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:

Personalized phosphoproteomics identifies functional signaling

Abstract

Protein phosphorylation dynamically integrates environmental and cellular information to control biological processes. Identifying functional phosphorylation amongst the thousands of phosphosites regulated by a perturbation at a global scale is a major challenge. Here we introduce ‘personalized phosphoproteomics’, a combination of experimental and computational analyses to link signaling with biological function by utilizing human phenotypic variance. We measure individual subject phosphoproteome responses to interventions with corresponding phenotypes measured in parallel. Applying this approach to investigate how exercise potentiates insulin signaling in human skeletal muscle, we identify both known and previously unidentified phosphosites on proteins involved in glucose metabolism. This includes a cooperative relationship between mTOR and AMPK whereby the former directly phosphorylates the latter on S377, for which we find a role in metabolic regulation. These results establish personalized phosphoproteomics as a general approach for investigating the signal transduction underlying complex biology.

This is a preview of subscription content, access via your institution

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Phosphoproteomes of extensive physiological perturbations retain unique subject-specific signatures.
Fig. 2: Individualized analysis of human phosphoproteomes defines invariable and variable phosphosites across humans.
Fig. 3: Insulin signaling in human skeletal muscle is rewired by exercise.
Fig. 4: Personalized phosphoproteomics prioritizes functional, phenotype-associated signaling.
Fig. 5: An independent validation cohort replicates personalized phosphoproteomics findings.
Fig. 6: mTORC1 phosphorylates AMPKα2 S377 to promote cellular proliferation during glucose withdrawal without inhibition of AMPK.

Similar content being viewed by others

Data availability

RAW data, MaxQuant and Spectronaut output tables have been deposited in the PRIDE proteomeXchange repository (https://www.ebi.ac.uk/pride/) with accession no. PXD027198. Processed data can be explored online at www.personalizedphospho.com.

Code availability

We have uploaded scripts and input data to https://github.com/eliseneedham/PersonalisedPhos

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  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Lundby, A. et al. Oncogenic mutations rewire signaling pathways by switching protein recruitment to phosphotyrosine sites. Cell 179, 543–560 (2019).

    Article  CAS  PubMed  Google Scholar 

  4. 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  CAS  PubMed  Google Scholar 

  5. Bodenmiller, B. et al. Phosphoproteomic analysis reveals interconnected system-wide responses to perturbations of kinases and phosphatases in yeast. Sci. Signal. 3, rs4 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Needham, E. J., Parker, B. L., Burykin, T., James, D. E. & Humphrey, S. J. Illuminating the dark phosphoproteome. Sci. Signal. 12, eaau8645 (2019).

    Article  CAS  PubMed  Google Scholar 

  7. Oliveira, A. P. et al. Regulation of yeast central metabolism by enzyme phosphorylation. Mol. Syst. Biol. 8, 623 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Gonçalves, E. et al. Systematic analysis of transcriptional and post-transcriptional regulation of metabolism in yeast. PLoS Comput. Biol. 13, e1005297 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Ochoa, D. et al. The functional landscape of the human phosphoproteome. Nat. Biotechnol. 38, 365–373 (2020).

    Article  CAS  PubMed  Google Scholar 

  10. Hoffman, N. J. et al. Global phosphoproteomic analysis of human skeletal muscle reveals a network of exercise-regulated kinases and AMPK substrates. Cell Metab. 22, 922–935 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Hingst, J. R. et al. Exercise-induced molecular mechanisms promoting glycogen supercompensation in human skeletal muscle. Mol. Metab. 16, 24–34 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  13. Humphrey, S. J., Karayel, O., James, D. E. & Mann, M. High-throughput and high-sensitivity phosphoproteomics with the EasyPhos platform. Nat. Protoc. 13, 1897–1916 (2018).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Hojlund, K. et al. Human ATP synthase beta is phosphorylated at multiple sites and shows abnormal phosphorylation at specific sites in insulin-resistant muscle. Diabetologia 53, 541–551 (2010).

