Proteomics in Systems Biology Methods and Protocols
Proteomics in Systems Biology Methods and Protocols
Proteomics
in Systems
Biology
Methods and Protocols
METHODS IN MOLECULAR BIOLOGY
Series Editor
John M. Walker
School of Life and Medical Sciences
University of Hertfordshire
Hatfield, Hertfordshire, AL10 9AB, UK
Edited by
Jörg Reinders
Institute of Functional Genomics, University of Regensburg, Regensburg, Germany
Editor
Jörg Reinders
Institute of Functional Genomics
University of Regensburg
Regensburg, Germany
Proteomics techniques have constantly been developed further through the last decade and
have been applied successfully for all kinds of samples and biological or medical questions.
Nowadays, they are established methods in many research labs, and proteomic studies can
be accomplished with good reliability and coverage on a routine basis. Therefore,
proteomics can be used as a powerful tool in functional genomics and systems biology
studies. Current challenges are thus the implementation of proteomic analyses in these
comprehensive studies. This applies for both sample generation and preparation to ensure
consistency over several levels of analyses like genomics, transcriptomics, and metabolomics
and integration of the multilevel data to generate biological knowledge. This book gives an
overview of contemporary quantitative proteomics methods and data interpretation
approaches and also gives examples of how to implement proteomics into systems biology.
v
Contents
Preface. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . v
Contributors . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . ix
vii
viii Contents
Index . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 311
Contributors
ix
x Contributors
MANFRED WUHRER • Center for Proteomics and Metabolomics, Leiden University Medical
Center, Leiden, The Netherlands; Division of BioAnalytical Chemistry, VU University
Amsterdam, Amsterdam, The Netherlands
RENÉ P. ZAHEDI • Leibniz-Institut für Analytische Wissenschaften—ISAS—e.V.,
Dortmund, Germany
MAKSIM ZAKHARTSEV • Plant Systems Biology, Plant Physiology, University of Hohenheim,
Stuttgart, Germany
Chapter 1
Abstract
The proteome is the functional entity of the cell, and perturbations of a cellular system almost always cause
changes in the proteome. These changes are a molecular fingerprint, allowing characterization and a
greater understanding of the effect of the perturbation on the cell as a whole. Monitoring these changes
has therefore given great insight into cellular responses to stress and disease states, and analytical platforms
to comprehensively analyze the proteome are thus extremely important tools in biological research. Mass
spectrometry has evolved as the most relevant technology to characterize proteomes in a comprehensive
way. However, due to a lack of throughput capacity of mass spectrometry-based proteomics, researchers
frequently use measurement of mRNA levels to approximate proteome changes. Growing evidence of
substantial differences between mRNA and protein levels as well as recent improvements in mass
spectrometry-based proteomics are heralding an increased use of mass spectrometry for comprehensive
proteome mapping. Here we describe the use of multiplexed quantitative proteomics using isobaric label-
ing with tandem mass tags (TMT) for the simultaneous quantitative analysis of five cancer cell proteomes
in biological duplicates in one mass spectrometry experiment.
1 Introduction
Proteins are the primary functional units of the cell, and as such,
information about their abundance, interaction partners, and mod-
ifications is critical for understanding both healthy and abnormal
cellular function. Traditionally, such work has been accomplished
on a protein-by-protein basis through genetic or biochemical tech-
niques. More recently, large-scale approaches attempting to moni-
tor an entire proteome—all proteins expressed in a cell or tissue—in
one step have become accessible [1, 2]. Such a holistic approach
allows identification of proteome imbalances and changes in func-
tional networks, enabling us to study and probe the state of a cell
in an unbiased and rapid fashion.
Jörg Reinders (ed.), Proteomics in Systems Biology: Methods and Protocols, Methods in Molecular Biology, vol. 1394,
DOI 10.1007/978-1-4939-3341-9_1, © Springer Science+Business Media New York 2016
1
2 Amanda Edwards and Wilhelm Haas
2 Materials
2.1 Cell Culture 1. Cell lines: This protocol is applicable for the proteomic analysis
of any adherent human cell line. Detached cell lines can also be
used, with modifications to the cell culture protocols.
2. Cell media: Use culture media appropriate for the chosen cell lines.
3. 1× sterile phosphate-buffered saline (PBS).
4. 0.25 % Trypsin.
4 Amanda Edwards and Wilhelm Haas
4. Vacuum manifold.
5. SepPak 1 cc (50 mg) C18 Cartridges (Waters).
6. Savant SC100 SpeedVac Concentrator.
7. High-pressure liquid chromatography system (ex: Agilent
1260 Infinity Quaternary LC System).
8. Agilent ZORBAX Extend-C18 column (4.6 mm × 250 mm, 5
μm particle size).
9. Deep-well 96-well plates.
10. Orbitrap Fusion (Thermo Fisher Scientific).
11. Easy-nLC 1000 (Thermo Fisher Scientific).
12. Resins: Magic C4 resin (5 μm, 100 Å, Michrom Bioresources),
Maccel C18AQ resin (3 μm, 200 Å, Nest Group), and GP-C18
(1.8 μm, 120 Å, Sepax Technologies).
3 Methods
3.1 Cell Culture 1. Grow each of the five cell lines to 90 % confluence in duplicate
in 10 cm2 dishes (a total of ten samples).
2. Prior to collecting the cells, wash gently twice with 2.5 mL
pre-warmed sterile 1× PBS (see Note 1).
3. Add 2 mL pre-warmed trypsin to each 10 cm2 dish, covering the
cell layer completely. Incubate for 5 min at 37 °C. Add 3 mL
pre-warmed media to each 10 cm2 dish, and collect each cell
mixture in 15 mL Falcon tubes.
4. Pellet cells by centrifuging at 500 × g for 5 min. Discard the
supernatant. Wash the cell pellet once with sterile 1× PBS (see
Note 2).
3.2 Cell Lysis Note: All subsequent steps should be performed at room temperature,
as the SDS in the lysis buffer will precipitate at cold temperatures.
1. Resuspend each cell pellet in 0.5 mL lysis buffer, pipetting up
and down to disrupt the cell pellet (see Note 3).
2. Lyse the cells by passing the resuspended cells ten times
through a 21-gauge needle (see Note 4). Transfer the suspen-
sion to 1.5 mL Eppendorf tubes.
3. Clear away cellular debris by centrifuging at 16,000 × g for
5 min. Collect the supernatant (see Note 5).
3.4 Digestion 1. Resuspend the protein pellet in 0.5 mL digestion buffer (see
of Proteins Note 12).
2. Add 2.5 μL LysC stock (5 μg) to each pellet. Vortex. Centrifuge
briefly at 3000 × g. Incubate overnight at room temperature,
agitating gently on a tabletop vortexer.
3. Add 12.5 μL trypsin stock (5 μg) to each tube. Vortex.
Centrifuge briefly at 3000 × g. Incubate at 37 °C for 6 h.
4. Acidify the reaction with 25 μL 10 % TFA (final concentration
TFA = 0.5 %). Vortex. Centrifuge at 16,000 × g for 5 min, and
collect the supernatant (see Note 13).
3.7 TMT Labeling 1. Resuspend the TMT reagent according to the manufacturer’s
of Peptides instructions in anhydrous acetonitrile (see Note 20).
2. Resuspend the peptides in 50 μL 200 mM HEPES (pH 8.5),
30 % anhydrous ACN (see Note 21). Vortex. Centrifuge briefly
at 3000 × g. Sonicate for 5 min.
3. Add 5 μL of TMT reagent to each peptide solution, with 1
TMT label used for each of the ten samples (126, 127n, 127c,
128n, 128c, 129n, 129c, 130n, 130c, and 131).
4. Incubate the reaction mixtures at room temperature for 1 h.
5. Quench the reaction by adding 6 μL of 200 mM HEPES
(pH 8.5), 5 % hydroxylamine. Incubate at room temperature
for 15 min.
6. Acidify the mixture by adding 50 μL of 1 % TFA. Combine all
ten samples into one sample, as they are now all distinctly
labeled.
8 Amanda Edwards and Wilhelm Haas
3.9 Mass The details of the LC-MS2/MS3 methods will depend on the
Spectrometry Analysis instrumentation available. Here, we describe a method using an
Easy-nLC 1000 (Thermo Fisher Scientific) with chilled autosam-
pler and an Orbitrap Fusion mass spectrometer (Thermo Fisher
Scientific)
1. Sample preparation: Resuspend each fraction in 8 μL 5 % ACN,
5 % formic acid and sonicate to ensure full suspension of all
peptides. Inject 3 μL of each sample for chromatographic sepa-
ration and mass spectrometry analysis.
2. Nanospray liquid chromatography method: Separate peptides
over a 100 μm inner diameter microcapillary column, packed
in-house with 0.5 cm of Magic C4 resin, 0.5 cm of Maccell
C18 resin, and 29 cm of GP-C18 resin. Use a 6–25 % gradient
of MS buffer B over 165 min at 300 nL/min to elute the pep-
tides. End the gradient with a 10-min wash with 100 % MS
buffer B to clear all remaining peptides off the column, and
re-equilibrate the column with 9 μL of 100 % MS buffer A to
prepare the column for subsequent runs.
3. Mass spectrometry method: Begin acquisition with a full MS1
spectrum acquired in the Orbitrap, and use synchronous pre-
cursor selection to isolate the ten highest intensity peptides for
MS2 analysis. Following CID fragmentation of these peptides,
perform MS2 scans in the linear ion trap. Once again, use syn-
chronous precursor selection to isolate the ten highest inten-
sity peptides for MultiNotch MS3 analysis [25]. Following
HCD fragmentation of the peptides, perform MS3 scans in the
Orbitrap for maximum sensitivity (see Note 23).
Multiplexed Quantitative Proteomics for High-Throughput Comprehensive Proteome… 9
Fig. 1 An overview of the workflow of a multiplexed quantitative proteomics measurement, from cell culture to
mass spectrometer
3.10 Data Analysis As above, the details of the data analysis will depend on the specific
search algorithms and software used. While we use Sequest [26] to
match peptide spectra to sequences, a variety of other options are
available (e.g., Mascot, X!Tandem). However, some parameters
should be universally applied.
1. Specific search parameters include digestion enzyme, static
peptide modifications, variable peptide modifications, and pre-
cursor ion tolerance. In this case, select trypsin as the enzyme,
10 Amanda Edwards and Wilhelm Haas
4 Notes
References
1. Kim M, Pinto S, Getnet D et al (2014) A draft 8. Wu Y, Williams E, Dubuis S et al (2014)
map of the human proteome. Nature Multilayered genetic and omics dissection of
509:575–581 mitochondrial activity in a mouse reference
2. Wilhelm M, Schlegl J, Hahne H et al (2014) population. Cell 158:1415–1430
Mass-spectrometry-based draft of the human 9. Zhang B, Wang J, Wang X et al (2014)
proteome. Nature 509:582–587 Proteogenomic characterization of human
3. Wang Z, Gerstein M, Snyder M (2009) RNA- colon and rectal cancer. Nature 513:382–387
Seq: a revolutionary tool for transcriptomics. 10. Gygi S, Rist B, Gerber S et al (1999)
Nat Rev Genet 10:57–63 Quantitative analysis of complex protein mix-
4. Li G, Burkhardt D, Gross C et al (2014) tures using isotope-coded affinity tags. Nat
Quantifying absolute protein synthesis rates Biotechnol 17:994–999
reveals principles underlying allocation of cel- 11. Ong S, Blagoev B, Kratchmarova I et al (2002)
lular resources. Cell 157:624–635 Stable isotope labeling by amino acids in cell
5. Torres E, Dephoure N, Panneerselvam A et al culture, SILAC, as a simple and accurate
(2010) Identification of aneuploidy-tolerating approach to expression proteomics. Mol Cell
mutations. Cell 143:71–83 Proteomics 1:376–386
6. Stingele S, Stoehr G, Peplowska K et al (2012) 12. Syka J, Marto J, Bai D et al (2004) Novel linear
Global analysis of genome, transcriptome and quadrupole ion trap/FT mass spectrometer:
proteome reveals the response to aneuploidy in performance characterization and use in the
human cells. Mol Syst Biol 8:608 comparative analysis of histone H3 post-
7. Dephoure N, Hwang S, O’Sullivan C et al translational modification. J Proteome Res
(2014) Quantitative proteomic analysis reveals 3:621–626
posttranslational responses to aneuploidy in 13. Olsen J, de Godoy L, Li G et al (2005) Parts
yeast. Elife 3, e03023 per million mass accuracy on an Orbitrap mass
Multiplexed Quantitative Proteomics for High-Throughput Comprehensive Proteome… 13
spectrometer via lock mass injection into a investigate host-pathogen interaction. Cell
C-trap. Mol Cell Proteomics 12:2010–2021 157:1460–1472
14. Hsu J, Huang S, Chow N et al (2003) Stable- 23. Wenger C, Lee M, Hebert A et al (2011)
isotope dimethyl labeling for quantitative pro- Gas-phase purification enables accurate, multi-
teomics. Anal Chem 75:6843–6852 plexed proteome quantification with isobaric
15. Wilson-Grady J, Haas W, Gygi S (2013) tagging. Nat Methods 8:933–935
Quantitative comparison of the fasted and re- 24. Ting L, Rad R, Gygi S et al (2011) MS3 elimi-
fed mouse liver phosphoproteomes using lower nates ratio distortion in isobaric multiplexed
pH reductive dimethylation. Methods 61: quantitative proteomics. Nat Methods 8:
277–286 937–940
16. Wu Y, Wang F, Liu Z et al (2014) Five-plex 25. McAlister G, Nusinow D, Jedrychowski M
isotope dimethyl labeling for quantitative et al (2014) MultiNotch MS3 enables accurate,
proteomics. Chem Commun (Camb) 50: sensitive, and multiplexed detection of differ-
1708–1710 ential expression across cancer cell line pro-
17. Blagoev B, Ong S, Kratchmarova I et al (2004) teomes. Anal Chem 86:7150–7158
Temporal analysis of phosphotyrosine- 26. Eng J, McCormack A, Yates J (1994) An
dependent signaling networks by quantitative approach to correlate tandem mass spectral
proteomics. Nat Biotechnol 22:1139–1145 data of peptides with amino acid sequences in a
18. Thompson A, Schäfer J, Kuhn K et al (2003) protein database. J Am Soc Mass Spectrom
Tandem mass tags: a novel quantification strat- 5:976–989
egy for comparative analysis of complex pro- 27. Peng J, Elias J, Thoreen C et al (2003)
tein mixtures by MS/MS. Anal Chem 75: Evaluation of multidimensional chromatogra-
1895–1904 phy coupled with tandem mass spectrometry
19. Ross P, Huang Y, Marchese J et al (2004) (LC/LC-MS/MS) for large-scale protein anal-
Multiplexed protein quantitation in ysis: the yeast proteome. J Proteome Res
Saccharomyces cerevisiae using amine-reactive 1:43–50
isobaric tagging reagents. Mol Cell Proteomics 28. Elias J, Gygi S (2007) Target-decoy search
3:1154–1169 strategy for increased confidence in large-scale
20. Choe L, D’Ascenzo M, Relkin N et al (2007) protein identifications by mass spectrometry.
8‐Plex quantitation of changes in cerebrospi- Nat Methods 4:207–214
nal fluid protein expression in subjects under- 29. Elias J, Gygi S (2009) Target-decoy search
going intravenous immunoglobulin treatment strategy for mass spectrometry-based pro-
for Alzheimer’s disease. Proteomics teomics. Methods Mol Biol 604:55–71
7:3651–3660 30. Huttlin E, Jedrychowski M, Elias J et al (2010) A
21. McAlister G, Huttlin E, Haas W et al (2012) tissue-specific atlas of mouse protein phosphoryla-
Increasing the multiplexing capacity of TMTs tion and expression. Cell 143:1174–1189
using reporter ion isotopologues with isobaric 31. Wühr M, Haas W, McAlister G et al (2012)
masses. Anal Chem 84:7469–7478 Accurate multiplexed proteomics at the MS2
22. Weekes M, Tomasec P, Huttlin E et al (2014) level using the complement reporter ion cluster.
Quantitative temporal viromics: an approach to Anal Chem 84:9214–9221
Chapter 2
Abstract
Among a variety of global quantification strategies utilized in mass spectrometry (MS)-based proteomics,
isobaric tags for relative and absolute quantitation (iTRAQ) are an attractive option for examining the rela-
tive amounts of proteins in different samples. The inherent complexity of mammalian proteomes and the
diversity of protein physicochemical properties mean that complete proteome coverage is still unlikely
from a single analytical method. Numerous options exist for reducing protein sample complexity and
resolving digested peptides prior to MS analysis. Indeed, the reliability and efficiency of protein identifica-
tion and quantitation from an iTRAQ workflow strongly depend on sample preparation upstream of
MS. Here we describe our methods for: (1) total protein extraction from immortalized cells; (2) subcel-
lular fractionation of murine tissue; (3) protein sample desalting, digestion, and iTRAQ labeling; (4)
peptide separation by strong cation-exchange high-performance liquid chromatography; and (5) peptide
separation by isoelectric focusing.
1 Introduction
Jörg Reinders (ed.), Proteomics in Systems Biology: Methods and Protocols, Methods in Molecular Biology, vol. 1394,
DOI 10.1007/978-1-4939-3341-9_2, © Springer Science+Business Media New York 2016
15
16 Christos Spanos and J. Bernadette Moore
2 Materials
2.3 Protein 1. 2 ml ZEBA columns (Pierce Scientific, UK; see Note 6).
Desalting, Digestion, 2. RIPA buffer and PI as before.
and iTRAQ Labeling
3. Vacuum centrifuge (Eppendorf, UK).
4. Low-bind microcentrifuge tubes (see Note 7).
18 Christos Spanos and J. Bernadette Moore
2.4 Peptide 1. HPLC instrument (Hewlett Packard 1100 series) with autos-
Separation by Strong ampler (Agilent Technologies 1200).
Cation-Exchange HPLC 2. Polysulfoethyl A chromatography column (100 × 94 2.1
mm—300 Å; Hichrom Ltd, UK).
3. Guard cartridge (Hichrom Ltd, UK; see Note 9).
4. Buffer A: 10 mM KH2PO4 (pH 2.75), 25 % acetonitrile
(see Note 10).
5. Buffer B: 10 mM KH2PO4, 1 M KCl (pH 2.75), 25 % acetonitrile
(see Note 10).
6. Vacuum centrifuge (Eppendorf, UK).
2.6 Sample Cleanup 1. 0.1 % (v/v) trifluoroacetic acid (TFA; Sigma Aldrich, UK) in
Prior to Mass HPLC-grade H2O.
Spectrometry 2. ZipTips C18 with 0.6 μl bed of chromatography media
(Millipore, USA).
3. “Wet solution”: 25 % acetonitrile in HPLC-grade H2O.
4. “Equilibration and wash solution”: 0.1 % TFA in HPLC-
grade H2O.
5. “Elution solution”: 50 % acetonitrile/0.1 % TFA in HPLC-
grade H2O.
3 Methods
3.1 Total Protein 1. Add protease inhibitor cocktail to RIPA buffer to a final con-
Extraction centration of 1.5×; keep on ice. Bring a microcentrifuge for the
from Hepatocellular 10,000 × g spin to 4 °C.
Carcinoma Cell Line 2. Following your preferred cell treatment, detach cells from flask
surface using trypsin and centrifuge cell suspension for 5 min
at 200 × g RT.
Sample Preparation for iTRAQ-based Proteomics 19
3. Wash cell pellets with 1× PBS and centrifuge again for 5 min at
1200 × g.
4. Resuspend cell pellet in 350 μl RIPA buffer containing PI and
leave on ice for 10 min.
5. Vortex lysed cells, then place on QIAshredder column, and
centrifuge for 2 min at 10,000 × g.
6. Measure protein concentration of resulting eluate using
BCA assay.
3.3 Protein 1. Remove storage solution from 2 ml ZEBA columns (see Note 6)
Desalting/Buffer by centrifuging at 2000 × g for 2 min. For tissue samples use
Exchange, Digestion, RIPA for buffer exchange; for cells use ddH2O for desalting.
and iTRAQ Labeling Add 1 ml RIPA buffer with PI or ddH2O to column resin and
centrifuge for 2 min at 2000 × g; repeat three times. Then add
samples to column resin and centrifuge for 2 min at 2000 × g.
Process BSA control alongside protein samples up until iTRAQ
labeling (step 9).
20 Christos Spanos and J. Bernadette Moore
3.4 Peptide 1. The dried, digested, and iTRAQ-labeled sample (one tube)
Separation by Strong was resuspended in 100 μl of buffer A.
Cation-Exchange HPLC 2. Set up guard cartridge (see Note 9) before HPLC column.
3. Condition the HPLC column by introducing buffer A (see
Note 10) for 30 min at a flow rate of 1.5 ml/min.
4. Use digested BSA sample to check both the efficiency of the
tryptic digestion and the separation efficiency of the column
using the stepped gradient (Table 1).
Sample Preparation for iTRAQ-based Proteomics 21
Table 1
The stepped gradient that was followed during the SCX approach. The
pressure was set to 400 bar
(continued)
22 Christos Spanos and J. Bernadette Moore
Table 1
(continued)
4 Notes
Acknowledgements
References
1. Moore JB, Weeks ME (2011) Proteomics and 6. Nagaraj N, Kulak NA, Cox J et al (2012)
systems biology: current and future applications System-wide perturbation analysis with nearly
in the nutritional sciences. Adv Nutr 2:355–364 complete coverage of the yeast proteome by
2. Evans C, Noirel J, Ow SY et al (2012) An insight single-shot ultra HPLC runs on a bench top
into iTRAQ: where do we stand now? Anal Orbitrap. Mol Cell Proteomics 11:M111.
Bioanal Chem 404:1011–1027 013722
3. Christoforou AL, Lilley KS (2012) Isobaric tag- 7. Mann M, Kulak NA, Nagaraj N et al (2013) The
ging approaches in quantitative proteomics: the coming age of complete, accurate, and ubiqui-
ups and downs. Anal Bioanal Chem tous proteomes. Mol Cell 49:583–590
404:1029–1037 8. Kruger T, Lehmann T, Rhode H (2013) Effect
4. Ow SY, Salim M, Noirel J et al (2009) iTRAQ of quality characteristics of single sample prepa-
underestimation in simple and complex mixtures: ration steps in the precision and coverage of pro-
“the good, the bad and the ugly”. J Proteome teomic studies—a review. Anal Chim Acta
Res 8:5347–5355 776:1–10
5. Karp NA, Huber W, Sadowski PG et al (2010) 9. Vandermarliere E, Mueller M, Martens L (2013)
Addressing accuracy and precision issues in Getting intimate with trypsin, the leading prote-
iTRAQ quantitation. Mol Cell Proteomics ase in proteomics. Mass Spectrom Rev
9:1885–1897 32:453–465
Chapter 3
Abstract
Altered and abnormal levels of proteins and their phosphorylation states are associated with many disorders.
Detection and quantification of such perturbations may provide a better understanding of pathological
conditions and help finding candidates for treatment or biomarkers. Over the years, isobaric mass tags for
relative quantification of proteins and protein phosphorylation by mass spectrometry have become increas-
ingly popular. One of the most commonly used isobaric chemical tags is iTRAQ (isobaric tag for relative
and absolute quantitation). In a typical iTRAQ-8plex experiment, a multiplexed sample amounts for up to
800 μg of peptides. Using state-of-the-art LC-MS approaches, only a fraction (~5 %) of such a sample is
required to generate comprehensive quantitative data on the global proteome level, so that the bulk of the
sample can be simultaneously used for quantitative phosphoproteomics. Here, we provide a simple and
straightforward protocol to perform quantitative analyses of both proteome and phosphoproteome from
the same sample using iTRAQ.
1 Introduction
Jörg Reinders (ed.), Proteomics in Systems Biology: Methods and Protocols, Methods in Molecular Biology, vol. 1394,
DOI 10.1007/978-1-4939-3341-9_3, © Springer Science+Business Media New York 2016
25
26 Fiorella A. Solari et al.
2 Materials
2.1 Cell Lysis 1. Biological sample obtained from primary cells or tissue, e.g.,
of Biological Samples 100 μg of fibroblasts.
2. Lysis buffer: 50 mM Tris–HCl (pH 7.8, adjust with HCl), 150
mM sodium chloride (NaCl) containing 1 % (w/v) sodium
dodecyl sulfate (SDS). Add one tablet of phosphatase inhibitor
cocktail phosSTOP (Roche Diagnostics) to 10 mL of buffer.
3. Benzonase (Novagen) and 1 M magnesium chloride (MgCl2)
solution.
4. Refrigerated benchtop centrifuge (Eppendorf) and a vortex mixer.
5. Determination of protein concentration: Bicinchoninic acid
(BCA) protein assay.
2.3 Digestion Control 1. HPLC: UltiMate 3000 rapid separation liquid chromatography
by Monolithic (RSLC) (Thermo Fisher Scientific) or similar HPLC system.
Reversed-Phase 2. HPLC column: PepSwift monolithic trap column 200
Chromatography μm × 5 mm and PepSwift monolithic capillary column 200
μm × 5 cm (both Thermo Fisher Scientific).
3. HPLC buffer A: 0.1 % TFA.
4. HPLC buffer B: 0.08 % TFA, 84 % ACN.
2.5 iTRAQ Labeling 1. iTRAQ reagents: 8 plex kit (113–119, 121) from AB SCIEX.
2. Dissolution buffer: 0.5 M triethylammonium bicarbonate
(TEAB), pH 8.5.
3. Reagent dilution solvent: 100 % isopropanol, LC-MS grade.
4. Thermomixer.
2.8 LC-MS/MS 1. HPLC: UltiMate 3000 nano RSLC system (Thermo Fisher
Scientific) with nano-UV cell for quality control or a similar
HPLC system.
2. HPLC column: Acclaim PepMap100 C18 trap column 100
μm × 2 cm, and Acclaim PepMap100 C18 main column 75
μm × 50 cm (both Thermo Fisher Scientific).
3. HPLC loading buffer: 0.1 % TFA.
4. HPLC solvent A: 0.1 % FA.
5. HPLC solvent B: 0.1 % FA, 84 % ACN.
6. Mass spectrometer: Q-Exactive mass spectrometer (Thermo
Fisher Scientific) or other MS systems that can provide high
mass accuracy and high resolution for both MS and MS/MS
scans, as well as beam-type CID fragmentation (or HCD in
Thermo instruments).
2.9 Data Analysis 1. Data analysis software: Proteome Discoverer (version 1.4,
Thermo Scientific) using the following nodes.
(a) Search algorithms: Mascot [14] (version 2.4.1, Matrix
Science), Sequest [15], and MS Amanda [16].
(b) Quantification: Reporter ion quantifier.
(c) Phosphorylation site assignment: PhosphoRS [17].
(d) False discovery rate (FDR) estimator: Peptide validator.
3 Methods
Make sure that you treat all samples completely the same way during
all mentioned steps until labeling, in order to keep the technical
variation as low as possible. Any differences during sample treatment
may affect the quantitative results.
Two Birds with One Stone: Parallel Quantification of Proteome and Phosphoproteome… 29
3.1 Cell Lysis 1. The volume of the lysis buffer depends on the number of cells.
and Determination Typically, 100 μL of buffer is required to solubilize for, e.g.,
of Protein 106 HeLa cells. Promote homogenization by mechanical/
Concentration shearing forces, induced by pipetting and ultrasonication.
2. To hydrolyze the DNA, add 3 μL of benzonase per 200 μL of
cell lysate plus 2 mM MgCl2 and incubate at 37 °C for 30 min.
3. Clarify the lysates by centrifugation. Use a precooled (4 °C)
centrifuge and spin down the sample tubes at 18,000 rcf for
30 min.
4. Collect the supernatant in a new LoBind Eppendorf tube and
determine the protein concentration using BCA assay accord-
ing to the manufacturer’s instructions. See Note 3.
3.4 SPEC Sample 1. To improve the reproducibility of this step, use a vacuum
Cleanup (Desalting) manifold system for peptide desalting. Use C18 AR 4 mg
material (Agilent) tips.
(a) Activation: Three times with 100 μL of 100 % ACN.
(b) Equilibration: Three times with 100 μL of 0.1 % TFA.
(c) Sample loading: Place the sample on the material and
reload the flow through three times.
(d) Washing: Three times with 100 μL of 0.1 % TFA.
(e) Peptides elution: Two times with 100 μL of 60 % ACN in
0.1 % TFA
2. To control the reproducibility of the desalting procedure, take
a small but equal volume (e.g., 2 μL) of eluate from each sam-
ple and dry it completely in the SpeedVac. Snap freeze the
remaining volume using liquid nitrogen and store the frozen
eluates at −80 °C until further use. See Note 8.
3. Measure the samples (2 μL) using nano-LC with UV detection
(214 nm) or LC-MS. If they look reproducible, take 100 μg
aliquots from each sample, dry them in the SpeedVac, and
proceed with iTRAQ labeling.
3.5 iTRAQ Labeling 1. Resolubilize each peptide pellet (~100 μg) in 30 μL of dissolution
buffer (0.5 M TEAB, pH 8.5).
2. Perform the labeling reaction as per the manufacturer’s instruc-
tions (iTRAQ-8plex, AB SCIEX).
3. After incubation, combine all the eight differentially labeled
samples in a new 1.5 mL LoBind Eppendorf tube.
4. Divide the multiplexed sample into two parts. Use one part
corresponding to ~5 % (~40 μg) for complete (global)
proteome analysis and the second part (~95 % or 760 μg) for
the enrichment of phosphopeptides.
5. Dry both parts completely in the SpeedVac and store the pellets
at −80 °C until further use.
Two Birds with One Stone: Parallel Quantification of Proteome and Phosphoproteome… 31
Fig. 1 Pre-fractionation of an iTRAQ-labeled sample on a high-pH C18 reversed-phase (RP) system. The pep-
tides are separated using a binary gradient (buffer A: 10 mM NH4HCO2, buffer B: 10 mM, NH4HCO2, 84 % ACN,
both pH 8.0) ranging from 3 to 50 % buffer B in 65 min. In total, 16 fractions are collected at 1-min intervals
using a concatenation approach. Separation of the peptides on high-pH RP columns and subsequent concat-
enation of the eluted fractions reduce sample complexity, improve selectivity, and increase proteome coverage
[23]. See Note 10
32 Fiorella A. Solari et al.
Fig. 2 Serial enrichment of phosphopeptides from an iTRAQ-labeled sample using TiO2 beads. It is recom-
mendable to perform a second round of enrichment (from Step 2) to enhance the phosphoproteome coverage.
The supernatants (1–3), which contain mostly non-phosphorylated peptides, can be combined and used for,
e.g., global proteome analysis. See Note 9
Two Birds with One Stone: Parallel Quantification of Proteome and Phosphoproteome… 33
3.8 SPE Cleanup 1. Prepare a C18 stage tip following the instructions from
of the step 17, but use C18 material instead of C8 material.
Phosphopeptide- 2. Weigh in 5 mg of Oligo R3 material and add 200 μL of 70 %
Enriched Sample ACN in 0.1 % TFA.
3. Fill the tip with 10 μL of R3 material (the height of the R3
material should be between 2 and 3 mm).
4. For the following steps, use an air-filled syringe to allow the
passage of the liquid through the material.
5. Activate the material with 50 μL of 100 % ACN. Repeat this
step twice.
6. Equilibrate the material with 50 μL of 0.1 % TFA. Repeat this
step twice.
7. Load the acidified phosphopeptides (from step 23 of
Subheading 3.8) onto the stage tip. Reload the flow-through
once more.
8. Wash the material with 50 μL of 0.1 % TFA. Repeat this step
twice.
9. Finally, elute the phosphopeptides from the material with 50
μL of 98 % ACN in 0.1 % TFA, in an HPLC vial, and directly
proceed with HILIC fractionation.
Fig. 3 An off-line-HILIC fractionation of phosphopeptides. The peptides (previously enriched using TiO2 beads)
are separated using a binary gradient (solvent A: 0.1 % TFA, 98 % ACN, solvent B: 0.1 % TFA) ranging from 15
to 40 % of solvent B in 37.5 min. HILIC fractionation of a pre-enriched sample improves the specificity and
increases the phosphoproteome coverage [26]
Fig. 4 MS/MS spectrum of the phosphopeptide iTRAQ-QPGLRQPsPSHDGSLSPLQDR (s = phosphorylated serine) acquired at 17,500 resolution in the Orbitrap mass
analyzer. The low-mass region contains the iTRAQ 8plex reporter ions that represent the relative abundances of the same peptide in eight different biological
samples
Two Birds with One Stone: Parallel Quantification of Proteome and Phosphoproteome… 37
3.11 Data Analysis 1. Search the mass spectrometry (MS) raw data against a human
Uniprot database (see Note 11) with Proteome Discoverer
(PD) software. Use a mere “target” database as PD generates
random decoy hits on the fly.
2. In PD, use the spectrum selector node for precursor ion selec-
tion to process MS raw data with default settings.
3. Use the reporter ion quantifier node and select iTRAQ-8plex
as the quantification method. See Note 12.
4. To maximize the number of peptide spectrum matches (PSMs),
include different search algorithms (Mascot, SEQUEST, and
MS Amanda) using the same set of parameters, i.e., precursor
and fragment ion tolerances of 10 ppm and 0.02 Da for MS
and MS/MS, respectively; trypsin as enzyme with a maximum
of two missed cleavages; carbamidomethylation of Cys,
iTRAQ-8plex on N-terminus, and Lys as fixed modifications;
oxidation of Met, and phosphorylation (only for the phospho-
proteome data) of Ser, Thr, and Tyr as variable modifications.
5. Use the peptide validator node to filter the data with a false
discovery rate (FDR) of 1 %.
6. For the phosphoproteome data analysis, add the PhosphoRS
[17] (version 3.1) node to score localization probabilities for
the identified phosphorylation sites.
2. Phosphoproteome:
(a) Export the phosphopeptide PSM list from PD to Microsoft
Excel. See Note 13.
(b) Select only unique phospho-PSMs that were used for
quantification.
(c) Calculate the normalization factor as described previously
(from Subheading 3.12) and normalize each iTRAQ channel
(see Subheading 3.12, steps 4(a)–(d)).
(d) With the help of ready-to-use Excel macro provided
by Mechtler lab (http://ms.imp.ac.at/?goto=phosphors),
determine the confident phosphorylation sites for each
peptide, as well as the position of the phosphorylation
within the protein sequence. (The analysis should be done
in the same Excel worksheet.)
(e) In Excel concatenate: (1) peptide sequence, (2) protein
accession, and (3) phospho-RS phosphorylation site
(considering only those with probabilities ≥95 %) to define
distinguishable phosphopeptide PSMs. See Note 14.
(f) Sort the data according to the concatenated row to group
phospho-PSMs that belong together. For those, determine
the median normalized abundance values per iTRAQ
channel, as well as the relative standard deviation.
