+ All Categories
Home > Documents > [Methods in Molecular Biology] Mass Spectrometry Data Analysis in Proteomics Volume 1007 ||...

[Methods in Molecular Biology] Mass Spectrometry Data Analysis in Proteomics Volume 1007 ||...

Date post: 16-Dec-2016
Category:
Upload: rune
View: 223 times
Download: 9 times
Share this document with a friend
19
Chapter 16 Proteomic Strategies to Characterize Signaling Pathways H.C. Harsha, Sneha M. Pinto, and Akhilesh Pandey Abstract Cells respond to external stimuli by transducing signals through a series of intracellular molecules and eliciting an appropriate response. The cascade of events through which the signals are transduced include post-translational modifications such as phosphorylation and ubiquitylation in addition to formation of multi-protein complexes. Improvements in biological mass spectrometry and protein/peptide microarray technology have tremendously improved our ability to probe proteins, protein complexes, and signaling pathways in a high-throughput fashion. Today, a single mass spectrometry-based investigation of a signaling pathway has the potential to uncover the large majority of known signaling intermediates painstakingly characterized over decades in addition to discovering a number of novel ones. Here, we discuss various proteomic strategies to characterize signaling pathways and provide protocols for phosphoproteomic analysis. Key words Signaling pathways, Mass spectrometry, Phosphorylation, Enrichment of phosphopro- teomes, Quantitative phosphoproteomics, Cancer 1 Introduction 1.1 Mass Spectrometry to Characterize Signaling Pathway Intermediates The last decade has witnessed tremendous progress in mass spectrometry approaches that has enabled unbiased global protein profiling studies. Enrichment and fractionation strategies have also evolved that enable deeper sampling of specific sub-proteomes of interest. Phosphorylation is one of the major post-translational modifications (PTMs) through which signals are transduced in a cell. Methods to enrich phosphorylated peptides have contributed significantly to our ability to characterize phosphoproteomes of cells and tissues. One major methodology that is used for enrich- ment is the use of antibodies that specifically enrich for tyrosine [13] or serine/threonine [4] phosphorylated proteins. These anti- phosphotyrosine and anti-phospho-serine/threonine antibodies also enable visualization of global activation of signaling molecules upon a stimulus. Following receptor stimulation using an appro- priate ligand, signaling intermediates that are phosphorylated in an inducible manner can be immunoprecipitated and visualized by Rune Matthiesen (ed.), Mass Spectrometry Data Analysis in Proteomics, Methods in Molecular Biology, vol. 1007, DOI 10.1007/978-1-62703-392-3_16, # Springer Science+Business Media, LLC 2013 359
Transcript
Page 1: [Methods in Molecular Biology] Mass Spectrometry Data Analysis in Proteomics Volume 1007 || Proteomic Strategies to Characterize Signaling Pathways

Chapter 16

Proteomic Strategies to Characterize Signaling Pathways

H.C. Harsha, Sneha M. Pinto, and Akhilesh Pandey

Abstract

Cells respond to external stimuli by transducing signals through a series of intracellular molecules andeliciting an appropriate response. The cascade of events through which the signals are transduced includepost-translational modifications such as phosphorylation and ubiquitylation in addition to formation ofmulti-protein complexes. Improvements in biological mass spectrometry and protein/peptide microarraytechnology have tremendously improved our ability to probe proteins, protein complexes, and signalingpathways in a high-throughput fashion. Today, a single mass spectrometry-based investigation of a signalingpathway has the potential to uncover the large majority of known signaling intermediates painstakinglycharacterized over decades in addition to discovering a number of novel ones. Here, we discuss variousproteomic strategies to characterize signaling pathways and provide protocols for phosphoproteomicanalysis.

Key words Signaling pathways, Mass spectrometry, Phosphorylation, Enrichment of phosphopro-teomes, Quantitative phosphoproteomics, Cancer

1 Introduction

1.1 Mass

Spectrometry

to Characterize

Signaling Pathway

Intermediates

The last decade has witnessed tremendous progress in massspectrometry approaches that has enabled unbiased global proteinprofiling studies. Enrichment and fractionation strategies have alsoevolved that enable deeper sampling of specific sub-proteomes ofinterest. Phosphorylation is one of the major post-translationalmodifications (PTMs) through which signals are transduced in acell. Methods to enrich phosphorylated peptides have contributedsignificantly to our ability to characterize phosphoproteomes ofcells and tissues. One major methodology that is used for enrich-ment is the use of antibodies that specifically enrich for tyrosine[1–3] or serine/threonine [4] phosphorylated proteins. These anti-phosphotyrosine and anti-phospho-serine/threonine antibodiesalso enable visualization of global activation of signaling moleculesupon a stimulus. Following receptor stimulation using an appro-priate ligand, signaling intermediates that are phosphorylated in aninducible manner can be immunoprecipitated and visualized by

Rune Matthiesen (ed.), Mass Spectrometry Data Analysis in Proteomics, Methods in Molecular Biology, vol. 1007,DOI 10.1007/978-1-62703-392-3_16, # Springer Science+Business Media, LLC 2013

359

Page 2: [Methods in Molecular Biology] Mass Spectrometry Data Analysis in Proteomics Volume 1007 || Proteomic Strategies to Characterize Signaling Pathways

Western blotting using the same antibodies. Although thesephosphorylated proteins can be visualized as distinct bands byWestern blotting, the identity of these proteins cannot be deter-mined. Tyrosine kinase signaling pathways were some of the firstpathways to be characterized using mass spectrometry. This waslargely enabled owing to the availability of robust antibodies toenrich tyrosine phosphoproteome. Since then, phosphoproteomicapproaches have been extensively used to characterize severalsignaling intermediates of growth factor and cytokine receptorsignaling pathways [4–6]. In addition, this method has been usedto characterize various immune signaling pathways as well as cancersignaling pathways. Salomon et al. employed phosphoproteomicsstrategy and showed tyrosine phosphorylation dynamics of tyrosinephosphosites in human T-cell signaling and BCR-ABL signaling[7]. In recent years, phosphoproteomic approaches in conjunctionwith stable isotope labeling strategies have resulted in characteriza-tion of thousands of molecules in a signaling pathway in a singleexperiment [8, 9].

1.2 Experimental

Strategies for

Enrichment of

Phosphoproteomes

Low stoichiometry of phosphoproteome necessitates use of enrich-ment methods to obtain detectable amounts of phosphopeptidesfrom biological samples. Effective enrichment of tyrosine phospho-proteins/peptides has been achieved using anti-phosphotyrosineantibodies. In particular, three monoclonal antibodies, 4G10,p-Tyr-100, and pY20, have been extensively used for tyrosine phos-phoprotein/peptide enrichment. These antibodies are known tohave overlapping but somewhat different specificities based on thesequence context surrounding the phosphotyrosine residues [10].A combination of these antibodies is likely to yield better coverageof tyrosine phosphoproteome than using each antibody in isolation.

In addition to antibody-based approaches, strategies that relyon chromatography-based methods as well as metal affinity-basedenrichment approaches referred to as immobilized metal affinitychromatography (IMAC) have also evolved for enrichment ofphosphopeptides [11–14]. Chromatography-based methods forpreferential enrichment of phosphopeptides include strong cationexchange (SCX) chromatography [15], strong anion exchange(SAX) chromatography [16], hydrophilic interaction liquid chro-matography (HILIC) [17], and electrostatic repulsion-hydrophilicinteraction chromatography (ERLIC) [18]. Chromatography-based methods often utilize the difference in the physicochemicalcharacteristics of the phosphopeptide as discriminating factorsto separate phosphopeptides from non-phosphorylated peptides.For example, phosphopeptides elute in the early fractions in SCXchromatography (at pH 2.7)-based fractionation as tryptic phos-phopeptides possess a lower net charge owing to negatively chargedphosphate moieties.

