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:

A mechano-signalling network linking microtubules, myosin IIA filaments and integrin-based adhesions

A Publisher Correction to this article was published on 30 May 2019

This article has been updated

Abstract

The interrelationship between microtubules and the actin cytoskeleton in mechanoregulation of integrin-mediated adhesions is poorly understood. Here, we show that the effects of microtubules on two major types of cell-matrix adhesion, focal adhesions and podosomes, are mediated by KANK family proteins connecting the adhesion protein talin with microtubule tips. Both total microtubule disruption and microtubule uncoupling from adhesions by manipulations with KANKs trigger a massive assembly of myosin IIA filaments, augmenting focal adhesions and disrupting podosomes. Myosin IIA filaments are indispensable effectors in the microtubule-driven regulation of integrin-mediated adhesions. Myosin IIA filament assembly depends on Rho activation by the RhoGEF GEF-H1, which is trapped by microtubules when they are connected with integrin-mediated adhesions via KANK proteins but released after their disconnection. Thus, microtubule capture by integrin-mediated adhesions modulates the GEF-H1-dependent effect of microtubules on the assembly of myosin IIA filaments. Subsequent actomyosin reorganization then remodels the focal adhesions and podosomes, closing the regulatory loop.

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: Effects of microtubule uncoupling from integrin adhesions by manipulations with KANK proteins.
Fig. 2: KANK proteins are required for targeting microtubules to focal adhesions.
Fig. 3: Myosin II filaments mediate the effect of microtubules on integrin-based adhesions.
Fig. 4: Recovery of functional podosomes in KANK1-depleted cells on ROCK inhibition.
Fig. 5: Function of GEF-H1 in KANK-mediated regulation of integrin-based adhesions.
Fig. 6: Interplay between microtubules, myosin IIA filaments and integrin adhesions.

Similar content being viewed by others

Data availability

All data generated or analysed during this study are included in this published article (and its Supplementary Information files). Raw datasets generated during and/or analysed during the current study are available from the corresponding authors on reasonable request.

Code availability

Custom-written code used to analyse the data in the current study is available from the corresponding authors on reasonable request.

Change history

  • 30 May 2019

    An amendment to this paper has been published and can be accessed via a link at the top of the paper.

References

  1. Kasza, K. E. et al. The cell as a material. Curr. Opin. Cell Biol. 19, 101–107 (2007).

    Article  CAS  Google Scholar 

  2. Geiger, B., Spatz, J. P. & Bershadsky, A. D. Environmental sensing through focal adhesions. Nat. Rev. Mol. Cell Biol. 10, 21–33 (2009).

    Article  CAS  Google Scholar 

  3. Kanchanawong, P. et al. Nanoscale architecture of integrin-based cell adhesions. Nature 468, 580–584 (2010).

    Article  CAS  Google Scholar 

  4. Horton, E. R. et al. The integrin adhesome network at a glance. J. Cell Sci. 129, 4159–4163 (2016).

    Article  CAS  Google Scholar 

  5. Bershadsky, A., Chausovsky, A., Becker, E., Lyubimova, A. & Geiger, B. Involvement of microtubules in the control of adhesion-dependent signal transduction. Curr. Biol. 6, 1279–1289 (1996).

    Article  CAS  Google Scholar 

  6. Enomoto, T. Microtubule disruption induces the formation of actin stress fibers and focal adhesions in cultured cells: possible involvement of the rho signal cascade. Cell Struct. Funct. 21, 317–326 (1996).

    Article  CAS  Google Scholar 

  7. Ezratty, E. J., Partridge, M. A. & Gundersen, G. G. Microtubule-induced focal adhesion disassembly is mediated by dynamin and focal adhesion kinase. Nat. Cell Biol. 7, 581–590 (2005).

    Article  CAS  Google Scholar 

  8. Bouchet, B. P. et al. Talin-KANK1 interaction controls the recruitment of cortical microtubule stabilizing complexes to focal adhesions. eLife. 5, e18124 (2016).

