Transcript
Page 1: Lectin Chromatography/Mass Spectrometry … Chromatography/Mass Spectrometry Discovery Workflow ... The lines were grown to 75−80% confluence in the appropriate ... (FCS) or phenol

Lectin Chromatography/Mass Spectrometry Discovery WorkflowIdentifies Putative Biomarkers of Aggressive Breast CancersPenelope M. Drake,† Birgit Schilling,‡ Richard K. Niles,† Akraporn Prakobphol,† Bensheng Li,‡

Kwanyoung Jung,§ Wonryeon Cho,∥ Miles Braten,† Halina D. Inerowicz,§ Katherine Williams,†

Matthew Albertolle,† Jason M. Held,‡ Demetris Iacovides,⊥ Dylan J. Sorensen,‡ Obi L. Griffith,⊥

Eric Johansen,† Anna M. Zawadzka,‡ Michael P. Cusack,‡ Simon Allen,† Matthew Gormley,†

Steven C. Hall,† H. Ewa Witkowska,† Joe W. Gray,⊥,¶ Fred Regnier,§ Bradford W. Gibson,*,‡,#and Susan J. Fisher*,†

†Department of Obstetrics, Gynecology and Reproductive Sciences, University of California San Francisco, 513 Parnassus Avenue,Box 0665, San Francisco, California 94143, United States

‡Buck Institute for Research on Aging, 8001 Redwood Boulevard, Novato, California 94945, United States§Department of Chemistry and Bindley Bioscience Center, Purdue University, 201 South University Street HANS B054,West Lafayette, Indiana 47907, United States

∥Bio-Nano Chemistry, Wonkwang University, 344-2 Shinyong-dong, Iksan, Jonbuk 570-749, Korea⊥Life Sciences Division, Lawrence Berkeley National Laboratory, Berkeley, California 94720, United States¶Department of Biomedical Engineering, Oregon Health and Science University, Portland, Oregon 97238, United States#Department, of Pharmaceutical Chemistry, Box 0446, University of California, San Francisco, California 94143, United States

*S Supporting Information

ABSTRACT: We used a lectin chromatography/MS-based approach to screen conditionedmedium from a panel of luminal (less aggressive) and triple negative (more aggressive)breast cancer cell lines (n = 5/subtype). The samples were fractionated using the lectinsAleuria aurantia (AAL) and Sambucus nigra agglutinin (SNA), which recognize fucose andsialic acid, respectively. The bound fractions were enzymatically N-deglycosylated andanalyzed by LC−MS/MS. In total, we identified 533 glycoproteins, ∼90% of which werecomponents of the cell surface or extracellular matrix. We observed 1011 glycosites, 100 ofwhich were solely detected in ≥3 triple negative lines. Statistical analyses suggested that anumber of these glycosites were triple negative-specific and thus potential biomarkers forthis tumor subtype. An analysis of RNaseq data revealed that approximately half of themRNAs encoding the protein scaffolds that carried potential biomarker glycosites were up-regulated in triple negative vs luminal cell lines, and that a number of genes encodingfucosyl- or sialyltransferases were differentially expressed between the two subtypes,suggesting that alterations in glycosylation may also drive candidate identification. Notably, the glycoproteins from which theseputative biomarker candidates were derived are involved in cancer-related processes. Thus, they may represent novel therapeutictargets for this aggressive tumor subtype.KEYWORDS: lectins, lectin chromatography, breast cancer, triple-negative, SNA, AAL, sialic acid, fucose

■ INTRODUCTIONThe intense interest in biomarker discovery is a reflection ofthe clinical need for tests with a high degree of sensitivity andspecificity for diagnosing diseases, predicting their courses, aswell as monitoring responses to therapy and disease recurrence.Technological breakthroughs in separation strategies andmass spectrometry (MS) have enabled rapid identification andquantification of large numbers of proteins in biological samples.1

Nonetheless, their complexity requires extensive fractionationto access low abundance proteins, such as those released fromnascent tumors. Alternatively, and technically less challenging,is the design of capture approaches that exploit disease biology

for the purpose of biomarker identification.2 For many reasons,glycosylation is an attractive target. First, the biology allows forthe rational design of discovery efforts. For example, changes inthe glycosylation machinery can be identified from microarraydata and translated in structural terms, providing a compellingrationale for designing lectin-based strategies to enrich glyco-peptides carrying disease-related carbohydrate motifs. Second,one protein can carry many copies of an altered glycan, whichmay also be added to other scaffolds. Thus, there is an important

