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:

Twinfilin uncaps filament barbed ends to promote turnover of lamellipodial actin networks

A Publisher Correction to this article was published on 02 March 2021

This article has been updated

Abstract

Coordinated polymerization of actin filaments provides force for cell migration, morphogenesis and endocytosis. Capping protein (CP) is a central regulator of actin dynamics in all eukaryotes. It binds to actin filament (F-actin) barbed ends with high affinity and slow dissociation kinetics to prevent filament polymerization and depolymerization. However, in cells, CP displays remarkably rapid dynamics within F-actin networks, but the underlying mechanism remains unclear. Here, we report that the conserved cytoskeletal regulator twinfilin is responsible for CP’s rapid dynamics and specific localization in cells. Depletion of twinfilin led to stable association between CP and cellular F-actin arrays, as well as to its retrograde movement throughout leading-edge lamellipodia. These were accompanied by diminished F-actin turnover rates. In vitro single-filament imaging approaches revealed that twinfilin directly promotes dissociation of CP from filament barbed ends, while enabling subsequent filament depolymerization. These results uncover a bipartite mechanism that controls how actin cytoskeleton-mediated forces are generated in cells.

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: KO of twinfilins leads to F-actin accumulation in the lamellipodia and perinuclear region.
Fig. 2: Twinfilin regulates actin dynamics in lamellipodia.
Fig. 3: Twinfilin does not accelerate barbed-end depolymerization of ADP-actin filaments.
Fig. 4: Twinfilin and CP display different localizations and dynamics in lamellipodia.
Fig. 5: Twinfilin regulates CP dynamics.
Fig. 6: Twinfilin accelerates filament barbed end uncapping in vitro.
Fig. 7: The actin-binding function of twinfilin is required for filament uncapping.
Fig. 8: Localization of twinfilin in lamellipodia and its role in regulation of CP dynamics.

Similar content being viewed by others

Data availability

Examples of all FRAP experiments over 5–9 time points as well as Excel spreadsheets summarizing the measured fluorescence intensity values in each cell are available at FigShare (https://doi.org/10.6084/m9.figshare.13221638.v1). Other original data of the study are available on reasonable request from the corresponding author. Source data are provided with this paper.

Change history

References

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

    Article  CAS  PubMed  Google Scholar 

  2. Svitkina, T. Ultrastructure of the actin cytoskeleton. Curr. Opin. Cell Biol. 54, 1–8 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Senju, Y. & Lappalainen, P. Regulation of actin dynamics by PI(4,5)P2 in cell migration and endocytosis. Curr. Opin. Cell Biol. 56, 7–13 (2019).

    Article  CAS  PubMed  Google Scholar 

  4. Rottner, K. & Schaks, M. Assembling actin filaments for protrusion. Curr. Opin. Cell Biol. 56, 53–63 (2019).

    Article  CAS  PubMed  Google Scholar 

  5. De Melo, L. et al. Evolutionary conservation of actin-binding proteins in Trypanosoma cruzi and unusual subcellular localization of the actin homologue. Parasitology 135, 955–965 (2008).

    Article  PubMed  Google Scholar 

  6. Pollard, T. D. & Borisy, G. G. Cellular motility driven by assembly and disassembly of actin filaments. Cell 112, 453–465 (2003).

    Article  CAS  PubMed  Google Scholar 

  7. Cooper, J. & Schafer, D. Control of actin assembly and disassembly at filament ends. Curr. Opin. Cell Biol. 12, 97–103 (2000).

    Article  CAS  PubMed  Google Scholar 

  8. Carlier, M.-F. & Pantaloni, D. Control of actin dynamics in cell motility. J. Mol. Biol. 269, 459–467 (1997).

    Article  CAS  PubMed  Google Scholar 

  9. Edwards, M. et al. Capping protein regulators fine-tune actin assembly dynamics. Nat. Rev. Mol. Cell Biol. 15, 677–689 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Shekhar, S., Pernier, J. & Carlier, M.-F. Regulators of actin filament barbed ends at a glance. J. Cell Sci. 129, 1085–1091 (2016).

    CAS  PubMed  Google Scholar 

  11. Loisel, T., Boujemaa, R., Pantaloni, D. & Carlier, M.-F. Reconstitution of actin-based motility of Listeria and Shigella using pure proteins. Nature 401, 613–616 (1999).

