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:

Compressive forces stabilize microtubules in living cells

Abstract

Microtubules are cytoskeleton components with unique mechanical and dynamic properties. They are rigid polymers that alternate phases of growth and shrinkage. Nonetheless, the cells can display a subset of stable microtubules, but it is unclear whether microtubule dynamics and mechanical properties are related. Recent in vitro studies suggest that microtubules have mechano-responsive properties, being able to stabilize their lattice by self-repair on physical damage. Here we study how microtubules respond to cycles of compressive forces in living cells and find that microtubules become distorted, less dynamic and more stable. This mechano-stabilization depends on CLASP2, which relocates from the end to the deformed shaft of microtubules. This process seems to be instrumental for cell migration in confined spaces. Overall, these results demonstrate that microtubules in living cells have mechano-responsive properties that allow them to resist and even counteract the forces to which they are subjected, being a central mediator of cellular mechano-responses.

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: MTs are deformed and stabilized in response to SCC in RPE1 cells.
Fig. 2: MTs are stabilized in response to SCC in enucleated RPE1 cells (cytoplasts).
Fig. 3: Characteristic timescales of MT mechano-stabilization.
Fig. 4: CLASP2 is enriched along the MT shaft in response to SCC.
Fig. 5: CLASP2 supports MT mechano-stabilization.
Fig. 6: Cell migration through constrictions depends on the mechano-stabilization of MTs.

Similar content being viewed by others

Data availability

Raw data are available from the corresponding authors upon request. Source data are provided with this paper.

Code availability

The computational code for image and data analysis is available via figshare at https://doi.org/10.6084/m9.figshare.22295881.v1.

References

  1. Heisenberg, C.-P. P. & Bellaïche, Y. Forces in tissue morphogenesis and patterning. Cell 153, 948–962 (2013).

    Article  CAS  Google Scholar 

  2. Akhmanova, A. & Kapitein, L. C. Mechanisms of microtubule organization in differentiated animal cells. Nat. Rev. Mol. Cell Biol. 23, 541–558 (2022).

    Article  CAS  Google Scholar 

  3. Hamant, O., Inoue, D., Bouchez, D., Dumais, J. & Mjolsness, E. Are microtubules tension sensors? Nat. Commun. 10, 2360 (2019).

    Article  Google Scholar 

  4. Gittes, F., Mickey, B., Nettleton, J. & Howard, J. Flexural rigidity of microtubules and actin filaments measured from thermal fluctuations in shape. J. Cell Biol. 120, 923–934 (1993).

    Article  CAS  Google Scholar 

  5. Garzon-Coral, C., Fantana, H. A. & Howard, J. A force-generating machinery maintains the spindle at the cell center during mitosis. Science 352, 1124–1127 (2016).

    Article  CAS  Google Scholar 

  6. Brangwynne, C. P. et al. Microtubules can bear enhanced compressive loads in living cells because of lateral reinforcement. J. Cell Biol. 173, 733–741 (2006).

    Article  CAS  Google Scholar 

  7. Brouhard, G. J. & Rice, L. M. Microtubule dynamics: an interplay of biochemistry and mechanics. Nat. Rev. Mol. Cell Biol. 19, 451–463 (2018).

  8. Akhmanova, A. & Steinmetz, M. O. Control of microtubule organization and dynamics: two ends in the limelight. Nat. Rev. Mol. Cell Biol. 16, 711–726 (2015).

    Article  CAS  Google Scholar 

  9. Komarova, Y. A., Akhmanova, A., Kojima, S. I., Galjart, N. & Borisy, G. G. Cytoplasmic linker proteins promote microtubule rescue in vivo. J. Cell Biol. 159, 589–599 (2002).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  11. Kaverina, I. et al. Tensile stress stimulates microtubule outgrowth in living cells. J. Cell Sci. 115, 2283–2291 (2002).

    Article  CAS  Google Scholar 

  12. Colin, L. et al. Cortical tension overrides geometrical cues to orient microtubules in confined protoplasts. Proc. Natl Acad. Sci. USA 117, 32731–32738 (2020).

    Article  CAS  Google Scholar 

  13. Janson, M. E. et al. Dynamic instability of microtubules is regulated by force. J. Cell Biol. 161, 1029–1034 (2003).

    Article  CAS  Google Scholar 

  14. 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 

  15. Rafiq, N. B. M. et al. A mechano-signalling network linking microtubules, myosin IIA filaments and integrin-based adhesions. Nat. Mater. 18, 638–649 (2019).

