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:

Adherens junction treadmilling during collective migration

Abstract

Collective cell migration is essential for both physiological and pathological processes. Adherens junctions (AJs) maintain the integrity of the migrating cell group and promote cell coordination while allowing cellular rearrangements. Here, we show that AJs undergo a continuous treadmilling along the lateral sides of adjacent leading cells. The treadmilling is driven by an actin-dependent rearward movement of AJs and is supported by the polarized recycling of N-cadherin. N-cadherin is mainly internalized at the cell rear and then recycled to the leading edge where it accumulates before being incorporated into forming AJs at the front of lateral cell–cell contacts. The polarized dynamics of AJs is controlled by a front-to-rear gradient of p120-catenin phosphorylation, which regulates polarized trafficking of N-cadherin. Perturbation of the GSK3-dependent phosphorylation of p120-catenin impacts on the stability of AJs, and the polarity and speed of leading cells during collective migration.

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: Continuous retrograde flow of N-cadherin-mediated adherens junctions (AJs) between adjacent migrating cells.
Figure 2: Coupling between the retrograde flow of AJs and the retrograde flow of transverse actin cables.
Figure 3: Leading-edge accumulation of N-cadherin–catenin complexes and formation of new AJs at the front of lateral cell–cell contacts depend on p120-ctn.
Figure 4: Polarized trafficking of N-cadherin from the cell rear to the leading edge.
Figure 5: Involvement of N-cadherin polarized recycling in AJ treadmilling.
Figure 6: Polarized regulation of p120-ctn association with N-cadherin.
Figure 7: GSK3-dependent regulation of p120-ctn phosphorylation and AJ dynamics during collective cell migration.
Figure 8: Role of p120-dependent AJ dynamics during collective migration.

Similar content being viewed by others

References

  1. Friedl, P. & Gilmour, D. Collective cell migration in morphogenesis, regeneration and cancer. Nat. Rev. Mol. Cell Biol. 10, 445–457 (2009).

    Article  CAS  PubMed  Google Scholar 

  2. Friedl, P., Locker, J., Sahai, E. & Segall, J. E. Classifying collective cancer cell invasion. Nat. Cell Biol. 14, 777–783 (2012).

    Article  PubMed  Google Scholar 

  3. Gnanaguru, G. et al. Laminins containing the β2 and γ3 chains regulate astrocyte migration and angiogenesis in the retina. Development 140, 2050–2060 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Chu, Y., Hughes, S. & Chan-Ling, T. Differentiation and migration of astrocyte precursor cells and astrocytes in human fetal retina: Relevance to optic nerve coloboma. FASEB J. 15, 2013–2015 (2001).

    Article  CAS  PubMed  Google Scholar 

  5. Fruttiger, M. Development of the mouse retinal vasculature: Angiogenesis versus vasculogenesis. Invest. Ophthalmol. Vis. Sci. 43, 522–527 (2002).

    PubMed  Google Scholar 

  6. Faber-Elman, A., Solomon, A., Abraham, J. A., Marikovsky, M. & Schwartz, M. Involvement of wound-associated factors in rat brain astrocyte migratory response to axonal injury: In vitro simulation. J. Clin. Invest. 97, 162–171 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Sofroniew, M. V. Molecular dissection of reactive astrogliosis and glial scar formation. Trends Neurosci. 32, 638–647 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Etienne-Manneville, S. Control of polarized cell morphology and motility by adherens junctions. Semin. Cell Dev. Biol. 22, 850–857 (2011).