(g) If the experiment consists of replicates, determine student’s
t-test and significantly change phosphopeptides as done
for the global proteome.
4 Notes
References
1. Macek B, Mann M, Olsen JV (2009) Global protein phosphorylation. J Am Soc Mass
and site-specific quantitative phosphopro- Spectrom 18(11):1932–1944
teomics: principles and applications. Annu Rev 7. Beck F, Geiger J, Gambaryan S et al (2013)
Pharmacol Toxicol 49(1):199–221 Time-resolved characterization of cAMP/
2. Batalha IL, Lowe CR, Roque ACA (2012) PKA-dependent signaling reveals that platelet
Platforms for enrichment of phosphorylated inhibition is a concerted process involving
proteins and peptides in proteomics. Trends multiple signaling pathways. Blood 123(5):
Biotechnol 30(2):100–110 e1–e10
3. Boja ES, Phillips D, French SA et al (2009) 8. Wu Y-B, Dai J, Yang XL et al (2009) Concurrent
Quantitative mitochondrial phosphoproteomics quantification of proteome and phosphopro-
using iTRAQ on an LTQ-Orbitrap with high teome to reveal system-wide association of
energy collision dissociation. J Proteome Res protein phosphorylation and gene expression.
8(10):4665–4675 Mol Cell Proteomics 8(12):2809–2826
4. Jones AE, Nühse T (2011) Phosphoproteomics 9. Ross PL, Huang YN, Marchese JN et al (2004)
using iTRAQ. In: Dissmeyer N, Schnittger A Multiplexed protein quantitation in
(eds) Plant kinases. Humana Press, New York, Saccharomyces cerevisiae using amine-reactive
pp 287–302 isobaric tagging reagents. Mol Cell Proteomics
5. Mertins P, Udeahi ND, Clauser KR et al 3(12):1154–1169
(2012) iTRAQ labeling is superior to mTRAQ 10. Gilar M, Olivova P, Daly AE et al (2005) Two-
for quantitative global proteomics and phos- dimensional separation of peptides using
phoproteomics. Mol Cell Proteomics RP-RP-HPLC system with different pH in first
11(6):M111.014423 and second separation dimensions. J Sep Sci
6. Liang X, Fonnum G, Hajivandi M et al (2007) 28(14):1694–1703
Quantitative comparison of IMAC and TiO2 11. Thingholm TE, Jorgensen TJ, Jensen ON et al
surfaces used in the study of regulated, dynamic (2006) Highly selective enrichment of phos-
Two Birds with One Stone: Parallel Quantification of Proteome and Phosphoproteome… 41
phorylated peptides using titanium dioxide. 21. Batth TS, Francavilla C, Olsen JV (2014)
Nat Protoc 1(4):1929–1935 Off-line high-pH reversed-phase fractionation
12. Alpert AJ (1990) Hydrophilic-interaction for in-depth phosphoproteomics. J Proteome
chromatography for the separation of peptides, Res 13(12):6176–6186
nucleic acids and other polar compounds. 22. Castillo A, Roig-Navarro AF, Pozo OJ (2007)
J Chromatogr 499:177–196 Secondary interactions, an unexpected problem
13. Thingholm TE, Larsen MR (2009) The use of emerged between hydroxyl containing analytes
titanium dioxide micro-columns to selectively and fused silica capillaries in anion-exchange
isolate phosphopeptides from proteolytic micro-liquid chromatography. J Chromatogr A
digests. Methods Mol Biol 527:57–66 1172(2):179–185
14. Perkins DN, Pappin DJ, Creasy DM et al 23. Yang F, Shen Y, Camp DG et al (2012) High
(1994) Probability-based protein identification pH reversed-phase chromatography with
by searching sequence databases using mass fraction concatenation for two-dimensional
spectrometry data. Electrophoresis 20(18): proteomic analysis. Expert Rev Proteomics
3551–3567 9(2):129–134
15. Geer LY, Markey SP, Kowalak JA et al (2004) 24. Larsen MR, Thingholm TE, Jensen ON et al
Open mass spectrometry search algorithm. (2005) Highly selective enrichment of phos-
J Proteome Res 3(5):958–964 phorylated peptides from peptide mixtures
16. Eng JK, McCormack AL, Yates JR (1994) An using titanium dioxide microcolumns. Mol
approach to correlate tandem mass spectral Cell Proteomics 4(7):873–886
data of peptides with amino acid sequences in a 25. Dickhut C, Radau S, Zahedi RP (2014)
protein database. J Am Soc Mass Spectrom Fast, efficient, and quality-controlled phos-
5(11):976–989 phopeptide enrichment from minute sample
17. Taus T, Kocher T, Pichler P et al (2011) amounts using titanium dioxide. In: Martins-
Universal and confident phosphorylation site de-Souza D (ed) Shotgun proteomics.
localization using phosphoRS. J Proteome Res Springer, New York, pp 417–430
10(12):5354–5362 26. Engholm-Keller K, Birck P, Storling J et al
18. Dickhut C, Feldmann I, Lambert J et al (2014) (2012) TiSH—a robust and sensitive global
Impact of digestion conditions on phosphopro- phosphoproteomics strategy employing a
teomics. J Proteome Res 13(6):2761–2770 combination of TiO2, SIMAC, and HILIC.
J Proteomics 75(18):5749–5761
19. Burkhart JM, Schumbrutzki C, Wortelkamp S
et al (2012) Systematic and quantitative com- 27. Wisniewski JR, Zougman A, Nagaraj N et al
parison of digest efficiency and specificity (2009) Universal sample preparation method
reveals the impact of trypsin quality on for proteome analysis. Nat Methods 6(5):
MS-based proteomics. J Proteomics 75(4): 359–362
1454–1462 28. Manza LL, Stamer SL, Ham AJ et al (2005)
20. Toll H, Oberacher H, Swart R et al (2005) Sample preparation and digestion for pro-
Separation, detection, and identification of teomic analyses using spin filters. Proteomics
peptides by ion-pair reversed-phase high- 5(7):1742–1745
performance liquid chromatography-electrospray 29. Kollipara L, Zahedi RP (2013) Protein car-
ionization mass spectrometry at high and low bamylation: in vivo modification or in vitro
pH. J Chromatogr A 1079(1–2):274–286 artefact? Proteomics 13(6):941–944
Chapter 4
Abstract
Biology and especially systems biology projects increasingly require the capability to detect and quantify
specific sets of proteins across multiple samples, for example the components of a biological pathway
through a set of perturbation-response experiments. Targeted proteomics based on selected reaction mon-
itoring (SRM) has emerged as an ideal tool to this purpose, and complements the discovery capabilities of
shotgun proteomics methods. SRM experiments rely on the development of specific, quantitative mass
spectrometric assays for each protein of interest and their application to the quantification of the protein
set in various biological samples. SRM measurements are multiplexed, namely, multiple proteins can be
quantified simultaneously, and are characterized by a high reproducibility and a broad dynamic range. We
provide here a practical guide to the development of SRM assays targeting a set of proteins of interest and
to their application to complex biological samples.
Key words Selected reaction monitoring, Targeted proteomics, Protein quantitation, Assay design,
Assay validation
1 Introduction
Jörg Reinders (ed.), Proteomics in Systems Biology: Methods and Protocols, Methods in Molecular Biology, vol. 1394,
DOI 10.1007/978-1-4939-3341-9_4, © Springer Science+Business Media New York 2016
43
44 Yuehan Feng and Paola Picotti
given peptide are monitored only for a short time interval, centered
around the known elution time of the peptide. This decreases the
number of concurrent SRM transitions measured at a given chro-
matographic time, allowing optimizing cycle time and dwell time
while measuring larger numbers of peptides per MS analysis. Using
this approach, up to 150 proteins or several hundred peptides [3]
can be simultaneously monitored in a 30-min LC-SRM run. Once
developed, SRM assays are stable and applicable to measuring the
target proteins in a variety of samples at high throughput. The
application of SRM in biological and biomedical projects has been
further promoted by a growing number of open-access databases
[4] providing experimentally validated SRM coordinates and by
automated or semiautomated data analysis tools [5, 6].
In the following, we provide a basic protocol for developing
SRM assays and conducting SRM measurements, assuming that a
list of proteins of interest has been defined.
2 Materials
2.2 LC-SRM/MS 1. Buffer A: HPLC-grade water with 0.1 % (v/v) formic acid.
2. Buffer B: HPLC-grade acetonitrile with 0.1 % (v/v) formic
acid.
3. A QqQ spectrometer equipped with a nano-electrospray ion
source and interfaced with a liquid chromatography (LC) system
operating in the nanoliter/min flow rate range.
4. A chromatographic column for nano-LC separation, packed
with C18 resin (20 cm length × 75 μm diameter).
5. Optional
(a) iRT Kit (Biognosys, Switzerland).
46 Yuehan Feng and Paola Picotti
3 Methods
3.1 Sample 1. Depending on the sample type under investigation (e.g., bac-
Preparation teria, yeast, mammalian cells, human tissue, or plasma) differ-
ent protein extraction procedures are used. After protein
extraction, estimate the concentration of the protein solution
using the BCA assay. Typical protein extraction buffers may
contain reagents that interfere with the BCA assay (e.g., DTT)
or with the subsequent MS analysis (e.g., detergents such as
sodium dodecyl sulfate or NP-40) and should thus be omitted
or diluted prior to the BCA assay.
2. The extraction buffer should contain a chaotropic agent at
high concentration, such as 8 M urea, to achieve complete pro-
tein denaturation. If the protein extract derives from a protein
precipitation step (for example using cold acetone), add the
denaturation buffer to the protein pellet to a final protein con-
centration of 2–3 mg/ml.
3. Adjust the pH of the protein extract to ~8.
4. Add the reduction buffer to a final concentration of 5 mM
TCEP–HCl, followed by incubation for 30 min at 37 °C to
reduce disulfide bridges.
5. Add the alkylation buffer to a final IAA concentration of 40
mM, followed by incubation at 25 °C in the dark for 45 min to
alkylate free cysteine residues.
6. Dilute the protein solution with 0.1 M AmBiC to a final con-
centration of 6 M urea to prevent denaturation of the protease
used in the first digestion step.
7. To achieve completeness of the digestion reaction, two proteases
are sequentially applied, Lys-C (cleaving at the C-terminus of
lysine) followed by trypsin (cleaving at the C-terminus of lysine
and arginine). Add first Lys-C to the protein solution to an
enzyme-to-substrate (E:S) ratio of 1:100, and incubate the
reaction mixture at 37 °C for 4 h. Lys-C tolerates harsher con-
ditions (e.g., higher denaturant concentration) compared to
trypsin. Therefore, dilute the proteolytic mixture again prior
to addition of trypsin to a final concentration of 2 M urea.
Add then trypsin to an E:S ratio of 1:100 and incubate the
proteolytic mixture at 37 °C overnight.
8. Stop the tryptic digestion by addition of formic acid to pH <3.
9. Desalt the peptide mixture using Sep-Pak tC18 Vac cartridges,
according to the manufacturer’s instruction, and using acetoni-
trile for the initial wash of the cartridges, 0.1 % formic acid for
cartridge equilibration and peptide desalting, and 60–80 %
acetonitrile for peptide elution. Evaporate the peptide mixture
eluted in 60–80 % acetonitrile in a vacuum centrifuge to dryness.
Selected Reaction Monitoring to Measure Proteins of Interest in Complex Samples… 47
3.2 SRM A protein SRM assay includes SRM transitions for at least one
Assay Design unique peptide generated upon digestion of the target protein, and
ideally comprises transitions for multiple peptides from that pro-
tein to improve reliability in the protein detection and quantifica-
tion steps. An SRM assay for a peptide in turn minimally includes
the mass-to-charge ratio (m/z) of a peptide precursor ion and
those of a set of fragments generated upon collision-induced dis-
sociation of the precursor. The design of protein SRM assays
involves selection of optimal peptide and fragment ion coordinates
and is followed by refinement and validation of the assays, after
which the assays can be applied to the quantification of the target
proteins. The information required to design SRM assays can be
obtained from in silico analyses, databases, or shotgun proteomics
data, as described below (Fig. 1).
In silico design: Ideal peptides for SRM uniquely map to the target
protein and show good ionization and fragmentation proper-
ties, resulting in intense SRM peaks. If the number of target
proteins is limited (for example, below 20), ideal peptides and
their fragments can simply be selected by testing the MS
performance of all unique peptides generated upon tryptic
digestion of each protein [7]. This involves retrieval of the target
Peak Statistical
Validation evaluation analysis
SRM assays
Refinement
Fig. 1 Workflow of SRM-based targeted proteomic analyses. Coordinates of protein SRM assays are generated
by in silico design, retrieved from Web-based repositories or derived from shotgun proteomics experiments.
The assay coordinates are validated using MS/MS spectra filtered to an acceptable FDR or synthetic peptides.
The assay coordinates are measured by SRM for assay refinement and the final SRM transitions are applied to
perform quantitative SRM measurements across the samples of interest. The SRM peak data are scrutinized
using tools such as Skyline and statistically evaluated
48 Yuehan Feng and Paola Picotti
3.3 SRM Assay The coordinates of an SRM assay require validation [2]; that is, it
Validation should be confirmed that the chosen transitions and associated
and Refinement retention times detect the target peptide and not a false-positive
peak, based on an acceptable error rate. SRM assays from databases
or shotgun data are typically already validated by statistically fil-
tered LC-MS/MS spectra or other approaches, and can in theory
be directly applied to protein quantification with the precautions
described above. SRM assays from in silico design and computa-
tional prediction require validation by alternative approaches. One
option is the acquisition of MS/MS spectra for each target peptide
from the biological sample containing the protein of interest, their
assignment to peptide sequences via database search, and statistical
filtering to the chosen error rate. Another simple and cost-efficient
possibility is the (MS/)MS analysis of either crude synthetic peptide
analogues [15] of the targeted peptides (see Note 7) or tryptic
digests of recombinant proteins [16]. These options are particu-
larly useful when the targeted proteins are of low abundance and
thus difficult to detect in the available biological samples or if the
samples of interest are precious (e.g., from patients) and can only
be used in the final quantification step.
For the refinement of SRM assays, the SRM coordinates derived
from Subheading 3.2 are measured in SRM mode using the biologi-
cal samples of interest. The resulting peaks are inspected to select a
minimal set of suitable transitions and associated retention time (for
scheduled SRM measurements) that defines the final quantitative
assay for the protein. The three to five most intense transitions for
the best ionizing peptides are typically chosen in this step. Shouldered
peaks, likely resulting from multiple co-elutingpeptides sharing the
same transition(s), should be discarded. Retention times of SRM
peaks at the peak apex are annotated, if the aim is a scheduled SRM
experiment. Note that the extracted retention times are valid only
if the chromatographic settings remain the same in the quantifica-
tion step (see Note 4).
Once the assays are validated as described above, SRM assay
refinement can be performed with the tool Skyline.
1. First, open the Skyline (.sky) file from which the assays were
derived, to enable matching of the measured transitions to the
associated identifiers.
2. Import the raw SRM data as Results.
3. Inspect the peaks associated to each peptide transition group.
Important criteria to evaluate if a peptide was identified and
where in the chromatogram include (1) co-elution and shape
similarity of all transitions for that peptide; (2) retention time
of the selected peak matching the RT extracted during the
validation step; and (3) relative intensities of fragment ions
matching those extracted from the reference spectral library
Selected Reaction Monitoring to Measure Proteins of Interest in Complex Samples… 51
3.5 Final Validated and refined SRM coordinates constitute the final SRM
Quantitative Analysis assay for a protein. Final SRM assays are directly applied to mea-
sure the target proteins using scheduled or unscheduled SRM
acquisition. The resulting data are manually inspected or processed
automatically using software tools such as the mProphet [6], to
evaluate detection and (relative) amount of the protein in each
sample. Manual data evaluation should include application of the
constraints mentioned above in Subheading 3.3 to minimize the
52 Yuehan Feng and Paola Picotti
4 Notes
Acknowledgements
References
1. Aebersold R, Mann M (2003) Mass 11. Mohammed Y, Domanski D, Jackson AM et al
spectrometry-based proteomics. Nature 422: (2014) PeptidePicker: a scientific workflow
198–207 with web interface for selecting appropriate
2. Picotti P, Aebersold R (2012) Selected reaction peptides for targeted proteomics experiments.
monitoring-based proteomics: workflows, J Proteomics 106:151–161
potential, pitfalls and future directions. Nat 12. Deutsch EW, Lam H, Aebersold R (2008)
Methods 9:555–566 PeptideAtlas: a resource for target selection for
3. Soste M, Hrabakova R, Wanka S et al (2014) A emerging targeted proteomics workflows.
sentinel protein assay for simultaneously quan- EMBO Rep 9:429–434
tifying cellular processes. Nat Methods 13. Whiteaker JR, Halusa GN, Hoofnagle AN et al
11:1045–1048 (2014) CPTAC assay portal: a repository of
4. Kusebauch U, Deutsch EW, Campbell DS et al targeted proteomic assays. Nat Methods 11:
(2014) Using PeptideAtlas, SRMAtlas, and 703–704
PASSEL: comprehensive resources for discov- 14. Sharma V, Eckels J, Taylor GK et al (2014)
ery and targeted proteomics. Curr Protoc Panorama: a targeted proteomics knowledge
Bioinformatics 46:13.25.11–13.25.28 base. J Proteome Res 13:4505–4510
5. MacLean B, Tomazela DM, Shulman N et al 15. Picotti P, Rinner O, Stallmach R et al (2010)
(2010) Skyline: an open source document High-throughput generation of selected
editor for creating and analyzing targeted reaction-monitoring assays for proteins and
proteomics experiments. Bioinformatics 26: proteomes. Nat Methods 7:43–46
966–968 16. Stergachis AB, MacLean B, Lee K et al (2011)
6. Reiter L, Rinner O, Picotti P et al (2011) Rapid empirical discovery of optimal peptides
mProphet: automated data processing and sta- for targeted proteomics. Nat Methods
tistical validation for large-scale SRM experi- 8:1041–1043
ments. Nat Methods 8:430–435 17. Choi M, Chang CY, Clough T et al (2014)
7. Bereman MS, MacLean B, Tomazela DM et al MSstats: an R package for statistical analysis of
(2012) The development of selected reaction quantitative mass spectrometry-based proteomic
monitoring methods for targeted proteomics experiments. Bioinformatics 30:2524–2526
via empirical refinement. Proteomics 18. Escher C, Reiter L, MacLean B et al (2012)
12:1134–1141 Using iRT, a normalized retention time for
8. Mallick P, Schirle M, Chen SS et al (2007) more targeted measurement of peptides.
Computational prediction of proteotypic pep- Proteomics 12:1111–1121
tides for quantitative proteomics. Nat 19. de Graaf EL, Altelaar AF, van Breukelen B et al
Biotechnol 25:125–131 (2011) Improving SRM assay development: a
9. Fusaro VA, Mani DR, Mesirov JP et al (2009) global comparison between triple quadrupole,
Prediction of high-responding peptides for tar- ion trap, and higher energy CID peptide frag-
geted protein assays by mass spectrometry. Nat mentation spectra. J Proteome Res
Biotechnol 27:190–198 10:4334–4341
10. Qeli E, Omasits U, Goetze S et al (2014) 20. Maclean B, Tomazella DM, Abatiello SE et al
Improved prediction of peptide detectability for (2010) Effect of collision energy optimization
targeted proteomics using a rank-based algo- on the measurement of peptides by selected
rithm and organism-specific data. J Proteomics reaction monitoring (SRM) mass spectrome-
108:269–283 try. Anal Chem 82:10116–10124
Chapter 5
Abstract
As cells develop and differentiate, they change in function and morphology, which often precede earlier
changes in signaling and metabolic control. Here we present a selected reaction monitoring (SRM) approach
which allows for the parallel quantification of metabolic regulators and their downstream targets.
In particular we explain and describe how to monitor abundance changes of glycolytic enzymes upon
PPARγ activation by using a label-free or a stable isotope-labeled standard peptide (SIS peptides) approach
applying triple-quadrupole mass spectrometry. We further outline how to fractionate the cell lysate into
cytosolic and nuclear fractions to enhance the sensitivity of the measurements and to investigate the
dynamic concentration changes in those compartments.
Key words Metabolic control, Adipocytes, Selected reaction monitoring, SIS peptides, Quantification,
Mass spectrometry, Proteomics
1 Introduction
In the past adipose tissue was just seen as a storage depot for free
fatty acids. Recent studies and models replaced this with the idea
that adipose tissue is a complex, essential, and highly active meta-
bolic and endocrine organ [1, 2]. Adipocytes also play a central
role in lipid and glucose metabolism and produce a large number
of hormones and cytokines, whose dysregulation is known to be
involved in metabolic syndrome, diabetes mellitus, and vascular
diseases. Therefore it affects the entire set of organ systems in our
body [3, 4].
The increasing rate of obesity in our society has led to intense
interest in understanding the mechanisms underlying the formation
of adipose tissue and its capacity to store fat [5–7]. The nuclear
receptor and lipid-binding protein PPARγ is a master regulator of
the formation and function of mature fat cells [8]. PPARγ is
expressed and activated during adipocyte differentiation, and
artificial induction of PPARγ in cells with adipogenic differentiation
Jörg Reinders (ed.), Proteomics in Systems Biology: Methods and Protocols, Methods in Molecular Biology, vol. 1394,
DOI 10.1007/978-1-4939-3341-9_5, © Springer Science+Business Media New York 2016
57
58 Andreas Hentschel and Robert Ahrends
Fig. 1 Selected reaction monitoring (SRM) total ion chromatogram of peptides derived from glycolysis proteins
out of an OP9 cytosolic protein extraction. The total ion chromatogram showing measurements of 50 proteins
(150 peptides and 900 transitions total) during an 80-min experiment on a single sample consisting of 1 μg of
total cytosolic protein digest. The inset panels display chromatograms of individual monitored peptides derived
from 1 to 8: L-lactate dehydrogenase A, tubulin beta 5 (control), glyceraldehyde-3-phosphate dehydrogenase,
fructose-bisphosphate aldolase A, pyruvate kinase, triosephosphate isomerase, phosphoglycerate mutase and
alpha-enolase
112 nM, and 250 nM). (b) Transitions of the control protein tubulin beta 5 derived from same sample and analysis. The SIS peptides for absolute quantification were
added in an amount of 500 fmol each, whereby every sample contained 1 μg of total cytosolic protein digest
61
62 Andreas Hentschel and Robert Ahrends
2 Materials
2.1 Cell Culture 1. Cells: OP9, a stromal cell line from mouse bone marrow, was
of OP9 Cells purchased from the Tokyo Metropolitan Institute of Medical
Science.
2. High-serum culture media: MEMα containing 20 % fetal
bovine serum (FBS) (Biochrome), 1 % penicillin/streptomycin
(PSG) (GIBCO).
3. Low-serum culture media: MEMα containing 10 % fetal bovine
serum (FBS) (Biochrome), 1 % penicillin/streptomycin (PSG)
(GIBCO).
4. Cell culture dishes: Cell culture flasks (T75) (Sarstedt), dispos-
able filter units, pore size 0.2 μm (Thermo) sterile serological
pipettes 5, 10, 25 ml (VWR).
5. Rosiglitazone stock solution: 1 mg/ml Rosiglitazone in
DMSO (Sigma), store at −80 °C.
6. Differentiation: Low-serum media containing 1 μM
rosiglitazone.
2.4 Peptide Cleanup 1. Buffer A: 0.05 % Trifluoroacetic acid (TFA) (pH 3.0); can also
Procedure use 5 % acetonitrile, 0.2 % formic acid, but can lose small
peptides.
2. Buffer B: 80 % Acetonitrile (ACN), 0.2 % formic acid (FA)
(pH below 4.0).
3. 100 % Methanol (Biosolve, ULC/MS).
4. Filters: SepPak C18 (100 mg, 1 cc), Waters #WAT023590,
1 mg capacity.
2.5 HPLC/MS 1. Standard peptides: Heavy arginine ([13C6] [15N4])- and lysine
Conditions ([13C6])-labeled peptides can be purchased from multiple ven-
and Materials dors (see Note 6). We use our own synthesized heavy peptides
and store them in 30 % ACN and 0.1 % TFA solution. Combine
all the heavy peptides needed for the experiment to a final con-
centration of 1.6 μM and store the peptide mix, as well as the
remainder of the concentrated individual peptide solutions,
aliquoted at −80 °C.
2. HPLC buffers: A: 0.1 % FA in HPLC-grade water, B: 84 %
ACN in 0.1 % FA.
3. Nano-HPLC instrument: Dionex UltiMate 3000 UHPLC
equipped with an Acclaim PepMap RSLC c18 reversed-phase
main column (Thermo) with 75 μm × 25 cm and 2 μm, 100 Å
particles, and an Acclaim PepMap 100 c18 reversed-phase pre
column (Thermo) with 100 μm × 2 cm and 5 μm, 100 Å
particles.
4. MS instrument: TSQ Vantage (Thermo Fisher Scientific)
triple-quadrupole mass spectrometer.
2.6 Data Analysis 1. Xcalibur 2.2.44, data analysis and instrument control.
Software 2. Skyline 2.5 software suite—available for free from the MacCoss
Lab website: https://brendanx-uw1.gs.washington.edu/lab-
key/project/home/software/Skyline/begin.view.
3. Microsoft Excel.
4. Origin 9.1.
64 Andreas Hentschel and Robert Ahrends
3 Methods
3.1 Cell Culture Each step must be carried out under sterile conditions.
1. Culture OP9 cells in high-serum culture media in cell culture
flasks as needed. Seed them out at 10 % of confluence (1E + 5 in
T15, 5E + 5 in T75, 1E + 6 in T175). Change the media every
2 days. After 4 days the cells are almost confluent (see Note 1).
2. For the experiment prepare 6 × T75 cell culture flasks with 10 %
confluence and incubate them for 3 days (should contain three
to four million cells after this period). Change the media on
third day to low-serum culture media and add rosiglitazone to
three of the flasks (end concentration 0.25 μM). Treat the
other ones just with DMSO with the same concentration.
Incubate all flasks for 48 h at 37 °C with 5 % CO2 in humidi-
fied atmosphere.
3.3 Cell Lysis 1. Resuspend the pellets in 100 μl lysis buffer (buffer A) per million
and Fractionated cells; incubate on ice for 10 min, flicking the tube every minute.
Extraction Can add more lysis buffer if needed.
Monitoring PPARG-Induced Changes in Glycolysis 65
3.4 Protein Digest 1. Centrifuge all fractions at max speed with the centrifuge
(20,000 × g) for 20 min at 4 °C. Carefully remove the superna-
tant leaving the protein pellet intact.
2. Allow the acetone to evaporate from the uncapped tube at
room temperature for 20 min. Do not over-dry pellet, or it
may not dissolve properly.
3. Resuspend the nuclear and cytosolic pellet in 6 M urea to
solubilize (10 μl per million cells at room temperature). Leave
for at least 1 h in urea, vortexing often. If the pellet is not dis-
solving you can use sonication in a water bath for 3–5 min.
4. Dilute samples to less than or equal to 2 M urea using 10 mM
NH4HCO3. Measure protein concentration with BCA kit. Make
a duplicate assay (standard and each sample). 1:5 dilution of each
sample is recommended to stay within the standard range.
66 Andreas Hentschel and Robert Ahrends
3.5 Peptide Cleanup 1. Wash cartridges (SepPak C18) with 3× volumes (1 volume is
Procedure equivalent of 1 ml) of methanol (100 %), fast flow (with vac-
uum applied). This gets rid of contaminants in the cartridge.
Make sure not to run the filter dry. Stop when meniscus is just
above it.
2. Equilibrate with 3× volumes of buffer A, fast flow (with vac-
uum applied).
3. Load sample (pH should be <3), slow flow, best by gravity.
4. Wash and desalt with 3× volumes of buffer A, fast flow (with
vacuum applied).
5. Elute peptides with 1 ml buffer B, slow flow (gravity). Elute the
remaining fluid by vacuum past filter (after gravity elution).
6. Vacuum centrifuge sample to dryness and resuspend in 2 %
acetonitrile and 0.1 % FA (want a concentration of 1 μg/μl as
stock). This can take up to 5 h, depending on the vacuum
centrifuge. Best to use a high-performance solvent-resistant
vacuum centrifuge connected to a vacuum pump of sufficient
power.
3.6 Setting Before starting with the measurements you have to set up the right
Up the SRM Method method containing the proteins and peptides of interest including
in Skyline the transitions for the SIS peptides.
1. First, set up the Skyline document with the following
preferences:
Peptide settings
(a) Digestion: Trypsin [KR|P], missed cleavages 0
(b) Filter: Min length 8, max length 25, exclude N-terminal
AAs 25, you can also exclude all peptides that contain specific
AAs like Cys or Met, as needed
(c) Modifications: Carbamidomethyl (C)
Monitoring PPARG-Induced Changes in Glycolysis 67
Transition settings
(a) Filter: Precursor charges 2 and 3, ion charges 1, ion types y
(b) Product ions: 6 ions
2. After those preparations are done, load a spectral library con-
taining the proteome of the species of interest (see Note 7).
3. Copy the amino acid sequences of each protein from Table 1
and paste it directly into the new Skyline document. The pro-
gram will automatically rank the best transitions for each peptide
according to the loaded spectral library (see Note 8).
4. Export the method (File/Export/Transition list) and choose
multiple methods. For max concurrent transitions choose 100
or 120 and method type: standard.
5. You can now import all the method files into the TSQ Vantage
program to search for all the peptides with their transitions and
get the right retention times.
List of proteins, peptides, and transitions for monitoring PPARγ-induced glycolytic changes. The transition list for PPARγ and other nuclear proteins
can be found in Ahrends et al. (2014, Science, DOI: 10.1126/science.1252079)
y5 646.35 624.36 1
y4 519.30 527.31 1
DAVLNAWAEDVDLR 793.8941++ 798.8982++ y10 1188.56 1198.57 1
y9 1074.52 1084.52 1
69
y8 1003.48 1013.49 1
(continued)
Table 1
70
(continued)
y5 570.36 578.37 1
VTLTPEEEAR 572.7959++ 577.8000++ y8 944.47 954.47 1
y7 831.38 841.39 1
y6 730.34 740.34 1
P17751 Triosephosphate isomerase Tpi1 IAVAAQNCYK 569.2897++ 573.2968++ y8 953.45 961.46 1
y7 854.38 862.39 1
y6 783.35 791.35 1
DLGATWVVLGHSER 770.3993++ 775.4035++ y8 896.49 906.50 1
y7 797.43 807.43 1
y5 585.27 595.28 1
VVLAYEPVWAIGTGK 801.9481++ 805.9552++ y9 928.53 936.53 1
y7 732.40 740.41 1
y6 546.32 554.33 1
O08528 Hexokinase-2 Hk2 LPLGFTFSFPCHQTK 890.4480++ 894.4551++ y8 1004.46 1012.47 1
y6 770.36 778.37 1
y5 673.31 681.32 1
NILIDFTK 482.2791++ 486.2862++ y6 736.42 744.43 1
y5 623.34 631.35 1
y4 510.26 518.27 1
P17710 Hexokinase-1 Hk1 LSDEILIDILTR 700.9034++ 705.9075++ y7 843.53 853.53 1
y6 730.45 740.45 1
y5 617.36 627.37 1
GDFIALDLGGSSFR 727.8673++ 732.8715++ y10 1022.53 1032.53 1
y9 951.49 961.49 1
y6 610.29 620.30 1
SANLVAATLGAILNR 742.4332++ 747.4373++ y11 1098.66 1108.67 1
y10 999.59 1009.60 1
y6 643.39 653.39 1
Q9DBJ1 Phosphoglycerate mutase 1 Pgam1 VLIAAHGNSLR 575.8382++ 580.8423++ y8 825.43 835.44 1
y7 754.3955 764.40 1
y6 683.35 693.36 1
YADLTEDQLPSCESLK 934.9328++ 938.9399++ y10 1176.56 1184.57 1
y8 933.47 941.48 1
y7 820.39 828.40 1
ALPFWNEEIVPQIK 842.4589++ 846.4660++ y9 1069.59 1077.60 1
y5 584.38 592.39 1
y4 485.31 493.32 1
Q9QXG4 Acetyl-coenzyme A Acss2 VAFYWEGNEPGETTK 864.3992++ 868.4063++ y10 1061.47 1069.48 1
synthetase y9 932.43 940.44 1
y6 632.32 640.33 1
AELGMNDSPSQSPPVK 828.8985++ 832.9056++ y11 1155.56 1163.57 1
y9 926.49 934.50 1
y8 839.46 847.47 1
IGPIATPDYIQNAPGLPK 933.0120++ 937.0191++ y12 1312.69 1320.70 1
y7 696.40 704.41 1
y6 582.36 590.37 1
P99024 Tubulin beta-5 chain Tubb5 ISVYYNEATGGK 651.3222++ 655.3293++ y9 1002.45 1010.46 1
y8 839.39 847.40 1
y7 676.33 684.34 1
Monitoring PPARG-Induced Changes in Glycolysis
y6 684.40 692.41 1
y5 571.32 579.33 1
72 Andreas Hentschel and Robert Ahrends
4 Notes
1. Do not allow the cells to reach full confluence until right before
the experiment. It is best to split the cells every 2 days to keep
them between 10 and 70 % confluence. OP9 cells are prone to
spontaneous differentiation if they grow too confluent.
2. At this point you can stain with trypan blue. Pipette approxi-
mately 2–5 μl from your sample onto an object slide, add 1–2 μl
of the trypan blue solution, and mix gently with the pipette.
Check under the microscope for the destroyed cells. If the
mechanical lysis was successful, you should only see blue stained
cell remnants. Everything that is not stained was not lysed.
3. This is a time-critical step because you can lose nuclear proteins
by diffusion if you wait too long to take the supernatant.
4. Do not exceed 500 mM KCl or you will extract histones.
5. For the further steps the histone fraction is not needed.
6. SIS peptides that are used in this protocol were synthesized
in-house without any modifications such as carbamidomethyl-
ation. SIS peptides without those modifications have to be
added to the digest before adding TCEP and IAA treatment.
If it is not possible to synthesize the peptides on your own
Monitoring PPARG-Induced Changes in Glycolysis 73
References
1. Kershaw EE, Flier JS (2004) Adipose tissue as 5. Van Gaal LF, Mertens IL, De Block CE (2006)
an endocrine organ. J Clin Endocrinol Metab Mechanisms linking obesity with cardiovascu-
89(6):2548–2556. doi:10.1210/jc.2004-0395 lar disease. Nature 444(7121):875–880.
