubMed  Google Scholar 

  14. Ferrell, J. E. & Machleder, E. M. The biochemical basis of an all-or-none cell fate switch in Xenopus oocytes. Science 280, 895–898 (1998).

    Article  CAS  PubMed  Google Scholar 

  15. Huang, C.-Y. F. & Ferrell, J. E. Ultrasensitivity in the mitogen-activated protein kinase cascade. Proc. Natl Acad. Sci. USA 93, 10078–10083 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Angeli, D., Ferrell, J. E. Jr & Sontag, E. D. Detection of multistability, bifurcations, and hysteresis in a large class of biological positive-feedback systems. Proc. Natl Acad. Sci. USA 101, 1822–1827 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Del Conte-Zerial, P. et al. Membrane identity and GTPase cascades regulated by toggle and cut-out switches. Mol. Syst. Biol. 4, 206 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Piekny, A. J. & Glotzer, M. Anillin is a scaffold protein that links RhoA, actin, and myosin during cytokinesis. Curr. Biol. 18, 30–36 (2008).

    Article  CAS  PubMed  Google Scholar 

  19. Tse, Y. C. et al. RhoA activation during polarization and cytokinesis of the early Caenorhabditis elegans embryo is differentially dependent on NOP-1 and CYK-4. Mol. Biol. Cell 23, 4020–4031 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Sun, L. et al. Mechanistic insights into the anchorage of the contractile ring by anillin and mid1. Dev. Cell 33, 413–426 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Lee, C. S., Choi, C. K., Shin, E. Y., Schwartz, M. A. & Kim, E. G. Myosin II directly binds and inhibits Dbl family guanine nucleotide exchange factors: a possible link to Rho family GTPases. J. Cell Biol. 190, 663–674 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Kuo, J. C., Han, X., Hsiao, C. T., Yates, J. R. III & Waterman, C. M. Analysis of the myosin-II-responsive focal adhesion proteome reveals a role for β-Pix in negative regulation of focal adhesion maturation. Nat. Cell Biol. 13, 383–393 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Wu, D., Asiedu, M. & Wei, Q. Myosin-interacting guanine exchange factor (MyoGEF) regulates the invasion activity of MDA-MB-231 breast cancer cells through activation of RhoA and RhoC. Oncogene 28, 2219–2230 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Yuce, O., Piekny, A. & Glotzer, M. An ECT2-centralspindlin complex regulates the localization and function of RhoA. J. Cell Biol. 170, 571–582 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Even-Ram, S. et al. Myosin IIA regulates cell motility and actomyosin-microtubule crosstalk. Nat. Cell Biol. 9, 299–309 (2007).

    Article  CAS  PubMed  Google Scholar 

  26. Bustos, R. I., Forget, M. A., Settleman, J. E. & Hansen, S. H. Coordination of Rho and Rac GTPase function via p190B RhoGAP. Curr. Biol. 18, 1606–1611 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Wu, M. H. et al. MCT-1 expression and PTEN deficiency synergistically promote neoplastic multinucleation through the Src/p190B signaling activation. Oncogene 33, 5109–5120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Wennerberg, K. et al. Rnd proteins function as RhoA antagonists by activating p190 RhoGAP. Curr. Biol. 13, 1106–1115 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Riou, P. et al. 14-3-3 proteins interact with a hybrid prenyl-phosphorylation motif to inhibit G proteins. Cell 153, 640–653 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Riento, K. et al. RhoE function is regulated by ROCK I-mediated phosphorylation. EMBO J. 24, 1170–1180 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Riento, K., Villalonga, P., Garg, R. & Ridley, A. Function and regulation of RhoE. Biochem. Soc. Trans. 33, 649–651 (2005).

    Article  CAS  PubMed  Google Scholar 

  32. Smith, A. L., Dohn, M. R., Brown, M. V. & Reynolds, A. B. Association of Rho-associated protein kinase 1 with E-cadherin complexes is mediated by p120-catenin. Mol. Biol. Cell 23, 99–110 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Hu, A., Wang, F. & Sellers, J. R. Mutations in human nonmuscle myosin IIA found in patients with May–Hegglin anomaly and Fechtner syndrome result in impaired enzymatic function. J. Biol. Chem. 277, 46512–46517 (2002).