    Article  CAS  PubMed  Google Scholar 

  12. Amatruda, J., Cannon, J., Tatchell, K., Hug, C. & Cooper, J. Disruption of the actin cytoskeleton in yeast capping protein mutants. Nature 344, 352–354 (1990).

    Article  CAS  PubMed  Google Scholar 

  13. Amatruda, J., Gattermeir, D., Karpova, T. & Cooper, J. Effects of null mutations and overexpression of capping protein on morphogenesis, actin distribution and polarized secretion in yeast. J. Cell Biol. 119, 1151–1162 (1992).

    Article  CAS  PubMed  Google Scholar 

  14. Mejillano, M. et al. Lamellipodial versus filopodial mode of the actin nanomachinery. Cell 118, 363–373 (2004).

    Article  CAS  PubMed  Google Scholar 

  15. Iwasa, J. & Mullins, R. D. Spatial and temporal relationships between actin-filament nucleation, capping, and disassembly. Curr. Biol. 17, 395–406 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Rogers, S., Wiedemann, U., Stuurman, N. & Vale, R. Molecular requirements for actin-based lamella formation in Drosophila S2 cells. J. Cell Biol. 162, 1079–1088 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Kim, K., Yamashita, A., Wear, M., Maéda, Y. & Cooper, J. Capping protein binding to actin in yeast: biochemical mechanism and physiological relevance. J. Cell Biol. 164, 567–580 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Sinnar, S., Antoku, S., Saffin, J.-M., Cooper, J. & Halpain, S. Capping protein is essential for cell migration in vivo and for filopodial morphology and dynamics. Mol. Biol. Cell 25, 2152–2160 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Akin, O. & Mullins, R. D. Capping protein increases the rate of actin-based motility by promoting filament nucleation by the Arp2/3 complex. Cell 133, 841–851 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Bhattacharya, N., Ghosh, S., Sept, D. & Cooper, J. Binding of myotrophin/V-1 to actin-capping protein: implications for how capping protein binds to the filament barbed end. J. Biol. Chem. 281, 31021–31030 (2006).

    Article  CAS  PubMed  Google Scholar 

  21. Jung, G. et al. V-1 regulates capping protein activity in vivo. Proc. Natl Acad. Sci. USA 113, E6610–E6619 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Fujiwara, I., Remmert, K., Piszczek, G. & Hammer, J. Capping protein regulatory cycle driven by CARMIL and V-1 may promote actin network assembly at protruding edges. Proc. Natl Acad. Sci. USA 111, E1970–E1979 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Taoka, M. et al. V-1, a protein expressed transiently during murine cerebellar development, regulates actin polymerization via interaction with capping protein. J. Biol. Chem. 278, 5864–5870 (2003).

    Article  CAS  PubMed  Google Scholar 

  24. Stark, B., Lanier, M. H. & Cooper, J. CARMIL family proteins as multidomain regulators of actin-based motility. Mol. Biol. Cell 28, 1713–1723 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  25. Takeda, S. et al. Two distinct mechanisms for actin capping protein regulation-steric and allosteric inhibition. PLoS Biol. 8, e1000416 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Uruno, T., Remmert, K. & Hammer, J. CARMIL is a potent capping protein antagonist: identification of a conserved CARMIL domain that inhibits the activity of capping protein and uncaps capped actin filaments. J. Biol. Chem. 281, 10635–10650 (2006).

    Article  CAS  PubMed  Google Scholar 

  27. Yang, C. et al. Mammalian CARMIL inhibits actin filament capping by capping protein. Dev. Cell 9, 209–221 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Hernandez-Valladares, M. et al. Structural characterization of a capping protein interaction motif defines a family of actin filament regulators. Nat. Struct. Mol. Biol. 17, 497–503 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Edwards, M., Liang, Y., Kim, T. & Cooper, J. A. Physiological role of the interaction between CARMIL1 and capping protein. Mol. Biol. Cell 24, 3047–3055 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Edwards, M., McConnell, P., Schafer, D. & Cooper, J. CPI motif interaction is necessary for capping protein function in cells. Nat. Commun. 6, 8415 (2015).

    Article  CAS  PubMed  Google Scholar 

  31. Park, L. et al. Cyclical action of the WASH Complex: FAM21 and capping protein drive WASH recycling, not initial recruitment. Dev. Cell 24, 169–181 (2013).

    Article  CAS  PubMed  Google Scholar 

  32. Zhao, J. et al. CD2AP links cortactin and capping protein at the cell periphery to facilitate formation of lamellipodia. Mol. Cell. Biol. 33, 38–47 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Lanier, M. H., McConnell, P. & Cooper, J. Cell migration and invadopodia formation require a membrane-binding domain of CARMIL2. J. Biol. Chem. 291, 1076–1091 (2016).