    Article  CAS  PubMed  Google Scholar 

  9. Oda, H. & Takeichi, M. Evolution: Structural and functional diversity of cadherin at the adherens junction. J. Cell Biol. 193, 1137–1146 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Taguchi, K., Ishiuchi, T. & Takeichi, M. Mechanosensitive EPLIN-dependent remodeling of adherens junctions regulates epithelial reshaping. J. Cell Biol. 194, 643–656 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Twiss, F. et al. Vinculin-dependent Cadherin mechanosensing regulates efficient epithelial barrier formation. Biol. Open 1, 1128–1140 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Gloushankova, N. A. et al. Dynamics of contacts between lamellae of fibroblasts: Essential role of the actin cytoskeleton. Proc. Natl Acad. Sci. USA 95, 4362–4367 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Shaye, D. D., Casanova, J. & Llimargas, M. Modulation of intracellular trafficking regulates cell intercalation in the Drosophila trachea. Nat. Cell Biol. 10, 964–970 (2008).

    Article  CAS  PubMed  Google Scholar 

  14. Arboleda-Estudillo, Y. et al. Movement directionality in collective migration of germ layer progenitors. Curr. Biol. 20, 161–169 (2010).

    Article  CAS  PubMed  Google Scholar 

  15. Ewald, A. J., Brenot, A., Duong, M., Chan, B. S. & Werb, Z. Collective epithelial migration and cell rearrangements drive mammary branching morphogenesis. Dev. Cell 14, 570–581 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Theveneau, E. et al. Collective chemotaxis requires contact-dependent cell polarity. Dev. Cell 19, 39–53 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Ganz, A. et al. Traction forces exerted through N-cadherin contacts. Biol. Cell 98, 721–730 (2006).

    Article  CAS  PubMed  Google Scholar 

  18. Smutny, M. & Yap, A. S. Neighborly relations: Cadherins and mechanotransduction. J. Cell Biol. 189, 1075–1077 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Ladoux, B. et al. Strength dependence of cadherin-mediated adhesions. Biophys. J. 98, 534–542 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Harris, T. J. & Tepass, U. Adherens junctions: From molecules to morphogenesis. Nat. Rev. Mol. Cell Biol. 11, 502–514 (2010).

    Article  CAS  PubMed  Google Scholar 

  21. Etienne-Manneville, S. Adherens junctions during cell migration. Subcell. Biochem. 60, 225–249 (2012).

    Article  CAS  PubMed  Google Scholar 

  22. Takeichi, M. Cadherins: A molecular family important in selective cell–cell adhesion. Annu. Rev. Biochem. 59, 237–252 (1990).

    Article  CAS  PubMed  Google Scholar 

  23. Angst, B. D., Marcozzi, C. & Magee, A. I. The cadherin superfamily: Diversity in form and function. J. Cell Sci. 114, 629–641 (2001).

    CAS  PubMed  Google Scholar 

  24. Meng, W. & Takeichi, M. Adherens junction: Molecular architecture and regulation. Cold Spring Harb. Perspect. Biol. 1, a002899 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  25. le Duc, Q. et al. Vinculin potentiates E-cadherin mechanosensing and is recruited to actin-anchored sites within adherens junctions in a myosin II-dependent manner. J. Cell Biol. 189, 1107–1115 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Liu, Z. et al. Mechanical tugging force regulates the size of cell–cell junctions. Proc. Natl Acad. Sci. USA 107, 9944–9949 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Camand, E., Peglion, F., Osmani, N., Sanson, M. & Etienne-Manneville, S. N-cadherin expression level modulates integrin-mediated polarity and strongly impacts on the speed and directionality of glial cell migration. J. Cell Sci. 125, 844–857 (2012).

    Article  CAS  PubMed  Google Scholar 

  28. Dupin, I., Camand, E. & Etienne-Manneville, S. Classical cadherins control nucleus and centrosome position and cell polarity. J. Cell Biol. 185, 779–786 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Micalizzi, D. S., Farabaugh, S. M. & Ford, H. L. Epithelial-mesenchymal transition in cancer: Parallels between normal development and tumor progression. J. Mammary. Gland Biol. Neoplasia 15, 117–134 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  30. Wheelock, M. J., Shintani, Y., Maeda, M., Fukumoto, Y. & Johnson, K. R. Cadherin switching. J. Cell Sci. 121, 727–735 (2008).