    Article  PubMed  Google Scholar 

  34. Beach, J. R. & Egelhoff, T. T. Myosin II recruitment during cytokinesis independent of centralspindlin-mediated phosphorylation. J. Biol. Chem. 284, 27377–27383 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Otani, T., Ichii, T., Aono, S. & Takeichi, M. Cdc42 GEF Tuba regulates the junctional configuration of simple epithelial cells. J. Cell Biol. 175, 135–146 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Simoes Sde, M., Mainieri, A. & Zallen, J. A. Rho GTPase and Shroom direct planar polarized actomyosin contractility during convergent extension. J. Cell Biol. 204, 575–589 (2014).

    Article  CAS  PubMed  Google Scholar 

  37. Zhang, B. & Zheng, Y. Regulation of RhoA GTP hydrolysis by the GTPase-activating proteins p190, p50RhoGAP, Bcr, and 3BP-1. Biochemistry 37, 5249–5257 (1998).

    Article  CAS  PubMed  Google Scholar 

  38. Omelchenko, T. & Hall, A. Myosin-IXA regulates collective epithelial cell migration by targeting RhoGAP activity to cell–cell junctions. Curr. Biol. 22, 278–288 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Elbediwy, A. et al. Epithelial junction formation requires confinement of Cdc42 activity by a novel SH3BP1 complex. J. Cell Biol. 198, 677–693 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Gierer, A. & Meinhardt, H. A theory of biological pattern formation. Kybernetik 12, 30–39 (1972).

    Article  CAS  PubMed  Google Scholar 

  41. Kondo, S. & Miura, T. Reaction–diffusion model as a framework for understanding biological pattern formation. Science 329, 1616–1620 (2010).

    Article  CAS  PubMed  Google Scholar 

  42. Shewan, A. M. et al. Myosin 2 is a key Rho kinase target necessary for the local concentration of E-cadherin at cell–cell contacts. Mol. Biol. Cell 16, 4531–4532 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Schramek, D. et al. Direct in vivo RNAi screen unveils myosin IIa as a tumor suppressor of squamous cell carcinomas. Science 343, 309–313 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Vicente-Manzanares, M., Ma, X., Adelstein, R. S. & Horwitz, A. R. Non-muscle myosin II takes centre stage in cell adhesion and migration. Nat. Rev. Mol. Cell Biol. 10, 778–790 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Nishimura, T. & Takeichi, M. Shroom3-mediated recruitment of Rho kinases to the apical cell junctions regulates epithelial and neuroepithelial planar remodeling. Development 135, 1493–1502 (2008).

    Article  CAS  PubMed  Google Scholar 

  46. Moore, T. et al. Self-organizing actomyosin patterns on the cell cortex at epithelial cell–cell junctions. Biophys. J. 107, 2652–2661 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Munjal, A., Philippe, J. M., Munro, E. & Lecuit, T. A self-organized biomechanical network drives shape changes during tissue morphogenesis. Nature 524, 351–355 (2015).

    Article  CAS  PubMed  Google Scholar 

  48. Bois, J. S., Julicher, F. & Grill, S. W. Pattern formation in active fluids. Phys. Rev. Lett. 106, 028103 (2011).

    Article  CAS  PubMed  Google Scholar 

  49. Goehring, N. W. et al. Polarization of PAR proteins by advective triggering of a pattern-forming system. Science 334, 1137–1141 (2011).

    Article  CAS  PubMed  Google Scholar 

  50. Smutny, M. et al. Multicomponent analysis of junctional movements regulated by myosin II isoforms at the epithelial zonula adherens. PLoS ONE 6, e22458 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Liu, Z., Adams, H. C. III & Whitehead, I. P. The rho-specific guanine nucleotide exchange factor Dbs regulates breast cancer cell migration. J. Biol. Chem. 284, 15771–15780 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. McLachlan, R. W. & Yap, A. S. Protein tyrosine phosphatase activity is necessary for E-cadherin-activated Src signaling. Cytoskeleton (Hoboken) 68, 32–43 (2011).

    Article  CAS  Google Scholar 

  53. Priya, R. & Gomez, G. A. Measurement of junctional protein dynamics using fluorescence recovery after photobleaching (FRAP). Bio-Protocol 3, e937 (2013).

