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:

Kank2 activates talin, reduces force transduction across integrins and induces central adhesion formation

Abstract

Integrin-based adhesions play critical roles in cell migration. Talin activates integrins and flexibly connects integrins to the actomyosin cytoskeleton, thereby serving as a ‘molecular clutch’ that transmits forces to the extracellular matrix to drive cell migration. Here we identify the evolutionarily conserved Kank protein family as novel components of focal adhesions (FAs). Kank proteins accumulate at the lateral border of FAs, which we term the FA belt, and in central sliding adhesions, where they directly bind the talin rod domain through the Kank amino-terminal (KN) motif and induce talin and integrin activation. In addition, Kank proteins diminish the talin–actomyosin linkage, which curbs force transmission across integrins, leading to reduced integrin–ligand bond strength, slippage between integrin and ligand, central adhesion formation and sliding, and reduced cell migration speed. Our data identify Kank proteins as talin activators that decrease the grip between the integrin–talin complex and actomyosin to regulate cell migration velocity.

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: Kank2 is a novel FA protein.
Figure 2: Kank2 is targeted to FAs through the KN motif.
Figure 3: Kank2 curbs cell migration by inducing adhesion sliding.
Figure 4: Adhesion sliding occurs at the interface between integrin and ligand.
Figure 5: Kank2 impairs force transmission across integrins.
Figure 6: Kank2 directly binds the talin rod.
Figure 7: Kank2 induces talin and integrin activation.
Figure 8: Kank2 decreases F-actin binding to talin-ABS2.

Similar content being viewed by others

References

  1. Vicente-Manzanares, M. & Horwitz, A. R. Adhesion dynamics at a glance. J. Cell Sci. 124, 3923–3927 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Parsons, J. T., Horwitz, A. R. & Schwartz, M. A. Cell adhesion: integrating cytoskeletal dynamics and cellular tension. Nat. Rev. Mol. Cell Biol. 11, 633–643 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. 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 

  4. Hoffman, B. D., Grashoff, C. & Schwartz, M. A. Dynamic molecular processes mediate cellular mechanotransduction. Nature 475, 316–323 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Giannone, G., Mege, R. M. & Thoumine, O. Multi-level molecular clutches in motile cell processes. Trends Cell Biol. 19, 475–486 (2009).

    CAS  PubMed  Google Scholar 

  6. Critchley, D. R. Biochemical and structural properties of the integrin-associated cytoskeletal protein talin. Annu. Rev. Biophys. 38, 235–254 (2009).

    CAS  PubMed  Google Scholar 

  7. Smilenov, L. B., Mikhailov, A., Pelham, R. J., Marcantonio, E. E. & Gundersen, G. G. Focal adhesion motility revealed in stationary fibroblasts. Science 286, 1172–1174 (1999).

    CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  9. 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 

  10. 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 

  11. Atherton, P. et al. Vinculin controls talin engagement with the actomyosin machinery. Nat. Commun. 6, 10038 (2015).

    CAS  PubMed  Google Scholar 

  12. Calderwood, D. A., Campbell, I. D. & Critchley, D. R. Talins and kindlins: partners in integrin-mediated adhesion. Nat. Rev. Mol. Cell Biol. 14, 503–517 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Goult, B. T. et al. Structural studies on full-length talin1 reveal a compact auto-inhibited dimer: implications for talin activation. J. Struct. Biol. 184, 21–32 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Song, X. et al. A novel membrane-dependent on/off switch mechanism of talin FERM domain at sites of cell adhesion. Cell Res. 22, 1533–1545 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Goksoy, E. et al. Structural basis for the autoinhibition of talin in regulating integrin activation. Mol. Cell 31, 124–133 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Chang, Y. C. et al. Structural and mechanistic insights into the recruitment of talin by RIAM in integrin signaling. Structure 22, 1810–1820 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Han, J. et al. Reconstructing and deconstructing agonist-induced activation of integrin αIIbβ3. Curr. Biol. 16, 1796–1806 (2006).

    CAS  PubMed  Google Scholar 

  18. Lafuente, E. M. et al. RIAM, an Ena/VASP and Profilin ligand, interacts with Rap1-GTP and mediates Rap1-induced adhesion. Dev. Cell 7, 585–595 (2004).