    Article  PubMed  Google Scholar 

  34. Schafer, D., Jennings, P. & Cooper, J. Dynamics of capping protein and actin assembly in vitro: uncapping barbed ends by polyphosphoinositides. J. Cell Biol. 135, 169–179 (1996).

    Article  CAS  PubMed  Google Scholar 

  35. Wioland, H. et al. ADF/cofilin accelerates actin dynamics by severing filaments and promoting their depolymerization at both ends. Curr. Biol. 27, 1956–1967 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Kim, T., Cooper, J. & Sept, D. The interaction of capping protein with the barbed end of the actin filament. J. Mol. Biol. 404, 794–802 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Lai, F. 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 

  38. Miyoshi, T. et al. Actin turnover-dependent fast dissociation of capping protein in the dendritic nucleation actin network: evidence of frequent filament severing. J. Cell Biol. 175, 947–955 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Fujiwara, I., Remmert, K. & Hammer, J. Direct observation of the uncapping of capping protein-capped actin filaments by CARMIL homology domain 3. J. Biol. Chem. 285, 2707–2720 (2010).

    Article  CAS  PubMed  Google Scholar 

  40. Poukkula, M., Kremneva, E., Serlachius, M. & Lappalainen, P. Actin-depolymerizing factor homology domain: a conserved fold performing diverse roles in cytoskeletal dynamics. Cytoskeleton 68, 471–490 (2011).

    Article  CAS  PubMed  Google Scholar 

  41. Falck, S. et al. Biological role and structural mechanism of twinfilin-capping protein interaction. EMBO J. 23, 3010–3019 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Hakala, M., Kalimeri, M., Enkavi, G., Vattulainen, I. & Lappalainen, P. Molecular mechanism for inhibition of twinfilin by phosphoinositides. J. Biol. Chem. 293, 4818–4829 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Johnston, A. et al. A novel mode of capping protein-regulation by twinfilin. eLife 7, e41313 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Goode, B., Drubin, D. & Lappalainen, P. Regulation of the cortical actin cytoskeleton in budding yeast by twinfilin, a ubiquitous actin monomer-sequestering protein. J. Cell Biol. 142, 723–733 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Vartiainen, M., Ojala, P., Auvinen, P., Peränen, J. & Lappalainen, P. Mouse A6/twinfilin is an actin monomer-binding protein that localizes to the regions of rapid actin dynamics. Mol. Cell Biol. 20, 1772–1783 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Ojala, P. et al. The two ADF-H domains of twinfilin play functionally distinct roles in interactions with actin monomers. Mol. Biol. Cell 13, 3811–3821 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Helfer, E. et al. Mammalian twinfilin sequesters ADP-G-actin and caps filament barbed ends: implications in motility. EMBO J. 25, 1184–1195 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Johnston, A., Collins, A. & Goode, B. High-speed depolymerization at actin filament ends jointly catalysed by twinfilin and Srv2/CAP. Nat. Cell Biol. 17, 1504–1511 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Hilton, D., Aguilar, R., Johnston, A. & Goode, B. Species-specific functions of twinfilin in actin filament depolymerization. J. Mol. Biol. 430, 3323–3336 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Wahlström, G. et al. Twinfilin is required for actin-dependent developmental processes in Drosophila. J. Cell Biol. 155, 787–796 (2001).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Wang, D. et al. Drosophila twinfilin is required for cell migration and synaptic endocytosis. J. Cell Sci. 123, 1546–1556 (2010).

    Article  CAS  PubMed  Google Scholar 

  52. Stritt, S. et al. Twinfilin 2a is a regulator of platelet reactivity and turnover in mice. Blood 130, 1746–1756 (2017).

    Article  CAS  PubMed  Google Scholar 

  53. Bockhorn, J. et al. MicroRNA-30c inhibits human breast tumour chemotherapy resistance by regulating TWF1 and IL-11. Nat. Commun. 4, e1393 (2013).

    Article  Google Scholar 

  54. Nevalainen, E., Skwarek-Maruszewska, A., Braun, A., Moser, M. & Lappalainen, P. Two biochemically distinct and tissue-specific twinfilin isoforms are generated from the mouse Twf2 gene by alternative promoter usage. Biochem. J. 417, 593–600 (2009).