    Article  Google Scholar 

  54. MATLAB and Statistics Toolbox Release 2012b (MATLAB, 2012)

  55. Itoh, R. E. et al. Activation of rac and cdc42 video imaged by fluorescent resonance energy transfer-based single-molecule probes in the membrane of living cells. Mol. Cell. Biol. 22, 6582–6591 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Pertz, O., Hodgson, L., Klemke, R. L. & Hahn, K. M. Spatiotemporal dynamics of RhoA activity in migrating cells. Nature 440, 1069–1072 (2006).

    Article  CAS  PubMed  Google Scholar 

  57. Leerberg, J. M. et al. Tension-sensitive actin assembly supports contractility at the epithelial zonula adherens. Curr. Biol. 24, 1689–1699 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank our laboratory colleagues for their unstinting support and advice during the course of this project, and our colleagues elsewhere for their kind gifts of reagents. We also thank M. Zerial and L. Tucker-Kellogg for thought-provoking discussions, M. Naghibosadat for her help with cloning and E. Moussa for his help in nano-ablation analysis. This work was supported by the National Health and Medical Research Council of Australia (1044041, 1037320, 1067405), The Kids Cancer Project of the Oncology Research Foundation, the EMPathy National Collaborative Research Program (CG-10-04) of the National Breast Cancer Foundation (Australia), a University of Queensland Early Career Research Grant to G.A.G. (2012003354) and an ANZ Trustees PhD Scholarship in Medical Research to R.P.

Author information

Authors and Affiliations

Authors

Contributions

R.P., G.A.G. and A.S.Y. conceived the project and designed the experiments. R.P. performed most of the experiments in collaboration with G.A.G. for nano-ablation, FRAP and Rho biosensor experiments, with assistance from S.V. for western blotting and immunoprecipitation experiments. G.A.G. and N.A.H. performed the mathematical modelling. H.L.C. performed experiments. S.B. performed the cloning and characterization of AHPH mutants. R.P. and G.A.G. analysed the data. R.P., G.A.G. and A.S.Y. wrote the paper.

Corresponding authors

Correspondence to Guillermo A. Gomez or Alpha S. Yap.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 2 Myosin II supports a stable Rho zone at the ZA.

(a) RhoA, E-cadherin and ZO-1 localization in TCA-fixed MCF-7 cells. (b) Domain organization of human full-length Anillin (FL-Anillin) and the respective constructs used in this study. The residues are numbered according to the Uniprot Id Q9NQW6. FBD (Formin binding domain); MBD (Myosin binding domain); ABD (Actin binding domain); AHD (Anillin homology domain), which is composed of RBD (Rho binding domain) and C2 domain. The AHPH (location biosensor) is composed of AHD and PH (Pleckstrin homology domain). (c) Representative images and fluorescence line-scan analysis of GFP-AHPH and endogenous RhoA at the ZA of MCF-7 cells. White line (merged image) represents the position where fluorescence intensity was measured. (d) Localization of RFP-UtrCH and GFP-tagged AHPH, AHPH + C3-T (C3 Transferase, 1 μg ml−1; 3 h), Δ AH, AHPHC2mut, AHPHA740D, AHPHE758K and AHPHA740D E758K in MCF-7 cells. (e) Lysates from control or ECT2 knockdown MCF-7 cells immunoblotted for ECT2 and GAPDH (loading control). (f) GTP-RhoA distribution in control or ECT2 knockdown MCF-7 cells. (g) Control (Ctrl) or E-cadherin RNAi (E-cad KD) MCF-7 cells fixed and immunostained for GFP (GFP AHPH), E-cadherin (E-cad) and ZO-1. (h) Lysates from control (Ctrl) or E-cadherin RNAi (E-cad KD) MCF-7 cells immunoblotted for E-cadherin and β-tubulin (loading control). (i) GTP-RhoA pull-down from control (Ctrl) or E-cadherin RNAi (E-cad KD) or NMIIA RNAi (NMIIA KD) MCF-7 cells using RBD domain of Rhotekin and immunoblotted for RhoA and β-tubulin. (j) NMIIA and E-cadherin localization in MCF-7 cells treated with vehicle (DMSO) or blebbistatin (BLB, 100 μM, 2 h). (k) RhoA localization in Caco2 and MDCK treated with DMSO or blebbistatin (100 μM, 2 h). (l,m) Lysates from Control (Ctrl), NMIIA knockdown or NMIIB knockdown MCF-7 cells immunoblotted for NMIIA, NMIIB and β-tubulin (loading control). Scale bars, 10 μm. DIC images corresponding to Supplementary Fig. 1d are shown in Supplementary Fig. 7.