    CAS  PubMed  Google Scholar 

  19. Legate, K. R. et al. Integrin adhesion and force coupling are independently regulated by localized PtdIns(4,5)2 synthesis. EMBO J. 30, 4539–4553 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Comrie, W. A., Babich, A. & Burkhardt, J. K. F-actin flow drives affinity maturation and spatial organization of LFA-1 at the immunological synapse. J. Cell Biol. 208, 475–491 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. 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 

  22. Legate, K. R., Montag, D., Bottcher, R. T., Takahashi, S. & Fassler, R. Comparative phenotypic analysis of the two major splice isoforms of phosphatidylinositol phosphate kinase type Igamma in vivo. J. Cell Sci. 125, 5636–5646 (2012).

    CAS  PubMed  Google Scholar 

  23. Stritt, S. et al. Rap1-GTP-interacting adaptor molecule (RIAM) is dispensable for platelet integrin activation and function in mice. Blood 125, 219–222 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Klapproth, S. et al. Loss of the Rap-1 effector RIAM results in leukocyte adhesion deficiency due to impaired β2 integrin function in mice. Blood 126, 2704–2712 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Kakinuma, N., Zhu, Y., Wang, Y., Roy, B. C. & Kiyama, R. Kank proteins: structure, functions and diseases. Cell. Mol. Life Sci. 66, 2651–2659 (2009).

    CAS  PubMed  Google Scholar 

  26. Ding, M., Goncharov, A., Jin, Y. & Chisholm., A. D. C. elegans ankyrin repeat protein VAB-19 is a component of epidermal attachment structures and is essential for epidermal morphogenesis. Development 130, 5791–5801 (2003).

    CAS  PubMed  Google Scholar 

  27. Yang, Y., Lee, W. S., Tang, X. & Wadsworth, W. G. Extracellular matrix regulates UNC-6 (netrin) axon guidance by controlling the direction of intracellular UNC-40 (DCC) outgrowth activity. PLoS ONE 9, e97258 (2014).

    PubMed  PubMed Central  Google Scholar 

  28. Ihara, S. et al. Basement membrane sliding and targeted adhesion remodels tissue boundaries during uterine-vulval attachment in Caenorhabditis elegans. Nat. Cell Biol. 13, 641–651 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Lerer, I. et al. Deletion of the ANKRD15 gene at 9p24.3 causes parent-of-origin-dependent inheritance of familial cerebral palsy. Human Mol. Genet. 14, 3911–3920 (2005).

    CAS  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  32. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Schiller, H. B., Friedel, C. C., Boulegue, C. & Fassler, R. Quantitative proteomics of the integrin adhesome show a myosin II-dependent recruitment of LIM domain proteins. EMBO Rep. 12, 259–266 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. 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 

  35. Bottcher, R. T. et al. Sorting nexin 17 prevents lysosomal degradation of β1 integrins by binding to the β1-integrin tail. Nat. Cell Biol. 14, 584–592 (2012).

    PubMed  Google Scholar 

  36. Zamir, E. et al. Dynamics and segregation of cell-matrix adhesions in cultured fibroblasts. Nat. Cell Biol. 2, 191–196 (2000).

    CAS  PubMed  Google Scholar 

  37. Wang, X. et al. Integrin Molecular Tension within Motile Focal Adhesions. Biophys. J. 109, 2259–2267 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Morimatsu, M., Mekhdjian, A. H., Chang, A. C., Tan, S. J. & Dunn, A. R. Visualizing the interior architecture of focal adhesions with high-resolution traction maps. Nano. Lett. 15, 2220–2228 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Theodosiou, M. et al. Kindlin-2 cooperates with talin to activate integrins and induces cell spreading by directly binding paxillin. Elife 5, e10130 (2016).

    PubMed  PubMed Central  Google Scholar 

  41. Gingras, A. R. et al. Central region of talin has a unique fold that binds vinculin and actin. J. Biol. Chem. 285, 29577–29587 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Himmel, M. et al. Control of high affinity interactions in the talin C terminus: how talin domains coordinate protein dynamics in cell adhesions. J. Biol. Chem. 284, 13832–13842 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Rossier, O. et al. Integrins β1 and β3 exhibit distinct dynamic nanoscale organizations inside focal adhesions. Nat. Cell Biol. 14, 1057–1067 (2012).

