829
Views
42
CrossRef citations to date
0
Altmetric
Editorial

Quantitative proteomics and biomarker discovery in human cancer

, &
Pages 115-118 | Published online: 09 Jan 2014

Early diagnosis and prevention is crucial for reducing the mortality and morbidity of cancer. Current clinical examination, such as mammography and invasive needle or surgical evaluation for breast cancer, and chest x-ray for lung cancer, for example, is often not sufficiently sensitive for early diagnosis. In order to detect cancer in its early stages, it is necessary to identify biomarkers for asymptomatic patients who may have cancer. Biomarkers could be proteins, metabolites or electrolytes whose differential expression indicates the presence of disease Citation[1]. Biomarkers of protein origin are powerful, both in monitoring the development of cancer and in determining the efficacy and safety of drugs. Novel biomarkers are urgently needed for the early diagnosis and treatment of cancer. Nowadays, the remarkable progress in proteomics technology has offered unprecedented opportunity for biomarker discovery.

Traditionally, 2DE has been the standard method for comparing protein expression between normal and disease-perturbed states. However, it is now known that 2DE lacks resolution and sensitivity. Only abundant proteins can be resolved on a 2D gel. Biomarkers, which are usually low in abundance, are rarely found with the 2DE method. Gel-free or mass spectrometry (MS)-based proteomics techniques are emerging as the choice for quantitatively measuring protein levels, due to better sensitivity and reproducibility over 2DE-based methods Citation[2,3]. However, quantitative proteomic profiling of complex biological samples for the purposes of biomarker discovery remains a challenge in proteomics. Multiple innovative profiling techniques have been introduced with the aims of comprehensively identifying and quantifying proteins. These can be approximately divided into two categories: isotope-labeled and label-free MS.

Isotope-labeled mass spectrometry

Isotope-labeling methods have been developed that introduce stable isotope tags to proteins via chemical reactions using isotope-coded affinity tags (ICAT) Citation[4,5] and isobaric tag for relative and absolute quantitation (iTRAQ) Citation[6], enzymatic labeling, for example using 18O water for trypsin digestion Citation[7], or via metabolic labeling (stable isotope labeling of amino acids in cell culture [SILAC]) Citation[8]. The pioneering ICAT technology selectively targets peptides containing a specific amino acid (cysteine) with a stable isotope-coded internal reference or standard Citation[9]. The extracted proteins from treatment and control samples are labeled with either light or heavy ICAT reagents by reacting with cysteinyl thiols on the proteins. Peptides containing the labeled and unlabeled ICAT tags are recovered by avidin affinity chromatography and are then analyzed by liquid chromatography (LC)-MS/MS. Differential protein expression is determined by the isotope peak ratio of the peptide. Enrichment of low-abundance proteins can be performed through cell lysate fractionation Citation[10]. ICAT technology has been widely used for protein identification and quantification in mammalian, liver and breast tumor cells Citation[11]. However, disadvantages of ICAT analyses are obvious: it is only applicable to proteins containing cysteine, it can only identify 300–400 proteins, far fewer than the 2DE method, and the peptides contain large labels, which makes database searching more difficult, especially for short peptides Citation[10].

Isobaric tag for relative and absolute quantitation is another labeling technique first developed by Ross et al., which use special isobaric tags to label proteins extracted from samples for comparison Citation[12]. The proteins from control and treatment samples have the same mass after reacting with the iTRAQ reagents. These peptides give four ion species of different masses upon collision-induced dissociation. These ions allow samples to be quantified in MS/MS mode Citation[12]. The amine specificity of the labeling reagents makes most peptides amenable to this labeling strategy with no loss of information. This is especially important for proteins with post-translational modifications, such as phosphorylation and glycosylation. In addition, the multiplexing capacity of these reagents allows for comparison among multiple cellular states. However, as a chemical labeling method, iTRAQ may generate side products during labeling and cause some loss of analytic sensitivity because chemical strategies involve a derivatization step that might not be complete.