    Article  CAS  PubMed  Google Scholar 

  55. Abella, J. V. G. et al. Isoform diversity in the Arp2/3 complex determines actin filament dynamics. Nat. Cell Biol. 18, 76–86 (2016).

    Article  CAS  PubMed  Google Scholar 

  56. Kapustina, M., Read, T. A. & Vitriol, E. A. Simultaneous quantification of actin monomer and filament dynamics with modeling-assisted analysis of photoactivation. J. Cell Sci. 129, 4633–4643 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Burnette, D. T. et al. A role for actin arcs in the leading-edge advance of migrating cells. Nat. Cell Biol. 13, 371–381 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Carlier, M. F., Romet-Lemonne, G. & Jégou, A. Actin filament dynamics using microfluidics. Methods Enzymol. 540, 3–17 (2014).

    Article  CAS  PubMed  Google Scholar 

  59. Jégou, A., Carlier, M. F. & Romet-Lemonne, G. Microfluidics pushes forward microscopy analysis of actin dynamics. Bioarchitecture 1, 271–276 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Pollard, T. Rate constants for the reactions of ATP- and ADP-actin with the ends of actin filaments. J. Cell Biol. 103, 2747–2754 (1986).

    Article  CAS  PubMed  Google Scholar 

  61. Carlier, M. F., Criquet, P., Pantaloni, D. & Korn, E. D. Interaction of cytochalasin D with actin filaments in the presence of ADP and ATP. J. Biol. Chem. 261, 2041–2050 (1986).

    Article  CAS  PubMed  Google Scholar 

  62. Fujiwara, I., Zweifel, M. E., Courtemanche, N. & Pollard, T. D. Latrunculin A accelerates actin filament depolymerization in addition to sequestering actin monomers. Curr. Biol. 28, 3183–3192 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  63. Coue, M. & Korn, E. D. Interaction of plasma gelsolin with G-actin and F-actin in the presence and absence of calcium ions. J. Biol. Chem. 260, 15033–15041 (1985).

    Article  CAS  PubMed  Google Scholar 

  64. Kuhn, J. R. & Pollard, T. D. Real-time measurements of actin filament polymerization by total internal reflection fluorescence microscopy. Biophys. J. 88, 1387–1402 (2005).

    Article  CAS  PubMed  Google Scholar 

  65. Paavilainen, V. et al. Structural basis and evolutionary origin of actin filament capping by twinfilin. Proc. Natl Acad. Sci. USA 104, 3113–3118 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Palmgren, S., Ojala, P., Wear, M., Cooper, J. & Lappalainen, P. Interactions with PIP2, ADP-actin monomers, and capping protein regulate the activity and localization of yeast twinfilin. J. Cell Biol. 155, 251–260 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Vartiainen, M., Sarkkinen, E., Matilainen, T., Salminen, M. & Lappalainen, P. Mammals have two twinfilin isoforms whose subcellular localizations and tissue distributions are differentially regulated. J. Biol. Chem. 278, 34347–34355 (2003).

    Article  CAS  PubMed  Google Scholar 

  68. Schafer, D. et al. Visualization and molecular analysis of actin assembly in living cells. J. Cell Biol. 143, 1919–1930 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. Kaksonen, M., Toret, C. P. & Drubin, D. G. A modular design for the clathrin- and actin-mediated endocytosis machinery. Cell 123, 305–320 (2005).

    Article  CAS  PubMed  Google Scholar 

  70. Liang, Y., Niederstrasser, H., Edwards, M., Jackson, C. E. & Cooper, J. A. Distinct roles for CARMIL isoforms in cell migration. Mol. Biol. Cell 20, 5290–5305 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  71. Paavilainen, V. et al. Structural conservation between the actin monomer-binding sites of twinfilin and actin-depolymerizing factor (ADF)/cofilin. J. Biol. Chem. 277, 43089–43095 (2002).

    Article  CAS  PubMed  Google Scholar 

  72. Paavilainen, V., Oksanen, E., Goldman, A. & Lappalainen, P. Structure of the actin-depolymerizing factor homology domain in complex with actin. J. Cell Biol. 182, 51–59 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Shekhar, S., Hoeprich, G. J., Gelles, J. & Goode, B. L. Twinfilin bypasses assembly conditions and actin filament aging to drive barbed end depolymerization. J. Cell Biol. 220, e202006022 (2021).