    Article  Google Scholar 

  9. Sun, Z. et al. Kank2 activates talin, reduces force transduction across integrins and induces central adhesion formation. Nat. Cell Biol. 18, 941–953 (2016).

    Article  CAS  Google Scholar 

  10. Chen, N. P., Sun, Z. & Fassler, R. The Kank family proteins in adhesion dynamics. Curr. Opin. Cell Biol. 54, 130–136 (2018).

    Article  CAS  Google Scholar 

  11. van der Vaart, B. et al. CFEOM1-associated kinesin KIF21A is a cortical microtubule growth inhibitor. Dev. Cell 27, 145–160 (2013).

    Article  Google Scholar 

  12. Lansbergen, G. et al. CLASPs attach microtubule plus ends to the cell cortex through a complex with LL5beta. Dev. Cell 11, 21–32 (2006).

    Article  CAS  Google Scholar 

  13. Clohisey, S. M., Dzhindzhev, N. S. & Ohkura, H. Kank Is an EB1 interacting protein that localises to muscle-tendon attachment sites in Drosophila. PloS ONE 9, e106112 (2014).

    Article  Google Scholar 

  14. Murphy, D. A. & Courtneidge, S. A. The ‘ins’ and ‘outs’ of podosomes and invadopodia: characteristics, formation and function. Nat. Rev. Mol. Cell Biol. 12, 413–426 (2011).

    Article  CAS  Google Scholar 

  15. Cox, S. & Jones, G. E. Imaging cells at the nanoscale. Int. J. Biochem. Cell Biol. 45, 1669–1678 (2013).

    Article  CAS  Google Scholar 

  16. Yu, C. H. et al. Integrin-matrix clusters form podosome-like adhesions in the absence of traction forces. Cell Rep. 5, 1456–1468 (2013).

    Article  CAS  Google Scholar 

  17. Linder, S., Hufner, K., Wintergerst, U. & Aepfelbacher, M. Microtubule-dependent formation of podosomal adhesion structures in primary human macrophages. J. Cell Sci. 113, 4165–4176 (2000).

    CAS  Google Scholar 

  18. Meddens, M. B. et al. Actomyosin-dependent dynamic spatial patterns of cytoskeletal components drive mesoscale podosome organization. Nat. Commun. 7, 13127 (2016).

    Article  CAS  Google Scholar 

  19. Monypenny, J. et al. Role of WASP in cell polarity and podosome dynamics of myeloid cells. Eur. J. Cell Biol. 90, 198–204 (2011).

    Article  CAS  Google Scholar 

  20. Kaverina, I., Krylyshkina, O. & Small, J. V. Microtubule targeting of substrate contacts promotes their relaxation and dissociation. J. Cell Biol. 146, 1033–1044 (1999).

    Article  CAS  Google Scholar 

  21. Hu, S. et al. Long-range self-organization of cytoskeletal myosin II filament stacks. Nat. Cell Biol. 19, 133–141 (2017).

    Article  CAS  Google Scholar 

  22. Ito, S. et al. Induced cortical tension restores functional junctions in adhesion-defective carcinoma cells. Nat. Commun. 8, 1834 (2017).

    Article  Google Scholar 

  23. Vicente-Manzanares, M., Zareno, J., Whitmore, L., Choi, C. K. & Horwitz, A. F. Regulation of protrusion, adhesion dynamics, and polarity by myosins IIA and IIB in migrating cells. J. Cell Biol. 176, 573–580 (2007).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  25. Riveline, D. et al. Focal contacts as mechanosensors: externally applied local mechanical force induces growth of focal contacts by an mDia1-dependent and ROCK-independent mechanism. J. Cell Biol. 153, 1175–1186 (2001).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  27. Kakinuma, N., Roy, B. C., Zhu, Y., Wang, Y. & Kiyama, R. Kank regulates RhoA-dependent formation of actin stress fibers and cell migration via 14-3-3 in PI3K-Akt signaling. J. Cell Biol. 181, 537–549 (2008).

    Article  CAS  Google Scholar 

  28. Linder, S. The matrix corroded: podosomes and invadopodia in extracellular matrix degradation. Trends Cell Biol. 17, 107–117 (2007).