    Article  CAS  PubMed  Google Scholar 

  16. Batista, T. M. et al. A cell-autonomous signature of dysregulated protein phosphorylation underlies muscle insulin resistance in type 2 diabetes. Cell Metab. 32, 844–859 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Vaag, A., Henriksen, J. E. & Beck-Nielsen, H. Decreased insulin activation of glycogen synthase in skeletal muscles in young nonobese Caucasian first-degree relatives of patients with non-insulin-dependent diabetes mellitus. J. Clin. Invest. 89, 782–788 (1992).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Um, S. H. et al. Absence of S6K1 protects against age- and diet-induced obesity while enhancing insulin sensitivity. Nature 431, 200–205 (2004).

    Article  CAS  PubMed  Google Scholar 

  19. Carraro, F., Stuart, C. A., Hartl, W. H., Rosenblatt, J. & Wolfe, R. R. Effect of exercise and recovery on muscle protein synthesis in human subjects. Am. J. Physiol. 259, E470–E476 (1990).

    CAS  PubMed  Google Scholar 

  20. Krycer, J. R. et al. Dynamic metabolomics reveals that insulin primes the adipocyte for glucose metabolism. Cell Rep. 21, 3536–3547 (2017).

    Article  CAS  PubMed  Google Scholar 

  21. Phan, J. & Reue, K. Lipin, a lipodystrophy and obesity gene. Cell Metab. 1, 73–83 (2005).

    Article  CAS  PubMed  Google Scholar 

  22. Hardie, D. G., Ross, F. A. & Hawley, S. A. AMPK: a nutrient and energy sensor that maintains energy homeostasis. Nat. Rev. Mol. Cell Biol. 13, 251–262 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Kjøbsted, R. et al. Enhanced muscle insulin sensitivity after contraction/exercise is mediated by AMPK. Diabetes 66, 598–612 (2017).

    Article  CAS  PubMed  Google Scholar 

  24. Lopez-Mejia, I. C. et al. CDK4 phosphorylates AMPKalpha2 to inhibit its activity and repress fatty acid oxidation. Mol. Cell 68, 336–349.e6 (2017).

    Article  CAS  PubMed  Google Scholar 

  25. Rodrik-Outmezguine, V. S. et al. Overcoming mTOR resistance mutations with a new-generation mTOR inhibitor. Nature 534, 272–276 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Ling, N. X. Y. et al. mTORC1 directly inhibits AMPK to promote cell proliferation under nutrient stress. Nat. Metab. 2, 41–49 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Inoki, K., Zhu, T. & Guan, K.-L. TSC2 mediates cellular energy response to control cell growth and survival. Cell 115, 577–590 (2003).

    Article  CAS  PubMed  Google Scholar 

  28. Gwinn, D. M. et al. AMPK phosphorylation of raptor mediates a metabolic checkpoint. Mol. Cell 30, 214–226 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Mertins, P. et al. Proteogenomics connects somatic mutations to signalling in breast cancer. Nature 534, 55–62 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Gillette, M. A. et al. Proteogenomic characterization reveals therapeutic vulnerabilities in lung adenocarcinoma. Cell 182, 200–225 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Wojtaszewski, J. F., Hansen, B. F., Kiens, B. & Richter, E. A. Insulin signaling in human skeletal muscle: time course and effect of exercise. Diabetes 46, 1775–1781 (1997).

    Article  CAS  PubMed  Google Scholar 

  32. Ong, S.-E. et al. Stable isotope labeling by amino acids in cell culture, SILAC, as a simple and accurate approach to expression proteomics. Mol. Cell. Proteomics 1, 376–386 (2002).

    Article  CAS  PubMed  Google Scholar 

  33. Cox, J. et al. Accurate proteome-wide label-free quantification by delayed normalization and maximal peptide ratio extraction, termed MaxLFQ. Mol. Cell. Proteomics 13, 2513–2526 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. 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  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  36. Bekker-Jensen, D. B. et al. Rapid and site-specific deep phosphoproteome profiling by data-independent acquisition without the need for spectral libraries. Nat. Commun. 11, 787 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Yang, P. et al. Multi-omic profiling reveals dynamics of the phased progression of pluripotency. Cell Syst. 8, 427–445 (2019).