360 H.C. Harsha et al.

Page 3: [Methods in Molecular Biology] Mass Spectrometry Data Analysis in Proteomics Volume 1007 || Proteomic Strategies to Characterize Signaling Pathways

Several immobilized metal affinity matrices have beenemployed including Fe3+, Ga3+, and Zr4+ [13, 19, 20]. One ofthe commonly employed methods for phosphopeptides is TiO2-based enrichment. This method is now routinely used to profilethousands of phosphorylation sites in various experimental con-texts. Some studies have also employed combinatorial strategieswhere SCX chromatography is first used to enrich phosphopeptidesfollowed by subsequent IMAC/TiO2-based enrichment for betterselectivity and specificity [21–23].

Antibody-based strategies are superior for enrichment of tyro-sine phosphopeptides, whereas TiO2- [24–26] and IMAC-basedstrategies have been found to be efficient at enriching serine/thre-onine phosphopeptides. Large-scale signaling studies have takenadvantage of this ability to identify and characterize large numberof mediators of cell signaling. In addition to phosphorylation, massspectrometric methods are also being used to investigate role ofacetylation, glycosylation, and ubiquitination in signal transductionpathways. For instance, the interplay of phosphorylation andO-GlcNAcylation and its implications on signaling is already welldocumented [27].

1.3 Quantitative

Phosphoproteomic

Studies that Have

Characterized Specific

Signaling Pathways

Several signaling pathways have now been characterized using massspectrometry-based global phosphoproteomic profiling. Thesestudies typically use cells that are either stimulated by specificligands followed by phosphoproteomics to identify pathway sub-strates (Fig. 1). Proteins that undergo phosphorylation upon acti-vation of specific signaling pathways are concluded as downstreameffectors of respective signaling pathways. EGF signaling was oneof the first signaling pathways to be characterized in a comprehen-sive fashion by mass spectrometry. Blagoev et al. employed aSILAC (stable isotope labeling by amino acids in cell culture)-based strategy coupled to an antibody-based affinity purificationmethod for enriching EGF-induced phosphoproteins. The experi-ment assessed protein phosphorylation dynamics across five timepoints post-EGF stimulation and identified about 81 effectors in asingle experiment [28]. Subsequently, Zhang et al. studied EGFRactivation in a time-dependent manner by employing both anti-body- and IMAC-based enrichment strategies and identified sev-eral downstream effectors [29]. Olsen et al. characterizedhundreds of site-specific changes downstream of EGF stimulationby employing a TiO2-based phosphopeptide enrichment strategy[30]. More recently, similar phosphoproteomic strategies havebeen employed to characterize kinase signaling pathways includingc-Src [31–35], ERBB2 [36, 37], Insulin [38], EphB2 [39, 40],FGF [41, 42], FGF-2 [43, 44], IL-2 [45], TGF-β [46], and TSLP[47] among others.

Proteomic Strategies to Characterize Signaling Pathways 361

Page 4: [Methods in Molecular Biology] Mass Spectrometry Data Analysis in Proteomics Volume 1007 || Proteomic Strategies to Characterize Signaling Pathways

No treatment Treatment with ligand

Grow in regular media Cells grown in stable isotope labeledamino acid contaning media

Rel

ativ

e A

bund

ance

m/z0

100 Pathway substrate

Unchanged by ligand

LC-MS analysis

Phosphopeptide enrichment

Fig. 1 Identification of pathway substrates by phosphoproteomics. Phosphopep-tides are enriched from stimulated and unstimulated cellular states and analyzedby mass spectrometry. Proteins/peptides from ligand-stimulated state areencoded using stable isotopes (asterisk) which introduces a mass shift thusfacilitating determination of relative abundance by mass spectrometry. Redpeptides represent an example of a pathway substrate that undergoes anincrease in phosphorylation after stimulation of cells with a specific ligand

Page 5: [Methods in Molecular Biology] Mass Spectrometry Data Analysis in Proteomics Volume 1007 || Proteomic Strategies to Characterize Signaling Pathways

1.4 Mass

Spectrometry to

Determine Dynamic

States of Proteins

in Signaling Pathways

Incorporation of quantitative proteomic strategies such as SILAC,iTRAQ, and TMT has tremendously improved our ability to deter-mine quantitative differences in protein expression across cellularstates [48–50]. These labeling strategies and label-free strategieshave played a vital role in subsequent years to characterize dynamicchanges in protein modifications as well as variations in proteincomplexes in signaling pathways [28, 29, 51, 52]. To characterizedynamic states of protein complexes in signaling pathways, recruit-ment of proteins upon activation of specific pathways can be deter-mined by mass spectrometry.

1.5 Determining

Kinase-Substrate

Motifs from High-

Throughput

Phosphoproteomic

Data

Kinases are known to recognize their substrates based on specificsequence motifs flanking the phosphorylation site. Identification ofspecific kinase substrate motifs provides opportunity to predictpotential targets of kinases. Global phosphoproteomic studies gen-erate large amounts of phosphopeptide data that can be utilized toidentify enriched sequences flanking phosphorylation sites. Tilldate, more than 300 kinase substrate motifs have been describedin the literature [53].

Over the past decade, several computational methods/toolshave been developed to predict potential PTM sites in proteinsequences. These predict phosphorylation sites in a kinase-dependent or kinase-independent fashion [54]. Kinase-dependentapproaches are based on the presumption that each kinase has aunique sequence specificity and thus substrates of these kinases canbe predicted using information available on kinase-specific motifsfrom literature curated databases such as Phospho.ELM [55],PhosphoSite [56], and HPRD [57]. Several prediction tools havebeen developed such as Scansite [58] and NetPhorest [59] whichpredict kinases from linear motifs using artificial neural networksor position-specific scoring matrices (PSSM). NetPhos [60] andKinasePhos [61] are other prediction tools that predict kinasephosphorylation sites based on available dataset of known phos-phorylation sites.

1.6 Mass

Spectrometry

Approaches to

Determine Aberrantly

Activated Signaling

Pathways in Diseases

Global phosphoproteomic profiling studies are now beingextensively carried out to characterize aberrantly regulated signaltransduction pathways in various diseases including cancers. Dys-regulation of kinase signaling pathways in cancers is now wellunderstood [62] and has led to the development of targetedtherapeutic strategies where a mutant kinase or an aberrantly acti-vated kinase is targeted for therapeutic purposes. Examples of thisinclude chronic myelogenous leukemia (CML) where BCR-ABL, afusion kinase is the therapeutic target and is treated with a smallmolecule inhibitor imatinib [63]; non-small cell lung cancer wherea mutant EGFR is targeted using small molecule inhibitor erlotinib[64]; and breast cancers where HER2/neu is targeted using anantibody [65].

Proteomic Strategies to Characterize Signaling Pathways 363

Page 6: [Methods in Molecular Biology] Mass Spectrometry Data Analysis in Proteomics Volume 1007 || Proteomic Strategies to Characterize Signaling Pathways

These successful outcomes have provided an impetus todeveloping strategies for identifying aberrant kinases in other diseasesincluding various cancers. We have previously shown the utility ofphosphoproteomics in identifying aberrantly activated kinase signal-ing pathways in pancreatic cancer [66]. Using an antibody-basedstrategy to enrich and compare tyrosine phosphoproteins from anon-neoplastic cell line and a neoplastic cell line of pancreas, aberrantactivation of EGFR pathway was identified in a subset of pancreaticcancers. The role of this activation of EGFR in driving cancer cellproliferation was tested by carrying out mouse xenograft studieswhere the implanted tumors were shown to regress upon treatmentwith an EGFR inhibitor. Other cancers where phosphoproteomicprofiling studies have been employed to identify driver kinases orkinase-driven signaling pathways include leukemia [67], sarcomas[68], non-small cell lung cancers [69], renal cell carcinoma [70],glioblastoma [71], melanoma [72], and colon cancer [73].

