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:

Structural basis for pure antagonism of integrin αVβ3 by a high-affinity form of fibronectin

Abstract

Integrins are important therapeutic targets. However, current RGD-based anti-integrin drugs are also partial agonists, inducing conformational changes that trigger potentially fatal immune reactions and paradoxical cell adhesion. Here we describe the first crystal structure of αVβ3 bound to a physiologic ligand, the tenth type III RGD domain of wild-type fibronectin (wtFN10), or to a high-affinity mutant (hFN10) shown here to act as a pure antagonist. Comparison of these structures revealed a central π-π interaction between Trp1496 in the RGD-containing loop of hFN10 and Tyr122 of the β3 subunit that blocked conformational changes triggered by wtFN10 and trapped hFN10-bound αVβ3 in an inactive conformation. Removing the Trp1496 or Tyr122 side chains or reorienting Trp1496 away from Tyr122 converted hFN10 into a partial agonist. These findings offer new insights into the mechanism of integrin activation and a basis for the design of RGD-based pure antagonists.

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: Binding properties of hFN10 and wtFN10 to αVβ3.
Figure 2: Structures of αVβ3 bound to FN10.
Figure 3: αVβ3–FN10 interfaces, conformational changes and structure validation.
Figure 4: RGD-containing loop structures in wild-type and modified FN10.

Similar content being viewed by others

Accession codes

Primary accessions

Protein Data Bank

Referenced accessions

Protein Data Bank

References

  1. Hynes, R.O. Integrins: bidirectional, allosteric signaling machines. Cell 110, 673–687 (2002).

    Article  CAS  PubMed  Google Scholar 

  2. Xiong, J.P. et al. Crystal structure of the extracellular segment of integrin αVβ3 . Science 294, 339–345 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Arnaout, M.A., Goodman, S.L. & Xiong, J.P. Structure and mechanics of integrin-based cell adhesion. Curr. Opin. Cell Biol. 19, 495–507 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Xiong, J.P. et al. Crystal structure of the complete integrin αVβ3 ectodomain plus an α/β transmembrane fragment. J. Cell Biol. 186, 589–600 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Honda, S. et al. Topography of ligand-induced binding sites, including a novel cation-sensitive epitope (AP5) at the amino terminus, of the human integrin β3 subunit. J. Biol. Chem. 270, 11947–11954 (1995).

    Article  CAS  PubMed  Google Scholar 

  6. Frelinger, A.L. III, Du, X.P., Plow, E.F. & Ginsberg, M.H. Monoclonal antibodies to ligand-occupied conformers of integrin αIIbβ3 (glycoprotein IIb-IIIa) alter receptor affinity, specificity, and function. J. Biol. Chem. 266, 17106–17111 (1991).

    CAS  PubMed  Google Scholar 

  7. Raborn, J., Wang, W. & Luo, B.H. Regulation of integrin αIIbβ3 ligand binding and signaling by the metal ion binding sites in the βI domain. Biochemistry 50, 2084–2091 (2011).

    Article  CAS  PubMed  Google Scholar 

  8. Friedland, J.C., Lee, M.H. & Boettiger, D. Mechanically activated integrin switch controls α5β1 function. Science 323, 642–644 (2009).

    Article  CAS  PubMed  Google Scholar 

  9. Cox, D., Brennan, M. & Moran, N. Integrins as therapeutic targets: lessons and opportunities. Nat. Rev. Drug Discov. 9, 804–820 (2010).

    Article  CAS  PubMed  Google Scholar 

  10. Gerber, E.E. et al. Integrin-modulating therapy prevents fibrosis and autoimmunity in mouse models of scleroderma. Nature 503, 126–130 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Maile, L.A. et al. A monoclonal antibody against αVβ3 integrin inhibits development of atherosclerotic lesions in diabetic pigs. Sci. Transl. Med. 2, 18ra11 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Aster, R.H., Curtis, B.R., McFarland, J.G. & Bougie, D.W. Drug-induced immune thrombocytopenia: pathogenesis, diagnosis, and management. J. Thromb. Haemost. 7, 911–918 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Xiong, J.P. et al. Crystal structure of the extracellular segment of integrin αVβ3 in complex with an Arg-Gly-Asp ligand. Science 296, 151–155 (2002).