    CAS  PubMed  Google Scholar 

  44. Medves, S. et al. Multiple oligomerization domains of KANK1-PDGFRβ are required for JAK2-independent hematopoietic cell proliferation and signaling via STAT5 and ERK. Haematologica 96, 1406–1414 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Iskratsch, T. et al. FHOD1 is needed for directed forces and adhesion maturation during cell spreading and migration. Dev. Cell 27, 545–559 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Stehbens, S. J. et al. CLASPs link focal-adhesion-associated microtubule capture to localized exocytosis and adhesion site turnover. Nat. Cell Biol. 16, 561–573 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Grigoriev, I. et al. Rab6 regulates transport and targeting of exocytotic carriers. Dev. Cell 13, 305–314 (2007).

    CAS  PubMed  Google Scholar 

  48. Lansbergen, G. et al. CLASPs attach microtubule plus ends to the cell cortex through a complex with LL5β. Dev. Cell 11, 21–32 (2006).

    CAS  PubMed  Google Scholar 

  49. Cox, J. & Mann, M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat. Biotechnol. 26, 1367–1372 (2008).

    CAS  PubMed  Google Scholar 

  50. Shevchenko, A., Tomas, H., Havlis, J., Olsen, J. V. & Mann, M. In-gel digestion for mass spectrometric characterization of proteins and proteomes. Nat. Protoc. 1, 2856–2860 (2006).

    CAS  PubMed  Google Scholar 

  51. Ciobanasu, C., Faivre, B. & LeClainche, C. Actomyosin-dependent formation of the mechanosensitive talin-vinculin complex reinforces actin anchoring. Nat. Commun. 5, 3095 (2014).

    PubMed  Google Scholar 

  52. Pfeifer, A., Kessler, T., Silletti, S., Cheresh, D. A. & Verma, I. M. Suppression of angiogenesis by lentiviral delivery of PEX, a noncatalytic fragment of matrix metalloproteinase 2. Proc. Natl Acad. Sci. USA 97, 12227–12232 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Frojmovic, M. M., O’ Toole, T. E., Plow, E. F., Loftus, J. C. & Ginsberg, M. H. Platelet glycoprotein IIb-IIIa (α IIb β 3 integrin) confers fibrinogen- and activation-dependent aggregation on heterologous cells. Blood 78, 369–376 (1991).

    CAS  PubMed  Google Scholar 

  54. Shattil, S. J., Hoxie, J. A., Cunningham, M. & Brass, L. F. Changes in the platelet membrane glycoprotein IIb.IIIa complex during platelet activation. J. Biol. Chem. 260, 11107–11114 (1985).

    CAS  PubMed  Google Scholar 

  55. Azioune, A., Carpi, N., Tseng, Q., Thery, M. & Piel, M. Protein Micropatterns: a Direct Printing Protocol Using Deep UVs. Method Cell Biol. 97, 133–146 (2010).

    CAS  Google Scholar 

Download references

Acknowledgements

We thank N. Nagaraj for technical support, and C. Grashoff, O. Rosslier, G. Giannone and R. Böttcher for discussions and reading of the manuscript. The work was supported by the National Institutes of Health R01GM112998 (to A.R.D.), Stanford Graduate Fellowship (to S.T.) and ERC, DFG and Max Planck Society (to R.F.).

Author information

Authors and Affiliations

Authors

Contributions

Z.S. and R.F. initiated the project, designed the experiments and wrote the paper; Z.S. performed the vast majority of the experiments; H.-Y.T., S.T., F.S., L.K., D.D. and A.A.W. performed experiments; Z.S., S.T., N.M., M.T., A.R.D. and R.F. analysed data; all authors read and approved the manuscript.