Received: December 8, 2011Published: February 6, 2012

Article

pubs.acs.org/jpr

© 2012 American Chemical Society 2508 dx.doi.org/10.1021/pr201206w | J. Proteome Res. 2012, 11, 2508−2520

Page 2: Lectin Chromatography/Mass Spectrometry … Chromatography/Mass Spectrometry Discovery Workflow ... The lines were grown to 75−80% confluence in the appropriate ... (FCS) or phenol

amplification effect, which could enable the detection of manyfewer abnormal cells than would otherwise be possible. Finally,glycosylation acts to shield the peptide backbone from pro-teolytic degradation.3 Thus, in theory, glycan-based biomarkersare likely to be more stable in a variety of disease settings thanunmodified proteins, which are often more labile.Glycosylation is altered in a number of pathologies, but its

relationship to cancer is particularly well-defined at phenotypicand, to a lesser degree, functional levels. For example, many ofthe most widely used clinical tests detect glycoproteins andcarbohydrate structures. These include carcinoembryonic anti-gen (CEA), commonly used as a marker of colorectal cancer;CA-125, frequently employed to diagnose ovarian cancer; CA19-9, the most commonly used biomarker for diagnosingpancreatic cancer; CA 15-3, used to monitor the metastasis ofbreast cancer;4 and prostate-specific antigen (PSA).5−8 Inaddition, glycan-specific antibodies and lectins are used for thecytological and histological evaluation of glycosylation for thepurpose of guiding diagnoses and enabling more accurateprognoses, for example, anti-Lewis (Le)x antibodies for bladdercancer, and the lectins Helix pomatia agglutinin (HPA) andUlex europaeus I agglutinin (UEA 1) for breast cancer.1 This isdue to the fact that increases in fucosylation and sialylation ofN-linked structures and truncation of O-linked oligosaccharidesoccur in many tumor types. The expression of Le antigens, suchas sialyl Lex, can also be indicative of disease progression, asthese structures play important roles in promoting metastasisby virtue of their well-known ability to mediate cell traffickingand extravasation.9,10

Breast cancer is now recognized to be a collection of distinctneoplastic diseases with different molecular and clinical attri-butes. Breast tumors can be stratified into five intrinsic subtypesand a “normal-like” group according to features such as mRNAexpression.11 Interestingly, these molecularly defined cohorts,which include luminal, basal-like, and claudin-low, are also predic-tive of clinical outcomes such as disease severity and treatmentresponse.12−14 Specifically, luminal tumors tend to be lessaggressive with better survival rates, while basal-like andclaudin-low lesions have generally worse prognoses.15 Addi-tionally, the expression of a therapeutic target such as theestrogen receptor (ER) or human epidermal growth factorreceptor 2 (HER2/ErbB2) determines tumor susceptibility todrugs that interact with these molecules.16,17 Triple negativebreast cancers (TNBC) express neither ER nor the progesteronereceptor (PR) and moderate levels of HER2. This clinicallyimportant, heterogeneous category includes most basal-like andclaudin-low tumors.18,19 TNBCs have poor survival rates andlack specific therapeutic targets, limiting treatment options andmaking early detection a priority.We hypothesized that biomarkers specific for these tumors

could be identified by a comparative analysis of the repertoireof secreted or shed glycoproteins in a panel of breast cancer celllines that have been extensively characterized at genomic andtranscriptional levels.20−22 Based on gene expression, the linescan be clustered into subsets that mirror the molecular char-acteristics of primary breast tumors. Thus, these panels areuseful tools for studying subtype-specific behavior, such as drugresponses and alternative splicing.20,23 Here, we used a subsetof cells from this collection for biomarker discovery. Specifi-cally, we analyzed conditioned medium (CM) from 5 luminaland 5 triple negative cell lines. The samples were distributed tothree laboratories: University of California San Francisco (UCSF),the Buck Institute for Research on Aging, and Purdue University.

