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:

Forces generated by lamellipodial actin filament elongation regulate the WAVE complex during cell migration

Abstract

Actin filaments generate mechanical forces that drive membrane movements during trafficking, endocytosis and cell migration. Reciprocally, adaptations of actin networks to forces regulate their assembly and architecture. Yet, a demonstration of forces acting on actin regulators at actin assembly sites in cells is missing. Here we show that local forces arising from actin filament elongation mechanically control WAVE regulatory complex (WRC) dynamics and function, that is, Arp2/3 complex activation in the lamellipodium. Single-protein tracking revealed WRC lateral movements along the lamellipodium tip, driven by elongation of actin filaments and correlating with WRC turnover. The use of optical tweezers to mechanically manipulate functional WRC showed that piconewton forces, as generated by single-filament elongation, dissociated WRC from the lamellipodium tip. WRC activation correlated with its trapping, dwell time and the binding strength at the lamellipodium tip. WRC crosslinking, hindering its mechanical dissociation, increased WRC dwell time and Arp2/3-dependent membrane protrusion. Thus, forces generated by individual actin filaments on their regulators can mechanically tune their turnover and hence activity during cell 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

Fig. 1: WRC diffusion trapping at the lamellipodium tip correlates with its activation state.
Fig. 2: WRC displays lateral movements along the lamellipodium tip.
Fig. 3: WRC lateral movements are driven by actin filaments elongation and correlate with fast turnover.
Fig. 4: Mechanical forces exerted by physical contact with elongating actin filaments drive FMNL2 lateral movements.
Fig. 5: Characterization of a functional transmembrane WRC enabling optical tweezers in situ manipulation.
Fig. 6: WRC activation correlates with its mechanical connections to the lamellipodium tip.
Fig. 7: Positive correlation between WRC increased dwell time at the lamellipodium tip and rates of membrane protrusion.

Similar content being viewed by others

Data availability

All data supporting the findings of this study are available from the corresponding author on reasonable request. Source data are provided with this paper.

References

  1. Krause, M. & Gautreau, A. Steering cell migration: lamellipodium dynamics and the regulation of directional persistence. Nat. Rev. Mol. Cell Biol. 15, 577–590 (2014).

    Article  CAS  PubMed  Google Scholar 

  2. Schaks, M., Giannone, G. & Rottner, K. Actin dynamics in cell migration. Essays Biochem. 63, 483–495 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Block, J. et al. FMNL2 drives actin-based protrusion and migration downstream of Cdc42. Curr. Biol. 22, 1005–1012 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Dang, I. et al. Inhibitory signalling to the Arp2/3 complex steers cell migration. Nature 503, 281–284 (2013).

    Article  CAS  PubMed  Google Scholar 

  5. Mehidi, A. et al. Transient activations of Rac1 at the lamellipodium tip trigger membrane protrusion. Curr. Biol. 29, 2852–2866 (2019).

    Article  CAS  PubMed  Google Scholar 

  6. Fort, L. et al. Fam49/CYRI interacts with Rac1 and locally suppresses protrusions. Nat. Cell Biol. 20, 1159–1171 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Schaks, M. et al. Distinct interaction sites of Rac GTPase with WAVE regulatory complex have non-redundant functions in vivo. Curr. Biol. 28, 3674–3684 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Chen, Z. et al. Structure and control of the actin regulatory WAVE complex. Nature 468, 533–538 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Lebensohn, A. M. & Kirschner, M. W. Activation of the WAVE complex by coincident signals controls actin assembly. Mol. Cell 36, 512–524 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Steffen, A. et al. Sra-1 and Nap1 link Rac to actin assembly driving lamellipodia formation. EMBO J. 23, 749–759 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Innocenti, M. et al. Abi1 is essential for the formation and activation of a WAVE2 signalling complex. Nat. Cell Biol. 6, 319–327 (2004).

    Article  CAS  PubMed  Google Scholar 

  12. Suetsugu, S. et al. Optimization of WAVE2 complex-induced actin polymerization by membrane-bound IRSp53, PIP3 and Rac. J. Cell Biol. 173, 571–585 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Svitkina, T. M. et al. Mechanism of filopodia initiation by reorganization of a dendritic network. J. Cell Biol. 160, 409–421 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Kage, F. et al. FMNL formins boost lamellipodial force generation. Nat. Commun. 8, 14832 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Mejillano, M. R. et al. Lamellipodial versus filopodial mode of the actin nanomachinery: pivotal role of the filament barbed end. Cell 118, 363–373 (2004).

    Article  CAS  PubMed  Google Scholar 

  16. Ridley, A. J., Paterson, H. F., Johnston, C. L., Diekmann, D. & Hall, A. The small GTP-binding protein rac regulates growth factor-induced membrane ruffling. Cell 70, 401–410 (1992).

    Article  CAS  PubMed  Google Scholar 

  17. Wu, Y. I. et al. A genetically encoded photoactivatable Rac controls the motility of living cells. Nature 461, 104–108 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Paszek, M. J. et al. The cancer glycocalyx mechanically primes integrin-mediated growth and survival. Nature 511, 319–325 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Sinha, B. et al. Cells respond to mechanical stress by rapid disassembly of caveolae. Cell 144, 402–413 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Iskratsch, T., Wolfenson, H. & Sheetz, M. P. Appreciating force and shape—the rise of mechanotransduction in cell biology. Nat. Rev. Mol. Cell Biol. 15, 825–833 (2014).

    Article  CAS  PubMed  Google Scholar 

  21. Massou, S. et al. Cell stretching is amplified by active actin remodelling to deform and recruit proteins in mechanosensitive structures. Nat. Cell Biol. 22, 1011–1023 (2020).

    Article  CAS  PubMed  Google Scholar 

  22. Friedland, J. C., Lee, M. H. & Boettiger, D. Mechanically activated integrin switch controls α5β1 function. Science 323, 642–644 (2009).

    Article  CAS  PubMed  Google Scholar 

  23. Giannone, G., Jiang, G., Sutton, D. H., Critchley, D. R. & Sheetz, M. P. Talin1 is critical for force-dependent reinforcement of initial integrin-cytoskeleton bonds but not tyrosine kinase activation. J. Cell Biol. 163, 409–419 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Jiang, G., Giannone, G., Critchley, D. R., Fukumoto, E. & Sheetz, M. P. Two-piconewton slip bond between fibronectin and the cytoskeleton depends on talin. Nature 424, 334–337 (2003).

