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.

  • Matters Arising
  • Published:

Identification of phosphosites that alter protein thermal stability

Matters Arising to this article was published on 17 June 2021

The Original Article was published on 05 August 2019

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: Analysis of phosphosite effects on protein stability.
Fig. 2: Examples of phosphosites that alter protein thermal stability.

Data availability

The MS proteomic data generated for this study were deposited to the ProteomeXchange Consortium via the PRIDE partner repository with the dataset identifier PXD016750.

Code availability

All code to reproduce analysis and data figures is available at https://gitlab.com/public_villenlab/dali_phospho_thermalstability.

References

  1. Hornbeck, P. V. et al. PhosphoSitePlus, 2014: mutations, PTMs and recalibrations. Nucleic Acids Res. 43, D512–D520 (2015).

    Article  CAS  Google Scholar 

  2. Huang, J. X. et al. High throughput discovery of functional protein modifications by Hotspot Thermal Profiling. Nat. Methods 16, 894–901 (2019).

    Article  CAS  Google Scholar 

  3. Potel, C. M. et al. Impact of phosphorylation on thermal stability of proteins. Nat. Methods https://doi.org/10.1038/s41592-021-01177-5 (2021).

  4. Gaetani, M. et al. Proteome Integral Solubility Alteration: a high-throughput proteomics assay for target deconvolution. J. Proteome Res. 18, 4027–4037 (2019).

    Article  CAS  Google Scholar 

  5. Savitski, M. M. et al. Tracking cancer drugs in living cells by thermal profiling of the proteome. Science 346, 1255784 (2014).

    Article  Google Scholar 

  6. Finley, D., Bartel, B. & Varshavsky, A. The tails of ubiquitin precursors are ribosomal proteins whose fusion to ubiquitin facilitates ribosome biogenesis. Nature 338, 394–401 (1989).

    Article  CAS  Google Scholar 

  7. Kane, P. M. et al. Protein splicing converts the yeast TFP1 gene product to the 69-kD subunit of the vacuolar H+-adenosine triphosphatase. Science 250, 651–657 (1990).

    Article  CAS  Google Scholar 

  8. Holt, L. J. et al. Global analysis of Cdk1 substrate phosphorylation sites provides insights into evolution. Science 325, 1682–1686 (2009).

    Article  CAS  Google Scholar 

  9. Dephoure, N. et al. A quantitative atlas of mitotic phosphorylation. Proc. Natl Acad. Sci. USA 105, 10762–10767 (2008).

    Article  CAS  Google Scholar 

  10. Imami, K. et al. Phosphorylation of the ribosomal protein RPL12/uL11 affects translation during mitosis. Mol. Cell 72, 84–98 (2018).

    Article  CAS  Google Scholar 

  11. Viéitez, C. et al. Towards a systematic map of the functional role of protein phosphorylation. Preprint at bioRxiv https://doi.org/10.1101/872770 (2019).

  12. Leutert, M., Rodriguez-Mias, R. A., Fukuda, N. K. & Villén, J. R2–P2 rapid-robotic phosphoproteomics enables multidimensional cell signaling studies. Mol. Syst. Biol. 15, e9021 (2019).

    Article  CAS  Google Scholar 

  13. Cox, J. & Mann, M. MaxQuant enables high peptide identification rates, individualized p.p.b.-range mass accuracies and proteome-wide protein quantification. Nat. Biotechnol. 26, 1367–1372 (2008).

    Article  CAS  Google Scholar 

  14. Benjamini, Y. & Hochberg, Y. Controlling the false discovery rate: a practical and powerful approach to multiple testing. J. R. Stat. Soc. Ser. B (Methodol.) 57, 289–300 (1995).

    Google Scholar 

  15. Pu, S., Wong, J., Turner, B., Cho, E. & Wodak, S. J. Up-to-date catalogues of yeast protein complexes. Nucleic Acids Res. 37, 825–831 (2009).

