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.

  • Perspective
  • Published:

Proteomics: a pragmatic perspective

Abstract

The evolution of mass spectrometry–based proteomic technologies has advanced our understanding of the complex and dynamic nature of proteomes while concurrently revealing that no 'one-size-fits-all' proteomic strategy can be used to address all biological questions. Whereas some techniques, such as those for analyzing protein complexes, have matured and are broadly applied with great success, others, such as global quantitative protein expression profiling for biomarker discovery, are still confined to a few expert laboratories. In this Perspective, we attempt to distill the wide array of conceivable proteomic approaches into a compact canon of techniques suited to asking and answering specific types of biological questions. By discussing the relationship between the complexity of a biological sample and the difficulty of implementing the appropriate analysis approach, we contrast areas of proteomics broadly usable today with those that require significant technical and conceptual development. We hope to provide nonexperts with a guide for calibrating expectations of what can realistically be learned from a proteomics experiment and for gauging the planning and execution effort. We further provide a detailed supplement explaining the most common techniques in proteomics.

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: Conceptual organization of proteomic experiments.
Figure 2: Applications of proteomic technologies.
Figure 3: Technologies for proteomics.
Figure 4: Protein identification and quantification by mass spectrometry.

Similar content being viewed by others

References

  1. Wasinger, V.C. et al. Progress with gene-product mapping of the Mollicutes: Mycoplasma genitalium. Electrophoresis 16, 1090–1094 (1995).

    Article  CAS  PubMed  Google Scholar 

  2. Ducret, A., Van Oostveen, I., Eng, J.K., Yates, J.R. III & Aebersold, R. High throughput protein characterization by automated reverse-phase chromatography/electrospray tandem mass spectrometry. Protein Sci. 7, 706–719 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Washburn, M.P., Wolters, D. & Yates, J.R. III. Large-scale analysis of the yeast proteome by multidimensional protein identification technology. Nat. Biotechnol. 19, 242–247 (2001).

    Article  CAS  PubMed  Google Scholar 

  4. Wilm, M. et al. Femtomole sequencing of proteins from polyacrylamide gels by nano-electrospray mass spectrometry. Nature 379, 466–469 (1996).

    Article  CAS  PubMed  Google Scholar 

  5. Aebersold, R. Constellations in a cellular universe. Nature 422, 115–116 (2003).

    Article  CAS  PubMed  Google Scholar 

  6. Keshishian, H. et al. Quantification of cardiovascular biomarkers in patient plasma by targeted mass spectrometry and stable isotope dilution. Mol. Cell Proteomics 8, 2339–2349 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Omenn, G.S. et al. Overview of the HUPO Plasma Proteome Project: results from the pilot phase with 35 collaborating laboratories and multiple analytical groups, generating a core dataset of 3020 proteins and a publicly-available database. Proteomics 5, 3226–3245 (2005).

    Article  CAS  PubMed  Google Scholar 

  8. de Godoy, L.M. et al. Comprehensive mass-spectrometry-based proteome quantification of haploid versus diploid yeast. Nature 455, 1251–1254 (2008).

    Article  CAS  PubMed  Google Scholar 

  9. Rush, J. et al. Immunoaffinity profiling of tyrosine phosphorylation in cancer cells. Nat. Biotechnol. 23, 94–101 (2005).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  11. Bouwmeester, T. et al. A physical and functional map of the human TNF-alpha/NF-κB signal transduction pathway. Nat. Cell Biol. 6, 97–105 (2004).

    Article  CAS  PubMed  Google Scholar 

  12. Muzio, M. et al. FLICE, a novel FADD-homologous ICE/CED-3-like protease, is recruited to the CD95 (Fas/APO-1) death–inducing signaling complex. Cell 85, 817–827 (1996).

    Article  CAS  PubMed  Google Scholar 

  13. Heck, A.J. Native mass spectrometry: a bridge between interactomics and structural biology. Nat. Methods 5, 927–933 (2008).

    Article  CAS  PubMed  Google Scholar 

  14. Sharon, M. & Robinson, C.V. The role of mass spectrometry in structure elucidation of dynamic protein complexes. Annu. Rev. Biochem. 76, 167–193 (2007).