Phosphoproteomic approaches have also been employed tostudy phosphorylation changes resulting from specific drug treat-ment [74]. Huang et al. employed phosphotyrosine strategy toexplore therapeutic strategy for glioblastoma [75]. Phosphopro-teomics is also being employed to investigate drug resistance andescape mechanisms in cancers. Rexer et al. generated six lapatinibresistant HER2 overexpressing human breast cancer cell lines toinvestigate resistance mechanisms. The resistant lines were found toutilize Src family kinase-driven pathways as escape mechanism [76].Andersen et al. carried out pathway-based phosphoprofiling toidentify biomarkers to predict responsiveness to targeted therapy.They showed pPRAS40 (Thr246) as a predictor of PI3K pathwayactivation and AKT inhibitor sensitivity [77].

1.7 Protein and

Peptide Microarray

Strategies to Study

Signaling

In addition to mass spectrometry, protein and peptide microarraystrategies also offer an opportunity to characterize signaling path-ways in a high-throughput manner. Protein microarrays have beenused in the past to characterize signaling pathways, to identifykinase substrates, and to study recruitment potential of intracellularphosphorylated regions of receptor tyrosine kinases [78–80].Reverse-phase protein microarrays (RPPAs) have been extensivelyused for studying aberrantly activated signaling pathways in cancers[81]. RPPAs are often printed with normal and cancer cell/tissuelysates and are probed using antibodies against activated forms orphosphorylated forms of specific proteins. Using this methodology,hundreds of cell/tissue lysates can be probed in a single experimentin an array format [82]. Protein microarrays have been used exten-sively for characterizing signaling pathways in various cancersincluding prostate cancer [83], ovarian cancer [84], and T-cellacute lymphoblastic leukemia [85]. In addition, protein microarraytechnology has also been used to identify molecular networksassociated with antiestrogen resistance in breast cancers [86], to

364 H.C. Harsha et al.

Page 7: [Methods in Molecular Biology] Mass Spectrometry Data Analysis in Proteomics Volume 1007 || Proteomic Strategies to Characterize Signaling Pathways

monitor response to kinase inhibitors [87], and to study aberrantactivation of signaling molecules in head and neck cancers [88].Peptide microarrays are also being used for high-throughputscreening of kinase substrates and also to characterize motifs recog-nized by specific kinases [33, 89].

1.8 Challenges

in Phosphorylation

Analysis

The most commonly used fragmentation method in proteomics iscollision-induced dissociation (CID). Fragmentation of serine/threonine phosphopeptides by CID generally yields fragmentsthat have lost the phosphate moieties. These precursor ions thathave lost the phosphate moiety generally dominate the MS/MSspectrum. This insufficient fragmentation of the phosphopeptidedirectly impacts peptide identification and phosphorylation sitedetermination. Electron transfer dissociation (ETD) has beenshown to be a better strategy for fragmentation while dealingwith labile modifications like O-GlcNAc and serine/threoninephosphorylation [26]. Trypsin remains the most widely used pro-tease in mass spectrometry-based proteomic analysis due to itsnumerous advantages; however, the routine use of trypsin forprotein digestion does not yield a comprehensive picture of allpotential phosphorylation events because some of the tryptic pep-tides are undetectable in MS (e.g., either too short or too long).Employing multiple proteases that have site-specific cleavage activ-ity have been shown to significantly improve sequence coverage inglobal proteomic analysis and the same strategy can be used tocomplement and improve phosphoproteome coverage [91].

Search engines such as Sequest, Mascot, and X!Tandem areemployed for protein identifications from MS fragment spectra.However, the fragmentation information may not be sufficient toaccurately assign the position of phosphorylation site. Sometimes,manual inspection MS/MS spectra is carried out to deduce themodified residue, although this is a daunting task and not feasiblefor large-scale phosphoproteomic experiments involving thousandsof MS/MS spectra. The exact localization of phosphorylation sitesnecessitates comparison of the observed spectra with theoreticalspectra containing all possible permutations of the phosphategroup being placed on possible sequence position. Computationaltools such as Ascore [92], MaxQuant [93], PhosCalc [94], andPhosphoRS [95] among others, have been developed which utilizealgorithms to improve peptide identification and localization of thesite of phosphorylation when multiple sites of phosphorylation maybe present. Ascore and PhosphoRS are probability-based scoringsystems which utilize peptide sequence identified in protein data-base search to generate a list of all possible combination of phos-phopeptides. The site localization by all these tools depends uponsite-determining fragment ions. Based on these ions, these algo-rithms calculate site-specific scores based on binomial probabilitiesand individual site probabilities.

Proteomic Strategies to Characterize Signaling Pathways 365

Page 8: [Methods in Molecular Biology] Mass Spectrometry Data Analysis in Proteomics Volume 1007 || Proteomic Strategies to Characterize Signaling Pathways

1.9 Signal

Transduction Pathway

Resources

Signaling pathways have been painstakingly characterized over theyears by careful work on individual effectors in a particular signalingpathway. This information is scattered in the scientific literature.There are various public resources including KEGG [96], Biocarta,and NetPath that provide organized information on signaling path-ways and pathway intermediates. NetPath is one of the mostdetailed pathway resources of human signaling pathways [97].

Phosphoproteomic studies generate enormous amount ofdata. One of the goals of phosphoproteomic studies in diseasecontexts is to identify pathways that are altered in pathologicalconditions. The readout obtained in these studies is the sum totalof signaling activity in those biological samples. Several bioinfor-matics approaches are being employed to determine activated sig-naling pathways. Most of these approaches rely on datasets fromknown literature and some form of enrichment analysis to identify ahigher likelihood of a given pathway to be altered. Various types ofexperimental data are utilized for this including co-expression,protein–protein interactions, colocalization and also known infor-mation on the involvement of molecules in specific pathways basedon the existing literature. We anticipate that, in the future, signaltransduction pathway resources will become truly comprehensiveand enable development of better tools and algorithms to discoveraberrant signaling pathways from high-throughput phosphopro-teomic and other proteomic studies.

2 Materials

2.1 Materials

for Characterizing

Signaling Pathways

Using Quantitative

Phosphoproteomics

1. Cell culture medium depleted of lysine and arginine aminoacids.

2. Stable isotope-labeled amino acids—13C6-lysine (CambridgeIsotopes, Catalog # CLM-2247-H-1) and 13C6-arginine(Cambridge Isotopes, Catalog # CLM-2265-H-1).

3. Fetal bovine serum.

4. Appropriate ligand to stimulate the pathway under investiga-tion.

5. Urea (Thermo Scientific, Catalog # 29700).

6. HEPES (Sigma-Aldrich, Catalog # H4034-100G).

7. Complete protease inhibitor cocktail tablets (Roche, Catalog #12715300).

8. Sodium orthovanadate (Sigma-Aldrich, Catalog # S6508).

9. Sodium pyrophosphate (Sigma-Aldrich, Catalog # S6422).

10. β-glycerophosphate (Sigma-Aldrich, Catalog #50020).

11. Dithiothreitol (DTT) (Sigma-Aldrich, Catalog # D9779-10G).

366 H.C. Harsha et al.

Page 9: [Methods in Molecular Biology] Mass Spectrometry Data Analysis in Proteomics Volume 1007 || Proteomic Strategies to Characterize Signaling Pathways

12. Iodoacetamide (IAA) (Sigma-Aldrich, Catalog # I1149-5G).

13. Sequencing grademodified trypsin (Promega,Catalog#V5111).

14. Sep-Pak Classic C18 cartridges (WATERS, Catalog #WAT051910).

15. Trifluoroacetic acid (TFA) (Fisher Scientific Catalog #A116-50).

16. Pierce BCA protein assay kit (Thermo Scientific, Catalog #23225).

17. Triethylammonium bicarbonate (TEABC stock) buffer 1.0 M,pH 8.4–8.6 (Sigma-Aldrich, Catalog #. 17902) 100 ml(>98.5 % purity).

18. XBridge C18, 5 μm 250 � 4.6 mm analytical column(Waters, Milford, MA, Catalog #. 186003117) for bRPLCfractionation.

19. Titansphere 10 or 5 μm (GL Sciences, Inc., Japan Catalog #5020-75010).

20. Anti-phosphotyrosine antibody 4G10 agarose conjugate(Millipore, Catalog # 16-101) or anti-phosphotyrosine mousemAb (p-Tyr-100) (sepharose conjugate) (Cell Signaling Tech-nology, Catalog # 9419).

