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:

The mechanical stability of proteins regulates their translocation rate into the cell nucleus

Abstract

A cell’s ability to react to mechanical stimuli is known to be affected by the transport of transcription factors, the proteins responsible for regulating transcription of DNA into RNA, across the membrane enveloping its nucleus. Yet the molecular mechanisms by which mechanical cues control this process remain unclear. Here we show that one such protein, myocardin-related transcription factor A (MRTFA), is imported into the nucleus at a rate that is inversely correlated with its nanomechanical stability, but independent of its thermodynamic stability. Attaching mechanically stable proteins to MRTFA results in reduced gene expression and the subsequent slowing down of cell migration. We conclude that the mechanical unfolding of proteins regulates their nuclear translocation rate, and highlight the role of the nuclear pore complex as a selective mechanosensor that is capable of detecting forces as low as 10 pN. The modulation of the mechanical stability of transcription factors may represent a general strategy for the control of gene expression.

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: Protein unfolding modulates the kinetics of nuclear import.
Fig. 2: The kinetics of MRTFA nuclear import is regulated by its mechanical properties.
Fig. 3: The NPC is highly mechanoselective.
Fig. 4: The mechanical selectivity of the NPC probed with optogenetic constructs.
Fig. 5: Mechanically stable MRTFA constructs downregulate gene expression and cellular motility.

Similar content being viewed by others

Data availability

Data supporting this research can be obtained from the corresponding author on reasonable request.

References

  1. Vogel, V. & Sheetz, M. Local force and geometry sensing regulate cell functions. Nat. Rev. Mol. Cell Biol. 7, 265–275 (2006).

    Article  Google Scholar 

  2. Dupont, S. et al. Role of YAP/TAZ in mechanotransduction. Nature 474, 179–183 (2011).

    Article  Google Scholar 

  3. Markiewicz, E. et al. The inner nuclear membrane protein emerin regulates beta-catenin activity by restricting its accumulation in the nucleus. EMBO J. 25, 3275–3285 (2006).

    Article  Google Scholar 

  4. Moon, H. S., Even-Ram, S., Kleinman, H. K. & Cha, H. J. Zyxin is upregulated in the nucleus by thymosin beta4 in SiHa cells. Exp. Cell Res. 312, 3425–3431 (2006).

    Article  Google Scholar 

  5. Fedorchak, G. R., Kaminski, A. & Lammerding, J. Cellular mechanosensing: getting to the nucleus of it all. Prog. Biophys. Mol. Biol. 115, 76–92 (2014).

    Article  Google Scholar 

  6. Medjkane, S., Perez-Sanchez, C., Gaggioli, C., Sahai, E. & Treisman, R. Myocardin-related transcription factors and SRF are required for cytoskeletal dynamics and experimental metastasis. Nat. Cell Biol. 11, 257–268 (2009).

    Article  Google Scholar 

  7. Olson, E. N. & Nordheim, A. Linking actin dynamics and gene transcription to drive cellular motile functions. Nat. Rev. Mol. Cell Biol. 11, 353–365 (2010).

    Article  Google Scholar 

  8. Jain, N., Iyer, K. V., Kumar, A. & Shivashankar, G. V. Cell geometric constraints induce modular gene-expression patterns via redistribution of HDAC3 regulated by actomyosin contractility. Proc. Natl Acad. Sci. USA 110, 11349–11354 (2013).

    Article  ADS  Google Scholar 

  9. Connelly, J. T. et al. Actin and serum response factor transduce physical cues from the microenvironment to regulate epidermal stem cell fate decisions. Nat. Cell Biol. 12, 711–718 (2010).

    Article  Google Scholar 

  10. Miralles, F., Posern, G., Zaromytidou, A. I. & Treisman, R. Actin dynamics control SRF activity by regulation of its coactivator MAL. Cell 113, 329–342 (2003).

    Article  Google Scholar 

  11. Ho, C. Y., Jaalouk, D. E., Vartiainen, M. K. & Lammerding, J. Lamin A/C and emerin regulate MKL1-SRF activity by modulating actin dynamics. Nature 497, 507–511 (2013).

    Article  ADS  Google Scholar 

  12. Pawlowski, R., Rajakyla, E. K., Vartiainen, M. K. & Treisman, R. An actin-regulated importin alpha/beta-dependent extended bipartite NLS directs nuclear import of MRTF-A. EMBO J. 29, 3448–3458 (2010).

