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:

Mechanical regulation of a molecular clutch defines force transmission and transduction in response to matrix rigidity

Abstract

Cell function depends on tissue rigidity, which cells probe by applying and transmitting forces to their extracellular matrix, and then transducing them into biochemical signals. Here we show that in response to matrix rigidity and density, force transmission and transduction are explained by the mechanical properties of the actin–talin–integrin–fibronectin clutch. We demonstrate that force transmission is regulated by a dynamic clutch mechanism, which unveils its fundamental biphasic force/rigidity relationship on talin depletion. Force transduction is triggered by talin unfolding above a stiffness threshold. Below this threshold, integrins unbind and release force before talin can unfold. Above the threshold, talin unfolds and binds to vinculin, leading to adhesion growth and YAP nuclear translocation. Matrix density, myosin contractility, integrin ligation and talin mechanical stability differently and nonlinearly regulate both force transmission and the transduction threshold. In all cases, coupling of talin unfolding dynamics to a theoretical clutch model quantitatively predicts cell response.

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

Figure 1: Talin sets a rigidity threshold that triggers increased force transmission, adhesion maturation and YAP nuclear translocation.
Figure 2: Talin depletion affects β3 ligation, FAK phosphorylation and stress fibre formation only above the stiffness threshold.
Figure 3: The balance between clutch unbinding and talin unfolding predicts the force/rigidity curves and the rigidity threshold for mechanotransduction.
Figure 4: The rigidity threshold requires an intact integrin–cytoskeletal link mediated by full-length talin.
Figure 5: The rigidity threshold is mediated by talin unfolding under force and subsequent vinculin binding.
Figure 6: The elements of the molecular clutch tune force transmission and the rigidity threshold required for talin unfolding and YAP nuclear translocation.

Similar content being viewed by others

References

  1. Moore, S. W., Roca-Cusachs, P. & Sheetz, M. P. Stretchy proteins on stretchy substrates: the important elements of integrin-mediated rigidity sensing. Dev. Cell 19, 194–206 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Paszek, M. J. et al. Tensional homeostasis and the malignant phenotype. Cancer Cell 8, 241–254 (2005).

    CAS  PubMed  Google Scholar 

  3. Engler, A. J., Sen, S., Sweeney, H. L. & Discher, D. E. Matrix elasticity directs stem cell lineage specification. Cell 126, 677–689 (2006).

    CAS  PubMed  Google Scholar 

  4. Chan, C. E. & Odde, D. J. Traction dynamics of filopodia on compliant substrates. Science 322, 1687–1691 (2008).

    CAS  PubMed  Google Scholar 

  5. Elosegui-Artola, A. et al. Rigidity sensing and adaptation through regulation of integrin types. Nat. Mater. 13, 631–637 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Case, L. B. & Waterman, C. M. Integration of actin dynamics and cell adhesion by a three-dimensional, mechanosensitive molecular clutch. Nat. Cell Biol. 17, 955–963 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Plotnikov, S. V., Pasapera, A. M., Sabass, B. & Waterman, C. M. Force fluctuations within focal adhesions mediate ECM-rigidity sensing to guide directed cell migration. Cell 151, 1513–1527 (2012).

    CAS  PubMed  Google Scholar 

  8. Gupta, M. et al. Adaptive rheology and ordering of cell cytoskeleton govern matrix rigidity sensing. Nat. Commun. 6, 7525 (2015).

    CAS  PubMed  Google Scholar 

  9. Oakes, P. W., Banerjee, S., Marchetti, M. C. & Gardel, M. L. Geometry regulates traction stresses in adherent cells. Biophys. J. 107, 825–833 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  11. Ghibaudo, M. et al. Traction forces and rigidity sensing regulate cell functions. Soft Matter 4, 1836–1843 (2008).

    CAS  Google Scholar 

  12. Schiller, H. B. et al. β1- and αv-class integrins cooperate to regulate myosin II during rigidity sensing of fibronectin-based microenvironments. Nat. Cell Biol. 15, 625–636 (2013).

    CAS  PubMed  Google Scholar 

  13. Ghassemi, S. et al. Cells test substrate rigidity by local contractions on sub-micrometer pillars. Proc. Natl Acad. Sci. USA 109, 5328–5333 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Califano, J. P. & Reinhart-King, C. A. Substrate stiffness and cell area predict cellular traction stresses in single cells and cells in contact. Cell. Mol. Bioeng. 3, 68–75 (2010).