    Article  CAS  PubMed  Google Scholar 

  31. Baum, B., Settleman, J. & Quinlan, M. P. Transitions between epithelial and mesenchymal states in development and disease. Semin. Cell Dev. Biol. 19, 294–308 (2008).

    Article  CAS  PubMed  Google Scholar 

  32. Etienne-Manneville, S. In vitro assay of primary astrocyte migration as a tool to study Rho GTPase function in cell polarization. Methods Enzymol. 406, 565–578 (2006).

    Article  CAS  PubMed  Google Scholar 

  33. Maruthamuthu, V., Aratyn-Schaus, Y. & Gardel, M. L. Conserved F-actin dynamics and force transmission at cell adhesions. Curr. Opin. Cell Biol. 22, 583–588 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Bard, L. et al. A molecular clutch between the actin flow and N-cadherin adhesions drives growth cone migration. J. Neurosci. 28, 5879–5890 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Sabatini, P. J., Zhang, M., Silverman-Gavrila, R. V. & Bendeck, M. P. Cadherins at cell-autonomous membrane contacts control macropinocytosis. J. Cell Sci. 124, 2013–2020 (2011).

    Article  CAS  PubMed  Google Scholar 

  36. Sharma, M. & Henderson, B. R. IQ-domain GTPase-activating protein 1 regulates beta-catenin at membrane ruffles and its role in macropinocytosis of N-cadherin and adenomatous polyposis coli. J. Biol. Chem. 282, 8545–8556 (2007).

    Article  CAS  PubMed  Google Scholar 

  37. Theisen, C. S., Wahl, J. K. 3rd, Johnson, K. R. & Wheelock, M. J. NHERF links the N-cadherin/catenin complex to the platelet-derived growth factor receptor to modulate the actin cytoskeleton and regulate cell motility. Mol. Biol. Cell 18, 1220–1232 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Davis, M. A., Ireton, R. C. & Reynolds, A. B. A core function for p120-catenin in cadherin turnover. J. Cell Biol. 163, 525–534 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Reynolds, A. B. & Carnahan, R. H. Regulation of cadherin stability and turnover by p120ctn: Implications in disease and cancer. Semin. Cell Dev. Biol. 15, 657–663 (2004).

    Article  CAS  PubMed  Google Scholar 

  40. Xiao, K. et al. p120-Catenin regulates clathrin-dependent endocytosis of VE-cadherin. Mol. Biol. Cell 16, 5141–5151 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Troyanovsky, R. B., Sokolov, E. P. & Troyanovsky, S. M. Endocytosis of cadherin from intracellular junctions is the driving force for cadherin adhesive dimer disassembly. Mol. Biol. Cell 17, 3484–3493 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Levayer, R., Pelissier-Monier, A. & Lecuit, T. Spatial regulation of Dia and Myosin-II by RhoGEF2 controls initiation of E-cadherin endocytosis during epithelial morphogenesis. Nat. Cell Biol. 13, 529–540 (2011).

    Article  CAS  PubMed  Google Scholar 

  43. Macia, E. et al. Dynasore, a cell-permeable inhibitor of dynamin. Dev. Cell 10, 839–850 (2006).

    Article  CAS  PubMed  Google Scholar 

  44. Fukumoto, Y., Shintani, Y., Reynolds, A. B., Johnson, K. R. & Wheelock, M. J. The regulatory or phosphorylation domain of p120 catenin controls E-cadherin dynamics at the plasma membrane. Exp. Cell Res. 314, 52–67 (2008).

    Article  CAS  PubMed  Google Scholar 

  45. Etienne-Manneville, S. & Hall, A. Cdc42 regulates GSK-3beta and adenomatous polyposis coli to control cell polarity. Nature 421, 753–756 (2003).

    Article  CAS  PubMed  Google Scholar 

  46. Etienne-Manneville, S. & Hall, A. Cdc42 regulates GSK3 and adenomatous polyposis coli (APC) to control cell polarity. Nature 421, 753–756 (2003).