    Article  CAS  Google Scholar 

  16. The PyMOL Molecular Graphics System v. 1.2r3pre (Schrödinger, LLC).

  17. Groll, M. et al. Structure of 20S proteasome from yeast at 2.4 Å resolution. Nature 386, 463–471 (1997).

    Article  CAS  Google Scholar 

  18. Bulfer, S. L., Brunzelle, J. S. & Trievel, R. C. Crystal structure of Saccharomyces cerevisiae Aro8, a putative α-aminoadipate aminotransferase. Protein Sci. 22, 1417–1424 (2013).

    Article  CAS  Google Scholar 

  19. Jogl, G., Rozovsky, S., McDermott, A. E. & Tong, L. Optimal alignment for enzymatic proton transfer: structure of the Michaelis complex of triosephosphate isomerase at 1.2-Å resolution. Proc. Natl Acad. Sci. USA 100, 50–55 (2003).

    Article  Google Scholar 

  20. Didierjean, C. et al. Crystal structure of two ternary complexes of phosphorylating glyceraldehyde-3-phosphate dehydrogenase from Bacillus stearothermophilus with NAD and d-glyceraldehyde 3-phosphate. J. Biol. Chem. 278, 12968–12976 (2003).

    Article  CAS  Google Scholar 

  21. Kim, H., Feil, I. K., Verlinde, C. L. M. J., Petra, P. H. & Hol, W. G. J. Crystal structure of glycosomal glyceraldehyde-3-phosphate dehydrogenase from Leishmania mexicana: implications for structure-based drug design and a new position for the inorganic phosphate binding site. Biochemistry 34, 14975–14986 (1995).

    Article  CAS  Google Scholar 

  22. Wagih, O. et al. A resource of variant effect predictions of single nucleotide variants in model organisms. Mol. Syst. Biol. 14, e8430 (2018).

    Article  Google Scholar 

  23. Eng, J. K., Jahan, T. A. & Hoopmann, M. R. Comet: an open-source MS/MS sequence database search tool. Proteomics 13, 22–24 (2013).

    Article  CAS  Google Scholar 

  24. Käll, L., Canterbury, J. D., Weston, J., Noble, W. S. & MacCoss, M. J. Semi-supervised learning for peptide identification from shotgun proteomics datasets. Nat. Methods 4, 923–925 (2007).

    Article  Google Scholar 

  25. Beausoleil, S. A., Villén, J., Gerber, S. A., Rush, J. & Gygi, S. P. A probability-based approach for high-throughput protein phosphorylation analysis and site localization. Nat. Biotechnol. 24, 1285–1292 (2006).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank members of the Villén laboratory for scientific discussions, in particular, B. Ruiz, M. Leutert and A. Hogrebe. We thank A. Llovet Soto and J. Eng for software developments on the data analysis pipeline. I.R.S. and K.N.H. were supported by NIH training grant T32HG000035. A.S.V. was supported by NIH training grant T32LM012419. Most of this work was supported by NIH grant R35GM119536 to J.V. The Villén laboratory is additionally supported by NIH grants R01AG056359, R01NS098329 and RM1HG010461; Human Frontiers Science Program grant RGP0034/2018; a research program grant from the WM Keck Foundation; and the University of Washington Proteome Resource UWPR95794.

Author information

Authors and Affiliations

Authors

Contributions

I.R.S., K.N.H., R.A.R.-M. and J.V. conceived the study and designed experiments. I.R.S. conducted experiments with advice from K.N.H., R.A.R.-M. and J.V. and assistance from A.A.B. I.R.S. analyzed data with advice from R.A.R.-M. and A.S.V. J.V. supervised the study. I.R.S. and J.V. wrote the paper, and all authors edited it.

Corresponding author

Correspondence to Judit Villén.

Ethics declarations

Competing interests

The authors declare no competing interests.