    Article  CAS  Google Scholar 

  16. Schaedel, L. et al. Microtubules self-repair in response to mechanical stress. Nat. Mater. 14, 1156–1163 (2015).

    Article  CAS  Google Scholar 

  17. Aumeier, C. et al. Self-repair promotes microtubule rescue. Nat. Cell Biol. 18, 1054–1064 (2016).

    Article  CAS  Google Scholar 

  18. Théry, M. & Blanchoin, L. Microtubule self-repair. Curr. Opin. Cell Biol. 68, 144–154 (2021).

    Article  Google Scholar 

  19. Faust, U. et al. Cyclic stress at mHz frequencies aligns fibroblasts in direction of zero strain. PLoS ONE 6, e28963 (2011).

    Article  CAS  Google Scholar 

  20. Janke, C. & Magiera, M. M. The tubulin code and its role in controlling microtubule properties and functions. Nat. Rev. Mol. Cell Biol. 21, 307–326 (2020).

    Article  CAS  Google Scholar 

  21. Vasquez, R. J., Howell, B., Yvon, A. M. C., Wadsworth, P. & Cassimeris, L. Nanomolar concentrations of nocodazole alter microtubule dynamic instability in vivo and in vitro. Mol. Biol. Cell 8, 973–985 (1997).

    Article  CAS  Google Scholar 

  22. 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  Google Scholar 

  23. Livne, A., Bouchbinder, E. & Geiger, B. Cell reorientation under cyclic stretching. Nat. Commun. 5, 3938 (2014).

    Article  CAS  Google Scholar 

  24. Jungbauer, S., Gao, H., Spatz, J. P. & Kemkemer, R. Two characteristic regimes in frequency-dependent dynamic reorientation of fibroblasts on cyclically stretched substrates. Biophys. J. 95, 3470–3478 (2008).

    Article  CAS  Google Scholar 

  25. Bernal, R., Hemelryck, M., Van, Gurchenkov, B. & Cuvelier, D. Actin stress fibers response and adaptation under stretch. Int. J. Mol. Sci. 23, 5095 (2022).

    Article  CAS  Google Scholar 

  26. Alam, S. G. et al. The nucleus is an intracellular propagator of tensile forces in NIH 3T3 fibroblasts. J. Cell Sci. 128, 1901–1911 (2015).

    Article  CAS  Google Scholar 

  27. Cadot, B. et al. Nuclear movement during myotube formation is microtubule and dynein dependent and is regulated by Cdc42, Par6 and Par3. EMBO Rep. 13, 741–749 (2012).

    Article  CAS  Google Scholar 

  28. Jimenez, A. J. et al. Acto-myosin network geometry defines centrosome position. Curr. Biol. 31, 1206–1220.e5 (2021).

    Article  CAS  Google Scholar 

  29. Webster, D. R., Gundersen, G. G., Bulinski, J. C. & Borisy, G. G. Differential turnover of tyrosinated and detyrosinated microtubules. Proc. Natl Acad. Sci. USA 84, 9040–9044 (1987).

    Article  CAS  Google Scholar 

  30. Reid, T. A. et al. Structural state recognition facilitates tip tracking of EB1 at growing microtubule ends. eLife 8, e48117 (2019).

    Article  CAS  Google Scholar 

  31. Aher, A. et al. CLASP mediates microtubule repair by restricting lattice damage and regulating tubulin incorporation. Curr. Biol. 30, 2175–2183.e6 (2020).

    Article  CAS  Google Scholar 

  32. Wittmann, T. & Waterman-Storer, C. M. Spatial regulation of CLASP affinity for microtubules by Rac1 and GSK3β in migrating epithelial cells. J. Cell Biol. 169, 929–939 (2005).

    Article  CAS  Google Scholar 

  33. Xu, T. et al. SOAX: a software for quantification of 3D biopolymer networks. Sci. Rep. 5, 9081 (2015).

    Article  Google Scholar 

  34. Lawrence, E. J., Arpag, G., Norris, S. R. & Zanic, M. Human CLASP2 specifically regulates microtubule catastrophe and rescue. Mol. Biol. Cell 29, 1168–1177 (2018).

    Article  CAS  Google Scholar 

  35. Drabek, K. et al. Role of CLASP2 in microtubule stabilization and the regulation of persistent motility. Curr. Biol. 16, 2259–2264 (2006).

    Article  CAS  Google Scholar 

  36. Mimori-Kiyosue, Y. et al. CLASP1 and CLASP2 bind to EB1 and regulate microtubule plus-end dynamics at the cell cortex. J. Cell Biol. 168, 141–153 (2005).

    Article  CAS  Google Scholar 

  37. Thiam, H. R. et al. Perinuclear Arp2/3-driven actin polymerization enables nuclear deformation to facilitate cell migration through complex environments. Nat. Commun. 7, 10997 (2016).