    Article  CAS  PubMed  Google Scholar 

  47. Etienne-Manneville, S. & Hall, A. Integrin-mediated activation of Cdc42 controls cell polarity in migrating astrocytes through PKCzeta. Cell 106, 489–498 (2001).

    Article  CAS  PubMed  Google Scholar 

  48. Caswell, P. & Norman, J. Endocytic transport of integrins during cell migration and invasion. Trends Cell Biol. 18, 257–263 (2008).

    Article  CAS  PubMed  Google Scholar 

  49. Caswell, P. T., Vadrevu, S. & Norman, J. C. Integrins: Masters and slaves of endocytic transport. Nat. Rev. Mol. Cell Biol. 10, 843–853 (2009).

    Article  CAS  PubMed  Google Scholar 

  50. Kowalczyk, A. P. & Nanes, B. A. Adherens junction turnover: Regulating adhesion through cadherin endocytosis, degradation, and recycling. Subcell Biochem. 60, 197–222 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Ulrich, F. & Heisenberg, C. P. Probing E-cadherin endocytosis by morpholino-mediated Rab5 knockdown in zebrafish. Methods Mol. Biol. 440, 371–387 (2008).

    Article  CAS  PubMed  Google Scholar 

  52. Lohia, M., Qin, Y. & Macara, I. G. The Scribble polarity protein stabilizes E-cadherin/p120-catenin binding and blocks retrieval of E-cadherin to the Golgi. PLoS ONE 7, e51130 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Chiasson, C. M., Wittich, K. B., Vincent, P. A., Faundez, V. & Kowalczyk, A. P. p120-catenin inhibits VE-cadherin internalization through a Rho-independent mechanism. Mol. Biol. Cell 20, 1970–1980 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Hoshino, T. et al. Regulation of E-cadherin endocytosis by nectin through afadin, Rap1, and p120ctn. J. Biol. Chem. 280, 24095–24103 (2005).

    Article  CAS  PubMed  Google Scholar 

  55. Ishiyama, N. et al. Dynamic and static interactions between p120 catenin and E-cadherin regulate the stability of cell–cell adhesion. Cell 141, 117–128 (2010).

    Article  CAS  PubMed  Google Scholar 

  56. Nanes, B. A. et al. p120-catenin binding masks an endocytic signal conserved in classical cadherins. J. Cell Biol. 199, 365–380 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Ewald, A. J. et al. Mammary collective cell migration involves transient loss of epithelial features and individual cell migration within the epithelium. J. Cell Sci. 125, 2638–2654 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Kametani, Y. & Takeichi, M. Basal-to-apical cadherin flow at cell junctions. Nat. Cell Biol. 9, 92–98 (2007).

    Article  CAS  PubMed  Google Scholar 

  59. Dupin, I., Sakamoto, Y. & Etienne-Manneville, S. Cytoplasmic intermediate filaments mediate actin-driven positioning of the nucleus. J. Cell Sci. 124, 865–872 (2011).

    Article  CAS  PubMed  Google Scholar 

  60. Gomes, E. R., Jani, S. & Gundersen, G. G. Nuclear movement regulated by Cdc42, MRCK, myosin, and actin flow establishes MTOC polarization in migrating cells. Cell 121, 451–463 (2005).

    Article  CAS  PubMed  Google Scholar 

  61. Noren, N. K., Liu, B. P., Burridge, K. & Kreft, B. p120 catenin regulates the actin cytoskeleton via Rho family GTPases. J. Cell Biol. 150, 567–580 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Anastasiadis, P. Z. et al. Inhibition of RhoA by p120 catenin. Nat. Cell Biol. 2, 637–644 (2000).

    Article  CAS  PubMed  Google Scholar 

  63. Anastasiadis, P. Z. & Reynolds, A. B. The p120 catenin family: Complex roles in adhesion, signaling and cancer. J. Cell Sci. 113, 1319–1334 (2000).