    Article  CAS  PubMed  Google Scholar 

  15. Mann, M. & Jensen, O.N. Proteomic analysis of post-translational modifications. Nat. Biotechnol. 21, 255–261 (2003).

    Article  CAS  PubMed  Google Scholar 

  16. Ong, S.E., Mittler, G. & Mann, M. Identifying and quantifying in vivo methylation sites by heavy methyl SILAC. Nat. Methods 1, 119–126 (2004).

    Article  CAS  PubMed  Google Scholar 

  17. Denison, C., Kirkpatrick, D.S. & Gygi, S.P. Proteomic insights into ubiquitin and ubiquitin-like proteins. Curr. Opin. Chem. Biol. 9, 69–75 (2005).

    Article  CAS  PubMed  Google Scholar 

  18. Zaia, J. Mass spectrometry of oligosaccharides. Mass Spectrom. Rev. 23, 161–227 (2004).

    Article  CAS  PubMed  Google Scholar 

  19. Gerber, S.A., Rush, J., Stemman, O., Kirschner, M.W. & Gygi, S.P. Absolute quantification of proteins and phosphoproteins from cell lysates by tandem MS. Proc. Natl. Acad. Sci. USA 100, 6940–6945 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Steen, H., Jebanathirajah, J.A., Springer, M. & Kirschner, M.W. Stable isotope-free relative and absolute quantitation of protein phosphorylation stoichiometry by MS. Proc. Natl. Acad. Sci. USA 102, 3948–3953 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Zhang, X., Jin, Q.K., Carr, S.A. & Annan, R.S. N-terminal peptide labeling strategy for incorporation of isotopic tags: a method for the determination of site-specific absolute phosphorylation stoichiometry. Rapid Commun. Mass Spectrom. 16, 2325–2332 (2002).

    Article  CAS  PubMed  Google Scholar 

  22. Kirkpatrick, D.S., Gerber, S.A. & Gygi, S.P. The absolute quantification strategy: a general procedure for the quantification of proteins and post-translational modifications. Methods 35, 265–273 (2005).

    Article  CAS  PubMed  Google Scholar 

  23. Gerber, S.A., Kettenbach, A.N., Rush, J. & Gygi, S.P. The absolute quantification strategy: application to phosphorylation profiling of human separase serine 1126. Methods Mol. Biol. 359, 71–86 (2007).

    Article  CAS  PubMed  Google Scholar 

  24. Rudd, P.M. et al. The glycosylation of the complement regulatory protein, human erythrocyte CD59. J. Biol. Chem. 272, 7229–7244 (1997).

    Article  CAS  PubMed  Google Scholar 

  25. Phanstiel, D. et al. Mass spectrometry identifies and quantifies 74 unique histone H4 isoforms in differentiating human embryonic stem cells. Proc. Natl. Acad. Sci. USA 105, 4093–4098 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Siuti, N. & Kelleher, N.L. Decoding protein modifications using top-down mass spectrometry. Nat. Methods 4, 817–821 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Mayya, V., Rezual, K., Wu, L., Fong, M.B. & Han, D.K. Absolute quantification of multisite phosphorylation by selective reaction monitoring mass spectrometry: determination of inhibitory phosphorylation status of cyclin-dependent kinases. Mol. Cell. Proteomics 5, 1146–1157 (2006).

    Article  CAS  PubMed  Google Scholar 

  28. Desiere, F. et al. Integration with the human genome of peptide sequences obtained by high-throughput mass spectrometry. Genome Biol. 6, R9 (2005).

    Article  PubMed  Google Scholar 

  29. Margulies, M. et al. Genome sequencing in microfabricated high-density picolitre reactors. Nature 437, 376–380 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Alberts, B. The cell as a collection of protein machines: preparing the next generation of molecular biologists. Cell 92, 291–294 (1998).

    Article  CAS  PubMed  Google Scholar 

  31. Neubauer, G. et al. Identification of the proteins of the yeast U1 small nuclear ribonucleoprotein complex by mass spectrometry. Proc. Natl. Acad. Sci. USA 94, 385–390 (1997).

    Article  CAS  PubMed  PubMed Central  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. Blagoev, B. et al. A proteomics strategy to elucidate functional protein-protein interactions applied to EGF signaling. Nat. Biotechnol. 21, 315–318 (2003).

    Article  CAS  PubMed  Google Scholar 

  34. Rual, J.F. et al. Towards a proteome-scale map of the human protein-protein interaction network. Nature 437, 1173–1178 (2005).