    Article  CAS  Google Scholar 

  38. Cross, R. A. Microtubule lattice plasticity. Curr. Opin. Cell Biol. 56, 88–93 (2019).

    Article  CAS  Google Scholar 

  39. Webster, D. R. & Borisy, G. G. Microtubules are acetylated in domains that turn over slowly. J. Cell Sci. 92, 57–65 (1989).

    Article  Google Scholar 

  40. Ambrose, C., Allard, J. F., Cytrynbaum, E. N. & Wasteneys, G. O. A CLASP-modulated cell edge barrier mechanism drives cell-wide cortical microtubule organization in Arabidopsis. Nat. Commun. 2, 430 (2011).

    Article  Google Scholar 

  41. Bouchet, B. P. & Akhmanova, A. Microtubules in 3D cell motility. J. Cell Sci. 130, 39–50 (2017).

    Article  CAS  Google Scholar 

  42. Wyatt, T. P. J. et al. Actomyosin controls planarity and folding of epithelia in response to compression. Nat. Mater. 19, 109–117 (2020).

    Article  CAS  Google Scholar 

  43. Uchida, K., Scarborough, E. A. & Prosser, B. L. Cardiomyocyte microtubules: control of mechanics, transport, and remodeling. Annu. Rev. Physiol. 84, 257–283 (2022).

    Article  Google Scholar 

  44. Robison, P. et al. Detyrosinated microtubules buckle and bear load in contracting cardiomyocytes. Science 352, aaf0659 (2016).

    Article  Google Scholar 

  45. Chen, C. Y. et al. Suppression of detyrosinated microtubules improves cardiomyocyte function in human heart failure. Nat. Med. 24, 1225–1233 (2018).

    Article  CAS  Google Scholar 

  46. Liu, Y. J. et al. Confinement and low adhesion induce fast amoeboid migration of slow mesenchymal cells. Cell 160, 659–672 (2015).

    Article  CAS  Google Scholar 

  47. Delarue, M. et al. Compressive stress inhibits proliferation in tumor spheroids through a volume limitation. Biophys. J. 107, 1821–1828 (2014).

    Article  CAS  Google Scholar 

  48. Nam, S. et al. Cell cycle progression in confining microenvironments is regulated by a growth-responsive TRPV4-PI3K/Akt-p27Kip1 signaling axis. Sci. Adv. 5, eaaw6171 (2019).

    Article  CAS  Google Scholar 

  49. Tse, J. M. et al. Mechanical compression drives cancer cells toward invasive phenotype. Proc. Natl Acad. Sci. USA 109, 911–916 (2012).

    Article  CAS  Google Scholar 

  50. Lacroix, B. et al. In situ imaging in C. elegans reveals developmental regulation of microtubule dynamics. Dev. Cell 29, 203–216 (2014).

    Article  CAS  Google Scholar 

  51. Azioune, A. et al. Robust method for high-throughput surface patterning of deformable substrates. Langmuir 27, 7349–7352 (2011).

    Article  CAS  Google Scholar 

  52. Iguiñiz, N., Frisenda, R., Bratschitsch, R. & Castellanos-Gomez, A. Revisiting the buckling metrology method to determine the Young’s modulus of 2D materials. Adv. Mater. 31, 1807150 (2019).

    Article  Google Scholar 

  53. Aillaud, C. et al. Evidence for new C-terminally truncated variants of α- and β-tubulins. Mol. Biol. Cell 27, 640–653 (2016).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  55. Akhmanova, A. et al. CLASPs are CLIP-115 and -170 associating proteins involved in the regional regulation of microtubule dynamics in motile fibroblasts. Cell 104, 923–935 (2001).

    Article  CAS  Google Scholar 

  56. Chandrakar, P. et al. Confinement controls the bend instability of three-dimensional active liquid crystals. Phys. Rev. Lett. 125, 257801 (2020).

    Article  CAS  Google Scholar 

  57. Püspöki, Z., Storath, M., Sage, D. & Unser, M. Transforms and operators for directional bioimage analysis: a survey. Adv. Anat. Embryol. Cell Biol. 219, 69–93 (2016).

    Article  Google Scholar 

  58. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  CAS  Google Scholar 

  59. Sage, D., Prodanov, D., Tinevez, J.-Y. & Schindelin, J. MIJ: making interoperablility between ImageJ and Matlab possible. In ImageJ User and Developer Conference 2426 (2012).

  60. Tinevez, J.-Y. et al. TrackMate: an open and extensible platform for single-particle tracking. Methods 115, 80–90 (2017).

