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.

  • Letter
  • Published:

A point mutation in KINDLIN3 ablates activation of three integrin subfamilies in humans

Abstract

Monogenic deficiency diseases provide unique opportunities to define the contributions of individual molecules to human physiology and to identify pathologies arising from their dysfunction. Here we describe a deficiency disease in two human siblings that presented with severe bleeding, frequent infections and osteopetrosis at an early age. These symptoms are consistent with but more severe than those reported for people with leukocyte adhesion deficiency III (LAD-III). Mechanistically, these symptoms arose from an inability to activate the integrins expressed on hematopoietic cells, including platelets and leukocytes. Immortalized lymphocyte cell lines isolated from the two individuals showed integrin activation defects. Several proteins previously implicated in integrin activation, including Ras-associated protein-1 (RAP1)1 and calcium and diacylglycerol-regulated guanine nucleotide exchange factor-1 (CALDAG-GEF1)2, were present and functional in these cell lines. The genetic basis for this disease was traced to a point mutation in the coding region of the KINDLIN3 (official gene symbol FERMT3) gene3. When wild-type KINDLIN-3 was expressed in the immortalized lymphocytes, their integrins became responsive to activation signals. These results identify a genetic disease that severely compromises the health of the affected individuals and establish an essential role of KINDLIN-3 in integrin activation in humans. Furthermore, allogeneic bone marrow transplantation was shown to alleviate the symptoms of the disease.

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: Analysis of platelet activation in subjects.
Figure 2: Impaired function of αMβ2, α4β1 and αVβ3 integrins on subjects' cells.
Figure 3: Osteopetrosis in subjects was rescued by BMT.
Figure 4: Subjects have a point mutation in KINDLIN3.

Similar content being viewed by others

Accession codes

Accessions

GenBank/EMBL/DDBJ

References

  1. Hattori, M. & Minato, N. Rap1 GTPase: functions, regulation, and malignancy. J. Biochem. 134, 479–484 (2003).

    Article  CAS  Google Scholar 

  2. Crittenden, J.R. et al. CalDAG-GEFI integrates signaling for platelet aggregation and thrombus formation. Nat. Med. 10, 982–986 (2004).

    Article  CAS  Google Scholar 

  3. Ussar, S., Wang, H.V., Linder, S., Fassler, R. & Moser, M. The Kindlins: subcellular localization and expression during murine development. Exp. Cell Res. 312, 3142–3151 (2006).

    Article  CAS  Google Scholar 

  4. Rogalski, T.M., Mullen, G.P., Gilbert, M.M., Williams, B.D. & Moerman, D.G. The UNC-112 gene in Caenorhabditis elegans encodes a novel component of cell-matrix adhesion structures required for integrin localization in the muscle cell membrane. J. Cell Biol. 150, 253–264 (2000).

    Article  CAS  Google Scholar 

  5. Shi, X. et al. The MIG-2/integrin interaction strengthens cell-matrix adhesion and modulates cell motility. J. Biol. Chem. 282, 20455–20466 (2007).

    Article  CAS  Google Scholar 

  6. Siegel, D.H. et al. Loss of kindlin-1, a human homolog of the Caenorhabditis elegans actin-extracellular-matrix linker protein UNC-112, causes Kindler syndrome. Am. J. Hum. Genet. 73, 174–187 (2003).

    Article  CAS  Google Scholar 

  7. Dowling, J.J. et al. Kindlin-2 is an essential component of intercalated discs and is required for vertebrate cardiac structure and function. Circ. Res. 102, 423–431 (2008).

    Article  CAS  Google Scholar 

  8. Kruger, M. et al. SILAC mouse for quantitative proteomics uncovers kindlin-3 as an essential factor for red blood cell function. Cell 134, 353–364 (2008).

    Article  Google Scholar 

  9. Moser, M., Nieswandt, B., Ussar, S., Pozgajova, M. & Fassler, R. Kindlin-3 is essential for integrin activation and platelet aggregation. Nat. Med. 14, 325–330 (2008).

    Article  CAS  Google Scholar 

  10. Dowling, J.J., Vreede, A.P., Kim, S., Golden, J. & Feldman, E.L. Kindlin-2 is required for myocyte elongation and is essential for myogenesis. BMC Cell Biol. 9, 36 (2008).

    Article  Google Scholar 

  11. Ma, Y.Q., Qin, J., Wu, C. & Plow, E.F. Kindlin-2 (Mig-2): a co-activator of beta3 integrins. J. Cell Biol. 181, 439–446 (2008).

    Article  CAS  Google Scholar 

  12. Alon, R. et al. A novel genetic leukocyte adhesion deficiency in subsecond triggering of integrin avidity by endothelial chemokines results in impaired leukocyte arrest on vascular endothelium under shear flow. Blood 101, 4437–4445 (2003).

    Article  CAS  Google Scholar 

  13. Kuijpers, T.W. et al. Natural history and early diagnosis of LAD-1/variant syndrome. Blood 109, 3529–3537 (2007).

    Article  CAS  Google Scholar 

  14. Quinn, M.J., Byzova, T.V., Qin, J., Topol, E.J. & Plow, E.F. Integrin αIIbβ3 and its antagonism. Arterioscler. Thromb. Vasc. Biol. 23, 945–952 (2003).