    PubMed  Google Scholar 

  15. Etienne, J. et al. Cells as liquid motors: mechanosensitivity emerges from collective dynamics of actomyosin cortex. Proc. Natl Acad. Sci. USA 112, 2740–2745 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Roca-Cusachs, P., Iskratsch, T. & Sheetz, M. P. Finding the weakest link—exploring integrin-mediated mechanical molecular pathways. J. Cell Sci. 125, 3025–3038 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Schoen, I., Pruitt, B. L. & Vogel, V. The Yin-Yang of rigidity sensing: how forces and mechanical properties regulate the cellular response to materials. Annu. Rev. Mater. Res. 43, 589–618 (2013).

    CAS  Google Scholar 

  18. Austen, K. et al. Extracellular rigidity sensing by talin isoform-specific mechanical linkages. Nat. Cell Biol. 17, 1597–1606 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Margadant, F. et al. Mechanotransduction in vivo by repeated talin stretch-relaxation events depends upon vinculin. PLoS Biol. 9, e1001223 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Roca-Cusachs, P., Gauthier, N. C., del Rio, A. & Sheetz, M. P. Clustering of α5β1 integrins determines adhesion strength whereas αvβ3 and talin enable mechanotransduction. Proc. Natl Acad. Sci. USA 106, 16245–16250 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Zhang, X. et al. Talin depletion reveals independence of initial cell spreading from integrin activation and traction. Nat. Cell Biol. 10, 1062–1068 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. del Rio, A. et al. Stretching single talin rod molecules activates vinculin binding. Science 323, 638–641 (2009).

    CAS  PubMed  Google Scholar 

  23. Chen, H., Choudhury, D. M. & Craig, S. W. Coincidence of actin filaments and talin is required to activate vinculin. J. Biol. Chem. 281, 40389–40398 (2006).

    CAS  PubMed  Google Scholar 

  24. Wegener, K. L. et al. Structural basis of integrin activation by talin. Cell 128, 171–182 (2007).

    CAS  PubMed  Google Scholar 

  25. Yao, M. et al. Mechanical activation of vinculin binding to talin locks talin in an unfolded conformation. Sci. Rep. 4, 4610 (2014).

    PubMed  PubMed Central  Google Scholar 

  26. Kong, F., Garcia, A. J., Mould, A. P., Humphries, M. J. & Zhu, C. Demonstration of catch bonds between an integrin and its ligand. J. Cell Biol. 185, 1275–1284 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Roca-Cusachs, P. Integrin-dependent force transmission to the extracellular matrix by α-actinin triggers adhesion maturation. Proc. Natl Acad. Sci. USA 110, E1361–E1370 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Tanentzapf, G. & Brown, N. H. An interaction between integrin and the talin FERM domain mediates integrin activation but not linkage to the cytoskeleton. Nat. Cell Biol. 8, 601–606 (2006).

    CAS  PubMed  Google Scholar 

  29. Hirata, H., Tatsumi, H., Lim, C. T. & Sokabe, M. Force-dependent vinculin binding to talin in live cells: a crucial step in anchoring the actin cytoskeleton to focal adhesions. Am. J. Physiol. Cell Physiol. 306, C607–C620 (2014).

    CAS  PubMed  Google Scholar 

  30. Cohen, D. M., Kutscher, B., Chen, H., Murphy, D. B. & Craig, S. W. A conformational switch in vinculin drives formation and dynamics of a talin-vinculin complex at focal adhesions. J. Biol. Chem. 281, 16006–16015 (2006).

    CAS  PubMed  Google Scholar 

  31. Humphries, J. D. et al. Vinculin controls focal adhesion formation by direct interactions with talin and actin. J. Cell Biol. 179, 1043–1057 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Bangasser, B. L., Rosenfeld, S. S. & Odde, D. J. Determinants of maximal force transmission in a motor-clutch model of cell traction in a compliant microenvironment. Biophys. J. 105, 581–592 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Bangasser, B. L. & Odde, D. J. Master equation-based analysis of a motor-clutch model for cell traction force. Cell. Mol. Bioeng. 6, 449–459 (2013).