Each group analyzed the samples using our recently publishedmethod for lectin affinity chromatographic enrichment and LC−MS/MS analyses.24 Overall, we identified 533 glycoproteins,including 1011 N-linked glycosylation sites (glycosites). Of these,100 were solely detected in ≥3 triple negative lines. Interestingly,many in the latter category were from glycoproteins that are up-regulated in the claudin-low subtype,21 involved in cancerprogression (e.g., epithelial to mesenchymal transition) and/ormetastasis.25

■ MATERIALS AND METHODSCell Culture and Production of Conditioned Media

All cells were cultured as described in Neve et al.21 To generatethe CM, we cultured 10 breast cancer cell lines (Table 1) that

were derived from 5 luminal (SKBR3, SUM52 PE,MDAMB175, UACC 812, and MDAMA361) and 5 triplenegative tumors (MDA468, BT549, HS578T, MDAMB231,and HCC38). CM was prepared and trypsin digested at Site M.The lines were grown to 75−80% confluence in the appropriateculture medium.21 Then they were washed with fresh mediumwithout fetal calf serum (FCS) or phenol red and incubated for10 min at 37 °C. This process was repeated twice before thecells were incubated in fresh medium (without FCS and phenolred) for 18−20 h. At the end of the culture period, the cellsretained their original morphologies with no evidence ofapoptosis. The CM was harvested and centrifuged at 2000× gfor 10 min. The supernatant was concentrated using Milliporecentrifugal filter units (MWCO 3K) and dialyzed againstphosphate buffered saline (PBS).Lectin Blotting and Staining

Biotinylated and fluoresceinated lectins were purchased fromVector Laboratories. Blotting: Cell lysates were separated bySDS-PAGE (4−12% gels) and transferred to nitrocellulosemembranes. Unless otherwise indicated, the following bufferwas used for all steps, including blocking, washing, and reagentdilution/incubation: 0.25 M Tris-Cl, pH 8.0, 0.5 M NaCl,0.5% NP-40. Blots were incubated in buffer for 1 h to blocknonspecific binding, then exposed to a solution of ∼5 μg/mL ofbiotinylated lectin for 2 h. Blots were washed 3 × 5 min withcopious amounts of buffer. Then, membranes were reacted withABC reagent (Vector Laboratories) for 1 h and washed again asbefore. Finally, bound lectin was detected using 3,3-diamino-benzidine (DAB, Vector Laboratories) prepared in water according

Table 1. Luminal and Triple-Negative Breast Cancer CellLines

cell linea tumor subtype ERb PRc HER2d diagnosis

SKBR3 Luminal − − Yes AdenocarcinomaSUM52 PE Luminal + − No CarcinomaMDAMB175 Luminal + − No IDCe

UACC 812 Luminal + − Yes IDCMDAMA361 Luminal + − Yes AdenocarcinomaMDA468 Basal A − − No AdenocarcinomaBT549 Claudin-low − − No IDC, papillaryHS5787 Claudin-low − − No IDCMDAMB 231 Claudin-low − − No AdenocarcinomaHCC38 Claudin-low − − No Ductal carcinoma

aThis table is populated with information from Neve et al., 2006.21bEstrogen receptor expression. cProgesterone receptor expression.dHuman epidermal growth factor receptor 2 overexpression. eInvasiveductal carcinoma.

Journal of Proteome Research Article

dx.doi.org/10.1021/pr201206w | J. Proteome Res. 2012, 11, 2508−25202509

Page 3: Lectin Chromatography/Mass Spectrometry … Chromatography/Mass Spectrometry Discovery Workflow ... The lines were grown to 75−80% confluence in the appropriate ... (FCS) or phenol

to the manufacturer’s instructions. Staining: cell surface labelingof nonpermeabilized cells was performed as described,26 exceptthat fluoresceinated lectins, rather than antibodies, were used.Trypsin Digestion

First, protein concentrations of the CM samples were deter-mined by amino acid analysis. Then, CM samples were digestedand desalted using a published method that incorporatesdenaturation with 6 M urea.27 As previously described,24

samples were spiked with 25 and 50 pmol of trypsin-digestedcontrol glycopeptides from commercial yeast invertase andhuman lactoferrin (Sigma, St. Louis, MO), respectively.Peptides were stored at −80 °C prior to analyses.Preparation of Lectin Columns