    Article  CAS  PubMed  Google Scholar 

  25. Wang, Y. et al. Visualizing the mechanical activation of Src. Nature 434, 1040–1045 (2005).

    Article  CAS  PubMed  Google Scholar 

  26. Coste, B. et al. Piezo proteins are pore-forming subunits of mechanically activated channels. Nature 483, 176–181 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Sawada, Y. et al. Force sensing by mechanical extension of the Src family kinase substrate p130Cas. Cell 127, 1015–1026 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. del Rio, A. et al. Stretching single talin rod molecules activates vinculin binding. Science 323, 638–641 (2009).

    Article  PubMed  Google Scholar 

  29. Hirata, H., Tatsumi, H. & Sokabe, M. Mechanical forces facilitate actin polymerization at focal adhesions in a zyxin-dependent manner. J. Cell Sci. 121, 2795–2804 (2008).

    Article  CAS  PubMed  Google Scholar 

  30. Ladoux, B. & Mège, R. M. Mechanobiology of collective cell behaviours. Nat. Rev. Mol. Cell Biol. 18, 743–757 (2017).

    Article  CAS  PubMed  Google Scholar 

  31. Zimmermann, D. et al. Mechanoregulated inhibition of formin facilitates contractile actomyosin ring assembly. Nat. Commun. 8, 703 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  32. Jégou, A., Carlier, M.-F. & Romet-Lemonne, G. Formin mDia1 senses and generates mechanical forces on actin filaments. Nat. Commun. 4, 1883 (2013).

    Article  PubMed  Google Scholar 

  33. Courtemanche, N., Lee, J. Y., Pollard, T. D. & Greene, E. C. Tension modulates actin filament polymerization mediated by formin and profilin. Proc. Natl Acad. Sci. USA 110, 9752–9757 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Wioland, H., Jegou, A. & Romet-Lemonne, G. Torsional stress generated by ADF/cofilin on cross-linked actin filaments boosts their severing. Proc. Natl Acad. Sci. USA 116, 2595–2602 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Chaudhuri, O., Parekh, S. H. & Fletcher, D. A. Reversible stress softening of actin networks. Nature 445, 295–298 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Bieling, P. et al. Force feedback controls motor activity and mechanical properties of self-assembling branched actin networks. Cell 164, 115–127 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Parekh, S. H., Chaudhuri, O., Theriot, J. A. & Fletcher, D. A. Loading history determines the velocity of actin-network growth. Nat. Cell Biol. 7, 1219–1223 (2005).

    Article  PubMed  Google Scholar 

  38. Giannone, G. et al. Lamellipodial actin mechanically links myosin activity with adhesion-site formation. Cell 128, 561–575 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Giannone, G. et al. Periodic lamellipodial contractions correlate with rearward actin waves. Cell 116, 431–443 (2004).

    Article  CAS  PubMed  Google Scholar 

  40. Houk, A. R. et al. Membrane tension maintains cell polarity by confining signals to the leading edge during neutrophil migration. Cell 148, 175–188 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Mueller, J. et al. Load adaptation of lamellipodial actin networks. Cell 171, 188–200 (2017).

    Article  CAS  PubMed  Google Scholar 

  42. Kovar, D. R. & Pollard, T. D. Insertional assembly of actin filament barbed ends in association with formins produces piconewton forces. Proc. Natl Acad. Sci. USA 101, 14725–14730 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Footer, M. J., Kerssemakers, J. W. J., Theriot, J. A. & Dogterom, M. Direct measurement of force generation by actin filament polymerization using an optical trap. Proc. Natl Acad. Sci. USA 104, 2181–2186 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Rossier, O. et al. Integrins β1 and β3 exhibit distinct dynamic nanoscale organizations inside focal adhesions. Nat. Cell Biol. 14, 1057–1067 (2012).

    Article  CAS  PubMed  Google Scholar 

  45. Chazeau, A. et al. Nanoscale segregation of actin nucleation and elongation factors determines dendritic spine protrusion. EMBO J. 33, 2745–2764 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Manley, S. et al. High-density mapping of single-molecule trajectories with photoactivated localization microscopy. Nat. Methods 5, 155–157 (2008).

    Article  CAS  PubMed  Google Scholar 

  47. Gautreau, A. et al. Purification and architecture of the ubiquitous Wave complex. Proc. Natl Acad. Sci. USA 101, 4379–4383 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Millius, A., Watanabe, N. & Weiner, O. D. Diffusion, capture and recycling of SCAR/WAVE and Arp2/3 complexes observed in cells by single-molecule imaging. J. Cell Sci. 125, 1165–1176 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Chen, B. et al. Rac1 GTPase activates the WAVE regulatory complex through two distinct binding sites. eLife 6, e29795 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Weisswange, I., Newsome, T. P., Schleich, S. & Way, M. The rate of N-WASP exchange limits the extent of ARP2/3-complex-dependent actin-based motility. Nature 458, 87–91 (2009).

    Article  CAS  PubMed  Google Scholar 

  51. Case, L. B., Zhang, X., Ditlev, J. A. & Rosen, M. K. Stoichiometry controls activity of phase-separated clusters of actin signaling proteins. Science 363, 1093–1097 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Weiner, O. D., Marganski, W. A., Wu, L. F., Altschuler, S. J. & Kirschner, M. W. An actin-based wave generator organizes cell motility. PLoS Biol. 5, 2053–2063 (2007).

    Article  CAS  Google Scholar 

  53. Lai, F. P. L. et al. Arp2/3 complex interactions and actin network turnover in lamellipodia. EMBO J. 27, 982–992 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Smith, B. A. et al. Three-color single molecule imaging shows WASP detachment from Arp2/3 complex triggers actin filament branch formation. eLife 2, e01008 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Svitkina, T. M. & Borisy, G. G. Arp2/3 complex and actin depolymerizing factor/cofilin in dendritic organization and treadmilling of actin filament array in lamellipodia. J. Cell Biol. 145, 1009–1026 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Forscher, P. & Smith, S. J. Actions of cytochalasins on the organization of actin filaments and microtubules in a neuronal growth cone. J. Cell Biol. 107, 1505–1516 (1988).

    Article  CAS  PubMed  Google Scholar 

  57. Co, C., Wong, D. T., Gierke, S., Chang, V. & Taunton, J. Mechanism of actin network attachment to moving membranes: barbed end capture by N-WASP WH2 domains. Cell 128, 901–913 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Bieling, P. et al. WH2 and proline-rich domains of WASP‐family proteins collaborate to accelerate actin filament elongation. EMBO J. 37, 102–121 (2018).

