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:

Protein-adaptive differential scanning fluorimetry using conformationally responsive dyes

Abstract

Differential scanning fluorimetry (DSF) is a technique that reports protein thermal stability via the selective recognition of unfolded states by fluorogenic dyes. However, DSF applications remain limited by protein incompatibilities with existing DSF dyes. Here we overcome this obstacle with the development of a protein-adaptive DSF platform (paDSF) that combines a dye library ‘Aurora’ with a streamlined procedure to identify protein–dye pairs on demand. paDSF was successfully applied to 94% (66 of 70) of proteins, tripling the previous compatibility and delivering assays for 66 functionally and biochemically diverse proteins, including 10 from severe acute respiratory syndrome coronavirus 2. We find that paDSF can be used to monitor biological processes that were previously inaccessible, demonstrated for the interdomain allostery of O-GlcNAc transferase. The chemical diversity and varied selectivities of Aurora dyes suggest that paDSF functionality may be readily extended. paDSF is a generalizable tool to interrogate protein stability, dynamics and ligand binding.

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

Fig. 1: Resolution of DSF incompatibility with a systematically curated dye library.
Fig. 2: Streamlined creation of paDSF assays for SARS-CoV-2 proteins.
Fig. 3: paDSF enables observation of allostery in OGT.
Fig. 4: Emergence of structural trends within paDSF dyes.

Similar content being viewed by others

Data availability

All data are available in the main text or Supplementary Information. A subset of the Aurora library is only available through a material transfer agreement with North Carolina State University. Raw paDSF data are available at ref. 35, and processed pDSF data at https://doi.org/10.5281/zenodo.10028702 (ref. 44). An annotated video of the primary liquid handling steps of a dye screen is available at ref. 23.

Code availability

All code and source data are available in Supplementary Information or upon request. DSFWorld is available at https://doi.org/10.5281/zenodo.8432909 (ref. 45).

References

  1. Schreiber, S. L. A chemical biology view of bioactive small molecules and a binder-based approach to connect biology to precision medicines. Isr. J. Chem. 59, 52–59 (2019).

    Article  CAS  PubMed  Google Scholar 

  2. Garlick, J. M. & Mapp, A. K. Selective modulation of dynamic protein complexes. Cell Chem. Biol. 27, 986–997 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Chattopadhyay, G. & Varadarajan, R. Facile measurement of protein stability and folding kinetics using a nano differential scanning fluorimeter. Protein Sci. 28, 1127–1134 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Greenfield, N. J. Using circular dichroism collected as a function of temperature to determine the thermodynamics of protein unfolding and binding interactions. Nat. Protoc. 1, 2527–2535 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Freire, E. Differential scanning calorimetry. Methods Mol. Biol. 40, 191–218 (1995).

    CAS  PubMed  Google Scholar 

  6. Atsavapranee, B., Stark, C. D., Sunden, F., Thompson, S. & Fordyce, P. M. Fundamentals to function: quantitative and scalable approaches for measuring protein stability. Cell Syst. 12, 547–560 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. 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 

  8. Semisotnov, G. V. et al. Study of the ‘molten globule’ intermediate state in protein folding by a hydrophobic fluorescent probe. Biopolymers 31, 119–128 (1991).

    Article  CAS  PubMed  Google Scholar 

  9. Simeonov, A. Recent developments in the use of differential scanning fluorometry in protein and small molecule discovery and characterization. Expert Opin. Drug Discov. 8, 1071–1082 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Gao, K., Oerlemans, R. & Groves, M. R. Theory and applications of differential scanning fluorimetry in early-stage drug discovery. Biophys. Rev. 12, 85–104 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  11. Biter, A. B., de la Peña, A. H., Thapar, R., Lin, J. Z. & Phillips, K. J. DSF guided refolding as a novel method of protein production. Sci. Rep. 6, 18906 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Lee, M. E., Dou, X., Zhu, Y. & Phillips, K. J. Refolding proteins from inclusion bodies using differential scanning fluorimetry guided (DGR) protein refolding and melttraceur web. Curr. Protoc. Mol. Biol. 125, e78 (2019).

    Article  PubMed  Google Scholar 

  13. Ristic, M., Rosa, N., Seabrook, S. A. & Newman, J. Formulation screening by differential scanning fluorimetry: how often does it work? Acta Crystallogr. F 71, 1359–1364 (2015).

