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:

Spatially resolved proteomic mapping in living cells with the engineered peroxidase APEX2

Abstract

This protocol describes a method to obtain spatially resolved proteomic maps of specific compartments within living mammalian cells. An engineered peroxidase, APEX2, is genetically targeted to a cellular region of interest. Upon the addition of hydrogen peroxide for 1 min to cells preloaded with a biotin-phenol substrate, APEX2 generates biotin-phenoxyl radicals that covalently tag proximal endogenous proteins. Cells are then lysed, and biotinylated proteins are enriched with streptavidin beads and identified by mass spectrometry. We describe the generation of an appropriate APEX2 fusion construct, proteomic sample preparation, and mass spectrometric data acquisition and analysis. A two-state stable isotope labeling by amino acids in cell culture (SILAC) protocol is used for proteomic mapping of membrane-enclosed cellular compartments from which APEX2-generated biotin-phenoxyl radicals cannot escape. For mapping of open cellular regions, we instead use a 'ratiometric' three-state SILAC protocol for high spatial specificity. Isotopic labeling of proteins takes 5–7 cell doublings. Generation of the biotinylated proteomic sample takes 1 d, acquiring the mass spectrometric data takes 2–5 d and analysis of the data to obtain the final proteomic list takes 1 week.

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: Live-cell proteomics using APEX.
Figure 2: Sample data showing characterization of APEX localization and activity leading up to a proteomic experiment.
Figure 3: Filtering the mass spectrometric data to obtain a final proteomic list.

Similar content being viewed by others

References

  1. Andersen, J.S. & Mann, M. Organellar proteomics: turning inventories into insights. EMBO Rep. 7, 874–879 (2006).

    Article  CAS  Google Scholar 

  2. ten Have, S., Boulon, S., Ahmad, Y. & Lamond, A.I. Mass spectrometry-based immuno-precipitation proteomics—the user's guide. Proteomics 11, 1153–1159 (2011).

    Article  CAS  Google Scholar 

  3. Brunner, Y., Schvartz, D., Couté, Y. & Sanchez, J.-C. Proteomics of regulated secretory organelles. Mass Spectrom. Rev. 28, 844–867 (2009).

    Article  CAS  Google Scholar 

  4. Rhee, H.-W. et al. Proteomic mapping of mitochondria in living cells via spatially restricted enzymatic tagging. Science 339, 1328–1331 (2013).

    Article  CAS  Google Scholar 

  5. Hung, V. et al. Proteomic mapping of the human mitochondrial intermembrane space in live cells via ratiometric APEX tagging. Mol. Cell 55, 332–341 (2014).

    Article  CAS  Google Scholar 

  6. 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  Google Scholar 

  7. Ross, P.L. et al. Multiplexed protein quantitation in Saccharomyces cerevisiae using amine-reactive isobaric tagging reagents. Mol. Cell. Proteomics 3, 1154–1169 (2004).

    Article  CAS  Google Scholar 

  8. Thompson, A. et al. Tandem mass tags: a novel quantification strategy for comparative analysis of complex protein mixtures by MS/MS. Anal. Chem. 75, 1895–1904 (2003).

    Article  CAS  Google Scholar 

  9. Lam, S.S. et al. Directed evolution of APEX2 for electron microscopy and proximity labeling. Nat. Methods 12, 51–54 (2015).

    Article  CAS  Google Scholar 

  10. Martell, J.D. et al. Engineered ascorbate peroxidase as a genetically encoded reporter for electron microscopy. Nat. Biotechnol. 30, 1143–1148 (2012).

    Article  CAS  Google Scholar 

  11. Wishart, J.F. & Rao, B.S.M. Recent Trends in Radiation Chemistry (World Scientific, 2010).

  12. Mortensen, A. & Skibsted, L.H. Importance of carotenoid structure in radical-scavenging reactions. J. Agric. Food Chem. 45, 2970–2977 (1997).

    Article  CAS  Google Scholar 

  13. Bhaskar, B. et al. A novel heme and peroxide-dependent tryptophan–tyrosine cross-link in a mutant of cytochrome c peroxidase. J. Mol. Biol. 328, 157–166 (2003).

    Article  CAS  Google Scholar 

  14. Rogers, M.S. et al. Cross-link formation of the cysteine 228–tyrosine 272 catalytic cofactor of galactose oxidase does not require dioxygen. Biochemistry 47, 10428–10439 (2008).

    Article  CAS  Google Scholar 

  15. Amini, F., Kodadek, T. & Brown, K.C. Protein affinity labeling mediated by genetically encoded peptide tags. Angew. Chem. Int. Ed. 41, 356–359 (2002).

    Article  CAS  Google Scholar 

  16. Colombini, M. A candidate for the permeability pathway of the outer mitochondrial membrane. Nature 279, 643–645 (1979).

    Article  CAS  Google Scholar 

  17. Chen, C.L. et al. Proteomic mapping in live Drosophila tissues using an engineered ascorbate peroxidase. Proc. Natl. Acad. Sci. USA 112, 12093–12098 (2015).

    Article  CAS  Google Scholar 

  18. Jing, J. et al. Proteomic mapping of ER-PM junctions identifies STIMATE as a regulator of Ca2+ influx. Nat. Cell Biol. 17, 1339–1347 (2015).