Supplementary Figure 3 p190B RhoGAP degrades the junctional Rho zone when NMII is inactivated.

(ae) ECT2 (a), MgcRacGAP (RacGAP) (b), p190A Rho GAP (p190A), Myosin IIA and Myosin IIB (c,e) localization in control (Ctrl), NMIIA RNAi (IIA KD), NMIIB RNAi (IIB KD), DMSO or blebbistatin (BLB, 100 μM, 2 h) treated MCF-7 cells. (f,g) Lysates from control (Ctrl), NMIIA RNAi (IIA KD), NMIIB RNAi (IIB KD), DMSO or blebbistatin (BLB, 100 μM, 2 h) treated MCF-7 cells immunoblotted for p190B Rho GAP and β-tubulin (loading control). (h) Rac 1 localization in control (Ctrl), NMIIA RNAi (IIA KD), NMIIB RNAi (IIB KD), DMSO or blebbistatin (BLB, 100 μM, 2 h) treated MCF-7 cells. (i) MCF-7 cells transfected with non-targeting control siRNA or Rac1 siRNA (Rac1KD) were treated with DMSO or with blebbistatin (BLB; 100 μM, 2 h), then fixed and stained for p190B Rho GAP. (j) Lysates from MCF-7 cells transfected with non-targeting control siRNA (Ctrl) or transfected with siRNA against Rac1 (Rac1KD) were immunoblotted for Rac1 and β-tubulin (loading control). (k) MCF-7 cells were treated with DMSO or blebbistatin (BLB; 100 μM, 2 h) alone or in combination with Rac inhibitors EHT1864 (10 μM, 12 h) or NSC23766 (50 μM, 12 h), fixed with methanol and stained for p190B GAP. (l) MCF-7 cells transduced with lentivirus encoding non-targeting control shRNA (Ctrl), NMIIA (IIAKD) or NMIIB (IIB KD) shRNA were transfected with the Raichu-Rac FRET biosensor and FRET measurements were performed as described in the Methods section. Average emission ratios were calculated at the apical junctions. Data represent mean ± s.e.m. of 30 contacts (n = 30) and the result is representative of two independent experiments. Statistical information, P values and source data are in Supplementary Table 2. (m) MCF-7 cells were treated with DMSO or treated with blebbistatin (BLB) (100 μM, 2 h) alone or in combination with Src inhibitors SU6656 (10 μM) or PP2 (10 μM)) for 4 h. Cells were fixed with methanol and stained for p190B GAP. Scale bars, 10 μm.

Supplementary Figure 4 Rnd3 recruits p190B to the ZA when NMII is inhibited.

(a) MCF-7 cells transfected with non-targeting control siRNA or siRNA against Rnd3 (Rnd3 KD) or Rnd1 (Rnd1 KD) and treated with vehicle control DMSO or blebbistatin (BLB; 100 μM, 2 h) were fixed with methanol and stained for p190B Rho GAP and E-cadherin. Representative confocal images were acquired at the apical junctions. (b) Lysates from MCF-7 cells transfected with non-targeting control siRNA (Ctrl) or siRNA targeted against Rnd3 (Rnd3 KD) were immunoblotted for Rnd3 and β-tubulin (loading control). (c) Lysates from MCF-7 cells transfected with Rnd3siRNA along with GFP-tagged Rnd3 constructs (WT, AIIA, S240A, T55V) were immunoblotted for GFP and β-tubulin (loading control). (d) MCF-7 cells transfected with non-targeting siRNA (Ctrl siRNA) or Rnd3 siRNA (Rnd3 KD) and reconstituted with low levels of GFP, or GFP-tagged WT Rnd3 (WT), AIIA Rnd3 (AIIA), S240A Rnd3 (S240A), or T55V Rnd3 (T55V) were treated with either DMSO or blebbistatin (BLB) (100 μM, 2 h), fixed with TCA and stained for RhoA. Asterisks () indicate GFP-tagged Rnd3 expressing cells. (e) MCF-7 cells transfected with GFP-AHPH and non-targeting siRNA (Ctrl siRNA) or Rnd3 siRNA (Rnd3 KD) and reconstituted with low levels of mCherry, or mCherry-tagged WT Rnd3 (WT), AIIA Rnd3 (AIIA), S240A Rnd3 (S240A), T55V Rnd3 (T55V) were treated with either DMSO or blebbistatin (BLB, 100 μM, 2 h) fixed with TCA and stained for GFP. Asterisks () indicate mCherry-tagged Rnd3 expressing cells. Scale bars, 10 μm.