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 Reproducibility and robustness of Dali compared to HTP.

a, Scatter plot and Pearson correlation between the mean Tm for unmodified peptides observed in the phosphopeptide enriched samples (n = 10) and the mean Tm for their corresponding proteins (n = 11). Results from the Huang et al. data reanalysis conducted by us. b, Scatter plot and Pearson correlation as in a with Rs values obtained from the Dali method (n = 6).

Extended Data Fig. 2 Phosphosites that significantly alter protein thermal stability using two different statistical settings.

Volcano plots showing ΔTm for mean phosphopeptide isoform to mean protein counterpart in the x axis, and the two-sided Student’s t-test probability in the y axis. a, Huang et al. implementation shows a P value because multiple hypothesis correction was not applied. Significant phosphopeptide isoforms (blue) are defined by P value < 0.05. b, Our proposed analysis consolidates data from MS reanalysis prior to statistical testing using a two-sided Welch’s t-test, which is performed assuming unequal variances between phosphopeptide isoform and proteins. Benjamini-Hochberg adjustment was used to correct P values for multiple hypothesis testing. Significant phosphopeptide isoforms (blue) are defined by q value < 0.05.

Extended Data Fig. 3 Examples of significant hits on proteins that undergo posttranslational splicing or cleavage.

a, Rs values for observed VMA1 unmodified peptides identified in phosphopeptide-enriched samples and proteome samples displayed across the length of VMA1. Spliced products from amino acid 2–283 and 738–1031 are joined to generate the V-type proton ATPase catalytic subunit A proteoform, extinguishing the 284–737 segment. Peptides derived from the proteome samples are colored in gray and significant unmodified peptides found in the phosphopeptide-enriched sample are in red. b, Similar plot to a for RPS31, which is cleaved to generate ubiquitin (1–76 amino acid segment) and 40 S ribosomal protein S31 (77–152 amino acid segment) proteins.

Extended Data Fig. 4 Examples of phosphosites that alter protein thermal stability and are located at protein interfaces.

Rs boxplots for a, ARO8 S59, b, TPI1 S79, and c, GAPDH S201 phosphopeptide isoforms and their protein counterparts. All boxplots show results from n = 6 biological replicates, and the line represents the median, the box designates the interquartile range (IQR), and the whiskers define 1.5*IQR from the box ends. ARO8 S59, TPI1 S79, and GAPDH S201 reside at dimerization interfaces as shown in the structures to the right (PDB accession: 4JE5, 1NEY, and 3PYM, respectively). Phosphomimetic mutations ARO8 S59E and TPI1 S79E are predicted to disrupt protein interfaces (ΔΔGpred = 3.78 and ΔΔGpred =8.04 respectively). Additionally, TPI1 S79E mutation is predicted to alter protein conformational stability (ΔΔGpred = 2.39). ΔΔGpred > 2 is predicted to be destabilizing.

Extended Data Fig. 5 Examples of phosphosites that alter protein thermal stability on glycolytic enzymes.

a, Rs values for PGK1 and all measured PGK1 phosphopeptide isoforms, with significantly destabilizing phosphosite S331 shown in red. ΔΔGpred for all glutamic acid phosphomimetic substitutions were obtained from mutfunc, with ΔΔGpred > 2 considered likely destabilizing. b, GAPDH S149 phosphopeptide is shared across all GAPDH paralogs (TDH1, TDH2, and TDH3). Boxplot shows Rs values and distributions for peptides unique to one isoform (TDH1, TDH2, TDH3), peptides shared among all GAPDH isoforms (all), all peptides for TDH3, and the S149 phosphopeptide isoform. Bottom panel shows localization of S149 on the GAPDH structure near the binding site of the enzyme substrate. Boxplots show results from 6 biological replicates, the line represents the median, the box designates the interquartile range (IQR), and the whiskers define 1.5*IQR from the box ends.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Smith, I.R., Hess, K.N., Bakhtina, A.A. et al. Identification of phosphosites that alter protein thermal stability. Nat Methods 18, 760–762 (2021). https://doi.org/10.1038/s41592-021-01178-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41592-021-01178-4

This article is cited by

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