Skip to main content
Log in

Label-free quantification of membrane-ligand interactions using backscattering interferometry

  • Letter
  • Published:

From Nature Biotechnology

View current issue Submit your manuscript

Abstract

Although membrane proteins are ubiquitous within all living organisms and represent the majority of drug targets, a general method for direct, label-free measurement of ligand binding to native membranes has not been reported. Here we show that backscattering interferometry (BSI) can accurately quantify ligand-receptor binding affinities in a variety of membrane environments. By detecting minute changes in the refractive index of a solution, BSI allows binding interactions of proteins with their ligands to be measured at picomolar concentrations. Equilibrium binding constants in the micromolar to picomolar range were obtained for small- and large-molecule interactions in both synthetic and cell-derived membranes without the use of labels or supporting substrates. The simple and low-cost hardware, high sensitivity and label-free nature of BSI should make it readily applicable to the study of many membrane-associated proteins of biochemical and pharmacological interest.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Figure 1: Experimental components.
Figure 2: Representative plots of BSI signal versus ligand concentration for the determination of binding constants for the following pairs of molecules (membrane-bound species + ligand).

Similar content being viewed by others

References

  1. Krummel, M.F. & Davis, M.M. Dynamics of the immunological synapse: finding, establishing and solidifying a connection. Curr. Opin. Immunol. 14, 66–74 (2002).

    Article  CAS  Google Scholar 

  2. Overington, J.P., Al-Lazikani, B. & Hopkins, A.L. How many drug targets are there? Nat. Rev. Drug Discov. 5, 993–996 (2006).

    Article  CAS  Google Scholar 

  3. Wise, A., Gearing, K. & Rees, S. Target validation of G-protein coupled receptors. Drug Discov. Today 7, 235–246 (2002).

    Article  CAS  Google Scholar 

  4. Markov, D.A., Swinney, K. & Bornhop, D.J. Label-free molecular interaction determinations with nanoscale interferometry. J. Am. Chem. Soc. 126, 16659–16664 (2004).

    Article  CAS  Google Scholar 

  5. Sorensen, H.S., Larsen, N.B., Latham, J.C., Bornhop, D.J. & Andersen, P.E. Highly sensitive biosensing based on interference from light scattering in capillary tubes. Appl. Phys. Lett. 89, 151108 (2006).

    Article  Google Scholar 

  6. Bornhop, D.J. et al. Free-solution, label-free molecular interactions studied by back-scattering interferometry. Science 317, 1732–1736 (2007).

    Article  CAS  Google Scholar 

  7. Kussrow, A. et al. Measurement of monovalent and polyvalent carbohydrate-lectin binding by back-scattering interferometry. Anal. Chem. 81, 4889–4897 (2009).

    Article  CAS  Google Scholar 

  8. Sonnino, S., Mauri, L., Chigorno, V. & Prinetti, A. Gangliosides as components of lipid membrane domains. Glycobiology 17, 1R–13R (2007).

    Article  CAS  Google Scholar 

  9. Fishman, P.H., Pacuszka, T. & Orlandi, P.A. Gangliosides as receptors for bacterial enterotoxins. Adv. Lipid Res. 25, 165–187 (1993).

    CAS  PubMed  Google Scholar 

  10. Kuziemko, G.M., Stroh, M. & Stevens, R.C. Cholera toxin binding affinity and specificity for gangliosides determined by surface plasmon resonance. Biochemistry 35, 6375–6384 (1996).

    Article  CAS  Google Scholar 

  11. Fang, Y., Frutos, A. & Lahiri, J. Ganglioside microarrays for toxin detection. Langmuir 19, 1500–1505 (2003).

    Article  CAS  Google Scholar 

  12. Cannon, B. et al. Cholesterol modulated antibody binding in supported lipid membranes as determined by total internal reflectance microscopy on a microfabricated high-throughput glass chip. Langmuir 21, 9666–9674 (2005).

    Article  CAS  Google Scholar 

  13. Brian, A.A. & McConnell, H.M. Allogenic stimulation of cyto-toxic T-cell by supported planar membranes. Proc. Natl. Acad. Sci. USA 81, 6159–6163 (1984).

    Article  CAS  Google Scholar 

  14. Mossman, K.D., Campi, G., Groves, J.T. & Dustin, M.L. Altered TCR signaling from geometrically repatterned immunological synapses. Science 310, 1191–1193 (2005).

    Article  CAS  Google Scholar 

  15. Cravatt, B.F. & Lichtman, A.H. Fatty acid amide hydrolase: an emerging therapeutic target in the endocannabinoid system. Curr. Opin. Chem. Biol. 7, 469–475 (2003).

    Article  CAS  Google Scholar 

  16. Devane, W.A. et al. Isolation and structure of a brain constituent that binds to the cannabinoid receptor. Science 258, 1946–1949 (1992).

    Article  CAS  Google Scholar 

  17. Mileni, M. et al. Binding and inactivation mechanism of a humanized fatty acid amide hydrolase by α-ketoheterocycle inhibitors revealed from co-crystal structures. J. Am. Chem. Soc. 131, 10497–10506 (2009).

    Article  CAS  Google Scholar 

  18. Boger, D.L. et al. Discovery of a potent, selective, and efficacious class of reversible alpha-ketoheterocycle inhibitors of fatty acid amide hydrolase effective as analgesics. J. Med. Chem. 48, 1849–1856 (2005).