    Article  CAS  Google Scholar 

  29. Wang, Y. & McNiven, M. A. Invasive matrix degradation at focal adhesions occurs via protease recruitment by a FAK-p130Cas complex. J. Cell Biol. 196, 375–385 (2012).

    Article  CAS  Google Scholar 

  30. Krendel, M., Zenke, F. T. & Bokoch, G. M. Nucleotide exchange factor GEF-H1 mediates cross-talk between microtubules and the actin cytoskeleton. Nat. Cell Biol. 4, 294–301 (2002).

    Article  CAS  Google Scholar 

  31. Kwan, K. M. & Kirschner, M. W. A microtubule-binding Rho-GEF controls cell morphology during convergent extension of Xenopus laevis. Development 132, 4599–4610 (2005).

    Article  CAS  Google Scholar 

  32. Jiu, Y. et al. Vimentin intermediate filaments control actin stress fiber assembly through GEF-H1 and RhoA. J. Cell Sci. 130, 892–902 (2017).

    Article  CAS  Google Scholar 

  33. Ren, Y., Li, R., Zheng, Y. & Busch, H. Cloning and characterization of GEF-H1, a microtubule-associated guanine nucleotide exchange factor for Rac and Rho GTPases. J. Biol. Chem. 273, 34954–34960 (1998).

    Article  CAS  Google Scholar 

  34. Chang, Y. C., Nalbant, P., Birkenfeld, J., Chang, Z. F. & Bokoch, G. M. GEF-H1 couples nocodazole-induced microtubule disassembly to cell contractility via RhoA. Mol. Biol. Cell 19, 2147–2153 (2008).

    Article  CAS  Google Scholar 

  35. Gee, H. Y. et al. KANK deficiency leads to podocyte dysfunction and nephrotic syndrome. J. Clin. Invest. 125, 2375–2384 (2015).

    Article  Google Scholar 

  36. Portran, D., Schaedel, L., Xu, Z., Thery, M. & Nachury, M. V. Tubulin acetylation protects long-lived microtubules against mechanical ageing. Nat. Cell Biol. 19, 391–398 (2017).

    Article  CAS  Google Scholar 

  37. Xu, Z. et al. Microtubules acquire resistance from mechanical breakage through intralumenal acetylation. Science 356, 328–332 (2017).

    Article  CAS  Google Scholar 

  38. Samereier, M. et al. EB1 contributes to proper front-to-back polarity in neutrophil-like HL-60 cells. Eur. J. Cell Biol. 96, 143–153 (2017).

    Article  CAS  Google Scholar 

  39. Bershadsky, A., Kozlov, M. & Geiger, B. Adhesion-mediated mechanosensitivity: a time to experiment, and a time to theorize. Curr. Opin. Cell Biol. 18, 472–481 (2006).

    Article  CAS  Google Scholar 

  40. Oakes, P. W., Beckham, Y., Stricker, J. & Gardel, M. L. Tension is required but not sufficient for focal adhesion maturation without a stress fiber template. J. Cell Biol. 196, 363–374 (2012).

    Article  CAS  Google Scholar 

  41. Choi, C. K. et al. Actin and alpha-actinin orchestrate the assembly and maturation of nascent adhesions in a myosin II motor-independent manner. Nat. Cell Biol. 10, 1039–1050 (2008).

    Article  CAS  Google Scholar 

  42. Oakes, P. W. & Gardel, M. L. Stressing the limits of focal adhesion mechanosensitivity. Curr. Opin. Cell Biol. 30, 68–73 (2014).

    Article  CAS  Google Scholar 

  43. Rafiq, N. B. et al. Podosome assembly is controlled by the GTPase ARF1 and its nucleotide exchange factor ARNO. J. Cell Biol. 216, 181–197 (2017).

    Article  CAS  Google Scholar 

  44. Elliott, H. et al. Myosin II controls cellular branching morphogenesis and migration in three dimensions by minimizing cell-surface curvature. Nat. Cell Biol. 17, 137–147 (2015).