Supplementary Figure 5 ROCK-1 phosphorylates Rnd3 to support junctional Rho signalling.

(a) Confluent MCF-7 cells were fixed with methanol and stained for ROCK-2 and E-cadherin. Representative images were acquired at the apical junctions of the cells by confocal microscopy. (b,c) Confluent MCF-7 cells were treated with vehicle (0.5% glycerol, Ctrl) or with Rho-inhibitor C3 Transferase (C3T, 1 μg ml−1) for 3 h, fixed with methanol and stained for ROCK-1 and ZO-1. Representative images were acquired at the apical junctions of the cells by confocal microscopy (b) and the fluorescence intensity at cell junctions was quantitated by linescan analysis (c). Data represent mean ± s.e.m. of three individual experiments (n = 3), P < 0.05 Student’s t-test. Statistical information, P values and source data are in Supplementary Table 2. (d) HEK293T cells expressing GFP-Rnd3 were treated with either PBS or Y-27632 (30 μM, 1 h). Rnd3 was immunoprecipitated by GFP-trap and lysates were immunoblotted for GFP and phospho-serine. (e) MCF-7 cells transfected with Rnd3 siRNA and reconstituted with GFP tagged WT Rnd3 (WT), AIIA Rnd3 (AIIA), S240A Rnd3 (S240A) were treated with either PBS or Y-27632 (30 μM, 1 h). Representative images were acquired at the apical junctions of the cells by confocal microscopy. (f) Lysates from MCF-7 cells treated either with PBS or Y-27632 (30 μM, 1 h), or blebbistatin (BLB, 100 μM, 2 h) and blotted for Rnd3 and β-tubulin (loading control). (g) MCF-7 cells were transfected with non-targeting Ctrl siRNA (Ctrl) or ROCK-1 siRNA (ROCK-1KD) and 48 h later treated with either blebbistatin (100 μM, 2 h) alone (BLB) or in combination with ROCK-1 KD (ROCK-1 KD + BLB). Cells were fixed with methanol and stained for p190B Rho GAP or fixed with TCA and stained for RhoA. Representative images were acquired at the apical junctions of the cells by confocal microscopy. (h,i) Lysates from MCF-7 cells transfected with non-targeting control siRNA (ctrl) or ROCK-1 siRNA (ROCK-1 KD) and immunoblotted for ROCK-1, ROCK-2 and β-tubulin (loading control). Scale bars, 10 μm. Corresponding DIC images for 4e are shown in Supplementary Fig. 7.

Supplementary Figure 6 Myosin II scaffolds ROCK-1 at the epithelial Zonula Adherens.

(a,b) MCF-7 cells transduced with lentivirus encoding non-targeting control shRNA (Ctrl) or shRNA against NMIIA (IIAKD) or shRNA against NMIIB (IIB KD) were fixed with methanol and stained for ROCK-1, NMIIA and NMIIB. Representative images were acquired at the apical junctions of the cells by confocal microscopy. (c) HEK293T cells were transfected with GFP-ROCK-1 and treated with blebbistatin (BLB, 100 μM, 2 h). ROCK-1 was immunoprecipitated using GFP-TRAP and lysates were immunoblotted for NMIIA, NMIIB and GFP. (d) HEK293T cells were transfected with GFP-FL NMIIA (full-length) or GFP-NMIIA-ROD and treated with Y-27632 (30 μM, 1 h). Immunoprecipitation was performed using GFP-TRAP and lysates were immunoblotted for ROCK-1 and GFP. (e,g) Lysates from MCF-7 cells transfected with non-targeting control siRNA (Ctrl) or NMIIA siRNA (NMIIAKD, e), Cdc42siRNA (Cdc42 KD, f), or N-WASP siRNA (N-WASP KD, g) and immunoblotted for NMIIA, Cdc42 or N-WASP, respectively, as well as β-tubulin or GAPDH (loading control). (h) MCF-7 cells transfected with non-targeting control siRNA (Ctrl) or siRNA targeted against Cdc42 (Cdc42 KD) or N-WASP(N-WASP KD) were fixed with methanol and stained for ROCK-1 and ZO-1. Representative images were acquired at the apical junctions of the cells by confocal microscopy. (i) MCF-7 cells transfected with non-targeting control siRNA (Ctrl) or siRNA targeted against Cdc42 (Cdc42 KD) or N-WASP (N-WASP KD were fixed with TCA and stained for RhoA. Representative images were acquired at the apical junctions of the cells by confocal microscopy. (j) MCF-7 cells transfected with non-targeting control siRNA (Ctrl) or siRNA targeted against Cdc42 (Cdc42 KD) or N-WASP (N-WASP KD) were fixed with methanol and stained for p190B Rho GAP, E-cadherin or ZO-1. Representative images were acquired at the apical junctions of the cells by confocal microscopy. Scale bars, 10 μm.