    CAS  PubMed  Google Scholar 

  64. Charrasse, S., Meriane, M., Comunale, F., Blangy, A. & Gauthier-Rouviere, C. N-cadherin-dependent cell–cell contact regulates Rho GTPases and beta-catenin localization in mouse C2C12 myoblasts. J. Cell Biol. 158, 953–965 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Stairs, D. B. et al. Deletion of p120-catenin results in a tumor microenvironment with inflammation and cancer that establishes it as a tumor suppressor gene. Cancer Cell 19, 470–483 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Mary, S. et al. Biogenesis of N-cadherin-dependent cell–cell contacts in living fibroblasts is a microtubule-dependent kinesin-driven mechanism. Mol. Biol. Cell 13, 285–301 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Reynolds, A. B., Herbert, L., Cleveland, J. L., Berg, S. T. & Gaut, J. R. p120, a novel substrate of protein tyrosine kinase receptors and of p60v-src, is related to cadherin-binding factors beta-catenin, plakoglobin and armadillo. Oncogene 7, 2439–2445 (1992).

    CAS  PubMed  Google Scholar 

  68. Riedl, J. et al. Lifeact: A versatile marker to visualize F-actin. Nat. Methods 5, 605–607 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. McKinney, S. A., Murphy, C. S., Hazelwood, K. L., Davidson, M. W. & Looger, L. L. A bright and photostable photoconvertible fluorescent protein. Nat. Methods 6, 131–133 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Etienne-Manneville, S., Manneville, J., Nicholls, S., Ferenczi, M. A. & Hall, A. Cdc42 and Par6/PKCζ regulate the spatially localized association of Dlg1 and APC to control cell polarization. J. Cell Biol. 170, 895–901 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are particularly grateful to M. Piel (Institut Curie, France), A.B. Reynolds (Vanderbilt University, USA), A. Ridley (King’s College, UK), C. Gauthier-Rouvière (CRBM, France), B. Goud (Institut Curie, France), M. Cohen-Salmon (College de France, France), D. Riveline (IGBMC, France) and L. Looger (Janelia Farm, USA) for plasmids and reagents. We thank J-Y. Tinevez and E. Perret from the Plate-Forme d’Imagerie Dynamique/IMAGOPOLE, of Institut Pasteur, and J-B. Manneville for critical reading of the manuscript. F.P. is financially supported by the University Paris VI and VII, the Association pour la Recherche contre le Cancer and the Fondation pour la Recherche Medicale; and F.L. by the Institut National du Cancer. This work was supported by the Institut National du Cancer, l’Association pour la Recherche contre le Cancer, and La Ligue contre le Cancer. We also would like to thank D. Porquet and G. Porquet for contributing to the funding of this study.

Author information

Authors and Affiliations

Authors

Contributions

F.P. conceived and performed all experiments except those shown in Figs 1c, 2a, 6h and 8g and Supplementary Figs 1d, 2 and 5b, which were conceived and performed by F.L. S.E-M. contributed to the conception of the experiments, the interpretation of the data and wrote the manuscript.

Corresponding author

Correspondence to Sandrine Etienne-Manneville.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 N-cadherin-GFP forms cadherin/catenins complexes with the endogenous proteins.

Fluorescence images showing the immunostaining of endogenous N-cadherin (a), anti-p120-ctn (b), anti-beta-catenin (c) in N-cadherin-GFP (Ncad-GFP) expressing astrocytes. (a) Note the colocalization of Ncad-GFP with the endogenous adherens junction proteins both at cell-cell contacts (arrows) and at the leading edge (arrowheads). (d) Immunostaining of endogenous N-cadherin (left) together with a membrane dye DiI (right) of migrating astrocytes 8 h after wounding. Higher magnification images of the leading edge (boxed area) are shown in the respective coloured rectangles on the right. The graph shows the fluorescence intensity profiles in both fluorescence channels along the two coloured lines. White arrows: direction of migration. Scale bar, 10 μm.

Supplementary Figure 2 Cell polarity in first and second row cells migrating in a 2D wound-healing assay.