    Article  CAS  PubMed  Google Scholar 

  35. Uetz, P. et al. A comprehensive analysis of protein-protein interactions in Saccharomyces cerevisiae. Nature 403, 623–627 (2000).

    Article  CAS  PubMed  Google Scholar 

  36. Bauer, A. & Kuster, B. Affinity purification-mass spectrometry. Powerful tools for the characterization of protein complexes. Eur. J. Biochem. 270, 570–578 (2003).

    Article  CAS  PubMed  Google Scholar 

  37. Gingras, A.C., Gstaiger, M., Raught, B. & Aebersold, R. Analysis of protein complexes using mass spectrometry. Nat. Rev. Mol. Cell Biol. 8, 645–654 (2007).

    Article  CAS  PubMed  Google Scholar 

  38. Poser, I. et al. BAC TransgeneOmics: a high-throughput method for exploration of protein function in mammals. Nat. Methods 5, 409–415 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Rigaut, G. et al. A generic protein purification method for protein complex characterization and proteome exploration. Nat. Biotechnol. 17, 1030–1032 (1999).

    Article  CAS  PubMed  Google Scholar 

  40. Schmitt-Ulms, G. et al. Time-controlled transcardiac perfusion cross-linking for the study of protein interactions in complex tissues. Nat. Biotechnol. 22, 724–731 (2004).

    Article  CAS  PubMed  Google Scholar 

  41. Andersen, J.S. et al. Proteomic characterization of the human centrosome by protein correlation profiling. Nature 426, 570–574 (2003).

    Article  CAS  PubMed  Google Scholar 

  42. Pflieger, D. et al. Quantitative proteomic analysis of protein complexes: concurrent identification of interactors and their state of phosphorylation. Mol. Cell. Proteomics 7, 326–346 (2008).

    Article  CAS  PubMed  Google Scholar 

  43. Andersen, J.S. et al. Nucleolar proteome dynamics. Nature 433, 77–83 (2005).

    Article  CAS  PubMed  Google Scholar 

  44. Alber, F. et al. Determining the architectures of macromolecular assemblies. Nature 450, 683–694 (2007).

    Article  CAS  PubMed  Google Scholar 

  45. Alber, F. et al. The molecular architecture of the nuclear pore complex. Nature 450, 695–701 (2007).

    Article  CAS  PubMed  Google Scholar 

  46. Hochleitner, E.O., Sondermann, P. & Lottspeich, F. Determination of the stoichiometry of protein complexes using liquid chromatography with fluorescence and mass spectrometric detection of fluorescently labeled proteolytic peptides. Proteomics 4, 669–676 (2004).

    Article  CAS  PubMed  Google Scholar 

  47. Menetret, J.F. et al. Single copies of Sec61 and TRAP associate with a nontranslating mammalian ribosome. Structure 16, 1126–1137 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Nanavati, D., Gucek, M., Milne, J.L., Subramaniam, S. & Markey, S.P. Stoichiometry and absolute quantification of proteins with mass spectrometry using fluorescent and isotope-labeled concatenated peptide standards. Mol. Cell. Proteomics 7, 442–447 (2008).

    Article  CAS  PubMed  Google Scholar 

  49. Hernandez, H. & Robinson, C.V. Determining the stoichiometry and interactions of macromolecular assemblies from mass spectrometry. Nat. Protoc. 2, 715–726 (2007).

    Article  CAS  PubMed  Google Scholar 

  50. Lorenzen, K., Olia, A.S., Uetrecht, C., Cingolani, G. & Heck, A.J. Determination of stoichiometry and conformational changes in the first step of the P22 tail assembly. J. Mol. Biol. 379, 385–396 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Gavin, A.C. et al. Functional organization of the yeast proteome by systematic analysis of protein complexes. Nature 415, 141–147 (2002).

    Article  CAS  PubMed  Google Scholar 

  52. Maiolica, A. et al. Structural analysis of multiprotein complexes by cross-linking, mass spectrometry, and database searching. Mol. Cell. Proteomics 6, 2200–2211 (2007).

    Article  CAS  PubMed  Google Scholar 

  53. Sinz, A. Chemical cross-linking and mass spectrometry to map three-dimensional protein structures and protein-protein interactions. Mass Spectrom. Rev. 25, 663–682 (2006).