2. Rosen ED, Spiegelman BM (2006) Adipocytes doi:10.1038/nature05487
as regulators of energy balance and glucose 6. Kahn SE, Hull RL, Utzschneider KM (2006)
homeostasis. Nature 444(7121):847–853. Mechanisms linking obesity to insulin resistance
doi:10.1038/nature05483 and type 2 diabetes. Nature 444(7121):840–
3. Tilg H, Moschen AR (2008) Inflammatory 846. doi:10.1038/nature05482
mechanisms in the regulation of insulin resis- 7. Olshansky SJ, Passaro DJ, Hershow RC et al
tance. Mol Med 14(3–4):222–231. (2005) A potential decline in life expectancy in
doi:10.2119/2007-00119.Tilg the United States in the 21st century. N Engl
4. Hajer GR, van Haeften TW, Visseren FL J Med 352(11):1138–1145. doi:10.1056/
(2008) Adipose tissue dysfunction in obesity, NEJMsr043743
diabetes, and vascular diseases. Eur Heart 8. Rosen ED, Walkey CJ, Puigserver P et al (2000)
J 29(24):2959–2971. doi:10.1093/eurheartj/ Transcriptional regulation of adipogenesis.
ehn387 Genes Dev 14(11):1293–1307
74 Andreas Hentschel and Robert Ahrends
Abstract
When deciding to perform a quantitative proteomics analysis, selectivity, sensitivity, and reproducibility are
important criteria to consider. The use of multiple reaction monitoring (MRM) has emerged as a powerful
proteomics technique in that regard since it avoids many of the problems typically observed in discovery-
based analyses. A prerequisite for such a targeted approach is that the protein targets are known, either as
a result of previous global proteomics experiments or because a specific hypothesis is to be tested. When
guidelines that have been established in the pharmaceutical industry many decades ago are taken into
account, setting up an MRM assay is relatively straightforward. Typically, proteotypic peptides with favor-
able mass spectrometric properties are synthesized with a heavy isotope for each protein that is to be moni-
tored. Retention times and calibration curves are determined using triple-quadrupole mass spectrometers.
The use of iRT peptide standards is both recommended and fully integrated into the bioinformatics pipe-
line. Digested biological samples are mixed with the heavy and iRT standards and quantified. Here we
present a generic protocol for the development of an MRM assay.
1 Introduction
Jörg Reinders (ed.), Proteomics in Systems Biology: Methods and Protocols, Methods in Molecular Biology, vol. 1394,
DOI 10.1007/978-1-4939-3341-9_6, © Springer Science+Business Media New York 2016
75
76 Annie Moradian et al.
Fig. 1 The two operational modes of a QTRAP instrument. (a) QTRAP mode. Selected ions of separated
peptides pass through Q1 and are fragmented in Q2, all fragment ions pass through Q3, and a full MS/MS
spectrum is recorded. This mode is particularly useful in the early stages of assay development when identities
of peptides need to be confirmed. (b) QQQ mode. Selected ions of separated peptides pass through Q1 and are
fragmented in Q2, and all selected fragment ions pass through Q3. Co-eluting transition ion pairs are recorded.
This mode is particularly useful in maximizing sensitivity of known peptides
3.0
VEA
2.5
YIL
2.0
AGG
Intensity (10Ÿ6)
1.5
TPV...T...
TPV...S...
1.0
DAV
GDL
TGF
GTF
0.5
LGG
FLL
0.0
10 20 30 40 50
Retention Time
Fig. 2 Elution profile of 11 iRT peptides in a 45-min gradient. Peptides are LGGNETQVR (LGG), AGGSSEPVTGLADK
(AGG), VEATFGVDESANK (VEA), YILAGVESNK (YIL), TPVISGGPYYER (TPV…S...), TPVITGAPYYER (TPV…T...), GDLD
AASYYAPVR (GLD), DAVTPADFSEWSK (DAV), TGFIIDPGGVIR (TGF), GTFIIDPAAIVR (GTF), FLLQFGAQGSPLFK (FLL)
Fig. 3 Example of a calibration curve for a standard peptide covering a dynamic range from 30 to 500,000
attomol. This curve can be used to quantify unknown samples. If an unknown sample has a response of
100,000 AU, the sample contains 550 attomol of that particular peptide
2 Materials
2.1 Chemicals 1. Solvent A: 0.2 % Formic acid. Add 2 mL of formic acid (for
mass spectrometry; 98 %) to 998 mL of water (HPLC gradient
2.1.1 LC and LC
grade quality).
Samples
2. Solvent B: Acetonitrile containing 0.2 % formic acid. Add 2
mL of formic acid (for mass spectrometry; 98 %) to 998 mL of
acetonitrile (HPLC gradient grade quality).
3. Synthetic heavy standard peptides (either prepared in-house or
commercially).
4. Synthetic iRT peptides (either prepared in-house or
commercially).
5. Standard solvent (30 % acetonitrile, 70 % of 0.2 % formic acid).
Add 30 mL of acetonitrile to a 69.8 mL of water and 0.2 mL
of formic acid.
6. All reagents should be of the highest purity available.
2.1.2 FASP 1. Lysis buffer. Add 5 g sodium dodecyl sulfate (SDS), 1.58 g
(100 mM) Tris/HCl (pH 7.6; adjust with HCl as necessary),
1.54 g (100 mM) D,L-dithiothreitol (DTT), supplemented
with 1× protease inhibitor cocktail (complete, EDTA free,
Roche) and 17.42 mg (1 mM) phenylmethanesulfonyl fluoride
(PMSF) brought up to 100 mL with ddH2O.
Targeted Tempo-Spatial Proteomics 79
2.1.3 Preparation 1. Heavy standard proteotypic peptides of target proteins are syn-
of Heavy Standard Peptide thesized, either commercially or if available in-house (see Note 2).
Solutions 2. Prepare solutions of peptide standards at a nominal concentra-
tion of 1 pmol/μL using 30 % acetonitrile and 70 % of 0.2 %
formic acid (see Note 3).
3. Eppendorf tubes; pipettes.
2.2 Liquid 1. QTRAP 6500 (AB Sciex, Framingham, MA) with an Eksigent
Chromatography- ekspert nanoLC 425 pump, ekspert nanoLC400 autosampler,
Mass Spectrometry ekspert cHiPLC, and Analyst software. The LC system is coupled
(LC-MS/MS) to a NanoSpray III Source and Heated Interface (AB/Sciex).
2. CHiPLC Chrom XP C18-CL 3 μm trap column, 120 Å (200
μm × 0.5 mm).
80 Annie Moradian et al.
3 Methods
3.3 LC-MRM 1. For the separation and analysis of the samples, a nanoLC
of Heavy Proteotypic QTRAP 6500 (AB Sciex, Framingham, MA) can be used
Standard Peptide (see Note 5).
Solutions 2. The peptides are separated using a CHiPLC Chrom XP
C18-CL 3 μm column, 120 Å (75 μm × 150 mm) equipped
with a CHiPLC Chrom XP C18-CL 3 μm trap column, 120 Å
(200 μm × 0.5 mm) at a column temperature of 45 °C and a
flow rate of 300 nL/min. Solvent A is 0.2 % formic acid and
solvent B is 98.8 % acetonitrile containing 0.2 % formic acid.
Linear gradients from 5 to 30 % B are applied within 45 min,
30–90 % B in 2 min, followed by 100 % B for 10 min.
Preliminary mass spectra are recorded in positive ion mode
acquiring data from the transition lists, initially in the QTRAP
Targeted Tempo-Spatial Proteomics 83
3.4 Data Analysis Import the wiff (or appropriate raw) files of all measurements into
Skyline software (see Note 19).
4 Notes
19. The files are imported and show target masses, individual tran-
sitions per peptide, peak areas, and retention times over the
multiple analysis. Considerable manual intervention is needed
to confirm assignment of peaks.
Acknowledgement
References
1. Picotti P, Aebersold R (2012) Selected reaction Proteomics 12(8):1111–1121. doi:10.1002/
monitoring-based proteomics: workflows, pmic.201100463
potential, pitfalls and future directions. Nat 7. MacLean B, Tomazela DM, Shulman N et al
Methods 9(6):555–566. doi:10.1038/ (2010) Skyline: an open source document editor
nmeth.2015 for creating and analyzing targeted proteomics
2. Carr SA, Abbatiello SE, Ackermann BL et al experiments. Bioinformatics 26(7):966–968.
(2014) Targeted peptide measurements in biology doi:10.1093/bioinformatics/btq054
and medicine: best practices for mass spectrometry- 8. Wisniewski JR, Zougman A, Nagaraj N et al
based assay development using a fit-for-purpose (2009) Universal sample preparation method
approach. Mol Cell Proteomics 13(3):907–917. for proteome analysis. Nat Methods 6(5):359–
doi:10.1074/mcp.M113.036095 362. doi:10.1038/nmeth.1322
3. Huttenhain R, Soste M, Selevsek N et al (2012) 9. Hess S, Akermann M, Ropte D et al (2001)
Reproducible quantification of cancer-associated Rapid and sensitive LC separation of new
proteins in body fluids using targeted pro- impurities in trimethoprim. J Pharm Biomed
teomics. Sci Transl Med 4(142):142ra194. Anal 25(3–4):531–538
doi:10.1126/scitranslmed.3003989 10. Hess S, Dolker M, Haferburg D et al (2001)
4. Kuzyk MA, Smith D, Yang J et al (2009) Separation, analyses and syntheses of trime-
Multiple reaction monitoring-based, multi- thoprim impurities. Pharmazie 56(4):306–310
plexed, absolute quantitation of 45 proteins in 11. Hess S, Muller CE, Frobenius W et al (2000)
human plasma. Mol Cell Proteomics 8(8):1860– 7-deazaadenines bearing polar substituents:
1877. doi:10.1074/mcp.M800540-MCP200 structure-activity relationships of new A(1) and
5. Picotti P, Rinner O, Stallmach R et al (2010) A(3) adenosine receptor antagonists. J Med
High-throughput generation of selected Chem 43(24):4636–4646. doi:10.1021/
reaction-monitoring assays for proteins and Jm000967d
proteomes. Nat Methods 7(1):43–46. 12. Hess S, Teubert U, Ortwein J et al (2001)
doi:10.1038/nmeth.1408 Profiling indomethacin impurities using high-
6. Escher C, Reiter L, MacLean B et al (2012) performance liquid chromatography and
Using iRT, a normalized retention time for nuclear magnetic resonance. Eur J Pharm Sci
more targeted measurement of peptides. 14(4):301–311
Chapter 7
Abstract
Mass spectrometry-based phosphoproteomics has been rapidly spread based on the advancement of mass
spectrometry and development of efficient enrichment techniques for phosphorylated proteins or peptides.
Non-targeted approach has been employed in most of the studies for phosphoproteome analysis. However,
targeted approach using selected/multiple reaction monitoring (SRM/MRM) is an indispensible tech-
nique used for the quantitation of known targets especially when we have many samples to quantitate
phosphorylation events on proteins in biological or clinical samples. We herein describe the application of
a large-scale phosphoproteome analysis and SRM-based quantitation for the systematic discovery and
validation of biomarkers.
1 Introduction
Jörg Reinders (ed.), Proteomics in Systems Biology: Methods and Protocols, Methods in Molecular Biology, vol. 1394,
DOI 10.1007/978-1-4939-3341-9_7, © Springer Science+Business Media New York 2016
87
88 Jun Adachi et al.
2 Materials
Tissues Cells
Non-targeted analysis Targeted analysis
Lysates Lysates
2 mg/sample 0.5 mg/sample
PTS method
Stable isotope
Peptides Peptides labeled peptides
Fe-IMAC
Phosphopeptides Phosphopeptides
Data analysis
Target selection
Fig. 1 Workflows of non-targeted analysis and targeted (SRM) analysis. In non-targeted analysis, proteins are
denatured and digested using phase transfer surfactant (PTS) method. Phosphopeptides are enriched using
Fe-IMAC, followed by SCX off-line fractionation and LC-MS/MS. In the SRM analysis, the individual sample is
processed by PTS method and spiked with a mixture of the isotope-labeled peptides. The resulting sample is
applied to Fe-IMAC to enrich the phosphopeptides and analyzed by SRM analysis
2.4 Strong Cation- 1. Buffer A: 25 % Acetonitrile, 10 mM H3PO4 (pH 3). Mix 250
Exchange mL of acetonitrile, approximately 650 mL of water, and 685
Chromatography μL of phosphoric acid. Adjust to pH 3.0 by adding KOH solu-
tion and to 1 L with water.
2. Buffer B: 25 % Acetonitrile, 10 mM H3PO4 (pH 3), 1 M KCl.
Mix 250 mL of acetonitrile, approximately 650 mL of water,
685 μL of phosphoric acid, and 74.55 g of KCl. Adjust to pH
3.0 by adding KOH solution and to 1 L with water.
3. HPLC system, e.g., Prominence UFLC (Shimadzu, Kyoto,
Japan).
4. Strong cation-exchange (SCX) column, e.g., 50 mm × 2.1 mm,
5 μm, 300 Å, ZORBAX 300SCX (Agilent Technology, Santa
Clara, CA).
2.6 Targeted (SRM/ Basically same reagents and equipment are used except for those
MRM-Based) LC-MS/ listed below.
MS Analysis 1. Stable isotope-labeled peptides (SI peptides) (crude grade)
(Thermo Fisher Scientific) (see Note 7).
2. Mass spectrometer for SRM analysis, e.g., TSQ Vantage triple-
quadruple mass spectrometer (Thermo Scientific).
3. Software for targeted (SRM/MRM) proteomic analysis, e.g.,
Pinpoint 1.2 (Thermo Scientific), which is a software to quan-
titate the peak areas (quantitative data of targeted peptides)
from the raw data of SRM analysis as well as to develop the
SRM methods.
4. 2 % Acetonitrile, 0.1 % TFA.
5. 2 % Acetonitrile, 0.1 % TFA, 25 μg/mL EDTA.
6. 1 pmol/μL BSA digest solution.
3 Methods
3.1 Sample 1. Chill the stainless tissue pulverizer in liquid nitrogen. Place a
Preparation piece of frozen tissue in the chilled device and pulverize the
for Tissue Sample tissue by striking the device with a mallet several times.
2. Check the size of crushed particles. Keep pulverizing the par-
ticles until there are no large pieces left in the particles.
3. Transfer the grinded tissue into the chilled tube. Store at −80
°C.
4. Take the required amount of pulverized tissue sample into a
microcentrifuge tube (see Note 8). If a high level of blood
contamination is predicted, wash the sample by an appropriate
volume of PBS.
5. Add cold lysis buffer, approximately 15 μL per 1 mg of tissue
(see Note 9). Suspend the tissue by pipetting.
6. Immediately place the sample tubes into an ice-cold water bath
in the Bioruptor-UCD-250 sonicator. Homogenize the sam-
ple by the sonication for 10 min (10 cycles of 30 s on/30 s off)
with the amplitude set to 250 W (see Note 10).
7. Centrifuge the sample at 100,000 × g for 30 min at 4 °C. Collect
the supernatant into a new tube. Place a small amount of the
92 Jun Adachi et al.
3.2 Sample 1. Wash cells in the tissue culture dish by directly adding cold
Preparation PBS and rocking gently. Aspirate PBS and repeat. Keep tissue
for Cultured Cells culture dish on ice throughout.
2. Add appropriate volume of ice-cold lysis buffer to the dish,
approximately 1 mL for a 100 mm tissue culture dish.
3. Scrape cells from the surface using a rubber spatula. Transfer
the cells into the chilled tube.
4. Sonicate the samples in an ice-cold water bath using the
Bioruptor-UCD-250 sonicator. Homogenize the sample by
the sonication for 10 min (10 cycles of 30 s on/30 s off) with
the amplitude set to 250 W. Repeat the sonication if the sam-
ple is still viscose.
5. Take a small amount of the sample into another tube to deter-
mine the protein concentration. Store the remainder at −80 °C.
6. The protein concentration is determined by a DC protein assay
kit using BSA as the standard.
3.3 Protein Digestion 1. Add the homogenate to a new tube. Dilute the homogenate
with the lysis buffer to a concentration that is constant across
all samples (see Note 11).
2. Reduce cysteine residues with 10 mM dithiothreitol (DTT) for
30 min.
3. Alkylate the residues with 50 mM iodoacetamide (IAA) for
30 min in the dark.
4. Dilute the sample five times with PTS-A buffer.
5. Digest the sample by 1:100 (w/w) trypsin for 12 h at 37 °C.
6. Add an equal volume of ethyl acetate. Acidify the sample by
adding 1/200 volume of trifluoroacetic acid (TFA) in order to
transfer the detergents from the water layer into the ethyl ace-
tate layer while most of the peptides remain in the water layer.
Mix the ethyl acetate and water layers well by vortexing the
tube. Centrifuge the tube at 10,000 × g for 10 min at room
temperature to separate the sample into two layers. Discard the
upper ethyl acetate layer.
7. Dry the aqueous layer using Speed Vac and store at −80 °C.
3.5 Phosphopeptide 1. Dissolve the tryptic digests prepared from the tissue samples or
Enrichment cultured cells in 60 % acetonitrile and 0.1 % TFA solution.
by Fe-IMAC for Non- 2. Load the digests to the Fe-IMAC resin. Centrifuge at 150 × g
targeted for 2 min and discard the flow through.
Phosphoproteome 3. Wash the resin by 60 % acetonitrile and 0.1 % TFA solution (3
Analysis mL of the solution per 1 mL of the resin) and centrifuge at
150 × g for 2 min to wash off the non-phosphopeptides. Repeat
this step twice more.
4. Add 2 % acetonitrile and 0.1 % TFA solution (3 mL of the
solution per 1 mL of the resin) to the resin. Centrifuge at
150 × g for 2 min and discard the flow through.
5. Elute phosphopeptides by 1 % phosphate solution (1 mL of
the solution per 1 mL of the resin) and centrifuge at 150 × g for
2 min. Collect the elute into a tube. Repeat this step once
more and then collect the second elute into the same tube.
6. Desalt the elute with a disposable solid-phase extraction (SPE)
device such as Sep-Pak C18 cartridge.
7. Dry the sample using Speed Vac and store at −80 °C.
3.6 Strong Cation- 1. Dissolve the sample in mobile buffer A (see Note 15).
Exchange 2. Apply the sample to an HPLC and separate on an SCX column
Chromatography using a linear gradient of mobile buffer A and B and sequen-
for Non-targeted tially collect eluate every 1 min (see Note 16).
Phosphoproteome
Analysis
94 Jun Adachi et al.
3.7 LC-MS/MS 1. Add 10 μL of 2 % acetonitrile and 0.1 % TFA to each sample vial.
for Non-targeted 2. Vortex each vial for 1 min and then spin down.
Phosphoproteome 3. Set the operating parameters of the mass spectrometer (see
Analysis Note 18).
4. Analyze each sample by LC-MS/MS (see Note 19).
5. Apply the acquired raw file to data analysis software with search
engine to identify and quantify the phosphopeptides (see Note 20)
6. Select the phosphopeptide that has to be validated in the sub-
sequent SRM analysis.
3.9 Targeted Analysis 1. In order to acquire target information (retention time, mass of
by LC-SRM/MS target ion, and transition ions) prior to the analysis, prepare a
mixture of the stable isotope-labeled peptide (SI peptides),
which has the same sequence as the phosphopeptide selected
from the results of non-targeted analysis. Analyze the mixture
by LC-MS/MS using data-dependent mode (see Note 23).
2. Create a primary method for the subsequent SRM analysis by
analyzing the acquired MS data and selecting the precursor
ions of each target observed with a strong signal intensity
(doubly, triply, or higher charged ions) and the product ions
generated from the precursor ion with a strong signal intensity
(see Note 24).
3. Optimize the parameters (m/z of product ions and collision
energy (CE)) of the SRM method (see Note 25).
4. Add 10 μL of 2 % acetonitrile, 0.1 % TFA, and 25 μg/mL
EDTA to each sample.
5. Vortex each vial for 1 min to dissolve the peptides and then
spin down.
6. Set the optimized SRM method and other operating parame-
ters for the SRM analysis (see Note 26).
7. Analyze each sample by LC-SRM/MS (see Note 27)
8. Analyze the acquired raw data by the software (see Note 28).
Target peptides are compared across the samples by extracted
ion chromatogram (XIC) intensity of each SRM transition and
then normalize the values of the endogenous targeted peptides
to those of the corresponding SI peptides.
4 Notes
References
1. Sharma K, D’Souza RC, Tyanova S et al (2014) 3. Masuda T, Tomita M, Ishihama Y (2008) Phase
Ultradeep human phosphoproteome reveals a transfer surfactant-aided trypsin digestion for
distinct regulatory nature of tyr and ser/thr- membrane proteome analysis. J Proteome Res
based signaling. Cell Rep 8(5):1583–1594. 7(2):731–740. doi:10.1021/pr700658q
doi:10.1016/j.celrep.2014.07.036 4. Rappsilber J, Mann M, Ishihama Y (2007)
2. Narumi R, Murakami T, Kuga T et al (2012) A Protocol for micro-purification, enrichment,
strategy for large-scale phosphoproteomics and pre-fractionation and storage of peptides for
SRM-based validation of human breast cancer proteomics using StageTips. Nat Protoc
tissue samples. J Proteome Res 11(11):5311– 2(8):1896–1906. doi:10.1038/nprot.2007.26
5322. doi:10.1021/pr3005474 1,nprot.2007.261 [pii]
Chapter 8
Abstract
Precise quantification is a major issue in contemporary proteomics. Both stable-isotope-labeling and label-
free methods have been established for differential protein quantification and both approaches have differ-
ent advantages and disadvantages. The present protocol uses the superior precision of label-free
SWATH-mass spectrometry to test for suitability of cell lines for a SILAC-labeling approach as systematic
regulations may be introduced upon incorporation of the “heavy” amino acids. The SILAC-labeled cell
cultures can afterwards be used for further analyses where stable-isotope-labeling is mandatory or has
substantial advantages over label-free approaches such as pulse-chase-experiments and differential protein
interaction analyses based on co-immunoprecipitation. As SWATH-mass spectrometry avoids the missing-
value-problem typically caused by undersampling in highly complex samples and shows superior precision
for the quantification, it is better suited for the detection of systematic changes caused by the SILAC-
labeling and thus, can serve as a useful tool to test cell lines for changes upon SILAC-labeling.
1 Introduction
Jörg Reinders (ed.), Proteomics in Systems Biology: Methods and Protocols, Methods in Molecular Biology, vol. 1394,
DOI 10.1007/978-1-4939-3341-9_8, © Springer Science+Business Media New York 2016
101
102 Yvonne Reinders et al.
Fig. 1 Distribution of the coefficients of variation (CV) for the SILAC- and the
SWATH-based quantification of the same HMB2-cell lysates. Both methods
were accomplished in triplicate. Only CVs for proteins identified and quantified
in all of the three SILAC-runs were included in this graph. Due to the undersam-
pling in the SILAC-based approach, substantially fewer proteins (719) are con-
tained in the SILAC-violin plot than in the SWATH-violin plot (2202) because the
SWATH-methodology eliminates undersampling. Furthermore, the precision of
the SWATH-technique (most frequent CV ~8 %) is far superior to the SILAC-
method (most frequent CV ~25 %)
2 Materials
2.1 Cell Culture All items are given for the cell line tested in our lab and should be
for SILAC-Labeling adapted to the respective cell line to be tested (see Note 3).
1. HMB2 cell line.
2. Dulbecco’s Modified Eagle Medium (DMEM) without argi-
nine and lysine.
3. Penicillin and streptomycin.
4. L-glutamine.
13
5. C6,15N2-lysine/13C6,15N4-arginine and the respective unla-
beled lysine and arginine.
6. Dialyzed fetal calf serum (FCS).
7. Phosphate buffered saline (PBS).
8. Trypsin.
9. Lysis buffer: 20 mM sodium phosphate, pH 7.4, 0.2 % SDS.
10. Sonicator.
11. Kit for determination of total protein amount or material for
your method of choice.
3 Methods
3.1 Cell Culture 1. Seed the cells into “light” and “heavy” SILAC DMEM media
for SILAC-Labeling supplemented with 10 % dialyzed FCS, 400 U/mL penicillin,
50 μg/mL streptomycin, and 300 μg/mL glutamine at the
required density (see Note 4).
Testing Suitability of Cell Cultures for SILAC-Experiments Using SWATH-Mass… 105
2. Cultivate the cells for at least five cell doubling times with fresh
medium every other day to ensure almost complete labeling of
all proteins.
3. Check for equal growth of the cells in both media regularly.
4. Wash the cells three times with PBS before harvesting the cells.
5. Detach adherent cells using 500 μg/L trypsin.
6. Lyse the cells by sonication in 1 mL lysis buffer.
7. Determine protein concentrations by your method of choice.
3.3 Nano-LC-MS/ HPLC settings can be changed according to the depth of the
MS-Analysis analysis that shall be achieved; in our lab 2–4 h runs are typically
accomplished. The HPLC conditions are the same for the SILAC-
runs with combined samples (also used for library generation) and
the SWATH-runs of the individual samples (see Note 6).
1. Load 1 μg of protein digest on a 2 cm, 100 μm I.D. C18-trapping
column (particle size 5 μm) with a flow rate of 5 μL/min.
2. Separate the samples on a 25 cm long 75 μm I.D. C18 column
(3 μm particle size) at a flow rate of 300 nL/min using a
212 min long gradient from 4 to 40 % B.
3. Operate the mass spectrometer at a TOP25 method (0.25 s for
TOF-MS from 350 to 1250 m/z, 0.1 s per MS/MS from 100
to 1500 m/z) for the SILAC-runs (see Note 7).
4. For the SWATH runs a 50 ms TOF-MS scan is followed by 37
windows of 25 m/z (100 ms per window) spanning the mass
range of 350–1250 m/z.
3.4 Data Analysis 1. Search the IDA-data using Protein Pilot against the
Uniprot-database.
2. Apply a 1 % false discovery rate (FDR) for the identifications.
106 Yvonne Reinders et al.
4 Notes
Acknowledgements
References
1. Putz S, Reinders J, Reinders Y et al (2005) targeted proteomics. Curr Top Med Chem
Mass spectrometry-based peptide quantifica- 14(3):344–350
tion: applications and limitations. Expert Rev 7. Kiyonami R, Domon B (2010) Selected reac-
Proteomics 2(3):381–392 tion monitoring applied to quantitative pro-
2. Ong SE, Blagoev B, Kratchmarova I et al teomics. Methods Mol Biol 658:155–166
(2002) Stable isotope labeling by amino acids 8. Fierro-Monti I, Racle J, Hernandez C et al
in cell culture, SILAC, as a simple and accurate (2013) A novel pulse-chase SILAC strategy mea-
approach to expression proteomics. Mol Cell sures changes in protein decay and synthesis rates
Proteomics 1(5):376–386 induced by perturbation of proteostasis with an
3. Ong S, Mann M (2006) A practical recipe for Hsp90 inhibitor. PLoS One 8(11):e80423.
stable isotope labeling by amino acids in cell doi:10.1371/journal.pone.0080423
culture (SILAC). Nat Protoc 1(6):2650–2660. 9. Takahashi Y (2015) Co-immunoprecipitation
doi:10.1038/nprot.2006.427 from transfected cells. Methods Mol Biol
4. Neilson KA, Ali NA, Muralidharan S et al 1278:381–389. doi:10.1007/978-1-4939-
(2011) Less label, more free: approaches in 2425-7_25
label-free quantitative mass spectrometry. 10. Wisniewski JR, Zougman A, Nagaraj N et al
Proteomics 11(4):535–553. doi:10.1002/ (2009) Universal sample preparation method
pmic.201000553 for proteome analysis. Nat Methods 6(5):359–
5. Gillet LC, Navarro P, Tate S et al (2012) Targeted 362. doi:10.1038/nmeth.1322
data extraction of the MS/MS spectra generated 11. Benjamini Y, Hochberg Y (1995) Controlling
by data-independent acquisition: a new concept the false discovery rate: a practical and powerful
for consistent and accurate proteome analysis. approach to multiple testing. J Roy Statist Soc
Mol Cell Proteomics 11(6):O111.016717. Ser B 57(1):289–300. doi:10.2307/2346101
doi:10.1074/mcp.O111.016717 12. Dunn OJ (1961) Multiple comparisons among
6. Gomez YR, Gallien S, Huerta V et al (2014) means. J Am Stat Assoc 56(293):52–64.
Characterization of protein complexes using doi:10.1080/01621459.1961.10482090
Chapter 9
Abstract
During the last 15 years, the combination of chemical cross-linking and high-resolution mass spectrometry
(MS) has matured into an alternative approach for analyzing 3D-structures of proteins and protein com-
plexes. Using the distance constraints imposed by the cross-links, models of the protein or protein complex
under investigation can be created. The majority of cross-linking studies are currently conducted with
homobifunctional amine-reactive cross-linkers. We extend this “traditional” cross-linking/MS strategy by
adding complementary photo-cross-linking data. For this, the diazirine-containing unnatural amino acids
photo-leucine and photo-methionine are incorporated into the proteins and cross-link formation is
induced by UV-A irradiation. The advantage of the photo-cross-linking strategy is that it is not restricted
to lysine residues and that hydrophobic regions in proteins can be targeted, which is advantageous for
investigating membrane proteins. We consider the strategy of combining cross-linkers with orthogonal
reactivities and distances to be ideally suited for maximizing the amount of structural information that can
be gained from a cross-linking experiment.
1 Introduction
Jörg Reinders (ed.), Proteomics in Systems Biology: Methods and Protocols, Methods in Molecular Biology, vol. 1394,
DOI 10.1007/978-1-4939-3341-9_9, © Springer Science+Business Media New York 2016
109
110 Philip Lössl and Andrea Sinz
Fig. 1 Amine- and photo-reactive cross-linking of proteins. (a) Amine-reactive NHS esters typically connect
lysine residues (shown in blue and red). NHS esters are usually employed as equimolar mixture of non-
deuterated and deuterated derivatives to facilitate the identification of cross-linked products in the mass
spectra based on their characteristic isotope patterns. The cross-linker BS2G (black ) possesses a spacer
length of 7.7 Å, but Cα–Cα distances of up to 29 Å can be bridged [14]. (b) Photo-Leu (red-black ) as an
example for photo-reactive diazirines. Upon irradiation with UV-A light (~365 nm) a reactive carbene (free
electron pair shown in black ) is created that can insert into NH or CH groups (blue ). The carbene can also
create an alkene or react with water, resulting in the formation of an alcohol [30] (see Fig. 3)
Cross-Linking/MS for Protein 3D-Structure Analysis 111
2 Materials
Prepare all solutions using ultrapure water, i.e., Milli-Q water, and
use reagents at the highest available purity. Amine-reactive chemi-
cal cross-linkers (NHS esters) should be stored dry under inert gas
(helium or nitrogen) and stock solutions have to be prepared in
dimethylsulfoxide (DMSO) directly before use. The photo-reactive
diazirine-containing amino acids L-photo-methionine and
L-photo-leucine can be commercially obtained from Thermo
Fisher Scientific and have to be stored under light exclusion.
112 Philip Lössl and Andrea Sinz
2.2 Incorporation 1. Cell culture dishes and Triple Flasks (Thermo Fisher Scientific).
of Photo-reactive 2. Cell culture medium: Dulbecco’s Modified Eagle Medium
Amino Acids into DMEM/F12 (Gibco/Life Technologies), 10 % (v/v) FCS,
Proteins in HEK 293 1 % (v/v) penicillin/streptomycin solution and 1 μg/μL
Cells puromycin.
3. Dulbecco’s Modified Eagle Limiting Medium DMEM-LM,
leucine- and methionine-depleted (Gibco/Life Technologies).
4. Fetal calf serum (FCS, Biochrom).
5. Penicillin/streptomycin solution.
6. Puromycin.
7. Phosphate-buffered saline (PBS): 140 mM NaCl, 10 mM KCl,
8 mM Na2HPO4, 2 mM KHPO4, pH 7.4.
8. 0.05 % (w/v) trypsin–EDTA solution (T/E).
9. 2 M Tris–HCl, pH 8.0.
10. L-photo-leucine and L-photo-methionine (Thermo Fisher
Scientific).
11. HEK293 EBNA cells stably transfected with the protein-
coding DNA sequence (see Note 9).
2.3 Photochemical After incorporation of the photo-reactive amino acids photo-Met and
Cross-Linking photo-Leu, we conduct the photo-cross-linking reactions in a home-
built UV irradiation chamber [22]. The chamber contains fluores-
cence lamps (40 W; emission spectrum between 305 and 420 nm,
emission maximum at 360 nm (CLEO Performance R UV-A-
fluorescence lamp, Phillips)). The UV-A sensor for quantifying the
irradiation energy was purchased from Kühnast (see Note 7).
7. TFA (Sigma).
8. Digestion buffer, freshly prepared: dissolve one trypsin, GluC
or chymotrypsin aliquot in 80 μL of 50 mM ammonium bicar-
bonate to obtain a solution of 13 ng/μL, store on ice.
2.7 Liquid In our lab, we use the following LC/MS setup, consisting of an
Chromatography/Mass Ultimate 3000 RSLC nano-HPLC system (Dionex) that is directly
Spectrometry (LC/MS) coupled to the nano-ESI source (EASY Spray source, Thermo
Fisher Scientific) of an Orbitrap Fusion Tribrid mass spectrometer
(Thermo Fisher Scientific) [23]. Alternatively, an LTQ-Orbitrap
XL mass spectrometer (Thermo Fisher Scientific) equipped with
nano-ESI source (Proxeon) is employed. Other nano-HPLC sys-
tems or mass spectrometers, such as FTICR or Q-TOF instru-
ments equipped with a nano-ESI source, can be used as well.
2.8 Software 1. Perform MS and MS/MS data acquisition and data analysis
for Data Acquisition with XCalibur 3.0 in combination with DCMS link 2.14.
and Analysis 2. Analyze cross-linked products with StavroX 3.4.5 (www.
StavroX.com) [24] that allows an automatic analysis and anno-
tation of MS and MS/MS data.
3. For obtaining optimum results, the following settings should be
used: Maximum mass deviation of 3 ppm between calculated and
experimental precursor masses and signal-to-noise ratio of ≥2.
4. Consider primary amino groups (Lys side chains and N-termini)
as well as hydroxyl groups (Thr, Ser, Tyr) as potential cross-
linking sites for BS2G [25, 26].
5. Consider all 20 amino acid residues as cross-linking sites for
UV-A-induced cross-linking of photo-Met and photo-Leu.
6. Oxidation of Met should be set as variable modification for all
cross-linked proteins.
7. Include carbamidomethylation as fixed modification for Cys.
8. Consider two missed cleavage sites for each amino acid (cleav-
age sites: Lys and Arg for trypsin; Tyr, Trp, and Phe for chy-
motrypsin; Glu and Asp for GluC).
3 Methods
3.2 Incorporation All steps that require manual handling of cultured cells must be
of Photo-reactive conducted under a laminar flow box. Solutions and materials have
Amino Acids into to be sterile-packaged or autoclaved prior to use.