    Article  CAS  PubMed  Google Scholar 

  74. Kotila, T. et al. Mechanism of synergistic actin filament pointed end depolymerization by cyclase-associated protein and cofilin. Nat. Commun. 10, 5320 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  75. Shekhar, S., Chung, J., Kondev, J., Gelles, J. & Goode, B. Synergy between cyclase-associated protein and cofilin accelerates actin filament depolymerization by two orders of magnitude. Nat. Commun. 10, 5319 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  76. Poukkula, M. et al. GMF promotes leading-edge dynamics and collective cell migration in vivo. Curr. Biol. 24, 2533–2540 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Haynes, E. M. et al. GMFβ controls branched actin content and lamellipodial retraction in fibroblasts. J. Cell Biol. 209, 803–812 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  78. Pelkmans, L. et al. Genome-wide analysis of human kinases in clathrin- and caveolae/raft-mediated endocytosis. Nature 436, 78–86 (2005).

    Article  CAS  PubMed  Google Scholar 

  79. Meacham, C., Ho, E., Dubrovsky, E., Gertler, F. & Hemann, M. In vivo RNAi screening identifies regulators of actin dynamics as key determinants of lymphoma progression. Nat. Genet. 41, 1133–1137 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Li, Q. et al. Attenuation of microRNA-1 derepresses the cytoskeleton regulatory protein twinfilin-1 to provoke cardiac hypertrophy. J. Cell Sci. 123, 2680–2680 (2010).

    Article  Google Scholar 

  81. Bockhorn, J. et al. MicroRNA-30c targets cytoskeleton genes involved in breast cancer cell invasion. Breast Cancer Res. Treat. 137, 373–382 (2013).

    Article  CAS  PubMed  Google Scholar 

  82. Peng, A., Belyantseva, I., Hsu, P., Friedman, T. & Heller, S. Twinfilin-2 regulates actin filament lengths in cochlear stereocilia. J. Neurosci. 29, 15083–15088 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Ran, F. A. et al. Genome engineering using the CRISPR-Cas9 system. Nat. Protoc. 8, 2281–2308 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  84. Rao, J. N., Madasu, Y. & Dominguez, R. Mechanism of actin filament pointed-end capping by tropomodulin. Science 345, 463–467 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  85. Bancaud, A., Huet, S., Rabut, G. & Ellenberg, J. Fluorescence perturbation techniques to study mobility and molecular dynamics of proteins in live cells: FRAP, photoactivation, photoconversion, and FLIP. Cold Spring Harb. Protoc. 5, 1303–1325 (2010).

    Google Scholar 

  86. Kumari, R. et al. Tropomodulins control the balance between protrusive and contractile structures by stabilizing actin-tropomyosin filaments. Curr. Biol. 30, 767–778 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  87. Remmert, K., Uruno, T. & Hammer, J. A. Purification of capping protein using the capping protein binding site of CARMIL as an affinity matrix. Protein Expr. Purif. 67, 113–119 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  88. Kaplan, E. L. & Meier, P. Nonparametric estimation from incomplete observations. J. Am. Stat. Assoc. 53, 457–481 (1958).

    Article  Google Scholar 

Download references

Acknowledgements

We thank V. Paavilainen and J. Saarikangas for reading the manuscript. We acknowledge staff at the Institute of Biotechnology Light Microscopy Unit, Biomedicum Imaging Unit and Institute of Molecular Medicine Finland High Content Imaging and Analysis Unit for providing support in imaging and image analysis. We thank M. Tirkkonen for technical assistance. This study was supported by grants from the Academy of Finland (no. 302161) and Cancer Society Finland (no. 4705949; to P.L.); the Doctoral School in Health Sciences at the University of Helsinki (to M.H. and T.K.) and from the Agence Nationale de la Recherche (Grant Muscactin; to G.R.-L.) and from the European Research Council (no. StG-679116; to A.J.).

Author information

Authors and Affiliations

Authors

Contributions

P.L. and M.H. crafted the original idea, and M.H., H.W., T.K., A.J., G.R.-L. and P.L. designed the experiments. M.H., M.T., T.K., A.J. and H.W. performed the experiments, and M.H., A.J. and H.W. analysed the data. M.H. and P.L. drafted the manuscript with contributions from all of the authors. P.L., A.J. and G.R.-L. acquired the funding.