    Article  CAS  PubMed  Google Scholar 

  54. Ewing, R.M. et al. Large-scale mapping of human protein-protein interactions by mass spectrometry. Mol. Syst. Biol. 3, 89 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  55. Gavin, A.C. et al. Proteome survey reveals modularity of the yeast cell machinery. Nature 440, 631–636 (2006).

    Article  CAS  PubMed  Google Scholar 

  56. Krogan, N.J. et al. Global landscape of protein complexes in the yeast Saccharomyces cerevisiae. Nature 440, 637–643 (2006).

    Article  CAS  PubMed  Google Scholar 

  57. Ho, Y. et al. Systematic identification of protein complexes in Saccharomyces cerevisiae by mass spectrometry. Nature 415, 180–183 (2002).

    Article  CAS  PubMed  Google Scholar 

  58. Kolch, W. Meaningful relationships: the regulation of the Ras/Raf/MEK/ERK pathway by protein interactions. Biochem. J. 351, 289–305 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Schubert, P., Hoffman, M.D., Sniatynski, M.J. & Kast, J. Advances in the analysis of dynamic protein complexes by proteomics and data processing. Anal. Bioanal. Chem. 386, 482–493 (2006).

    Article  CAS  PubMed  Google Scholar 

  60. White, F.M. Quantitative phosphoproteomic analysis of signaling network dynamics. Curr. Opin. Biotechnol. 19, 404–409 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Kung, L.A. & Snyder, M. Proteome chips for whole-organism assays. Nat. Rev. Mol. Cell Biol. 7, 617–622 (2006).

    Article  CAS  PubMed  Google Scholar 

  62. Paweletz, C.P. et al. Reverse phase protein microarrays which capture disease progression show activation of pro-survival pathways at the cancer invasion front. Oncogene 20, 1981–1989 (2001).

    Article  CAS  PubMed  Google Scholar 

  63. Speer, R. et al. Molecular network analysis using reverse phase protein microarrays for patient tailored therapy. Adv. Exp. Med. Biol. 610, 177–186 (2008).

    Article  CAS  PubMed  Google Scholar 

  64. Zhu, H. et al. Global analysis of protein activities using proteome chips. Science 293, 2101–2105 (2001).

    Article  CAS  PubMed  Google Scholar 

  65. Huang, P.H. & White, F.M. Phosphoproteomics: unraveling the signaling web. Mol. Cell 31, 777–781 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Picotti, P., Bodenmiller, B., Mueller, L.N., Domon, B. & Aebersold, R. Full dynamic range proteome analysis of S. cerevisiae by targeted proteomics. Cell 138, 795–806 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Van, P.T. et al. Halobacterium salinarum NRC-1 PeptideAtlas: toward strategies for targeted proteomics and improved proteome coverage. J. Proteome Res. 7, 3755–3764 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  68. King, N.L. et al. Analysis of the Saccharomyces cerevisiae proteome with PeptideAtlas. Genome Biol. 7, R106 (2006).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  69. Chen, E.I., Hewel, J., Felding-Habermann, B. & Yates, J.R. III. Large scale protein profiling by combination of protein fractionation and multidimensional protein identification technology (MudPIT). Mol. Cell. Proteomics 5, 53–56 (2006).

    Article  CAS  PubMed  Google Scholar 

  70. Malmstrom, J. et al. Optimized peptide separation and identification for mass spectrometry based proteomics via free-flow electrophoresis. J. Proteome Res. 5, 2241–2249 (2006).

    Article  PubMed  CAS  Google Scholar 

  71. Hubner, N.C., Ren, S. & Mann, M. Peptide separation with immobilized pI strips is an attractive alternative to in-gel protein digestion for proteome analysis. Proteomics 8, 4862–4872 (2008).

    Article  CAS  PubMed  Google Scholar 

  72. Chen, E.I., McClatchy, D., Park, S.K. & Yates, J.R. III. Comparisons of mass spectrometry compatible surfactants for global analysis of the mammalian brain proteome. Anal. Chem. 80, 8694–8701 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Graumann, J. et al. Stable isotope labeling by amino acids in cell culture (SILAC) and proteome quantitation of mouse embryonic stem cells to a depth of 5,111 proteins. Mol. Cell. Proteomics 7, 672–683 (2008).

    Article  CAS  PubMed  Google Scholar 

  74. Schirle, M., Heurtier, M.A. & Kuster, B. Profiling core proteomes of human cell lines by one-dimensional PAGE and liquid chromatography-tandem mass spectrometry. Mol. Cell. Proteomics 2, 1297–1305 (2003).