    Article  CAS  Google Scholar 

  19. Mick, D.U. et al. Proteomics of primary cilia by proximity labeling. Dev. Cell 35, 497–512 (2015).

    Article  CAS  Google Scholar 

  20. Polianskyte, Z. et al. LACTB is a filament-forming protein localized in mitochondria. Proc. Natl. Acad. Sci. USA 106, 18960–18965 (2009).

    Article  CAS  Google Scholar 

  21. Chapman-Smith, A. & Cronan, J.E. Molecular biology of biotin attachment to proteins. J. Nutr. 129, 477S–484S (1999).

    Article  CAS  Google Scholar 

  22. 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 

  23. Choi-Rhee, E., Schulman, H. & Cronan, J.E. Promiscuous protein biotinylation by Escherichia coli biotin protein ligase. Protein Sci. 13, 3043–3050 (2004).

    Article  CAS  Google Scholar 

  24. Roux, K.J., Kim, D.I., Raida, M. & Burke, B. A promiscuous biotin ligase fusion protein identifies proximal and interacting proteins in mammalian cells. J. Cell Biol. 196, 801–810 (2012).

    Article  CAS  Google Scholar 

  25. Morriswood, B. et al. Novel bilobe components in Trypanosoma brucei identified using proximity-dependent biotinylation. Eukaryot. Cell 12, 356–367 (2013).

    Article  CAS  Google Scholar 

  26. Kim, D.I. et al. Probing nuclear pore complex architecture with proximity-dependent biotinylation. Proc. Natl. Acad. Sci. USA 111, E2453–E2461 (2014).

    Article  CAS  Google Scholar 

  27. Firat-Karalar, E.N., Rauniyar, N., Yates, J.R. III & Stearns, T. Proximity interactions among centrosome components identify regulators of centriole duplication. Curr. Biol. 24, 664–670 (2014).

    Article  CAS  Google Scholar 

  28. DeMoss, J.A., Genuth, S.M. & Novelli, G.D. The enzymatic activation of amino acids via their acyl-adenylate derivatives. Proc. Natl. Acad. Sci. USA 42, 325–332 (1956).

    Article  CAS  Google Scholar 

  29. Ryan, B.J., Carolan, N. & Ó'Fágáin, C. Horseradish and soybean peroxidases: comparable tools for alternative niches? Trends Biotechnol. 24, 355–363 (2006).

    Article  CAS  Google Scholar 

  30. Ellisman, M.H., Deerinck, T.J., Shu, X. & Sosinsky, G.E. Picking faces out of a crowd: genetic labels for identification of proteins in correlated light and electron microscopy imaging. Methods Cell Biol. 111, 139–155 (2012).

    Article  CAS  Google Scholar 

  31. Hopkins, C., Gibson, A., Stinchcombe, J. & Futter, C. Methods Enzymol. 327, 35–45 (2000).

    Article  CAS  Google Scholar 

  32. Jiang, S. et al. A proteomics approach to the cell-surface interactome using the enzyme-mediated activation of radical sources reaction. Proteomics 12, 54–62 (2012).

    Article  CAS  Google Scholar 

  33. Hubner, N.C. et al. Quantitative proteomics combined with BAC TransgeneOmics reveals in vivo protein interactions. J. Cell Biol. 189, 739–754 (2010).

    Article  CAS  Google Scholar 

  34. Sancak, Y. et al. EMRE is an essential component of the mitochondrial calcium uniporter complex. Science 342, 1379–1382 (2013).

    Article  CAS  Google Scholar 

  35. 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  CAS  Google Scholar 

  36. Cox, J. et al. Andromeda: a peptide search engine integrated into the MaxQuant environment. J. Proteome Res. 10, 1794–1805 (2011).

    Article  CAS  Google Scholar 

  37. Ashburner, M. et al. Gene Ontology: tool for the unification of biology. Nat. Genet. 25, 25–29 (2000).

    Article  CAS  Google Scholar 

  38. Pagliarini, D.J. et al. A mitochondrial protein compendium elucidates complex I disease biology. Cell 134, 112–123 (2008).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank H.-W. Rhee, P. Zou and J. Martell for help with EM samples. E. Vasile (Koch Institute Microscopy Core Facility) acquired the EM images. We thank O. Aygun and S. Han for their edits to the manuscript. V.H., S.S.L. and K.J.C. were supported by National Science Foundation Graduate Research Fellowships. S.S.L. was also supported by a National Defense Science and Engineering Graduate Fellowship. Funding was provided by the US National Institutes of Health (NIH R01 CA186568 to A.Y.T.) and the Howard Hughes Medical Institute Collaborative Initiative Award (to A.Y.T. and S.A.C.).

Author information

Authors and Affiliations

Authors

Contributions

V.H. performed the fluorescence characterization of matrix-APEX labeling and the proteomic data analysis. V.H., N.D.U. and A.Y.T. wrote the paper. All authors edited the paper.

Corresponding author

Correspondence to Alice Y Ting.

Ethics declarations

Competing interests

The Massachusetts Institute of Technology has submitted a patent application on this work.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hung, V., Udeshi, N., Lam, S. et al. Spatially resolved proteomic mapping in living cells with the engineered peroxidase APEX2. Nat Protoc 11, 456–475 (2016). https://doi.org/10.1038/nprot.2016.018

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nprot.2016.018

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