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:

Self-repair protects microtubules from destruction by molecular motors

Abstract

Microtubule instability stems from the low energy of tubulin dimer interactions, which sets the growing polymer close to its disassembly conditions. Molecular motors use ATP hydrolysis to produce mechanical work and move on microtubules. This raises the possibility that the mechanical work produced by walking motors can break dimer interactions and trigger microtubule disassembly. We tested this hypothesis by studying the interplay between microtubules and moving molecular motors in vitro. Our results show that molecular motors can remove tubulin dimers from the lattice and rapidly destroy microtubules. We also found that dimer removal by motors was compensated for by the insertion of free tubulin dimers into the microtubule lattice. This self-repair mechanism allows microtubules to survive the damage induced by molecular motors as they move along their tracks. Our study reveals the existence of coupling between the motion of molecular motors and the renewal of the microtubule lattice.

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: Gliding assays destroy non-stabilized microtubules.
Fig. 2: Motility assays destroy non-stabilized microtubules.
Fig. 3: Single dyneins can damage the microtubule lattice.
Fig. 4: Free tubulin dimers prevent microtubule destruction by kinesin and dynein.
Fig. 5: Molecular motors enhance tubulin exchange in the microtubule lattice.

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.

References

  1. Drummond, D. R. Regulation of microtubule dynamics by kinesins. Semin. Cell Dev. Biol. 22, 927–934 (2011).

    Article  CAS  Google Scholar 

  2. Hibbel, A. et al. Kinesin Kip2 enhances microtubule growth in vitro through length-dependent feedback on polymerization and catastrophe. Elife 4, e10542 (2015).

    Article  Google Scholar 

  3. Laan, L. et al. Cortical dynein controls microtubule dynamics to generate pulling forces that position microtubule asters. Cell 148, 502–514 (2012).

    Article  CAS  Google Scholar 

  4. Hunter, A. W. et al. The kinesin-related protein MCAK is a microtubule depolymerase that forms an ATP-hydrolyzing complex at microtubule ends. Mol. Cell 11, 445–457 (2003).

    Article  CAS  Google Scholar 

  5. Arellano-Santoyo, H. et al. A tubulin binding switch underlies Kip3/kinesin-8 depolymerase activity. Dev. Cell 42, 37–51.e8 (2017).

    Article  CAS  Google Scholar 

  6. Schaedel, L. et al. Lattice defects induce microtubule self-renewal. Nat. Phys. 15, 830–838 (2019).

    Article  CAS  Google Scholar 

  7. Peet, D. R., Burroughs, N. J. & Cross, R. A. Kinesin expands and stabilizes the GDP-microtubule lattice. Nat. Nanotechnol. 13, 386–391 (2018).

    Article  CAS  Google Scholar 

  8. Shima, T. et al. Kinesin-binding–triggered conformation switching of microtubules contributes to polarized transport. J. Cell Biol. 217, 4164–4183 (2018).

    Article  CAS  Google Scholar 

  9. Dumont, E. L. P., Do, C. & Hess, H. Molecular wear of microtubules propelled by surface-adhered kinesins. Nat. Nanotechnol. 10, 166–169 (2015).

    Article  CAS  Google Scholar 

  10. VanDelinder, V., Adams, P. G. & Bachand, G. D. Mechanical splitting of microtubules into protofilament bundles by surface-bound kinesin-1. Sci. Rep. 6, 39408 (2016).

    Article  CAS  Google Scholar 

  11. Howard, J., Hudspeth, A. J. & Vale, R. D. Movement of microtubules by single kinesin molecules. Nature 342, 154–158 (1989).

    Article  CAS  Google Scholar 

  12. Vale, R. D. et al. Direct observation of single kinesin molecules moving along microtubules. Nature 380, 451–453 (1996).

    Article  CAS  Google Scholar 

  13. Portran, D., Gaillard, J., Vantard, M. & Théry, M. Quantification of MAP and molecular motor activities on geometrically controlled microtubule networks. Cytoskeleton 70, 12–23 (2013).