Proteins 1. Add HEK293 EBNA cells to 10 mL of cell culture medium.
2. Culture the cells on a 57 cm2 cell culture dish in an incubator
with a water-saturated 5 % CO2 atmosphere at 37 °C.
3. Exchange the medium when the contained pH indicator turns
yellow indicating acidification.
4. Monitor the cell growth every other day by checking the cell
confluence under a microscope. Once a confluence of 80 % is
reached, passage the cells to new dishes:
(a) Wash the cells with 5 mL PBS.
(b) Add 1.5 mL T/E and incubate for 5 min at 37 °C to
cleave off the cells from the plate.
(c) Stop proteolysis by adding 8.5 mL of cell culture medium.
(d) Pellet the cells by centrifuging them for 5 min at 500 × g in
a 15-mL Falcon tube.
(e) Resuspend the cell pellet in 5 mL fresh cell culture
medium.
(f) Distribute the suspension on fresh cell culture dishes.
5. When the cells reach 80 % confluence again, replace the cell
culture medium with FCS-free medium (containing only
DMEM/F12, penicillin/streptomycin, and puromycin) and
incubate them for another 24 h (see Note 10).
6. Remove the FCS-free medium and add 10 mL of DMEM-LM
supplemented with 4 mM L-photo-Leu and 2 mM L-photo-
Met (see Notes 8 and 10).
7. Replace the medium after 2 days, storing the first batch of
supernatant at −20 °C.
8. Collect the second batch after 2 days of incubation.
9. Equilibrate the collected medium by adding Tris–HCl buffer
(pH 8) to a final concentration of 12 mM.
116 Philip Lössl and Andrea Sinz
3.4 SDS-PAGE 1. Load your samples on a Precise™ Protein Gel 4–20 % acryl-
amide gradient gel.
2. Perform the gel electrophoresis at a constant current of 25 mA.
3. After the electrophoresis is finished, keep the gel in fixing solu-
tion for 1 h.
4. Rinse the gel two times with Milli-Q H2O.
5. Stain the gel overnight in colloidal Coomassie staining
solution.
6. Destain the gel by shaking it in Milli-Q H2O.
7. Inspect the gel for signals that might represent the cross-linked
protein complex (Fig. 3).
3.5 In-Gel Digestion Working under a laminar flow box wearing gloves and sleeve pro-
tectors is recommended in order to avoid keratin and dust con-
tamination of samples. The laminar flow box should include an
incubation shaker suitable for Eppendorf tubes.
3.5.1 Excise Protein 1. Wash the Coomassie-stained gel twice for 10 min with Milli-Q
Bands from the SDS Gel H2O on a shaker.
2. Excise gel bands of interest using a sterile scalpel and transfer
gel bands to a clean glass plate. Cut the gel band into cubes of
ca. 1 mm3 and transfer them into an Eppendorf tube.
Cross-Linking/MS for Protein 3D-Structure Analysis 117
3.5.2 Reduce 1. Add 100 μL Milli-Q H2O to the gel pieces, shake for 10 min.
and Alkylate Proteins 2. Add 500 μL ACN. Shake for 5 min and discard the liquid.
3. Add 50 μL reduction buffer. Incubate at 56 °C for 30 min,
then let the solution cool down to room temperature.
118 Philip Lössl and Andrea Sinz
Fig. 3 SDS-PAGE analysis of nidogen-1 and laminin γ1 short arm after cross-
linking with BS2G. Nidogen-1 and laminin γ1 short arm were cross-linked both
separately and after co-incubation at molar ratios of 1:1 and 1:2. M molecular
weight marker
4. Add 500 μL ACN. Shake for 5 min and discard the liquid.
5. Add 50 μL alkylation buffer. Incubate at room temperature in
the dark for 30 min.
6. Add 500 μL ACN. Shake for 5 min and discard the liquid.
3.5.3 Wash and Destain 1. Add 100 μL ACN/100 mM ammonium bicarbonate (1:1),
Gel Pieces shake for 10–30 min to destain the gel pieces.
2. Add 500 μL ACN. Shake for 5 min and discard the liquid.
3. Repeat steps 1 and 2 if necessary.
3.5.4 In-Gel Digestion 1. Bring the gel pieces to complete dryness before adding the
digestion buffer (protease in ammonium bicarbonate).
2. Per gel band, add 25 μL trypsin digestion buffer and incubate
gel pieces for 1 h on ice. In case a double digestion is per-
formed, add GluC (or another protease) first and incubate the
gel pieces for 1 h on ice, then add trypsin and incubate for
another 1 h on ice.
3. Add 50 mM ammonium bicarbonate. The gel pieces have to
be covered completely.
4. Incubate overnight at 37 °C.
3.5.5 Extract Peptides 1. Stop the digestion by adding the double volume of extraction
solution.
2. Shake for 15 min at 37 °C, remove the supernatant, and trans-
fer it to a new Eppendorf tube.
Cross-Linking/MS for Protein 3D-Structure Analysis 119
3.6 In-Solution 1. Incubate proteins for 1 h with a 20-fold excess (v/v) of pre-
Digestion cooled acetone to precipitate them from solution.
3.6.1 Denature Proteins 2. Centrifuge the sample for 30 min at 15,000 × g (4 °C).
3. Let the pellet dry at room temperature.
4. Solubilize the pellet in 25 μL denaturing solution.
3.6.3 In-Solution 1. Dilute the sample with 60 μL Milli-Q H2O to avoid denatur-
Digestion ation of the proteases by urea.
2. Add trypsin digestion buffer to an enzyme/protein ratio of
1:50 (v/v) and incubate the sample at 37 °C for 2 h. In case a
double digestion is performed, add GluC (or another prote-
ase) first and incubate for 1 h at room temperature, then add
trypsin and incubate at 37 °C for 2 h.
3. Acidify the sample to a pH value below 4 (check with pH
paper) and concentrate it in a vacuum concentrator to a vol-
ume between 60 and 120 μL.
4. Peptide mixtures are ready to be analyzed by nano-HPLC/
nano-ESI-Orbitrap mass spectrometry. Before MS analysis,
samples may be stored at −20 °C.
3.8 Data Analysis Data analysis of cross-linked peptides is performed with the StavroX
software allowing lysines and protein N-termini as reaction sites of
NHS-ester based cross-linkers (Fig. 1). As NHS-esters have also
been found to react with serines, threonines, and tyrosines, these
reaction sites should also be taken into account [25, 26]. For
photo-cross-links induced by diazirines, all 20 amino acids were
considered as potential reaction sites (see Note 13, Fig. 1).
Please note that trypsin will cleave at modified lysines with low
frequency, therefore the number of missed cleavage sites has to be
set to a higher value for cross-link identification. Filtering of putative
cross-linked candidates is facilitated by the application of isotope-
labeled D0/D4 cross-linkers, which generate a characteristic isotope
pattern for cross-linked peptides. To identify this isotope pattern,
experimental mass lists obtained by high-resolution mass spectrom-
etry are filitered for monoisotopic deconvoluted masses (using the
Proteome Discoverer software, Thermo Fisher Scientific) that show
a mass difference of 4 amu. This reduced experimental mass list is
then compared to a theoretical mass list of all possibly cross-linked
peptides, allowing a maximum mass deviation of 3 ppm. This allows
automated identification of potentially cross-linked peptides, for
which MS/MS data have to be evaluated. Both the D0 and the D4
cross-linked species have to be isolated and fragmented, so the MS/
MS data can also be checked for the characteristic isotope patterns
(see Subheading 3.7, step 6). To further automate the MS/MS-based
identification of cross-linked species, the StavroX software calculates
the corresponding theoretical fragment ions and compares them to
the experimental mass spectra (Fig. 4). Subsequently, StavroX assigns
Cross-Linking/MS for Protein 3D-Structure Analysis 121
3.9 Use Cross-Links 1. Make sure that the numbering of the identified cross-links
as Distance complies with the atom numbering in the PDB files and/or
Constraints amino acid sequences used as input for the modeling process
for Rosetta-Based (see Note 16).
Computational 2. List all identified cross-links in a tab-separated constraint file,
Modeling as exemplified in Table 1 (see Note 17). Constraints obtained
from different cross-linking experiments may be listed in the
same file.
3. Enforce the specified distance constraints by the command line
flag “-constraints:cst_fa_file</path/to/constraint/file>”, if
working with high-resolution models where all amino acid side
chain atoms are modeled, or “-constraints:cst_file</path/to/co
nstraint/file>”, if working with low-resolution models where
amino acid side chains are represented by a centroid sphere.
Table 1
Overview of cross-links in the structurally solved region of the nidogen-1/laminin γ1 short arm
complex. Cα–Cα distances of cross-linked residues indicated in the crystal structure (PDB entry
1NPE) and in the best-scoring models created by Rosetta
4 Notes
Table 2
Information to be included in a Rosetta constraint file. The file should be tab-separated and must not
include any column headings. All listed information has to be in accordance with the input PDB file
Acknowledgements
References
1. Gavin A, Bösche M, Krause R et al (2002) 4. Tang X, Bruce JE (2010) A new cross-linking
Functional organization of the yeast proteome by strategy: protein interaction reporter (PIR) tech-
systematic analysis of protein complexes. Nature nology for protein-protein interaction studies. Mol
415(6868):141–147. doi:10.1038/415141a Biosyst 6(6):939–947. doi:10.1039/b920876c
2. Petrotchenko EV, Borchers CH (2010) 5. Calabrese AN, Pukala TL (2013) Chemical
Crosslinking combined with mass spectrome- cross-linking and mass spectrometry for the
try for structural proteomics. Mass Spectrom structural analysis of protein assemblies. Aust
Rev 29(6):862–876. doi:10.1002/mas.20293 J Chem 66(7):749. doi:10.1071/CH13164
3. Sinz A (2014) The advancement of chemical 6. Kalisman N, Adams CM, Levitt M (2012)
cross-linking and mass spectrometry: from sin- Subunit order of eukaryotic TRiC/CCT chap-
gle proteins to protein interaction networks. eronin by cross-linking, mass spectrometry,
Expert Rev Proteomics 11:733–743 and combinatorial homology modeling. Proc
Cross-Linking/MS for Protein 3D-Structure Analysis 127
Abstract
Matrix assisted laser desorption/ionization mass spectrometry imaging (MALDI MSI) is a technique to
visualize molecular features of tissues based on mass detection. This chapter focuses on MALDI MSI of
peptides and provides detailed operational instructions for sample preparation of cryoconserved and
formalin-fixed paraffin-embedded (FFPE) tissue. Besides sample preparation we provide protocols for the
MALDI measurement, tissue staining, and data analysis. On-tissue digestion and matrix application are
described for two different commercially available and commonly used spraying devices: the SunCollect
(SunChrom) and the ImagePrep (Bruker Daltonik GmbH).
Key words MALDI imaging, MSI, Peptide, Trypsin, Matrix application, ImagePrep, SunCollect,
Formalin-fixed paraffin-embedded (FFPE), Cryoconserved
1 Introduction
Jörg Reinders (ed.), Proteomics in Systems Biology: Methods and Protocols, Methods in Molecular Biology, vol. 1394,
DOI 10.1007/978-1-4939-3341-9_10, © Springer Science+Business Media New York 2016
129
130 Birte Beine et al.
2 Materials
2.6 Materials 1. Liquid Tip Ex (water based) to set marks for geometric orien-
for Setting Reference tation of the tissue.
Points on the ITO Slide 2. Solvent-resistant pen to set fine marks for coregistration (e.g.,
laboratory marker, #6130603, Paul Marienfeld GmbH &
Co. KG).
5. Ethanol, 100 %.
6. Xylene, 100 %.
7. Mounting medium (e.g., Richard-Allan Scientific Cytoseal
XYL, Thermo Fisher Scientific).
8. Cover glass.
3 Methods
3.1 Fresh Frozen The following description is based on the assumption that all steps
Tissue Sample of tissue preparation need to be performed including the tissue
Preparation conservation process. If you work for example with clinical samples
this process is in most cases already done and you can skip
Subheading 3.1.1 and start directly with the sectioning of the tis-
sue (Subheading 3.1.2) (see Note 3).
3.1.1 Sample Collection Ideal samples are snap or flash frozen in the gas phase of either
and Snap Freezing liquid nitrogen or isopentane directly after dissection in order to
avoid autolysis and to preserve the molecular composition of the
tissue (see Note 4).
1. In case of nitrogen do not drop the sample directly into the
nitrogen to avoid cracks within the sample. Rather place the
sample if small enough in a tube or vial and cap it tightly or
wrap in aluminum foil and submerge in liquid nitrogen for
flash freezing (see Note 5).
2. Supervise the temperature with a thermometer when using
isopentane to make sure that it does not reach −78 °C (subli-
mation temperature of CO2) (see Note 6).
3. If samples are contaminated with blood it is advisable to briefly
wash the tissue in cold phosphate buffered saline (PBS) prior
to freezing.
4. Place frozen samples in pre-labeled and prechilled containers.
The samples can be stored at −80 °C or used for subsequent
cutting of tissue sections.
134 Birte Beine et al.
3.1.2 Fresh Frozen A cryostat is used to cut the samples into 10 μm thin tissue sections.
Tissue Sectioning
1. Remove the frozen sample from the freezer and allow it to
equilibrate in the cryostat chamber to the desired temperature
for approximately 30 min, depending on the size of the sample
(see Note 7).
2. Fix the sample on the metal holder using optimal cutting tem-
perature (OCT) compound (see Note 8). Complete embed-
ding of the sample in OCT should be avoided since it can lead
to suppressed ion formation and intensity during the
measurement.
3. Cut samples into 10 μm thin tissue slices using the glass anti-
roll plate to prevent upward curling of the cut section. Remove
the glass plate and directly pick up the section from the stage
onto a conductive indium tin oxide (ITO) glass slide (see Note
9). Static attraction will draw the section to adhere quickly to
the warm slide. Folding and curling of the section has to be
avoided during this “thaw mounting” process. Assure com-
plete adhesion of the tissue by placing the back of the slide
shortly on the back of the hand. Perform this step also inside
the cryostat (see Note 10).
4. Let the sample dry inside the cryostat (see Note 11). Then
place the dried sample into a precooled, labeled air tight con-
tainer. Samples can either be processed right away or in that
condition be stored at ultra-low temperature (−80 °C) until
analysis (see Note 12).
3.1.3 Drying 1. Samples taken from the −80 °C freezer are dried in a vacuum
and Washing of the Sample desiccator for about 30 min (see Note 13).
Prior to Enzyme Application 2. The slides are washed for 1 min per step in a series of ethanol
steps 70 %/70 %/100 %. Slightly moving of the slides while in
solution enhances the washing process. This step is primarily
done to remove salts, lipids and other contaminants.
3. After washing dry the tissue once again in the vacuum desicca-
tor for about 30 min.
3.1.4 Setting Reference Reference points for orientation and instrument settings need to
Points and Scanning be placed on the glass slide close to the tissue prior to enzyme and
matrix application. Figure 1 shows and example for reference point
placement.
1. Draw crosses with Tip Ex (water based) in some distance to
the tissue to prevent contamination (see Note 14).
2. Then write letters using a solvent-resistant pen very close to
the tissue (see Fig. 1).
3. Scan the tissue with 2400 dpi resolution using a flatbed scan-
ner. This scan is used for teaching in order to transfer the
Tissue MALDI Mass Spectrometry Imaging (MALDI MSI) of Peptides 135
Fig. 1 Sample slide with reference points. The crosses are made with Tipp-Ex. The letters are written very close
to the tissue using a solvent-resistant pen
3.2 FFPE Tissue The following description is based on the assumption that all steps
Sample Preparation of tissue preparation need to be performed including the fixation
and embedding process of the tissue. If you work for example with
clinical samples in most of the cases these processes are already
done in the clinic and you can skip reading Subheading 3.2.1 (see
Note 3).
3.2.1 Tissue Fixation, 1. After dissection of the tissue transfer the sample immediately
Dehydration and Paraffin into 4 % formaldehyde. To remove blood or other fluids per-
Embedding form a brief washing step with ice cold phosphate buffered
saline (PBS) prior to fixation.
2. A standardized routine fixation, dehydration, and paraffin
embedding in pathology department is performed overnight
in an automated manner (e.g., using the Shandon Pathcentre
Tissue Processor, Thermo Fisher Scientific). A typical program
for tissue fixation is displayed in Table 1 (see Note 15).
3.2.2 FFPE Tissue It is much easier to generate very thin sections (2–6 μm) from
Sectioning FFPE tissue than from cryoconserved tissue. Instead of using a
cryostat at subzero temperatures, FFPE sectioning is performed
using a microtome at room temperature.
1. Preheat the water bath and flattening table for tissue section-
ing to 39 °C.
2. Place the paraffin block with the tissue into the specimen
holder (see Note 16).
3. Cut the sample into 5 μm thin sections and transfer the sec-
tions into the water bath using small paint brushes. Wait until
136 Birte Beine et al.
Table 1
Program for tissue fixation using the Shandon Pathcentre. The protocol was kindly provided by the
pathology department of the University Hospital RWTH Aachen (Germany) directed by Prof. Dr. med.
R. Knüchel-Clarke
3.2.4 Antigen Retrieval In this protocol antigen retrieval is achieved using a heat induced
citric acid antigen retrieval method [14].
1. To prevent bubble formation during the antigen retrieval pro-
cedure prepare a histology glass jar with a rack filled up with
dummy glass slides leaving one space free for your sample.
2. Place the slide into 0.01 M citric acid buffer (pH 6) and put
the glass jar into the microwave. Start the 600 W program for
3 min. Afterwards reduce the power to 90 W for 10 more min-
utes (see Note 19). Avoid strong boiling.
3. To proceed with the antigen retrieval prepare a heat plate to
150 °C and place the glass jar onto the heat plate and incubate
for another 30 min directly after the incubation in the micro-
wave. Let the sample cool down for 15 min on the bench top.
4. To optimize the pH for the subsequent enzymatic digestion,
incubate the sample twice in 0.01 M NH4HCO3 solution once
the citric acid buffer has cooled down. Prepare a histology
glass jar with the solution. Use fresh solution for the second
1 min incubation.
3.2.5 Setting Reference Apply the reference points and scan the tissue as described in
Points and Scanning Subheading 3.1.4.
The tissue preparation continuous with the enzyme applica-
tion and digest of the tissue (see Subheading “Protease Application
and Digest—SunCollect/SunDigest” or “Protease Application
and Digest—ImagePrep”).
3.3 Enzyme This protocol provides the experimental procedures of enzyme and
and Matrix Application matrix application with two different instruments which are the
for Peptide MALDI MSI most common commercially available devices for these applica-
tions; the SunCollect (SunChrom) and the ImagePrep (Bruker
Daltonik GmbH).
There are two main differences comparing both instruments
regarding their spraying mechanism.
The SunCollect device uses pneumatic atomization to spray
enzyme and matrix solution onto the sample. The spray head is
flexible and moves quickly and continuously during the prepara-
tion process over the tissue. The spaying process is regulated by
software and individual spraying sessions can be programmed.
Parameters which can be modified are for example the distance of
138 Birte Beine et al.
the spaying head to the sample, the speed of the spay head while
moving over the tissue and the liquid flow per minute. Those are
only some of the parameters which can be modified for optimizing
the spraying result. For detailed information please read the manu-
factures’ manual.
TheImagePrep device is an automatic spraying device, which
uses vibrational vaporization for spray production. The spray head
is fixed and the spray must be optimized to reach the sample which
must be positioned in a defined distance towards the spray head.
The ImagePrep software is preset with some default methods for
protease and matrix applications and is controlled by a sensor
which is positioned near the tissue. Each method consists of one or
more phases each with individual spraying, incubating or drying
conditions. Parameters of the different phases can be individually
modified as well as the number of spraying phases can be changed.
As before for detailed information please read the manufactures’
manual. Figure 2 provides an overview of the two instruments
together with a detailed view of the actual spraying device.
3.3.1 Protease Enzyme application is a crucial step in tissue preparation for pep-
Application and Digest tide MALDI MSI. There are several parameters which need to be
considered: optimal digestion of the proteins requires a humid
environment but delocalization due to diffusion needs to be mini-
mized to remain the spatial resolution of the peptides. Therefore
the spray for enzyme application needs to produce very small drop-
lets and at the same time provide sufficient humidity for enzyme
activity. In the following section we describe procedures that fulfill
these requirements either using the SunCollect (Subheading
“Protease Application and Digest—SunCollect/SunDigest”) or
the ImagePrep (Subheading “Protease Application and Digest—
ImagePrep”) spraying device.
The enzymatic digestion requires an environment with high
relative humidity (RH) and temperature. When using trypsin, tem-
peratures of 37 °C are the lowest temperature level for sufficient
digestion. In the MALDI MSI community a lot of self-made
instrumentations (digestion chambers) for efficient enzyme diges-
tion are used. In this protocol we provide two different types of
digestion chambers: the SunDigest, a sensor regulated and pro-
grammable automatic device and a self-made digestion chamber
using pipette boxes.
Fig. 2 SunCollect and ImagePrep instrument. In addition to an overview of the two instruments are detailed
views of the spraying devices given. A1–A3 show the SunCollect device. B1–B3 illustrates the ImagePrep
device
3.4 Matrix Matrix application is another critical step of the sample preparation
Application for MALDI MSI. During matrix application it is necessary to use
solvents and water to extract the peptides from the tissue and to
induce the cocrystallization of the matrix and the analyte. Crystal
size formation and extraction efficiency have a large impact on the
MSI result. Thus it is the overall goal during matrix application to
achieve efficient peptide extraction while maintaining good spatial
resolution.
3.4.1 Matrix 1. Place the tissue in the SunCollect instrument. When teaching
Application—SunCollect was applied for enzyme application no further teaching is nec-
essary. Otherwise teach the instrument (see Subheading
“Protease Application and Digest—SunCollect/SunDigest”).
2. Use a 2500 μl syringe for matrix application and fill it with
matrix solution. Place the syringe into the pump and connect
it with the capillary (see Note 29).
3. Use the software to design a spraying program for matrix appli-
cation of your sample (see Note 30). The program for matrix
application differs considerably to the one for enzyme applica-
tion. To initiate crystal formation flow rates of 10–20 μl/min
should be applied. The flow rate per minute increases with the
number of layers. The maximal flow rate to prevent diffusion
should be 40 μl/min.
4. Hold an empty glass slide below the spray and check for a con-
tinuous and fine spray.
5. Remove the glass slide and start the matrix application
program.
6. After matrix application fix the slide into the MTP Slide
Adapter II. Apply a peptide standard in the middle of the steel
frame of the adapter for external calibration: according to the
dried droplet method, mix equal volumes (1 μl) of calibration
standard sample with matrix solution and dry at room
temperature.
7. Clean the capillary by filling the syringe with 70 % ACN. Use
a flow rate of 40 μl/min and let it run for at least 15 min.
While flushing the capillary with ACN clean the spray tip
with the same solvent using a pipette. Let several drops run
down the capillary tip without touching the end of the capil-
lary with the pipette tip.
Continue with the MALDI MSI measurement (see Subheading 4).
3.4.2 Matrix 1. Start off with testing the sprayer. Fill the glass vial 2/3 with
Application—ImagePrep methanol and choose in the display of the ImagePrep Spray
(See Note 31) Head → Adjust → Test Spray. First test 100 % and then 10 %. With
the first setting the spray should reach the round plate on the
opposite site of the chamber whereas the 10 % should at least
142 Birte Beine et al.
reach the end of the placement area (with the sensor in the mid-
dle) for the slide. If not adjust the offset from for example 4–5 %.
2. Remove the methanol residues by wiping the inside of the
chamber with a paper towel.
3. Adjust the power and modulation for the matrix. Load the
prepared matrix into the glass vial and choose adjustment in
the display of the instrument. By default is the power set to 25
and the modulation to 30 for DHB and 15 and 40 for
HCCA. The matrix spray should reach the end of the place-
ment area of the slide. If necessary adjust the power or modu-
lation (see Note 32).
4. Once again clean the inside of the chamber especially the area
above the sensor. Afterwards place the sample slide on top
without covering the sensor with the tissue (see Note 33). If
more than one tissue is on the slide, position the sensor in
between the two samples. Lay a cover glass on top of the slide,
above the sensor.
5. Start the NIVI-Logger (part of the ImagePrep software that
runs on a computer) to monitor the spray cycles.
6. Start the run using the Bruker standard program
DHB_for_digest_nsh01 or HCCA_nsh04.
7. Wait at least until the first 3 cycles of the second phase are
finished and control the profile of the logger. An increase of
the measured curve should be visible if not adjust the spray
power boost (see Note 34).
8. Once the program has finished, stop the logger, take out the
slide and remove the matrix from the short edges of the slides
with 100 % ethanol before fixing the slide into the MTP Slide
Adapter II (see Note 35).
9. Apply a peptide standard in the middle of the steel frame of
the adapter for external calibration: according to the dried
droplet method, mix equal volumes (1 μl) of calibration stan-
dard sample with matrix solution and dry at room
temperature.
10. Clean the inside of the ImagePrep with methanol by running
the corresponding clean program. Afterwards dry the inside of
the chamber with a paper towel.
Continue with the MALDI MSI measurement.
3.5 MALDI MSI The following section describes the peptide MALDI MSI measure-
Measurement ment using a MALDI-TOF mass spectrometer (ultrafleXtreme,
Bruker Daltonik GmbH) controlled by the Bruker software
FlexControl and FlexImaging.
1. Open FlexControl.
Tissue MALDI Mass Spectrometry Imaging (MALDI MSI) of Peptides 143
2. Load the MTP Slide Adapter II with the fixed ITO slide into
the instrument.
3. Open FlexImaging and create a file for your sample, chose the
desired autoXecute method and raster width. Use the Tip Ex
crosses on the slide to coordinate the sample and correlate it to
the previously made scan of the slide.
4. Define the measurement region on the tissue.
5. In FlexControl: choose reflector mode with a set mass range of
m/z 600–4000. Save those settings in a FlexControl method.
The method should also contain a statistical recalibration of
the spectra. For this purpose integrate the flexAnalysis method
ImageID_StatRecal.FAMSMethod [15].
6. Load the generated FlexControl method and measure the
external calibrants on the target frame. The error of each pep-
tide should not exceed 20 ppm. Apply the calibration and save
the method.
7. Finally determine the necessary laser power for the tissue. Save
the method and start the run using FlexImaging.
3.6 HE-Staining The unique characteristic of MALDI MSI compared to other mass
of the Analyzed Tissue spectrometry based analyses is the ability to maintain the spatial
resolution of the analyzed molecules. It is thus possible to compare
the MS data directly to the histological features, which are visible
after staining the tissue. Standard histological staining protocols
can be used to stain the tissue after the measurement. Check which
kind of staining is the best to answer your questions. In the follow-
ing we provide a fast hematoxylin–eosin (HE) staining which is
suitable for many tissue types. Hematoxylin stains nuclei blue,
while eosin results in a pink staining of the connective tissue.
1. Remove the matrix by washing the tissue in 70 % ethanol. For
this purpose move the slide up and down for several times.
2. Incubate the tissue in Meyers’ hematoxylin solution for 1 min
(see Note 36).
3. Incubate the tissue in tab water for 4 min. This step causes a
pH shift and intensifies the staining of hematoxylin.
4. Dip the slide twice into 0.25 % eosin solution.
5. Dip the slide twice into distilled water.
6. Repeat dipping the slide in 70 % ethanol.
7. Incubate the slide for 20 s each in 96 %, 100 %, and again 100
% ethanol.
8. For complete dehydration incubate the slide 5 min in 100 %
xylene. Then dry the slide completely.
9. Put one drop of mounting medium on the tissue and place a
cover slip on top. Try to avoid air bubbles since they will dete-
144 Birte Beine et al.
3.7 Data Analysis For the analysis of the MALDI MSI data we use SCiLS Lab. This
Using SCiLS Lab software allows you to analyze several tissues simultaneously, which
is important when comparing different samples in one study or
comparing different methods.
1. Import the mis-files from FlexImaging into SCiLS Lab. You
can arrange your samples in the way you want them to be dis-
played (rotation is possible as well). SCiLS Lab converts the
data into a so-called h5-file. The generated h5-file contains all
the data you need for further analysis (see Note 37).
2. In SCiLS Lab you have several options for data analysis. To
gain a first impression it is a good starting point to run the
“Segmentation pipeline” and let the software perform all neces-
sary steps automatically. This pipeline contains preprocessing
steps such as baseline removal, normalization and the genera-
tion of an overview spectrum for all the samples in your data
set. Furthermore peak picking, alignment, spatial denoising
and segmentation are performed.
3. To evaluate different data sets statistical and structure analysis
are suitable tools. The software provides a large amount of
analysis tools so that only some selected examples can be pro-
vided. For statistical analysis two different component analyses
can be performed in SCiLS Lab: PCA (principal component
analysis) and pLSA (probabilistic latent semantic analysis)
whereby the latter one can also be seen as a form of structure
analysis (see Note 38).
4. When you use a model tissue like rat brain it is possible to
judge the quality of your different methods using an additional
strategy: counting the number of m/z values, which show a
structured image for a well-known and highly abundant pro-
tein, e.g., myelin basic protein (MBP) in rat brain (see Note
39). For this strategy you have to first perform an in-silico
digest of MBP with trypsin using for example MS-Digest from
the ProteinProspector website (http://prospector.ucsf.edu/
prospector/cgi-bin/msform.cgi?form=msdigest) to obtain a
list of MBP specific peptides (see Note 40). For the in-silico
digest defined settings must be used: maximal number of
missed cleavages 1, oxidation of methionine as variable modi-
fication, peptide mass range 800–4000 Da and MALDI-TOF/
TOF as instrument type. In case of MBP this results in a list of
45 peptides respectively 45 monoisotopic m/z values.
Tissue MALDI Mass Spectrometry Imaging (MALDI MSI) of Peptides 145
This list can be imported into the h5 data set (see Note 41). It
is then possible to visualize the images of each individual m/z
value allowing for example a peak shift of ±0.3 Da. It is now
possible to count the number of visible MPB structures and
compare them between you different experiments.
4 Notes
10. It has been reported that ITO slides modified with poly-L-
lysine improved the adherence of the cells to the substrate
[16].
11. Do not place the slide on the metal ground of the cryostat
while letting the cut tissue section dry inside the instrument.
The direct contact with the metal will lead to freeze artifacts.
We therefore recommend placing a vacuum formed slide
holder inside the cryostat to overcome this problem.
12. In general it is advisable to use flash frozen samples right away
for imaging rather than storing them long term. The same
applies for the cut tissue sections on ITO glass slides. In case
of long-term storage we recommend to put each slide in an
individual container even though there might be space for
more but by doing so we avoid temperature changes when
opening the box to remove single slides. We found
LockMailer™ with screw caps to be the best storage device for
that purpose.
13. Drying of the samples is also possible in a regular desiccator.
14. Use only little amount of Tip Ex to maintain an even surface
otherwise you will have problems to cover the subsequently
HE stained sample with a cover glass after MALDI MSI
measurement.
15. Fixation time can be reduced to ~30 min in case of small
biopsy samples whereas some bloody or fatty tissues as well as
some fetal tissues require longer fixation times. Make sure to
check the optimal conditions for your tissue sample prior to
fixation.
16. We recommend removing surplus paraffin wax at the edges of
the block to minimize the sectioning area.
17. If the flattening of the tissue is not sufficient after 5–10 min,
temperature needs to be optimized.
18. We recommend using a histology glass jar with molded glass
cover in which the slides are in an upright position.
19. During this process it is necessary to constantly check for bub-
ble formation and to remove bubbles by simply pulling the
slide holder upwards.
20. Put the syringe into a plastic bag to avoid contamination.
21. To find the perfect enzyme spray settings for your tissue you
should test one to three slides before you prepare a real sam-
ple. Test enzyme spray settings and digestion settings and
compare the MALDI MSI results in regard to intensities, spa-
tial resolution and digest efficiency.
22. We spray for at least 1 min before starting the program.
Tissue MALDI Mass Spectrometry Imaging (MALDI MSI) of Peptides 147
Fig. 3 Overview of different crystal shapes and distributions using the SunCollect spraying device. HCCA (7
mg/ml, 50 % ACN, 0.2 % TFA) matrix was applied onto rat brain tissue sections only the parameter of the z
position during matrix spray was changed (see Note 29). (a) and (b) display images with different magnitude
(×20 and ×40)
Acknowledgement
We would like to thank Julian Elm for his skilful technical assis-
tance during the method development process. We thank Dennis
Trede for excellent support dealing with SCiLS Lab. Furthermore
150 Birte Beine et al.
References
1. Castaing R, Slodzian G (1962) Microanalyse 10. Delvolve AM, Woods AS (2011) Optimization
par emission ionique secondaire. J Microscopie of automated matrix deposition for biomolec-
1:395–410 ular mapping using a spotter. J Mass Spectrom
2. Trim PJ, Djidja MC, Muharib T et al (2012) 46(10):1046–1050. doi:10.1002/jms.1986
Instrumentation and software for mass spec- 11. Yang J, Caprioli RM (2011) Matrix sublima-
trometry imaging—making the most of what tion/recrystallization for imaging proteins by
you’ve got. J Proteomics 75(16):4931–4940. mass spectrometry at high spatial resolution.
doi:10.1016/j.jprot.2012.07.016 Anal Chem 83(14):5728–5734. doi:10.1021/
3. Groseclose MR, Andersson M, Hardesty WM ac200998a
et al (2007) Identification of proteins directly 12. Jaskolla TW, Karas M, Roth U et al (2009)
from tissue: in situ tryptic digestions coupled Comparison between vacuum sublimed matri-
with imaging mass spectrometry. J Mass ces and conventional dried droplet preparation
Spectrom 42(2):254–262. doi:10.1002/ in MALDI-TOF mass spectrometry. J Am Soc
jms.1177 Mass Spectrom 20(6):1104–1114.
4. Monroe EB, Annangudi SP, Hatcher NG et al doi:10.1016/j.jasms.2009.02.010
(2008) SIMS and MALDI MS imaging of the 13. Ait-Belkacem R, Sellami L, Villard C et al
spinal cord. Proteomics 8(18):3746–3754. (2012) Mass spectrometry imaging is moving
doi:10.1002/pmic.200800127 toward drug protein co-localization. Trends
5. Murphy RC, Hankin JA, Barkley RM (2009) Biotechnol 30(9):466–474. doi:10.1016/j.
Imaging of lipid species by MALDI mass spec- tibtech.2012.05.006
trometry. J Lipid Res 50 Suppl:S317–S322. 14. Gustafsson OJ, Eddes JS, Meding S et al
doi:10.1194/jlr.R800051-JLR200 (2013) Matrix-assisted laser desorption/ion-
6. Khatib-Shahidi S, Andersson M, Herman JL ization imaging protocol for in situ character-
et al (2006) Direct molecular analysis of whole- ization of tryptic peptide identity and
body animal tissue sections by imaging MALDI distribution in formalin-fixed tissue. Rapid
mass spectrometry. Anal Chem 78(18):6448– Commun Mass Spectrom 27(6):655–670.