Corresponding author

Correspondence to Pekka Lappalainen.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Extended data

Extended Data Fig. 1 Generation of twinfilin-1, twinfilin-2, and twinfilin-1/twinfilin-2 knockout B16-F1 cells.

a-b, Western blot analysis of wild-type and twinfilin knockout cells performed with anti-TWF-1 (panel a) and anti-TWF-2 (panel b) antibodies. Anti-α-tubulin antibody was used as a loading control. c, Sanger sequencing of the third exon of twinfilin-1 gene from genomic DNA of wild-type and twf1/twf2-KO g3 knockout cell lines. Sequences obtained with forward and reverse primers for the third exon of twinfilin-1 are shown. d, Sanger sequencing of the third exon of twinfilin-2 from genomic DNA of wild-type and twf1/twf2-KO g3 knockout cell lines. Sequences with forward and reverse primers for the third exon of twinfilin-2 are shown. e-f, MiSeq NGS sequencing results of twinfilin-1 exon 3 (panel e), and twinfilin-2 exon 3 (panel f) of wild-type and twf1/twf2-KO-g3 cells. Due to chromosome amplification, twinfilin-1 knockout was a result of two allele variants and twinfilin-2 knockout a result of four allele variants. All mutant alleles lead to premature stop-codons in the region encoding twinfilins’ N- terminal ADF-H domain. Numbering of nucleotide and amino acid sequences represent the first nucleotide and amino acid residues shown, respectively. All Western blot experiments were repeated three times. Unprocessed Western blots are provided in Source Data Extended Data Fig. 1.

Source data

Extended Data Fig. 2 Knockout of twinfilins does not affect cellular levels of actin and actin-binding proteins.

Identical amounts of lysates of wild-type and twf1/twf2 KO B16-F1 cells were loaded on SDS-PAGE gels and protein levels were determined with Western blot. a, Levels of β-actin. Anti-α-tubulin antibody was used as a loading control. We note that anti-β-actin antibody remained in the blot even after vigorous stripping of the membrane and was thus visible also in anti-α-tubulin detection. Protein levels of b, Capping Protein, c, cofilin-1, d, profilin-1, e, cyclase-associated protein-1, and f, CARMIL-1. Anti-α-tubulin or anti-histone H3 antibodies were used as a loading controls. All Western blot experiments were repeated three times, except in panel c where experiment was repeated two times. Unprocessed Western blots are provided in Source Data Extended Data Fig. 2.

Source data

Extended Data Fig. 3 Twinfilin-1/twinfilin-2 knockout leads to an accumulation of F-actin on endosomes at the perinuclear region.

a, Representative examples of phalloidin staining of wild-type B16-F1, and twf1/twf2-KO cells after 7.5 min uptake of 20 µg/ml AlexaFluor-647 transferrin. Scale bar = 20 µm. The dotted square indicates the perinuclear region magnified in the insert. b, Pearson’s correlation analysis of AlexaFluor-568 phalloidin and AlexaFluor-647 transferrin. Each data point represents individual cell (n = 19 for wild type and n = 21 for knockout cells) from one experiment with mean and standard deviations shown. c, Normalized F-actin intensities in transferrin-positive endosomes of wild-type and twf1/twf2-KO cells as measured from AlexaFluor-555 phalloidin stained cells after 7.5 min intake of 20 µg/ml AlexaFluor-647 phalloidin. The median, 25th and 75th percentiles and the minimum and maximum values of the data are shown. Numbers of measured cells were: B16-F1 wt = 2,518, twf1/2-KO-g3 = 3,637, twf1/2-KO-g3 + EGFP-TWF1 = 158 from two experiments. Statistical source data are provided in Source Data Extended Data Fig. 3.

Source data

Extended Data Fig. 4 Twinfilin slows down the barbed end depolymerization of ADP-actin filaments anchored to the glass bottom of open chambers.