    Article  CAS  PubMed  Google Scholar 

  75. Kuster, B., Schirle, M., Mallick, P. & Aebersold, R. Scoring proteomes with proteotypic peptide probes. Nat. Rev. Mol. Cell Biol. 6, 577–583 (2005).

    Article  CAS  PubMed  Google Scholar 

  76. Mallick, P. et al. Computational prediction of proteotypic peptides for quantitative proteomics. Nat. Biotechnol. 25, 125–131 (2007).

    Article  CAS  PubMed  Google Scholar 

  77. Tabb, D.L. et al. Repeatability and reproducibility in proteomic identifications by liquid chromatography-tandem mass spectrometry. J. Proteome Res. 9, 761–776 (2009).

    Article  CAS  Google Scholar 

  78. Swaney, D.L., Wenger, C.D. & Coon, J.J. Value of using multiple proteases for large-scale mass spectrometry-based proteomics. J. Proteome Res. 9, 1323–1329 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Gruhler, A. et al. Quantitative phosphoproteomics applied to the yeast pheromone signaling pathway. Mol. Cell. Proteomics 4, 310–327 (2005).

    Article  CAS  PubMed  Google Scholar 

  80. Pinkse, M.W. 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).

    Article  CAS  PubMed  Google Scholar 

  81. Reiland, S. et al. Large-scale Arabidopsis phosphoproteome profiling reveals novel chloroplast kinase substrates and phosphorylation networks. Plant Physiol. 150, 889–903 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  82. Villen, J., Beausoleil, S.A., Gerber, S.A. & Gygi, S.P. Large-scale phosphorylation analysis of mouse liver. Proc. Natl. Acad. Sci. USA 104, 1488–1493 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  83. Zielinska, D.F., Gnad, F., Jedrusik-Bode, M., Wisniewski, J.R. & Mann, M. Caenorhabditis elegans has a phosphoproteome atypical for metazoans that is enriched in developmental and sex determination proteins. J. Proteome Res. 8, 4039–4049 (2009).

    Article  CAS  PubMed  Google Scholar 

  84. Lemeer, S. et al. Endogenous phosphotyrosine signaling in zebrafish embryos. Mol. Cell. Proteomics 6, 2088–2099 (2007).

    Article  CAS  PubMed  Google Scholar 

  85. Zhang, Y. et al. Time-resolved mass spectrometry of tyrosine phosphorylation sites in the epidermal growth factor receptor signaling network reveals dynamic modules. Mol. Cell. Proteomics 4, 1240–1250 (2005).

    Article  CAS  PubMed  Google Scholar 

  86. Au, C.E. et al. Organellar proteomics to create the cell map. Curr. Opin. Cell Biol. 19, 376–385 (2007).

    Article  CAS  PubMed  Google Scholar 

  87. Lilley, K.S. & Dupree, P. Plant organelle proteomics. Curr. Opin. Plant Biol. 10, 594–599 (2007).

    Article  CAS  PubMed  Google Scholar 

  88. Dunkley, T.P., Watson, R., Griffin, J.L., Dupree, P. & Lilley, K.S. Localization of organelle proteins by isotope tagging (LOPIT). Mol. Cell. Proteomics 3, 1128–1134 (2004).

    Article  CAS  PubMed  Google Scholar 

  89. Jang, J.H. & Hanash, S. Profiling of the cell surface proteome. Proteomics 3, 1947–1954 (2003).

    Article  CAS  PubMed  Google Scholar 

  90. Wollscheid, B. et al. Mass-spectrometric identification and relative quantification of N-linked cell surface glycoproteins. Nat. Biotechnol. 27, 378–386 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Zhang, H., Li, X.J., Martin, D.B. & Aebersold, R. Identification and quantification of N-linked glycoproteins using hydrazide chemistry, stable isotope labeling and mass spectrometry. Nat. Biotechnol. 21, 660–666 (2003).

    Article  CAS  PubMed  Google Scholar 

  92. Bantscheff, M., Schirle, M., Sweetman, G., Rick, J. & Kuster, B. Quantitative mass spectrometry in proteomics: a critical review. Anal. Bioanal. Chem. 389, 1017–1031 (2007).