    Article  CAS  Google Scholar 

  14. Chari, A. et al. ProteoPlex: stability optimization of macromolecular complexes by sparse-matrix screening of chemical space. Nat. Methods 12, 859–865 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Ahmed, S., Bhasin, M., Manjunath, K. & Varadarajan, R. Prediction of residue-specific contributions to binding and thermal stability using yeast surface display. Front. Mol. Biosci. 8, 800819 (2021).

    Article  CAS  PubMed  Google Scholar 

  16. Menzen, T. & Friess, W. High-throughput melting-temperature analysis of a monoclonal antibody by differential scanning fluorimetry in the presence of surfactants. J. Pharm. Sci. 102, 415–428 (2013).

    Article  CAS  PubMed  Google Scholar 

  17. Wu, T. et al. Three essential resources to improve differential scanning fluorimetry (DSF) experiments. Preprint at bioRxiv https://doi.org/10.1101/2020.03.22.002543 (2020).

  18. Alexandrov, A. I., Mileni, M., Chien, E. Y. T., Hanson, M. A. & Stevens, R. C. Microscale fluorescent thermal stability assay for membrane proteins. Structure 16, 351–359 (2008).

    Article  CAS  PubMed  Google Scholar 

  19. Ihmels, H. Dyes in modern organic chemistry. Beilstein J. Org. Chem. 15, 2798–2800 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Condello, C. et al. Structural heterogeneity and intersubject variability of Aβ in familial and sporadic Alzheimer’s disease. Proc. Natl Acad. Sci. USA https://doi.org/10.1073/pnas.1714966115 (2018).

  21. Kuenemann, M. A. et al. Weaver’s historic accessible collection of synthetic dyes: a cheminformatics analysis. Chem. Sci. 8, 4334–4339 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Protein-adaptive DSF data explorer. shinyapps https://padsfdyes.shinyapps.io/Exp1243_heatmap_cache/ (2024).

  23. Gestwicki, J. Dye screening visual protocol. Zenodo https://doi.org/10.5281/zenodo.100231977 (2024).

  24. Schiavina, M., Pontoriero, L., Uversky, V. N., Felli, I. C. & Pierattelli, R. The highly flexible disordered regions of the SARS-CoV-2 nucleocapsid N protein within the 1–248 residue construct: sequence-specific resonance assignments through NMR. Biomol. NMR Assign. 15, 219–227 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Giri, R. et al. Understanding COVID-19 via comparative analysis of dark proteomes of SARS-CoV-2, human SARS and bat SARS-like coronaviruses. Cell. Mol. Life Sci. 78, 1655–1688 (2021).

    Article  CAS  PubMed  Google Scholar 

  26. Cubuk, J. et al. The SARS-CoV-2 nucleocapsid protein is dynamic, disordered, and phase separates with RNA. Nat. Commun. 12, 1936 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Wang, S. et al. Targeting liquid–liquid phase separation of SARS-CoV-2 nucleocapsid protein promotes innate antiviral immunity by elevating MAVS activity. Nat. Cell Biol. 23, 718–732 (2021).

    Article  CAS  PubMed  Google Scholar 

  28. Krafcikova, P., Silhan, J., Nencka, R. & Boura, E. Structural analysis of the SARS-CoV-2 methyltransferase complex involved in RNA cap creation bound to sinefungin. Nat. Commun. 11, 3717 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Lin, S. et al. Crystal structure of SARS-CoV-2 nsp10 bound to nsp14-ExoN domain reveals an exoribonuclease with both structural and functional integrity. Nucleic Acids Res. 49, 5382–5392 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Yoshimoto, F. K. The proteins of severe acute respiratory syndrome coronavirus-2 (SARS CoV-2 or n-COV19), the cause of COVID-19. Protein J. 39, 198–216 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Fu, Z. et al. The complex structure of GRL0617 and SARS-CoV-2 PLpro reveals a hot spot for antiviral drug discovery. Nat. Commun. 12, 488 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Schuller, M. et al. Fragment binding to the Nsp3 macrodomain of SARS-CoV-2 identified through crystallographic screening and computational docking. Sci. Adv. 7, eabf8711 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  33. Virdi, R. S. et al. Discovery of drug-like ligands for the Mac1 domain of SARS-CoV-2 Nsp3. SLAS Discov. https://doi.org/10.1177/2472555220960428 (2020).