21. 2, 5-Dihydroxybenzoic acid (DHB) (Sigma-Aldrich, Catalog#. 149357-100G).

22. Ammonium hydroxide (J.T. Baker, Catalog #. 9721-01).

23. Empore C18 Disk (Catalog # 98060402181).

2.2 Solvents 1. Solvent A: 7 mM TEABC in water (Add 7 ml TEABC stockto 1 L) (pH 9).

2. Solvent B: 7 mM TEABC in 90 % acetonitrile (ACN) (JTBaker, 99.9 % pure), (Add 7 ml TEABC stock to 1 L of 90 %ACN).

3. Wash solution 1(80 % ACN, 1 % TFA, HPLC grade).

4. DHB solution (5 % 2,5-dihydroxybenzoic acid (DHB), i.e.,50 mg of DHB in 1 ml of Wash solution 1).

5. Ammonia solution (2 % NH4OH, pH ~10.5, i.e., 20 μl ofNH4OH in 1 ml of 40 % ACN).

2.3 Equipment 1. Centrifuge with refrigeration.

2. Sonicator—Branson Sonifier 250.

3. Lyophilizer.

4. High-performance liquid chromatography system.

5. High-resolution mass spectrometer interfaced with nanoscaleliquid chromatography system.

Proteomic Strategies to Characterize Signaling Pathways 367

Page 10: [Methods in Molecular Biology] Mass Spectrometry Data Analysis in Proteomics Volume 1007 || Proteomic Strategies to Characterize Signaling Pathways

3 Methods

3.1 Stable Isotope

Labeling by Amino

Acids in Cell Culture

For SILAC-based experiments, it is essential to adapt the cells inmedia supplemented with heavy isotopic forms of lysine and argi-nine amino acids (http://www.silac.org). Also, refer to Harshaet al. [90] for a detailed protocol on adapting the cells to SILACmedia. To carry out deep proteomic profiling of tyrosine andserine/threonine phosphoproteome, it is desirable to obtain25–30 mg of proteins after combining lysates from both unlabeledand labeled cell population. Estimating one million cells to yieldapproximately 100 μg of protein, one would require ~1.5 � 108

for an experiment. Some laboratories are able to use significantlylower amount of starting material without compromising the depthof sampling—we anticipate that more and more laboratories will beable to do this in the future.

1. Culture 150 million cells in SILAC medium and same amountof cells in standard medium.

2. Once cells have reached the desired density, serum starve bothcell populations overnight.

3. Stimulate the labeled cell population with appropriate ligand forpredetermined time period (seeNote 1).

4. Lyse the cells in 9 M urea lysis buffer.

5. Sonicate cell lysate for efficient lysis (3 Woutput, duty cycle-40,3 cycles of 10–15 bursts per cycle).

6. Centrifuge the lysates at 12,000 � g for 15 min.

7. Collect the supernatant and estimate protein concentrationusing the BCA method.

8. Pool lysates equivalent to 13 mg protein from both unstimu-lated and stimulated conditions.

9. Reduce the proteins by adding DTT to a final concentration of5 mM and incubating at 56 �C for 45 min.

10. Bring the lysate to room temperature and alkylate the proteinsby adding IAA to a final concentration of 10 mM and incubat-ing in the dark for 30 min.

11. Dilute the lysate to obtain a final urea concentration of �2 Musing HEPES buffer.

12. Add trypsin to obtain an enzyme to substrate ratio of 1:20(w/w).

13. Incubate at 37 �C overnight.

14. Acidify the peptide digest by adding TFA to a final concentra-tion of 1 %.

15. Centrifuge at 4,000 � g for 15 min.

368 H.C. Harsha et al.

Page 11: [Methods in Molecular Biology] Mass Spectrometry Data Analysis in Proteomics Volume 1007 || Proteomic Strategies to Characterize Signaling Pathways

16. Collect the supernatant and clean up the sample using C18

Sep-Pak cartridge.

17. Aliquot 5 mg equivalent peptide digest into a separate tube.

18. Freeze the tubes containing 5 mg equivalent peptide and20 mg equivalent peptide digest using liquid nitrogen/dryice/�80 �C freezer.

19. Lyophilize the samples for 2–3 days (see Note 2).

3.2 Enrichment

of Tyrosine

Phosphopeptides

1. Dissolve 20 mg Sep-Pak purified lyophilized peptide digest in1.5 ml of HEPES buffer.

2. Sonicate the tube in a water bath sonicator for 10 min tofacilitate dissolution of peptides.

3. Centrifuge the tube at 12,000 � g for 5 min and recover thesupernatant into a fresh tube.

4. Check the pH using pH paper strip and ensure that the pH is7–8. If not, adjust the pH using 1 M Tris base (see Note 3).

5. Resuspend one vial of agarose conjugated pY100 anti-phosphotyrosine antibody from Cell Signaling Technologyor 50 μg equivalent agarose conjugated 4G10 anti-phosphotyrosine antibody fromMillipore in 1�HEPES buffer(see Note 4).

6. Gently mix by inverting the tubes 4–5 times to obtain homog-enous suspension (see Note 5).

7. Spin the tubes at 4,000 � g for 1 min at 4 �C and carefullyremove and discard the supernatant leaving 50 μl of bufferbehind (see Note 6).

8. Transfer the peptide sample into the microfuge tube containingthe washed agarose-conjugated anti-phosphotyrosine antibody.

9. Incubate the tube on a rotator at 4 �C for 2 h. Ensure that thespeed is adjusted to maintain a homogenous suspension ofthe beads.

10. Centrifuge the tube at 500 � g for 1 min and discard thesupernatant.

11. Wash the beads by adding 1.5 ml of HEPES buffer and mixingit gently by inverting the tube 4–5 times to obtain a homoge-nous suspension.

12. Aspirate the supernatant using a pipette.

13. Repeat steps 10–12 two times.

14. Wash the beads twice in 1 ml distilled water as described insteps 10–12.

15. For eluting the phosphopeptides, add 75 μl of 0.1 % TFA andtap it gently to obtain homogenous suspension.

Proteomic Strategies to Characterize Signaling Pathways 369

Page 12: [Methods in Molecular Biology] Mass Spectrometry Data Analysis in Proteomics Volume 1007 || Proteomic Strategies to Characterize Signaling Pathways

16. Leave the tube at room temperature for 5 min.

17. Centrifuge at 500 � g for 1 min.

18. Aspirate the supernatant without disturbing the beads using apipette with a gel loading tip.

19. Repeat steps 15–18.

20. Clean the eluate using C18 ZipTip.

3.3 C18 StageTip

Clean Up

1. Prepare a C18 StageTip using Empore C18 disk. Wash the C18

disk in the following order:

l 80 % ACN, 0.5 % acetic acid solution.

l 50 % ACN, 0.1 % formic acid.

l 0.15 % TFA.

2. Load the combined eluate onto a C18 StageTip. Elute thesample into microfuge tube and reload the eluate.

3. Wash the bound peptides with 35 μl of 0.15 % TFA. Check forthe presence of beads. Ensure that no beads are present.

4. Elute the bound peptides with 10 μl of 50 % ACN, 0.1 % formicacid (see Note 7).

3.4 Enrichment of

Phosphopeptides

using TiO2

1. Dissolve the Sep-Pak purified and lyophilized peptidedigest (5–10 mg) in 2 ml of basic RPLC Solution A (pH 9)(see Note 8).

2. Centrifuge the tube at 16,000 � g for 5 min at 4 �C andtransfer the supernatant to a microfuge tube.

3. Fractionate the peptide digest using basic reverse phase chro-matography. Collect fraction in a 96-well plate (1 ml capacity)(see Note 9).

4. Dry the fractions at freezing temperature.

5. Add 400 μl of 80 % acetonitrile with 0.1 % TFA to A1 to A12wells in the 96-well plate and concatenate fractions fromA1–H1, A2–H2. . .A12–H12 to obtain a total of 12 fractions.Dry the samples using speed vac fitted with refrigeratedvapor trap.

