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:

High-throughput and high-sensitivity phosphoproteomics with the EasyPhos platform

Abstract

Mass spectrometry has transformed the field of cell signaling by enabling global studies of dynamic protein phosphorylation (‘phosphoproteomics’). Recent developments are enabling increasingly sophisticated phosphoproteomics studies, but practical challenges remain. The EasyPhos workflow addresses these and is sufficiently streamlined to enable the analysis of hundreds of phosphoproteomes at a depth of >10,000 quantified phosphorylation sites. Here we present a detailed and updated workflow that further ensures high performance in sample-limited conditions while also reducing sample preparation time. By eliminating protein precipitation steps and performing the entire protocol, including digestion, in a single 96-well plate, we now greatly minimize opportunities for sample loss and variability. This results in very high reproducibility and a small sample size requirement (≤200 μg of protein starting material). After cell culture or tissue collection, the protocol takes 1 d, whereas mass spectrometry measurements require ~1 h per sample. Applied to glioblastoma cells acutely treated with epidermal growth factor (EGF), EasyPhos quantified 20,132 distinct phosphopeptides from 200 μg of protein in less than 1 d of measurement time, revealing thousands of EGF-regulated phosphorylation events.

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: Schematic of the EasyPhos platform for high-throughput phosphoproteomics.
Fig. 2: Performance evaluation of the EasyPhos method using limited starting material.
Fig. 3: Evaluation of the performance of the updated EasyPhos workflow with respect to starting material, MS instruments, and LC gradient duration.
Fig. 4: Demonstration of the phosphoproteomics workflow.

Similar content being viewed by others

References

  1. Humphrey, S. J., James, D. E. & Mann, M. Protein phosphorylation: a major switch mechanism for metabolic regulation. Trends Endocrinol. Metab. 26, 676–687 (2015).

    Article  PubMed  CAS  Google Scholar 

  2. Sharma, K. et al. Ultradeep human phosphoproteome reveals a distinct regulatory nature of Tyr and Ser/Thr-based signaling. Cell Rep. 8, 1583–1594 (2014).

    Article  PubMed  CAS  Google Scholar 

  3. Ficarro, S. B. et al. Phosphoproteome analysis by mass spectrometry and its application to Saccharomyces cerevisiae. Nat. Biotechnol. 20, 301–305 (2002).

    Article  PubMed  CAS  Google Scholar 

  4. Olsen, J. V. et al. Global, in vivo, and site-specific phosphorylation dynamics in signaling networks. Cell 127, 635–648 (2006).

    Article  PubMed  CAS  Google Scholar 

  5. Lemeer, S. & Heck, A. J. The phosphoproteomics data explosion. Curr. Opin. Chem. Biol. 13, 414–420 (2009).

    Article  PubMed  CAS  Google Scholar 

  6. Riley, N. M. & Coon, J. J. Phosphoproteomics in the age of rapid and deep proteome profiling. Anal. Chem. 88, 74–94 (2016).

    Article  PubMed  CAS  Google Scholar 

  7. Rigbolt, K. T. G. & Blagoev, B. Quantitative phosphoproteomics to characterize signaling networks. Semin. Cell Dev. Biol. 23, 863–871 (2012).

    Article  PubMed  CAS  Google Scholar 

  8. Olsen, J. V. & Mann, M. Status of large-scale analysis of post-translational modifications by mass spectrometry. Mol. Cell. Proteomics 12, 3444–3452 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  9. Jünger, M. A. & Aebersold, R. Mass spectrometry-driven phosphoproteomics: patterning the systems biology mosaic. Wiley Interdiscip. Rev. Dev. Biol. 3, 83–112 (2014).

    Article  PubMed  CAS  Google Scholar 

  10. Humphrey, S. J., Azimifar, S. B. & Mann, M. High-throughput phosphoproteomics reveals in vivo insulin signaling dynamics. Nat. Biotechnol. 33, 990–995 (2015).