The depolymerization of individual ADP-actin filaments was recorded during 8 minutes in the presence or absence of 2 µM TWF1 in F-buffer (see Methods). Filament surface anchoring induces pauses during depolymerization that need to be excluded from the analysis in order to quantify filament depolymerization rates. We note that the presence of twinfilin reduces the frequency of observable pauses. a, Depolymerization rate measurements excluding pauses are in agreement with measurements performed on single filaments in microfluidics experiments shown Fig. 3b (n = 30 filaments for both conditions, N = 2 biological repeats). The median, 25th and 75th percentiles and the minimum and maximum values of the data are shown. b, The global depolymerization rates, including pauses, are lower and very similar to the values reported from previous experiments in open chambers assays43,48,49 (n = 30 filaments for both conditions, N = 2 biological repeats). The median, 25th and 75th percentiles and the minimum and maximum values of the data are shown. c, Two representative kymographs showing the depolymerization of actin filaments in buffer (left), exhibiting a 3-minute-long pause, or in the presence of 2 µM TWF1 (right). d, Depolymerization of 3.4 µM actin filaments (5% pyrene-label) capped at their pointed ends with 0.5 µM tropomodulin-1 was followed by decrease in pyrene-actin fluorescence. In absence of TWF1 (black squares), no depolymerization was observed. Addition of 7 µM C-terminal ADF-H domain of TWF1, which binds ADP-actin monomers with nanomolar affinity, induces filament barbed end depolymerization (magenta circles) due to ADP-actin monomer sequestration. In the presence of 7 µM full-length TWF1, a slower actin filament depolymerization was detected (cyan triangles), indicating that full-length TWF1 slows down actin depolymerization by associating with filament barbed ends. A representative example from two independent experiments is shown. Statistical source data are provided in Source Data Extended Data Fig. 4.

Source data

Extended Data Fig. 5 CARMIL-1 increases CP dynamics in vitro.

a, Survival fraction of capped filament barbed ends, exposed to 1 µM G-actin with either 0 or 0.5 µM twinfilin, and stabilized by phalloidin or not. Survival fractions measured in depolymerization conditions are shown (from Fig. 6b) as comparison. Following label order, n = 480, 170, 32, 46, 31, 50 filaments from N = 8, 3, 1, 1, 1, 1 experiments. Filaments were exposed to rhodamine-phalloidin throughout the experiment, and were estimated to be >70% phalloidin-saturated, based on rhodamine fluorescence intensity. Fraction of uncapped filament barbed ends are shown as thick lines with 95% confidence intervals as shaded surfaces and with single exponential fits as thin lines. b, CARMIL and TWF1 accelerate uncapping in vitro, CARMIL being more efficient. Intermediate uncapping efficiency was detected, when both proteins were included in the assay. In the same order as figure labels, n = 480, 170, 60, 120, 120 filaments from N = 8, 3, 1, 2, 2 experiments. Fraction of uncapped filament barbed ends are shown as thick lines with 95% confidence intervals as shaded surfaces and with single exponential fits as thin lines. Statistical source data are provided in Source Data Extended Data Fig. 5.

Source data

Extended Data Fig. 6 CARMIL-1 cannot rescue CP localization and dynamics in twf1/twf2-knockout cells.

a, Representative examples of localization of EGFP-CARMIL1 in B16-F1 wild-type and twf1/twf2-KO cells. Scale bar = 10 µm. b, Line profile analysis of EGFP-CARMIL1 localization compared to AlexaFluor-568-phalloidin staining. Data represent mean of n = 13 (wild type) and n = 14 (knockout) cells from two experiments with error bars representing standard deviation. c, CARMIL-1 was depleted by siRNA in twf-1/twf-2 knockout cells, and the protein levels were detected by Western blot with anti-CARMIL-1 antibody. Histone H3 antibody was used as a loading control (left). Representative Western blot and immunofluorescence images from two experiments of twf1/twf2 KO and twf/twf2 KO + CARML-1 siRNA cells (right). Scale bar = 10 µm. d, Representative example of FRAP experiments of EGFP-CP dynamics in twf-1/twf-2 knockout cells over-expressing mCherry-CARMIL-1 (left). Scale bars = 5 µm. Recovery of EGFP-CP in knockout cells over-expressing mCherry-CARMIL-1 was compared to EGFP-CP recovery in wild-type and twf-1/twf-2 KO B16-F1 cells (data from Fig. 5) (right). Data represent the mean of n = 13 measurements from two experiments with shaded surfaces indicating standard deviations. Halftime of EGFP-CP fluorescence recovery in twf-1/twf-2 knockout cells overexpressing mCherry-CARMIL-1 was 14.58 s. Statistical source data are provided in Source Data Extended Data Fig. 6.