6. Reconstitute the fractions with 100 μl of 5 % DHB solution.Ensure the peptides are completely dissolved (see Note 10).

7. Suspend TiO2 beads (50 mg) in 100 μl of 5 % DHB solutionand incubate for 15 min on the rotator at room temp (seeNote 11).

8. Centrifuge briefly and discard supernatant.

9. Add 2 μl of beads to each tube (each fraction may have~500 μg–1 mg of peptides) (TiO2: peptide ratio of 1:1) andincubate the peptide–DHB–TiO2 mix on a rotator for 15 minat room temperature (see Note 12).

370 H.C. Harsha et al.

Page 13: [Methods in Molecular Biology] Mass Spectrometry Data Analysis in Proteomics Volume 1007 || Proteomic Strategies to Characterize Signaling Pathways

10. Centrifuge the tubes at 1,500 � g for 1 min.

11. Transfer the entire content to StageTip with C8 plug.

12. Elute the unbound peptides into an Eppendorf tube.

13. Wash the peptide–TiO2 beads with 100 μl of wash solution 1.

14. Repeat step 12.

15. Elute phosphopeptides from TiO2 beads in 30 μl of 2 %NH4OH solution into collection tubes containing 20 μl of0.1 % TFA.

16. Repeat the elution step twice.

17. Dry the samples in speedvac without heating.

18. Resuspend dried peptides in 0.1 % TFA and desalt them usingC18 StageTip (or ZipTip).

4 Notes

1. The cells under study can also be reverse labeled to carry out abiological replicate experiment.

2. Extended period of lyophilization facilitates removal of tracesof TFA as low pH affects immunoaffinity purification.

3. While adjusting pH of the peptide digest prior to immunoaffi-nity purification, Tris–base must be added in small aliquots of5 μl until the desired pH of 7–8 is reached.

4. Anti-phosphotyrosine antibodies pY100 and 4G10 have over-lapping and slightly varied specificities. Using a mixture of thesetwo antibodies yields larger number of tyrosine phosphopep-tides as compared to using only one of them.

5. Antibody conjugated beads should be mixed gently to create ahomogenous suspension. Do not vortex or agitate the beads.

6. Do not try to aspirate all of the liquid as this may result in loss ofbeads.

7. If beads are accidentally loaded onto the StageTip, the anti-body-conjugated to the beads also bind to C18. Using a higherpercentage of organic solvent for elution may result in theelution of these antibodies which will interfere with MS analy-sis. With 50 % ACN, the proteins do not elute.

8. Dissolve the lyophilized peptide digests with bRPLC solvent A.The solvent should be added in 1 ml aliquot each time. Soni-cate the tubes in water bath sonicator for 5 min to ensurecomplete dissolution of peptides.

9. 50 μl of 1 % Formic acid should be added to 96 wells prior tofraction collection to neutralize peptides.

Proteomic Strategies to Characterize Signaling Pathways 371

Page 14: [Methods in Molecular Biology] Mass Spectrometry Data Analysis in Proteomics Volume 1007 || Proteomic Strategies to Characterize Signaling Pathways

10. To ensure complete dissolution of the peptides, the tubes canbe kept on thermomixer set at 100�g for 20 min at 20 �Cto ensure complete dissolution.

11. TiO2 bead suspension in 5 % dihydrobenzoic acid turnsbrownish.

12. Alternatively, the tubes can be kept on thermomixer set at100�g for 15–30 min at 20 �C.

References

1. Wang JY (1988) Antibodies for phosphotyro-sine: analytical and preparative tool for tyrosyl-phosphorylated proteins. Anal Biochem172:1–7

2. Frackelton AR Jr, Posner M, Kannan B,Mermelstein F (1991) Generation of monoclo-nal antibodies against phosphotyrosineand their use for affinity purification ofphosphotyrosine-containing proteins. Meth-ods Enzymol 201:79–92

3. Kamps MP (1991) Generation and use of anti-phosphotyrosine antibodies for immunoblot-ting. Methods Enzymol 201:101–110

4. Gronborg M, Kristiansen TZ, Stensballe A,Andersen JS, Ohara O, Mann M, Jensen ON,Pandey A (2002) A mass spectrometry-basedproteomic approach for identification ofserine/threonine-phosphorylated proteins byenrichment with phospho-specific antibodies:identification of a novel protein, Frigg, as aprotein kinase A substrate. Mol Cell Proteo-mics 1:517–527

5. Pandey A, Podtelejnikov AV, Blagoev B, Bus-telo XR, Mann M, Lodish HF (2000) Analysisof receptor signaling pathways by mass spec-trometry: identification of vav-2 as a substrateof the epidermal and platelet-derived growthfactor receptors. Proc Natl Acad Sci U S A97:179–184

6. Pandey A, Fernandez MM, Steen H, BlagoevB, Nielsen MM, Roche S, MannM, Lodish HF(2000) Identification of a novel immunorecep-tor tyrosine-based activation motif-containingmolecule, STAM2, by mass spectrometry andits involvement in growth factor and cytokinereceptor signaling pathways. J Biol Chem275:38633–38639

7. Salomon AR, Ficarro SB, Brill LM, Brinker A,Phung QT, Ericson C, Sauer K, Brock A, HornDM, Schultz PG, Peters EC (2003) Profilingof tyrosine phosphorylation pathways inhuman cells using mass spectrometry. ProcNatl Acad Sci U S A 100:443–448

8. Kosako H, Nagano K (2011) Quantitativephosphoproteomics strategies for understand-ing protein kinase-mediated signal transduc-tion pathways. Expert Rev Proteomics 8:81–94

9. Stasyk T, Huber LA (2012) Mapping in vivosignal transduction defects by phosphoproteo-mics. Trends Mol Med 18:43–51

10. Tinti M, Nardozza AP, Ferrari E, Sacco F, Cor-allino S, Castagnoli L, Cesareni G (2012) The4G10, pY20 and p-TYR-100 antibody specific-ity: profiling by peptide microarrays. N Bio-technol 29:571–577

11. Ficarro SB, McCleland ML, Stukenberg PT,Burke DJ, Ross MM, Shabanowitz J, HuntDF, White FM (2002) Phosphoproteome anal-ysis by mass spectrometry and its application toSaccharomyces cerevisiae. Nat Biotechnol20:301–305

12. Muszynska G, Dobrowolska G, Medin A,Ekman P, Porath JO (1992) Model studies oniron(III) ion affinity chromatography. II. Inter-action of immobilized iron(III) ions with phos-phorylated amino acids, peptides and proteins.J Chromatogr 604:19–28

13. Posewitz MC, Tempst P (1999) Immobilizedgallium(III) affinity chromatography of phos-phopeptides. Anal Chem 71:2883–2892

14. Cao P, Stults JT (1999) Phosphopeptide anal-ysis by on-line immobilized metal-ion affinitychromatography-capillary electrophoresis-electrospray ionization mass spectrometry. JChromatogr A 853:225–235

15. Beausoleil SA, Jedrychowski M, Schwartz D,Elias JE, Villen J, Li J, Cohn MA, Cantley LC,Gygi SP (2004) Large-scale characterization ofHeLa cell nuclear phosphoproteins. Proc NatlAcad Sci U S A 101:12130–12135

16. Han G, Ye M, Zhou H, Jiang X, Feng S, JiangX, Tian R, Wan D, Zou H, Gu J (2008) Large-scale phosphoproteome analysis of human livertissue by enrichment and fractionation of phos-phopeptides with strong anion exchange chro-matography. Proteomics 8:1346–1361

372 H.C. Harsha et al.

Page 15: [Methods in Molecular Biology] Mass Spectrometry Data Analysis in Proteomics Volume 1007 || Proteomic Strategies to Characterize Signaling Pathways

17. McNulty DE, Annan RS (2008) Hydrophilicinteraction chromatography reduces the com-plexity of the phosphoproteome and improvesglobal phosphopeptide isolation and detection.Mol Cell Proteomics 7:971–980

