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.

  • Protocol
  • Published:

The use of differential scanning fluorimetry to detect ligand interactions that promote protein stability

Abstract

Differential scanning fluorimetry (DSF) is a rapid and inexpensive screening method to identify low-molecular-weight ligands that bind and stabilize purified proteins. The temperature at which a protein unfolds is measured by an increase in the fluorescence of a dye with affinity for hydrophobic parts of the protein, which are exposed as the protein unfolds. A simple fitting procedure allows quick calculation of the transition midpoint; the difference in the temperature of this midpoint in the presence and absence of ligand is related to the binding affinity of the small molecule, which can be a low-molecular-weight compound, a peptide or a nucleic acid. DSF is best performed using a conventional real-time PCR instrument. Ligand solutions from a storage plate are added to a solution of protein and dye, distributed into the wells of the PCR plate and fluorescence intensity measured as the temperature is raised gradually. Results can be obtained in a single day.

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: Optical properties and fluorescence signal in the presence of lysozyme (native versus denatured) for selected dyes that can be used for DSF.
Figure 2: Typical recording of fluorescence intensity versus temperature for the unfolding of protein (citrate synthase) in the presence of SYPRO orange.
Figure 3: Optimum excitation of SYPRO orange in protein buffer and gain in fluorescence upon protein unfolding.
Figure 4: Layout for a 96-well plate, enabling screening of sets of 29 compounds under three different conditions in parallel.
Figure 5: Concentration-dependent stabilization of citrate synthase by oxaloacetate.
Figure 6: Visualization of the screen results for citrate synthase against physiologically relevant compounds.

Similar content being viewed by others

References

  1. Schellman, J.A. Temperature, stability, and the hydrophobic interaction. Biophys. J. 73, 2960–2964 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Privalov, P.L. Stability of proteins: small globular proteins. Adv. Protein Chem. 33, 167–241 (1979).

    Article  CAS  PubMed  Google Scholar 

  3. Brandts, J.F. Study of strong to ultratight protein interactions using differential scanning calorimetry. Biochemistry 29, 6927–6940 (1990).

    Article  CAS  PubMed  Google Scholar 

  4. Matulis, D., Kranz, J.K., Salemme, F.R. & Todd, M.J. Thermodynamic stability of carbonic anhydrase: measurements of binding affinity and stoichiometry using ThermoFluor. Biochemistry 44, 5258–5266 (2005).

    Article  CAS  PubMed  Google Scholar 

  5. Senisterra, G.A. et al. Screening for ligands using a generic and high-throughput light-scattering-based assay. J. Biomol. Screen. 11, 940–948 (2006).

    Article  CAS  PubMed  Google Scholar 

  6. Vedadi, M. et al. Chemical screening methods to identify ligands that promote protein stability, protein crystallization, and structure determination. Proc. Natl. Acad. Sci. USA 103, 15835–15840 (2006).

    Article  CAS  PubMed  Google Scholar 

  7. Holdgate, G.A. & Ward, W.H. Measurements of binding thermodynamics in drug discovery. Drug Discov. Today 10, 1543–1550 (2005).

    Article  CAS  PubMed  Google Scholar 

  8. Poklar, N., Lah, J., Salobir, M., Macek, P. & Vesnaver, G. pH and temperature-induced molten globule-like denatured states of equinatoxin II: a study by UV-melting, DSC, far- and near-UV CD spectroscopy, and ANS fluorescence. Biochemistry 36, 14345–14352 (1997).

    Article  CAS  PubMed  Google Scholar 

  9. Pantoliano, M.W. et al. High-density miniaturized thermal shift assays as a general strategy for drug discovery. J. Biomol. Screen. 6, 429–440 (2001).

    Article  CAS  PubMed  Google Scholar 

  10. Lo, M.-C. et al. Evaluation of fluorescence-based thermal shift assays for hit identification in drug discovery. Anal. Biochem. 332, 153–159 (2004).

    Article  CAS  PubMed  Google Scholar 

  11. Ericsson, U.B., Hallberg, M.B., DeTitta, G.T., Dekker, N. & Nordlund, P. Thermofluor-based high-throughput stability optimization of proteins for structural studies. Anal. Biochem. 357, 289–298 (2006).

    Article  CAS  PubMed  Google Scholar 

  12. Epps, D.E., Sarver, R.W., Rogers, J.M., Herberg, J.T. & Tomich, P.K. The ligand affinity of proteins measured by isothermal denaturation kinetics. Anal. Biochem. 292, 40–50 (2001).

    Article  CAS  PubMed  Google Scholar 

  13. Plotnikov, V. et al. An autosampling differential scanning calorimeter instrument for studying molecular interactions. Assay Drug Dev. Technol. 1 1 (Part 1): 83–90 (2002).

    Article  CAS  PubMed  Google Scholar 

  14. Pace, C.N., Vajdos, F., Fee, L., Grimsley, G. & Gray, T. How to measure and predict the molar absorption coefficient of a protein. Protein Sci. 4, 2411–2423 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Greenfield, N.J. Using circular dichroism spectra to estimate protein secondary structure. Nat. Protoc. 1, 2876–2890 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Dawson, R.M.C., Elliott, D.C., Elliott, W.H. & Jones, K.M. Data for Biochemical Research 3rd edn. (Oxford University Press, Oxford, UK, 1986).

    Google Scholar 

Download references

Acknowledgements

We are grateful to Oleg Y. Fedorov and Guillermo Senisterra for fruitful discussions, and to Aled M. Edwards and Patrick J. Finerty Jr. for critically reading the manuscript. The Structural Genomics Consortium is a registered charity (number 1097737) that receives funds from the Canadian Institutes for Health Research, the Canadian Foundation for Innovation, Genome Canada through the Ontario Genomics Institute, GlaxoSmithKline, Karolinska Institutet, the Knut and Alice Wallenberg Foundation, the Ontario Innovation Trust, the Ontario Ministry for Research and Innovation, Merck & Co. Inc., the Novartis Research Foundation, the Swedish Agency for Innovation Systems, the Swedish Foundation for Strategic Research and the Wellcome Trust.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Frank H Niesen.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Rights and permissions

Reprints and permissions

About this article

Cite this article

Niesen, F., Berglund, H. & Vedadi, M. The use of differential scanning fluorimetry to detect ligand interactions that promote protein stability. Nat Protoc 2, 2212–2221 (2007). https://doi.org/10.1038/nprot.2007.321

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nprot.2007.321

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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