Corresponding author

Correspondence to Reinhard Fässler.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1

(a) SILAC ratio plot showing specific β1wt tail interactors with high β1 wt/β1 scr and low β1 scr/β1 wt SILAC ratios in reverse SILAC labeling experiments and with high MS intensities in the FA-enriched proteome of FN-seeded fibroblasts in red. Dab-2 (green) and SNX17 (grey) show a high β1 wt/β1 scr SILAC ration but were absent from the FA-enriched proteome. (b) Split channels of Fig. 1b. Mouse fibroblasts seeded on FN for 3h and immunostained for Kank2 (green), Kindlin-2 (red) and DAPI (blue). (c) Mouse fibroblasts seeded on FN for 3h and immunostained for Kank2 (green), Talin (red) and DAPI (blue). (d) Confocal image of immunostained (Kank2, green; Kindlin-2, red; DAPI, blue) wild-type fibroblasts seeded for 3h on a FN-coated micropattern. The boxed areas show Kank2 puncta around paxillin-positive FAs. (e) Mouse fibroblasts seeded on FN for 5h and immunostained for β1 integrin (Itgb1, green), β3 integrin (Itgb3, red), Paxillin (Pxn, blue) and DAPI (grey). (f,g) Mouse fibroblasts expressing Kank2-GFP seeded for 5h on FN and immunostained for β1 integrin (Itgb1, red) or active β1 integrin (9EG7, red), Paxillin (Pxn, blue) and DAPI (grey). Scale bar in b-g, 10 μm.

Supplementary Figure 2

(a) MS intensities of Kank1-4 in the FA-enriched sub-proteome and total proteome of wild-type mouse kidney fibroblasts (mean ± s.d.; n = 3 independent MS measurements). (b) Western blot analysis of Kank2 and β1 integrin (Itgb1) in cells expressing scrambled (shScr) and Kank2-specific shRNAs. Actin served to control protein loading. (c) Western blot analysis of Kank2 before and after shRNA-mediated knockdown using polyclonal anti-Kank2, and of indicated GFP-tagged Kank2 constructs using anti-GFP antibodies, respectively. Vinculin is used as loading control.

Supplementary Figure 3

(a,b) Immunofluorescence of GFP-tagged Kank2 constructs (green) co-stained for Liprin-β1 (a, red) or ELKS (b, red), Paxillin (Pxn; blue) and DAPI (grey). (c) Line profile analysis of GFP-tagged FL-Kank2 co-stained with Liprin-β1 and ELKS. Full arrowheads indicate the position of FA belt and open arrowheads indicate Liprin-β1- and ELKS-enriched regions outside the FA belt. Scale bar, 2 μm. (d,e) Western blot testing GFP-tagged Kank-KN (d), FL-Kank2 and Kank2ΔKN (e) binding to biotinylated β1 integrin tails. FL and mutant Kank2 proteins were detected with the anti-GFP antibody, and Kindlin-2 was used to control the β1 tail pull-down assay. (f) GFP-tagged Kank1, Kank3 and Kank4 expressed in Kank2-depleted fibroblasts and co-stained for Paxillin (Pxn; red), F-actin (blue) and DAPI (grey). Scale bars in a,b,f, 10 μm.

Supplementary Figure 4

(a) Time lapse images of cells stably expressing GFP-tagged FL-Kank2 (green) and Paxillin-TagRFP (Pxn-TagRFP; red) during isotropic spreading on FN. Arrow heads indicate Kank2 recruitment to the proximal FA border. (b) Normalized Pxn-TagRFP intensities during FA disassembly behind lamella in cells expressing FL-Kank2 (red) or Kank2ΔKN (blue) (mean ± s.d.; n = 8 cells for FL-Kank2, and n = 10 cells for Kank2ΔKN pooled from 4 independent experiments). (c) FA disassembly rates calculated from (b) as rate constants in a one-phase exponential decay (mean ± s.d.). (d) Ten overlaid, sequential time lapse images of Vinculin-mCherry in the cell centre temporally colour-coded using spectrum look up tables (LUT). Corresponding time lapse images of GFP are shown as projection of average intensity. Scale bar, 5 μm. (e) Percentage of adhesion sites with sliding velocity above 0.3 μm min−1 in indicated cell lines (mean ± s.d., n = 5 cells from 3 independent experiments; P values were calculated using one-way ANOVA Tukey test). (f) 2-D random migration of indicated cells on FN (box plot with median, 1st–3rd quantile box, minimal to maximal whiskers; shScr, n = 52 cells; shKank2#1, n = 58 cells; shKank2#2, n = 60 cells; data aggregated from 4 independent experiments; P values were calculated using Student t-test). (g) Cell spreading area of indicated cells at different time points (n ≥ 50 cells for each condition). (h) Percentage of adhesion sites with sliding velocity above 0.3 μm min−1 in Kank2-depleted fibroblasts stably expressing Paxillin-TagRFP and Kank2-GFP on FN treated with or without 50 μM broad spectrum MMP inhibitor Gm6001 for 5h (mean ± s.d., n = 5 cells from 3 independent experiments).