  38. Needham, E. J. et al. Phosphoproteomics of acute cell stressors targeting exercise signaling networks reveal drug interactions regulating protein secretion. Cell Rep. 29, 1524–1538 (2019).

    Article  CAS  PubMed  Google Scholar 

  39. Ritchie, M. E. et al. limma powers differential expression analyses for RNA-sequencing and microarray studies. Nucleic Acids Res. 43, e47 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Google Scholar 

  41. Conway, J. R., Lex, A. & Gehlenborg, N. UpSetR: an R package for the visualization of intersecting sets and their properties. Bioinformatics 33, 2938–2940 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Storey, J., Bass, A., Dabney, A., Robinson, D. & Warnes, G. qvalue: Q-value estimation for false discovery rate control. https://www.bioconductor.org/packages/devel/bioc/manuals/qvalue/man/qvalue.pdf (2021).

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

    Article  CAS  PubMed  Google Scholar 

  44. Ochoa, D. et al. An atlas of human kinase regulation. Mol. Syst. Biol. 12, 888 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Chen, J. et al. The trans-ancestral genomic architecture of glycemic traits. Nat. Genet. 53, 840–860 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Dickinson, M. E. et al. High-throughput discovery of novel developmental phenotypes. Nature 537, 508–514 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank S. Kurdyukov, A. Zadoorian, I. Beck Nielsen, B. Bolmgren and L. Hansen for technical assistance. We thank E. Pickering and members of the Metabolic Systems Biology group, Pfizer Cardiovascular and Metabolic Diseases Research Unit and the Molecular Physiology section of the University of Copenhagen for feedback and discussions. AMPKα double-knockout MEFs were kindly provided by B. Viollet. This research was facilitated by access to Sydney Mass Spectrometry, a core research facility at the University of Sydney. This work was funded by: Pfizer, Inc.; Australian Government Research Training Program Scholarship and the University of Sydney Val Street Scholarship (to E.J.N.); ARC Laureate Fellowship (no. FL200100096 to D.E.J.); NHMRC Project Grant (no. APP1122376 to B.L.P.); APP1161262 (to J.S.O.); ARC Discovery Project Grant (no. DP180101682 to J.P.); Danish Council for In-dependent Research Medical Sciences (no. FSS 8020-00288 to J.F.P.W.); Ministry of Culture Denmark (no. FPK 2016-0027 to J.F.P.W.); The Novo Nordisk Foundation (no. NNF16OC 0023046 to J.F.P.W.); The University of Sydney University of Copenhagen Partnership Collaboration Awards (to J.F.P.W. and D.E.J.); the Victorian Government’s Operational Infrastructure Support Program (to J.S.O.); a research grant from the Danish Diabetes Academy, which is funded by the Novo Nordisk Foundation, grant number NNF17SA0031406 (J.O.).

Author information

Authors and Affiliations

Authors

Contributions

E.J.N., S.J.H., D.E.J., J.F.P.W. and C.P. conceptualized the study. J.R.H., S.J.H., E.J.N., B.L.P., G.Y., N.X.Y.L., K.R.M., J.O., J.M.K., K.H., E.A.R., B.K. and J.F.P.W. conducted investigations. E.J.N., S.J.H., J.R.H., J.F.P.W., B.L.P., E.A.R., B.K. and J.P. were responsible for methodology. E.J.N. performed formal analysis. E.J.N. and S.J.H. were responsible for visualization. Software was the responsibility of E.J.N. and S.J.H. N.X.Y.L., J.S.O. and D.E.J. oversaw resources. Project administration was performed by C.P., D.E.J., J.F.P.W. and S.J.H. Funding was acquired by D.E.J., J.F.P.W. and C.P. S.J.H., D.E.J., J.F.P.W., C.P. and J.P. supervised the project. E.J.N., S.J.H. and D.E.J. wrote the original draft. Writing, review and editing were undertaken by all authors.