Absolute quantification (AQUA), uses synthesized isotopically labeled peptides that mimic native peptides as internal standards Citation[13,14]. The method has the potential for high-throughput and multiplexed sample analysis Citation[15]. SILAC, which takes a similar strategy but utilizes different media containing light or heavy forms of particular amino acids, has emerged as a popular label-based quantification technique Citation[16]. It was first developed by Mann et al., based on metabolic incorporation of ‘light-’ or ‘heavy-’ form amino acids into the proteins in living cultured cells Citation[8]. Usually, heavily labeled (13C or 15N) arginine, lysine or both are used in culture medium to ensure complete labeling of every trypsinized peptide fragment. In experiments, one cell population is fed with regular amino acids, while the other is fed with 13C- or 15N-labeled amino acids. After several rounds of cell division, heavy amino acids will be incorporated into newly synthesized proteins. In the MS spectrum, the light and heavy peptides will show up as two distinct peaks separated by the incremental mass of the labeled amino acids. By comparing the signal intensity, relative quantification can be achieved. Owing to its simplicity in principle, SILAC is widely used for biomarker discovery Citation[17], cell signaling dynamics Citation[18], identification of posttranslational modification sites Citation[19,20], protein–protein interaction Citation[21–23] and subcellular proteomics Citation[24].

The dynamics of protein turnover is another key feature to the understanding of regulation of protein expression in cells Citation[25,26]. Recently, we developed a general method for determination of protein synthesis rate using labeling of amino acids with deuterium or 15N at low enrichment Citation[27,28]. This method, ‘modified SILAC’ (mSILAC), can measure protein synthesis rate quantitatively, based on analysis of mass isotopmer distribution (MIDA) of the newly synthesized protein Citation[28]. Once precursor enrichment is known, protein synthesis is determined from isotopomer distribution. In experiments with 30–50% enriched 15N amino acids, incorporation of 15N amino acids result in sufficient mass shift in the new protein, the 15N enrichment can be estimated from the mass shift by curve fitting, and the expected isotopomer distribution of the new peptide can be generated by the concatenation function. Synthesis rate is then calculated by multiple linear regression analysis of the observed peptide spectrum on the expected new and the old (unlabeled) spectra Citation[28]. This method obviates the need for the use of a 100% newly synthesized protein as a reference as in Vogt’s methods Citation[29,30]. The concatenation function provides an ideal 100% labeled spectrum and multiple regression analysis uses all the information from the mass spectrum. Our mathematical algorithm represents a major improvement in the calculation of protein synthesis rate, permitting the use of isotope labeling of protein through the pathways of amino acid metabolism with low cost isotopes Citation[27,28].

For small peptides, another method, multiple-reaction monitoring (MRM), attempts to monitor prespecified ions (and their daughter product ions after MS/MS) as specific signatures for a particular protein instead of aiming to measure everything in a sample. In this method, initial selective scanning for a particular precursor ion in the first MS is followed by scans of a particular transition. Only one of the ions produced during collision-induced dissociation is selected with the aid of prior knowledge of reliable precursors and transition state Citation[31]. Due to limitations in mass spectrometer dynamic range and resolution of chromatography separation, MRM is frequently combined with immunodepletion, size-exclusion chromatography Citation[32] or enrichment of stable isotope standards with capture by antipeptide antibody Citation[33]. These hybrid methods are particularly attractive because they can precisely measure the quantity of low-abundance proteins, which are otherwise bypassed in other conventional methods. MRM is comparable to ELISA with regard to sensitivity but obviate the necessity of antibody, which is costly to obtain and sometimes unavailable. MRM is normally more accurate than ELISA because it does not have propensity of cross-reaction as antibodies have.

Label-free mass spectrometry

With the advances of new instrumentation, computing power and advanced bioinformatics, generic label-free LC-MS shotgun screening methods, such as MudPIT, have been alternatives for relative and absolute protein quantitation in biological samples. Without requiring modification of the analytes, the label-free approaches were based on the observation that MS intensity is linearly proportional to ion concentration, even in complex samples such as serum Citation[34]. However, it was later observed that not all peptides are equally detectable because of competition between ions, dynamic-range limitation and sensitivity of MS instrumentation Citation[35]. A new technique, known as spectral counting, circumvented these pitfalls by not looking at differences in ion peak areas or intensities, as used in isotope-based methods Citation[35]. Nevertheless, the main bottleneck for label-free LC-MS still lies in computer softwares and sample preparation. Although many software tools are available, there is a large space for improvement, such as user interface and calculation efficiency of workflow. Moreover, the accuracy of label-free quantitation depends largely on experimental set-up, such as protein extraction and sample stability, which is illustrated by the fact that the quantitative reproducibility of technical replicates is much better than that of experimental replicates Citation[36,37]. With improvements in the efficiency of data analysis workflow, label-free MS will potentially be widely used for biomarker discovery and validation.