  34. Gahbauer, S. et al. Iterative computational design and crystallographic screening identifies potent inhibitors targeting the Nsp3 macrodomain of SARS-CoV-2. Proc. Natl Acad. Sci. USA 120, e2212931120 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Gestwicki, J. Data_S2_dye_screening_results. Zenodo https://doi.org/10.5281/zenodo.10028692 (2023).

  36. Milardi, D., La Rosa, C. & Grasso, D. Extended theoretical analysis of irreversible protein thermal unfolding. Biophys. Chem. 52, 183–189 (1994).

    Article  CAS  PubMed  Google Scholar 

  37. Myers, J. K., Pace, C. N. & Scholtz, J. M. Denaturant m values and heat capacity changes: relation to changes in accessible surface areas of protein unfolding. Protein Sci. 4, 2138–2148 (1995).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Rees, D. C. & Robertson, A. D. Some thermodynamic implications for the thermostability of proteins. Protein Sci. 10, 1187–1194 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Levine, Z. G. & Walker, S. The biochemistry of O-GlcNAc transferase: which functions make it essential in mammalian cells? Annu. Rev. Biochem. 85, 631–657 (2016).

    Article  CAS  PubMed  Google Scholar 

  40. Alteen, M. G. et al. Potent De Novo macrocyclic peptides that inhibit O-GlcNAc transferase through an Allosteric mechanism. Angew. Chem. Int. Ed. Engl. 62, e202215671 (2022).

    Article  PubMed  Google Scholar 

  41. Gestwicki, J. Supplementary Table S2_protein_screening_conditions. Zenodo https://doi.org/10.5281/zenodo.10480848 (2024).

  42. Carpenter, A. Visualizing and analyzing proteins in Python. Medium https://towardsdatascience.com/visualizing-and-analyzing-proteins-in-python-bd99521ccd (2021).

  43. Welcome to UCSF Dye Screening. shinyapps https://ucsfdyescreens.shinyapps.io/home/ (2024).

  44. Gestwicki, J. Data S3_dye_screen_results_raw_data. Zenodo https://doi.org/10.5281/zenodo.10028702 (2023).

  45. taiawu. taiawu/dsfworld: DSFworld ShinyApp website. Zenodo https://doi.org/10.5281/zenodo.8432909 (2023).

Download references

Acknowledgements

Specific material contributions are listed in Supplementary Information. The authors thank M. Jacobson, J. Fraser, K. Shokat, D. Wassarman, D. Schwarz, D. Elnatan, H. Shao, M. Moore and S. Williams for helpful discussions. Proteins were generously donated by A. Ambrose, A. Thwin, D. Wassarman, E. Thompson, A. Freeberg, G. Correy, H. Shao, H. Saunders, J. Swan, M. Janowska, M. Ramirz-Alvarado, M. Ravalin, M. Gupta, M. McCabe, M. Schoof, R. Freilich and V. Long from the labs of M. Arkin, A. Maria Cuervo, J. Fraser, J. Gestwicki, J. Gross, R. Klevit, G. Narlikar, K. Shokat, D. Southworth, R. Stroud, K. Verba, P. Walter and J. Ward. Several SARS-CoV-2 proteins were provided by the UCSF QBI Coronavirus Research Group (QCRG). We are grateful for additional support from M. Capps and Z. Karakesisoglu (Analytik Jena); C. Brown and D. Schubach (Exciton-Luxottica); D. Beniston (ChemBridge Corporation); K. Kitto, the Home Away students; and S. Gahbauer, S. Vigneron, J. Lee, D. Duncan and N. Saul (UCSF). Funding was provided by National Institute of Health U19 (J.E.G., AI171110); Sandler Program for Breakthrough Biomedical Research (J.E.G.); Tau Consortium (J.E.G.); National Science Foundation Graduate Research Fellowship (T.W., 1000259744); National Institute of Standards and Technology (N.R.V., 60NANB19D115); National Institutes of Health K99 (O.T.J., K99NS128717); National Institutes of Health F32 (E.C.C., F32AG076281); National Institutes of Health R01 (J.E.G., N.R.V., Z.M., 1R01GM141299‐01); National Institutes of Health R01 (C.L.P., GM107069, R01GM121507 and R35GM141859); GlycoNet, the Canadian Glycomics Network (D.J.V., CD-1); Canadian Cancer Society Research Institute (D.J.V., CCSRI-706825); and Canada Research Chairs program (D.J.V., Tier I CRC in Chemical Biology).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization: T.W., J.E.G., M.G.A. and D.J.V. Data curation: T.W. Formal analysis: T.W. and Z.J.G.-D. Funding acquisition: T.W. and J.E.G. Investigation: T.W., J.C.Y., A.S., M.G.A., A.S.W., Z.M., C.L.P., N.R.V. and D.F. Methodology: T.W., J.C.Y. and A.S. Visualization: T.W. Project administration: J.E.G. Supervision: J.E.G. Writing—original draft: T.W., E.C.C., O.T.J., Z.J.G.-D. and J.E.G. Writing—review and editing: T.W., M.G.A., D.J.V. and J.E.G.