    Article  CAS  PubMed  Google Scholar 

  93. Griffin, N.M. et al. Label-free, normalized quantification of complex mass spectrometry data for proteomic analysis. Nat. Biotechnol. 28, 83–89 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  94. Saito, A. et al. AYUMS: an algorithm for completely automatic quantitation based on LC-MS/MS proteome data and its application to the analysis of signal transduction. BMC Bioinformatics 8, 15 (2007).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  95. Wolf-Yadlin, A., Hautaniemi, S., Lauffenburger, D.A. & White, F.M. Multiple reaction monitoring for robust quantitative proteomic analysis of cellular signaling networks. Proc. Natl. Acad. Sci. USA 104, 5860–5865 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  96. Ono, M. et al. Label-free quantitative proteomics using large peptide data sets generated by nanoflow liquid chromatography and mass spectrometry. Mol. Cell. Proteomics 5, 1338–1347 (2006).

    Article  CAS  PubMed  Google Scholar 

  97. Parish, R. Comparison of linear regression methods when both variables contain error: relation to clinical studies. Ann. Pharmacother. 23, 891–898 (1989).

    CAS  Google Scholar 

  98. Mueller, L.N. et al. SuperHirn - a novel tool for high resolution LC-MS-based peptide/protein profiling. Proteomics 7, 3470–3480 (2007).

    Article  CAS  PubMed  Google Scholar 

  99. Cox, J. et al. A practical guide to the MaxQuant computational platform for SILAC-based quantitative proteomics. Nat. Protoc. 4, 698–705 (2009).

    Article  CAS  PubMed  Google Scholar 

  100. 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 

  101. Han, D.K., Eng, J., Zhou, H. & Aebersold, R. Quantitative profiling of differentiation-induced microsomal proteins using isotope-coded affinity tags and mass spectrometry. Nat. Biotechnol. 19, 946–951 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  102. Faca, V. et al. Quantitative analysis of acrylamide labeled serum proteins by LC-MS/MS. J. Proteome Res. 5, 2009–2018 (2006).

    Article  CAS  PubMed  Google Scholar 

  103. Rauch, A. et al. Computational Proteomics Analysis System (CPAS): an extensible, open-source analytic system for evaluating and publishing proteomic data and high throughput biological experiments. J. Proteome Res. 5, 112–121 (2006).

    Article  CAS  PubMed  Google Scholar 

  104. Jaffe, J.D. et al. PEPPeR, a platform for experimental proteomic pattern recognition. Mol. Cell. Proteomics 5, 1927–1941 (2006).

    Article  CAS  PubMed  Google Scholar 

  105. Park, S.K., Venable, J.D., Xu, T. & Yates, J.R. III. A quantitative analysis software tool for mass spectrometry-based proteomics. Nat. Methods 5, 319–322 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Du, X. et al. A computational strategy to analyze label-free temporal bottom-up proteomics data. J. Proteome Res. 7, 2595–2604 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  107. Domon, B. & Aebersold, R. Three strategies for quantitative proteomics and their use. Nat. Biotechnol. 28, 710–721 (2010).

    Article  CAS  PubMed  Google Scholar 

  108. Zhang, R. & Regnier, F.E. Minimizing resolution of isotopically coded peptides in comparative proteomics. J. Proteome Res. 1, 139–147 (2002).

    Article  CAS  PubMed  Google Scholar 

  109. Zhang, R., Sioma, C.S., Wang, S. & Regnier, F.E. Fractionation of isotopically labeled peptides in quantitative proteomics. Anal. Chem. 73, 5142–5149 (2001).

    Article  CAS  PubMed  Google Scholar 

  110. Zhang, Y. et al. A robust error model for iTRAQ quantification reveals divergent signaling between oncogenic FLT3 mutants in acute myeloid leukemia. Mol. Cell Proteomics 7, 780–790 (2009).

    Google Scholar 

  111. Faca, V. et al. Contribution of protein fractionation to depth of analysis of the serum and plasma proteomes. J. Proteome Res. 6, 3558–3565 (2007).

    Article  CAS  PubMed  Google Scholar 

  112. Liu, Y., Patricelli, M.P. & Cravatt, B.F. Activity-based protein profiling: the serine hydrolases. Proc. Natl. Acad. Sci. USA 96, 14694–14699 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  113. Bantscheff, M. et al. Quantitative chemical proteomics reveals mechanisms of action of clinical ABL kinase inhibitors. Nat. Biotechnol. 25, 1035–1044 (2007).