    Article  CAS  Google Scholar 

  45. Vasiliev, J. M. et al. Effect of colcemid on the locomotory behaviour of fibroblasts. J. Embryol. Exp. Morphol. 24, 625–640 (1970).

    CAS  Google Scholar 

  46. Small, J. V., Geiger, B., Kaverina, I. & Bershadsky, A. How do microtubules guide migrating cells? Nat. Rev. Mol. Cell Biol. 3, 957–964 (2002).

    Article  CAS  Google Scholar 

  47. Jones, G. E., Zicha, D., Dunn, G. A., Blundell, M. & Thrasher, A. Restoration of podosomes and chemotaxis in Wiskott-Aldrich syndrome macrophages following induced expression of WASp. Int. J. Biochem. Cell Biol. 34, 806–815 (2002).

    Article  CAS  Google Scholar 

  48. Ballestrem, C., Magid, N., Zonis, J., Shtutman, M. & Bershadsky, A. in Cell Motility (eds Ridley, A., Peckham, M. & Clark, P.) 75–99 (Wiley, 2004).

  49. Weng, S., Shao, Y., Chen, W. & Fu, J. Mechanosensitive subcellular rheostasis drives emergent single-cell mechanical homeostasis. Nat. Mater. 15, 961–967 (2016).

    Article  CAS  Google Scholar 

  50. Ballestrem, C., Hinz, B., Imhof, B. A. & Wehrle-Haller, B. Marching at the front and dragging behind: differential αVβ3-integrin turnover regulates focal adhesion behavior. J. Cell Biol. 155, 1319–1332 (2001).

    Article  CAS  Google Scholar 

  51. Rid, R., Schiefermeier, N., Grigoriev, I., Small, J. V. & Kaverina, I. The last but not the least: the origin and significance of trailing adhesions in fibroblastic cells. Cell Motil. Cytoskel. 61, 161–171 (2005).

    Article  Google Scholar 

  52. Pan, C. Q., Liou, Y.-c. & Low, B. C. Active Mek2 as a regulatory scaffold that promotes Pin1 binding to BPGAP1 to suppress BPGAP1-induced acute Erk activation and cell migration. J. Cell Sci. 123, 903–916 (2010).

    Article  CAS  Google Scholar 

  53. Sabass, B., Gardel, M. L., Waterman, C. M. & Schwarz, U. S. High resolution traction force microscopy based on experimental and computational advances. Biophys. J. 94, 207–220 (2008).

    Article  CAS  Google Scholar 

  54. Liu, J. et al. Talin determines the nanoscale architecture of focal adhesions. Proc. Natl Acad. Sci. USA 112, E4864–E4873 (2015).

    Article  CAS  Google Scholar 

  55. Zhang, Z., Nishimura, Y. & Kanchanawong, P. Extracting microtubule networks from superresolution single-molecule localization microscopy data. Mol. Biol. Cell 28, 333–345 (2017).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank A. Akhmanova (Utrecht University, Netherlands), R. Fäasler (Max Plank Institute for Biochemistry, Martinsried, Germany), C. M. Waterman (National Institutes of Health, USA), R. Zaidel-Bar (Mechanobiology Institute, Singapore) and M. Dodding for providing constructs used in this study. We are grateful to A. Akhmanova for useful discussions and constructive criticism. We thank D. Pitta de Araujo (MBI Science Communications Unit) for help with Fig. 6. This research is supported by the National Research Foundation, Prime Minister’s Office, Singapore, and the Ministry of Education under the Research Centres of Excellence programme through the Mechanobiology Institute, Singapore (ref no. R-714-006-006-271) (A.D.B., N.B.M.R., V.T. and M.N.) and Singapore Ministry of Education Academic Research Fund Tier 3 MOE grant no. MOE2016-T3-1-002 (A.D.B., Y.N.). A.D.B. also acknowledges support from a Maimonides Israeli–France grant (Israeli Ministry of Science Technology and Space) and EU Marie Skłodowska-Curie Network InCeM (project ID 642866) at the Weizmann Institute of Science. N.B.M.R. is also funded by a joint National University of Singapore–King’s College London graduate studentship. G.E.J. is supported by the Medical Research Council, UK (G1100041, MR/K015664) and the generous provision of a visiting professorship from the Mechanobiology Institute, Singapore. P.K. and Z.Z. are funded by the Ministry of Education Academic Research Fund Tier 2 (MOE-T2-1-124), the Mechanobiology Institute seed funding, the National Research Foundation Fellowship (NRF-NRFF-2011-04) and the National Research Foundation Competitive Research Programme (NRF2012NRF-CRP001-084).