The columns were prepared at Site M from a single batch oflectin-conjugated beads and distributed to all the laboratories.Briefly, Sambucus nigra agglutinin (SNA) and Aleuria aurantialectin (AAL) were purchased from Vector Laboratories(Burlingame, CA). Lectins (20 mg) were suspended at 5−10 mg/mL in PBS and conjugated to 330 mg of POROS-ALbeads (Applied Biosystems, Foster City, CA) as previouslydescribed.24 Unconjugated protein was removed by washing thebeads (5 × 5 mL of 1 M sodium chloride) before they werepacked into 3 individual 4.6 × 50 mm PEEK HPLC columns.Routine storage was in PBS with 0.02% sodium azide at 4 °Cfor up to 6 months. Columns were reused for up to 75 affinityseparations without degradation of the performance character-istics as assessed by glycopeptide enrichment and total numberof glycopeptides recovered from digested human plasma.Lectin Chromatography

Instrumentation. The HPLC systems employed werestandardized in terms of injection volume, transfer line lengths,dead volume minimization, and common UV elution profiles.Site M used a Paradigm MG4 HPLC system equipped with aCTC PAL robot configured as an autosampler and fractioncollector (Michrom Bioresources). At Site X, a Waters systemincluding 1525 Binary HPLC equipped with a 717 plus Auto-sampler and a Fraction Collector III was employed. Site S useda Shimadzu 20AD HPLC system equipped with a SIL-20ACautosampler; fractions were collected manually.Mobile Phases. Buffer A was 25 mM Tris buffer, pH 7.4,

50 mM sodium chloride, 10 mM calcium chloride, and 10 mMmagnesium chloride; Buffer B was 0.5 M acetic acid.Affinity Separation. Routinely, ∼100 μg of digested

protein was diluted into Buffer A, applied to the lectin column,and separated using the following 3 step gradient: (1) Sampleload: Buffer A for 9.0 min at 80 μL/min; (2) Sample elution:Buffer B for 4.8 min at 500 μL/min; and (3) Re-equilibration:Buffer A for 6.0 min at 3000 μL/min. The bound fraction,collected from 9.0 to 14.25 min, was desalted using Oasis HLBcartridges as described above. Eluted samples were neutralizedby the addition of 0.5 M ammonium bicarbonate and con-centrated to <100 μL by vacuum centrifugation. Further detailsare described in the accompanying SOP (SupplementaryDocument 1, Supporting Information).PNGase F Digestion

N-linked glycopeptides in the bound fractions were deglycosy-lated by treatment with PNGase F (Glycerol-free, New EnglandBiolabs; Ipswich, MA) as previously described.24 Followingdeglycosylation, samples were desalted and concentrated usingC18 ZipTips (Millipore; Billerica, MA) or MicroSpin Columns,5−200 μL (The Nest Group, Inc.; Southborough, MA).