    Article  CAS  PubMed  Google Scholar 

  114. Cravatt, B.F., Wright, A.T. & Kozarich, J.W. Activity-based protein profiling: from enzyme chemistry to proteomic chemistry. Annu. Rev. Biochem. 77, 383–414 (2008).

    Article  CAS  PubMed  Google Scholar 

  115. Rikova, K. et al. Global survey of phosphotyrosine signaling identifies oncogenic kinases in lung cancer. Cell 131, 1190–1203 (2007).

    Article  CAS  PubMed  Google Scholar 

  116. Blethrow, J.D., Glavy, J.S., Morgan, D.O. & Shokat, K.M. Covalent capture of kinase-specific phosphopeptides reveals Cdk1-cyclin B substrates. Proc. Natl. Acad. Sci. USA 105, 1442–1447 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  117. Emmert-Buck, M.R. et al. Laser capture microdissection. Science 274, 998–1001 (1996).

    Article  CAS  PubMed  Google Scholar 

  118. Lu, Q. et al. Analysis of mouse brain microvascular endothelium using immuno-laser capture microdissection coupled to a hybrid linear ion trap with Fourier transform-mass spectrometry proteomics platform. Electrophoresis 29, 2689–2695 (2008).

    Article  CAS  PubMed  Google Scholar 

  119. Johann, D.J. et al. Approaching solid tumor heterogeneity on a cellular basis by tissue proteomics using laser capture microdissection and biological mass spectrometry. J. Proteome Res. 8, 2310–2318 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  120. Reimel, B.A. et al. Proteomics on fixed tissue specimens - a review. Curr. Proteomics 6, 63–69 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  121. Faca, V.M. et al. A mouse to human search for plasma proteome changes associated with pancreatic tumor development. PLoS Med. 5, e123 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  122. Harsha, H.C. et al. A compendium of potential biomarkers of pancreatic cancer. PLoS Med. 6, e1000046 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  123. Bandhakavi, S., Stone, M.D., Onsongo, G., Van Riper, S.K. & Griffin, T.J. A dynamic range compression and three-dimensional peptide fractionation analysis platform expands proteome coverage and the diagnostic potential of whole saliva. J. Proteome Res. 8, 5590–5600 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  124. Righetti, P.G., Boschetti, E., Lomas, L. & Citterio, A. Protein equalizer technology: the quest for a “democratic proteome”. Proteomics 6, 3980–3992 (2006).

    Article  CAS  PubMed  Google Scholar 

  125. Brand, J., Haslberger, T., Zolg, W., Pestlin, G. & Palme, S. Depletion efficiency and recovery of trace markers from a multiparameter immunodepletion column. Proteomics 6, 3236–3242 (2006).

    Article  CAS  PubMed  Google Scholar 

  126. Seam, N. et al. Quality control of serum albumin depletion for proteomic analysis. Clin. Chem. 53, 1915–1920 (2007).

    Article  CAS  PubMed  Google Scholar 

  127. Anderson, N.L. et al. Mass spectrometric quantitation of peptides and proteins using Stable Isotope Standards and Capture by Anti-Peptide Antibodies (SISCAPA). J. Proteome Res. 3, 235–244 (2004).

    Article  CAS  PubMed  Google Scholar 

  128. Kuhn, E. et al. Developing multiplexed assays for troponin I and interleukin-33 in plasma by peptide immunoaffinity enrichment and targeted mass spectrometry. Clin. Chem. 55, 1108–1117 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  129. Pitteri, S.J. et al. Integrated proteomic analysis of human cancer cells and plasma from tumor bearing mice for ovarian cancer biomarker discovery. PLoS ONE 4, e7916 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  130. Katayama, H. et al. Application of serum proteomics to the Women's Health Initiative conjugated equine estrogens trial reveals a multitude of effects relevant to clinical findings. Genome Med. 1, 47 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  131. Faça, V., Wang, H. & Hanash, S. Proteomic global profiling for cancer biomarker discovery. Methods Mol. Biol. 492, 309–320 (2009).

    Article  PubMed  CAS  Google Scholar 

  132. Faça, V.M. & Hanash, S.M. In-depth proteomics to define the cell surface and secretome of ovarian cancer cells and processes of protein shedding. Cancer Res. 69, 728–730 (2009).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  133. Faça, V.M. et al. Proteomic analysis of ovarian cancer cells reveals dynamic processes of protein secretion and shedding of extra-cellular domains. PLoS ONE 3, e2425 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  134. Hanash, S.M., Pitteri, S.J. & Faça, V.M. Mining the plasma proteome for cancer biomarkers. Nature 452, 571–579 (2008).