Author information

Authors and Affiliations

Authors

Contributions

A.D.B. conceived and designed the project together with V.V., P.K. and G.E.J. N.B.M.R. and Y.N. equally designed and performed all experiments and prepared the manuscript; S.V.P., V.T., Z.Z., S.S. and M.N. provided assistance in carrying out experiments and discussed results. A.D.B., N.B.M.R. and Y.N., together with V.V., P.K. and G.E.J., discussed results and prepared the manuscript.

Corresponding authors

Correspondence to Pakorn Kanchanawong, Gareth E. Jones or Alexander D. Bershadsky.

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.

Supplementary information

Supplementary Information

Supplementary Notes, Supplementary Figs. 1–13, Supplementary Table 1, Supplementary Video Legends 1–17, Supplementary Refs. 1–39

Reporting Summary

Supplementary Video 1

The microtubule plus ends of an HT1080 cell transfected with control siRNA are accumulated at focal adhesions concentrated within adhesive islands

Supplementary Video 2

The microtubule plus ends were randomly distributed over adhesive islands and spaces between them in an HT1080 cell depleted of KANK1 and KANK2

Supplementary Video 3

Microtubule outgrowth induced transient disassembly of focal adhesions in an HT1080 cell

Supplementary Video 4

Rapamycin-induced linking of two parts of the KANK1 molecule triggered disassembly of focal adhesions in an HT1080 cell

Supplementary Video 5

Microtubule disruption triggered massive assembly of myosin II filaments and disassembly of podosomes in a THP-1 cell

Supplementary Video 6

Microtubule disruption triggered massive assembly of myosin II filaments and augmentation of focal adhesion in an HT1080 cell

Supplementary Video 7

Microtubule outgrowth induced disassembly of myosin II filaments in an HT1080 cell

Supplementary Video 8

Transient decrease in traction forces after microtubule outgrowth in an HT1080 cell

Supplementary Video 9

KANK2 knockdown suppressed disassembly of myosin II filaments induced by microtubule outgrowth in an HT1080 cell

Supplementary Video 10

Activation of RhoA by CN03 augmented myosin II filament formation and podosome disruption in a THP-1 cell

Supplementary Video 11

Dynamics of podosomes and myosin II filaments in a control THP-1 cell

Supplementary Video 12

ROCK inhibition by Y-27632 did not affect podosome dynamics in a THP-1 cell

Supplementary Video 13

Inhibition of ROCK by Y-27632 disrupted myosin II filaments and prevented nocodazole-induced disassembly of podosomes in a THP-1 cell

Supplementary Video 14

Disruption of myosin II filaments by Y-27632 resulted in recovery of podosomes in a THP-1 cell pretreated with nocodazole

Supplementary Video 15

Disruption of myosin II filaments by Y-27632 resulted in recovery of podosomes in a KANK1-depleted THP-1 cell

Supplementary Video 16

Constitutively active RhoA and ROCK prevented the disruption of focal adhesions on microtubule outgrowth in HT1080 cells

Supplementary Video 17

GEF-H1 knockdown suppressed disruption of myosin II filaments and prevented disassembly of focal adhesions on microtubule outgrowth in an HT1080 cell

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Rafiq, N.B.M., Nishimura, Y., Plotnikov, S.V. et al. A mechano-signalling network linking microtubules, myosin IIA filaments and integrin-based adhesions. Nat. Mater. 18, 638–649 (2019). https://doi.org/10.1038/s41563-019-0371-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41563-019-0371-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