6456. doi:10.1021/ac060788p doi:10.1002/rcm.6488
7. Diehl HC, Beine B, Elm J et al (2015) The 15. Mann M, Hojrup P, Roepstorff P (1993) Use
challenge of on-tissue digestion for MALDI- of mass spectrometric molecular weight infor-
MSI—a comparison of different protocols to mation to identify proteins in sequence data-
improve imaging experiments. Anal Bioanal bases. Biol Mass Spectrom 22(6):338–345.
Chem. doi:10.1007/s00216-014-8345-z doi:10.1002/bms.1200220605
8. Oezdemir RF, Gaisa NT, Lindemann-Docter K 16. Zavalin A, Todd EM, Rawhouser PD et al
et al (2012) Proteomic tissue profiling for the (2012) Direct imaging of single cells and tissue
improvement of grading of noninvasive papillary at sub-cellular spatial resolution using transmis-
urothelial neoplasia. Clin Biochem 45(1–2):7–11. sion geometry MALDI MS. J Mass Spectrom
doi:10.1016/j.clinbiochem.2011.09.013 47(11):1473–1481. doi:10.1002/jms.3108
9. Casadonte R, Caprioli RM (2011) Proteomic 17. Hanselmann M, Kirchner M, Renard BY et al
analysis of formalin-fixed paraffin-embedded (2008) Concise representation of mass spec-
tissue by MALDI imaging mass spectrometry. trometry images by probabilistic latent seman-
Nat Protoc 6(11):1695–1709. doi:10.1038/ tic analysis. Anal Chem 80(24):9649–9658.
nprot.2011.388 doi:10.1021/ac801303x
Chapter 11
Abstract
Ethyl esterification is a technique for the chemical modification of sialylated glycans, leading to enhanced
stability when performing matrix-assisted laser desorption/ionization (MALDI)-mass spectrometry (MS),
as well as allowing the efficient detection of both sialylated and non-sialylated glycans in positive ion mode.
In addition, the method shows specific reaction products for α2,3- and α2,6-linked sialic acids, leading to
an MS distinguishable mass difference. Here, we describe the ethyl esterification protocol for 96 glycan
samples, including enzymatic N-glycan release, the aforementioned ethyl esterification, glycan enrichment,
MALDI target preparation, and the MS(/MS) measurement.
1 Introduction
Jörg Reinders (ed.), Proteomics in Systems Biology: Methods and Protocols, Methods in Molecular Biology, vol. 1394,
DOI 10.1007/978-1-4939-3341-9_11, © Springer Science+Business Media New York 2016
151
152 Karli R. Reiding et al.
2 Materials
2.3 Sepharose HILIC 1. Sepharose bead slurry: 25 % Cl-4B Sepharose beads (45–
Solid Phase Extraction 165 μm, GE Healthcare, Uppsala, Sweden) in 20 % ethanol.
Transfer 500 μL beads to a 2 mL tube. Spin down the beads
and remove the supernatant. Add 1.5 mL 20 % ethanol. Make
fresh before using.
2. 85 % acetonitrile (ACN). Measure 150 mL water in a 50 mL
graduated cylinder and add to a 1 L screw-cap glass bottle.
Using a 1 L graduated cylinder, measure 850 mL ACN and
add to the glass bottle. Store at 4 °C, but bring to room
temperature before usage.
154 Karli R. Reiding et al.
3 Methods
3.1 PNGase F 1. Add 4 μL 2 % SDS to each well of a 96-well plate (see Note 8).
N-Glycan Release 2. Add 2 μL glycoprotein sample to each well and mix briefly by
pipetting up and down (see Notes 9 and 10).
3. Cover the plate with adhesive tape to prevent evaporation (see
Note 11).
4. Incubate the plate for 10 min in a 60 °C oven to denature the
proteins (see Note 12).
5. Remove the plate from the oven, and allow it to return to
room temperature (±5 min).
6. Carefully remove adhesive tape, making sure any condensation
on the tape does not cause cross-contamination of the
samples.
7. Add 4 μL release solution to each well (see Note 8).
Ethyl Esterification for MALDI-MS Analysis of Protein Glycosylation 155
3.3 Sepharose HILIC 1. Carefully remove the adhesive tape and discard it (see Note 10).
Solid Phase Extraction 2. Add 20 μL pure ACN to each sample and mix briefly by pipet-
ting up and down (see Notes 9 and 15).
3. Place a 96-well filter plate on a vacuum manifold equipped
with a liquid collection box.
4. Transfer 20 μL Sepharose bead slurry to each well of the
96-well filter plate. The Sepharose will sediment rapidly, keep
it thoroughly suspended by mixing the solution before each
pipetting step (see Note 9).
5. Apply low vacuum until the liquid has flown through (see Note
16). However, make sure the filters and beads remain moist
(see Note 17). Any flow-through building up in the collection
box can best be discarded after steps 8 and 15 when the filter
plate is removed from the vacuum manifold.
6. Add 100 μL water to each well, then apply low vacuum until
the liquid has flown through (see Note 9). Repeat two addi-
tional times.
7. Add 100 μL of 85 % ACN to each well, then apply low vacuum
until the liquid has flown through. Repeat two additional times
(see Note 9).
8. Press the filter plate briefly on lint-free paper to remove any
solution adhering to the bottom, then place the filter plate on
an empty 96-well plate (see Note 18).
9. Briefly mix the ethyl esterified samples, then transfer them to
the filter plate (±38 μL). Apply the samples directly to the
beads (see Note 9).
156 Karli R. Reiding et al.
10. Place the filter plate (on the 96-well plate) on a shaking
platform, and incubate for 5 min at maximum velocity (see
Note 19).
11. Transfer the filter plate back to the vacuum manifold and apply
low vacuum until the liquid has flown through.
12. Apply 100 μL 85 % ACN 0.1 % TFA to each well, then apply
low vacuum until the liquid has flown through. Repeat two
additional times (see Notes 9 and 20).
13. Apply 100 μL 85 % ACN to each well, then apply low vacuum
until the liquid has flown through. Repeat two additional times
(see Note 9).
14. Apply additional vacuum to remove residual fluid from the fil-
ter plate.
15. Press the filter plate briefly on lint-free paper to remove solution
adhering to the bottom, then place it on a clean (new) 96-well
plate (see Note 21).
16. Add 30 μL water to each well for elution (see Note 9).
17. Place the stacked plates (filter plate on the 96-well plate) on a
shaking platform, and incubate for 5 min at maximum velocity
(see Note 22).
18. Transfer the stacked plates to a centrifuge equipped with plate-
holder inserts. Use a counter-weight if only one plate will be
centrifuged (see Note 23).
19. Centrifuge for 1 min at 200 × g (see Note 24).
20. The enriched ethyl esterified N-glycans are now in the flow-through.
Do not store the samples before measurement, as this may over
time lead to degradation of the lactonized reaction products.
3.5 MALDI-MS 1. Measure the samples in positive ion mode (see Note 28). When
Measurement preforming TOF-MS the resolution can, due to the derivatiza-
tion step, be increased by reflectron mode measurement (as
compared to linear mode measurement) without leading to
visible metastable decay of the sialylated glycan species.
2. Before sample measurement, use a standard for calibration (see
Note 29).
Ethyl Esterification for MALDI-MS Analysis of Protein Glycosylation 157
4 Notes
Table 1
Potential satellite signals relative to expected glycan peaks. Whereas amidation and incomplete
esterification affect only the sialic acids, and therefore change the relative distribution of the
analyzed glycans, the reducing end losses and salt variation have an impact on all species and
preserve the relative distribution. In addition to the displayed masses, the presence of peaks in the
spectrum with significantly lower resolution (metastable peaks) may also indicate incomplete
esterification or too high laser power settings
Table 2
The most commonly observed structurally informative mass differences from a given precursor ion.
Unless multiple fucoses are present on a structure, assigning a fucose to an antenna requires a
−221.09 signal to prove the lack of core fucosylation. Observed losses can be verified at the lower
m/z range of the spectrum as [M + Na]+ ions
Acknowledgements
References
1. Wormald MR, Dwek RA (1999) Glycoproteins: of plasma protein fucosylation. PLoS
glycan presentation and protein-fold stability. Genet 6(12), e1001256. doi:10.1371/jour-
Structure 7(7):R155–R160 nal.pgen.1001256
2. Crocker PR, Paulson JC, Varki A (2007) 6. Vanhooren V, Desmyter L, Liu XE et al (2007)
Siglecs and their roles in the immune N-glycomic changes in serum proteins during
system. Nat Rev Immunol 7(4):255–266. doi: human aging. Rejuvenation Res 10(4):521–
10.1038/nri2056 531a. doi:10.1089/rej.2007.0556
3. Alessandri L, Ouellette D, Acquah A et al 7. Reis CA, Osorio H, Silva L et al (2010)
(2012) Increased serum clearance of oligo- Alterations in glycosylation as biomarkers for
mannose species present on a human IgG1 cancer detection. J Clin Pathol 63(4):322–
molecule. MAbs 4(4):509–520. doi:10.4161/ 329. doi:10.1136/jcp.2009.071035
mabs.20450 8. Knezevic A, Gornik O, Polasek O et al (2010)
4. Moremen KW, Tiemeyer M, Nairn AV (2012) Effects of aging, body mass index, plasma lipid
Vertebrate protein glycosylation: diversity, syn- profiles, and smoking on human plasma
thesis and function. Nat Rev Mol Cell Biol N-glycans. Glycobiology 20(8):959–969.
13(7):448–462. doi:10.1038/nrm3383 doi:10.1093/glycob/cwq051
5. Lauc G, Essafi A, Huffman JE et al (2010) 9. Adamczyk B, Tharmalingam T, Rudd PM
Genomics meets glycomics—the first (2012) Glycans as cancer biomarkers. Biochim
GWAS study of human N-Glycome iden- Biophys Acta 1820(9):1347–1353. doi:10.
tifies HNF1alpha as a master regulator 1016/j.bbagen.2011.12.001
162 Karli R. Reiding et al.
Abstract
Zwitterionic hydrophilic interaction chromatography (ZIC-HILIC) solid-phase extraction (SPE) com-
bined with direct-infusion nanoESI mass spectrometry (MS) and tandem MS/MS is a well-suited method
for the analysis of protein N-glycosylation. A site-specific characterization of N-glycopeptides is achieved
by the combination of proteolytic digestions employing unspecific proteases, glycopeptide enrichment by
use of ZIC-HILIC SPE, and subsequent mass spectrometric analysis. The use of thermolysin or a mixture
of trypsin and chymotrypsin leads per se to a mass-based separation, that is, small nonglycosylated peptides
and almost exclusively glycopeptides at higher m/z values. As a result of their higher hydrophilicity
N-glycopeptides comprising short peptide backbones are preferably accumulated by the ZIC-HILIC-
based separation procedure. By employing this approach complications associated with low ionization
efficiencies of N-glycopeptides resulting from signal suppression in the presence of highly abundant non-
glycosylated peptides can be largely reduced. Here, we describe a simple protocol aimed at the enrichment
of N-glycopeptides derived from in-solution and in-gel digestions of SDS-PAGE-separated glycoproteins
preceding mass spectrometric analysis.
Key words N-glycosylation, Glycopeptides, In-solution digestion, In-gel digestion, NanoESI MS,
CID
1 Introduction
*The first two authors (Pohlentz and Marx) contributed equally and share the first authorship.
Jörg Reinders (ed.), Proteomics in Systems Biology: Methods and Protocols, Methods in Molecular Biology, vol. 1394,
DOI 10.1007/978-1-4939-3341-9_12, © Springer Science+Business Media New York 2016
163
164 Gottfried Pohlentz et al.
Fig. 1 Strategy for analysis of glycopeptides derived from in-solution and in-gel
digestions of glycoproteins
ZIC-HILIC Enrichment of Proteolytic N-Glycopeptides 167
Fig. 2 Direct-infusion nanoESI mass spectra obtained from tryptic/chymotryptic digestions of bovine IgG prep-
arations analyzed by different methods: IS = In-solution digest, IG = In-gel digest, ZT = ZipTip desalting,
ZH = ZIC-HILIC enrichment. The code used for peak annotation corresponds to the glycan structures listed in
Table 2 (column 1)—first number, the glycosylation site (see Table 1, columns 1 and 2)—letter, and the peptide
backbone (see Table 1, columns 1 and 3)—indices
Table 1
168
Synopsis of detected glycopeptides derived from in-solution and in-gel tryptic/chymotryptic digestions of bovine IgG (in-gel digestion of heavy chain)
after enrichment (for details see Table 2)
Table 2
Detected glycan structures in glycopeptides derived from in-solution and in-gel tryptic/chymotryptic
digestions of bovine IgG (in-gel digestion of heavy chain)
Bovine IgG
No./glycosylation sitea Glycan IS IS + ZH IG + ZT IG + ZH IG + ZT + ZH
1
1 N183 − + − + +
2
N183 − − − − −
1
2 N183 − + − − +
2
N183 − − − − −
1
3 N183 − + − − −
2
N183 − − − − −
1
4 N183 − + − − +
2
N183 − − − − −
1
5 N183 − + − − +
2
N183 − − − − −
1
6 N183 − + − + +
2
N183 − − − − −
1
7 N183 + + − + +
2
N183 − + − − +
1
8 N183 + + + + +
2
N183 − + − − +
1
9 N183 + + + + +
2
N183 − + − + +
1
10 N183 + + + + +
2
N183 + + + + +
1
11 N183 + + + + +
2
N183 − + − − +
1
12 N183 − + − − +
2
N183 − + − − −
1
13 N183 − + − − +
2
N183 − + − − −
1
14 N183 − + − − +
2
N183 − + − − +
2 Materials
2.5 Desalting 1. Solid-phase extraction C18-RP tips: ZipTip C18, Tip Size
of Extracted P10, (Merck Chemicals GmbH, Schwalbach, Germany).
Proteolytic Peptides 2. Wetting solution: Acetonitrile.
3. Binding solution: Trifluoroacetic acid (0.1 %).
4. Equilibration solution: Trifluoroacetic acid (0.1 %).
5. Wash solution: Trifluoroacetic acid (0.1 %).
6. Elution solution 1: Trifluoroacetic acid (0.1 %)/acetonitrile
(50/50).
7. Elution solution 2: Trifluoroacetic acid (0.1 %)/acetonitrile
(20/80).
8. Elution solution 3: Acetonitrile.
3 Methods
3.2 Desalting 1. Allow excess packing buffer to drain from the NAP™ 5 column
by Size-Exclusion by gravity to the top of the gel bed.
Chromatography 2. Apply 2.5 ml of 50 mM ammonium bicarbonate and allow the
(Sephadex G-25) buffer to drain out by gravity to the top of the gel bed. Repeat
this step four times.
3. Carefully add the sample obtained under Subheading 3.1 to
the column and allow the sample to penetrate the gel bed
completely.
4. Apply 500 μl of 50 mM ammonium bicarbonate and allow the
buffer to drain out by gravity to the top of the gel bed. The
flow through is dicarded.
5. Place an appropriate collection tube under the column, apply
500 μl of 50 mM ammonium bicarbonate, and allow the buf-
fer to drain out by gravity to the top of the gel bed.
6. The sample is dried by removal of the solvent in vacuo by use
of a centrifugal evaporator.
75 μl) to the column, and allow the sample to penetrate the gel
bed completely. Centrifuge the assembly for 4 min at 1000 × g.
7. The sample is dried by removal of the solvent in vacuo by use
of a centrifugal evaporator.
3.4 In-Solution 1. For in-solution digestion 100 pmol of either the reduced and
Digestion alkylated or the untreated glycoprotein is dissolved in 20 μl 10
mM ammonium bicarbonate (see Note 3).
2. Shake the mixture for 7 min at 95 °C (see Note 4) and chill
tubes to room temperature.
3. Add 1 μl of the protease solution and incubate the reaction
mixture overnight at 37 °C in a shaker (750 rpm) (see Note 5).
If thermolysin is employed as protease incubate at 65 °C in a
shaker (750 rpm).
4. The sample is dried by removal of the solvent in vacuo by use
of a centrifugal evaporator.
5. Add 50 μl water and dry in vacuo by use of a centrifugal evapo-
rator. Repeat this step at least once.
3.5 In-Gel Digestion If reduced and alkylated (glyco-)proteins have been separated by
use of one-dimensional or two-dimensional sodium dodecyl
sulfate-polyacrylamide gel electrophoresis (SDS-PAGE) and visu-
alized by either silver or Coomassie® brilliant blue staining glyco-
peptides are formed by in-gel digestion (see Note 3). The in-gel
digestion procedure follows the method described by Shevchenko
et al. [27].
1. Excise protein bands or spots by use of a clean scalpel.
2. Cut bands into small pieces (see Note 6) and transfer gel cubes
into a reaction tube of appropriate volume (0.5 or 1.5 ml).
3. Add 100 μl of 100 mM ammonium bicarbonate/acetonitrile
(50/50) and shake for 30 min. Subsequently, remove destain-
ing solution.
4. Add 500 μl of acetonitrile and shake for 10 min until gel pieces
shrink and become opaque. Remove supernatant and dry gel
pieces in vacuo by use of a centrifugal evaporator.
5. Add 30 μl of 10 mM ammonium bicarbonate and 10 μl of pro-
tease solution and incubate gel pieces for 30 min on an ice bath.
6. If digestion buffer solution is absorbed entirely add an appro-
priate amount of 10 mM ammonium bicarbonate and protease
solution (3/1, v/v) to cover gel pieces completely. Incubate
reaction mixture for 30 min on an ice bath (see Note 7).
7. Add an appropriate amount of 10 mM ammonium bicarbonate
to cover gel pieces completely and incubate the reaction mixture
overnight at 37 °C in a shaker (750 rpm). If thermolysin is
employed as protease incubate at 65 °C in a shaker (750 rpm).
ZIC-HILIC Enrichment of Proteolytic N-Glycopeptides 175
4 Notes
References
peptide and glycoprotein enrichment. Analyst 24. Reissner C, Stahn J, Breuer D et al (2014)
139:688–704 Dystroglycan binding to α-neurexin competes
18. Huang BY, Yang CK, Liu CP et al (2014) with neurexophilin-1 and neuroligin in the
Stationary phases for the enrichment of glyco- brain. J Biol Chem 289:27585–27603
proteins and glycopeptides. Electrophoresis 25. Gnanesh Kumar BS, Pohlentz G, Mormann M
35:2091–2107 et al (2013) Characterization of α-mannosidase
19. Hägglund P, Bunkenborg J, Elortza F et al from Dolichos lablab seeds: partial amino acid
(2004) A new strategy for identification of sequencing and N-glycan analysis. Protein
N-glycosylated proteins and unambiguous Expr Purif 89:7–15
assignment of their glycosylation sites using 26. Gnanesh Kumar BS, Pohlentz G, Schulte M
HILIC enrichment and partial deglycosylation. et al (2014) N-glycan analysis of mannose/
J Proteome Res 3:556–566 glucose specific lectin from Dolichos lablab
20. Thaysen-Andersen M, Højrup P (2006) seeds. Int J Biol Macromol 69:400–407
Enrichment and characterization of glycopep- 27. Shevchenko A, Tomas H, Havlis J et al (2006)
tides from gel-separated glycoproteins. Am In-gel digestion for mass spectrometric charac-
Biotechnol Lab 24:14–17 terization of proteins and proteomes. Nat
21. Picariello G, Ferranti P, Mamone G et al (2008) Protoc 1:2856–2860
Identification of N-linked glycoproteins in 28. Domon B, Costello CE (1988) A systematic
human milk by hydrophilic interaction liquid nomenclature for carbohydrate fragmentations
chromatography and mass spectrometry. in FAB-MS MS spectra of glycoconjugates.
Proteomics 8:3833–3847 Glycoconjugate J 5:397–409
22. Gnanesh Kumar BS, Pohlentz G, Schulte M 29. Mormann M, Eble J, Schwoeppe C et al (2008)
et al (2014) Jack bean α-mannosidase: amino Fragmentation of intra-peptide and inter-
acid sequencing and N-glycosylation analysis of peptide disulfide bonds of proteolytic peptides
a valuable glycomics tool. Glycobiology by nanoESI collision-induced dissociation.
24:252–261 Anal Bioanal Chem 392:831–838
23. Sharma A, Pohlentz G, Bobbili KB et al (2013) 30. Consortium for Functional Glycomics. CFG
The sequence and structure of snake gourd functionalglycomicsgateway. http://www.
(Trichosanthes anguina) seed lectin, a three- functionalglycomics.org/static/consortium/
chain nontoxic homologue of type II RIPs. Acta CFGnomenclature.pdf. Accessed 20 July
Crystallogr D Biol Crystallogr 69:1493–1503 2012
Chapter 13
Abstract
Identification of protein-protein interactions can be a critical step in understanding the function and
regulation of a particular protein and for exploring intracellular signaling cascades. Affinity purification
coupled to mass spectrometry (APMS) is a powerful method for isolating and characterizing protein
complexes. This approach involves the tagging and subsequent enrichment of a protein of interest along
with any stably associated proteins that bind to it, followed by the identification of the interacting proteins
using mass spectrometry. The protocol described here offers a quick and simple method for routine sample
preparation for APMS analysis of suitably tagged human cell lines.
Key words Affinity purification-mass spectrometry, Cell signaling pathway, Liquid chromatography-mass
spectrometry
1 Introduction
Jörg Reinders (ed.), Proteomics in Systems Biology: Methods and Protocols, Methods in Molecular Biology, vol. 1394,
DOI 10.1007/978-1-4939-3341-9_13, © Springer Science+Business Media New York 2016
181
182 Julian H.M. Kwan and Andrew Emili
2 Materials
3 Methods
4 Notes
References
1. Butland G, Peregrín-Alvarez JM, Li J et al tive map of physical and genetic interactions
(2005) Interaction network containing con- mediated by the hsp90 chaperone. Cell
served and essential protein complexes in 120:715–727. doi:10.1016/j.cell.2004.12.024
Escherichia coli. Nature 433:531–537. 3. Gavin A-C, Aloy P, Grandi P et al (2006)
doi:10.1038/nature03239 Proteome survey reveals modularity of the
2. Zhao R, Davey M, Hsu Y-C et al (2005) yeast cell machinery. Nature 440:631–636.
Navigating the chaperone network: an integra- doi:10.1038/nature04532
Simple and Effective Affinity Purification Procedures for Mass Spectrometry-Based… 187
Abstract
The mammalian immune system has evolved to respond to pathogenic, environmental, and cellular
changes in order to maintain the health of the host. These responses include the comparatively primitive
innate immune response, which represents a rapid and relatively nonspecific reaction to challenge by
pathogens and the more complex cellular adaptive immune response. This adaptive response evolves with
the pathogenic challenge, involves the cross talk of several cell types, and is highly specific to the pathogen
due to the liberation of peptide antigens and their presentation on the surface of affected cells. Together
these two forms of immunity provide a surveillance mechanism for the system-wide scrutiny of cellular
function, environment, and health. As such the immune system is best understood at a systems biology
level, and studies that combine gene expression, protein expression, and liberation of peptides for antigen
presentation can be combined to provide a detailed understanding of immunity. This chapter details our
experience in identifying peptide antigens and combining this information with more traditional pro-
teomics approaches to understand the generation of immune responses on a holistic level.
Key words Major histocompatibility complex, Human leukocyte antigens, Peptide ligands, Mass
spectrometry, Antigen presentation
1 Introduction
Jörg Reinders (ed.), Proteomics in Systems Biology: Methods and Protocols, Methods in Molecular Biology, vol. 1394,
DOI 10.1007/978-1-4939-3341-9_14, © Springer Science+Business Media New York 2016
189
190 Nadine L. Dudek et al.
Table 1
Commonly used monoclonal antibodies for MHC-peptide immunoaffinity chromatography
Fig. 1 A systems approach to antigen presentation using mass spectrometry. In this workflow, cells are infected
with virus in vitro. At various time points, cells are harvested and lysed. A sample of lysate is taken and subjected
to tryptic digestion. The remaining lysate is used to affinity purify MHC peptide complexes. Both tryptic peptides
and non-tryptic MHC peptides are subjected to RP-HPLC before mass spectrometric interrogation. Mass spectro-
metric analysis involves both a global discovery approach of LC-MS/MS and the targeted method of LC-MRM,
where a set of known peptides are detected and quantified. In this workflow it is possible to simultaneously
quantify the presentation of virus or host peptide-MHC complexes, and the levels of their source antigens at
multiple times during infection to develop a comprehensive picture of antigen presentation
2 Materials
2.1 Generation All solutions for cross-linking with the exception of triethanolamine
of MHC Eluate and DMP should be filtered through a 0.2 μM filter.
2.1.1 Preparation 1. Phosphate-buffered saline (PBS): 137 mM NaCl, 2.7 mM
of Cross-Linked KCl, 10 mM Na2HPO4, and 2 mM KH2PO4, pH 7.4.
Immunoaffinity Column 2. Purified monoclonal antibody (mAb) at 1–10 mg/ml in PBS:
Ideally the mAb should only recognize the class I or class II
allele of interest, although affinity and specificity issues fre-
quently require a compromise (Table 1, see Note 1).
3. Suitable column (e.g., disposable plastic Econo-Column from
BioRad).
4. Protein A resin (e.g., Repligen CaptivA™ PriMAB).
5. Borate wash buffer: 0.05 M Borate buffer pH 8.0. For 100 ml
of buffer, add 3.97 ml of 0.1 N NaOH to 50 ml of 0.1 M boric
acid/0.1 M KCl stock solution and make up to 100 ml with
MS-grade H2O.
6. Protein A wash buffer: 0.2 M Triethanolamine, pH 8.2 at
RT. Prepare this solution fresh and pH just prior to use.
7. Dimethyl pimelimidate (DMP) cross-linker: 40 mM DMP-
2HCl in 0.2 M triethanolamine pH 8.3. Prepare DMP by dis-
solving 250 mg DMP-2HCl (Sigma) in 22 ml 0.2 M
triethanolamine pH 8.2. Adjust pH to 8.3 with NaOH, and
bring to 24.1 ml. Prepare this solution fresh and pH just prior
to use (see Note 2).
8. Termination buffer: Ice-cold 0.2 M Tris, pH 8.0.
9. Stripping buffer: 0.1 M Citrate, pH 3.0.
2.1.2 Generation MHC-bound peptides may be affinity purified from whole tissue,
of Cell Lysate isolated primary cells, or transformed cells grown in culture (see
Note 3). The amount of material required is dependent on the
downstream application (LC-MS/MS vs. LC-MRM) and will be
highly dependent on the expression levels of MHC on the cells
contained within the sample (see Note 4). Transformed cells grown
in culture are the simplest sample type for MHC/peptide isolation
with cell numbers ranging between 5 × 107 and 1 × 109 per isola-
tion. Cells may be expanded, washed in PBS, and the pellets snap
frozen in liquid nitrogen for storage at −80 °C for up to 6 months.
1. 2× Lysis buffer: 0.5 % NP-40 (IGEPAL 630 from Sigma is the
equivalent), 50 mM Tris, pH 8.0 (from 1 M stock solution),
150 mM NaCl, protease inhibitor cocktail (cOmplete protease
inhibitor from Roche or equivalent, should be made up fresh
each time) in MS-grade H2O. Prepare lysis buffer just prior to
use and keep on ice.
194 Nadine L. Dudek et al.
2.1.3 Immunoaffinity MHC class I and II molecules can be eluted from the same sample
Purification of MHC Class I/ by using tandem columns. For human cell lines, we routinely pass
Class II Molecules the cell lysate through a class I column, followed sequentially by a
column specific for HLA DR, then HLA DQ, and finally HLA DP.
1. mAb cross-linked protein A resin from Subheading 2.1.1.
2. Pre-column (non-cross-linked protein A sepharose in suitable
column): The bed volume should be half that of the cross-
linked column; that is, for a 1 ml protein A-mAb column, use
a 0.5 ml pre-column.
3. Pepstatin A: 1 mg/ml stock in isopropanol, aliquot, and store
at −20 °C.
4. PMSF: 0.1 M stock in absolute ethanol, aliquot, and store at
−20 °C.
5. Wash buffer 1: 0.005 % IGEPAL 630, 50 mM Tris, pH 8.0,
150 mM NaCl, 5 mM EDTA, 100 μM PMSF, 1 μg/ml pep-
statin A in MS-grade H2O.
6. Wash buffer 2: 50 mM Tris, pH 8.0, 150 mM NaCl, in MS-
grade H2O.
7. Wash buffer 3: 50 mM Tris, pH 8.0, 450 mM NaCl, in MS-
grade H2O.
8. Wash buffer 4: 50 mM Tris, pH 8.0, in MS-grade H2O.
9. Elution buffer: 10 % acetic acid in MS-grade water (use best
grade glacial acetic acid, e.g., Sigma ACS grade).
2.3 Targeted Mass In cases where peptide epitopes are known, isotopically labeled
Spectrometric (AQUA) peptides can be used for absolute quantitation by tar-
Analysis geted LC-MRM (multiple reaction monitoring) [38, 39]. AQUA
of MHC Eluate peptides are composed of the same amino acid sequence as the
natural equivalent but bearing one or more heavy amino acids.
A Systems Approach to Understand Antigen Presentation and the Immune Response 195
2.4 Analysis In order to correlate the cellular proteome with the immun-
of Cellular Proteome opeptidome, a small sample of the MHC lysate is subjected to
Using FASP Digestion tryptic digestion and mass spectrometric analysis [40]. Samples
of MHC Lysate for tryptic digestion may be taken immediately after lysis or from
the flow through of the MHC-affinity column. Retaining the
flow through from the affinity column and freezing at −80 °C is
advised so that if required, more tryptic digestions can be per-
formed. We utilize the Expedeon FASP Protein Digestion Kit as
a convenient way of generating tryptic peptides. This kit is com-
patible with a number of reducing agents; however we recom-
mend TCEP.
1. Expedeon FASP Protein Digestion Kit (reagents except tryp-
sin and reducing agent are provided in the FASP kit).
2. Ammonium bicarbonate (in kit): 50 mM.
3. NaCl (in kit): 0.5 M.
4. Urea sample solution (in kit): 1 ml of Tris solution to one tube
of urea, vortex until powder is dissolved. Prepare just before
use.
5. 10× Iodoacetamide solution (in kit): Add 100 μl of urea
sample solution to one tube of iodoacetamide. Mix and dis-
solve by pipetting up and down. Transfer into clean
microfuge tube, wrap in foil, and keep on ice. Prepare just
before use.
6. Digestion solution (in kit): Dissolve one 1 μg tube of trypsin
in 75 μl of 50 mM ammonium bicarbonate solution and
keep on ice. Aim for 1:100 trypsin-to-protein ratio. Scale
amount of trypsin according to how much lysate is used, i.e.,
if loading 400 μg of protein, use 4 μg trypsin. Prepare just
before use.
7. Bradford reagent.
8. Trypsin (single shot 1 μg, Sigma).
9. 0.1 % FA.
10. TCEP: 0.5 M in 50 mM ammonium bicarbonate (Sigma).
196 Nadine L. Dudek et al.
3 Methods
3.1.2 Generation 1. Cells (5 × 107 to 1 × 109) can be grown in spinner flasks, biore-
of Cell Lysate actors, or roller bottles (see Note 7) to appropriate numbers,
washed in PBS, and harvested by centrifugation (2000 × g,
10 min at 4 °C). Pellets should be snap frozen in liquid nitrogen
and may be harvested iteratively for storage at −80 °C for up to
6 months. If collecting tissues, they should be rinsed in cold
PBS containing protease inhibitors and immediately snap fro-
zen without liquid for later processing.
2. Prepare a 2× concentrated solution of lysis buffer. Lysis buffer
is added at 2× to allow for the volume of the cell pellet to be
taken into account prior to adjustment of the concentration of
the lysate to 1×. Cells are lysed at 5 × 107–1 × 108 cells per ml
A Systems Approach to Understand Antigen Presentation and the Immune Response 197
3.1.3 Immunoaffinity 1. Using gravity flow or a peristaltic pump in a cold room, load
Purification of MHC Class I/ cell lysate onto a protein A sepharose pre-column that has been
Class II pre-equilibrated in 10 c.v. wash buffer 1. Multiple pre-columns
may be required depending on the size and type of sample and
should be replaced upon clogging.
2. Collect pre-cleared lysate and slowly load onto the cross-linked
mAb column(s). If gravity flow is too quick (<1 h for lysate to
pass), use a peristaltic pump. For small samples (1–4 ml lysate),
it is generally better to add lysate to several of 2 ml LoBind
Eppendorf tubes containing resin and rotate slowly end over
end at 4 °C for 1 h.
3. For maximal yield the lysate should be run through the col-
umn twice. Retain flow through and freeze at −80 °C for sub-
sequent FASP analysis. If multiple columns are being run (i.e.,
class I and II elutions from the same lysate), we routinely run
the lysate once only over each column. The columns can be set
up on a retort stand in tandem so that the flow through from
198 Nadine L. Dudek et al.
3.1.4 Separation of MHC A single RP-HPLC step may be used to isolate peptides if an
Eluate by RP-HPLC immunological readout is used to assay peptide fractions.
However biochemical analysis by mass spectrometry requires a
minimum of two dimensions of RP-HPLC to achieve sufficient
separation (see Note 8).
1. Peptides are separated from MHC heavy chain, β2m (for class
I molecules), leached antibody, and contaminating detergent
using a C18 reverse-phase column running on a mobile-phase
buffer A of 0.1 % TFA and buffer B of 80 % acetonitrile/0.1 %
TFA (see Note 9). We routinely use a 4.6 mm internal diame-
ter × 50 mm (or 100 mm for greater separation see Note 10)
long reversed-phase C18 endcapped HPLC column
(Chromolith Speed Rod, Merck) on an ÄKTAmicro™ HPLC
system (GE Healthcare).
2. Separate based on a rapid gradient of buffer A to B, which
results in 10–30 peptide-containing fractions (e.g., 2–40 % B
for 4 min, 40–45 % for 4 min, and a rapid 2 min increase to
100 % B; Fig. 2). Using this approach a small number of early
fractions contain greater than 95 % of the peptides, whilst the
later fractions contain IGEPAL 630 polymers which hamper
MS analysis severely and β2-microglobulin (see Note 11).
3. Collect fractions (500 μl) into LoBind Eppendorf tubes.
At this point fractions may be frozen at −80 °C.
A Systems Approach to Understand Antigen Presentation and the Immune Response 199
Fig. 2 Monolithic separation of affinity-purified MHC-peptide complexes and fractionation of bound peptides.