Corresponding author

Correspondence to Jason E. Gestwicki.

Ethics declarations

Competing interests

J.E.G. is a consultant for Contour and DiCE and a consultant and shareholder for Protego and Kaizen. All other authors declare that they have no competing interests.

Peer review

Peer review information

Nature Biotechnology thanks Matthew Groves, Bridget Wagner and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Representative examples of paDSF screening results.

For full dye screen results for all proteins, including comparisons of raw paDSF curves collected for each protein with different dyes, see data availability statement. (a) Results of a dye screen against the SYPRO-incompatible protein nuclear-hormone receptor 23. Far left panel: representative unsuccessful DSF conducted with SYPRO Orange. Right panels: examples of successful paDSF data, collected with six different paDSF dyes. Plots show dyes at four different concentrations, demonstrating a dose-responsive relationship between RFU and dye concentration. (b) Representative examples of resolution of four archetypes of SYPRO Orange incompatibility by paDSF dyes. (c) Examples of successful paDSF data collected for four additional proteins. As in panel A, dyes are plots show dyes at four different concentrations, demonstrating a dose-responsive relationship between RFU and dye concentration.

Extended Data Fig. 2 Apparent melting temperatures for paDSF dyes and SYPRO Orange.

Plots include data for the 60 protein test set, as well as the tested 10 SARS-CoV2 proteins. (a) For the subset of tested proteins compatible with SYPRO Orange, comparison of Tmas reported by Aurora library dyes and SYPRO Orange. Tmas between SYPRO Orange and Aurora dyes for the same protein are strongly correlated, with a Pearson R value of 0.97, p = 2.2 e-16. Statistical significance was determined by one-way t-test. Data shown includes 2,211 unique pairings(b) Tmas reported by all hit dyes from the Aurora library for all tested proteins. Green points = Mean Tma between all hit Aurora dyes for a given protein. Error bars represent ± standard error of Tmas between different Aurora dyes. Experiments were performed in triplicate. For proteins compatible with SYPRO Orange, Tma reported by SYPRO Orange is displayed as orange ‘X’s. Grey points: Tma of minor unfolding transitions, defined as secondary or tertiary transitions with lower RFU in a given unfolding curve.

Extended Data Fig. 3 Screenshot of interactive online database for all dye screen data.

All dye screen data is presented in an interactive form at https://padsfdyes.shinyapps.io/Exp1243_heatmap_cache/. For full dye screen results for all proteins, including comparisons of raw paDSF curves collected for each protein with different dyes, see Extended Data Fig. 2.

Extended Data Fig. 4 Apparent thermal shifts with paDSF dyes for SARS-CoV2 proteins and resolution of screening artifacts.

(a) Thermal upshift in the presence of ADP ribose with nsp3 mac1, with 6 paDSF dyes and SYPRO orange. Error bars represent +/- standard deviation. Experiments were performed in triplicate. (b) Thermal upshift in the presence of GRL0617 with PLPro, with 3 paDSF dyes. Representative curves are shown. (c) Protein-independent activation of SYPRO Orange observed for compounds which produced uninterpretable raw DSF data from a 320-compound pyridine pilot screen using SYPRO Orange. Representative curves are shown. (d) Comparison of DSF data collected using SYPRO Orange and dye T004, demonstrating improved compound compatibility of T004 over SYPRO Orange. Individual duplicates are shown.

Extended Data Fig. 5 Raw and analyzed paDSF with OGT, catalytic domains and TPR domain.