18. Alpert AJ (2008) Electrostatic repulsionhydrophilic interaction chromatography forisocratic separation of charged solutes andselective isolation of phosphopeptides. AnalChem 80:62–76

19. Andersson L, Porath J (1986) Isolation ofphosphoproteins by immobilized metal (Fe3+)affinity chromatography. Anal Biochem154:250–254

20. Feng S, Ye M, Zhou H, Jiang X, Jiang X, ZouH, Gong B (2007) Immobilized zirconium ionaffinity chromatography for specific enrich-ment of phosphopeptides in phosphoproteomeanalysis. Mol Cell Proteomics 6:1656–1665

21. Gruhler A, Olsen JV, Mohammed S, Morten-sen P, Faergeman NJ, Mann M, Jensen ON(2005) Quantitative phosphoproteomicsapplied to the yeast pheromone signaling path-way. Mol Cell Proteomics 4:310–327

22. Zhou H, Low TY, Hennrich ML, van derToorn H, Schwend T, Zou H, Mohammed S,Heck AJ (2011) Enhancing the identificationof phosphopeptides from putative basophilickinase substrates using Ti (IV) based IMACenrichment. Mol Cell Proteomics 10(M110):006452

23. Villen J, Beausoleil SA, Gerber SA, Gygi SP(2007) Large-scale phosphorylation analysisof mouse liver. Proc Natl Acad Sci U S A104:1488–1493

24. Sano A, Nakamura H (2004) Titania as achemo-affinity support for the column-switching HPLC analysis of phosphopeptides:application to the characterization of phos-phorylation sites in proteins by combinationwith protease digestion and electrospray ioni-zation mass spectrometry. Anal Sci20:861–864

25. LarsenMR, Thingholm TE, Jensen ON, Roep-storff P, Jorgensen TJ (2005) Highly selectiveenrichment of phosphorylated peptides frompeptide mixtures using titanium dioxide micro-columns. Mol Cell Proteomics 4:873–886

26. Molina H, Horn DM, Tang N, Mathivanan S,Pandey A (2007) Global proteomic profiling ofphosphopeptides using electron transfer disso-ciation tandem mass spectrometry. Proc NatlAcad Sci U S A 104:2199–2204

27. Wang Z, Udeshi ND, Slawson C, ComptonPD, Sakabe K, Cheung WD, Shabanowitz J,Hunt DF, Hart GW (2010) Extensive crosstalk

between O-GlcNAcylation and phosphoryla-tion regulates cytokinesis. Sci Signal 3:ra2

28. Blagoev B, Ong SE, Kratchmarova I, Mann M(2004) Temporal analysis of phosphotyrosine-dependent signaling networks by quantitativeproteomics. Nat Biotechnol 22:1139–1145

29. Zhang Y, Wolf-Yadlin A, Ross PL, Pappin DJ,Rush J, Lauffenburger DA, White FM (2005)Time-resolved mass spectrometry of tyrosinephosphorylation sites in the epidermal growthfactor receptor signalingnetwork reveals dynamicmodules. Mol Cell Proteomics 4:1240–1250

30. Olsen JV, Blagoev B, Gnad F, Macek B, KumarC, Mortensen P, Mann M (2006) Global,in vivo, and site-specific phosphorylationdynamics in signaling networks. Cell127:635–648

31. Qiao Y,Molina H, Pandey A, Zhang J, Cole PA(2006) Chemical rescue of a mutant enzyme inliving cells. Science 311:1293–1297

32. Luo W, Slebos RJ, Hill S, Li M, Brabek J,Amanchy R, Chaerkady R, Pandey A,Ham AJ, Hanks SK (2008) Global impact ofoncogenic Src on a phosphotyrosine proteome.J Proteome Res 7:3447–3460

33. Amanchy R, Zhong J, Molina H, Chaerkady R,Iwahori A, Kalume DE, Gronborg M, Joore J,Cope L, Pandey A (2008) Identification of c-Srctyrosine kinase substrates usingmass spectrometryand peptide microarrays. J Proteome Res7:3900–3910

34. Amanchy R, Zhong J, Hong R, Kim JH, GucekM, Cole RN, Molina H, Pandey A (2009)Identification of c-Src tyrosine kinase sub-strates in platelet-derived growth factor recep-tor signaling. Mol Oncol 3:439–450

35. Martinez-Ferrando I, Chaerkady R, Zhong J,Molina H, Kishore H, Herbst-Robinson K,Dancy BM, Katju V, Bose R, Zhang J, PandeyA, Cole PA (2012) Identification of targets ofc-Src tyrosine kinase by chemical complemen-tation and phosphoproteomics. Mol Cell Pro-teomics 11:355–369

36. Bose R, Molina H, Patterson AS, Bitok JK,Periaswamy B, Bader JS, Pandey A, Cole PA(2006) Phosphoproteomic analysis of Her2/neu signaling and inhibition. Proc Natl AcadSci U S A 103:9773–9778

37. Mukherji M, Brill LM, Ficarro SB, HamptonGM, Schultz PG (2006) A phosphoproteomicanalysis of the ErbB2 receptor tyrosine kinasesignaling pathways. Biochemistry45:15529–15540

38. Kruger M, Kratchmarova I, Blagoev B, TsengYH, Kahn CR, Mann M (2008) Dissection ofthe insulin signaling pathway via quantitative

Proteomic Strategies to Characterize Signaling Pathways 373

Page 16: [Methods in Molecular Biology] Mass Spectrometry Data Analysis in Proteomics Volume 1007 || Proteomic Strategies to Characterize Signaling Pathways

phosphoproteomics. Proc Natl Acad Sci U S A105:2451–2456

39. Zhang G, Spellman DS, Skolnik EY, NeubertTA (2006) Quantitative phosphotyrosine pro-teomics of EphB2 signaling by stable isotopelabeling with amino acids in cell culture(SILAC). J Proteome Res 5:581–588

40. Zhang G, Fenyo D, Neubert TA (2008)Screening for EphB signaling effectors usingSILAC with a linear ion trap-orbitrap massspectrometer. J Proteome Res 7:4715–4726

41. Hinsby AM, Olsen JV, Mann M (2004) Tyro-sine phosphoproteomics of fibroblast growthfactor signaling: a role for insulin receptorsubstrate-4. J Biol Chem 279:46438–46447

42. Cunningham DL, Sweet SM, Cooper HJ,Heath JK (2010) Differential phosphoproteo-mics of fibroblast growth factor signaling: iden-tification of Src family kinase-mediatedphosphorylation events. J Proteome Res9:2317–2328

43. Ding VM, Boersema PJ, Foong LY, PreisingerC, Koh G, Natarajan S, Lee DY, Boekhorst J,Snel B, Lemeer S, Heck AJ, Choo A (2011)Tyrosine phosphorylation profiling in FGF-2-stimulated human embryonic stem cells. PLoSOne 6:e17538

44. Zoumaro-Djayoon AD, Ding V, Foong LY,Choo A, Heck AJ, Munoz J (2011) Investigat-ing the role of FGF-2 in stem cell maintenanceby global phosphoproteomics profiling. Prote-omics 11:3962–3971

45. Osinalde N, Moss H, Arrizabalaga O, Omaet-xebarria MJ, Blagoev B, Zubiaga AM, Full-aondo A, Arizmendi JM, Kratchmarova I(2011) Interleukin-2 signaling pathway analy-sis by quantitative phosphoproteomics. J Pro-teomics 75:177–191

46. Ali NA, Molloy MP (2011) Quantitative phos-phoproteomics of transforming growth factor-beta signaling in colon cancer cells. Proteomics11:3390–3401

47. Zhong J, KimMS, Chaerkady R, Wu X, HuangTC, Getnet D, Mitchell CJ, Palapetta SM,Sharma J, O’Meally RN, Cole RN, Yoda A,Moritz A, Loriaux MM, Rush J, WeinstockDM, Tyner JW, Pandey A (2012) TSLP Signal-ing Network Revealed by SILAC-Based Phos-phoproteomics. Mol Cell Proteomics 11(M112):017764