Supplementary Figure 7 Bistable properties of RhoA localization at cell–cell junctions.

(a) Using the model and constants as described in Supplementary Table 1, time course plots for Rnd3 against RhoA are shown for three values of KNMIIAROCK1 = 1.4: (left); KNMIIAROCK1 = 2.3 (centre); KNMIIAROCK1 = 1.4 (right) KNMIIAROCK1 = 0.5. In each, initial concentrations of RhoA and Rnd3 were set in a range between 0 and 3.5 and all other concentrations equal to unity. Simulations were run for t = 0 to t = 5,000, with each line corresponding to a time course. Arrows denote the direction of increased time and red circles denote stable points. (b,c) Convergence behaviour of Rnd3 and RhoA upon (b) varying initial values of NMIIA for the network in Fig. 7a and; (c) varying the strength of negative feedback from Rnd3 to ROCK-1 for the network present in Scheme 4 (Supplementary Note). Using the model and constants as in Supplementary Table 1, the convergence behaviour for initial values of Rnd3 and RhoA in the range 0 to 3.5 were examined, with all other initial concentrations set to 1 apart from NMIIA which was allowed to take values 0, 1, 2, 3, 4 for the simulations in (b); and for the Hill constant KRnd3ROCK1 which was allowed to take values 0.05, 0.10, 0.15, 0.20, 0.25 and 0.30, and with initial concentration of NMIIA = 1, and b7 = 0.15 for simulations in (c). The red circles show the stable point concentrations for Rnd3 and RhoA that all initialisations converge towards. Each point in the plane then corresponds to an initialisation for Rnd3 and RhoA that will converge to one of the stable circles. Each line then denotes the boundary between those points in the plane that will converge to either the RhoA low/Rnd3 high state of junctional content and the RhoA high/Rnd3 low state). (d) A model for the maintenance of a robust Rho zone at the epithelial Zonula Adherens.

Supplementary Figure 8 Bright field/DIC images corresponding to Figs 1a, f, 2h, 3a, 3b, 5h and Supplementary Figs 1c, e, 4e.

Supplementary Figure 9 Uncropped western blots.

Supplementary Table 1 Sequences of siRNA and primers used for the study.

Supplementary information

Supplementary Information

Supplementary Information (PDF 2428 kb)

Supplementary Note

Supplementary Information (PDF 19518 kb)

Supplementary Table 2

Statistical source data (XLSX 3890 kb)

GFP-AHPH localizes at the zonula adherens of epithelial cells.

Z-stacks of GFP-AHPH and RFP-UtrCH acquired by spinning disc confocal microscopy. (AVI 967 kb)

GFP-AHPH exhibits stability on the time scale of minutes.

Time-lapse imaging of GFP-APHPH (transfected in MCF-7 cells) acquired over a span of 30 min. (AVI 3294 kb)

ROCK-1 inhibition causes accumulation of p190B Rho GAP at the cell–cell junctions.

MCF-7 cells were transfected with GFP-p190B RhoGAP and time-lapsed imaging was performed briefly before and after addition of Y-27632 (30 μM) (20-min post-treatment). (AVI 3327 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Priya, R., Gomez, G., Budnar, S. et al. Feedback regulation through myosin II confers robustness on RhoA signalling at E-cadherin junctions. Nat Cell Biol 17, 1282–1293 (2015). https://doi.org/10.1038/ncb3239

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3239

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