Representative UV trace showing fractionation of the eluted mixture of peptides and heavy chains from both
MHC class I and class II eluates. The early fractions contain MHC peptides and the later fractions contain heavy
chains, detergent, and leached antibody. The β2-microglobulin peak is highlighted for the MHC class I eluate
3.2 LC-MS/MS Two forms of mass spectrometry can be used to analyze fractions
Analysis containing HLA-bound peptides (Fig. 3). This may consist of the
of MHC Eluate traditional global LC-MS/MS analysis or more targeted method-
ologies such as multiple reaction monitoring. Global LC-MS/MS
is recommended for samples where the MHC peptide repertoire
composition is unknown. Peptide species are separated by an LC
gradient and paired MS and MS/MS spectra are acquired by the
mass spectrometer. Downstream analysis and identification of
acquired spectra are facilitated by either manual sequencing or,
preferably, the use of protein identification software algorithms
(e.g., Mascot (MatrixScience), ProteinPilot™ (SCIEX)).
Numerous factors can affect the resulting number of peptide
identifications from a global LC-MS/MS analysis, including initial
starting cell number, expression level of the MHC molecules at the
cell surface, efficiency of cell lysis, quantity of antibody used for
immunoaffinity capture, appropriate and sufficient upstream
HPLC fractionation, online LC gradient, and specific MS param-
eter settings. It is recommended to optimize these variables.
1. Place samples in a sonicating water bath for 5 min to detach
peptides bound to plastic.
200 Nadine L. Dudek et al.
Fig. 3 A comparison of information-dependent acquisition (IDA) or LC-MS/MS and LC-MRM analysis. In global
LC-MS/MS analysis a defined number of precursor ions are selected for fragmentation during each duty cycle
of the mass spectrometer. The selection of precursors is a stochastic process typically based on abundance or
ion intensity and limited to the top 10–50 most intense ions entering the instrument in that particular cycle. In
contrast, LC-MRM involves the selection of specific, predefined, precursors that are targeted for analysis and
detected based on a defined set of fragment ions. This allows relatively low-intensity ions to be selected in
preference to more abundant co-eluting species. Area under the curve quantitation of the MRM transitions can
then afford very accurate and specific quantitation
Fig. 4 Examples of biochemical analysis of peptides eluted from MHC class I and class II molecules. (a) A typical
total ion chromatogram (TIC) of LC-separated MHC-bound peptides analyzed on a 5600+ TripleTOF mass
spectrometer (SCIEX). (b–d) Annotated MS/MS spectra of various MHC-bound peptides. ARFDSDVEVY (panel b)
and phosphorylated RSLSPMS*GLFGSIW (panel d) were eluted from the MHC class I molecules HLA-B*27:05
and HLA-B*57:01, respectively. AGQLVFLATEGDHL (panel c) was eluted from human MHC class II molecules.
The insets of each panel show the corresponding precursor MS1 regions
3.3 Targeted Mass Targeted methodologies, e.g., LC-MRM, are of use when specific
Spectrometric peptide epitopes are known and a qualitative and/or quantitative
Analysis readout is desired. Here, MS instrument parameters are set to tar-
of MHC Eluate get only the peptides of interest, ignoring the rest of the sample
allowing low-abundance peptides to be detected in complex sam-
ples (see Note 14). LC-MRM also negates the need for high cell
numbers as such targeted methods are, by their nature, highly spe-
cific and more sensitive than global LC-MS/MS approaches. For
LC-MRM, synthetic peptides corresponding to those of interest
can be used to design and optimize MRM parameters [41] or
alternatively MS/MS data from discovery-based experiments can
be used to identify optimal transition parameters. The detection of
spiked AQUA peptides by LC-MRM allows integration of the area
202 Nadine L. Dudek et al.
under the curve of the light and heavy peptides, providing quanti-
tation of the light peptides. These values can be related back to the
starting number of cells, to give the most accurate assessment pos-
sible of epitope copy number per cell [38, 39].
1. Prepare AQUA peptide stock, we generally reconstitute pep-
tides at 5 mM in 100 % DMSO (choice of buffer will be user
dependent, see Note 5). Run approximately 50 fmol of peptide
diluted in 0.1 % FA on mass spectrometer to determine the
dominant precursor ion. Peptides are then run targeting the
dominant precursor ion and fragmented across a range of col-
lision energies (CE) in order to determine the optimal energy
to generate the most intense fragment ions [41]. Note that
each MRM transition can use its own CE value, so different
fragment ions will sometimes require different CE values.
Typically the top four most intense fragment ions are used to
build the MRM for a given peptide.
2. For each AQUA peptide perform a standard curve and choose
a concentration in the linear part of the curve that will be used
to spike MHC samples. Typically this will be between 1 and
100 fmol. Determine which fraction AQUA peptides elute
during RP-HPLC for initial separation on the C18 column
(prior to loading onto mass spec). By doing this it will be pos-
sible to run only fractions that contain the peptide of interest
which will substantially reduce instrument time depending on
the number of AQUA peptides being analyzed. Moreover, co-
elution of the AQUA peptide provides further confidence in
the detection of the target peptide.
3. Add isotopically labeled (AQUA) peptides of known concen-
tration into the MHC eluate immediately following elution
from the antibody affinity column. This will allow for the co-
elution of the light (i.e., endogenous) and heavy (i.e., AQUA)
peptide during RP-HPLC.
4. We routinely run LC-MRM experiments on a 5500 QTRAP
(SCIEX) with similar LC conditions as above. MRM transi-
tions are used with a dwell time between 5 and 40 ms, opti-
mized to result in a cycle time that will lead to at least eight
data points across a detected peptide. MS parameters are unit
resolution for Q1 and Q3, with the MRM experiment coupled
to an information-dependent acquisition (IDA) criterion set to
trigger an EPI scan (10,000 Da/s; rolling CE; unit resolution)
following any MRM transition exceeding 500 counts.
5. To quantitate the amount of the peptide of interest present in
each HPLC fraction, the area under each MRM transition
peak is calculated (using for example MultiQuant 2.0, SCIEX).
After peak integration, the area value for all MRM transitions
for the peptide is combined and compared against the area
A Systems Approach to Understand Antigen Presentation and the Immune Response 203
4 Notes
http://www.genscript.com/peptide_solubility_and_stablity.
html. Peptides should be quantified to an exact concentration
(amino acid analysis; DirectDetect (Merck Millipore)). AQUA
peptides are spiked into the sample immediately post-acid elu-
tion from the affinity column. The amount of each AQUA
peptide to add requires optimization on a per-peptide basis,
but typically ranges from 1 to 100 fmol per sample.
6. For each new antibody it is advisable to check the efficiency of
the cross-linking reaction by SDS-PAGE. Take a sample of the
original antibody (1) and a sample of the flow through (2)
following incubation with the resin. Take aliquots of resin
(25 μl) before the addition of DMP (3) and after incubation
with DMP (4). It is also advisable to concentrate the flow
through from the citric acid wash (5) using a 15–30 kDa cut-
off concentrator to ~500 μl (this is done to see how much
antibody has been left unbound), and take 25 μl to add to 2×
SDS-PAGE loading dye. Samples should be run on a reducing
12 % SDS-PAGE gel and Commassie stained. Heavy and light
chains of the antibody should be seen in samples 1 and 3.
There should be little antibody in samples 2, 4, and 5. If the
antibody has not been used for DMP cross-linking before,
perform a small-scale purification to test that the antibody still
retains its binding affinity.
7. Cells can be expanded by standard tissue culture techniques;
however particular care should be taken with adherent cells.
Removing cells with trypsin can strip coatings from treated tis-
sue culture plastics, which may interfere with subsequent mass
spectral analysis. We routinely passage cells using trypsin; how-
ever when harvesting for snap freezing we remove cells using
5–10 mM EDTA in PBS.
8. Column and mobile phase choice for multidimensional RP-
HPLC are dictated by sample composition but consider-
ations should include altered ion pair agent, altered mobile
phase pH, altered stationary-phase ligand or mode, and col-
umn dimensions.
9. A common practice to separate peptides from heavy chain and
β2m in the case of class I molecules is to use a low-protein-
binding spin filter. In our experience, this results in significant
loss of peptides, irrespective of filter brand or pre-blocking
with BSA. We find that separation using RP-HPLC increases
the yield of peptides at least tenfold.
10. We routinely separate β2m from heavy chains using a 4.6 mm
internal diameter × 50 or 100 mm long reversed-phase C18
endcapped HPLC column (Chromolith Speed Rod, Merck).
The choice between 50 and 100 mm is dependent on the cell
number and whether the samples are to be used for LC-MS/MS
A Systems Approach to Understand Antigen Presentation and the Immune Response 207
15. The FASP kit comes with 30 kDa cutoff spin filters; we how-
ever prefer to use 10 kDa cutoff spin filters from Pall.
16. Mass spectrometric analysis can be performed on the FASP
digest without fractionation; however we find that this dra-
matically reduces the number of protein identifications. We
recommend an off-line separation by RP-HPLC; alternatively
other separation methods such as SCX may be used.
References
1. Madden DR (1995) The three-dimensional 12. Scull KE, Dudek NL, Corbett AJ et al (2012)
structure of peptide-MHC complexes. Annu Secreted HLA recapitulates the immunopepti-
Rev Immunol 15:587–622 dome and allows in-depth coverage of HLA
2. Stern LJ, Brown JH, Jardetzky TS et al (1994) A*02:01 ligands. Mol Immunol 51(2):136–142
Crystal structure of the human class II MHC 13. Illing PT, Vivian JP, Dudek NL et al (2012)
protein HLA-DR1 complexed with an influ- Immune self-reactivity triggered by drug-
enza virus peptide. Nature 368(6468): modified HLA-peptide repertoire. Nature
215–221 486(7404):554–558
3. Fremont DH, Hendrickson WA, Marrack P 14. Dudek NL, Tan CT, Gorasia DG et al (2012)
et al (1996) Structures of an MHC class II Constitutive and inflammatory immunopepti-
molecule with covalently bound single pep- dome of pancreatic beta-cells. Diabetes
tides. Science 272(5264):1001–1004 61(11):3018–3025
4. Garcia KC, Teyton L, Wilson IA (1999) 15. Urban RG, Chicz RM, Lane WS et al (1994)
Structural basis of T cell recognition. Annu A subset of HLA-B27 molecules contains pep-
Rev Immunol 17:369–397 tides much longer than nonamers. Proc Natl
5. Falk K, Rötzschke O, Deres K et al (1991) Acad Sci U S A 91(4):1534–1538
Identification of naturally processed viral non- 16. Purcell AW, Kelly AJ, Peh CA et al (2000)
apeptides allows their quantification in infected Endogenous and exogenous factors contribut-
cells and suggests an allele-specific T cell epit- ing to the surface expression of HLA B27 on
ope forecast. J Exp Med 174:425–434 mutant APC. Hum Immunol 61(2):120–130
6. Sijts AJ, Neisig A, Neefjes J et al (1996) Two 17. Storkus WJ, Howell DN, Salter RD et al
Listeria monocytogenes CTL epitopes are pro- (1987) NK susceptibility varies inversely with
cessed from the same antigen with different target cell class I HLA antigen expression.
efficiencies. J Immunol 156(2):683–692 J Immunol 138(6):1657–1659
7. Rotzschke O, Falk K, Deres K et al (1990) 18. Macdonald WA, Purcell AW, Mifsud NA et al
Isolation and analysis of naturally processed (2003) A naturally selected dimorphism within
viral peptides as recognized by cytotoxic the HLA-B44 supertype alters class I struc-
T cells. Nature 348(6298):252–254 ture, peptide repertoire, and T cell recogni-
8. Storkus WJ, Zeh HJ, Maeurer MJ et al (1993) tion. J Exp Med 198(5):679–691
Identification of human melanoma peptides 19. Zernich D, Purcell AW, Macdonald WA et al
recognized by class I restricted tumor infiltrat- (2004) Natural HLA class I polymorphism
ing T lymphocytes. J Immunol 151(7): controls the pathway of antigen presentation
3719–3727 and susceptibility to viral evasion. J Exp Med
9. Storkus WJ, Zeh HJ, Salter RD et al (1993) 200(1):13–24
Identification of T-cell epitopes: rapid isolation 20. Scally SW, Petersen J, Law SC et al (2013) A
of class I-presented peptides from viable cells molecular basis for the association of the HLA-
by mild acid elution. J Immunother 14(2): DRB1 locus, citrullination, and rheumatoid
94–103 arthritis. J Exp Med 210(12):2569–2582
10. Falk K, Rötzschke O, Stevanovic S et al (1991) 21. Ringrose JH, Yard BA, Muijsers A et al (1996)
Allele-specific motifs revealed by sequencing of Comparison of peptides eluted from the
self-peptides eluted from MHC molecules. groove of HLA-B27 from Salmonella infected
Nature 351:290–296 and non-infected cells. Clin Rheumatol 15
11. Rammensee HG, Falk K, Rotzschke O (1993) Suppl 1:74–78
Peptides naturally presented by MHC class I 22. van Els CA, Herberts CA, van der Heeft E
molecules. Annu Rev Immunol 11:213–244 et al (2000) A single naturally processed
A Systems Approach to Understand Antigen Presentation and the Immune Response 209
measles virus peptide fully dominates the 34. Ozato K, Sachs DH (1981) Monoclonal anti-
HLA-A*0201-associated peptide display bodies to mouse MHC antigens. III. Hybridoma
and is mutated at its anchor position in per- antibodies reacting to antigens of the H-2b
sistent viral strains. Eur J Immunol haplotype reveal genetic control of isotype
30(4):1172–1181 expression. J Immunol 126(1):317–321
23. Nepom BS, Nepom GT, Coleman M et al 35. Kappler JW, Skidmore B, White J et al (1981)
(1996) Critical contribution of beta chain resi- Antigen-inducible, H-2-restricted, interleukin-
due 57 in peptide binding ability of both 2-producing T cell hybridomas. Lack of inde-
HLA-DR and -DQ molecules. Proc Natl Acad pendent antigen and H-2 recognition. J Exp
Sci U S A 93(14):7202–7206 Med 153(5):1198–1214
24. Lampson LA, Levy R (1980) Two populations 36. Janeway CA Jr, Conrad PJ, Lerner EA et al
of Ia-like molecules on a human B cell line. (1984) Monoclonal antibodies specific for Ia
J Immunol 125(1):293–299 glycoproteins raised by immunization with
25. Gorga JC, Knudsen PJ, Foran JA et al (1986) activated T cells: possible role of T cellbound
Immunochemically purified DR antigens in Ia antigens as targets of immunoregulatory T
liposomes stimulate xenogeneic cytolytic T cells. J Immunol 132(2):662–667
cells in secondary in vitro cultures. Cell 37. Oi VT, Jones PP, Goding JW et al (1978)
Immunol 103(1):160–173 Properties of monoclonal antibodies to mouse
26. Watson AJ, DeMars R, Trowbridge IS et al Ig allotypes, H-2, and Ia antigens. Curr Top
(1983) Detection of a novel human class II Microbiol Immunol 81:115–120
HLA antigen. Nature 304(5924):358–361 38. Croft NP, Smith SA, Wong YC et al (2013)
27. Parham P, Brodsky FM (1981) Partial purifica- Kinetics of antigen expression and epitope pre-
tion and some properties of BB7.2. A cyto- sentation during virus infection. PLoS Pathog
toxic monoclonal antibody with specificity for 9(1), e1003129
HLA-A2 and a variant of HLA-A28. Hum 39. Tan CT, Croft NP, Dudek NL et al (2011)
Immunol 3(4):277–299 Direct quantitation of MHC-bound peptide
28. Ellis SA, Taylor C, McMichael A (1982) epitopes by selected reaction monitoring.
Recognition of HLA-B27 and related antigen Proteomics 11(11):2336–2340
by a monoclonal antibody. Hum Immunol 40. Wisniewski JR, Zougman A, Nagaraj N et al
5(1):49–59 (2009) Universal sample preparation method
29. Schittenhelm RB, Dudek NL, Croft NP et al for proteome analysis. Nat Methods 6(5):359–
(2014) A comprehensive analysis of constitu- 362. doi:10.1038/nmeth.1322
tive naturally processed and presented HLA- 41. Lange V, Picotti P, Domon B et al (2008)
C*04:01 (Cw4)-specific peptides. Tissue Selected reaction monitoring for quantitative
Antigens 83(3):174–179 proteomics: a tutorial. Mol Syst Biol 4:222
30. Braud VM, Allan DS, Wilson D et al (1998) 42. Edwards PA, Smith CM, Neville AM et al
TAP- and tapasin-dependent HLA-E sur- (1982) A human-hybridoma system based on
face expression correlates with the binding a fast-growing mutant of the ARH-77 plasma
of an MHC class I leader peptide. Curr Biol cell leukemia-derived line. Eur J Immunol
8(1):1–10 12(8):641–648
31. Thomas R, Apps R, Qi Y et al (2009) HLA-C 43. Alexander J, Payne JA, Murray R et al (1989)
cell surface expression and control of HIV/ Differential transport requirements of HLA
AIDS correlate with a variant upstream of and H-2 class I glycoproteins. Immunogenetics
HLA-C. Nat Genet 41(12):1290–1294 29(6):380–388
32. Hammerling GJ, Rusch E, Tada N et al (1982) 44. Nelson CA, Roof RW, McCourt DW et al
Localization of allodeterminants on H-2Kb (1992) Identification of the naturally pro-
antigens determined with monoclonal anti- cessed form of hen egg white lysozyme bound
bodies and H-2 mutant mice. Proc Natl Acad to the murine major histocompatibility com-
Sci U S A 79(15):4737–4741 plex class II molecule I-Ak. Proc Natl Acad Sci
33. Straus DS, Stroynowski I, Schiffer SG et al U S A 89(16):7380–7383
(1985) Expression of hybrid class I genes of 45. Escher C, Reiter L, MacLean B et al (2012)
the major histocompatibility complex in mouse Using iRT, a normalized retention time for
L cells. Proc Natl Acad Sci U S A 82(18): more targeted measurement of peptides.
6245–6249 Proteomics 12(8):1111–1121
Chapter 15
Abstract
Chemical proteomics provides a powerful means to gain systems-level insight into the mode of action of
small molecules and/or natural products. In contrast to high-throughput screening efforts which only
interrogate selected subproteomes such as kinases and often only consider individual domains, the meth-
odology presented herein allows for the determination of the molecular targets of small molecules or drugs
in a more relevant physiological setting. As such, the compound of interest is exposed to the entire variety
of cellular proteins considering all naturally occurring posttranslational modifications and activation states.
Samples prepared according to the procedures described in this protocol are compatible with lysates from
cultured cell lines, primary cells, or samples from biopsies. In combination with state-of-the-art mass spec-
trometry techniques this approach grants access to a comprehensive view of small molecule-target protein
interactions.
Key words Drug discovery, Target deconvolution, Chemical proteomics, Mode of action
1 Introduction
Jörg Reinders (ed.), Proteomics in Systems Biology: Methods and Protocols, Methods in Molecular Biology, vol. 1394,
DOI 10.1007/978-1-4939-3341-9_15, © Springer Science+Business Media New York 2016
211
212 Kilian V.M. Huber and Giulio Superti-Furga
small molecules or natural products are often both time and cost
intensive and usually only cover a selected range of protein classes,
e.g., kinases or histone deacetylases. Furthermore, these assays do
not provide any information if the targets identified in the screen
are “real-and-relevant” interactors as such issues as differential tis-
sue expression or even posttranslational modifications are not
considered.
Several target deconvolution approaches have been developed
covering a wide range of biochemical and genetic techniques as
well as in silico methods [2, 4]. Among all those affinity-based
approaches have contributed to the discovery of a number of
important drug classes such as immunosuppressants [5, 6] and his-
tone deacetylase (HDAC) inhibitors [7]. The combination of this
methodology with protein mass spectrometry has been termed
“chemical proteomics” and has lately been applied to diverse areas
of research including the elucidation of the molecular mechanism
of thalidomide teratogenicity [8] as well as the identification of
novel regulators of necroptosis signaling [9] and potential antican-
cer targets [10]. In a typical chemical proteomic experiment, the
small molecule or compound of interest is incubated with a rele-
vant cell lysate which can be prepared from tissue culture cell lines
or even primary cells and biopsy samples. Using the small-molecule
compound as bait by means of a drug matrix, cellular interactors
are captured and purified by affinity enrichment. After washing,
the eluted proteins are digested to peptides and can subsequently
be identified by mass spectrometry followed by bioinformatic anal-
ysis (Fig. 1). This procedure requires the compound of interest
itself or a corresponding analogue to be amenable to chemical
derivatization in order to be immobilized on the solid phase.
The design of a suitable analogue is facilitated by prior knowledge
of structure-activity relationships (SAR) or co-crystal structures of
annotated targets. Alternatively, the so-called east–west approach
may be applied to compounds devoid of those data [11]. The concept
of this strategy is to prepare two coupleable analogues of the
compound of interest of which each is modified at a different,
preferably most distant site.
An alternative chemical proteomic approach is activity-based
probe profiling (ABPP) which does not require chemical modifica-
tion of the query compound by taking advantage of a reactive
probe which binds covalently to a given class of proteins, e.g.,
serine hydrolases [12–14]. In this case, comparison of treated versus
untreated sample yields the putative interactors. Recently, another
proteomic target identification strategy based on ligand-induced
thermal stabilization of proteins has been established [15]. This
methodology termed thermal stability profiling does also not
depend on chemical derivatization and instead uses the unmodified
original compound of interest allowing for the detection of protein-
ligand interactions in intact living cells. For a general overview of
Fig. 1 Schematic outline of a chemical proteomic experiment. A drug matrix consisting of the immobilized small molecule on a solid phase is incubated with lysates
from either cell lines, primary cells, or biopsy samples. After incubation, contaminants are removed by extensive washing and finally target proteins are eluted and
analyzed by mass spectrometry followed by bioinformatic processing
Profiling of Small Molecules by Chemical Proteomics
213
214 Kilian V.M. Huber and Giulio Superti-Furga
2 Materials
5. EDTA.
6. NaCl.
7. NaOH.
8. Purified water.
9. Formic acid.
HEPES-NaOH-EDTA buffer pH 7.5: 50 mM HEPES, 0.5 μM
EDTA pH 8.0, 100 mM NaCl; adjust pH to 7.5 using NaOH,
prepare freshly, keep on ice.
3 Methods
3.1 Preparation 1. (a) For cell pellets, thaw pellets on ice and resuspend in lysis
of Whole-Cell Lysates buffer (depending on pellet size, rule-of-thumb 1:1 ratio),
(Timing 2 h) (See transfer into homogenizer/Dounce apparatus (e.g., 0.9 mm
Notes 2 and 3) syringe) and homogenize sample ten times (optional).
(b) For tissues, transfer the sample into a tissue homoge-
nizer/Dounce apparatus, wash with lysis buffer, and adjust to
desired volume; homogenize sample ten times.
2. Transfer the homogenate to a Falcon tube and incubate on ice
for 30 min.
3. Transfer homogenate to polycarbonate ultracentrifuge tubes,
balance tubes, and centrifuge lysate for 10 min at 4 °C at
20,000 × g.
4. Transfer supernatant to fresh polycarbonate ultracentrifuge
tubes, balance tubes, and centrifuge for 1 h at 4 °C at 90,000 × g.
5. Transfer supernatant (remove most of lipid layer, if possible)
to a fresh Falcon centrifuge tube, and keep on ice.
6. Determine protein concentration (e.g., Bradford).
7. Prepare lysate aliquots (e.g., 5–10 mg total protein) or use
directly for pull-downs (see Note 4).
216 Kilian V.M. Huber and Giulio Superti-Furga
3.2 Coupling 1. Pipet 100 μL of slurry (≈50 μL settled bed volume) in a 1.5
of Compounds mL Eppendorf tube. Caution: Always use filter tips and cut
to Sepharose Beads pipet tips for pipetting beads!
(Timing 3 Days) 2. Centrifuge beads for 3 min at room temperature at 75 × g, and
remove supernatant.
3. Add 50 μL of DMSO (abs) to beads, suspend gently by invert-
ing several times, centrifuge (as before), and discard
supernatant.
4. Add 500 μL DMSO (abs), resuspend beads gently by inverting
several times, transfer beads in a 2 mL Eppendorf tube, centri-
fuge (as before), and discard supernatant; repeat wash step
another two times.
5. Resuspend beads in 50 μL DMSO (abs).
6. Add 0.025 μmol of coupleable compound to 50 % bead slurry;
add 0.75 μL triethylamine, and mix carefully.
7. Incubate on roto-shaker for 16–24 h at RT with 10 rpm.
8. To check coupling efficiency, centrifuge beads (as before) and
remove 10 μL (≈5 nmol) from supernatant.
9. Check for remaining unreacted compound by HPLC; if there
are still significant amounts detected in the supernatant go
back to step 7 and extend coupling reaction time. Repeat steps
8 and 9.
10. Add 2.5 μL ethanolamine to drug-bead mixture in order to
block unreacted NHS-ester groups.
11. Incubate on roto-shaker for at least 8 h at room temperature
with 10 rpm.
12. Centrifuge beads (see above), remove supernatant, and wash
twice with 500 μL of DMSO (abs).
13. Proceed directly with pull-down and wash with lysis buffer.
Pausing point: Alternatively, the drug-bead matrix can be
stored for up to 2 weeks using the following procedure: Remove
supernatant and add 50 μL of isopropanol, resuspend beads gently
by inverting several times, centrifuge (as before), and discard
supernatant. Add 500 μL isopropanol, resuspend beads gently by
inverting several times, centrifuge (as before), and discard
supernatant; repeat wash step once. Add 50 μL isopropanol to
beads and resuspend gently; store coupled beads at 4 °C (away
from light) until further use.
3.4 Concluding The described procedures provide an effective means to reveal the
Remarks interactors of small molecules and natural compounds. However,
due to the high complexity of the samples and varying protein abun-
dance it is recommendable to include control pull-downs which can
be either the unreacted bead matrix itself or an unrelated compound
matrix to estimate random and unspecific binding. Alternatively,
lysates can be preincubated with the original, unmodified compound
of interest to determine competitive binding (Fig. 1). If coupled
with quantitative MS technologies such as iTRAQor TMT this
approach also allows for the determination of Kds.
218 Kilian V.M. Huber and Giulio Superti-Furga
4 Notes
References
1. Swinney DC, Anthony J (2011) How were mide teratogenicity. Science 327(5971):
new medicines discovered? Nat Rev Drug 1345–1350
Discov 10(7):507–519 9. Sun L, Wang H, Wang Z et al (2012) Mixed
2. Schenone M, Dancik V, Wagner BK et al lineage kinase domain-like protein mediates
(2013) Target identification and mechanism of necrosis signaling downstream of RIP3 kinase.
action in chemical biology and drug discovery. Cell 148(1–2):213–227
Nat Chem Biol 9(4):232–240 10. Huber KVM, Salah E, Radic B et al (2014)
3. Sillaber C, Herrmann H, Bennett K et al (2009) Stereospecific targeting of MTH1 by (S)-
Immunosuppression and atypical infections in crizotinib as an anticancer strategy. Nature
CML patients treated with dasatinib at 140 mg 508(7495):222–227
daily. Eur J Clin Invest 39(12):1098–1109 11. Rix U, Superti-Furga G (2009) Target profil-
4. Terstappen GC, Schlupen C, Raggiaschi R et al ing of small molecules by chemical proteomics.
(2007) Target deconvolution strategies in drug Nat Chem Biol 5(9):616–624
discovery. Nat Rev Drug Discov 6(11):891–903 12. Nomura DK, Dix MM, Cravatt BF (2010)
5. Brown EJ, Albers MW, Bum Shin T et al Activity-based protein profiling for biochemi-
(1994) A mammalian protein targeted by cal pathway discovery in cancer. Nat Rev
G1-arresting rapamycin-receptor complex. Cancer 10(9):630–638
Nature 369(6483):756–758 13. Savitski MM, Reinhard FBM, Franken H et al
6. Harding MW, Galat A, Uehling DE et al (2014) Tracking cancer drugs in living cells by
(1989) A receptor for the immuno-suppressant thermal profiling of the proteome. Science
FK506 is a cis-trans peptidyl-prolyl isomerase. 346(6205):1255784
Nature 341(6244):758–760 14. Huber KVM, Olek KM, Müller AC et al (2015)
7. Taunton J, Hassig CA, Schreiber SL (1996) A Proteome-wide drug and metabolite interaction
mammalian histone deacetylase related to the mapping by thermal-stability profiling. Nat
yeast transcriptional regulator Rpd3p. Science Meth 12(11):1055–1057
272(5260):408–411 15. Schirle M, Bantscheff M, Kuster B (2012) Mass
8. Ito T, Ando H, Suzuki T et al (2010) spectrometry-based proteomics in preclinical
Identification of a primary target of thalido- drug discovery. Chem Biol 19(1):72–84
Chapter 16
Abstract
Mass spectrometry-based methods allow for the direct, comprehensive analysis of expressed proteins and
their quantification among different conditions. However, in general identification of proteins by assigning
experimental mass spectra to peptide sequences of proteins relies on matching mass spectra to theoretical
spectra derived from genomic databases of organisms. This conventional approach limits the applicability
of proteomic methodologies to species for which a genome reference sequence is available. Recently,
RNA-sequencing (RNA-Seq) became a valuable tool to overcome this limitation by de novo construction
of databases for organisms for which no DNA sequence is available, or by refining existing genomic data-
bases with transcriptomic data. Here we present a generic pipeline to make use of transcriptomic data for
proteomics experiments. We show in particular how to efficiently fuel proteomic analysis workflows with
sample-specific RNA-sequencing databases. This approach is useful for the proteomic analysis of so far
unsequenced organisms, complex microbial metatranscriptomes/metaproteomes (for example in the
human body), and for refining current proteomics data analysis that solely relies on the genomic sequence
and predicted gene expression but not on validated gene products. Finally, the approach used in the here
presented protocol can help to improve the data quality of conventional proteomics experiments that can
be influenced by genetic variation or splicing events.
1 Introduction
Jörg Reinders (ed.), Proteomics in Systems Biology: Methods and Protocols, Methods in Molecular Biology, vol. 1394,
DOI 10.1007/978-1-4939-3341-9_16, © Springer Science+Business Media New York 2016
219
220 Toni Luge and Sascha Sauer
2 Materials
Fig. 1 Scheme of the bioinformatics workflow for the integrative analysis of shot-
gun mass spectrometry and RNA-Seq data. Proteins are subjected to LC-MS/MS
analysis whereas RNA, ideally isolated from the same samples used for pro-
teomics, is sequenced on next-generation sequencing (NGS) instruments [19].
Short read sequencing data is used to reconstruct protein sequences used as
sequence database in the peptide search engine for identification of peptides
and proteins from mass spectrometric raw data. Finally, identified proteins can
be de novo annotated based on sequence homology search to known proteins
3 Methods
Fig 2 Overview of the reconstruction of protein sequences from short read RNA-
sequencing (RNA-Seq) data. The Trinity pipeline [36] is used to build full-length
transcripts. This software first assembles contigs (sets of overlapping sequences)
in the Inchworm module. The Chrysalis module clusters contigs and processes
each cluster to a de Bruijn graph component. Butterfly, the third module, extracts
all probable sequences from the graph components and reports reconstructed
sequences and their isoforms. Protein sequences are inferred by the EMBOSS
tool “getorf” [37] and subjected to the Andromeda search engine [16] for peptide
and protein identification from mass spectrometry raw data
and reads that probably contain sequencing errors (see Note 4).
The reconstructed transcripts are translated to protein sequences
using the EMBOSS tool “getorf.” This file is subjected to the
Andromeda search engine for peptide identification from mass
spectrometry data (see Subheading 3.3).
1. Log in from your local working station to your Unix server
where Trinity is installed. For instance, use the free SSH and
telnet client PuTTY (or appropriate alternatives) when work-
ing on Windows operating system. Open terminal (“$” denotes
the shell prompt) and copy raw or if necessary pre-processed
sequencing data in FASTQ format to the working folder:
$ mkdir workingfolder
$ cp rawdata/Illumina_singleread_*.fq /
workingfolder
$ cd workingfolder
Sample Specific Databases for Proteomics 225
Fig 3 Main steps performed by the MaxQuant software [39] for the analysis of shotgun proteomic data acquired
on high-resolution liquid chromatography-coupled tandem mass spectrometry (LC-MS/MS) instruments. The
software suite contains algorithms to efficiently extract information from raw mass spectrometry data includ-
ing peptide and protein identities as well as their high-accuracy quantification. First features are detected on
MS1 level as three-dimensional objects in m/z, retention time, and intensity space. Isotope label partners are
identified, features quantified and normalized, and their masses calculated precisely. To achieve mass accura-
cies in the p.p.b. range, mass errors (Δm/z ) are estimated and mass offsets corrected by nonlinear mass
recalibration. Furthermore MS/MS spectra are preprocessed and filtered prior to the main MS/MS ion search
by the Andromeda search engine [16]. Peptide spectrum matches are scored and the false discovery rate
(FDR) controlled on peptide and protein level by decoy database searching [17]. Additionally MaxQuant
accounts for co-fragmented peptides occasionally observed due to the complexity of protein samples.