(a) Structural model of full-length OGT, based on crystal structures of the TPR (PDB 1W3B) and catalytic (PDB 5C1D) domains. Cartoons of the (b) full-length OGT construct, which includes the full TPR and catalytic domains, (c) catalytic domain construct, which includes the full catalytic domain and TPR repeats 9–13.5, and (d) TPR domain construct, which includes TPR repeats 1-10. (e–h) paDSF results from the 10 hit dyes for OGT, with raw RFU data displayed alongside fitted models for (E) OGT alone, fitted to DSFworld Model 4 (two sigmoids, with initial RFU (F) catalytic domain alone, fitted to DSFworld Model 2 (one sigmoids, with initial RFU (G) TPR domain alone, fitted to DSFworld Model 2 (one sigmoids, with initial RFU (H) overlay of raw data and final fits for all three constructs. (i) Tmas for the three constructs, extracted using the fits displayed in panels A-D. Data are presented as the mean of three technical replicates +/- standard deviation. No significant (ns) difference was found between Tma OGT-1 and Tma cat, nor Tma OGT-2 and Tma TPR. Significance determined using two-tailed t-test, and a significance threshold of p < 0.01.

Extended Data Fig. 6 Thermal shifts with UDP-GlcNAc from TW408 paDSF.

paDSF results using dye TW408 to monitor titration of UDP-GlcNAc with OGT, catalytic domain, or TPR domain. (a) Schematic of interaction of UDP-GlcNAc with full length and isolated catalytic domain, but not TPR domain, of OGT. (b) Raw RFU for each construct with 0–50 µM UltraPure. UDP-GlcNAc. Top panels: full temperature range (30–80 °C), bottom panels: zoomed in on temperatures relevant to the unfolding transition. (c) Comparison of resulting ∆Tmas. Left panel: Tma OGT1 and Tma cat; right panel: Tma OGT2 and Tma TPR. Thermal upshift occurs in Tma OGT1 and Tma cat (Tma OGT1 Spearman estimate = 0.92, p = 1.13e-10; Tma cat Spearman estimate = 0.94, p = 1.48e-12); but not Tma OGT2 or Tma TPR (Tma OGT2 Spearman estimate = 0.095, p = 0.66; Tma TPR Spearman estimate = 0.005, p = 0.98). ns = not significant (p > 0.05); *** p < 0.0001. Statistical significance was determined by two-way t-test. Data are the mean of three technical replicates +/- standard deviation.

Extended Data Fig. 7 Thermal shifts with L4 from TW408 paDSF.

paDSF results using dye TW408 to monitor titration of L4 with OGT, catalytic domain, or TPR domain. (a) Schematic of L4 with the four tested conditions: full OGT, catalytic domain, TPR domain, and a 1:1 mix of catalytic and TPR domain. All constructs were tested at 1 µM final; 1:1 mix had 1 µM catalytic domain and 1 mM TPR domain. (b) Raw RFU for each construct in the presence of 0–10 µM L4. (c) Comparison of resulting Tmas. Left panel: Tma OGT1, Tma cat:TPR 1, and Tma cat; right panel: Tma OGT2, Tma cat:TPR 2, and Tma TPR. Significant thermal upshifts with L4 treatment observed with Tma OGT2, Tma cat:TPR 2, and Tma TPR (Tma OGT2 spearman estimate = 0,95, p = 2.3e-12; Tma cat:TPR 2 spearman estimate = 0.97, p = 4.7e-15; Tma TPR spearman estimate = 0.96, p = 3.6e-14). Significant thermal downshift with L4 observed in Tma OGT1 (data in Fig. 3d, included here for reference, Tma OGT1 spearman estimate = −0.92, p = 6.2e-11). No dose-responsive thermal shift with L4 observed in Tma cat or Tma cat:TPR 1 (Tma cat spearman estimate = −0.27, p = 0.2; Tma cat:TPR 1 spearman estimate = −0.34, p = 0.09). ns = not significant (p > 0.05); *** p < 0.0001. Statistical significance was determined by two-way t-test. Data are presented as the mean of three technical replicates +/- standard deviation.

Extended Data Fig. 8 OGT oligomerization state is not affected by L4, but L4 treatment increases the temperature sensitivity of OGT catalytic activity.