    Article  CAS  Google Scholar 

  14. Carazo-Salas, R. E., Antony, C. & Nurse, P. The kinesin Klp2 mediates polarization of interphase microtubules in fission yeast. Science 309, 297–300 (2005).

    Article  CAS  Google Scholar 

  15. Braun, M., Drummond, D. R., Cross, R. A. & McAinsh, A. D. The kinesin-14 Klp2 organizes microtubules into parallel bundles by an ATP-dependent sorting mechanism. Nat. Cell Biol. 11, 724–730 (2009).

    Article  CAS  Google Scholar 

  16. Guo, H. et al. Mechanism and dynamics of breakage of fluorescent microtubules. Biophys. J. 90, 2093–2098 (2006).

    Article  CAS  Google Scholar 

  17. Vigers, G. P., Coue, M. & McIntosh, J. R. Fluorescent microtubules break up under illumination. J. Cell Biol. 107, 1011–1024 (1988).

    Article  CAS  Google Scholar 

  18. Mahamdeh, M., Simmert, S., Luchniak, A., Schäffer, E. & Howard, J. Label-free high-speed wide-field imaging of single microtubules using interference reflection microscopy. J. Microsc. 272, 60–66 (2018).

    Article  CAS  Google Scholar 

  19. Lasek, R. J. & Brady, S. T. Attachment of transported vesicles to microtubules in axoplasm is facilitated by AMP-PNP. Nature 316, 645–647 (1985).

    Article  CAS  Google Scholar 

  20. Braun, M. et al. The human kinesin-14 HSET tracks the tips of growing microtubules in vitro. Cytoskeleton 70, 515–521 (2013).

    Article  CAS  Google Scholar 

  21. Jonsson, E., Yamada, M., Vale, R. D. & Goshima, G. Clustering of a kinesin-14 motor enables processive retrograde microtubule-based transport in plants. Nat. Plants 1, 15087 (2015).

    Article  CAS  Google Scholar 

  22. DeSantis, M. E. et al. Lis1 has two opposing modes of regulating cytoplasmic dynein. Cell 170, 1197–1208.e12 (2017).

    Article  CAS  Google Scholar 

  23. Reck-Peterson, S. L. et al. Single-molecule analysis of dynein processivity and stepping behavior. Cell 126, 335–348 (2006).

    Article  CAS  Google Scholar 

  24. Block, S. M. Kinesin motor mechanics: binding, stepping, tracking, gating, and limping. Biophys. J. 92, 2986–2995 (2007).

    Article  CAS  Google Scholar 

  25. Dye, R. B., Flicker, P. F., Lien, D. Y. & Williams, R. C. End-stabilized microtubules observed in vitro: stability, subunit, interchange, and breakage. Cell Motil. Cytoskeleton 21, 171–186 (1992).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  27. Gennerich, A., Carter, A. P., Reck-Peterson, S. L. & Vale, R. D. Force-induced bidirectional stepping of cytoplasmic dynein. Cell 131, 952–965 (2007).

    Article  CAS  Google Scholar 

  28. Dimitrov, A. et al. Detection of GTP-tubulin conformation in vivo reveals a role for GTP remnants in microtubule rescues. Science 322, 1353–1356 (2008).

    Article  CAS  Google Scholar 

  29. de Forges, H. et al. Localized mechanical stress promotes microtubule rescue. Curr. Biol. 26, 3399–3406 (2016).

    Article  Google Scholar 

  30. 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 

  31. Nakata, T., Niwa, S., Okada, Y., Perez, F. & Hirokawa, N. Preferential binding of a kinesin-1 motor to GTP-tubulin-rich microtubules underlies polarized vesicle transport. J. Cell Biol. 194, 245–255 (2011).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  33. Vemu, A. et al. Severing enzymes amplify microtubule arrays through lattice GTP-tubulin incorporation. Science 361, eaau1504 (2018).