Histogram showing the percentage of cells with a correctly oriented centrosome in the first and second row near the wound, 8 h after wounding. Results are shown as mean ± s.e.m. of 3 independent experiments in each conditions (first row n = 440 cells, second row n = 307).

Supplementary Figure 3 p120 depletion does not alter N-cadherin-mediated cell-cell contacts in confluent non migrating astrocytes.

(a) Westernblot (WB) showing p120-ctn and tubulin expression levels in astrocytes nucleofected with control siRNA (si ctl) or siRNA specific for p120-ctn (si p120#1, si p120#2). (b) Histogram showing an average 54% and 79% decrease in p120 expression respectively for si p120#1 and si p120#2 expressing cells, compared to control. Data are mean ± s.e.m. of normalized protein expression analysed in n = 4 independent experiments using the LI-COR technology. P < 0.001, unpaired Student t-test. (c) Immunofluorescence images showing N-cadherin (green) and Hoechst (nuclei, magenta) staining of confluent astrocytes 4 days after nucleofection with si ctl and si p120-ctn (si p120#1, si p120#2). The yellow arrows highlight the similar organization of AJs in si CTL and si p120-ctn cells. (d) Primary astrocytes were nucleofected with the indicated siRNA and submitted, 3 days later, to a wound healing assay. The graph shows the percentage of migrating cells, in which the centrosome is located in the quadrant facing the wound in front of the nucleus, as an indication of cell polarization. Data are mean ± s.e.m. of more than 150 cells from n = 3 independent experiments. P < 0.05,P < 0.01, unpaired Students t test. Scale bar, 10 μm.

Supplementary Figure 4 Role of protein synthesis and membrane traffic in N-cadherin dynamics.

(a) Immunofluorescence images showing N-cadherin immunostaining in DMSO- or cycloheximide-treated astrocytes 8 h after wounding. Yellow arrowheads point to sites of N-cadherin accumulation at the cell leading edge. The histogram on the right shows, for both conditions, the percentage of cells with an N-cadherin accumulation at the leading edge. Data are given as mean ± s.e.m. of more than 100 cells from n = 3 independent experiments. The difference in N-cadherin accumulation is not statistically significant: ns: P = 0.065, unpaired Student t-test. (b) Immunostaining of endogenous N-cadherin in DMSO- or Nocodazole-treated astrocytes, and GFP or Rab5S34N-myc (Rab5-DN) transfected cells, after 8 h of migration. The images and the corresponding analytical graphs showing the intensity at the leading edges in each representative cells refer to the data given by the graph in the main Fig. 5b. (c) Immunofluorescence images (from time-lapse acquisition) of dynasore (80 μM)-treated confluent astrocytes. Scale bar, 10 μm.

Supplementary Figure 5 p120-ctn tyrosine phosphorylation is not changed during collective cell migration.

(a) Cell lysates were obtained at the indicated time after wounding and were analysed by westernblotting (WB) using antibodies specifically recognizing phosphorylated Tyr96 or Tyr228 of p120-ctn and p120-ctn independently of its phosphorylation state (p120 total). (b) Immunofluorescence images showing N-cadherin and phospho T310-p120-ctn in migrating astrocytes nucleofected with the indicated siRNA or p120-ctn construct. Scale bar 10 μm.

Supplementary Figure 6 GSK3 depletion by siRNA.

Primary astrocytes were nucleofected with the indicated siRNAs. Three days later, cells were lyzed and analysed by westernblotting using anti-GSK3 and anti-tubulin. The quantitative analysis, using the LI-COR technology, of GSK3 expression level compared to tubulin is shown under the westernblot.

Supplementary Figure 7 Table summarizing the effects of p120-ctn depletion, GSK3 inhibition and inhibition of GSK3-dependent phosphorylation of p120-ctn on AJs maintenance and dynamics during collective cell migration.

The + or− signs in the table represent the level of the evaluated biological process (+++ being the highest level of activity and −−−, the lowest).

Supplementary Figure 8 Original Western blots referring to main figures 6d, g, 7a and Supplementary Figs 3.a, 5.a and 6.