    Article  PubMed  CAS  Google Scholar 

  11. Robles, M. S., Humphrey, S. J. & Mann, M. Phosphorylation is a central mechanism for circadian control of metabolism and physiology. Cell Metab. 25, 118–127 (2017).

    Article  PubMed  CAS  Google Scholar 

  12. Liu, J. J. et al. In vivo brain GPCR signaling elucidated by phosphoproteomics. Science 360, eaao4927 (2018).

  13. Sacco, F. et al. Glucose-regulated and drug-perturbed phosphoproteome reveals molecular mechanisms controlling insulin secretion. Nat. Commun. 7, 13250 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  14. Steger, M. et al. Phosphoproteomics reveals that Parkinson’s disease kinase LRRK2 regulates a subset of Rab GTPases. eLife 5, e12813 (2016).

  15. Courcelles, M. et al. Phosphoproteome dynamics reveal novel ERK1/2 MAP kinase substrates with broad spectrum of functions. Mol. Syst. Biol. 9, 129–57 (2013).

    Google Scholar 

  16. Kanshin, E., Bergeron-Sandoval, L. P., Isik, S. S., Thibault, P. & Michnick, S. W. A cell-signaling network temporally resolves specific versus promiscuous phosphorylation. Cell Rep. 10, 1202–1214 (2015).

    Article  PubMed  CAS  Google Scholar 

  17. Posewitz, M. C. & Tempst, P. Immobilized gallium(III) affinity chromatography of phosphopeptides. Anal. Chem. 71, 2883–2892 (1999).

    Article  PubMed  CAS  Google Scholar 

  18. Andersson, L. & Porath, J. Isolation of phosphoproteins by immobilized metal (Fe3+) affinity chromatography. Anal. Biochem. 154, 250–254 (1986).

  19. Villén, J. & Gygi, S. P. The SCX/IMAC enrichment approach for global phosphorylation analysis by mass spectrometry. Nat. Protoc. 3, 1630–1638 (2008).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Zhou, H. et al. Robust phosphoproteome enrichment using monodisperse microsphere–based immobilized titanium (IV) ion affinity chromatography. Nat. Protoc. 8, 461–480 (2013).

    Article  PubMed  CAS  Google Scholar 

  21. Larsen, M. R., Thingholm, T. E., Jensen, O. N., Roepstorff, P. & Jørgensen, T. J. D. Highly selective enrichment of phosphorylated peptides from peptide mixtures using titanium dioxide microcolumns. Mol. Cell. Proteomics 4, 873–886 (2005).

    Article  PubMed  CAS  Google Scholar 

  22. Liang, S.-S., Makamba, H., Huang, S.-Y. & Chen, S.-H. Nano-titanium dioxide composites for the enrichment of phosphopeptides. J. Chromatogr. A 1116, 38–45 (2006).

    Article  PubMed  CAS  Google Scholar 

  23. Sugiyama, N. et al. Phosphopeptide enrichment by aliphatic hydroxy acid-modified metal oxide chromatography for nano-LC-MS/MS in proteomics applications. Mol. Cell. Proteomics 6, 1103–1109 (2007).

    Article  PubMed  CAS  Google Scholar 

  24. Thingholm, T. E., Jørgensen, T. J. D., Jensen, O. N. & Larsen, M. R. Highly selective enrichment of phosphorylated peptides using titanium dioxide. Nat. Protoc. 1, 1929–1935 (2006).

    Article  PubMed  CAS  Google Scholar 

  25. Macek, B., Mann, M. & Olsen, J. V. Global and site-specific quantitative phosphoproteomics: principles and applications. Annu. Rev. Pharmacol. Toxicol. 49, 199–221 (2009).

    Article  PubMed  CAS  Google Scholar 

  26. Pinkse, M. W. H. H. et al. Highly robust, automated, and sensitive online TiO2-based phosphoproteomics applied to study endogenous phosphorylation in Drosophila melanogaster. J. Proteome Res. 7, 687–697 (2008).