    Article  CAS  Google Scholar 

  61. Mardia, K. V. & Jupp, P. E. Statistics of Directional Data 2nd edn (John Wiley & Sons, 2000).

  62. Ruhnow, F., Zwicker, D. & Diez, S. Tracking single particles and elongated filaments with nanometer precision. Biophys. J. 100, 2820–2828 (2011).

    Article  CAS  Google Scholar 

  63. Girão, H. et al. CLASP2 binding to curved microtubule tips promotes flux and stabilizes kinetochore attachments. J. Cell Biol. 219, e201905080 (2020).

    Google Scholar 

  64. Reth, M. Matching cellular dimensions with molecular sizes. Nat. Immunol. 14, 765–767 (2013).

    Article  CAS  Google Scholar 

  65. Mikhaylova, M. et al. Resolving bundled microtubules using anti-tubulin nanobodies. Nat. Commun. 6, 7933 (2015).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by the European Research Council (Consolidator Grant 771599 (ICEBERG) to M.T. and Advanced Grant 741773 (AAA) to L.B.), by the Bettencourt-Schueller Foundation, the Emergence program of the Ville de Paris and the Schlumberger Foundation for education and research. This project was also supported by the MuLife imaging facility, which is funded by GRAL, a program from the Chemistry Biology Health Graduate School of University Grenoble Alpes (ANR-17-EURE-0003). The work of D.M.R. and D.V. was supported by a grant from the National Institute of Health (R35GM136372). A.A. was supported by the Netherlands Organisation for Scientific Research (NWO) ECHO Grant 711.018.004. G.G. was supported by the INCA (AAP PLBIO no. 2020-109) and by the French National Research Agency (ANR-21-CE11-0004-01). M.D. was supported by the Fondation pour la Recherche Médicale (SPF201809007121).

Author information

Authors and Affiliations

Authors

Contributions

Y.L., M.T. and L.B. conceived the study and designed the overall experiments. Y.L., D.R. and F.N.V. conducted the experiments. D.C., M.D., T.P., M.P., G.G., D.V. and A.A. provided the materials and shared the methods. Y.L., O.K. and D.M.R. analysed the data. Y.L., M.T. and L.B. wrote the Article. All the authors reviewed, edited and approved the paper.

Corresponding authors

Correspondence to Laurent Blanchoin or Manuel Théry.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Materials thanks the anonymous reviewers for their contribution to the peer review of this work.

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 Figs. 1–10 and legends to Videos 1–5.

Reporting Summary

Supplementary Video 1

Real-time capture of a 12-s-long 10% SCC. In most of our experiments in this study, the cells were subjected to 10 cycles.

Supplementary Video 2

RPE1 cells were transfected to express GFP-EB1. The cells were imaged on a spinning-disc confocal microscope with a ×63/1.4 objective. The images were taken every second for 2 min. The video is displayed at 20 images per second, that is, ×20 acceleration. The same cell was recorded before (left images) and after (right images) 12 SCC. The bottom images show the overlay of the top images to reveal the EB1 trajectories.

Supplementary Video 3

RPE1 cells were transfected to express GFP-EB3. The cells were imaged on a spinning-disc confocal video microscope with a ×63/1.4 objective. The images were taken every second for 4 min. The video is displayed at 15 images per second, that is, ×15 acceleration. The images are displayed with a cyan look-up table.

Supplementary Video 4

WT and CLASP2−/− cells were treated with SiR-tubulin to reveal MTs and imaged on a spinning-disc confocal video microscope with a ×63/1.4 objective. The images were taken every 15 s during 15 min. The video is displayed at 10 images per second, that is, ×150 acceleration.

Supplementary Video 5

WT and CLASP2−/− cells were treated with Hoechst to visualize their nuclei and imaged in transmitted light and with fluorescence excitation through a ×20 objective. The images were taken every 10 min for 12 h. The video is displayed at 10 images per second, that is, ×6,000 acceleration. The positions of the nuclei were tracked using a TrackMate plug-in for Fiji.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 2

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 5a

Unprocessed western blots.

Source Data Fig. 6

Statistical source data.

Source Data Supplementary Fig. 2

Statistical source data.

Source Data Supplementary Fig. 3

Statistical source data.

Source Data Supplementary Fig. 4

Statistical source data.

Source Data Supplementary Fig. 5

Statistical source data.

Source Data Supplementary Fig. 6

Statistical source data.

Source Data Supplementary Fig. 8

Statistical source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Li, Y., Kučera, O., Cuvelier, D. et al. Compressive forces stabilize microtubules in living cells. Nat. Mater. 22, 913–924 (2023). https://doi.org/10.1038/s41563-023-01578-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41563-023-01578-1

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