48. Ong SE, Blagoev B, Kratchmarova I, Kristen-sen DB, Steen H, Pandey A, Mann M (2002)Stable isotope labeling by amino acids in cellculture, SILAC, as a simple and accurateapproach to expression proteomics. Mol CellProteomics 1:376–386

49. Ross PL, Huang YN, Marchese JN, WilliamsonB, Parker K, Hattan S, Khainovski N, Pillai S,Dey S, Daniels S, Purkayastha S, Juhasz P, Mar-tin S, Bartlet-Jones M, He F, Jacobson A, Pap-pin DJ (2004) Multiplexed proteinquantitation in Saccharomyces cerevisiae usingamine-reactive isobaric tagging reagents. MolCell Proteomics 3:1154–1169

50. Thompson A, Schafer J, Kuhn K, Kienle S,Schwarz J, Schmidt G, Neumann T, JohnstoneR, Mohammed AK, Hamon C (2003) Tandemmass tags: a novel quantification strategy forcomparative analysis of complex protein mix-tures by MS/MS. Anal Chem 75:1895–1904

51. Blagoev B, Kratchmarova I, Ong SE, NielsenM, Foster LJ, Mann M (2003) A proteomicsstrategy to elucidate functional protein-proteininteractions applied to EGF signaling. Nat Bio-technol 21:315–318

52. Wu SL, Kim J, Bandle RW, Liotta L, PetricoinE, Karger BL (2006) Dynamic profiling of thepost-translational modifications and interac-tion partners of epidermal growth factor recep-tor signaling after stimulation by epidermalgrowth factor using extended range proteomicanalysis (ERPA). Mol Cell Proteomics5:1610–1627

53. Amanchy R, Periaswamy B, Mathivanan S,Reddy R, Tattikota SG, Pandey A (2007) Acurated compendium of phosphorylationmotifs. Nat Biotechnol 25:285–286

54. Schwartz D, Chou MF, Church GM (2009)Predicting protein post-translational modifica-tions using meta-analysis of proteome scaledata sets. Mol Cell Proteomics 8:365–379

55. Diella F, Gould CM, Chica C, Via A, GibsonTJ (2008) Phospho.ELM: a database of phos-phorylation sites—update 2008. Nucleic AcidsRes 36:D240–D244

56. Hornbeck PV, Kornhauser JM, Tkachev S,Zhang B, Skrzypek E, Murray B, Latham V,Sullivan M (2012) PhosphoSitePlus: a compre-hensive resource for investigating the structureand function of experimentally determinedpost-translational modifications in man andmouse. Nucleic Acids Res 40:D261–D270

57. Keshava Prasad TS, Goel R, Kandasamy K,Keerthikumar S, Kumar S, Mathivanan S, Teliki-cherlaD,RajuR, ShafreenB,Venugopal A, Balak-rishnanL,MarimuthuA, Banerjee S, SomanathanDS,SebastianA,Rani S,Ray S,HarrysKishoreCJ,Kanth S, Ahmed M, Kashyap MK, Mohmood R,Ramachandra YL, Krishna V, Rahiman BA,Mohan S, Ranganathan P, Ramabadran S, Chaer-kady R, Pandey A (2009) Human Protein Refer-ence Database—2009 update. Nucleic Acids Res37:D767–D772

374 H.C. Harsha et al.

Page 17: [Methods in Molecular Biology] Mass Spectrometry Data Analysis in Proteomics Volume 1007 || Proteomic Strategies to Characterize Signaling Pathways

58. Obenauer JC, Cantley LC, Yaffe MB (2003)Scansite 2.0: proteome-wide prediction of cellsignaling interactions using short sequencemotifs. Nucleic Acids Res 31:3635–3641

59. Miller ML, Jensen LJ, Diella F, Jorgensen C,Tinti M, Li L, Hsiung M, Parker SA, BordeauxJ, Sicheritz-Ponten T, Olhovsky M, PasculescuA, Alexander J, Knapp S, Blom N, Bork P, Li S,Cesareni G, Pawson T, Turk BE, Yaffe MB,Brunak S, Linding R (2008) Linear motifatlas for phosphorylation-dependent signaling.Sci Signal 1(35):ra2

60. Blom N, Gammeltoft S, Brunak S (1999)Sequence and structure-based prediction ofeukaryotic protein phosphorylation sites. JMol Biol 294:1351–1362

61. Huang HD, Lee TY, Tzeng SW, Horng JT(2005) KinasePhos: a web tool for identifyingprotein kinase-specific phosphorylation sites.Nucleic Acids Res 33:W226–W229

62. Hanahan D, Weinberg RA (2000) The hall-marks of cancer. Cell 100:57–70

63. Druker BJ, Tamura S, Buchdunger E, Ohno S,Segal GM, Fanning S, Zimmermann J, LydonNB (1996) Effects of a selective inhibitor of theAbl tyrosine kinase on the growth of Bcr-Ablpositive cells. Nat Med 2:561–566

64. Lynch TJ, Bell DW, Sordella R, Gurubhagava-tula S, Okimoto RA, Brannigan BW,Harris PL,Haserlat SM, Supko JG, Haluska FG, LouisDN, Christiani DC, Settleman J, Haber DA(2004) Activating mutations in the epidermalgrowth factor receptor underlying responsive-ness of non-small-cell lung cancer to gefitinib.N Engl J Med 350:2129–2139

65. Slamon DJ, Leyland-Jones B, Shak S, Fuchs H,Paton V, Bajamonde A, Fleming T, EiermannW, Wolter J, Pegram M, Baselga J, Norton L(2001) Use of chemotherapy plus a monoclo-nal antibody against HER2 for metastaticbreast cancer that overexpresses HER2. NEngl J Med 344:783–792

66. Harsha HC, Jimeno A, Molina H, Mihalas AB,Goggins MG, Hruban RH, Schulick RD,Kamath U, Maitra A, Hidalgo M, Pandey A(2008) Activated epidermal growth factorreceptor as a novel target in pancreatic cancertherapy. J Proteome Res 7:4651–4658

67. Walters DK, Goss VL, Stoffregen EP, Gu TL,Lee K, Nardone J, McGreevey L, HeinrichMC, Deininger MW, Polakiewicz R, DrukerBJ (2006) Phosphoproteomic analysis ofAML cell lines identifies leukemic oncogenes.Leuk Res 30:1097–1104

68. Bai Y, Li J, Fang B, Edwards A, Zhang G, BuiM, Eschrich S, Altiok S, Koomen J, Haura EB

(2012) Phosphoproteomics identifies drivertyrosine kinases in sarcoma cell lines andtumors. Cancer Res 72:2501–2511

69. JunHJ, JohnsonH, Bronson RT, de Feraudy S,White F, Charest A (2012) The oncogenic lungcancer fusion kinase CD74-ROS activates anovel invasiveness pathway through E-Syt1phosphorylation. Cancer Res 72(15):3764–3774

70. Suwaki N, Vanhecke E, Atkins KM, Graf M,Swabey K, Huang P, Schraml P, Moch H, Cas-sidy AM, Brewer D, Al-Lazikani B, WorkmanP, De-Bono J, Kaye SB, Larkin J, Gore ME,Sawyers CL, Nelson P, Beer TM, Geng H, GaoL, Qian DZ, Alumkal JJ, Thomas G, ThomasGV (2011) A HIF-regulated VHL-PTP1B-Srcsignaling axis identifies a therapeutic target inrenal cell carcinoma. Sci Transl Med 3(85):ra47

71. Chumbalkar V, Latha K, Hwang Y,Maywald R,Hawley L, Sawaya R, Diao L, Baggerly K,Cavenee WK, Furnari FB, Bogler O (2011)Analysis of phosphotyrosine signaling inglioblastoma identifies STAT5 as a novel down-stream target of DeltaEGFR. J Proteome Res10:1343–1352

72. Old WM, Shabb JB, Houel S, Wang H, CoutsKL, Yen CY, Litman ES, Croy CH, Meyer-Arendt K, Miranda JG, Brown RA, Witze ES,Schweppe RE, Resing KA, Ahn NG (2009)Functional proteomics identifies targets ofphosphorylation by B-Raf signaling in mela-noma. Mol Cell 34:115–131