Red dotted-line boxes highlight the regions of the Western Blots shown in the figures. All Western blots were named according to the main figure to which they relate to. The blot marked by a black asterisk was used for the main figure 6d and the Supplementary Fig. 5a since the kinetics of p120-ctn phosphorylation on T130, Y228, Y96 during cell migration was assessed during the same experiments, using the same cell lysates.

Supplementary information

Supplementary Information

Supplementary Information (PDF 2643 kb)

AJs dynamics in confluent and migrating astrocytes (2D).

Fluorescence time-lapse acquisition of N-cadherin-GFP expressing astrocytes in a confluent monolayer (a, left) or migrating in a wound healing assay (4 h after wounding) (b, right). Note the absence of apparent lateral AJs movement in confluent cells, as opposed to migrating cells in which a treadmilling of AJs is observed. 1 frame/3 min for 2 h (a, left) and 1 frame/5 min for 3h20 (b, right). Scale bar, 10 μm. (AVI 4738 kb)

AJs dynamics migrating fibroblasts and endothelial cells (2D).

Phase contrast (left) and fluorescence (right) time-lapse acquisition of N-cadherin-GFP expressing NIH3T3 fibroblasts (higher panels) and VE-cadherin-GFP expressing EA.hy926 endothelial cells (lower panels) migrating in a wound healing assay (2 h after wounding) 1 frame/3 min for 2 h (a, left) and 1 frame/5 min for 3h20 Scale bar, 10 μm (AVI 1146 kb)

AJs dynamics in astrocytes migrating in a 3D matrix.

Fluorescence time-lapse acquisition of migrating astrocytes transfected with N-cadherin-GFP (1 frame/5 min). N-cadherin-GFP expressing astrocytes seeded within a thick layer of matrigel (3D), migrating out of a coherent group of cells (white arrows). Note the AJs retrograde flow (coloured arrowheads) in collectively migrating astrocytes. Scale bar, 50 μm. (AVI 4055 kb)

AJs dynamics in the first and second row of cells (2D).

Fluorescence time-lapse acquisition of N-cadherin-GFP expressing astrocytes migrating in a wound healing assay (4 h after wounding). Note the absence of AJs movement in the cell in the second row, compared to wound edge cells in which a treadmilling of AJs is observed. Scale bar, 10 μm. (AVI 3779 kb)

AJs and F-actin dynamics in polarized immobile astrocytes.

Fluorescence time-lapse acquisition of astrocytes transfected with N-cadherin-GFP (green) and Lifeact-Cherry (red) (1 frame/5 min). Astrocytes were plated on circular micropatterns to allow polarization in the absence of migration. Note the similar retrograde flow of transverse actin cables and AJs. Scale bar, 10 μm. (AVI 60100 kb)

N-cadherin molecules undergo an actual retrograde movement on the lateral junctions of collectively migrating cells.

Fluorescence time-lapse acquisition of migrating N-cadherin–EOS-expressing astrocytes. Photoconverted (red) N-cadherin–EOS at the front of the lateral junction (Fig. 1e) undergo a retrograde movement, opposite to the direction of migration (white arrow). 1 frame/5 min. Scale bar, 2 μm. (AVI 2433 kb)

AJs and F-actin dynamics in 2D migrating astrocytes.

Fluorescence time-lapse acquisition of migrating astrocytes transfected with N-cadherin-GFP and Lifeact-Cherry (1 frame/5 min). Note that AJs and transverse actin filaments undergo a similar retrograde flow, opposite to the direction of migration (white arrow). Scale bar, 10 μm. (AVI 15139 kb)

Leading edge N-cadherin moves to the side of the cell and participates in the formation of new AJs.

Fluorescence time-lapse acquisition of migrating astrocytes transfected with N-cadherin-GFP (1 frame/5 min) focusing on the front region of the cell. Note the lateral movement of cadherin clusters (colored arrows) participating in the formation of new AJs at the front of lateral intercellular contacts. Scale bar, 5 μm. (AVI 1133 kb)

p120-ctn depletion affects N-cadherin dynamics and formation of new AJs at the front side of migrating cells.