    Article  CAS  Google Scholar 

  19. Romero, F.A. et al. Potent and selective alpha-ketoheterocycle-based inhibitors of the anandamide and oleamide catabolizing enzyme, fatty acid amide hydrolase. J. Med. Chem. 50, 1058–1068 (2007).

    Article  CAS  Google Scholar 

  20. Garfunkle, J. et al. Optimization of the central heterocycle of alpha-ketoheterocycle inhibitors of fatty acid amide hydrolase. J. Med. Chem. 51, 4392–4403 (2008).

    Article  CAS  Google Scholar 

  21. Cheng, Y. & Prusoff, W.H. Relationship between the inhibition constant (KI) and the concentration of inhibitor which causes 50 per cent inhibition (I50) of an enzymatic reaction. Biochem. Pharmacol. 22, 3099–3108 (1973).

    Article  CAS  Google Scholar 

  22. Hesselgesser, J. et al. Identification and characterization of the CXCR4 chemokine receptor in human T cell lines: ligand binding, biological activity, and HIV-1 infectivity. J. Immunol. 160, 877–883 (1998).

    CAS  PubMed  Google Scholar 

  23. Pin, J.-P. et al. Activation mechanism of the heterodimeric GABAB receptor. Biochem. Pharmacol. 68, 1565–1572 (2004).

    Article  CAS  Google Scholar 

  24. Urwyler, S. et al. Positive allosteric modulation of native and recombinant gamma-aminobutyric acidB receptors by 2,6-di-tert-butyl-4-(3-hydroxy-2,2-dimethyl-propyl)-phenol (CGP7930) and its aldehyde analog CGP13501. Mol. Pharm. 60, 963–971 (2001).

    Article  CAS  Google Scholar 

  25. Bowery, N.G., Hill, D.R. & Hudson, A.L. [3Hl(-)baclofen: an improved ligand for GABAB sites. Neuropharmacology 24, 207–210 (1985).

    Article  CAS  Google Scholar 

  26. Howson, W., Mistry, J., Broekman, M. & Hills, J.M. Biological activity of 3-aminopropyl (methyl) phosphinic acid, a potent and selective GABAB agonist with CNS activity. Bioorg. Med. Chem. Lett. 3, 515–518 (1993).

    Article  CAS  Google Scholar 

  27. Froestl, W. et al. Phosphinic acid analogs of GABA. 1. New potent and selective GABAB agonists. J. Med. Chem. 38, 3297–3312 (1995).

    Article  CAS  Google Scholar 

  28. Brugger, F., Wicki, U., Olpe, H.R., Froestl, W. & Mickel, S. The action of new potent GABA-B receptor antagonists in the hemisected spinal cord preparation of the rat. Eur. J. Pharmacol. 235, 153–155 (1993).

    Article  CAS  Google Scholar 

  29. Latham, J.C., Stein, R.A., Bornhop, D.J. & Mchaourab, H.S. Free-solution label-free detection of α-crystallin chaperone interactions by back-scattering interferometry. Anal. Chem. 81, 1865–1871 (2009).

    Article  CAS  Google Scholar 

  30. Burns, J.M. et al. A novel chemokine receptor for SDF-1 and I-TAC involved in cell survival, cell adhesion, and tumor development. J. Exp. Med. 203, 2201–2213 (2006).

    Article  CAS  Google Scholar 

  31. Kaupmann, K. et al. Expression cloning of GABAB receptors uncovers similarity to metabotropic glutamate receptors. Nature 386, 239–246 (1997).

    Article  CAS  Google Scholar 

  32. Kussrow, A.K. et al. The potential of backscattering interferometry as an in vitro clinical diagnostic tool for the serological diagnosis of infectious disease. Analyst (Lond.) 135, 1535–1537 (2010).

    Article  CAS  Google Scholar 

  33. Kussrow, A., Baksh, M.M., Bornhop, D.J. & Finn, M.G. Universal sensing by transduction of antibody binding using backscattering interferometry. ChemBioChem 12, 367–370 (2011).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

This work was supported by the US National Institutes of Health (RO1 EB003537-01A2; U01 MH069062; and the Joint Center for Innovative Membrane Protein Technologies, Roadmap Grant GM073197) and The Skaggs Institute for Chemical Biology. We are grateful to J. Garfunkel and D. Boger of The Scripps Research Institute for samples of the FAAH inhibitors, R. Stevens of The Scripps Research Institute for samples of the FAAH protein, M. Hanes and T. Handel of the University of California, San Diego for the samples of the CXCL12 chemokine and K. Kaupmann of Novartis for the GABAB-transfected CHO cell line.

Author information

Authors and Affiliations

Authors

Contributions

M.M.B. developed methods for sample preparation, prepared samples for analysis and processed the raw BSI data; A.K.K. performed BSI measurements and processed the raw data; M.M. prepared the FAAH protein; M.M.B., A.K.K., M.G.F. and D.J.B. designed the project and wrote the manuscript.

Corresponding authors

Correspondence to M G Finn or Darryl J Bornhop.

Ethics declarations

Competing interests

D.J.B. has a financial interest in a company that is commercializing BSI. The other authors declare that they have no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Methods and Supplementary Figs. 1–3 (PDF 854 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Baksh, M., Kussrow, A., Mileni, M. et al. Label-free quantification of membrane-ligand interactions using backscattering interferometry. Nat Biotechnol 29, 357–360 (2011). https://doi.org/10.1038/nbt.1790

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.1790

  • Springer Nature America, Inc.

This article is cited by

Navigation