73. Leroy C, Fialin C, Sirvent A, Simon V, UrbachS, Poncet J, Robert B, Jouin P, Roche S (2009)Quantitative phosphoproteomics reveals a clus-ter of tyrosine kinases that mediates SRC inva-sive activity in advanced colon carcinoma cells.Cancer Res 69:2279–2286

74. Nagano K, Shinkawa T, Mutoh H, Kondoh O,Morimoto S, Inomata N, Ashihara M, Ishii N,Aoki Y, Haramura M (2009) Phosphoproteo-mic analysis of distinct tumor cell lines inresponse to nocodazole treatment. Proteomics9:2861–2874

75. Huang PH, Mukasa A, Bonavia R, Flynn RA,Brewer ZE, Cavenee WK, Furnari FB, WhiteFM (2007) Quantitative analysis of EGFRvIIIcellular signaling networks reveals a combina-torial therapeutic strategy for glioblastoma.Proc Natl Acad Sci U S A 104:12867–12872

76. Rexer BN, Ham AJ, Rinehart C, Hill S, Granja-Ingram NM, Gonzalez-Angulo AM, Mills GB,Dave B, Chang JC, Liebler DC, Arteaga CL(2011) Phosphoproteomic mass spectrometryprofiling links Src family kinases to escape fromHER2 tyrosine kinase inhibition. Oncogene30:4163–4174

Proteomic Strategies to Characterize Signaling Pathways 375

Page 18: [Methods in Molecular Biology] Mass Spectrometry Data Analysis in Proteomics Volume 1007 || Proteomic Strategies to Characterize Signaling Pathways

77. Andersen JN, Sathyanarayanan S, Di Bacco A,Chi A, Zhang T, Chen AH, Dolinski B, KrausM, Roberts B, Arthur W, Klinghoffer RA, Gar-gano D, Li L, Feldman I, Lynch B, Rush J,Hendrickson RC, Blume-Jensen P, PaweletzCP (2010) Pathway-based identification ofbiomarkers for targeted therapeutics: persona-lized oncology with PI3K pathway inhibitors.Sci Transl Med 2(43):ra55

78. De la Mota-Peynado A, Chernoff J, Beeser A(2011) Identification of the atypical MAPKErk3 as a novel substrate for p21-activatedkinase (Pak) activity. J Biol Chem286:13603–13611

79. Zach S, Felk S, Gillardon F (2010) Signal trans-duction protein array analysis links LRRK2 toSte20 kinases and PKC zeta that modulateneuronal plasticity. PLoS One 5:e13191

80. Kaushansky A, Gordus A, Chang B, Rush J,MacBeath G (2008) A quantitative study ofthe recruitment potential of all intracellulartyrosine residues on EGFR, FGFR1 andIGF1R. Mol Biosyst 4:643–653

81. Wilson B, Liotta LA, Petricoin E 3rd (2010)Monitoring proteins and protein networksusing reverse phase protein arrays. Dis Markers28:225–232

82. Chan SM, Ermann J, Su L, Fathman CG, UtzPJ (2004) Protein microarrays for multiplexanalysis of signal transduction pathways. NatMed 10:1390–1396

83. Paweletz CP, Charboneau L, Bichsel VE,Simone NL, Chen T, Gillespie JW,Emmert-Buck MR, Roth MJ, Petricoin IE,Liotta LA (2001) Reverse phase proteinmicroarrays which capture disease progres-sion show activation of pro-survival pathwaysat the cancer invasion front. Oncogene20:1981–1989

84. Wulfkuhle JD, Aquino JA, Calvert VS, FishmanDA, Coukos G, Liotta LA, Petricoin EF 3rd(2003) Signal pathway profiling of ovarian can-cer from human tissue specimens using reverse-phase protein microarrays. Proteomics3:2085–2090

85. Chan SM, Weng AP, Tibshirani R, Aster JC,Utz PJ (2007) Notch signals positively regulateactivity of the mTOR pathway in T-cell acutelymphoblastic leukemia. Blood 110:278–286

86. van Agthoven T, Godinho MF, Wulfkuhle JD,Petricoin EF 3rd, Dorssers LC (2012) Proteinpathway activation mapping reveals molecularnetworks associated with antiestrogen resis-tance in breast cancer cell lines. Int J Cancer131(9):1998–2007

87. Lavezzari G, Lackner MR (2012) Monitoringphosphoproteomic response to targeted kinaseinhibitors using reverse-phase protein microar-rays. Methods Mol Biol 795:203–215

88. Frederick MJ, VanMeter AJ, Gadhikar MA,Henderson YC, Yao H, Pickering CC, WilliamsMD, El-Naggar AK, Sandulache V, Tarco E,Myers JN, Clayman GL, Liotta LA, PetricoinEF 3rd, Calvert VS, Fodale V, Wang J, WeberRS (2011) Phosphoproteomic analysis of sig-naling pathways in head and neck squamouscell carcinoma patient samples. Am J Pathol178:548–571

89. Amanchy R, Kandasamy K,Mathivanan S, Peri-aswamy B, Reddy R, Yoon WH, Joore J, BeerMA, Cope L, Pandey A (2011) Identificationof novel phosphorylation motifs through anintegrative computational and experimentalanalysis of the human phosphoproteome.J Proteomics Bioinform 4:22–35

90. Harsha HC, Molina H, Pandey A (2008)Quantitative proteomics using stable isotopelabeling with amino acids in cell culture. NatProtoc 3:505–516

91. Bian Y, Ye M, Song C, Cheng K, Wang C, WeiX, Zhu J, Chen R, Wang F, Zou H (2012)Improve the coverage for the analysis of phos-phoproteome of HeLa cells by a tandem diges-tion approach. J Proteome Res 11:2828–2837

92. Beausoleil SA, Villen J, Gerber SA, Rush J,Gygi SP (2006) A probability-based approachfor high-throughput protein phosphorylationanalysis and site localization. Nat Biotechnol24:1285–1292

93. Cox J, Mann M (2008) MaxQuant enableshigh peptide identification rates, individualizedp.p.b.-range mass accuracies and proteome-wide protein quantification. Nat Biotechnol26:1367–1372

94. MacLean D, Burrell MA, Studholme DJ, JonesAM (2008) PhosCalc: a tool for evaluating thesites of peptide phosphorylation from massspectrometer data. BMC Res Notes 1:30

95. Taus T, Kocher T, Pichler P, Paschke C,Schmidt A, Henrich C, Mechtler K (2011)Universal and confident phosphorylation sitelocalization using phosphoRS. J ProteomeRes 10:5354–5362

96. Ogata H, Goto S, Sato K, Fujibuchi W, BonoH, Kanehisa M (1999) KEGG: Kyoto encyclo-pedia of genes and genomes. Nucleic Acids Res27:29–34

97. Kandasamy K, Mohan SS, Raju R, Keerthiku-mar S, Kumar GS, Venugopal AK, TelikicherlaD, Navarro JD, Mathivanan S, Pecquet C,

376 H.C. Harsha et al.

Page 19: [Methods in Molecular Biology] Mass Spectrometry Data Analysis in Proteomics Volume 1007 || Proteomic Strategies to Characterize Signaling Pathways

Gollapudi SK, Tattikota SG, Mohan S, Padhu-kasahasram H, Subbannayya Y, Goel R, JacobHK, Zhong J, Sekhar R, Nanjappa V, Balak-rishnan L, Subbaiah R, Ramachandra YL,Rahiman BA, Prasad TS, Lin JX, HoutmanJC, Desiderio S, Renauld JC, Constantinescu

SN, Ohara O, Hirano T, Kubo M, Singh S,Khatri P, Draghici S, Bader GD, Sander C,Leonard WJ, Pandey A (2010) NetPath: apublic resource of curated signal transductionpathways. Genome Biol 11:R3

Proteomic Strategies to Characterize Signaling Pathways 377


Recommended