Corresponding authors

Correspondence to David E. James, Jørgen F. P. Wojtaszewski or Sean J. Humphrey.

Ethics declarations

Competing interests

C.P. was an employee of Pfizer during the study. The other authors declare no competing interests.

Additional information

Peer review information Nature Biotechnology thanks the anonymous reviewers for their contribution to the peer review of this work.

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

Extended data

Extended Data Fig. 1 Quality control and subject-specific human phosphoproteome variation.

a, Correlation matrix of the technical replicates. b, Correlation matrix of the biological replicates. c, Quantified and regulated phosphopeptides, phosphosites and phosphoproteins. d-e, PCA of median normalised phosphosites quantified in all ground state samples. Points are numbered by subject and coloured by exercised (blue) and non-exercised (grey) legs. f-g, PCA of the subset of the phosphosites from d-e, with matched protein data quantified in all samples. h-i, PCA of the sites in f-g, normalized by their respective total protein levels by subtracting the median normalised protein log2 intensities from the median normalised phosphosite log2 intensities. Abundance of phosphorylated j, 14-3-3γ Y133, k, ROCK2 T1374 and l, ATP Synthase β S415 compared to their total protein abundances at ground state. m, Glucose uptake of the non-exercised (grey) and exercised (blue) legs during a hyperinsulinemic-euglycemic clamp expressed as area under the curve (two-sided paired t-test).

Extended Data Fig. 2 Exercise-and insulin regulated phosphosites, and comparison with phosphoproteomes measured immediately after acute exercise.

a, Volcano plots of the phosphosites regulated by exercise and insulin (two-way repeated measures ANOVA with planned contrasts). b, Upset plot, quantifying the regulated phosphosites (BH padj < 0.05 and fold change > 1.5) shared in the different conditions. c, Quantified phosphosites in this study compared to Hoffman et al. 2015. d, Regulated phosphosites by exercise recovery in this study, not considering the insulin-stimulated states, compared to Hoffman et al. 2015. For this study, regulated is padj < 0.05 and fold change > 1.5. For Hoffman et al., regulated is considered as padj < 0.05. e, Enrichment of kinase substrates in the ground state exercise-regulated phosphosites in this study compared to Hoffman et al. regulated phosphosites.

Extended Data Fig. 3 Patterns of signalling in response to exercise and insulin.

a, Hypothetical profiles that fit the groupings in Fig. 3 and Supplementary Table 4. b, Quantification of phosphosites in the groupings, with the proportion of phosphosites with an interaction between exercise and insulin shaded. Sites were considered to have an interaction if the interaction term padj < 0.05 and fold change >1.5.

Extended Data Fig. 4 Personalised phosphoproteomics extracts glucose uptake-associated phosphosites.

a, Ranks of phosphosites that passed filtering for glucose uptake correlations ordered by glucose uptake correlation significance, ANOVA significance and magnitude fold change. Kendall’s rank correlation (τ) was calculated on the -log10p and absolute log2FC values. b, Gene Ontology enrichment of the glucose uptake-associated phosphosites, (Fisher’s exact test), filtered for pathways with BH-padj < 0.05. Pearson’s correlation and BH padj of phosphorylated c, GYS1 S649 and d, VAMP2 S61 with glucose uptake, with prior exercise (blue outline) and insulin (orange centre). e, Overrepresentation of kinase substrates in positive glucose uptake-associated sites (Fisher’s exact test, BH-padj). f, The distribution of kinase substrates by correlation coefficient with glucose uptake (asterisk indicates empirical p < 0.05 from a Kolmogorov-Smirnov test ordered by correlation p-value). g, Quantification of Akt substrates in the classes defined in Fig. 3. Profiles of Akt regulatory sites h, T308 i, S473 and j, substrate GSK3β S9 k, Pearson’s correlation and BH padj of phosphorylated LPIN1 S889 with glucose uptake. l, LPIN1 sequence aligned to mTORC1 motif. m, Pearson’s correlation and BH padj of AGPAT1 protein levels with glucose uptake AUC, in ground state exercised (blue) or not exercised (grey) legs. Correlations and Kolmogorov-Smirnov tests were two-sided. Fisher’s exact tests were one-sided.