    Article  CAS  PubMed  Google Scholar 

  59. Dmitrieff, S. & Nédélec, F. Amplification of actin polymerization forces. J. Cell Biol. 212, 763–766 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Koestler, S. A., Auinger, S., Vinzenz, M., Rottner, K. & Small, J. V. Differentially oriented populations of actin filaments generated in lamellipodia collaborate in pushing and pausing at the cell front. Nat. Cell Biol. 10, 306–313 (2008).

    Article  CAS  PubMed  Google Scholar 

  61. Dolati, S. et al. On the relation between filament density, force generation and protrusion rate in mesenchymal cell motility. Mol. Biol. Cell 29, 2674–2686 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Higashida, C. et al. Actin polymerization-driven molecular movement of mDia1 in living cells. Science 303, 2007–2010 (2004).

    Article  CAS  PubMed  Google Scholar 

  63. Harris, E. S., Gauvin, T. J., Heimsath, E. G. & Higgs, H. N. Assembly of filopodia by the formin FRL2 (FMNL3). Cytoskeleton 67, 755–772 (2010).

    Article  CAS  PubMed  Google Scholar 

  64. Thompson, M. E., Heimsath, E. G., Gauvin, T. J., Higgs, H. N. & Jon Kull, F. FMNL3 FH2-actin structure gives insight into formin-mediated actin nucleation and elongation. Nat. Struct. Mol. Biol. 20, 111–118 (2013).

    Article  CAS  PubMed  Google Scholar 

  65. Fazal, F. M. & Block, S. M. Optical tweezers study life under tension. Nat. Photon. 5, 318–321 (2011).

    Article  CAS  Google Scholar 

  66. Huang, D. L., Bax, N. A., Buckley, C. D., Weis, W. I. & Dunn, A. R. Vinculin forms a directionally asymmetric catch bond with F-actin. Science 357, 703–706 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Leduc, C. et al. A highly specific gold nanoprobe for live-cell single-molecule imaging. Nano Lett. 13, 1489–1494 (2013).

    Article  CAS  PubMed  Google Scholar 

  68. Siton, O. et al. Cortactin releases the brakes in actin-based motility by enhancing WASP-VCA detachment from Arp2/3 branches. Curr. Biol. 21, 2092–2097 (2011).

    Article  CAS  PubMed  Google Scholar 

  69. Berro, J., Michelot, A., Blanchoin, L., Kovar, D. R. & Martiel, J. L. Attachment conditions control actin filament buckling and the production of forces. Biophys. J. 92, 2546–2558 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  70. Kaksonen, M., Toret, C. P. & Drubin, D. G. Harnessing actin dynamics for clathrin-mediated endocytosis. Nat. Rev. Mol. Cell Biol. 7, 404–414 (2006).

    Article  CAS  PubMed  Google Scholar 

  71. Derivery, E. et al. The Arp2/3 activator WASH controls the fission of endosomes through a large multiprotein complex. Dev. Cell 17, 712–723 (2009).

    Article  CAS  PubMed  Google Scholar 

  72. Welch, M. D. & Way, M. Arp2/3-mediated actin-based motility: a tail of pathogen abuse. Cell Host Microbe 14, 242–255 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Law, A. L. et al. Lamellipodin and the Scar/WAVE complex cooperate to promote cell migration in vivo. J. Cell Biol. 203, 673–689 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  74. Chen, X. J. et al. Ena/VASP proteins cooperate with the WAVE complex to regulate the actin cytoskeleton. Dev. Cell 30, 569–584 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  75. Abou-Kheir, W., Isaac, B., Yamaguchi, H. & Cox, D. Membrane targeting of WAVE2 is not sufficient for WAVE2-dependent actin polymerization: a role for IRSp53 in mediating the interaction between Rac and WAVE2. J. Cell Sci. 121, 379–390 (2008).

    Article  CAS  PubMed  Google Scholar 

  76. Chen, B. et al. The WAVE regulatory complex links diverse receptors to the actin cytoskeleton. Cell 156, 195–207 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Pipathsouk, A. et al. The WAVE complex associates with sites of saddle membrane curvature. J. Cell Biol. 220, e202003086 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Gautier, J. J. et al. Clathrin is required for Scar/Wave-mediated lamellipodium formation. J. Cell Sci. 124, 3414–3427 (2011).

    Article  CAS  PubMed  Google Scholar 

  79. Chia, P. H., Chen, B., Li, P., Rosen, M. K. & Shen, K. Local F-actin network links synapse formation and axon branching. Cell 156, 208–220 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Risca, V. I. et al. Actin filament curvature biases branching direction. Proc. Natl Acad. Sci. USA 109, 2913–2918 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. Law, A.-L. et al. Nance-Horan syndrome-like 1 protein negatively regulates Scar/WAVE-Arp2/3 activity and inhibits lamellipodia stability and cell migration. Nat. Commun. 12, 5687 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Su, J., Muranjan, M. & Sap, J. Receptor protein tyrosine phosphatase alpha activates Src-family kinases and controls integrin-mediated responses in fibroblasts. Curr. Biol. 9, 505–511 (1999).

    Article  CAS  PubMed  Google Scholar 

  83. Kage, F. et al. FMNL2 and -3 regulate Golgi architecture and anterograde transport downstream of Cdc42. Sci Rep. 7 (2017).

  84. Derivery, E. et al. Free Brick1 is a trimeric precursor in the assembly of a functional wave complex. PLoS ONE 3, e2462 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  85. Racine, V. et al. Multiple-target tracking of 3D fluorescent objects based on simulated annealing. In Proc. 3rd IEEE International Symposium on Biomedical Imaging 1–3, 1020–1023 (IEEE, 2006).

  86. Racine, V. et al. Visualization and quantification of vesicle trafficking on a three-dimensional cytoskeleton network in living cells. J. Microsc. 225, 214–228 (2007).

    Article  PubMed  Google Scholar 

  87. Izeddin, I. et al. Wavelet analysis for single molecule localization microscopy. Opt. Express 20, 2081–2095 (2012).

    Article  CAS  PubMed  Google Scholar 

  88. Kechkar, A., Nair, D., Heilemann, M., Choquet, D. & Sibarita, J.-B. Real-time analysis and visualization for single-molecule based super-resolution microscopy. PLoS ONE 8, e62918 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  89. Cheezum, M. K., Walker, W. F. & Guilford, W. H. Quantitative comparison of algorithms for tracking single fluorescent particles. Biophys. J. 81, 2378–2388 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  90. Gura Sadovsky, R., Brielle, S., Kaganovich, D. & England, J. L. Measurement of rapid protein diffusion in the cytoplasm by photo-converted intensity profile expansion. Cell Rep. 18, 2795–2806 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Orré, T., Mehidi, A., Massou, S., Rossier, O. & Giannone, G. Using single-protein tracking to study cell migration. Methods Mol. Biol. 1749, 291–311 (2018).