Phase contrast (left) and fluorescence (right) time-lapse acquisitions of migrating p120-ctn-depleted astrocytes expressing N-cadherin-GFP (1 frame/10 min). The acquisition starts a few minutes after wounding and shows that, in contrast to the continuous treadmilling of AJs observed in control cells (movie 1), new AJs formation at the front of lateral cell-cell contacts is altered while the retrograde flow of preexisting AJs is initiated (white arrowheads). Scale bar, 10 μm. (AVI 6429 kb)

Polarized N-cadherin vesicular trafficking from the rear to the front of migrating astrocytes.

Fluorescence time-lapse acquisition of N-cadherin-GFP expressing migrating astrocytes 8 h after wounding (1 frame/10 sec, inverted contrast) reveals trafficking of N-cadherin vesicles. Images were obtained in a median focal plane (that is situated approximately in the middle of the cell height) and thus do not clearly show the rear (which is located higher) and the front (which is located lower) of lateral AJs. Note the active trafficking of N-cadherin positive vesicles preferentially moving from the rear to the front of the cell. Scale bar, 10 μm. (AVI 12358 kb)

Altered membrane trafficking affects N-cadherin polarized recruitment to the leading edge and the formation of new AJs at the front side of migrating cells.

Fluorescence time-lapse acquisition of migrating N-cadherin-GFP expressing astrocytes (1 frame/5 min). Migration was initiated in DMSO containing cell culture medium. The dynamin inhibitor Dynasore was added after 170 min of time-lapse acquisition. The addition of new medium leads to a change of contrast in the movie. Scale bar, 7 μm. (AVI 7407 kb)

p120-ctn depletion increases astrocyte migration in 3D matrix.

Phase contrast time-lapse acquisitions of control(left) and p120-ctn-depleted (right) astrocytes seeded in a 3D matrigel. 1 frame/15 min for 6 h. Scale bar, 10 μm. (AVI 1374 kb)

p120-ctn reduces collective cell migration.

Phase contrast (left) and fluorescence (right) time-lapse acquisitions of migrating p120-ctn-depleted astrocytes expressing siRNA-resistant WT-p120-ctn-GFP (1 frame/20 min). The acquisition starts 6 h after wounding. Note that wound-edge p120-ctn-depleted cells, but not the cell expressing WT-p120-ctn-GFP, tend to separate from the rest of the monolayer. (AVI 2924 kb)

Inhibition of p120-ctn phosphorylation perturbs AJ dynamics and inhibits cell migration.

Phase contrast (left) and fluorescence (right) time-lapse acquisitions of migrating p120-ctn-depleted astrocytes expressing siRNA-resistant T310A-p120-ctn-GFP (1 frame/20 min). The acquisition starts 6 h after wounding. Note that wound-edge p120-ctn-depleted cells, but not the cell expressing T310-p120-ctn-GFP, tend to separate from the rest of the monolayer. In contrast to WT-p120-ctn-expressing cells (Supplementary Video 13), T310-p120-ctn-GFP expressing cells have a broader and less polarized leading edge, a sign of altered AJ formation at the front of lateral cell-cell contacts (Fig. 7e, g and Supplementary Videos 9 and 14). (AVI 2449 kb)

Gap junction dynamics in migrating astrocytes (2D).

Fluorescence time-lapse acquisition of Connexin43-GFP expressing astrocytes migrating in a wound healing assay (4 h after wounding). Note the absence of apparent retrograde movement of connexin-mediated gap junctions along the lateral sides of migrating astrocytes. 1 frame/3 min for 2 h Scale bar, 10 μm. (AVI 850 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Peglion, F., Llense, F. & Etienne-Manneville, S. Adherens junction treadmilling during collective migration. Nat Cell Biol 16, 639–651 (2014). https://doi.org/10.1038/ncb2985

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb2985

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