(a) Histograms of counts of MW from mass photometry, used to calculate molecular weights: OGT: 115 kDa, Catalytic domain; 82 kDa, TPR domain: 43, 86, 129 kDa (monomer, dimer, trimer). (b) OGT catalytic activity with a model substrate peptide (KKKYPGGSTPVSSANMM), with enzymatic activity allowed to proceed at 29, 32, 35, 38, 41, 44, 47, or 50 °C. Activity was measured as RLU using Promega UDP-Glo Assay, with higher RLU values corresponding to higher activity. Comparison of temperature-dependent loss of enzymatic activity (RLU) in the presence of increasing concentrations of L4. Solid lines and dots: OGT-containing samples; Dashed lines and Xs: protein-free control. (c) L4-dependent increase in temperature sensitivity of catalytic activity at 41 °C. In all panels, data are presented as the mean of three technical replicates +/- standard deviation.

Extended Data Fig. 9 Properties of the Aurora dye collection.

(a) Comparison of the chemical diversity of Aurora versus a representative diversity set of drug-like molecules that is used for high throughout drug screens. (b) cLogP of dyes and relationship to paDSF performance. (A) Comparison of cLogP, a calculated predictor of water solubility, to the paDSF performance of that dye. Filled circles: percent of tested proteins for which a dye was called a ‘hit’. Xs: percent of tested proteins for which a dye was called ‘sensitive’. No significant correlation was found between hit rate and cLogP (Pearson p value > 0.05, see panel for exact values). Statistical significance was determined with a two-way t-test. (c) Distribution of cLogP values for the full Aurora library (312 dyes), for reference. (d) Individual points represent a single dye, and dyes are arranged along the axis in order of increasing hit rate. Data are shown for the full Aurora library (312 dyes). Colored points indicate dyes which contain a given substructure, grey points indicate dyes lacking that substructure. A singe substructure is presented in each panel, and panels are shown only for sub-structures present in at least one dye. All points left of the grey vertical line, in the area labeled ‘Never hit’ were never a hit with any tested protein. Some dyes contain multiple substructures.

Extended Data Fig. 10 Additional analyses of the Aurora dyes.

(a) Distribution of dyes among the hit and sensitive dyes assigned for each protein. Individual histograms displaying the number of hit dyes for a given protein, binned by the global hit rates of the specific hit dyes. Total hit rates are divided into ten evenly spaced bins, containing hit rates of up to 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100% of all tested proteins. Green bars: for ‘hit’ assignment. Grey bars: for ‘sensitive’ assignment. The presence of both selective and promiscuous dye hits in an individual dye screen appears as the presence of hits in both lower hit rate (for example < 20%) and higher hit rate (for example >40%) bins. Results for individual proteins are presented in individual panels, with the protein name indicated in the panel title. (b) Weak positive correlation between protein sequence similarity and dye fingerprint. Comparison of the relationship between primary sequence similarity and dye screen outcomes shows a weak positive Pearson’s correlation: 0.19, p-value 3.9e-20. Statistical significance was determined by one-way ANOVA with Tukey’s post-hoc analysis. Data shown includes 2,211 unique pairings between 67 proteins (72 proteins tested total - 5 proteins tested only as hetero-complexes; see Supplementary Methods). To provide context, the number of dyes screened in common between two proteins is indicated in point size and color. Smaller, darker points: fewer dyes screened in common. Larger, lighter points: more dyes screened in common. See figure key for exact mappings. Calculations of the Jaccard Index (y axis values) and pairwise sequence similarity (x axis values) are described in Supplementary Methods.

Supplementary information

Supplementary Information

Table of contents, Supplementary Notes 1–4 and legends for additional supplementary files.

Reporting Summary

Supplementary Table 1

A table of the key dye properties, such as hit percentage, fluorescence wavelength, catalog information and cLogP.

Supplementary Table 2

A table of proteins, with detailed information about the screening conditions (for example, buffer and concentration) for each.

Supplementary Data 1

Complete list of calculated properties of the proteins, such as hydrophobicity, secondary structure, molecular weight and sequence.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wu, T., Yu, J.C., Suresh, A. et al. Protein-adaptive differential scanning fluorimetry using conformationally responsive dyes. Nat Biotechnol (2024). https://doi.org/10.1038/s41587-024-02158-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41587-024-02158-7

Search

Quick links

Nature Briefing: Translational Research

Sign up for the Nature Briefing: Translational Research newsletter — top stories in biotechnology, drug discovery and pharma.

Get what matters in translational research, free to your inbox weekly. Sign up for Nature Briefing: Translational Research