    Article  PubMed  Google Scholar 

  92. Orré, T. et al. Molecular motion and tridimensional nanoscale localization of kindlin control integrin activation in focal adhesions. Nat. Commun 12, 3104 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  93. Radhakrishnan, A. V. et al. in Methods in Molecular Biology Vol. 2217, 85–113 (Humana Press, 2021).

  94. Cao, L. et al. SPIN90 associates with mDia1 and the Arp2/3 complex to regulate cortical actin organization. Nat. Cell Biol. 22, 803–814 (2020).

    Article  CAS  PubMed  Google Scholar 

  95. Chamma, I. et al. Mapping the dynamics and nanoscale organization of synaptic adhesion proteins using monomeric streptavidin. Nat. Commun. 7, 10773 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank B. Tessier, R. Sterling and J. Carrere for technical assistance, M. Garcia, M. Lagardère and T. Orré for helpful discussions, C. Poujol and S. Marais (Bordeaux Imaging Center, BIC) for technical help and F. Cordelières (BIC) for support with kymograph analysis (Kymo Tool Box). We thank E. Verdier and N. Retailleau for technical help (IINS Cell Biology Facility, grant no. ANR-10-LABX-43). We would also like to thank J. B. Sibarita (IINS) for his support with sptPALM analysis and B. Wehrle-Haller (University of Geneva) and T. Stradal (Helmholtz Centre for Infection Research) for providing GFP-K80-β3-integrin expression plasmid and anti-Nap1 antibodies, respectively. J. Lakins (Center for Bioengineering and Tissue Regeneration, UCSF) is gratefully acknowledged for his help in using the PiggyBac system as well as for providing the Piggybac inducible and Hyperactive transposase plasmids. We acknowledge financial support from the French Ministry of Research and CNRS, ANR grant Integractome (G.G.), ANR grant FastNano (G.G.), Ligue Contre le Cancer (A.M.), Conseil Régional Aquitaine (A.M.), Fondation pour la Recherche Médicale (G.G. and A.M.) and the Deutsche Forschungsgemeinschaft (K.R.). We also acknowledge France-BioImaging infrastructure supported by the French National Research Agency (ANR-10-INBS-04).

Author information

Authors and Affiliations

Authors

Contributions

G.G. conceived and coordinated the project. G.G, A.M., K.R. and O.R. conceptualized the experiments. A.M. performed and analysed the sptPALM and laser trap experiments. O.R. performed pilot sptPALM experiments. F.K. designed and generated Nap1-KO cells and performed rescue experiments. M. Schaks carried out rescue experiments in Sra-1/PIR121 double-KO cells. M. Sainlos generated biotinylated nanobody. Z.K. and A.G. designed and generated protein constructs. Z.K. designed and generated TM-Nap1 and TM-Sra1 constructs and stable TM-Nap1 cell lines for biochemical analysis. M.C. performed sptPALM experiments using TM-Sra1, LatA and CytoD at lower concentrations. A.P. and A.G. performed immunoprecipitation and western blotting experiments. G.G., K.R. and A.M. wrote the manuscript, with input from all authors.

Corresponding author

Correspondence to Grégory Giannone.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Cell Biology thanks the anonymous reviewers for their contribution to the peer review of this work.

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Prototypic trajectories corresponding to the different diffusion modes: immobile, confined and free-diffusive.

a, Trajectories obtained from a sptPALM acquisition on mEos2-Nap1 in the lamellipodium of a spreading MEF (same cell as in Fig. 1a, 50 Hz, duration: 150 s). Trajectories are color-coded according to their diffusion modes: diffusive (gray), confined (yellow) and immobile (red) (top). b, Prototypic trajectories corresponding to distinct diffusion modes (immobile, confined and free-diffusive) highlighted in a. c, Illustration of the Mean Squared Displacement (MSD) computation from Δt to 4 x Δt using the equation in the inset, where xi and yi are the coordinates of the label position at time \(i \times {\Delta}t\). We defined the measured diffusion coefficient D as the slope of the affine regression line fitted to the n=1 to 4 values of the\(MSD\left( {n \times {\Delta}t} \right)\) (dotted line in the graph). d, Variation of the MSD over time for selected and highlighted trajectories in a and b. e, Variation of the MSD over time for all the trajectories of the sptPALM sequence with the same color-code as in a, mean ± s.e.m. for trajectories. Numerical source data are provided in Source data.

Source data

Extended Data Fig. 2 Diffusive behavior of WRC subunits at the lamellipodium tip and the membrane outside of the lamellipodium.

a, Super-resolution intensity image of mEos2-Abi1 in a MEF lamellipodium obtained by sptPALM (50 Hz, duration: 150 s) (left) (inset: α-actinin-GFP fluorescence image). Trajectories are color-coded according to their diffusion modes: diffusive (gray), confined (yellow), immobile (red) (right). b, Same as a for mEos2-Sra1. c, Same as a for mEos2-Wave2. d, Same as a for mEos2-Actin. e, Same as a for mEos2-CAAX. f, Same as a for cytosolic mEos2. g, Distribution of Log(D) at the Tip versus Outside (mean for cells). h, Fraction of diffusive, confined and immobile populations at the tip versus outside. Data are presented as mean ± s.e.m. for cells. Light, mid and dark shading refer to diffusive, confined and immobile populations, respectively. i, Diffusion coefficients (D) for free diffusive trajectories at Tip versus Outside, box plots displaying median (notch) and mean (square) ± percentiles (25-75%). Images in a-f are representative of 3, 3, 6, 8, 6 and 2 independent experiments that yielded similar results. g-h (for inside and outside), data pooled from independent experiments: mEos2-Nap1, n = 22 cells (pooled over 6 experiments); mEos2-Abi1, n = 11 cells (over 3 experiments); mEos2-Wave2, n = 15 cells (over 6 experiments); mEos2-Sra1, n = 11 cells (over 3 experiments) and cytosolic mEos2, n = 5 cells (over 2 experiments). i, Data pooled from cells in g-h and Fig. 1d,e, n is the number of trajectories: mEos2-ArpC5A, nTip = 1035, nOutside = 691; mEos2-Actin, nTip = 1948, nOutside = 6764; cytosolic mEos2, nTip = 26, nOutside = 111; mEos2-Nap1, nTip = 2976, nOutside = 12493; mEos2-Abi1, nTip = 4183, nOutside = 7233; mEos2-Wave2, nTip = 1936, nOutside = 6885; mEos2-Sra1, nTip = 1715, nOutside = 12564 and mEos2-CAAX, nTip = 455, nOutside = 4605. Where indicated, statistical significances were obtained using two-tailed unpaired t-test for fractions of immobilization (h) or non-parametric, two-tailed Mann-Whitney rank sum test for diffusion coefficient (i). Inside the lamellipodium tip and outside the lamellipodium, all the different conditions were compared (black P-values) (h-i). Inside the lamellipodium tip, each given condition was compared with the value obtained outside the lamellipodium (colored P-values) (h-i). Note that a significant fraction of the Wave2 protein displays a faster membrane free-diffusion which differs from all the other subunits of WRC that we tested in this study. This could correspond to a fraction of the Wave2 protein not incorporated in WRC12, which might diffuse as single proteins at the plasma membrane. Also see Supplementary Table 1. Numerical source data are provided in Source data.