    PubMed  Google Scholar 

  34. Saltel, F. et al. New PI(4,5)P2- and membrane proximal integrin-binding motifs in the talin head control β3-integrin clustering. J. Cell Biol. 187, 715–731 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Klapholz, B. et al. Alternative mechanisms for talin to mediate integrin function. Curr. Biol. 25, 847–857 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Iwamoto, D. V. & Calderwood, D. A. Regulation of integrin-mediated adhesions. Curr. Opin. Cell Biol. 36, 41–47 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Case, L. B. et al. Molecular mechanism of vinculin activation and nanoscale spatial organization in focal adhesions. Nat. Cell Biol. 17, 880–892 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Thievessen, I. et al. Vinculin–actin interaction couples actin retrograde flow to focal adhesions, but is dispensable for focal adhesion growth. J. Cell Biol. 202, 163–177 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Roca-Cusachs, P. et al. Micropatterning of single endothelial cell shape reveals a tight coupling between nuclear volume in G1 and proliferation. Biophys. J. 94, 4984–4995 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Swift, J. et al. Nuclear lamin-A scales with tissue stiffness and enhances matrix-directed differentiation. Science 341, 1240104 (2013).

    PubMed  PubMed Central  Google Scholar 

  41. DuFort, C. C., Paszek, M. J. & Weaver, V. M. Balancing forces: architectural control of mechanotransduction. Nat. Rev. Mol. Cell Biol. 12, 308–319 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Plouffe, S. W., Hong, A. W. & Guan, K. L. Disease implications of the Hippo/YAP pathway. Trends Mol. Med. 21, 212–222 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Bate, N. et al. Talin contains a C-terminal calpain2 cleavage site important in focal adhesion dynamics. PLoS ONE 7, e34461 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Simonson, W. T., Franco, S. J. & Huttenlocher, A. Talin1 regulates TCR-mediated LFA-1 function. J. Immunol. 177, 7707–7714 (2006).

    CAS  PubMed  Google Scholar 

  45. Pierschbacher, M. D. & Ruoslahti, E. Influence of stereochemistry of the sequence Arg-Gly-Asp-Xaa on binding-specificity in cell-adhesion. J. Biol. Chem. 262, 17294–17298 (1987).

    CAS  PubMed  Google Scholar 

  46. Frelinger, A. L. 3rd, Du, X. P., Plow, E. F. & Ginsberg, M. H. Monoclonal antibodies to ligand-occupied conformers of integrin α IIb β 3 (glycoprotein IIb-IIIa) alter receptor affinity, specificity, and function. J. Biol. Chem. 266, 17106–17111 (1991).

    CAS  PubMed  Google Scholar 

  47. Alcaraz, J. et al. Microrheology of human lung epithelial cells measured by atomic force microscopy. Biophys. J. 84, 2071–2079 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Hutter, J. L. & Bechhoefer, J. Calibration of atomic-force microscope tips. Rev. Sci. Instrum. 64, 1868–1873 (1993).

    CAS  Google Scholar 

  49. Serra-Picamal, X. et al. Mechanical waves during tissue expansion. Nat. Phys. 8, U628–U666 (2012).

    Google Scholar 

  50. Butler, J. P., Tolic-Norrelykke, I. M., Fabry, B. & Fredberg, J. J. Traction fields, moments, and strain energy that cells exert on their surroundings. Am. J. Physiol. Cell Physiol. 282, C595–C605 (2002).

    CAS  PubMed  Google Scholar 

  51. Elosegui-Artola, A. et al. Image analysis for the quantitative comparison of stress fibers and focal adhesions. PLoS ONE 9, e107393 (2014).

    PubMed  PubMed Central  Google Scholar 

  52. Chen, Y. et al. Fluorescence biomembrane force probe: concurrent quantitation of receptor-ligand kinetics and binding-induced intracellular signaling on a single cell. J. Vis. Exp. 102, e52975 (2015).

    Google Scholar 

  53. Takagi, J., Petre, B. M., Walz, T. & Springer, T. A. Global conformational rearrangements in integrin extracellular domains in outside-in and inside-out signaling. Cell 110, 599–611 (2002).

    CAS  PubMed  Google Scholar 

  54. Chesla, S. E., Selvaraj, P. & Zhu, C. Measuring two-dimensional receptor-ligand binding kinetics by micropipette. Biophys. J. 75, 1553–1572 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Chen, W., Zarnitsyna, V. I., Sarangapani, K. K., Huang, J. & Zhu, C. Measuring receptor-ligand binding kinetics on cell surfaces: from adhesion frequency to thermal fluctuation methods. Cell. Mol. Bioeng. 1, 276–288 (2008).