  27. Humphrey, S. J. et al. Dynamic adipocyte phosphoproteome reveals that Akt directly regulates mTORC2. Cell Metab. 17, 1009–1020 (2013).

  28. Wang, Y. et al. Reversed-phase chromatography with multiple fraction concatenation strategy for proteome profiling of human MCF10A cells. Proteomics 11, 2019–2026 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Batth, T. S., Francavilla, C. & Olsen, J. V. Off-line high-pH reversed-phase fractionation for in-depth phosphoproteomics. J. Proteome Res. 13, 6176–6186 (2014).

    Article  PubMed  CAS  Google Scholar 

  30. Batth, T. S. et al. Large-scale phosphoproteomics reveals Shp-2 phosphatase-dependent regulators of Pdgf receptor signaling. Cell Rep. 22, 2601–2614 (2018).

    Article  CAS  Google Scholar 

  31. de Graaf, E. L., Giansanti, P., Altelaar, A. F. M. & Heck, A. J. R. Single-step enrichment by Ti4+-IMAC and label-free quantitation enables in-depth monitoring of phosphorylation dynamics with high reproducibility and temporal resolution. Mol. Cell. Proteomics 13, 2426–2434 (2014).

  32. Masuda, T., Sugiyama, N., Tomita, M. & Ishihama, Y. Microscale phosphoproteome analysis of 10,000 cells from human cancer cell lines. Anal. Chem. 83, 7698–7703 (2011).

  33. Ren, D. et al. An improved trypsin digestion method minimizes digestion-induced modifications on proteins. Anal. Biochem. 392, 12–21 (2009).

    Article  PubMed  CAS  Google Scholar 

  34. Masuda, T., Tomita, M. & Ishihama, Y. Phase transfer surfactant-aided trypsin digestion for membrane proteome analysis. J. Proteome Res. 7, 731–740 (2008).

    Article  PubMed  CAS  Google Scholar 

  35. Kulak, N. A., Pichler, G., Paron, I., Nagaraj, N. & Mann, M. Minimal, encapsulated proteomic-sample processing applied to copy-number estimation in eukaryotic cells. Nat. Methods 11, 319–324 (2014).

    Article  PubMed  CAS  Google Scholar 

  36. Kokubu, M., Ishihama, Y., Sato, T., Nagasu, T. & Oda, Y. Specificity of immobilized metal affinity-based IMAC/C18 tip enrichment of phosphopeptides for protein phosphorylation analysis. Anal. Chem. 77, 5144–5154 (2005).

    Article  PubMed  CAS  Google Scholar 

  37. Scheltema, R. A. et al. The Q Exactive HF, a benchtop mass spectrometer with a pre-filter, high performance quadrupole and an ultra-high field Orbitrap analyzer. Mol. Cell. Proteomics 13, 3698–3708 (2014).

  38. Kelstrup, C. D. et al. Rapid and deep proteomes by faster sequencing on a benchtop quadrupole ultra-high-field Orbitrap mass spectrometer. J. Proteome Res. 13, 6187–6195 (2014).

  39. Shaffer, S. A., Prior, D. C., Anderson, G. A., Udseth, H. R. & Smith, R. D. An ion funnel interface for improved ion focusing and sensitivity using electrospray ionization mass spectrometry. Anal. Chem. 70, 4111–4119 (1998).

    Article  PubMed  CAS  Google Scholar 

  40. Kelstrup, C. D. et al. Performance evaluation of the Q Exactive HF-X for shotgun proteomics. J. Proteome Res. 17, 727–738 (2018).

    Article  PubMed  CAS  Google Scholar 

  41. Tyanova, S., Temu, T. & Cox, J. The MaxQuant computational platform for mass spectrometry-based shotgun proteomics. Nat. Protoc. 11, 2301–2319 (2016).

    Article  PubMed  CAS  Google Scholar 

  42. Rappsilber, J., Mann, M. & Ishihama, Y. Protocol for micro-purification, enrichment, pre-fractionation and storage of peptides for proteomics using StageTips. Nat. Protoc. 2, 1896–1906 (2007).