Source data

Extended Data Fig. 3 WRC and FMNL2 lateral movements are driven by actin filaments elongation.

a, Fluorescence image of a MEF lamellipodium labeled with α-actinin-GFP (left). Right, mEos2-Abi1 kymographs generated from a sptPALM time-lapse sequence (2 Hz) perpendicular to the edge (left panel, dashed lines). mEos2-Abi1 motions highlighted (white lines) and corresponding velocities (nm.s-1). b, Distributions of anterograde and retrograde perpendicular movements (duration > 3 s) for mEos2-ArpC5A (n = 535 movements, pooled over 8 cells from 5 independent experiments); mEos2-Nap1 (n = 161 movements, over 6 cells from 3 experiments) and mEos2-Abi1 (n = 143 movements, over 3 cells from 2 experiments). Fractions of anterograde and retrograde movements superior to displacements measured in fixed cells are shown in the plot. c, Fluorescence image of a MEF lamellipodium labeled with α-actinin-GFP (left). Scale bar, 3 µm. Right, mEos2-Abi1 kymographs generated from a sptPALM time-lapse sequence (2 Hz) tangential to the edge (left panel, dashed lines). mEos2-Abi1 motions highlighted (white lines) and corresponding velocities (nm.s-1). d, Same as c for mEos2-Wave2. e, Same as c for mEos2-Abi1 in a fixed MEF. f, Speed of lateral movements (duration > 3 s) for mEos2-Wave2, box plots displaying median (notch) and mean (square) ± percentiles (25-75%). g, Dwell times of lateral movements (duration > 1.5 s) for mEos2-Wave2, box plots displaying median (notch) and mean (square) ± percentiles (25-75%). f, g, Data pooled from independent experiments: mEos2-Wave2, nSpeed = 165 movements, nDwell-Time = 303 movements (pooled over 5 cells from 3 experiments). h, α-actinin-GFP images of a spreading MEF before (left) and after (middle left) acute treatment with LatA (200 nM). Same for LatA (1 µM) (before: middle right, after: right). i, Same as h for CytoD (100 nM, 1 µM). j, Same as c for mEos2-Abi1 in a live MEF treated with LatA (1 μM). k, Same as c for mEos3.2-Sra1 treated with LatA (200 nM). l, Same as c for mEos3.2-Sra1 treated with CytoD (100 nM). m, Same as c for mEos3.2-Sra1-AD treated with LatA (1 µM). n, Distribution of left and right lateral movements (duration > 3 s) for mEos2-Nap1 (n = 263 movements, pooled over 5 cells from 3 independent experiments);mEos2-Nap1 LatA 1 µM (n = 639 movements, over 6 cells from 3 experiments); mEos2-Abi1 (n = 342 movements, over 3 cells from 2 experiments); mEos2-Abi1 LatA 1 µM (n = 210 movements, over 4 cells from 2 experiments); mEos3.2-Sra1 (n = 164 movements, over 5 cells from 2 experiments); mEos3.2-Sra1 LatA 200 nM (n = 275 movements, over 5 cells from 2 experiments); mEos3.2-Sra1 LatA 1 µM (n = 562 movements, over 5 cells from 3 experiments); mEos3.2-Sra1 CytoD 100 nM (n = 327 movements, over 6 cells from 3 experiments); mEos3.2-Sra1 CytoD 1 µM (n = 361 movements, over 6 cells from 3 experiments); mEos3.2-Sra1-WCA* (n = 471 movements, over 6 cells from 3 experiments); mEos3.2-Sra1-AD (n = 62 movements, over 6 cells from 4 experiments); mEos3.2-Sra1-AD LatA 1 µM (n = 145 movements, over 6 cells from 2 experiments); mEos2-Wave2 (n = 165 movements, over 5 cells from 3 experiments); mEos2-Wave2-ΔWCA (n = 270 movements, over 3 cells from 2 experiments); mEos2-Wave2-ΔWCA LatA 1 µM (n = 155 movements, over 3 cells from 2 experiments); FMNL2-mEos2 (n = 479 movements, over 5 cells from 2 experiments); FMNL2-mEos2 LatA 1 µM (n = 206 movements, over 3 cells from 1 experiments); FMNL2-A272E-mEos2 (n = 488 movements, over 4 cells from 2 experiments); FMNL2-I704A-mEos2 (n = 200 movements, over 6 cells from 3 experiments) and fixed mEos2-Abi1 (n = 103 movements, over 2 cells from 1 experiment). Images in a, c-e and h-m are representative of 2, 2, 3, 1, 3, 3, 2, 3, 2, 3 and 3 independent experiments that yielded similar results. Fractions of left and right movements superior to displacements measured in fixed cells are shown in the plot. All results for each condition correspond to pooled data from several independent experiments. Also see Supplementary Table 2. Numerical source data are provided in Source data.

Source data

Extended Data Fig. 4 FRAP experiments show a decreasing order of turnover for Sra1-AD, Sra1 and WCA*.

a, GFP fluorescence images of GFP-Sra1 transfected in Sra1/PIR121-KO B16 cells (upper). GFP-Sra1 time-lapse images during a FRAP experiment (bottom). The outlined area in the upper panel is shown at a higher magnification in the bottom panel. Scale bar, 5 µm. b, Analysis of GFP-Sra1 fluorescence recovery in FRAP experiments performed as in a (mean ± s.e.m. for cells) (n = 24 cells). The recovery is quantified from the outlined area in a bottom. c, d, Same as a,b for GFP-Sra1-WCA* (n = 25 cells). e, f, Same as a,b for GFP-Sra1-AD (n = 30 cells). Images in a, c, d, are representative of 2 independent experiments that yielded similar results. b, d, f, Data pooled from 2 independent experiments. Numerical source data are provided in Source data.