    CAS  PubMed  Google Scholar 

  56. Chen, W., Lou, J. & Zhu, C. Forcing switch from short- to intermediate- and long-lived states of the αA domain generates LFA-1/ICAM-1 catch bonds. J. Biol. Chem. 285, 35967–35978 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We acknowledge support from the Spanish Ministry for Economy and Competitiveness (BFU2011-23111, BFU2012-38146 and BFU2014-52586-REDT), a Career Integration Grant within the seventh European Community Framework Programme (PCIG10-GA-2011-303848), the European Research Council (Grant Agreements 242993 and 240487), the Generalitat de Catalunya, Fundació La Caixa, Fundació la Marató de TV3 (project 20133330), and the National Institutes of Health (US NIH R01AI044902). A.E.-A., R.O. and C.P.-G. were supported respectively by a Juan de la Cierva Fellowship (Spanish Ministry of Economy and Competitiveness), a FI fellowship (Generalitat de Catalunya), and the fundació ‘La Caixa’. We thank R. Sunyer, J. Alcaraz, E. Bazellières, F. Rico, S. Garcia-Manyes, N. Bate, N. Berrow and the members of the P.R.-C. and X.T. laboratories for technical assistance and discussions.

Author information

Authors and Affiliations

Authors

Contributions

A.E.-A. and P.R.-C. conceived the study, A.E.-A., C.Z., X.T. and P.R.-C. designed the experiments, A.E.-A., R.O., Y.C., A.K., C.P.-G. and N.C. performed the experiments, P.R.-C. carried out the theoretical modelling, and A.E.-A. and P.R.-C. wrote the paper.

Corresponding author

Correspondence to Pere Roca-Cusachs.

Ethics declarations

Competing interests

The results have been protected under a patent application.

Integrated supplementary information

Supplementary Figure 4 Cell adhesion to fibronectin-coated gels is mediated by αvβ3 and α5β1 integrins.

(a) Images showing control and Talin 2 shRNA cells on 29 kPa fibronectin-coated polyacrylamide gels with or without blocking integrin α5β1 (using 10 μg ml−1 of BMB5 antibody), αvβ3 (using 0.5 mM of the specific Gpen peptide) or both. Scale bar is 50 μm. (b) Corresponding quantification of the percentage of spread cells (from left to right, n = 12, 12, 11, 11, 20, 11, 10, 12 fields of view)(, p ≤ 0.001, two-way Anova). Data show 1 out of 2 independent experiments. Blocking both integrins abolished cell adhesion almost completely.

Supplementary Figure 5 Talin1 Head L325R expression progressively reduces force transmission above but not below the rigidity threshold.

Traction forces exerted by control cells on 5 kPa gels (blue) and 29 kPa gels (red) as a function of the efficiency of transfection with Talin1 Head L325R. Values are compared to mean forces of untransfected control cells (left) and talin shRNA cells (right). Note that in Fig. 1, talin 1 Head L325R data represent averages for well transfected cells only. Dotted lines represent sigmoidal fits to the data. (Control: 5 kPa, n = 35 cells; 29 kPa, n = 12 cells. Control + Talin 1 Head: 5 kPa, n = 37 cells; 29 kPa, n = 42 cells. Talin 2 shRNA: 5 kPa, n = 34 cells; 29 kPa, n = 29 cells). Data show 1 out of 3 independent experiments.

Supplementary Figure 6 Further quantifications of integrin and pFAK in adhesions.

(a) quantification of integrin density from staining images of ligand bound β3 for Control cells (red, n = 20, 31, 24, 33, 20, 29 fields respectively for increasing rigidity measured in 8–10 cells) and Talin 2 shRNA cells (blue, n = 20, 21, 25, 29, 24, 29 fields measured in 8–9 cells). Data show 1 out of 3 independent experiments. Integrin densities were significantly different between control and depleted cells only above 5 kPa (P < 0.001, two-way Anova). (b) Quantification of the percentage of cell spreading area covered by pFAK-positive adhesions from staining images of Control cells (red. n = 11, 10, 17, 17, 17, 15 cells respectively for increasing rigidity) and Talin 2 shRNA cells (blue, n = 11, 10, 10, 12, 10, 10 cells) as a function of substrate stiffness. Data show 1 out of 3 independent experiments. Significant differences were observed only above 5 kPa (P = 0.039, two-way Anova).

Supplementary Figure 7 Dependence of cell area and myosin phosphorylation on substrate stiffness.