Conclusion

The recent development of MS-based strategies for absolute protein quantification offers great opportunity for biomarker discovery. While the prospects of this technology are exciting and promising, the current methodology is far from perfect. First, most current methods require complicated sample preparation, such as immunosubtraction, multidimensional LC separation, immunoaffinity and solid-phase extraction, in order to enhance the analytical dynamic range and detection sensitivity. To establish high-throughput pipelines, we should ideally have a one-step preparation. Second, useful and validated biomarkers are still rare based on these developed methods because low-abundance biomarkers are always immersed in large quantities of routine proteins, especially in plasma samples. There is a large space for improvement of sensitivity.

Financial & competing interests disclosure

This work is jointly funded by the Bone Biology Program of the Cancer and Smoking Related Disease Research Program and the Nebraska Tobacco Settlement Biomedical Research Program (289104–845610 to GX), and partially supported by a grant awarded to Wai-Nang Paul Lee from the UCLA Center of Excellence in Pancreatic disease (P01 AT003960–01) and Harbor-UCLA GCRC Mass Spectrometry Core (M01 RR00425–33). The authors have no other relevant affiliations or financial involvement with any organization or entity with a financial interest in or financial conflict with the subject matter or materials discussed in the manuscript apart from those disclosed.

No writing assistance was utilized in the production of this manuscript.