Source data

Extended Data Fig. 7 Overexpression of cofilin-1 does not rescue CP dynamics in twinfilin-deficient cells.

a, Representative example of EGFP-CP dynamics in lamellipodia of twf-1/twf-2 knockout cells overexpressing mCherry-cofilin-1 examined by a FRAP assay. Scale bars = 5 µm. b, Analysis of FRAP data from the mean of n = 9 individual measurements from two experiments with shaded surfaces indicating standard. Data of B16-F1 wild-type cells and twf-1/twf-2-knockout cells is the same as in Fig. 5. Half-time of EGFP-CP fluorescence recovery in twf-1/twf-2 knockout cells overexpressing mCherry-cofilin-1 was 20.95 s for bound fraction and 0.24 s for diffuse fraction. We note that longer observation time (60 s, see Fig. 5) is required for EGFP-CP recovery to reach the plateau. c, Western blot analysis of Cherry-cofilin-1 overexpression in twf1/twf2-KO B16-F1 cells. Anti-cofilin-1 antibody detection is shown. Based on band intensities in two experiments, we estimate 3-fold overexpression of cofilin-1 to normal levels. Unprocessed Western blot and statistical source data are provided in Source Data Extended Data Fig. 7.

Source data

Supplementary information

Reporting Summary

Supplementary Video 1

Lamellipodia protrusion of WT (left) and Twf1/Twf2-KO (right) B16-F1 cells on laminin-coated glass-bottom dishes imaged using DIC microscopy.

Supplementary Video 2

Representative examples of random migration of WT and Twf1/Twf2-KO-g3 B16-F1 cells imaged using phase-contrast microscopy. The tracks of cells used in the analysis are indicated by lines. Please note that tracking was stopped when cells divided or collided with each other.

Supplementary Video 3

Fluorescence recovery of eGFP–actin after photobleaching in WT (left) and Twf1/Twf2-KO (right) B16-F1 cells.

Supplementary Video 4

Fluorescence decay of PA–GFP–actin (left) after photoactivation in WT (top) and Twf1/Twf2-KO (middle) B16-F1 cells, and in Twf1/Twf2-KO cells expressing mCherry–TWF1 (rescue; bottom). Lamellipodia were marked with either mCherry–LifeAct or mCherry–TWF1 expression (right).

Supplementary Video 5

Fluorescence recovery of eGFP–TWF1 in a WT B16-F1 cell.

Supplementary Video 6

Fluorescence recovery of eGFP–CP in a WT B16-F1 cell.

Supplementary Video 7

Fluorescence recovery of eGFP–CP in a Twf1/Twf2-KO B16-F1 cell.

Supplementary Video 8

Fluorescence recovery of eGFP–CP (left) after photobleaching in WT (top) and Twf1/Twf2-KO (bottom) B16-F1 cells. Endosomal F-actin structures were visualized with coexpression of mCherry–LifeAct (right).

Supplementary Video 9

Fluorescence recovery of eGFP–CP (left) after photobleaching in Twf1/Twf2-KO B16-F1 cells overexpressing mCherry–CARMIL-1 (right).

Supplementary Video 10

Fluorescence recovery of eGFP–CP (left) after photobleaching in Twf1/Twf2-KO B16-F1 cells overexpressing mCherry–cofilin-1 (right).

Supplementary Video 11

Fluorescence recovery of eGFP–CP (left) after photobleaching in Twf1/Twf2-KO B16-F1 cells coexpressing mCherry–TWF1 WT (right).

Supplementary Video 12

Fluorescence recovery of eGFP–CP (left) after photobleaching in Twf1/Twf2-KO B16-F1 cells coexpressing mCherry–TWF1 tail mutant (F323A/K325A/K327A) (right).

Supplementary Video 13

Fluorescence recovery of eGFP–CP (left) after photobleaching in Twf1/Twf2-KO B16-F1 cells coexpressing mCherry–TWF1 ADF-H-domain mutant (R96A/K98A/R267A/R269A) (right).

Source data

Statistical Source Data Fig. 1

Statistical source data.

Statistical Source Data Fig. 2

Statistical source data.

Statistical Source Data Fig. 3

Statistical source data.

Statistical Source Data Fig. 4

Statistical source data.

Statistical Source Data Fig. 5

Statistical source data.

Statistical Source Data Fig. 6

Statistical source data.

Statistical Source Data Fig. 7

Statistical source data.

Statistical Source Data Fig. 8

Statistical source data.

Source Data Extended Data Fig. 1

Unprocessed western blots.

Source Data Extended Data Fig. 2

Unprocessed western blots.

Source Data Extended Data Fig. 3

Statistical source data.

Source Data Extended Data Fig. 4

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 6

Unprocessed western blots.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 7

Unprocessed western blots.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hakala, M., Wioland, H., Tolonen, M. et al. Twinfilin uncaps filament barbed ends to promote turnover of lamellipodial actin networks. Nat Cell Biol 23, 147–159 (2021). https://doi.org/10.1038/s41556-020-00629-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-020-00629-y

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