    Article  PubMed  CAS  Google Scholar 

  43. Thingholm, T. E., Larsen, M. R., Ingrell, C. R., Kassem, M. & Jensen, O. N. TiO2-based phosphoproteomic analysis of the plasma membrane and the effects of phosphatase inhibitor treatment. J. Proteome Res. 7, 3304–3313 (2008).

  44. Pan, C., Gnad, F., Olsen, J. V. & Mann, M. Quantitative phosphoproteome analysis of a mouse liver cell line reveals specificity of phosphatase inhibitors. Proteomics 8, 4534–4546 (2008).

    Article  PubMed  CAS  Google Scholar 

  45. Svensson, M. et al. Heat stabilization of the tissue proteome: a new technology for improved proteomics. J. Proteome Res. 8, 974–981 (2009).

    Article  PubMed  CAS  Google Scholar 

  46. Lundby, A. et al. Quantitative maps of protein phosphorylation sites across 14 different rat organs and tissues. Nat. Commun. 3, 876 (2012).

  47. 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  PubMed  CAS  Google Scholar 

  48. Tyanova, S. et al. The Perseus computational platform for comprehensive analysis of (prote)omics data. Nat. Methods 13, 731–740 (2016).

    Article  PubMed  CAS  Google Scholar 

  49. Vizcaíno, J. A. et al. ProteomeXchange provides globally coordinated proteomics data submission and dissemination. Nat. Biotechnol. 32, 223–226 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  50. Deutsch, E. W. et al. The ProteomeXchange consortium in 2017: supporting the cultural change in proteomics public data deposition. Nucleic Acids Res. 45, D1100–D1106 (2017).

    Article  PubMed  CAS  Google Scholar 

  51. Kyte, J. & Doolittle, R. F. A simple method for displaying the hydropathic character of a protein. J. Mol. Biol. 157, 105–132 (1982).

    Article  PubMed  CAS  Google Scholar 

Download references

Acknowledgements

We thank members of the Proteomics and Signal Transduction Group; the Metabolic Systems Biology Group; and the staff of SydneyMS for support and discussions; and, in particular, I. Paron, K. Mayr, and G. Sowa for MS technical assistance; J. Cox for bioinformatic tools; and N. Nagaraj, E. Humphrey, B. Parker, and M. Larance for technical discussions. This work was funded in part by the Max Planck Society for the Advancement of Science, the Novo Nordisk Foundation (grant NNF15CC0001) (M.M.), and the National Health and Medical Research Council (NHMRC) (grants GNT1061122 and GNT1086850) (D.E.J.). Thanks to J. Cobcroft for her generous funding support (S.J.H.).

Author information

Authors and Affiliations

Authors

Contributions

S.J.H. designed the studies and developed methods. S.J.H. and O.K. performed the phosphoproteomic experiments and analyzed the data. M.M. and D.E.J. supervised the project and wrote the manuscript with S.J.H. and O.K.

Corresponding authors

Correspondence to Sean J. Humphrey or Matthias Mann.

Ethics declarations

Competing interests

M.M., O.K., and D.E.J. declare that they have no competing interests. S.J.H. is an inventor on a provisional patent (no. 2018901179) relating to parts of the described protocol.

Additional information

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

Related links

Key references using this protocol

1. Humphrey, S. J., Azimifar, S. B. & Mann, M. Nat. Biotechnol. 33, 990–995 (2015) https://doi.org/10.1038/nbt.3327

2. Robles, S., Humphrey, S. J. & Mann, M. Cell Metab. 25, 118–127 (2017) https://doi.org/10.1016/j.cmet.2016.10.004

3. Sacco, F. et al. Nat. Commun. 7, 13250 (2016) https://doi.org/10.1038/ncomms13250

4. Steger, M. et al. eLife 5, e12813 (2016) https://doi.org/10.7554/eLife.12813

Integrated supplementary information

Supplementary Figure 1 Comparison of the original EasyPhos workflow and the updated protocol described here.