Supplementary Figure 5

(a) Western blot analysis of Kank2 and Liprin-β1 in wild-type cells expressing FL-Kank2-GFP together with scrambled (shScr) and two Liprin-β1-specific shRNAs. Vinculin served to control protein loading. (b) 2D correlation coefficient between GFP and inverted FRET signals in FL-Kank2-GFP expressing cells before and after Liprin-β1 depletion (dot plot and box plot; FL-Kank2 shScr, n = 30 cells; FL-Kank2 shLiprin-β1#1, n = 31 cells; FL-Kank2 shLiprin-β1#2, n = 33 cells; data aggregated from 3 independent experiments for each condition; P values were calculated using the Wilcoxon Rank Sum test). (c) Ratio between adhesion area with high tension (≥250 pN μm−2) and adhesion area with low tension (<250 pN μm−2) were calculated in cells expressing indicated constructs (dot plot and box plot; FL-Kank2, n = 24 cells; Kank2ΔCoil, n = 45 cells; FL-Kank2 shScr, n = 30 cells; FL-Kank2 shLiprin-β1#1, n = 31 cells; FL-Kank2 shLiprin-β1#2, n = 33 cells; data aggregated from 3 independent experiments for each condition; P values were calculated using the Wilcoxon Rank Sum test).

Supplementary Figure 6

(a) SDS-PAGE analysis of bacterially expressed and purified FL-Talin-1, Talin head domain (THD), Venus-His-Sumo-tagged Talin R4-R8 domain, GST-KN fusion protein and GST. (b) HEK293 cells expressing GFP-tagged Talin rod truncations were lysed, incubated with recombinant GST-KN, immunoprecipitated with anti-GFP antibody and analyzed with Western blot. Red asterisk marks the recombinant GST-KN protein.

Supplementary Figure 7

(a) Western blot and (b) densitometry of Talin binding to β1 integrin tails in cells treated with increasing doses of dexamethasone (Dox) to induce expression of either FL-Kank2 or Kank2ΔKN (mean ± s.d.; n = 3 independent experiments; P values were calculated using Student t-test). Asterisk marks additional Kank2 bands after transient induction of protein expression, which is likely due to post-translational modification. (c,d) Representative Western blot (c) and densitometry (d) of endogenous Kank2, Talin and Kindlin-2 binding to β1 integrin tails in the presence of increasing concentrations of recombinant KN-GST. (e) Western blot of endogenous Kank2, Talin and Kindlin-2 binding to β1 integrin tails in the presence of increasing concentrations of chemically synthesized KN peptide. (f) SDS-PAGE analysis of FL-Kank2 expressed and purified from HEK293 cells. (g) Western blot analysis of recombinant Talin-1 binding to β1 integrin tail in the presence of the KN peptide and increasing concentration of recombinant THD. (h) Western blot analysis of β1 integrin tail pull downs of endogenous Talin detected with an antibody recognizing THD in cells overexpressing the GFP-tagged Talin rod domain (Talin-Rod-GFP) in the presence or absence of KN peptide. Source data for b can be found in Supplementary Table 2.