    Article  Google Scholar 

Download references

Acknowledgements

This project benefited from several funding sources: European Research Council (ERC) Advanced 741773 (AAA) to L.B., ERC Consolidator 771599 (ICEBERG) to M.T.; Agence Nationale de la Recherche (ANR)-18-CE13-0001 to K.J. and M.T., Howard Hughes Medical Institute to S.L.R.-P and IRTELIS PhD programme from the CEA to S.T. Our imaging platform is supported by the Laboratory of Excellence Grenoble Alliance for Integrated Structural & Cell Biology (LabEX GRAL) (ANR-10-LABX-49-01) and the University Grenoble Alpes graduate school (Ecoles Universitaires de Recherche, CBH-EUR-GS, ANR-17-EURE-0003). We thank S. Diez (TU Dresden) for interesting discussions and sharing reagents.

Author information

Authors and Affiliations

Authors

Contributions

S.T. performed all experiments with motility assays; D.I. performed all experiments with gliding assays; J.G. purified and labelled tubulin and assisted S.T. and D.I. for all experiments; Z.M.H., M.E.D. and S.L.R.-P. purified dyneins; D.P., E.D. and C.L. discussed the founding hypothesis of the project; C.A. and L.S. performed preliminary experiments; K.J. analysed data; and L.B. and M.T. designed and directed the project.

Corresponding authors

Correspondence to Laurent Blanchoin or Manuel Théry.

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 Methods, references, Figs. 1–6, Tables 1–14 and video legends 1–12.

Reporting Summary

Supplementary Video 1

Gliding assay of stabilized microtubules. Taxol-treated (left) and GMPCPP-microtubules (right), labelled with 20% fluorescent tubulin, gliding on a layer of kinesin-1. Images were taken with a ×20 objective in epifluorescence microscopy. Acquisition frame rate, 1 image every 5 seconds over 15 minutes. Display, 25 images s–1. Scale bar, 100 µm.

Supplementary Video 2

Gliding assay of non-stabilized capped GDP-microtubules. Capped GDP-microtubules, labelled with 20% fluorescent tubulin, gliding on a layer of kinesin-1. Images were taken with a ×20 objective in epifluorescence microscopy. Acquisition frame rate, 1 image every 5 seconds over 15 minutes. Display, 25 images s–1. Scale bar, 100 µm.

Supplementary Video 3

Microtubule breakage during gliding. Capped GDP-microtubules, labelled with 20% fluorescent tubulin, gliding on a layer of kinesin-1. Microtubules were automatically detected and tracked in order to reposition the visualization field so as to keep the moving microtubule at the centre of the field. Images were taken with a ×63 objective in TIRF microscopy with ×1.5 magnifier. Acquisition frame rate, 1 image every 5 seconds over 15 minutes. Display, 5 images s–1. Scale bar, 20 µm.

Supplementary Video 4

Microtubule breakage upon kinesin motor walk. Capped GDP-microtubules, labelled with 20% fluorescent tubulin, were assembled from micropatterned seeds and served as tracks for the motility of kinesins (10 nM Klp2). Images were taken with a ×63 objective in TIRF microscopy with ×1.5 magnifier. Acquisition frame rate, 1 image every 3 seconds over 3 minutes. Display, 5 images s–1. Scale bar, 10 µm.

Supplementary Video 5

Microtubule breakage versus uncapping upon dynein motor walk. Capped GDP-microtubules, labelled with 20% fluorescent tubulin, were assembled from micropatterned seeds and served as tracks for the motility of kinesins (1 nM Klp2). Images were taken with a ×63 objective in TIRF microscopy with ×1.5 magnifier. Acquisition frame rate, 1 image every 3 seconds over 3 minutes. Display, 3 images s–1. Scale bar, 10 µm.

Supplementary Video 6

Single dynein motors walking on a capped microtubule. Capped GDP-microtubules, labelled with 20% fluorescent tubulin, were assembled from micropatterned seeds and served as tracks for the motility of dyneins (50 pM). Images were taken with a ×63 objective in TIRF microscopy with ×1.5 magnifier. Acquisition frame rate, 1 image every 3 seconds over 2 minutes. Display, 10 images s–1. Scale bar, 10 µm.