Source data

Extended Data Fig. 5 Diffusive behavior of WRC subunits after LatA treatment.

a, Super-resolution intensity image of mEos2-Nap1 in the lamellipodium of a MEF treated with LatA (1 μM) (50 Hz, duration: 150 s) (left) (inset: α-actinin-GFP fluorescence image). Trajectories are color-coded according to their diffusion modes: diffusive (gray), confined (yellow), immobile (red) (right). b, Same as a for mEos2-Abi1 in a MEF treated with LatA (1 μM). c, Same as a for mEos2-CAAX in a MEF treated with LatA (1 μM). d, Distribution of Log(D) at the Tip versus Outside (mean for cells). e, Fraction of diffusive, confined and immobile populations at the tip versus outside. Data are presented as mean ± s.e.m. for cells. Light, mid and dark shading refer to diffusive, confined and immobile populations, respectively. f, Diffusion coefficients (D) for free diffusive trajectories at Tip versus Outside, box plots displaying median (notch) and mean (square) ± percentiles (25-75%). Images in a-c are representative of 3, 5 and 2 independent experiments that yielded similar results. d-e, (for inside and outside), data pooled from independent experiments: mEos2-Nap1, n = 22 cells (pooled over 6 experiments); mEos2-Nap1 treated with 1 µM of LatA, n = 6 cells (over 3 experiments); mEos2-Abi1, n = 11 cells (over 3 experiments); mEos2-Abi1 treated with 1 µM of LatA, n = 9 cells (over 5 experiments); mEos2-CAAX, n = 7 cells (over 6 experiments) and mEos2-CAAX treated with 1 µM of LatA, n = 4 cells (over 2 experiments). k, Data pooled from cells in d-e, n is the number of trajectories: mEos2-Nap1, nTip = 2976, nOutside = 12493; mEos2-Nap1 treated with 1 µM of LatA, nTip = 399, nOutside = 667; mEos2-Abi1, nTip = 4183, nOutside = 7233; mEos2-Abi1 treated with 1 µM of LatA, nTip = 383, nOutside = 1191; mEos2-CAAX, nTip = 455, nOutside = 4605 and mEos2-CAAX treated with 1 µM of LatA, nTip = 314, nOutside = 1671. Where indicated, statistical significances were obtained using two-tailed unpaired t-test for fractions of immobilization (e) or non-parametric, two-tailed Mann-Whitney rank sum test for diffusion coefficient (f). Inside the lamellipodium tip and outside the lamellipodium, all the different conditions were compared (black P-values) (e,f). Inside the lamellipodium tip, each given condition was compared with the value obtained outside the lamellipodium (colored P-values) (e,f). Also see Supplementary Table 1. Numerical source data are provided in Source data.

Source data

Extended Data Fig. 6 Diffusive behavior of Wave2-ΔWCA, FMNL2, Sra1-AD after LatA treatment.

a, Super-resolution intensity image of mEos2-Wave2-ΔWCA in a MEF lamellipodium obtained by sptPALM (50 Hz, duration: 150 s) (left) (inset: α-actinin-GFP fluorescence image). Trajectories are color-coded according to their diffusion modes: diffusive (gray), confined (yellow), immobile (red) (right). b, Same as a for mEos2-Wave2-ΔWCA in a MEF treated with LatA (1 μM). c, Same as a for mEos2-Sra1-AD in a MEF treated with LatA (1 μM). d, Same as a for FMNL2-mEos2 in a MEF treated with LatA (1 μM). e, Distribution of Log(D) at the Tip versus Outside (mean for cells). f, Fraction of diffusive, confined and immobile populations at the tip versus outside. Data are presented as mean ± s.e.m. for cells. Light, mid and dark shading refer to diffusive, confined and immobile populations, respectively. g, Diffusion coefficients (D) for free diffusive trajectories at Tip versus Outside, box plots displaying median (notch) and mean (square) ± percentiles (25-75%). Images in a-d are representative of 3, 2, 4 and 3 independent experiments that yielded similar results. e-f, (for inside and outside), data pooled from independent experiments: mEos3.2-Sra1-AD, n = 7 cells (pooled over 4 experiments); mEos3.2-Sra1-AD treated with 1 µM of LatA, n = 7 cells (over 2 experiments); mEos2-Wave2, n = 15 cells (over 6 experiments); mEos2-Wave2-ΔWCA, n = 8 cells (over 3 experiments); mEos2-Wave2-ΔWCA treated with 1 µM of LatA, n = 4 cells (over 2 experiments); FMNL2-mEos2, n = 19 cells (over 5 experiments) and FMNL2-mEos2 treated with 1 µM of LatA, n = 6 cells (over 3 experiments). k, Data pooled from cells in i-j, n is the number of trajectories: mEos3.2-Sra1-AD, nTip = 398, nOutside = 2971; mEos3.2-Sra1-AD treated with 1 µM of LatA, nTip = 202, nOutside = 738; mEos2-Wave2, nTip = 1936, nOutside = 6885; mEos2-Wave2-ΔWCA, nTip = 1033, nOutside = 8281; mEos2-Wave2-ΔWCA treated with 1 µM of LatA, nTip = 69, nOutside = 255; FMNL2-mEos2, nTip = 3388, nOutside = 19567 and FMNL2-mEos2 treated with 1 µM of LatA, nTip = 743, nOutside = 3245. Where indicated, statistical significances were obtained using two-tailed unpaired t-test for fractions of immobilization (f) or non-parametric, two-tailed Mann-Whitney rank sum test for diffusion coefficient (g). Inside the lamellipodium tip and outside the lamellipodium, all the different conditions were compared (black P-values) (f,g). Inside the lamellipodium tip, each given condition was compared with the value obtained outside the lamellipodium (colored P-values) (f,g). Also see Supplementary Table 1. Numerical source data are provided in Source data.