Supplementary Figure 8

(a) Immunofluorescence of FL-Kank2-GFP (green) co-stained with endogenous RhoGDIα (red) and DAPI (blue). Scale bar, 10 μm. (b) Western blot (left) and densitometric (right) analysis of RhoA activation in Kank2-depleted cells expressing indicated Kank2 constructs seeded on FN for 1h (mean ± s.d.; n = 3 independent experiments; P values were calculated using one-way ANOVA Tukey test; N.S., no significance). (c) Western blot (left) and densitometric (right) analysis of myosin light chain (MLC) phosphorylation at serine-19 (pMLC-Ser19) in Kank2-depleted cells expressing indicated Kank2 constructs seeded on FN for 1h. Actin was used to control protein loading (mean ± s.d.; n = 4 independent experiments; P values were calculated using one-way ANOVA Tukey test; N.S., no significance). (d) Still images from a representative time lapse FRAP experiment of Talin-1-GFP shown in rainbow LUT. Talin-1-GFP was co-expressed with Cherry-tagged FL-Kank2 or Kank2ΔKN, respectively, in Kank2-deplted fibroblasts. Co-localization of Kank2-mCherry and Talin-1-GFP in the region of interest (ROI, red circle) was confirmed in pre-bleached images. Scale bar, 2 μm. Source data for b,c can be found in Supplementary Table 2.

Supplementary information

Supplementary Information

Supplementary Information (PDF 2890 kb)

Supplementary Table 1

Supplementary Information (XLSX 9 kb)

Supplementary Table 2

Supplementary Information (XLSX 14 kb)

FL-Kank2-GFP (green) and Paxillin-TagRFP (red) were stably expressed in Kank2-depleted mouse fibroblasts.

Confocal live cell imaging during isotropic cell spreading 5 min after plating on FN in both fluorescence channels and phase contrast. Scale bar, 10 μm. (AVI 1586 kb)

FL-Kank2-GFP (green) and Paxillin-TagRFP (red) were stably expressed in Kank2-depleted mouse kidney fibroblast.

Confocal live cell imaging of polarized cells 45 min after plating on FN in both fluorescence channels and phase contrast. Scale bar, 10 μm. (AVI 9783 kb)

FL-Kank2-GFP (green) and Paxillin-TagRFP (red) were stably expressed in Kank2-depleted mouse kidney fibroblast.

Confocal live cell imaging of migrating cells 4 h after plating on FN in both fluorescence channels and phase contrast. Scale bar, 10 μm. (AVI 6606 kb)

Kank2ΔKN-GFP (green) and Paxillin-TagRFP (red) were stably expressed in Kank2-depleted mouse kidney fibroblast.

Confocal live cell imaging of polarized cells 45 min after plating on FN in both fluorescence channels and in phase contrast. Scale bar, 10 μm. (AVI 7998 kb)

FL-Kank2-GFP (green) and Vinculin-mCherry (red) were expressed in Kank2-depleted mouse kidney fibroblast.

Confocal live cell imaging of migrating cells 4 h after plating on FN in both fluorescence channels and phase contrast. Scale bar, 10 μm. (AVI 6758 kb)

Kank2ΔKN-GFP (green) and Vinculin-mCherry (red) were expressed in Kank2-depleted mouse kidney fibroblast.

Confocal live cell imaging of migrating cells 4 h after plating on FN in both fluorescence channels and phase contrast. Scale bar, 10 μm. (AVI 3070 kb)

Kank2ΔCoil-GFP (green) and Vinculin-mCherry (red) were expressed in Kank2-depleted mouse kidney fibroblast.

Confocal live cell imaging of migrating cells 4h after plating on FN in both fluorescence channels and phase contrast. Scale bar, 10 μm. (AVI 5110 kb)

Kank2(1–670)-GFP (green) and Vinculin-mCherry (red) were expressed in Kank2-depleted mouse kidney fibroblast.

Confocal live cell imaging of migrating cells 4h after plating on FN in both fluorescence channels and phase contrast. Scale bar, 10 μm. (AVI 5302 kb)

GFP-tagged Kank2-KN (green) and Vinculin-mCherry (red) were expressed in Kank2-depleted mouse kidney fibroblast.

Confocal live cell imaging of migrating cells 4h after plating on FN in both fluorescence channels and phase contrast. Scale bar, 10 μm. (AVI 3059 kb)

GFP control (green) and Vinculin-mCherry (red) were expressed in Kank2-depleted mouse kidney fibroblast.

Confocal live cell imaging of migrating cells 4h after plating on FN in both fluorescence channels and in phase contrast. Scale bar, 10 μm. (AVI 0 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Sun, Z., Tseng, HY., Tan, S. et al. Kank2 activates talin, reduces force transduction across integrins and induces central adhesion formation. Nat Cell Biol 18, 941–953 (2016). https://doi.org/10.1038/ncb3402

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb3402

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