(a) Quantification of cell area in response to substrate stiffness for control and talin 2 shRNA cells (Control: n = 17, 12, 35, 42, 42, 12 cells respectively for increasing stiffness; Talin 2 shRNA: n = 10, 11, 34, 23, 25, 29 cells). Data show 1 out of 14 independent experiments. Talin depletion did not have a significant effect (two-way Anova). (b) For cells plated on gels of the indicated stiffness, representative western blots of talin, GAPDH as loading control, phosphorylated myosin light chain and total myosin light chain for Control and Talin 2 shRNA cells. (c) Corresponding quantification of the phosphorylated/total myosin light chain ratio (pooled from n = 3 independent experiments). No significant differences were found (two-way Anova). (d) Representative western blots of phosphorylated myosin light chain and total myosin light chain for wild-type MEF cells. (e) Corresponding quantification of the phosphorylated/total myosin light chain ratio (pooled from n = 3 independent experiments). No significant differences were found, suggesting that myosin phosphorylation is not significantly affected in MEF cells regardless of talin (one-way Anova).

Supplementary Figure 8 Further analyses on the effects of vinculin fragments.

(a) Average forces in response to substrate stiffness for cells transfected with Talin 2 shRNA + VD1 (red, n = 10, 13, 11, 11, 13, 10 cells, respectively for increasing stiffness) and Talin 2 shRNA + VD1 A501 (blue, n = 11, 11, 12, 11, 11, 10 cells). No significant differences were found between transfections (two-way Anova). Data show 1 out of 3 independent experiments. (b) Quantification of Nuclear/Cytosolic YAP ratio for the same conditions as in (a) (Talin 2 shRNA + VD1:n = 26, 20, 30, 29, 32, 32 cells respectively for increasing stiffness; Talin 2 shRNA + VD1 A501: n = 22, 21, 21, 23, 23, 24 cells). No significant differences were found between transfections (two-way Anova). Data show 1 out of 3 independent experiments. (c) Quantification of Nuclear/Cytosolic YAP ratio for control cells transfected with VD1 (red) and or VD1 A501 (blue) as a function of transfection efficiency (measured as the relative intensity of EGFP fluorescence) on 29 kPa polyacrylamide gels (Control + VD1:n = 59 cells; Control + VD1 A501: n = 49 cells). Data show 1 out of 3 independent experiments. Dashed lines are a sigmoidal fit to the experimental results for each condition. Further confirming the blocking role of VD1, increasing transfection efficiencies progressively decreased nuclear localization of YAP. In contrast, increasing efficiencies of transfection with VD1 A501 had no effect.

Supplementary Figure 9 Fibronectin coating densities.

Resulting fibronectin coating densities on the surface of polyacrylamide gels of 5 and 29 kPa coated with solutions containing 1, 10, or 100 μg ml−1 of fibronectin. n = 6 gels in all cases except 100 μg ml−1–29 kPa (5 gels). Data pooled from two independent experiments.

Supplementary Figure 10 Unprocessed versions of the western blots shown in Supplementary Fig. 4.

(a,b) blots corresponding to panel b in Supplementary Fig. 4. (c) blots corresponding to panel d in Supplementary Fig. 4. All measured bands corresponded to the molecular weights of the different proteins as detailed by antibody providers: talin (225–235 kDa), GADPH (36 kDa), and MLC (18 kDa). Note that blots do not show the entire molecular weight spectrum because membranes were cut before antibody incubation to incubate each band only with the relevant antibody.

Supplementary Table 1 Model parameters.
Supplementary Table 2 Polyacrylamide gel rigidities measured with AFM.
Supplementary Table 3 Statistical details of Fig. 6 panels.

Supplementary information

Supplementary Information

Supplementary Information (PDF 1057 kb)

41556_2016_BFncb3336_MOESM8_ESM.avi

Time-lapse of control cells transfected with lifeact-GFP and plated on fibronectin-coated substrates of increasing Young’s modulus (2-5-11-14-29 kPa from left to right). Scale bar is 20 μm. (AVI 1996 kb)

41556_2016_BFncb3336_MOESM9_ESM.avi

Time-lapse of talin 2 shRNA cells transfected with lifeact-GFP and plated on fibronectin-coated substrates of increasing Young’s modulus (2-5-11-14-29 kPa from left to right). Scale bar is 20 μm. (AVI 1733 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Elosegui-Artola, A., Oria, R., Chen, Y. et al. Mechanical regulation of a molecular clutch defines force transmission and transduction in response to matrix rigidity. Nat Cell Biol 18, 540–548 (2016). https://doi.org/10.1038/ncb3336

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3336

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