    Article  CAS  Google Scholar 

  15. Newman, P.J., Seligsohn, U., Lyman, S. & Coller, B.S. The molecular genetic basis of Glanzmann thrombasthenia in the Iraqi-Jewish and Arab populations in Israel. Proc. Natl. Acad. Sci. USA 88, 3160–3164 (1991).

    Article  CAS  Google Scholar 

  16. 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 

  17. Feng, X. et al. A Glanzmann's mutation in β3 integrin specifically impairs osteoclast function. J. Clin. Invest. 107, 1137–1144 (2001).

    Article  CAS  Google Scholar 

  18. Kasirer-Friede, A., Kahn, M.L. & Shattil, S.J. Platelet integrins and immunoreceptors. Immunol. Rev. 218, 247–264 (2007).

    Article  CAS  Google Scholar 

  19. Ginsberg, M.H., Partridge, A. & Shattil, S.J. Integrin regulation. Curr. Opin. Cell Biol. 17, 509–516 (2005).

    Article  CAS  Google Scholar 

  20. Moyle, M. et al. A hookworm glycoprotein that inhibits neutrophil function is a ligand of the integrin CD11b/CD18. J. Biol. Chem. 269, 10008–10015 (1994).

    CAS  PubMed  Google Scholar 

  21. Miura, Y. et al. Defective osteogenesis of the stromal stem cells predisposes CD18-null mice to osteoporosis. Proc. Natl. Acad. Sci. USA 102, 14022–14027 (2005).

    Article  CAS  Google Scholar 

  22. Kinashi, T. Intracellular signalling controlling integrin activation in lymphocytes. Nat. Rev. Immunol. 5, 546–559 (2005).

    Article  CAS  Google Scholar 

  23. Kinashi, T. et al. LAD-III, a leukocyte adhesion deficiency syndrome associated with defective Rap1 activation and impaired stabilization of integrin bonds. Blood 103, 1033–1036 (2004).

    Article  CAS  Google Scholar 

  24. Alon, R. & Etzioni, A. LAD-III a novel group of leukocyte integrin activation deficiencies. Trends Immunol. 24, 561–566 (2003).

    Article  CAS  Google Scholar 

  25. Mory, A. et al. Kindlin-3: a new gene involved in the pathogenesis of LAD-III. Blood 112, 2591 (2008).

    Article  CAS  Google Scholar 

  26. Moser, M. et al. Kindlin-3 is for required for β2 integrin–mediated leukocyte adhesion to endothelial cells. Nat. Med. advance online publication doi:10.1038/nm.1921 (22 February 2009).

  27. Larjava, H., Plow, E.F. & Wu, C. Kindlins: essential regulators of integrin signalling and cell-matrix adhesion. EMBO Rep. 9, 1203–1208 (2008).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank Roswell Park Cancer Institute Proteomics Resources, Lerner Research Institute Proteomics and Imaging Cores (J. Drazba) and Stranad Fellows I. Byzov and R. Swaninger for help with the project, A.Graybiel (Massachusetts Institute of Technology) for providing antibodies to CALDAG-GEF1 and L. Parise (University of North Carolina) for providing the GST-RALGDS fusion cDNA construct. This study was supported by HL073311 and HL071625 US National Institutes of Health grants.

Author information

Authors and Affiliations

Authors

Contributions

N.L.M. identified the Kindlin-3 mutation, performed molecular biology and protein biochemistry studies and wrote the manuscript; L.Z. contributed to study design and experiments on primary leukocytes from subjects; J.C. performed assays with EGFP-Kindlin-3 rescue and siRNA-mediated KINDLIN3 knockdown and western blotting; A.C. performed microscopy studies and FACS analysis; O.R. performed cell culture work and molecular biology; Y.-Q.M. performed molecular biology and Kindlin-3–specific antibody preparation; E.A.P. performed platelet studies; M.T. performed neutrophil analysis; D.P.L. and A.I.C. performed osteogenesis assays; S.B.S. originally described the subjects, designed clinical studies and wrote the manuscript; E.F.P. designed the studies, interpreted the results and wrote the manuscript; T.V.B. performed experiments with platelets and leukocytes, designed the general strategy, interpreted data and wrote the manuscript.

Corresponding author

Correspondence to Tatiana V Byzova.

Supplementary information

Supplementary Text and Figures

Supplementary Methods, Supplementary Figs. 1–6 and Supplementary Note (PDF 1270 kb)

Supplementary Movie 1

Movie 1 of subject's cells transfected with control GFP vector. Differential Interference contrast (DIC) and fluorescence are shown. Frames were recorded every 5 seconds. Subject's cells float but fail to adhere to fibronectin. (AVI 1015 kb)

Supplementary Movie 2

Movie 2 of subject's cells transfected with kindling-3-GFP fusion construct. Differential Interference contrast (DIC) and fluorescence are shown. Frames were recorded every 5 seconds. Upon expression of KINDLIN-3, subjects cells attach and spread on fibronectin. Note raffles formation and active movement of KINDLIN-3 overexpressing cells. Movies of representative cells are shown. (AVI 1099 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Malinin, N., Zhang, L., Choi, J. et al. A point mutation in KINDLIN3 ablates activation of three integrin subfamilies in humans. Nat Med 15, 313–318 (2009). https://doi.org/10.1038/nm.1917

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.1917

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