    Article  Google Scholar 

  13. Mouilleron, S., Langer, C. A., Guettler, S., McDonald, N. Q. & Treisman, R. Structure of a pentavalent G-actin•MRTF-A complex reveals how G-actin controls nucleocytoplasmic shuttling of a transcriptional coactivator. Sci. Signal. 4, ra40 (2011).

    Article  Google Scholar 

  14. Vartiainen, M. K., Guettler, S., Larijani, B. & Treisman, R. Nuclear actin regulates dynamic subcellular localization and activity of the SRF cofactor MAL. Science 316, 1749–1752 (2007).

    Article  ADS  Google Scholar 

  15. Baarlink, C., Wang, H. & Grosse, R. Nuclear actin network assembly by formins regulates the SRF coactivator MAL. Science 340, 864–867 (2013).

    Article  ADS  Google Scholar 

  16. Kim, S. J. et al. Integrative structure and functional anatomy of a nuclear pore complex. Nature 555, 475–482 (2018).

    Article  ADS  Google Scholar 

  17. Maillard, R. A. et al. ClpX(P) generates mechanical force to unfold and translocate its protein substrates. Cell 145, 459–469 (2011).

    Article  Google Scholar 

  18. Olivares, A. O., Kotamarthi, H. C., Stein, B. J., Sauer, R. T. & Baker, T. A. Effect of directional pulling on mechanical protein degradation by ATP-dependent proteolytic machines. Proc. Natl Acad. Sci. USA 4, ra40 (2011).

    Google Scholar 

  19. Rodriguez-Larrea, D. & Bayley, H. Multistep protein unfolding during nanopore translocation. Nat. Nanotechnol. 8, 288–295 (2013).

    Article  ADS  Google Scholar 

  20. Maimon, T., Elad, N., Dahan, I. & Medalia, O. The human nuclear pore complex as revealed by cryo-electron tomography. Structure 20, 998–1006 (2012).

    Article  Google Scholar 

  21. Lin, D. H. et al. Architecture of the symmetric core of the nuclear pore. Science 352, aaf1015 (2016).

    Article  ADS  Google Scholar 

  22. Lemke, E. A. The multiple faces of disordered nucleoporins. J. Mol. Biol. 428, 2011–2024 (2016).

    Article  Google Scholar 

  23. Grunwald, D. & Singer, R. H. Multiscale dynamics in nucleocytoplasmic transport. Curr. Opin. Cell Biol. 24, 100–106 (2012).

    Article  Google Scholar 

  24. Schmidt, H. B. & Gorlich, D. Transport selectivity of nuclear pores, phase separation, and membraneless organelles. Trends Biochem. Sci. 41, 46–61 (2016).

    Article  Google Scholar 

  25. Rout, M. P., Aitchison, J. D., Magnasco, M. O. & Chait, B. T. Virtual gating and nuclear transport: the hole picture. Trends Cell Biol. 13, 622–628 (2003).

    Article  Google Scholar 

  26. Lim, R. Y. et al. Nanomechanical basis of selective gating by the nuclear pore complex. Science 318, 640–643 (2007).

    Article  ADS  Google Scholar 

  27. Frey, S., Richter, R. P. & Gorlich, D. FG-rich repeats of nuclear pore proteins form a three-dimensional meshwork with hydrogel-like properties. Science 314, 815–817 (2006).

    Article  ADS  Google Scholar 

  28. Lim, R. Y. et al. Flexible phenylalanine–glycine nucleoporins as entropic barriers to nucleocytoplasmic transport. Proc. Natl Acad. Sci. USA 103, 9512–9517 (2006).

    Article  ADS  Google Scholar 

  29. Yamada, J. et al. A bimodal distribution of two distinct categories of intrinsically disordered structures with separate functions in FG nucleoporins. Mol. Cell Proteom. 9, 2205–2224 (2010).

    Article  Google Scholar 

  30. Peters, R. Translocation through the nuclear pore complex: selectivity and speed by reduction-of-dimensionality. Traffic 6, 421–427 (2005).

    Article  Google Scholar 

  31. Fisher, T. E., Oberhauser, A. F., Carrion-Vazquez, M., Marszalek, P. E. & Fernandez, J. M. The study of protein mechanics with the atomic force microscope. Trends Biochem. Sci. 24, 379–384 (1999).