    Article  CAS  PubMed  Google Scholar 

  14. Xiao, T., Takagi, J., Coller, B.S., Wang, J.H. & Springer, T.A. Structural basis for allostery in integrins and binding to fibrinogen-mimetic therapeutics. Nature 432, 59–67 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Mould, A.P., Barton, S.J., Askari, J.A., Craig, S.E. & Humphries, M.J. Role of ADMIDAS cation-binding site in ligand recognition by integrin α5β1 . J. Biol. Chem. 278, 51622–51629 (2003).

    Article  CAS  PubMed  Google Scholar 

  16. Gao, C. et al. Eptifibatide-induced thrombocytopenia and thrombosis in humans require FcγRIIa and the integrin β3 cytoplasmic domain. J. Clin. Invest. 119, 504–511 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Bassler, N. et al. A mechanistic model for paradoxical platelet activation by ligand-mimetic αIIbβ3 (GPIIb/IIIa) antagonists. Arterioscler. Thromb. Vasc. Biol. 27, e9–e15 (2007).

    Article  CAS  PubMed  Google Scholar 

  18. Alghisi, G.C., Ponsonnet, L. & Ruegg, C. The integrin antagonist cilengitide activates αVβ3, disrupts VE-cadherin localization at cell junctions and enhances permeability in endothelial cells. PLoS ONE 4, e4449 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Reynolds, A.R. et al. Stimulation of tumor growth and angiogenesis by low concentrations of RGD-mimetic integrin inhibitors. Nat. Med. 15, 392–400 (2009).

    Article  CAS  PubMed  Google Scholar 

  20. Zhu, J. et al. Structure-guided design of a high-affinity platelet integrin αIIbβ3 receptor antagonist that disrupts Mg2+ binding to the MIDAS. Sci. Transl. Med. 4, 125ra132 (2012).

    Article  Google Scholar 

  21. Bowditch, R.D. et al. Identification of a novel integrin binding site in fibronectin. Differential utilization by β3 integrins. J. Biol. Chem. 269, 10856–10863 (1994).

    CAS  PubMed  Google Scholar 

  22. Richards, J. et al. Engineered fibronectin type III domain with a RGDWXE sequence binds with enhanced affinity and specificity to human αvβ3 integrin. J. Mol. Biol. 326, 1475–1488 (2003).

    Article  CAS  PubMed  Google Scholar 

  23. Scarborough, R.M. Development of eptifibatide. Am. Heart J. 138, 1093–1104 (1999).

    Article  CAS  PubMed  Google Scholar 

  24. Mould, A.P. et al. Integrin activation involves a conformational change in the α1 helix of the β subunit A-domain. J. Biol. Chem. 277, 19800–19805 (2002).

    Article  CAS  PubMed  Google Scholar 

  25. Kendall, T., Mukai, L., Jannuzi, A.L. & Bunch, T.A. Identification of integrin β subunit mutations that alter affinity for extracellular matrix ligand. J. Biol. Chem. 286, 30981–30993 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. 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).

    Article  CAS  PubMed  Google Scholar 

  27. Adair, B.D. et al. Three-dimensional EM structure of the ectodomain of integrin αVβ3 in a complex with fibronectin. J. Cell Biol. 168, 1109–1118 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Cluzel, C. et al. The mechanisms and dynamics of αvβ3 integrin clustering in living cells. J. Cell Biol. 171, 383–392 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Felding-Habermann, B. et al. Involvement of tumor cell integrin αvβ3 in hematogenous metastasis of human melanoma cells. Clin. Exp. Metastasis 19, 427–436 (2002).

    Article  CAS  PubMed  Google Scholar 

  30. Nagae, M. et al. Crystal structure of α5β1 integrin ectodomain: atomic details of the fibronectin receptor. J. Cell Biol. 197, 131–140 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Sato, Y. et al. An N-glycosylation site on the β-propeller domain of the integrin α5 subunit plays key roles in both its function and site-specific modification by β1,4-N-acetylglucosaminyltransferase III. J. Biol. Chem. 284, 11873–11881 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Xie, C. et al. Structure of an integrin with an αI domain, complement receptor type 4. EMBO J. 29, 666–679 (2010).