Source data

Extended Data Fig. 7 Fast directed motions of FMNL2 inside and outside the lamellipodium.

a, Fluorescence image of α-actinin-GFP in a MEF co-transfected with FMNL2-mEos2 (left). Kymographs generated from a raw sptPALM acquisition (50 Hz) to measure FMNL2-mEos2 fast directed motions occurring in the lamellipodium (1) or outside the lamellipodium (2,3) (left panel, dashed lines). Fast motions of FMNL2-mEos2 highlighted (white lines) and corresponding velocities (nm.s-1). b, Distributions of speed for fast directed motions occurring in the lamellipodium (n = 29 events), or outside the lamellipodium (n = 38 events) for FMNL2-mEos2 (pooled over 3 cells from 2 experiments), box plots displaying median (notch) and mean (square) ± percentiles (25-75%). c, Super-resolution intensity image of FMNL2-mEos2 in the a MEF lamellipodium obtained by sptPALM (50 Hz, duration: 150 s) (left). Right, FMNL2-mEos2 kymographs generated from a sptPALM time-lapse sequence (2 Hz) along LOI corresponding to directed motions occurring within or outside the lamellipodium. d, same as c for FMNL2-I704A-mEos2. e, same as c for FMNL2-A272E-mEos2. f, density of directed motions, box plots displaying median (horizontal line) and mean (square) ± percentiles (25-75%) for each cell. Data pooled from independent experiments: mEos3.2-Sra1, n = 5 cells (pooled over 2 experiments); mEos3.2-Sra1-WCA*, n = 4 cells (over 2 experiments) and mEos3.2-Sra1-AD, n = 6 cells (over 3 experiments). Images in a, c-e are representative of 2, 2, 2 and 3 independent experiments that yielded similar results. Where indicated, statistical significance were obtained using two-tailed Mann-Whitney rank sum test for speed (b) and density (f) of directed motions. Also see Supplementary Table 3. Numerical source data are provided in Source data.

Source data

Extended Data Fig. 8 Sra1 lateral movements in B16 FMNL2/3 KO.

a, sptPALM image of mEos3.2-Sra1 in the lamellipodium of a FMNL2/3 KO B16 cell (50 Hz) (left) (inset: α-actinin-GFP fluorescence image). Trajectories are color-coded according to their diffusion modes: diffusive (gray), confined (yellow), immobile (red) (right). b, Same as a for mEos3.2-Sra1 in a FMNL2/3 KO B16 cell expressing FMNL2-GFP (insets: α-actinin-GFP and FMNL2-GFP fluorescence images). c, Distribution of Log(D) at the Tip versus Outside (mean for cells). d, Fraction of diffusive, confined and immobile populations at the tip versus outside. Data are presented as mean ± s.e.m. for cells. Light, mid and dark shading refer to diffusive, confined and immobile populations, respectively. e, Diffusion coefficients (D) for free diffusive trajectories at Tip versus Outside, box plots displaying median (notch) and mean (square) ± percentiles (25-75%). c,d, (for inside and outside), Data pooled from 2 independent experiments: mEos3.2-Sra1 (n = 11 cells) and mEos3.2-Sra1 in cells expressing FMNL2-GFP (n = 6 cells). e, Data pooled from cells in c,d, n is the number of trajectories: mEos3.2-Sra1, nTip = 3714, nOutside = 9469 and mEos3.2-Sra1 in cells expressing FMNL2-GFP, nTip = 1682, nOutside = 4311. f, Fluorescence image of the lamellipodium labeled with α-actinin-GFP (left) in a FMNL2/3 KO B16 cell. Scale bar, 3 µm. Right, mEos3.2-Sra1 kymographs generated from a sptPALM time-lapse sequence (2 Hz) tangential to the edge (left panel, dashed lines). mEos3.2-Sra1 motions highlighted (white lines) and corresponding velocities (nm.s-1). g, Fluorescence image of the lamellipodium labeled with FMNL2-GFP (left) in a FMNL2/3 KO B16 cell. Right, mEos3.2-Sra1 kymographs. h, Speed of lateral movements (duration > 3 s), box plots displaying median (notch) and mean (square) ± percentiles (25-75%). i, Dwell times of lateral movements (duration > 1.5 s), box plots displaying median (notch) and mean (square) ± percentiles (25-75%). j, Distribution of left and right of lateral movements (duration > 3 s). h-j, Data pooled from 2 independent experiments: mEos3.2-Sra1, nSpeed = 186 movements, nDwell-Time = 372 movements, nDisplacement = 186 movements (over 5 cells) and mEos3.2-Sra1, nSpeed = 124 movements, nDwell-Time = 309 movements, nDisplacement = 124 movements (over 5 cells). Images in a-b and f-g are representative of 2 independent experiments that yielded similar results. Where indicated, statistical significances were obtained using two-tailed unpaired t-test for fractions of immobilization (d) or non-parametric, two-tailed Mann-Whitney rank sum test for diffusion coefficient (e). Inside the lamellipodium tip and outside the lamellipodium, all the different conditions were compared (black P-values) (d,e). Inside the lamellipodium tip, each given condition was compared with the value obtained outside the lamellipodium (colored P-values) (d,e). Statistical significance were obtained using two-tailed Mann-Whitney rank sum test for speed (h) and dwell time (i) of lateral movements. Also see Supplementary Table 1 and Supplementary Table 2. Numerical source data are provided in Source data.

Source data

Extended Data Fig. 9 Diffusive behavior of transmembrane-Sra1 versus controls.

a, Schematic representation of TM-Sra and the transmembrane control protein, TM-ctr. b, Super-resolution intensity image of TM-ctr in a MEF lamellipodium obtained by sptPALM (50 Hz, duration: 150 s) (left) (inset: α-actinin-GFP fluorescence image). Trajectories are color-coded according to their diffusion modes: diffusive (gray), confined (yellow), immobile (red) (right). c, Same as b for TM-Sra1. d, Distribution of Log(D) at the Tip versus Outside (mean for cells). e, Fraction of diffusive, confined and immobile populations at the tip versus outside. Data are presented as mean ± s.e.m. for cells. Light, mid and dark shading refer to diffusive, confined and immobile populations, respectively. f, Diffusion coefficients (D) for free diffusive trajectories at Tip versus Outside, box plots displaying median (notch) and mean (square) ± percentiles (25-75%). d, e (for inside and outside), data pooled from independent experiments: mEos3.2-Sra1, n = 9 cells (pooled over 4 experiments); TM-Sra1, n = 9 cells (over 2 experiments) and mEos2-TM-ctr, n = 10 cells (over 2 experiments). f, Data pooled from cells in d-e and Fig. 5c,d, n is the number of trajectories: mEos2-Nap1, nTip = 2976, nOutside = 12493; TM-Nap1, nTip = 1820, nOutside = 6729; mEos3.2-Sra1, nTip = 921, nOutside = 3713; TM-Sra1, nTip = 629, nOutside = 3174 and mEos2-TM-ctr, nTip = 1893, nOutside = 16442. g, Western blotting of Nap1-deficient (Nap1-KO) and control B16-F1 cells expressing TM-Nap1 and using anti-Nap1 or anti-GFP antibodies, as indicated; tubulin expression was used as loading control. Blot in g is representative of 3 independent experiments that yielded similar results. h, Filamentous actin labeled with ATTO-594-labeled phalloidin in Sra1/PIR121-KO B16 cells (upper left and right). Arrowheads point to lamellipodia. GFP fluorescence images of TM-Sra1 (lower left) transfected into Sra1-KO B16-F1 melanoma cells or untransfected cells (lower right). i, Quantification of cells displaying lamellipodia in Sra1/PIR121-KO B16 cells transfected with TM-Sra1 (n = 80 cells pooled over 3 independent experiments) or untransfected (n = 55 cells pooled over 3 independent experiments) (mean ± s.e.m. for experiments). Images in b, c and h are representative of 2, 2 and 3 independent experiments that yielded similar results. Where indicated, statistical significances were obtained using two-tailed unpaired t-test for fractions of immobilization (e) or t-test for lamellipodia formation (i), or non-parametric, two-tailed Mann-Whitney rank sum test for diffusion coefficient (f). Inside the lamellipodium tip and outside the lamellipodium, all the different conditions were compared (black P-values) (e,f). Inside the lamellipodium tip, each given condition was compared with the value obtained outside the lamellipodium (colored P-values) (e,f). Also see Supplementary Table 1. Numerical source data are provided in Source data. Unprocessed blot from g is available in the Source Data.