    Article  Google Scholar 

  32. Mehlin, H., Daneholt, B. & Skoglund, U. Translocation of a specific premessenger ribonucleoprotein particle through the nuclear pore studied with electron microscope tomography. Cell 69, 605–613 (1992).

    Article  Google Scholar 

  33. Grunwald, D. & Singer, R. H. In vivo imaging of labelled endogenous beta-actin mRNA during nucleocytoplasmic transport. Nature 467, 604–607 (2010).

    Article  ADS  Google Scholar 

  34. Stevens, B. J. & Swift, H. RNA transport from nucleus to cytoplasm in Chironomus salivary glands. J. Cell Biol. 31, 55–77 (1966).

    Article  Google Scholar 

  35. Lowe, A. R. et al. Selectivity mechanism of the nuclear pore complex characterized by single cargo tracking. Nature 467, 600–603 (2010).

    Article  ADS  Google Scholar 

  36. Elosegui-Artola, A. et al. Force triggers YAP nuclear entry by regulating transport across nuclear pores. Cell 171, 1397–1410 e1314 (2017).

    Article  Google Scholar 

  37. Kudo, N. et al. Leptomycin B inhibition of signal-mediated nuclear export by direct binding to CRM1. Exp. Cell Res. 242, 540–547 (1998).

    Article  Google Scholar 

  38. Li, H., Carrion-Vazquez, M., Oberhauser, A. F., Marszalek, P. E. & Fernandez, J. M. Point mutations alter the mechanical stability of immunoglobulin modules. Nat. Struct. Biol. 7, 1117–1120 (2000).

    Article  Google Scholar 

  39. Kang, H. J., Coulibaly, F., Clow, F., Proft, T. & Baker, E. N. Stabilizing isopeptide bonds revealed in gram-positive bacterial pilus structure. Science 318, 1625–1628 (2007).

    Article  ADS  Google Scholar 

  40. Alegre-Cebollada, J., Badilla, C. L. & Fernandez, J. M. Isopeptide bonds block the mechanical extension of pili in pathogenic Streptococcus pyogenes. J. Biol. Chem. 285, 11235–11242 (2010).

    Article  Google Scholar 

  41. Kang, H. J. & Baker, E. N. Intramolecular isopeptide bonds give thermodynamic and proteolytic stability to the major pilin protein of Streptococcus pyogenes. J. Biol. Chem. 284, 20729–20737 (2009).

    Article  Google Scholar 

  42. Li, H. et al. Reverse engineering of the giant muscle protein titin. Nature 418, 998–1002 (2002).

    Article  ADS  Google Scholar 

  43. Li, H. & Fernandez, J. M. Mechanical design of the first proximal Ig domain of human cardiac titin revealed by single molecule force spectroscopy. J. Mol. Biol. 334, 75–86 (2003).

    Article  Google Scholar 

  44. Carrion-Vazquez, M. et al. Mechanical and chemical unfolding of a single protein: a comparison. Proc. Natl Acad. Sci. USA 96, 3694–3699 (1999).

    Article  ADS  Google Scholar 

  45. Randles, L. G., Rounsevell, R. W. & Clarke, J. Spectrin domains lose cooperativity in forced unfolding. Biophys. J. 92, 571–577 (2007).

    Article  Google Scholar 

  46. Perez-Jimenez, R., Garcia-Manyes, S., Ainavarapu, S. R. & Fernandez, J. M. Mechanical unfolding pathways of the enhanced yellow fluorescent protein revealed by single molecule force spectroscopy. J. Biol. Chem. 281, 40010–40014 (2006).

    Article  Google Scholar 

  47. Soderholm, J. F. et al. Importazole, a small molecule inhibitor of the transport receptor importin-beta. ACS Chem. Biol. 6, 700–708 (2011).

    Article  Google Scholar 

  48. Niopek, D., Wehler, P., Roensch, J., Eils, R. & Di Ventura, B. Optogenetic control of nuclear protein export. Nat. Commun. 7, 10624 (2016).

    Article  ADS  Google Scholar 

  49. Record, J. et al. Immunodeficiency and severe susceptibility to bacterial infection associated with a loss-of-function homozygous mutation of MKL1. Blood 126, 1527–1535 (2015).