Supplementary Video 7

Microtubule breakage upon dynein motor walk. Capped GDP-microtubules, labelled with 20% fluorescent tubulin, were assembled from micropatterned seeds and served as tracks for the motility of dyneins (50 pM). Images were taken with a ×63 objective in TIRF microscopy with ×1.5 magnifier. Acquisition frame rate, 1 image every 3 seconds over 3 minutes. Display, 5 images s–1. Scale bar, 10 µm.

Supplementary Video 8

Free tubulin dimers protect microtubules during gliding assay on kinesins. Capped GDP-microtubules, labelled with 20% fluorescent tubulin, gliding on a layer of kinesin-1 in the presence of unlabelled free tubulin dimers. Images were taken with a ×20 objective in epifluorescence microscopy. Acquisition frame rate, 1 image every 5 seconds over 15 minutes. Display, 25 images s–1. Scale bar, 100 µm.

Supplementary Video 9

Free tubulin dimers protect microtubules during gliding assay on dyneins. Capped GDP-microtubules, labelled with 20% fluorescent tubulin, gliding on a layer of dyneins in the presence of unlabelled free tubulin dimers. Images were taken with a ×20 objective in epifluorescence microscopy. Acquisition frame rate, 1 image every 5 seconds over 15 minutes. Display, 25 images s–1. Scale bar, 100 µm.

Supplementary Video 10

Free tubulin dimers protect microtubules during motility assay. Capped GDP-microtubules, labelled with 20% fluorescent tubulin, were assembled from micropatterned seeds and served as tracks for the motility of kinesins (10 nM Klp2) in the absence (left) or presence (right) of free tubulin dimers (14 µM). Images were taken with a ×63 objective in TIRF microscopy with ×1.5 magnifier. Acquisition frame rate, 1 image every 2 minutes over 1 hour. Display, 5 images s–1. Scale bar, 10 µm.

Supplementary Video 11

Free tubulin dimers incorporate along microtubules during gliding assay. Capped GDP-microtubules, with the shaft labelled with 3% red fluorescent tubulin and the stable ends labelled with 20% red fluorescent tubulin (middle column), gliding on a layer of kinesins (kinesin-1) in the presence of unlabelled fluorescent free tubulin dimers after a 30-minute-long exposure to 14 µM free green fluorescent tubulin dimers (left column). Images were taken with a ×63 objective in TIRF microscopy with ×1.5 magnifier. Acquisition frame rate, 1 image every second over 30 seconds. Display, 5 images s–1. Scale bar, 20 µm.

Supplementary Video 12

Free tubulin dimers incorporate along microtubules during motility assay. Capped GDP-microtubules, labelled with 3% red fluorescent tubulin, were assembled from micropatterned seeds and served as tracks for the motility of kinesins (10 nM Klp2) in the presence of green free tubulin dimers (14 µM) that were further replaced by unlabelled dimers prior to imaging. Images were taken with a ×63 objective in TIRF microscopy with ×1.5 magnifier. Acquisition frame rate, 1 image every second over 30 seconds. Display, 5 images s–1. Scale bar, 10 µm.

Source data

Source Data Fig. 1

Numerical data to generate the graphs and tables.

Source Data Fig. 2

Numerical data to generate the graphs and tables.

Source Data Fig. 3

Numerical data to generate the graphs and tables.

Source Data Fig. 4

Numerical data to generate the graphs and tables.

Source Data Fig. 5

Numerical data to generate the graphs and tables.

Source Data Supplementary Fig. 1

Numerical data to generate the graphs and tables.

Source Data Supplementary Fig. 2

Numerical data to generate the graphs and tables.

Source Data Supplementary Fig. 3

Numerical data to generate the graphs and tables.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Triclin, S., Inoue, D., Gaillard, J. et al. Self-repair protects microtubules from destruction by molecular motors. Nat. Mater. 20, 883–891 (2021). https://doi.org/10.1038/s41563-020-00905-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41563-020-00905-0

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