Optionally, isotope-labeled peptide pairs are re-quantified if one label partner is missing and peptide identifica-
tions matched between different LC-MS/MS runs after their retention time alignment. In the consolidation
phase proteins are assembled from peptide identifications, protein ratios calculated from peptide ratios by
robust median averaging, and results compiled in multiple cross referenced tab-delimited .txt files that are
used for further analysis steps in Perseus (www.maxquant.org) or R [38]
Fig. 4 Configuration of the RNA-Seq-derived protein sequences as search space in the Andromeda peptide
search engine of the MaxQuant computational proteomics platform [16, 39]. After installation of the MaxQuant
software package launch the “AndromedaConfig.exe” from the MaxQuant directory. The screenshot shows the
graphical user interface of version 1.3.0.5 with the EMBOSS “getorf”- processed Trinity de novo assembly
“Galaxy4-[getorf_on_data_1].fasta” selected as a new database
3.4 Annotation In the last step the novel sequences identified to be present in the
of Sequence Data sample on RNA and protein level are to be annotated based on
homology search to known sequence data. Annotating the whole
de novo-assembled transcriptome is time consuming. However,
taking the fraction that could be positively identified in the pro-
teomics experiment will reduce data amount and speed up the
analysis. Therefore the FASTA header of the protein sequences
reported in the MaxQuant output files is used to parse the custom
protein sequence database via regular expressions and extract the
228 Toni Luge and Sascha Sauer
4 Notes
Acknowledgement
References
1. Ong S-E, Blagoev B, Kratchmarova I et al variate hypergeometric analysis. J Proteome
(2002) Stable isotope labeling by amino acids Res 6:654–661
in cell culture, SILAC, as a simple and accurate 15. Shilov IV, Seymour SL, Patel AA et al (2007)
approach to expression proteomics. Mol Cell The Paragon Algorithm, a next generation
Proteomics 1:376–386 search engine that uses sequence temperature
2. Hsu J, Huang S, Chow N et al (2003) Stable- values and feature probabilities to identify pep-
isotope dimethyl labeling for quantitative pro- tides from tandem mass spectra. Mol Cell
teomics. Anal Chem 75:6843–6852 Proteomics 6:1638–1655
3. Wiese S, Reidegeld KA, Meyer HE et al (2007) 16. Cox J, Neuhauser N, Michalski A et al (2011)
Protein labeling by iTRAQ: a new tool for Andromeda: a peptide search engine integrated
quantitative mass spectrometry in proteome into the MaxQuant environment. J Proteome
research. Proteomics 7:340–350 Res 10:1794–1805
4. Gerber SA, Rush J, Stemman O et al (2003) 17. Nesvizhskii AI (2010) A survey of computa-
Absolute quantification of proteins and phos- tional methods and error rate estimation proce-
phoproteins from cell lysates by tandem dures for peptide and protein identification in
MS. Proc Natl Acad Sci U S A 100: shotgun proteomics. J Proteomics
6940–6945 73:2092–2123
5. Schwanhäusser B, Busse D, Li N et al (2011) 18. Wang X, Slebos RJC, Wang D et al (2012)
Global quantification of mammalian gene Protein identification using customized protein
expression control. Nature 473:337–342 sequence databases derived from RNA-Seq
6. Mann M, Kulak NA, Nagaraj N et al (2013) data. J Proteome Res 11:1009–1017
The coming age of complete, accurate, and 19. Metzker ML (2010) Sequencing technolo-
ubiquitous proteomes. Mol Cell 49:583–590 gies—the next generation. Nat Rev Genet
7. Gstaiger M, Aebersold R (2009) Applying 11:31–46
mass spectrometry-based proteomics to genet- 20. Evans VC, Barker G, Heesom KJ et al (2012)
ics, genomics and network biology. Nat Rev De novo derivation of proteomes from tran-
Genet 10:617–627 scriptomes for transcript and protein identifica-
8. Freiwald A, Weidner C, Witzke A et al (2013) tion. Nat Methods 9:1207–1211
Comprehensive proteomic data sets for study- 21. Lopez-Casado G, Covey PA, Bedinger PA et al
ing adipocyte-macrophage cell-cell communi- (2012) Enabling proteomic studies with RNA-
cation. Proteomics 13:3424–3428 Seq: the proteome of tomato pollen as a test
9. Meierhofer D, Weidner C, Hartmann L et al case. Proteomics 12:761–774
(2013) Protein sets define disease states and 22. Sheynkman GM, Shortreed MR, Frey BL et al
predict in vivo effects of drug treatment. Mol (2013) Discovery and mass spectrometric
Cell Proteomics 12:1965–1979 analysis of novel splice-junction peptides using
10. Mann M, Hendrickson RC, Pandey A (2001) RNA-Seq. Mol Cell Proteomics 12:2341–2353
Analysis of proteins and proteomes by mass 23. Adamidi C, Wang Y, Gruen D et al (2011) De
spectrometry. Annu Rev Biochem 70: novo assembly and validation of planaria tran-
437–473 scriptome by massive parallel sequencing and
11. Perkins DN, Pappin DJC, Creasy DM et al shotgun proteomics. Genome Res
(1999) Probability-based protein identification 21:1193–1200
by searching sequence databases using mass 24. He R, Kim M-J, Nelson W et al (2012) Next-
spectrometry data. Electrophoresis generation sequencing-based transcriptomic
20:3551–3567 and proteomic analysis of the common reed,
12. Eng JK, McCormack AL, Yates JR (1994) An Phragmites australis (Poaceae), reveals genes
approach to correlate tandem mass spectral involved in invasiveness and rhizome specificity.
data of peptides with amino acid sequences in a Am J Bot 99:232–247
protein database. J Am Soc Mass Spectrom 25. Song J, Sun R, Li D et al (2012) An improve-
5:976–989 ment of shotgun proteomics analysis by adding
13. Craig R, Beavis RC (2004) TANDEM: match- next-generation sequencing transcriptome data
ing proteins with tandem mass spectra. in orange. PLoS One 7, e39494
Bioinformatics 20:1466–1467 26. Romero-Rodríguez MC, Pascual J, Valledor L
14. Tabb DL, Fernando CG, Chambers MC et al (2014) Improving the quality of protein
(2007) MyriMatch: highly accurate tandem identification in non-model species.
mass spectral peptide identification by multi- Characterization of Quercus ilex seed and
232 Toni Luge and Sascha Sauer
Pinus radiata needle proteomes by using tion, visualization and analysis in functional
SEQUEST and custom databases. J Proteomics genomics research. Bioinformatics 21:
105:85–91 3674–3676
27. Wu X, Xu L, Gu W et al (2014) Iterative 34. Gillet LC, Navarro P, Tate S et al (2012)
genome correction largely improves proteomic Targeted data extraction of the MS/MS spectra
analysis of nonmodel organisms. J Proteome generated by data-independent acquisition: a
Res 13:2724–2734 new concept for consistent and accurate pro-
28. Woo S, Cha SW, Merrihew G et al (2014) teome analysis. Mol Cell Proteomics
Proteogenomic database construction driven 11:O111.016717
from large scale RNA-seq data. J Proteome Res 35. Egertson JD, Kuehn A, Merrihew GE et al
13:21–28 (2013) Multiplexed MS/MS for improved
29. Armengaud J, Trapp J, Pible O et al (2014) data-independent acquisition. Nat Methods
Non-model organisms, a species endangered 10:744–746
by proteogenomics. J Proteomics 105:5–18 36. Haas BJ, Papanicolaou A, Yassour M et al
30. Wang X, Zhang B (2014) Integrating genomic, (2013) De novo transcript sequence recon-
transcriptomic, and interactome data to improve struction from RNA-seq using the Trinity plat-
peptide and protein identification in shotgun form for reference generation and analysis. Nat
proteomics. J Proteome Res 13:2715–2723 Protoc 8:1494–1512
31. Wang X, Zhang B (2013) customProDB: an R 37. Rice P (2000) The European Molecular
package to generate customized protein data- Biology Open Software Suite EMBOSS: the
bases from RNA-Seq data for proteomics European Molecular Biology Open Software
search. Bioinformatics 29:3235–3237 Suite. Trends Genet 16:2–3
32. Luge T, Kube M, Freiwald A et al (2014) 38. R Development Core Team R (2011) R: a lan-
Transcriptomics assisted proteomic analysis of guage and environment for statistical comput-
Nicotiana occidentalis infected by “Candidatus ing. R Found Stat Comput 1:409
Phytoplasma mali” strain AT. Proteomics 39. Cox J, Matic I, Hilger M et al (2009) A practi-
14:1882–1889 cal guide to the MaxQuant computational plat-
33. Conesa A, Götz S, García-Gómez JM et al form for SILAC-based quantitative proteomics.
(2005) Blast2GO: a universal tool for annota- Nat Protoc 4:698–705
Chapter 17
Abstract
The proteomic study of non-model organisms, such as many crop plants, is challenging due to the lack
of comprehensive genome information. Changing environmental conditions require the study and selec-
tion of adapted cultivars. Mutations, inherent to cultivars, hamper protein identification and thus consid-
erably complicate the qualitative and quantitative comparison in large-scale systems biology approaches.
With this workflow, cultivar-specific mutations are detected from high-throughput comparative MS anal-
yses, by extracting sequence polymorphisms with de novo sequencing. Stringent criteria are suggested to
filter for confidential mutations. Subsequently, these polymorphisms complement the initially used data-
base, which is ready to use with any preferred database search algorithm. In our example, we thereby
identified 26 specific mutations in two cultivars of Pisum sativum and achieved an increased number
(17 %) of peptide spectrum matches.
Key words Proteomics, De novo sequencing, Polymorphism, Crop science, Cultivars, Mass spec-
trometry, Pisum sativum
1 Introduction
Jörg Reinders (ed.), Proteomics in Systems Biology: Methods and Protocols, Methods in Molecular Biology, vol. 1394,
DOI 10.1007/978-1-4939-3341-9_17, © Springer Science+Business Media New York 2016
233
234 Reinhard Turetschek et al.
Fig. 1 Workflow with main steps from initial DB compilation and de novo
sequence analysis to the application of the new organism aligned DB, explained
in detail as follows (cf. Subheadings 3.2–3.6)
2 Materials
2.1 Plant Material 1. Seeds from P. sativum ssp. cultivar Messire were provided by
the Institute for Sustainable Agriculture CISC (Department of
Plant Breeding, Cordoba, Spain). Cultivar Protecta was
obtained from Probstdorfer Saatzucht GmbH & Co KG
(Probstdorf, Austria).
236 Reinhard Turetschek et al.
3 Methods
3.3 Custom A composite protein-fasta file was created by merging the following
Database Design six databases:
Uniprot UniRef100 (all identical sequences and subfragments
with 11 or more residues are placed into a single record—http://
www.uniprot.org/help/uniref) sourced at 15-05-2013 from the
following Taxa:
1. Pisum sativum
2. Rhizobium leguminosarum
3. Glomus
4. Mycosphaerella
5. Legume-specific protein database (LegProt) [27] including
information from the following organisms: Pisum sativum,
Lotus japonicus, Medicago sativa, Glycine max, Lupinus albus,
Phaseolus vulgaris.
6. Processed dbEST NCBI sourced from http://www.coolsea-
sonfoodlegume.org/
Pisum Sativum Unigene v1, P. Sativum Unigene wa1, Pisum
Sativum Unigene v2;
Nucleotide sequences were six-frame-translated using mEM-
BOSS. For each accession number the longest continuous amino
acid sequence (longest ORF) within a frame was chosen. If multi-
ple sequences (of different frames) were of the same maximum
length, all of them were kept (each with a different accession num-
ber, including the frame number).
The 6 fasta files described above were combined, producing a
new fasta containing 135,754 entries. Protein sequences 100 %
identical in sequence and length were combined by subsequently
adding one header after the other, separating them by the follow-
ing characters “ __***__ ” (no matter if the redundancies origi-
nated from one or multiple fasta files). All other entries were simply
added to the end of the new file. The first accession number of the
header was repeatedly written at the very beginning of the header
line, separated by a “ | ” in order to consistently view and parse the
accession numbers.
3.4 De Novo Several automated software solutions for de novo sequencing are
Sequencing available to date. The outcome very much depends on the quality
and Homology Search of processed spectra and the selected algorithm [28]. Higher spec-
tra quality can be achieved by adaptation of the fragmentation (see
Note 1). Here, the de novo search was performed with PEAKS
[18] employing settings according to the resolution and mass
accuracy of the mass spectrometer used (see Subheading 2.3). By
calculating the narrowest possible mass error tolerance most occur-
ring PTMs cannot be mistaken for a mutation. Hence, a mass error
238 Reinhard Turetschek et al.
3.5 Evaluation Various automated programs [21, 23, 24] crucially simplify the
of Homology identification of sequence variance. However, these identifications
and Mutation must be critically filtered to obtain only confident sequence amend-
ments to the original database. First, exclusively proteotypic pep-
tides are added to the original database, because a variation in any
other peptide cannot be specifically attributed to one protein.
However, as a result of using a merged DB containing sequence
information of several RNA data (see Subheading 3.1), variations
are sometimes assigned to multiple protein entries with the same
function but slightly different sequences. In such case, the protein
entry with the most assigned peptides and highest coverage is cho-
sen for further processing. If the number of assigned peptides and
coverage is similar for several DB entries, all of them are chosen for
further processing. Second, the mutated peptide must not have a
non-mutated counterpart: if a mutated and a non-mutated peptide
are attributed to the same sequence in the database, the identified
mutation is likely to be a false positive. Third, the sequence of the
mutation must be confirmed by at least two MS/MS spectra.
Thereby, a de novo error—identifying a mutation—caused by a
low-quality spectrum is largely avoided and mutations gain confi-
dence. A typical quantitative shotgun proteomics experiment
requires the measurement of several replicates, which usually
acquire enough MS/MS to confirm mutations.
3.6 Database Confidently identified mutated amino acid residues are added to
Extension the database by copying the original fasta entries with the mutated
with Mutations sequence. Original entries must be kept, because the located muta-
tions may be characteristic for just one cultivar. The mutated
sequences are found in new entries with modified accession and
header (see example below).
Accession: ACU20233.1_m1
Header: unknown [glycine max] [Me_IV25]
A Proteomic Workflow Using High-Throughput De Novo Sequencing Towards… 239
3.8 Iterated Search The new extended DB was used for a standard DB search using the
with DB Search SEQUEST algorithm with the following settings: 5 ppm precursor
Algorithm mass tolerance, 0.5 Da fragment mass tolerance, acetylation of the
N-terminus, and oxidation of methionine as dynamic modifica-
tions. Minimum peptide confidence was set to medium, and mini-
mum Xcorr to 2. A minimum of two peptides per protein were
required for identification.
In the present study of P. sativum with the cultivars Messire
and Protecta we identified 48 variations to original DB entries, of
which 26 are mutations showing high cultivar specificity. Both cul-
tivars have five mutations in common. Messire showed 12 and
Protecta 9 characteristic mutations. Furthermore, 22 homologues
were identified from entries of different species (e.g., G. max from
the LegProt DB). The ratio of replaced and substituting amino
acids (Fig. 2) shows that most frequently valine and alanine are
both replacing and substituting other amino acids in our
experiment.
The number of peptide spectral matches (PSMs) shows how
many of fragmented ions match to the applied DB. Thus, a rather
complete DB will result in a higher number of PSMs compared to
an imperfect DB. Here Fig. 3 shows that the number of PSMs
increased significantly (17 %) for the two studied cultivars after
amending the initial DB with sequence variations. Besides improv-
ing protein identification, the elevated number of PSMs crucially
contributes to more accurate and confident quantification.
240 Reinhard Turetschek et al.
Fig. 2 Ratio of replaced (from the initial DB) and substituting amino acids
3000
# PSMs
2000
1000
0
Messire Protecta
original DB extended DB
Fig. 3 The total number of PSMs is significantly increased (student’s t-test, p < 0.05) in both cultivars when the
extended DB is applied; n = 24, error bars indicate standard error at 95 % confidence intervals
4 Notes
References
1. Fox JW, Ma L, Nelson K et al (2006) 12. Romero-Rodríguez MC, Pascual J, Valledor L
Comparison of indirect and direct approaches et al (2014) Improving the quality of protein
using ion-trap and Fourier transform ion identification in non-model species.
cyclotron resonance mass spectrometry for Characterization of Quercus ilex seed and
exploring viperid venom proteomes. Toxicon Pinus radiata needle proteomes by using
47(6):700–714 SEQUEST and custom databases. J Proteomics
2. Mackessy SP (2002) Biochemistry and phar- 105:85–91
macology of colubrid snake venoms. J Toxicol 13. Vanderschuren H, Lentz E, Zainuddin I et al
Toxin Rev 21(1–2):43–83 (2013) Proteomics of model and crop plant
3. Nawarak J, Sinchaikul S, Wu CY et al (2003) species: status, current limitations and strategic
Proteomics of snake venoms from Elapidae advances for crop improvement. J Proteomics
and Viperidae families by multidimensional 93:5–19
chromatographic methods. Electrophoresis 14. Morris HR, Williams DH, Ambler RP (1971)
24(16):2838–2854 Determination of the sequences of protein-
4. OmPraba G, Chapeaurouge A, Doley R et al derived peptides and peptide mixtures by mass
(2010) Identification of a novel family of snake spectrometry. Biochem J 125(1):189–201
venom proteins Veficolins from Cerberus ryn- 15. Johnson RS, Biemann K (1987) The primary
chops using a venom gland transcriptomics structure of thioredoxin from Chromatium
and proteomics approach. J Proteome Res vinosum determined by high-performance
9(4):1882–1893 tandem mass spectrometry. Biochemistry
5. Serrano SMT, Shannon JD, Wang D et al 26(5):1209–1214
(2005) A multifaceted analysis of viperid snake 16. Frank A, Pevzner P (2005) PepNovo: de novo
venoms by two-dimensional gel electrophore- peptide sequencing via probabilistic network
sis: an approach to understanding venom pro- modeling. Anal Chem 77(4):964–973
teomics. Proteomics 5(2):501–510 17. Fischer B, Roth V, Roos F et al (2005)
6. Wong ESW, Morgenstern D, Mofiz E et al NovoHMM: a hidden Markov model for de
(2012) Proteomics and deep sequencing com- novo peptide sequencing. Anal Chem
parison of seasonally active venom glands in 77(22):7265–7273
the platypus reveals novel venom peptides and 18. Ma B, Zhang K, Hendrie C et al (2003)
distinct expression profiles. Mol Cell PEAKS: powerful software for peptide de novo
Proteomics 11(11):1354–1364 sequencing by tandem mass spectrometry.
7. Abraham PE, Giannone RJ, Xiong W et al Rapid Commun Mass Spectrom 17(20):
(2014) Metaproteomics: extracting and min- 2337–2342
ing proteome information to characterize 19. Taylor JA, Johnson RS (1997) Sequence data-
metabolic activities in microbial communities. base searches via de novo peptide sequencing
Curr Protoc Bioinformatics 46:13.26. by tandem mass spectrometry. Rapid Commun
1–13.26.14 Mass Spectrom 11(9):1067–1075
8. Becher D, Bernhardt J, Fuchs S et al (2013) 20. Ma B, Johnson R (2012) De novo sequencing
Metaproteomics to unravel major microbial and homology searching. Mol Cell Proteomics
players in leaf litter and soil environments: 11(2):O111.014902
challenges and perspectives. Proteomics 21. Han Y, Ma B, Zhang K (2005) SPIDER: soft-
13(18–19):2895–2909 ware for protein identification from sequence
9. Hao C, Liu Q, Yang J et al (2008) tags with de novo sequencing error.
Metaproteomics: exploration of the functions J Bioinform Comput Biol 3(3):697–716
of microbial ecosystems. Chin J Appl Environ 22. Dasari S, Chambers MC, Slebos RJ et al
Biol 14(2):270–275 (2010) TagRecon: high-throughput mutation
10. Siggins A, Gunnigle E, Abram F (2012) identification through sequence tagging.
Exploring mixed microbial community func- J Proteome Res 9(4):1716–1726
tioning: recent advances in metaproteomics. 23. Searle BC, Dasari S, Turner M et al (2004)
FEMS Microbiol Ecol 80(2):265–280 High-throughput identification of proteins
11. Wang H-B, Zhang ZX, Li H et al (2011) and unanticipated sequence modifications
Characterization of metaproteomics in crop using a mass-based alignment algorithm for
rhizospheric soil. J Proteome Res 10(3): MS/MS de novo sequencing results. Anal
932–940 Chem 76(8):2220–2230
A Proteomic Workflow Using High-Throughput De Novo Sequencing Towards… 243
24. Tabb DL, Saraf A, Yates JR 3rd (2003) sequencing algorithms. J Proteome Res 5(11):
GutenTag: high-throughput sequence tagging 3018–3028
via an empirically derived fragmentation 29. Armengaud J, Trapp J, Pible O et al (2014)
model. Anal Chem 75(23):6415–6421 Non-model organisms, a species endangered
25. Lee FW, Lo SC (2008) The use of Trizol by proteogenomics. J Proteomics 105:5–18
reagent (phenol/guanidine isothiocyanate) for 30. Guthals A, Clauser KR, Frank AM et al
producing high quality two-dimensional gel (2013) Sequencing-grade de novo analysis of
electrophoretograms (2-DE) of dinoflagel- MS/MS triplets (CID/HCD/ETD) from
lates. J Microbiol Methods 73(1):26–32 overlapping peptides. J Proteome Res
26. Staudinger C, Mehmeti V, Turetschek R et al 12(6):2846–2857
(2012) Possible role of nutritional priming for 31. Frese CK, Altelaar AF, Hennrich ML et al
early salt and drought stress responses in (2011) Improved peptide identification by tar-
Medicago truncatula. Front Plant Sci 3:285 geted fragmentation using CID, HCD and
27. Lei Z, Dai X, Watson BS et al (2011) A legume ETD on an LTQ-Orbitrap Velos. J Proteome
specific protein database (LegProt) improves Res 10(5):2377–2388
the number of identified peptides, confidence 32. Gooley AA, Packer NH (1997) The impor-
scores and overall protein identification success tance of protein co- and post-translational
rates for legume proteomics. Phytochemistry modifications in proteome projects. In: Wilkins
72(10):1020–1027 MR et al (eds) Proteome research: new fron-
28. Pevtsov S, Fedulova I, Mirzaei H et al (2006) tiers in functional genomics. Springer, Berlin,
Performance evaluation of existing de novo pp 65–91
Chapter 18
Abstract
Quantitative proteomic experiments in recent years became almost routine in many aspects of biology.
Particularly the quantification of peptides and corresponding phosphorylated counterparts from a single experi-
ment is highly important for understanding of dynamics of signaling pathways. We developed an analytical
method to quantify phosphopeptides (pP) in relation to the quantity of the corresponding non-phosphorylated
parent peptides (P). We used mixed-mode solid-phase extraction to purify total peptides from tryptic digest and
separated them from most of the phosphorous-containing compounds (e.g., phospholipids, nucleotides) which
enhances pP enrichment on TiO2 beads. Phosphoproteomic data derived with this designed method allows
quantifying pP/P stoichiometry, and qualifying experimental data for mathematical modeling.
Key words Phosphopeptide enrichment, Mixed-mode solid-phase extraction, Metal oxide affinity
chromatography, Mathematical modeling
1 Introduction
Jörg Reinders (ed.), Proteomics in Systems Biology: Methods and Protocols, Methods in Molecular Biology, vol. 1394,
DOI 10.1007/978-1-4939-3341-9_18, © Springer Science+Business Media New York 2016
245
246 Maksim Zakhartsev et al.
(S)ignal
ATP ADP
receptor R
R′ level 1 R P
PP1
ATP ADP
Conserved moiety
Kinase K1
Kipool = Ki + K i′
K′2 level 2 K2 P
Ki K i′
+ =1
PP2 Ki pool Kipool
P
...
Kn−1
ATP ADP
target T
PPn
Fig. 1 A simplified hypothetical scheme of a signal (S) transfer from receptor (R) to
target (T) through a linear signal transduction pathway consisting of different
kinases (K). At each cascade level, only two interconvertable forms are shown:
active and inactive (marked by ′). The mechanism of activation is through phos-
phorylation by means of group transfer from phosphate-donor (e.g., ATP, GTP). The
active form of a species from a preceding level activates species at a subsequent
level. PPi—ith phosphatase. The figure was compiled from [1, 7]
1.2 Total Peptide The classical approach to purify total peptides from complex digest
Purification mixes exploits hydrophobic interactions between side chains of
hydrophobic amino acids and C18-groups of reverse-phase SPE sor-
bent [15]. However, this approach results in co-purification of a
variety of nonpolar components, such as lipids, pigments, or sterols.
This often is particularly problematic when working with strongly
pigmented plant tissues. At low pH (e.g., <3.0) the zwitterion of a
peptide becomes fully protonated (i.e., a weak cation or weak base)
which allows the use of strong cation exchanger (SCX) for their
purification, but it also results in co-purification of charged impuri-
ties such as nucleotides. Thus, a combination of the reverse-phase
and SCX modes provides enhanced peptide purification and addi-
tionally a better removal of nonpolar and charged impurities. Oasis®
MCX (mixed-mode cation-exchange and reversed-phase sorbent)
provides such dual modes of retention by (1) strong sulfonic-acid-
cation exchanger and (2) reversed-phase interactions in combination
with hydrophilic interactions on a single organic co-polymer. The
sorbent is highly selective and sensitive for extraction of basic com-
pounds from acidified biological matrices (such as plasma, urine,
bile, and pigmented plant tissue) and demonstrates very good capac-
ity for the peptide purification from tryptic protein digests. Sample
preparation based on use of mixed-mode solid phases provides supe-
rior removal of nearly all phospholipids and weaker acids, achieving
double-positive effects by (1) eliminating phosphorous-containing
compounds (phospholipids, nucleic acids, etc.) that strongly inter-
fere with phosphopeptide enrichment and (2) eliminating the major
sources of matrix effects (e.g., neutrals), a known case of ion
suppression, loss of sensitivity, and inaccurate quantification by liquid
chromatography with a tandem mass analyzer (LC-MS/MS).
1.3 Phosphopeptide The typical pP enrichment protocols rely on several unit operations,
(pP) Enrichment such as total peptide purification/desalting after the digest, pP
enrichment, and further desalting, in order to provide salt-free
samples for the LC-MS/MS analysis. Usually, all desalting steps are
248 Maksim Zakhartsev et al.
1.4 Peptide (P) There are different methods of peptide quantification in quantita-
and Phosphopeptide tive proteomics, which implement either absolute or relative pep-
(pP) Quantification tide quantifications. The quantification techniques are based on
the use of stable isotope labeling (e.g., SILAC), isobaric labeling
(e.g., TMT, iTRAQ), isotope-coded affinity tag (ICAT), internal
peptide standards [21], and label-free quantification (e.g., iBAQ).
The workflow we developed is based on label-free relative peptide
quantification (iBAQ by MaxQuant [22]) before and after pP
enrichment, using the amount (literally a mass in μg) of total pep-
tides and its loss during pP enrichment for the iBAQ value normal-
ization (Fig. 2). This approach allows us to estimate the degree of
phosphorylation of the signaling protein pool (see Note 1).
However, peptides and their phosphorylated counterparts have
significant differences in ionization/detection efficiency (so-called
flyability) [23]. Determination of flyability ratio for a particular
pP/P pair (based on the technical replicates and conserved moiety
assumption) allows to correct the signal intensities of the corre-
sponding species and to quantify the absolute phosphorylation stoi-
chiometry in each obtained pP/P pair [23]. This aspect also has to
be taken into account for the final quantification.
From Phosphoproteome to Modeling of Plant Signaling Pathways 249
Sample
(homogenate, CF, MF)
digest
mixed-mode SPE
on Oasis MCX
[tP]1
aliquot 1 aliquot 2
TiO2 -enrichment
[tP]2
LC-MS/MS LC-MS/MS
iBAQ1-j iBAQ2-j
2 Materials
2.3 In-Solution 1. Reduction buffer: 6.5 mM (or 1 μg/μL; w/v) DTT in water.
Digest 2. Alkylation buffer: 27 mM (or 5 μg/μL; w/v) iodoacetamide in
water.
3. 10 mM Tris–HCl, pH 8.0.
4. Lysyl endopeptidase (LysC) stock solution: 0.5 μg/μL
(Promega).
5. Trypsin stock solution: 0.5 μg/μL (Promega).
6. 2 % Trifluoroacetic acid (TFA) in water (v/v).
3. Water.
4. 2 % Formic acid in water (v/v).
5. 5 % Ammonium hydroxide in 80 % methanol (pH 11.0) (v/v).
6. Thermo-Strips and Caps, 8 × 0.2 mL (ThermoScientific).
3 Methods
3.2 Total Protein This protocol is adapted from [20]. All protein extraction steps
Extraction must be done on ice.
1. Weigh root samples (g of fresh weight; gFW), wrap them indi-
vidually in aluminum foil, and freeze them immediately in liq-
uid nitrogen (see Note 6).
2. Break the frozen samples into small pieces with the hammer
while keeping them wrapped in aluminum foil. Make sure that
the samples are constantly frozen after harvesting to avoid
rapid dephosphorylation.
3. Transfer the cell material into ice-cold homogenization buffer
in a ratio of 5 mL homogenization buffer per 1 gFW.
4. Resuspend the cell material thoroughly by gentle stirring or
shaking to get rid of clots.
5. Grind the sample manually in a Potter® homogenizer on ice,
50 smooth strokes per 7 mL sample.
6. Filter the homogenate through four layers of Miracloth® to get
rid of cell wall material and tissue debris.
7. Centrifuge the homogenate at 7.5 × 103 × g for 15 min at 4 °C
to get rid of unbroken cells and organelles.
8. Collect the supernatant.
9. Centrifuge the supernatant at 48 × 103 × g for 80 min at 4 °C to
precipitate microsomal vesicles.
10. Collect the supernatant which represents the cytosolic fraction
(i.e., water-soluble proteins, can be used for other
experiments).
11. Resuspend the pellet, which represents the microsomal frac-
tion (i.e., endomembranes and membrane-associated pro-
teins), in 500 μL of ice-cold UTU-buffer.
12. Rigorously vortex the suspension and ultra-sonicate it for
approximately 30 s.
13. Quantify total protein content in the microsomal fraction
using NanoOrange ® protein quantification kit and BSA stan-
dards in UTU buffer.
From Phosphoproteome to Modeling of Plant Signaling Pathways 253
3.5 C8-TiO2-StageTip 1. Activation of TiO2 beads: Activate TiO2 beads at 130 °C for
Column Preparation 30 min prior to use (see Note 3).
2. Preparing TiO2 bead stock suspension: Weigh 25 mg of TiO2
beads, resuspend it in 500 μL of 100 % methanol, and vortex
the suspension well.
3. Loading of TiO2 bead stock suspension: Load 20 μL of the
stock suspension (overall 1 mg; see Note 12) on top of the C8
disk in the 200 μL C8-StageTip (see Note 13).
4. Let the suspension settle down under the gravity force in order
to distribute the beads evenly.
5. Spin (see Note 14) the C8-TiO2-StageTip to force the solution
through.
4 Notes
ì iBAQ1. j
ï Pj =
ï m1
í (3)
ï pP = iBAQ 2. j ´ l
ï j m2
î
References
1. Klipp E, Liebermeister W (2006) Mathematical 8. Hein MY, Sharma K, Cox J et al (2013)
modeling of intracellular signaling pathways. Chapter 1—proteomic analysis of cellular
BMC Neurosci 7(Suppl 1):S10. doi:10.1186/ systems. In: Walhout AJM, Vidal M, Dekker
1471-2202-7-S1-S10 J (eds) Handbook of systems biology.
2. Mariottini C, Iyengar R (2013) Chapter 16—sys- Academic, San Diego, pp 3–25
tem biology of cell signaling. In: Walhout AJM, 9. Cox J, Mann M (2011) Quantitative, high-
Vidal M, Dekker J (eds) Handbook of systems resolution proteomics for data-driven systems biol-
biology. Academic, San Diego, pp 311–327 ogy. Annu Rev Biochem 80(1):273–299.
3. Duan G, Walther D, Schulze W (2013) doi:10.1146/annurev-biochem-061308-093216
Reconstruction and analysis of nutrient- 10. Choudhary C, Mann M (2010) Decoding sig-
induced phosphorylation networks in nalling networks by mass spectrometry-based
Arabidopsis thaliana. Front Plant Sci 4:540. proteomics. Nat Rev Mol Cell Biol 11(6):427–
doi:10.3389/fpls.2013.00540 439. doi:10.1038/nrm2900
4. Niittylä T, Fuglsang AT, Palmgren MG et al 11. Altelaar AFM, Munoz J, Heck AJR (2013)
(2007) Temporal analysis of sucrose-induced Next-generation proteomics: towards an inte-
phosphorylation changes in plasma membrane grative view of proteome dynamics. Nat Rev
proteins of arabidopsis. Mol Cell Proteomics Genet 14(1):35–48. doi:10.1038/nrg3356
6(10):1711–1726. doi:10.1074/mcp. 12. Olsen J, Macek B (2009) High accuracy mass
M700164-MCP200 spectrometry in large-scale analysis of protein
5. Schulze WX (2010) Proteomics approaches to phosphorylation. In: Lipton M, Paša-Tolic
understand protein phosphorylation in p athway L (eds) Mass spectrometry of proteins and
modulation. Curr Opin Plant Biol 13(3): peptides. Humana Press, Totowa, NJ,
279–286. doi:10.1016/j.pbi.2009.12.008 pp 131–142
6. Blagoev B, Ong S-E, Kratchmarova I et al 13. Schmelzle K, White FM (2006) Phosphoproteomic
(2004) Temporal analysis of phosphotyrosine- approaches to elucidate cellular signaling net-
dependent signaling networks by quantitative works. Curr Opin Biotechnol 17(4):406–414.
proteomics. Nat Biotechnol 22(9):1139–1145. doi:10.1016/j.copbio.2006.06.004
doi:10.1038/nbt1005 14. Larsen MR, Thingholm TE, Jensen ON et al
7. Kholodenko BN, Hoek JB, Westerhoff HV (2005) Highly selective enrichment of phos-
et al (1997) Quantification of information phorylated peptides from peptide mixtures
transfer via cellular signal transduction path- using titanium dioxide microcolumns. Mol
ways. FEBS Lett 414(2):430–434. Cell Proteomics 4(7):873–886. doi:10.1074/
doi:10.1016/S0014-5793(97)01018-1 mcp.T500007-MCP200
From Phosphoproteome to Modeling of Plant Signaling Pathways 259
15. Wisniewski JR, Zougman A, Nagaraj N et al H+ ATPase during pollen grain germination
(2009) Universal sample preparation method and tube growth. Planta 213(1):132–141.
for proteome analysis. Nat Methods 6(5): doi:10.1007/s004250000483
359–362. doi:10.1038/nmeth.1322 21. Pratt JM, Simpson DM, Doherty MK et al
16. Nakagami H (2014) StageTip-based (2006) Multiplexed absolute quantification for
HAMMOC, an efficient and inexpensive phos- proteomics using concatenated signature pep-
phopeptide enrichment method for plant shot- tides encoded by QconCAT genes. Nat Protoc
gun phosphoproteomics. In: Jorrin-Novo JV 1(2):1029–1043
et al (eds) Plant proteomics. Humana Press, 22. Cox J, Mann M (2008) MaxQuant enables
New York, pp 595–607 high peptide identification rates, individual-
17. Thingholm TE, Jorgensen TJD, Jensen ON ized p.p.b.-range mass accuracies and
et al (2006) Highly selective enrichment of proteome- wide protein quantification. Nat
phosphorylated peptides using titanium dioxide. Biotechnol 26(12):1367–1372. doi:10.1038/
Nat Protoc 1(4):1929–1935. doi:10.1038/ nbt.1511
nprot.2006.185 23. Steen H, Jebanathirajah JA, Springer M et al
18. Beckers GM, Hoehenwarter W, Röhrig H et al (2005) Stable isotope-free relative and abso-
(2014) Tandem metal-oxide affinity chroma- lute quantitation of protein phosphorylation
tography for enhanced depth of phosphopro- stoichiometr y by MS. Proc Natl Acad Sci
teome analysis. In: Jorrin-Novo JV et al (eds) U S A 102(11):3948–3953. doi:10.1073/
Plant proteomics. Humana Press, New York, pnas.0409536102
pp 621–632 24. Rappsilber J, Mann M, Ishihama Y (2007)
19. Colby T, Röhrig H, Harzen A et al (2011) Protocol for micro-purification, enrichment,
Modified metal-oxide affinity enrichment com- pre-fractionation and storage of peptides for
bined with 2D-PAGE and analysis of phospho- proteomics using StageTips. Nat Protoc 2(8):
proteomes. In: Dissmeyer N, Schnittger A 1896–1906
(eds) Plant kinases. Humana Press, New York, 25. Meier-Schellersheim M, Xu X, Angermann B
pp 273–286 et al (2006) Key role of local regulation in
20. Pertl H, Himly M, Gehwolf R et al (2001) chemosensing revealed by a new molecular
Molecular and physiological characterisation interaction-
based modeling method. PLoS
of a 14-3-3 protein from lily pollen grains reg- Comput Biol 2(7), e82. doi:10.1371/journal.
ulating the activity of the plasma membrane pcbi.0020082
Chapter 19
Abstract
In quantitative proteomics, large lists of identified and quantified proteins are used to answer biological
questions in a systemic approach. However, working with such extensive datasets can be challenging, espe-
cially when complex experimental designs are involved. Here, we demonstrate how to post-process large
quantitative datasets, detect proteins of interest, and annotate the data with biological knowledge. The
protocol presented can be achieved without advanced computational knowledge thanks to the user-friendly
Perseus interface (available from the MaxQuant website, www.maxquant.org). Various visualization tech-
niques facilitating the interpretation of quantitative results in complex biological systems are also
highlighted.