References

  • Rifai N, Gillette MA, Carr SA. Protein biomarker discovery and validation: the long and uncertain path to clinical utility. Nat. Biotechnol.24, 971–983 (2006).
  • Aebersold R, Mann M. Mass spectrometry-based proteomics. Nature.422, 198–207 (2003).
  • Xiao GG, Recker, RR, Deng, HW. Recent advances in proteomics and cancer biomarker discovery. Clin. Med. Oncol.2, 1–10 (2008).
  • Gygi SP, Rist B, Gerber SA et al. Quantitative analysis of complex protein mixtures using isotope-coded affinity tags. Nat. Biotechnol.17, 994–999 (1999).
  • Haqqani AS, Nesic M, Preston E et al. Characterization of vascular protein expression patterns in cerebral ischemia/reperfusion using laser capture microdissection and ICAT-nanoLC-MS/MS. FASEB J.19, 1809–1821 (2005).
  • Shadforth IP, Dunkley TP, Lilley KS et al. i-Tracker: for qauntitative proteomics using iTRAQ. BMC Genomics.6, 145 (2005).
  • Heller M, Mattou H, Menzel C et al. Trypsin catalyzed 16O-to-18O exchange for comparative proteomics: tandem mass spectrometry comparison using MALDI-TOF, ESI-QTOF, and ESI-ion trap mass spectrometers. J. Am. Soc. Mass Spectrom.14, 704–718 (2003).
  • Ong SE, Blagoev B, Kratchmarova I 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).
  • Shiio Y, Aebersold R. Quantitative proteome analysis using isotope-coded affinity tags and mass spectrometry. Nat. Protoc.1, 139–145 (2006).
  • Graves PR, Haystead TA. Molecular biologist’s guide to proteomics. Microbiol. Mol. Biol. Rev.66, 39–63 (2002).
  • Somiari RI, Somiari S, Russell S et al. Proteomics of breast carcinoma. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci.815, 215–225 (2005).
  • Ross PL, Huang YN, Marchese JN et al. Multiplexed protein quantitation in Saccharomyces cerevisiae using amine-reactive isobaric tagging reagents. Mol. Cell Proteomics.3, 1154–1169 (2004).
  • Gerber SA, Kettenbach AN, Rush J et al. The absolute quantification strategy: application to phosphorylation profiling of human separase serine 1126. Methods Mol. Biol.359, 71–86 (2007).
  • Kirkpatrick DS, Gerber SA, Gygi SP. The absolute quantification strategy: a general procedure for the quantification of proteins and post-translational modifications. Methods.35, 265–273 (2005).
  • Aggarwal K, Choe LH, Lee KH. Shotgun proteomics using the iTRAQ isobaric tags. Brief Funct. Genomic Proteomic.5, 112–120 (2006).
  • Mann M. Functional and quantitative proteomics using SILAC. Nat. Rev. Mol. Cell Biol.7, 952–958 (2006).
  • Gronborg M, Kristiansen TZ, Iwahori A et al. Biomarker discovery from pancreatic cancer secretome using a differential proteomic approach. Mol. Cell. Proteomics.5, 157–171 (2006).
  • Guerrera IC, Keep NH, Godovac-Zimmermann J. Proteomics study reveals cross-talk between Rho guanidine nucleotide dissociation inhibitor 1 post-translational modifications in epidermal growth factor stimulated fibroblasts. J. Proteome Res.6, 2623–2630 (2007).
  • Liang X, Fonnum G, Hajivandi M et al. Quantitative comparison of IMAC and TiO2 surfaces used in the study of regulated, dynamic protein phosphorylation. J. Am. Soc. Mass Spectrom.18, 1932–1944 (2007).
  • Soufi B, Jers C, Hansen ME et al. Insights from site-specific phosphoproteomics in bacteria. Biochim. Biophys. Acta.1784, 186–192 (2008).
  • Selbach M, Mann M. Protein interaction screening by quantitative immunoprecipitation combined with knockdown (QUICK). Nat. Methods.3, 981–983 (2006).
  • Bose R, Molina H, Patterson AS et al. Phosphoproteomic analysis of Her2/neu signaling and inhibition. Proc. Natl Acad. Sci. USA.103, 9773–9778 (2006).
  • Foster LJ, Rudich A, Talior I et al. Insulin-dependent interactions of proteins with GLUT4 revealed through stable isotope labeling by amino acids in cell culture (SILAC). J. Proteome Res.5, 64–75 (2006).
  • Waanders LF, Hanke S, Mann M. Top-down quantitation and characterization of SILAC-labeled proteins. J. Am. Soc. Mass Spectrom.18, 2058–2064 (2007).
  • Doherty MK, Whitehead C, McCormack H et al. Proteome dynamics in complex organisms: using stable isotopes to monitor individual protein turnover rates. Proteomics.5, 522–533 (2005).
  • Pratt JM, Petty J, Riba-Garcia I et al. Dynamics of protein turnover, a missing dimension in proteomics. Mol. Cell. Proteomics.1, 579–591 (2002).
  • Xiao GG, Garg M, Lim S et al. Determination of protein synthesis in vivo using labeling from deuterated water and analysis of MALDI-TOF spectrum. J. Appl. Physiol.104(3), 828–36 (2008).
  • Zhao Y, Lee WNP, Lim S et al. Quantitative proteomics: measuring protein synthesis using 15N amino acids labeling in pancreas cancer cells. Anal. Chem.81(2), 764–771 (2009).
  • Vogt JA, Schroer K, Holzer K et al. Protein abundance quantification in embryonic stem cells using incomplete metabolic labelling with 15N amino acids, matrix-assisted laser desorption/ionisation time-of-flight mass spectrometry, and analysis of relative isotopologue abundances of peptides. Rapid Commun. Mass Spectrom.17, 1273–1282 (2003).
  • Vogt JA, Hunzinger C, Schroer K et al. Determination of fractional synthesis rates of mouse hepatic proteins via metabolic 13C-labeling, MALDI-TOF MS and analysis of relative isotopologue abundances using average masses. Anal. Chem.77, 2034–2042 (2005).
  • Ong SE, Mann M. Mass spectrometry-based proteomics turns quantitative. Nat. Chem. Biol.1, 252–262 (2005).
  • Liao H, Wu J, Kuhn E et al. Use of mass spectrometry to identify protein biomarkers of disease severity in the synovial fluid and serum of patients with rheumatoid arthritis. Arthritis Rheum.50, 3792–3803 (2004).
  • Anderson NL, Anderson NG, Haines LR 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).
  • Chelius D, Bondarenko PV. Quantitative profiling of proteins in complex mixtures using liquid chromatography and mass spectrometry. J. Proteome Res.1, 317–323 (2002).
  • Zhang B, VerBerkmoes NC, Langston MA et al. Detecting differential and correlated protein expression in label-free shotgun proteomics. J. Proteome Res.5, 2909–2918 (2006).
  • Wang W, Zhou H, Lin H et al. Quantification of proteins and metabolites by mass spectrometry without isotopic labeling or spiked standards. Anal. Chem.75, 4818–4826 (2003).
  • Jaffe JD, Mani DR, Leptos KC et al. PEPPeR, a platform for experimental proteomic pattern recognition. Mol. Cell. Proteomics.5, 1927–1941 (2006).

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.