(a) Distribution of peptide length for phosphopeptides identified by both methods, or those uniquely identified by the updated workflow. (b) Fraction of phosphopeptides containing missed cleavages in the updated and original workflows, as well as three published deep phosphoproteome studies employing different strategies (SCX fractionation27, high pH reversed-phase fractionation29, and single-run31). (c) Phosphopeptide quantification as a proportion of the total quantifiable data points, i.e. total number of phosphopeptides quantified in any n sample ÷ (total number of phosphopeptides identified × n replicate samples measured).

Supplementary Figure 2 Evaluation of the sensitivity of the updated EasyPhos workflow with different starting materials and MS instruments.

(a) Phosphopeptides (left) and Class 1 phosphorylation sites (right) quantified from a dose-titration experiment spanning 25–750 µg of total protein material (determined by BCA assay) from untreated U87 cells using the protocol described here. Protein was pooled immediately after cell lysis and divided into four workflow replicates (n = 4) for each starting material condition. Only the amount of protein for a single replicate was carried forward for each independent workflow replicate. Samples were measured on a Q Exactive HF mass spectrometer using 90-min gradients. (b) Quantification of Class 1 phosphorylation sites from the experiment depicted in Figure 3a. Phosphopeptides from a dose-titration experiment spanning 12.5–400 µg of total protein starting material (determined by BCA assay) from untreated U87 cells using the protocol described here. Protein was pooled immediately after cell lysis and divided into four workflow replicates (n = 4) for each starting material condition. Only the amount of protein for a single replicate was carried forward for each independent workflow replicate. Samples were measured on either Q Exactive HF (“Instrument: HF”) or Q Exactive HF-X (“Instrument: HF-X”) mass spectrometers as indicated, using 60-min LC gradients. Data were processed with or without the “Match between runs” algorithm in MaxQuant enabled as indicated, to transfer MS2 identifications between samples. Error bars denote mean and standard deviation of the workflow replicates.

Supplementary Figure 3 Investigation of optimal MS instrument settings for phosphopeptide quantification on the Q Exactive HF-X mass spectrometer.

In these experiments data were acquired with replicate injections of the same sample, equivalent to enrichment from 100 µg of cell lysate per injection (a-d) or as otherwise indicated (e-f). Ranges were chosen based on instrument manufacturer recommendations. The effect of Ion funnel RF level on (a) number of MS/MS scans performed during a 60-min LC gradient, and (b) the number of phosphorylation sites quantified (all) and localized accurately to a single residue (MaxQuant phosphorylation site localization probability >0.75, Class 1). Effect of Normalized Collision Energy (NCE) on (c) MS2 scan identification, and (d) the number of phosphorylation sites quantified (all) and localized accurately to a single residue (MaxQuant phosphorylation site localization probability >0.75, Class 1). Effect of MS2 resolution and MS2 maximum ion injection time on the quantification of Class 1 phosphorylation sites with either (e) low phosphopeptide load (equiv. to enrichment from 50 µg of cell lysate) or (f) higher phosphopeptide load (equiv. to enrichment from 400 µg of cell lysate). The number of phosphorylation sites quantified are reported for each combined MS2 resolution and ion injection time setting.

Supplementary Figure 4 Schematic of the 96-well deep-well plate used for sample digestion and phosphopeptide enrichment.

Cut-away view shows characteristic features of the plate design that enable rapid and efficient sample aspiration. A disposable glass pipette attached to a vacuum line can be used to aspirate supernatants without disturbing the centrifuge-pelleted beads by sliding the pipette down the square corner of each well and stopping when reaching the “aspiration stop point” depicted above. Only a small amount of supernatant should remain in the well (~ 10 µl), making wash steps highly efficient without loss of beads.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–4

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Humphrey, S.J., Karayel, O., James, D.E. et al. High-throughput and high-sensitivity phosphoproteomics with the EasyPhos platform. Nat Protoc 13, 1897–1916 (2018). https://doi.org/10.1038/s41596-018-0014-9

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-018-0014-9

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: 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