    Article  CAS  PubMed  Google Scholar 

  34. Goodman, S.L. & Picard, M. Integrins as therapeutic targets. Trends Pharmacol. Sci. 33, 405–412 (2012).

    Article  CAS  PubMed  Google Scholar 

  35. Cheresh, D.A. Human endothelial cells synthesize and express an Arg-Gly-Asp–directed adhesion receptor involved in attachment to fibrinogen and von Willebrand factor. Proc. Natl. Acad. Sci. USA 84, 6471–6475 (1987).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Mehta, R.J. et al. Transmembrane-truncated αvβ3 integrin retains high affinity for ligand binding: evidence for an 'inside-out' suppressor? Biochem. J. 330, 861–869 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Cheng, M. et al. Mutation of a conserved asparagine in the I-like domain promotes constitutively active integrins αLβ2 and αIIbβ3 . J. Biol. Chem. 282, 18225–18232 (2007).

    Article  CAS  PubMed  Google Scholar 

  38. Leahy, D.J., Aukhil, I. & Erickson, H.P. 2.0 A crystal structure of a four-domain segment of human fibronectin encompassing the RGD loop and synergy region. Cell 84, 155–164 (1996).

    Article  CAS  PubMed  Google Scholar 

  39. Mitjans, F. et al. An anti-αv-integrin antibody that blocks integrin function inhibits the development of a human melanoma in nude mice. J. Cell Sci. 108, 2825–2838 (1995).

    CAS  PubMed  Google Scholar 

  40. Otwinowski, Z. & Minor, W. Processing of X-ray diffraction data collected in oscillation mode. Methods Enzymol. 276, 307–326 (1997).

    Article  CAS  PubMed  Google Scholar 

  41. Battye, T.G., Kontogiannis, L., Johnson, O., Powell, H.R. & Leslie, A.G. iMOSFLM: a new graphical interface for diffraction-image processing with MOSFLM. Acta Crystallogr. D Biol. Crystallogr. 67, 271–281 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. McCoy, A.J. et al. Phaser crystallographic software. J. Appl. Crystallogr. 40, 658–674 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Adams, P.D. et al. PHENIX: a comprehensive Python-based system for macromolecular structure solution. Acta Crystallogr. D Biol. Crystallogr. 66, 213–221 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Emsley, P. & Cowtan, K. Coot: model-building tools for molecular graphics. Acta Crystallogr. D Biol. Crystallogr. 60, 2126–2132 (2004).

    Article  PubMed  Google Scholar 

  45. Pettersen, E.F. et al. UCSF Chimera—a visualization system for exploratory research and analysis. J. Comput. Chem. 25, 1605–1612 (2004).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank H.P. Erickson (Duke University) for providing the FN7-10 plasmid, T.J. Kunicki (The Scripps Research Institute) for access to AP5 antibody, M. Ginsberg (University of California, San Diego) for providing LIBS-1 and LIBS-6 mAbs, G.A. Petsko (Brandeis University) for helpful discussions and Z. Ding and D. Mueller-Pompalla for expert technical assistance. This work was supported by grants DK088327, DK48549, DK096334 and DK007540 (M.A.A.) from the National Institute of Diabetes and Digestive and Kidney Diseases, US National Institutes of Health.

Author information

Authors and Affiliations

Authors

Contributions

M.A.A. conceived and designed experiments. J.F.V.A., J.-P.X. and S.L.G. made and purified proteins. J.F.V.A. and J.-P.X. performed the crystallographic studies. J.L.A., X.R., J.F.V.A., M.A.A. and B.D.A. performed the biophysical, biochemical and cell-based assays. M.A.A., J.F.V.A., J.-P.X. and J.L.A. interpreted data. M.A.A. wrote the manuscript with the assistance of S.L.G. and J.-P.X.

Corresponding author

Correspondence to M Amin Arnaout.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Formation of αVβ3–FN10 complexes and fitted standard curve used to measure their Stokes radii.

(a) Coomassie-stained reduced SDS-PAGE gels of size-fractionated αVβ3 following its incubation with wFN10 (lanes 2, 5) or hFN10 (lanes 3, 6) in 1 mM each of Ca2+/Mg2+ or 1 mM Mn2+. 40 μg of protein mixtures were loaded in lanes 2, 3, 5, and 6. Lanes 1, 4 molecular mass markers (small arrows from top to bottom: 250, 150, 100, 75, 50, 37, 25, 20, 15, 10 kDa). αVβ3 formed a stable complex with hFN10 in Ca2+/Mg2+ or Mn2+ but with wtFN10 only in Mn2+. (b) Protein standards were Thyroglobulin (Thy), Ferritin (Fer), Albumin (Alb), Ovalbumin (Ova), chymotrypsinogen A (Chy), and ribonuclease A (Rib). Elution volumes were expressed as the square root of the log ratio of elution volume (Ve)/Void volume (Vo). Linear regression curve fit is plotted.