Extended Data Fig. 5 An independent cohort replicates glucose uptake associations.

a, Correlation coefficients for the common sites that passed filtering to test for correlation to glucose uptake in both studies (significant regulation and quantification in ≥ 50% of the samples, discovery cohort: Pearson’s correlation, validation cohort: Kendall’s rank correlation) are plotted. Green points are reported mTOR/p70S6K substrates. b, Phosphosites that passed filtering to correlate with glucose uptake (regulated, quantified ≥ 50% samples) were ordered by -log10p of their correlation to glucose uptake, their ANOVA significance, or their magnitude fold change. Enrichment was calculated with Kolmogorov-Smirnov test with empirical p-values. c, Enrichment of Gene Ontology biological processes in the glucose uptake-associated phosphosites, calculated by Fisher’s exact test, filtered for pathways with BH padj < 0.05. d, Enrichment of kinase substrates amongst phosphosites positively correlated with glucose uptake (padj < 0.05, r > 0, Fisher’s exact test). e, Glucose uptake Kendall’s rank correlation coefficients and BH padj of phosphosites grouped by kinase. Asterisks indicate Kolmogorov-Smirnov test empirical p-value < 0.05. Correlations and Kolmogorov-Smirnov tests were two-sided. Fisher’s exact tests were one-sided.

Extended Data Fig. 6 mTORC1 phosphorylates AMPKα2 S377.

a, Repeated measures correlation profile of AMPK S377 (two-sided, BH-adjusted p-value). b, Quantification of phosphosites on AMPK from in vitro kinase assays with mTORC1 and different combinations of AMPK subunits. The γ subunit was kept as AMPKγ1. For each condition, n = 1. c, AMPK inhibitory site profiles in human skeletal muscle. d, Sequence alignment of AMPKα2 S377 across different species. S. cerevisiae SNF1 was used in the alignment. e, AMPKα2β2γ1 structure from PDB 6b2e. AMPKα2 residues 377–398 were not modelled so are included as a dashed line. AMPKα2 S377 is approximated as immediately adjacent to residue 376 and indicated in green. f, Real-time proliferation analysis of AMPKα1/2 KO MEF cells re-expressing WT or S377A AMPKα2 in media with 25 mM glucose. g, AMPK substrate phosphosite profiles in human skeletal muscle. h, mTORC1 substrate phosphosite profiles in human skeletal muscle. Data are presented as the mean +/− SEM represented by shading.

Supplementary information

Supplementary Information

Supplementary Note 1.

Reporting Summary

Supplementary Tables

Table 1: The post-exercise and insulin-regulated human skeletal muscle phosphoproteome, and associations with leg glucose uptake. Table 2: Inter- and intrasubject phosphoproteome CVs. Table 3: Comparison of 4-h post-exercise phosphoproteomes with those measured immediately after acute exercise. Table 4: Insulin-regulated, potentiated, emergent and divergent phosphosites with exercise. Table 5: Discovery and validation cohort subject characteristics. Table 6: Glucose uptake AUC and proteome correlations. Table 7: Validation cohort glucose uptake associations with discovery cohort glucose-uptake-associated phosphosites. Table 8: Glucose uptake associations of the combined discovery and validation cohorts. Table 9: Detection of AMPK phosphosites from mTORC1 in vitro kinase assays.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Needham, E.J., Hingst, J.R., Parker, B.L. et al. Personalized phosphoproteomics identifies functional signaling. Nat Biotechnol 40, 576–584 (2022). https://doi.org/10.1038/s41587-021-01099-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41587-021-01099-9

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