    Article  Google Scholar 

  50. Beck, M. & Hurt, E. The nuclear pore complex: understanding its function through structural insight. Nat. Rev. Mol. Cell Biol. 18, 73–89 (2017).

    Article  Google Scholar 

  51. von Appen, A. et al. In situ structural analysis of the human nuclear pore complex. Nature 526, 140–143 (2015).

    Article  ADS  Google Scholar 

  52. Bestembayeva, A. et al. Nanoscale stiffness topography reveals structure and mechanics of the transport barrier in intact nuclear pore complexes. Nat. Nanotechnol. 10, 60–64 (2015).

    Article  ADS  Google Scholar 

  53. Frey, S. et al. Surface properties determining passage rates of proteins through nuclear pores. Cell 174, 202–217 e209 (2018).

    Article  Google Scholar 

  54. Mohr, D., Frey, S., Fischer, T., Guttler, T. & Gorlich, D. Characterisation of the passive permeability barrier of nuclear pore complexes. EMBO J. 28, 2541–2553 (2009).

    Article  Google Scholar 

  55. Ketterer, P. et al. DNA origami scaffold for studying intrinsically disordered proteins of the nuclear pore complex. Nat. Commun. 9, 902 (2018).

    Article  ADS  Google Scholar 

  56. Popa, I., Kosuri, P., Alegre-Cebollada, J., Garcia-Manyes, S. & Fernandez, J. M. Force dependency of biochemical reactions measured by single-molecule force-clamp spectroscopy. Nat. Protoc. 8, 1261–1276 (2013).

    Article  Google Scholar 

  57. Rosa, N. et al. Meltdown: a tool to help in the interpretation of thermal melt curves acquired by differential scanning fluorimetry. J. Biomol. Screen. 20, 898–905 (2015).

    Article  Google Scholar 

  58. Rodriguez, L. G., Wu, X. & Guan, J. L. Wound-healing assay. Methods Mol. Biol. 294, 23–29 (2005).

    Google Scholar 

Download references

Acknowledgements

We thank M. Vartiainen (University of Helsinki) for sharing the MRTFA–GFP plasmid, M. Parsons (King’s College London) for sharing the pEBFP2–N1 and pEYFP–N1 vectors and the MDA-MB-231 cell line, and U. Eggert and M. Pfuhl (King’s College London) for providing HeLa and Top10 competent cells, respectively. We thank the Nikon Imaging Centre at King’s College London for assistance in setting up the cell-imaging experiments. We wish to thank G. Yang for help in qPCR analysis, and C. Nichols and S. Conte (King’s College London) for help with differential scanning fluorimetry experiments. A.E.M.B. is the recipient of a Sir Henry Wellcome fellowship (210887/Z/18/Z). V.S.R. was funded by the BHF Centre for Research Excellence at King’s College London. This work was supported by BHF grant PG/13/50/30426, the European Commission (Mechanocontrol, grant agreement SEP-210342844), EPSRC Fellowship K00641X/1, EPSRC Strategic Equipment Grant EP/M022536/1, Leverhulme Trust Project Grant RPG-2015-225, Leverhulme Trust Research Leadership Award RL-2016-015, Wellcome Trust Investigator Award 212218/Z/18/Z and Royal Society Wolfson Fellowship RSWF/R3/183006, all to S.G.-M.

Author information

Authors and Affiliations

Authors

Contributions

S.G.-M. conceived the research. E.I. designed and performed cell biology, live-cell imaging experiments, motility assays and qPCR experiments. A.S. designed and performed live-cell imaging analysis and kinetic modelling. P.R.-L., S.J.B., E.R. and A.L. performed molecular biology work. A.E.M.B. and Y.J.W. conducted single-molecule nanomechanical experiments and A.E.M.B. analysed data. E.R. with A.S. conducted and analysed differential scanning fluorimetry experiments. S.G.B., F.P. and V.S.R. acquired preliminary data. C.S. and P.R.-C. participated in data discussion. S.G.-M., E.I. and A.S. wrote the paper.

Corresponding author

Correspondence to Sergi Garcia-Manyes.

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 and Supplementary Figs. 1–17.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Infante, E., Stannard, A., Board, S.J. et al. The mechanical stability of proteins regulates their translocation rate into the cell nucleus. Nat. Phys. 15, 973–981 (2019). https://doi.org/10.1038/s41567-019-0551-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41567-019-0551-3

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