1 Introduction
Jörg Reinders (ed.), Proteomics in Systems Biology: Methods and Protocols, Methods in Molecular Biology, vol. 1394,
DOI 10.1007/978-1-4939-3341-9_19, © Springer Science+Business Media New York 2016
261
262 Elise Aasebø et al.
2 Material
3 Methods
3.1 Loading Data 1. Open the Perseus program (annotated with the Perseus logo)
in Perseus located in the main Perseus folder.
2. Download annotations: Click the blue tab indicated in Fig. 1a
and then Annotation download (Fig. 1b) in the menu thatw-
wshows up. Go to the provided Dropbox folder and download
the appropriate mainAnnot.txt.gz file in the OrganismSpecific
folder (in this example use mainAnnot.homo_sapiens.txt.gz),
put the downloaded annotation file into Perseus\conf\annotation.
Extract the zipped file (see Note 2).
3. Import your output file: Press the green upwards-pointing
arrow to load your data (Fig. 1c). When you hover over the
arrow you will get the message: Generic matrix upload.
4. After clicking the green arrow, a matrix opens where you can
upload your .txt file (Fig. 2), in this case use the
“proteinGroups_5cell-line-mix.txt” file (see Note 3). Transfer
quantitative data (ratios) into the Expression field (see Note 4).
You can also choose other parameters of interest, such as
“Number of proteins”, “Unique peptides”, etc. Put numerical
Interpretation of Quantitative Shotgun Proteomic Data 263
Fig. 1 When using Perseus for the first time, it is recommended to download
annotations (a, b). To import a data set (Generic matrix upload), press the green
arrow (c)
Fig. 2 Select the output file (.txt format) to upload (usually the ProteinGroups.txt file for MaxQuant).
The unique column headers in the output are listed in the left column. Select the columns to analyze in
Perseus and press the arrows to load them into the columns to the right. Be sure to load protein expression
data into the Expression fields and numerical information—such as the number of peptides or sequence
coverage—into the Numerical field
simply select the matrix and click the red cross. If you are using
Perseus version 1.5.0.15 or newer, you can (re-)name the
matrices by double clicking them (Fig. 3).
6. The MaxQuant output display the ratios as Heavy/Light, so in
order to compare all light samples against the heavy sample
(i.e., the internal standard) we have to invert the comparison
to get Light/Heavy. Go to: Basic → Transform, and write “1/
(x)” in the Transformation field.
7. Transform the expression values into log values (see Note 6).
Go to: Basic → Transform → choose “log2(x)”.
3.3 Normalization 1. The example dataset is now in log2 values; thus the normal
distribution should be centered on zero. Visualize the distri-
bution of the dataset by using a histogram. Go to:
Visualization → Histogram. Accept the default settings by
Interpretation of Quantitative Shotgun Proteomic Data 265
Fig. 3 Matrix overview. Each new task that changes the Data matrix will appear as a new matrix. It is possible
to navigate between different matrices, delete paths and (re-) name matrices, thus allowing inspecting inter-
mediate results
a b
Counts
Counts
0 5 0 5
MV4-11 MV4-11
Fig. 4 Histogram displaying the protein ratio distribution before (a) and after (b) normalization, exemplified by
two of the cell lines. The Histogram tab can be found next to the Data tab in the same matrix
3.4 Gene Annotation 1. Add categorical annotations. Go to: Annot. Columns → Add
annotation. Select the mainAnnot.homo_sapiens.txt file as the
Source (Fig. 5). Choose from “GOBP name” down to
“Keyword” in the Annotations to be added field and transfer
the selected annotations over to the empty field using the
arrow. Click OK.
266 Elise Aasebø et al.
Fig. 5 Add annotation—a prerequisite is that annotations are already downloaded (described in Subheading
3.1.2 and Fig. 1)
Fig. 6 Identify regulated proteins in each sample by using the Significance A test with the indicated parame-
ters. Regulated proteins are marked with the symbol “+” in the Data matrix
3.5 Statistical 1. Go to: Basic → Significance A. Select all cell lines and transfer
Evaluation them to the empty field using the arrow. In the Use for trunca-
tion field choose P value and use the default (0.05) as Threshold
value (Fig. 6). Click OK. The protein ratios that are significant
outliers relative to the sample population are now annotated
with “+” in the matrix.
2. Compare the cell lines derived from patients at time of diagno-
sis (MV4-11 and OCI-AML3) to the other cells lines derived
from patients during relapse (Molm-13, NB4 and THP-1).
Start by making two groups: Annot. rows → Categorical anno-
tations rows and specify the groups as “Diagnosis” or “Relapse”
as shown in Fig. 7. Click OK.
3. Do a two-samples t-test to compare the groups: Tests → Two
samples t-test. Select P value in the Use for truncation field and
keep 0.05 as Threshold value, and default settings for other
parameters (Fig. 8). Click OK.
Interpretation of Quantitative Shotgun Proteomic Data 267
Fig. 7 Categorical annotation rows is used to create groups. In this case we create “Group1” (default name)
and mark the samples with either “Diagnosis” or “Relapse”
Fig. 8 Find significantly expressed proteins between the groups “Diagnosis” or “Relapse” by applying a Two-
samples tests. Use the indicated parameters. The significantly expressed proteins are marked with the symbol
“+” in the Data matrix
3.6 Volcano Plot 1. Compare the two groups. Go to: Visualization → Scatter plot
and keep Columns in the Matrix access field. Click OK.
2. Find the Scatter plot tab next to the Data tab in the same
matrix. Select t-test difference and –Log t-test p value in the tabs
at right of the scatter plot (Fig. 9a).
3. To look for specific pathways or annotations go to the
Categories tab and select terms related to apoptosis (Fig. 9b).
4. Alternatively, go to: Misc. → Volcano Plot. Keep the default
parameters and click OK.
Fig. 9 Volcano plot. Start by creating a scatter plot and compare the t-test difference to the -Log t-test p value
(a) to visualize the volcano plot. Inspect enriched terms, such as “apoptosis,” by going to the Categories tab
(b). The proteins annotated by the term appear in red in the volcano plot
3. Find the PCA plot next to the Data tab and change from No
labels to All labels (Fig. 10a).
4. Color the groups “Diagnosis” and “Relapse” with two differ-
ent colors by clicking on the color symbol (Fig. 10b).
5. Select the proteins responsible for the separation in the PCA
plot by changing the mode from zoom (magnifying glass) to
select (square) (Fig. 10c). The selected proteins in this example
are marked in red in the PCA plot (Fig. 10d) and the protein
name is marked in blue in the box indicated in Fig. 10e.
3.8 Hierarchical 1. Normalize on the protein level: Before clustering you need to
Clustering normalize the protein values at the row level. Go to:
Normalization → Z-score and select Rows in the Matrix Access
field (see Note 8). Use default settings for the other parame-
ters. Click OK.
Interpretation of Quantitative Shotgun Proteomic Data 269
Fig. 10 Principal Component Analysis (PCA). Change from No labels to All labels (a) to reveal the sample names
of the dots in the PCA plot. Color the samples belonging to each group in different colors (b) to see if the cell
lines of one group cluster together. To look at the proteins that cause the separation of the samples, see the
box at the lower left. Change from the zoom to select option (c) and select the proteins to the left (d). Note that
the selected proteins appear with a blue line in the table at lower right (e)
Fig. 11 Hierarchical clustering. The clustering is performed both at protein (row) and sample (column) level. Specify the number of clusters (in this example two) at
both levels by clicking the symbols indicated in (a). Configure the row and column names by clicking the symbols indicated in (b). At the upper right there is a table
of Row clusters (c), you can select a cluster by clicking the line. The proteins of the selected cluster are displayed in the profile plot (d). By selecting Protein names
in (e), you can see the table of protein names in the Members tab
Interpretation of Quantitative Shotgun Proteomic Data 271
4 Notes
1. For more help and discussion with other users, visit the
Perseus Google Group at https://groups.google.com/
forum/#!forum/perseus-list.
2. You need a program to extract zipped files. Here we used
7-Zip File Manager, downloaded freely from the internet.
After installing 7-Zip, right click on the zipped folder and
“extract” the folder.
3. You can upload tab separated files, such as the .txt files from a
MaxQuant search, but also output files from other software
and types of experiments (genomics, transcriptomics, metabo-
lomics, etc.), as long as the file contains a unique column
header and is in the .txt format. In this example we look at the
protein expression data, but you can also explore and analyze
peptide and modification data with this software.
4. The expression values will differ between ratios (in case of
SILAC, TMT, iTRAQ, etc.) and intensities (in case of label
free), depending on your experiment.
5. In general you should have expression values in minimum 50
% of your samples, this allows for better statistics and cluster-
ing. It is also possible to specify the minimum number of valid
values in each group or in at least one group in the Mode field.
This can be relevant if a protein is expressed in one group and
absent in another group.
6. If you have intensity values you might use a base 10 logarith-
mic transformation, while base 2 logarithm is recommended
for ratios and is used in this example.
272 Elise Aasebø et al.
Acknowledgements
References
1. Aebersold R, Mann M (2003) Mass spectrometry- 3. Altelaar AF, Munoz J, Heck AJ (2013) Next-
based proteomics. Nature 422:198–207 generation proteomics: towards an integrative view
2. Olsen JV, Mann M (2013) Status of large-scale of proteome dynamics. Nat Rev Genet 14:35–48
analysis of post-translational modifications by 4. Vaudel M, Sickmann A, Martens L (2010)
mass spectrometry. Mol Cell Proteomics Peptide and protein quantification: a map of
12:3444–3452 the minefield. Proteomics 10:650–670
Interpretation of Quantitative Shotgun Proteomic Data 273
Abstract
Targeting subproteomes is a good strategy to decrease the complexity of a sample, for example in body
fluid biomarker studies. Glycoproteins are proteins with carbohydrates of varying size and structure
attached to the polypeptide chain, and it has been shown that glycosylation plays essential roles in several
vital cellular processes, making glycosylation a particularly interesting field of study. Here, we describe a
method for the enrichment of glycosylated peptides from trypsin digested proteins in human cerebrospinal
fluid. We also describe how to perform the data analysis on the mass spectrometry data for such samples,
focusing on site-specific identification of glycosylation sites, using user friendly open source software.
1 Introduction
Jörg Reinders (ed.), Proteomics in Systems Biology: Methods and Protocols, Methods in Molecular Biology, vol. 1394,
DOI 10.1007/978-1-4939-3341-9_20, © Springer Science+Business Media New York 2016
275
276 Astrid Guldbrandsen et al.
2 Materials
3 Methods
20. Add 200 μL 100 mM Ambic to the beads and wash for 5 min
at 20 °C.
21. Collect the supernatant and pool with the peptides collected
in step 19.
22. Acidify the sample by adding 7 μL 5 M HCl, drop by drop.
Keep lid open to avoid pressure building up inside the tube.
23. Add 200 μL 0.1 % FA and vortex.
24. Perform clean-up at 4 °C as is described in steps 8 and 12, but
this time use Oasis μElution plates (see Note 4) and 500 μL 80
% ACN–0.1 % FA for conditioning and elution and 500 μL 0.1
% FA for washing, and place the sample tube in the magnetic
rack before transfer to the Oasis plate to remove any remain-
ing beads. Elute with 150 μL × 2.
25. Transfer sample (300 μL) to a new Eppendorf tube, concen-
trate to dryness and freeze at −80 °C until LC-MS analysis.
26. Dissolve in appropriate solvent and volume for LC-MS analy-
sis (see Note 5).
3.2 Data The data interpretation consists of two main parts: (1) match the
Interpretation spectra to a database, and (2) interpret the matches to infer pro-
teins and glycosylation sites. For the first part, so-called search
engines are used, retrieving a list of Peptide Spectrum Matches
(PSMs). In the second, the PSMs are assembled into inferred pep-
tides and proteins, the quality of the identification results is evalu-
ated in order to limit the prevalence of false positive hits, and
post-translational modification (PTM) sites are inferred. More
details on the identification process can be found in the following
reviews [26–28].
In this chapter, the above task is demonstrated using user
friendly open source software, SearchGUI (version 1.20.8) and
PeptideShaker (version 0.33.6). These tools notably present the
advantage to support multiple search engines for PTM studies in a
user friendly environment. For more details on how to operate
these tools, please refer to the respective free tutorials [29] (http://
compomics.com/bioinformatics-for-proteomics). Note that the
concepts introduced here can be transposed to most proteomics
applications, like OpenMS [30, 31], the TransProteomic Pipeline
(TPP) [32], or MaxQuant [33].
3.3 Database Search 1. After starting SearchGUI, Click Add and select the spectrum
files to search in the Input & Output panel at the top of the
dialog shown in Fig. 1. Here, spectrum files consist of peak
lists of the original MS2 spectra in the mgf format (http://
www.matrixscience.com/help/data_file_help.html#GEN).
To convert raw data to the mgf format it is recommended to
use ProteoWizard [34]. In this experiment, fractionation has
been performed, so there is a total of 20 files to be searched.
280 Astrid Guldbrandsen et al.
Fig. 1 SearchGUI main dialog. The main dialog appearing when the tool is started. Spectrum files, search set-
tings, and output folder have to be loaded and specified. The search engines shown are all automatically
selected, but can be unchecked. The search can be automatically processed in PeptideShaker if this option is
selected
Fig. 2 SearchGUI search settings dialog. This dialog appears when editing the search settings, the parameters
used in this experiment are selected. The database (FASTA file) is the human complement of the UniProtKB/
Swiss-Prot database available from the UniProt website (www.uniprot.org)
3.5 Data Export Project features and result reports containing the possible glycosyl-
ation sites (at the protein, peptide, and PSMs level) can be exported
by pressing Export → Identification Features. In the Export Features
window displayed in Fig. 5 the type of report to export can be
chosen and custom reports created (see Notes 7 and 8).
4 Notes
right. At the bottom of the display, the protein sequence is displayed with the identified peptides color coded. Note that peptides can be navigated by clicking the
sequence. Deamidated asparagines (glycosylation sites) are highlighted in the peptide and protein sequences using the color coding from the search parameters.
More details on the project are available via the other tabs in the upper right corner
283
284 Astrid Guldbrandsen et al.
Fig. 4 PeptideShaker’s Spectrum & Fragment Ions display. The Spectrum & Fragment Ions panel displays
information on the annotation of the spectrum based on the selected peptide. At the top left, the intensity of
the fragment ions annotated on the spectrum is illustrated with bars at the possible fragmentation site on the
peptide sequence, where the modification is color coded. The intensities of b and y ions are in blue and red,
respectively. Here, the deamidation of the asparagine on the ENAT motif is displayed in brown and clearly
flanked with fragment ions. At the top center, a histogram shows the respective shares of annotated and not
annotated peaks, in green and grey, respectively, using intensity bins. At the top right, the m/z deviation of
every fragment ion is plotted against the fragment ion m/z. Below, the spectrum is displayed with the anno-
tated peaks in red. It is possible to customize the annotation of the spectrum using the menu at the bottom
Fig. 5 PeptideShaker export features dialog. This dialog is available from the Export → Identification Features.
Under the panel Reports, several default reports can be selected and exported, while custom made reports can
be created and edited by right-clicking the table
Acknowledgements
References
1. Gevaert K, Van Damme P, Ghesquiere B et al ical insights and biomedical implications. Cell
(2007) A la carte proteomics with an emphasis on Mol Life Sci 64:2133–2152
gel-free techniques. Proteomics 7:2698–2718 6. Varki A (1993) Biological roles of oligosaccha-
2. Zhang H, Li XJ, Martin DB et al (2003) rides: all of the theories are correct.
Identification and quantification of N-linked Glycobiology 3:97–130
glycoproteins using hydrazide chemistry, stable 7. Roth J (2002) Protein N-glycosylation along
isotope labeling and mass spectrometry. Nat the secretory pathway: relationship to organ-
Biotechnol 21:660–666 elle topography and function, protein quality
3. Gamblin DP, Scanlan EM, Davis BG (2009) control, and cell interactions. Chem Rev
Glycoprotein synthesis: an update. Chem Rev 102:285–303
109:131–163 8. Sato Y, Endo T (2010) Alteration of brain gly-
4. Shental-Bechor D, Levy Y (2008) Effect of gly- coproteins during aging. Geriatr Gerontol Int
cosylation on protein folding: a close look at 10 Suppl 1:S32–S40
thermodynamic stabilization. Proc Natl Acad 9. Ruggeri ZM, Mendolicchio GL (2007)
Sci U S A 105:8256–8261 Adhesion mechanisms in platelet function. Circ
5. Sola RJ, Rodriguez-Martinez JA, Griebenow K Res 100:1673–1685
(2007) Modulation of protein biophysical 10. Berger MS, Locher GW, Saurer S et al (1988)
properties by chemical glycosylation: biochem- Correlation of c-erbB-2 gene amplification and
286 Astrid Guldbrandsen et al.
protein expression in human breast carcinoma interface for simultaneous OMSSA and
with nodal status and nuclear grading. Cancer X!Tandem searches. Proteomics 11:996–999
Res 48:1238–1243 23. Vaudel M, Burkhart JM, Zahedi RP et al
11. Hudziak RM, Schlessinger J, Ullrich A (1987) (2015) PeptideShaker enables reanalysis of
Increased expression of the putative growth MS-derived proteomics data sets. Nature bio-
factor receptor p185HER2 causes transforma- technology 33:22–24
tion and tumorigenesis of NIH 3T3 cells. Proc 24. Vizcaino JA, Deutsch EW, Wang R et al (2014)
Natl Acad Sci U S A 84:7159–7163 ProteomeXchange provides globally coordi-
12. Vogelzang NJ, Lange PH, Goldman A et al nated proteomics data submission and dissemi-
(1982) Acute changes of alpha-fetoprotein and nation. Nat Biotechnol 32:223–226
human chorionic gonadotropin during induc- 25. Martens L, Hermjakob H, Jones P et al (2005)
tion chemotherapy of germ cell tumors. Cancer PRIDE: the proteomics identifications data-
Res 42:4855–4861 base. Proteomics 5:3537–3545
13. Bosl GJ, Lange PH, Fraley EE et al (1981) 26. Vaudel M, Sickmann A, Martens L (2012)
Human chorionic gonadotropin and alphafeto- Current methods for global proteome iden-
protein in the staging of nonseminomatous tes- tification. Expert Rev Proteomics 9:
ticular cancer. Cancer 47:328–332 519–532
14. Thompson DK, Haddow JE (1979) Serial 27. Nesvizhskii AI (2010) A survey of computa-
monitoring of serum alpha-fetoprotein and tional methods and error rate estimation pro-
chorionic gonadotropin in males with germ cedures for peptide and protein identification
cell tumors. Cancer 43:1820–1829 in shotgun proteomics. J Proteomics
15. Catalona WJ, Richie JP, Ahmann FR et al 73:2092–2123
(1994) Comparison of digital rectal examina- 28. Chalkley RJ, Clauser KR (2012) Modification
tion and serum prostate specific antigen in the site localization scoring: strategies and perfor-
early detection of prostate cancer: results of a mance. Mol Cell Proteomics 11:3–14
multicenter clinical trial of 6,630 men. J Urol 29. Vaudel M, Venne AS, Berven FS et al (2014)
151:1283–1290 Shedding light on black boxes in protein iden-
16. Canney PA, Moore M, Wilkinson PM et al tification. Proteomics 14:1001–1005
(1984) Ovarian cancer antigen CA125: a pro- 30. Kohlbacher O, Reinert K, Gropl C et al (2007)
spective clinical assessment of its role as a TOPP—the OpenMS proteomics pipeline.
tumour marker. Br J Cancer 50:765–769 Bioinformatics 23:e191–e197
17. Topol EJ, Byzova TV, Plow EF (1999) Platelet 31. Bertsch A, Gropl C, Reinert K et al (2011)
GPIIb-IIIa blockers. Lancet 353:227–231 OpenMS and TOPP: open source software for
18. Guldbrandsen A, Vethe H, Farag Y et al (2014) LC-MS data analysis. Methods Mol Biol 696:
In-depth characterization of the cerebrospinal 353–367
fluid proteome displayed through the CSF 32. Deutsch EW, Mendoza L, Shteynberg D et al
Proteome Resource (CSF-PR). Mol Cell (2010) A guided tour of the Trans-Proteomic
Proteomics 13(11):3152–63 Pipeline. Proteomics 10:1150–1159
19. Tian Y, Zhou Y, Elliott S et al (2007) Solid- 33. Cox J, Mann M (2008) MaxQuant enables
phase extraction of N-linked glycopeptides. high peptide identification rates, individualized
Nat Protoc 2:334–339 p.p.b.-range mass accuracies and proteome-
20. Berven FS, Ahmad R, Clauser KR et al (2010) wide protein quantification. Nat Biotechnol
Optimizing performance of glycopeptide cap- 26:1367–1372
ture for plasma proteomics. J Proteome Res 34. Kessner D, Chambers M, Burke R et al (2008)
9:1706–1715 ProteoWizard: open source software for rapid
21. Gonzalez J, Takao T, Hori H et al (1992) A proteomics tools development. Bioinformatics
method for determination of N-glycosylation 24:2534–2536
sites in glycoproteins by collision-induced 35. Apweiler R, Bairoch A, Wu CH et al (2004)
dissociation analysis in fast atom bombard- UniProt: the Universal Protein knowledge-
ment mass spectrometry: identification of the base. Nucleic Acids Res 32:D115–D119
positions of carbohydrate-linked asparagine 36. Vaudel M, Burkhart JM, Sickmann A et al
in recombinant alpha-amylase by treatment (2011) Peptide identification quality control.
with peptide-N-glycosidase F in 18O-labeled Proteomics 11:2105–2114
water. Anal Biochem 205:151–158 37. Flicek P, Amode MR, Barrell D et al (2014)
22. Vaudel M, Barsnes H, Berven FS et al (2011) Ensembl 2014. Nucleic Acids Res 42:
SearchGUI: an open-source graphical user D749–D755
Chapter 21
Abstract
In biological and medical sciences, high throughput analytical methods are now commonly used to investigate
samples of different conditions, e.g., patients versus controls. Systemic functional analyses emerged as a
reference method to go beyond a list of regulated compounds, and identify activated or inactivated bio-
logical functions. This approach holds the promise for a better understanding of biological systems, of the
mechanisms involved in disease progression, and thus improved diagnosis, prognosis, and treatment. In
this chapter, we present a simple workflow to conduct pathway analyses on biological data using the freely
available Reactome platform (http://www.reactome.org).
1 Introduction
Jörg Reinders (ed.), Proteomics in Systems Biology: Methods and Protocols, Methods in Molecular Biology, vol. 1394,
DOI 10.1007/978-1-4939-3341-9_21, © Springer Science+Business Media New York 2016
287
288 Luis Francisco Hernández Sánchez et al.
2 Material
3 Methods
Fig. 6 The option on the Tools menu at the homepage to go to the Pathway Browser of Reactome
13. In the upper right corner, click the down arrow button to
show the Diagram Key, describing the shapes corresponding to
every type of entity in the diagram. To hide the key, click the
arrow button again. Note that proteins are shown as rectangles
with rounded corners.
14. In the Hierarchy Panel to the left, select the reaction called
Transphosphorylation of JAK1 inside the same sub-pathway
Interferon gamma signaling.
15. Right click on the protein PTPN6, and select the option Other
pathways to show the diagram of other pathways related to that
protein. For the protein uniquely involved in this pathway, the
label No other pathways is shown.
16. On the Hierarchy Panel on the left, select Binding of STAT1 to
p-IFNGR1. Right click on the protein STAT1-1 and choose
Display interactors to show the compounds interacting with
this protein in this pathway. Here 10 out of 52 interactors
associated to this protein are highlighted with new entity boxes
with blue thick borders. Note the small white square at the top
right corner of the entity box of the protein indicating the
number of interactors (Fig. 10).
17. You can switch to other pathway diagrams at any time by
selecting another pathway, sub-pathway or reaction name in
the hierarchy panel. The details panel at the bottom will be
updated according to what is currently selected in the hierar-
chy panel or the pathway diagram.
This section describes how to analyze quantitative data using
Reactome with the dataset material indicated in Subheading 2,
item 1. First, the analysis of the list of protein accessions will be
used to demonstrate how to find pathways of interest, a use case
relevant to both qualitative and quantitative datasets. Second, the
quantitative information will be provided to Reactome along with
the protein identifiers.
1. From the Reactome homepage (Fig. 1), click Analyze Data to
show the analysis tools (Fig. 11).
2. Prepare a data file containing the list of proteins according to
the required format:
(a) Create a new text file and name it protein_list.txt.
(b) In Supplementary Table S2 of [15], go to the worksheet
called Results_Merged_Five_and_four_IS. There you will
find the table entitled Supplementary Table 2: Merged data
from the five and four cell lines experiments.
(c) Select the protein accessions, here the column called
Protein IDs, along with the header, copy the data into the
text file and save the file.
Systemic Analysis of Regulated Functional Networks 297
Fig. 10 The interactors of protein STAT1-1 in the Phosphorylation of STAT1 by JAK kinases reaction
(d) Change the first row of the file from Protein IDs to #GBM
Uniprot (see Note 1 and 2).
(e) Delete any empty lines after the protein list.
3. Upload the data file with the set of proteins to analyze:
(a) First click the Browse button to select your protein_list.txt
file.
(b) Make sure that the checkbox Project to human is selected,
as the identifiers to analyze are related to human
pathways.
(c) Click Analyze to upload the file and start the analysis of the
inserted data.
4. As an alternative, you can simply paste the information in the
textbox within the Analysis Tools section.
(a) In the Analyze Data screen, click Click here to paste you
data or try example data sets… to display the input field.
At the right you can also select example datasets.
298
Luis Francisco Hernández Sánchez et al.
Fig. 11 The Data Analysis screen with the Analysis tools of Reactome
Systemic Analysis of Regulated Functional Networks 299
Fig. 13 The diagram for Translation pathway partially colored in yellow to indicate the percentage of structures in the sample data set found in this pathway
301
302
Luis Francisco Hernández Sánchez et al.
Fig. 14 The diagram for Eukaryotic Translation Elongation sub-pathway with partially colored entity boxes
Systemic Analysis of Regulated Functional Networks 303
Fig. 15 The table of participating components of the protein set EEF1A1-like proteins highlighting in yellow the
table cells of the components found in the sample protein data set
Fig. 17 Diagram panel with analysis results coloring entities according to the submitted numerical data
305
306 Luis Francisco Hernández Sánchez et al.
4 Notes
Fig. 21 Diagram of the Lipoprotein metabolism pathway after the analysis of the submitted data
Systemic Analysis of Regulated Functional Networks 309
Acknowledgements
References
1. Mouse Genome Sequencing Consortium, outstanding challenges. PLoS Comput Biol 8,
Waterston RH, Lindblad-Toh K et al (2002) e1002375
Initial sequencing and comparative analysis of 7. Vizcaino JA, Mueller M, Hermjakob H et al
the mouse genome. Nature 420:520–562 (2009) Charting online OMICS resources: a
2. Rosen R (1968) A means toward a new holism. navigational chart for clinical researchers.
Science 161:34–35 Proteomics Clin Appl 3:18–29
3. Barsnes H, Martens L (2013) Crowdsourcing 8. Kanehisa M (1997) A database for post-
in proteomics: public resources lead to better genome analysis. Trends Genet 13:375–376
experiments. Amino Acids 44:1129–1137 9. Ogata H, Goto S, Sato K et al (1999) KEGG:
4. Vaudel M, Sickmann A, Martens L (2014) Kyoto Encyclopedia of Genes and Genomes.
Introduction to opportunities and pitfalls in Nucleic Acids Res 27:29–34
functional mass spectrometry based pro- 10. Pico AR, Kelder T, van Iersel MP et al (2008)
teomics. Biochim Biophys Acta 1844:12–20 WikiPathways: pathway editing for the people.
5. Muller T, Schrotter A, Loosse C et al (2011) PLoS Biol 6, e184
Sense and nonsense of pathway analysis software 11. Croft D, O’Kelly G, Wu G et al (2011)
in proteomics. J Proteome Res 10:5398–5408 Reactome: a database of reactions, pathways
6. Khatri P, Sirota M, Butte AJ (2012) Ten years and biological processes. Nucleic Acids Res
of pathway analysis: current approaches and 39:D691–D697
310 Luis Francisco Hernández Sánchez et al.
12. Takami H, Taniguchi T, Moriya Y et al (2012) nated proteomics data submission and dissemi-
Evaluation method for the potential function- nation. Nat Biotechnol 32:223–226
ome harbored in the genome and metage- 17. Martens L, Hermjakob H, Jones P et al (2005)
nome. BMC Genomics 13:699 PRIDE: the proteomics identifications data-
13. Shannon P, Markiel A, Ozier O et al (2003) base. Proteomics 5:3537–3545
Cytoscape: a software environment for inte- 18. Cox J, Mann M (2008) MaxQuant enables high
grated models of biomolecular interaction net- peptide identification rates, individualized p.p.b.-
works. Genome Res 13:2498–2504 range mass accuracies and proteome-wide protein
14. Kisseleva T, Bhattacharya S, Braunstein J et al quantification. Nat Biotechnol 26:1367–1372
(2002) Signaling through the JAK/STAT 19. Bertsch A, Gropl C, Reinert K et al (2011)
pathway, recent advances and future chal- OpenMS and TOPP: open source software for
lenges. Gene 285:1–24 LC-MS data analysis. Methods Mol Biol
15. Aasebo E, Vaudel M, Mjaavatten O et al 696:353–367
(2014) Performance of super-SILAC based 20. Deutsch EW, Mendoza L, Shteynberg D et al
quantitative proteomics for comparison of dif- (2010) A guided tour of the Trans-Proteomic
ferent acute myeloid leukemia (AML) cell Pipeline. Proteomics 10:1150–1159
lines. Proteomics 14(17-18):1971–6 21. Vaudel M, Burkhart JM, Zahedi RP et al (2015)
16. Vizcaino JA, Deutsch EW, Wang R et al (2014) PeptideShaker enables reanalysis of MS-derived
ProteomeXchange provides globally coordi- proteomics data sets. Nat Biotechnol 33:22–24
INDEX
A Glycolysis .....................................................................57–73
Glycopeptides .................................................. 163–178, 275,
Adipocytes ....................................................................57, 58 277–279, 282
Affinity purification-mass spectrometry (APMS), ..181–183 Glycoproteomics.......................................................275–284
Antigen presentation ................................................189–208
Assay design .................................................................47–49 H
Assay validation ...................................................... 50–51, 54
High-performance liquid chromatography
B (HPLC) ........................12, 16–18, 20–23, 27, 28,
30, 31, 34, 35, 39, 45, 63, 78, 81, 89, 90, 93, 94,
Biomarkers ..............................................44, 87, 88, 151, 275 96, 97, 104–106, 183, 185, 194, 198, 199, 202,
206, 215, 216
C
Human leukocyte antigen (HLA) ................... 189–192, 194,
Cell signaling pathways ............................................181–186 199, 201, 204, 205
Chemical cross-linking ......................109, 112, 114–115, 124 Hydrophilic interaction liquid chromatography
Chemical proteomics ................................................211–218 (HILIC) ...........................26, 28, 33–35, 40, 152,
Collision-induced dissociation (CID) ............... 8, 60, 97, 98, 155–156, 207
120, 122, 171, 172, 176, 178, 240
Cotton ...................................................................... 152, 160 I
Crop science .............................................................233–241 ImagePrep .........................................130, 131, 137–142, 145
Cryoconserved tissue ........................................ 131, 132, 135 Immobilized metal ion affinity chromatography
Cultivar..................................................... 234, 235, 238–240 (IMAC) ................................................ 88, 90, 94
Immune response .....................................................189–208
D
In-gel digestion ........................113, 116–119, 165, 168–169,
Data interpretation ................................................... 276, 279 171–172, 174–175
Data post-processing ........................................................261 In-solution digestions .......................113–114, 116, 119, 165,
De Novo Sequencing................................................233–241 171, 174, 175
Drug discovery .................................................................212 Isobaric labels ...............................................................3, 248
Isoelectric focusing ......................................... 16–18, 22, 220
E iTRAQ ................................... 3, 15–40, 87, 97, 98, 101, 217,
Enrichment .............................. 2, 3, 30, 32, 33, 39, 152, 158, 219, 248, 270
164–165, 167–169, 172, 177, 183, 212, 247, 261,
K
267, 272, 276–279
Ethyl esterification (EE) ..........................................151–161 Kinases ....................................... 59, 212, 245, 246, 289–292,
295, 297
F
L
Formalin fixed paraffin embedded (FFPE) ............. 130–132,
135–137 Label-free proteomics.......................................................102
Functional Networks ................................................287–307 Linkage-specific ....................................................... 152, 157
Functional proteomics ......................................................287 Liquid chromatography-mass spectrometry
(LC-MS) ...................... 25–28, 30, 35, 48, 50, 55,
G 79–80, 89–91, 94, 95, 97, 98, 114, 119–120, 185,
Gene expression................................................................229 192–194, 199–201, 205–207, 220, 222, 226, 236,
Gene/protein expression ..................................................221 251, 255, 276, 279
Jörg Reinders (ed.), Proteomics in Systems Biology: Methods and Protocols, Methods in Molecular Biology, vol. 1394,
DOI 10.1007/978-1-4939-3341-9, © Springer Science+Business Media New York 2016
311
PROTEOMICS IN SYSTEMS BIOLOGY: METHODS AND PROTOCOLS
312 Index