Supplementary Figure 2 Crystal structures and omit maps of αVβ3–FN10 complexes.

Ribbon diagrams of αVβ3 ectodomains bound to wtFN10 (a) or hFN10 (b). Both integrins are in the same orientation. αV chain is in blue and β3 chain is in light green (a) or pink (b). The four-αV domains (Propeller, Thigh, Calf-1 and Calf-2) and eight domains of β3 (PSI, βA, Hybrid [H], IE1-4 and βTD) are marked in (a). Orange spheres represent the five Mn2+ ions at the base of the Propeller and at the genu of αV. Mn2+ ions at LIMBS (gray), MIDAS (cyan) and ADMIDAS (magenta) are shown as spheres. Glycan carbons are indicated in the respective chain color. (c, d) σA weighted Fo-Fc omit electron density (ED) maps of the ligand-binding site and surrounding area for αVβ3-wtFN10 (c) and αVβ3-hFN10 (d) structures contoured at 4.0 σ, with FN10 protein omitted from the map calculation in each case.

Supplementary Figure 3 Significance of FN-glycan interaction in αVβ3–wtFN10 structure and IE2/Thigh and βTD/βA contacts found in the αVβ3–hFN10 structure.

(a) Adhesion (mean ± SD, n = 3 independent experiments) of HEK293T cells expressing αVβ3N339S or αVN266Qβ3N339S to immobilized full-length FN in presence of Ca2+/Mg2+. A540: absorbance at 540 nm. (b) Ribbon diagram showing the ionic and electrostatic interactions between β3's EGF-like domain 2 (IE2)(in pink) and αV's Thigh domain (in blue). σA weighted 2Fo-Fc map (gray, contoured at 1.0 σ) around the interacting residues is shown. The carboxyl oxygens of the β-genu residue Asp477 in β3-subunit IE2 H-bond to a carboxyl and carbonyl oxygens of Glu547 in the Thigh domain. OE1 and OE2 of the IE2 residue Glu500 H-bond to Glu550 OD1 and to Thr553 OG, respectively, with OE2 also forming a salt bridge with Lys503 of Thigh domain. (c) Ribbon diagram showing the intrachain H-bond between Ser674 in the CD loop of the βTD and Gln319 in the α6 helix of the βA domain. σA weighted 2Fo-Fc maps around Gln319 and the Asp672-Lys676 sequence is shown in gray contoured at 1.0 σ. α helices 1, 6 and 7 and strand F-α7 loop are labeled. The metal ions are colored as in supplementary Fig. 2.

Supplementary Figure 4 Crystal structure of αVβ3–hFN10/B complex.

Ribbon diagram of the αVβ3 ectodomain bound to hFN10/B (in light green). Integrin domains are labeled. Orange spheres represent the five Mn2+ ions at the base of the Propeller and at the αV genu. Glycan carbons are indicated in the respective chain color. Inset, σA weighted Fo-Fc omit map of the ligand-binding site and surrounding area for the αVβ3-hFN10/B structure contoured at 4.0 σ, with hFN10/B protein omitted from map calculation. Mn2+ ions at LIMBS, MIDAS and ADMIDAS are colored as in supplementary Fig. 2.

Supplementary Figure 5 Expression of the AP5 epitope on hFN10/B–bound αVβ3.

Histograms showing binding of fluoresceinated AP5 mAb to K562-αVβ3 cells in absence (control) or presence of wtFN10, hFN10 or hFN10/B in 1 mM MnCl2. Histograms show the results of two independent experiments (Exp).

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–5 (PDF 21877 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Van Agthoven, J., Xiong, JP., Alonso, J. et al. Structural basis for pure antagonism of integrin αVβ3 by a high-affinity form of fibronectin. Nat Struct Mol Biol 21, 383–388 (2014). https://doi.org/10.1038/nsmb.2797

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nsmb.2797

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