    Article  CAS  PubMed  Google Scholar 

  135. Caprioli, R.M., Farmer, T.B. & Gile, J. Molecular imaging of biological samples: localization of peptides and proteins using MALDI-TOF MS. Anal. Chem. 69, 4751–4760 (1997).

    Article  CAS  PubMed  Google Scholar 

  136. Cornett, D.S., Reyzer, M.L., Chaurand, P. & Caprioli, R.M. MALDI imaging mass spectrometry: molecular snapshots of biochemical systems. Nat. Methods 4, 828–833 (2007).

    Article  CAS  PubMed  Google Scholar 

  137. Hsieh, Y., Chen, J. & Korfmacher, W.A. Mapping pharmaceuticals in tissues using MALDI imaging mass spectrometry. J. Pharmacol. Toxicol. Methods 55, 193–200 (2007).

    Article  CAS  PubMed  Google Scholar 

  138. Woods, A.S. & Jackson, S.N. Brain tissue lipidomics: direct probing using matrix-assisted laser desorption/ionization mass spectrometry. AAPS J. 8, E391–E395 (2006).

    Article  PubMed  PubMed Central  Google Scholar 

  139. Taguchi, F. et al. Mass spectrometry to classify non-small-cell lung cancer patients for clinical outcome after treatment with epidermal growth factor receptor tyrosine kinase inhibitors: a multicohort cross-institutional study. J. Natl. Cancer Inst. 99, 838–846 (2007).

    Article  CAS  PubMed  Google Scholar 

  140. Deutsch, E.W. et al. Human Plasma PeptideAtlas. Proteomics 5, 3497–3500 (2005).

    Article  CAS  PubMed  Google Scholar 

  141. Deutsch, E.W., Lam, H. & Aebersold, R. PeptideAtlas: a resource for target selection for emerging targeted proteomics workflows. EMBO Rep. 9, 429–434 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  142. Zhang, Q. et al. A mouse plasma peptide atlas as a resource for disease proteomics. Genome Biol. 9, R93 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  143. Castellana, N.E. et al. Discovery and revision of Arabidopsis genes by proteogenomics. Proc. Natl. Acad. Sci. USA 105, 21034–21038 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  144. Gupta, N. et al. Comparative proteogenomics: combining mass spectrometry and comparative genomics to analyze multiple genomes. Genome Res. 18, 1133–1142 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  145. Gupta, N. et al. Whole proteome analysis of post-translational modifications: applications of mass-spectrometry for proteogenomic annotation. Genome Res. 17, 1362–1377 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  146. Tanner, S. et al. InsPecT: identification of posttranslationally modified peptides from tandem mass spectra. Anal. Chem. 77, 4626–4639 (2005).

    Article  CAS  PubMed  Google Scholar 

  147. Gerszten, R.E., Carr, S.A. & Sabatine, M. Integration of proteomic-based tools for improved biomarkers of myocardial injury. Clin. Chem. 56, 194–201 (2010).

    Article  CAS  PubMed  Google Scholar 

  148. Kentsis, A. et al. Discovery and validation of urine markers of acute pediatric appendicitis using high-accuracy mass spectrometry. Ann. Emerg. Med. 55, 62–70 (2010).

    Article  PubMed  Google Scholar 

  149. Rifai, N., Gillette, M.A. & Carr, S.A. Protein biomarker discovery and validation: the long and uncertain path to clinical utility. Nat. Biotechnol. 24, 971–983 (2006).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

The authors thank M. Bergman, C. Flinders, K. Kramer and S. Mumenthaler for comments on the manuscript. The work of P.M. was supported by NCI-1U54CA143907 and NCI-U54CA119367.

Author information

Authors and Affiliations

Authors

Corresponding authors

Correspondence to Parag Mallick or Bernhard Kuster.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Glossary, Supplementary Figure 1 and Supplementary Techniques (PDF 944 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Mallick, P., Kuster, B. Proteomics: a pragmatic perspective. Nat Biotechnol 28, 695–709 (2010). https://doi.org/10.1038/nbt.1658

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nbt.1658

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