Source data

Extended Data Fig. 10 Optical tweezer calibrations for pulling and connection-breaking assays.

a, Schematic representation of the optical tweezers binding assay. Nanobody-beads were held for 3 s on the cell surface 1 µm behind the tip of the lamellipodium using the laser trap. Beads were scored as bound if they remained focused in the plane of the membrane for more than 10 s after turning the laser tweezers off. b, Binding fractions as a function of the Nanobody-Biotin/BSA-Biotin ratio on beads for GFP-β3-integrin (n0/1 = 24 cells, n1/2 = 11 cells and n1/0 = 43 cells, pooled over 8, 3 and 15 independent experiments), TM-Nap1 (n0/1 = 6 cells, n1/2 = 46 cells and n1/2 = 29 cells, pooled over 2, 9 and 5 experiments) and TM-ctr (n0/1 = 3 cells, n1/2 = 12 cells and n1/0 = 5 cells, pooled over 1, 7 and 2 experiments), (mean ± s.e.m. for cells). c, Fractions of pulled beads below 1 pN (light histograms), between 1 pN-2 pN (mid shaded histograms) and above 2 pN (dark histograms), for TM-Nap1 (n = 5 experiments), TM-Sra1 (n = 3 experiments), TM-Sra1-WCA* (n = 3 experiments), TM-Sra1-AD (n = 3 experiments) and TM-ctr (n = 4 experiments), (mean ± s.e.m. for experiments). d, Pulled fraction as function of the maximal escapement force determined for defined trap stiffnesses. TM-Nap1 (n = 329 pulling assay), TM-Sra1 (n = 399 pulling assay), TM-Sra1-WCA* (n = 372 pulling assay), TM-Sra1-AD (n = 211 pulling assay) and TM-ctr (n = 157 pulling assay). c, d, Data pooled from independent experiments: TM-Nap1 (pooled over 28 cells from 5 experiments), TM-Sra1(pooled over 11 cells from 3 experiments), TM-Sra1-WCA* (pooled over 12 cells from 3 experiments), TM-Sra1-AD (pooled over 18 cells from 3 experiments) and TM-ctr (pooled over 9 cells from 4 experiments). e, Fractions of breaking forces bellow 1 pN (dark histograms), between 1 pN-2 pN (mid shaded histograms) and above 2 pN (light shaded histograms). TM-Nap1 (11 cells), TM-Sra1(9 cells), TM-Sra1-WCA* (22 cells), TM-Sra1-AD (4 cells) and TM-ctr (5 cells), (mean ± s.e.m. for cells). f, Distributions of breaking forces for nanobody-beads bound to TM-Nap1 (n = 54 breaking events), TM-Sra1 (n = 65 breaking events), TM-Sra1-WCA* (n = 218 breaking events), TM-Sra1-AD (n = 8 breaking events) and TM-ctr (n = 16 breaking events). e, f, Data pooled from independent experiments: TM-Nap1 (pooled over 11 cells from 5 experiments), TM-Sra1 (pooled over 9 cells from 5 experiments), TM-Sra1-WCA* (pooled over 22 cells from 8 experiments), TM-Sra1-AD (pooled over 4 cells from 3 experiments) and TM-ctr (pooled over 5 cells from 3 experiments). g, Using a piezoelectric motor, the sample was moved at increasing velocities. The increasing viscous drag force exerted on the bead triggered larger displacements until escapement from laser trap (left). Displacement versus time plots from the trap center as function of the viscous drag force at different laser powers (2.4, 3.9 and 7.9 mW) (right). h, Displacement amplitudes as function of the viscous drag force. Trap stiffnesses were calculated for different laser powers, and corresponded to the slope of displacement versus force curves. i, Trap stiffness as a function of laser power. j, Maximal trap force as function of laser power. Also see Supplementary Table 4. Numerical source data are provided in Source data.

Source data

Supplementary information

Reporting Summary

Supplementary Table

Supplementary Video 1

From a raw sptPALM acquisition to a super-resolution intensity image and corresponding trajectories.

Supplementary Video 2

Super-resolution time-lapse sequence of ArpC5A.

Supplementary Video 3

Super-resolution time-lapse sequence of Nap1.

Supplementary Video 4

Super-resolution time-lapse sequence of Abi1.

Supplementary Video 5

Super-resolution time-lapse sequence of Nap1 after LatA treatment.

Supplementary Video 6

Super-resolution time-lapse sequence of Abi1 after LatA treatment.

Supplementary Video 7

Super-resolution time-lapse sequence of Wave2-ΔWCA.

Supplementary Video 8

Super-resolution time-lapse sequence of Wave2-ΔWCA after LatA treatment.

Supplementary Video 9

Fast directed motions of FMNL2 at the lamellipodium tip and outside the lamellipodium.

Supplementary Video 10

Super-resolution time-lapse sequence of FMNL2-mEos2.

Supplementary Video 11

Super-resolution time-lapse sequence of FMNL2-mEos2 after LatA treatment.

Supplementary Video 12

Pulling assay for a ‘pulled’ nanobody-bead.

Supplementary Video 13

Pulling assay for a ‘not pulled’ nanobody-bead.

Supplementary Video 14

Breaking assay for TM-Nap1.

Supplementary Video 15

Breaking assay for TM-ctr.

Supplementary Video 16

TM-Sra1 crosslinking increases membrane protrusion rates.

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mehidi, A., Kage, F., Karatas, Z. et al. Forces generated by lamellipodial actin filament elongation regulate the WAVE complex during cell migration. Nat Cell Biol 23, 1148–1162 (2021). https://doi.org/10.1038/s41556-021-00786-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-021-00786-8

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