ESI-QqTOF Mass Spectrometric Analyses (Sites M and X)The peptides were separated using an Eksigent nano-LC 2DHPLC system (Eksigent, Dublin, CA), which was directly con-nected to a quadrupole time-of-flight (QqTOF) QSTAR Elitemass spectrometer (AB Sciex, Foster City, CA). We injected33% (v/v) of the bound material per run. Briefly, peptides wereapplied to a guard column (C18 Acclaim PepMap100, 300 μmI.D. × 5 mm, 5 μm particle size, 100 Å pore size; Dionex,Sunnyvale, CA) and washed with the aqueous loading solvent(2% solvent B in A, flow rate: 20 μL/min) for 10 min prior toseparation on a C18 Acclaim PepMap100 column (75 μm I.D. ×15 cm, 3 μm particle size, 100 Å pore size; Dionex, Sunnyvale,CA). Bound material was eluted at a flow rate of 300 nL/minusing the following gradients: 2−40% solvent B in A (from 0 to60 min), 40−90% solvent B in A (from 60 to 75 min), and at 90%solvent B in A (from 75 to 85 min), with a total runtime of120 min (including column equilibration). Solvent A consisted of0.1% formic acid in 98% H2O/2% acetonitrile and solvent B was0.1% formic acid in 98% acetonitrile/2% H2O. Spectra werecalibrated using MS/MS fragment-ions of a Glu-Fibrinogen Bpeptide standard. Advanced information dependent acquisitionwas employed for MS/MS data collection using QSTAR Elite(Analyst QS 2.0) specific features, including “Smart Collision”(fragment intensity multiplier set to 2.0) and “Smart Exit”(maximum accumulation time of 2.5 s) to obtain MS/MS spectrafor the six most abundant precursor ions following each surveyscan. To increase overall sampling efficiencies, two replicate nano-HPLC-MS/MS analyses were performed per sample.ESI-LTQ-Orbitrap XL Mass Spectrometric Analyses (Site S)The peptide mixtures were separated as described above usingan Agilent nanoflow 1100 HPLC system (Agilent, Santa Clara,CA) connected to a hybrid linear ion trap Orbitrap massspectrometer (LTQ Orbitrap XL, Thermo Fisher Scientific).The electrospray ionization emitter tip (Pico-tip emitter, F360-75-15-N-5-C10.5) was purchased from New Objective(Woburn, MA). The mass spectrometer, which was calibratedwith a solution of caffeine, MRFA and Ultramark 1621 accord-ing to the manufacturer’s instructions, was operated in the data-dependent mode. Full MS scans from m/z 350 to 1600 with afull width at half-maximum resolution of 30000 were acquiredas profile data, followed by MS/MS scans of the six mostabundant ions in the linear trap. Singly charged ions wereexcluded. A dynamic mass exclusion time was applied for 120 swith a repeat count of 1 and a repeat duration time of 30 s. Inall scan modes, one micro scan was applied.Database SearchesMass spectrometric data from all laboratories were analyzed atSite M using two bioinformatics database search engines withintegrated peak picking, ProteinPilot (AB Sciex) version4.0.8085 (revision 148085) using the Paragon Algorithm4.0.0.0, 148083,28 and Mascot version 2.2.04 using MascotDaemon version 2.2.2 (both Matrix Science). For the latter, thefollowing (default) data import filter options were used:precursor charge state +2 to +4, reject spectrum if <7 peaks orif precursor is <400 or >10000 m/z, remove peaks withintensity <0.001% of the highest peak; centroid all MS/MSdata, percentage height 50, and merge distance 0.1 atomic massunits. Peak lists for the Orbitrap LC−MS/MS data sets weregenerated using Mascot Distiller 2.3.2.0 (Matrix Science) withthe supplied processing parameter file Orbitrap_low_res_MS2_4.opt. The Orbitrap peak lists were saved in MGF formatwith Distiller preferences set to save MS/MS peaks as MH+ for

Journal of Proteome Research Article

dx.doi.org/10.1021/pr201206w | J. Proteome Res. 2012, 11, 2508−25202510

Page 4: Lectin Chromatography/Mass Spectrometry … Chromatography/Mass Spectrometry Discovery Workflow ... The lines were grown to 75−80% confluence in the appropriate ... (FCS) or phenol

input into Mascot and ProteinPilot search engines. All data weresearched using a merged database of 20293 protein sequencesincluding the publicly available human Swiss-Prot UniProtrelease 2010_09 plus 7 other proteins, which includes all 20286reviewed (formerly Swiss-Prot) Human Uniprot Entries, as wellas PNGase F (Q9XBM8|Q9XBM8_FLAME, P21163|PNGF_ELIMR) and Yeast Invertase (P10594|INV1_YEAST,P00724|INV2_YEAST, P10595|INV3_YEAST, P10596|IN-V4_YEAST, P10597|INV5_YEAST). ProteinPilot searcheswere performed as previously described.24 A ProteinPilot pep-tide confidence cutoff value of 98.8 was chosen, correspondingto a local FDR of 5%. For Mascot searches, the following param-eters were used: trypsin enzyme specificity, carbamidomethyl(Cys) as a fixed modification, and the following variable modifi-cations: deamidation of asparagine and glutamine residues, oxi-dization of methionines, acetylation at the protein N-terminus,cyclization of N-terminal glutamines, and two missed trypticcleavages. For QSTAR Elite data, a mass tolerance of 100 ppmand 0.4 Da was set for the precursor and product ions,respectively; whereas values of 10 ppm and 0.8 Da were appliedto Orbitrap data. Peptide-spectral matches with expectationvalues <0.026 were accepted. FDR analysis was performedusing the Mascot automatic decoy search. In all cases, thepeptide false-positive identification rate was <3%.Glycopeptide Assignment

Deglycosylated peptides were identified as previously de-scribed,24 on the basis of several criteria including the motifNxS/T, x ≠ proline, in which Asn was converted to Asp(reported by the search engine as Asn deamidation), and thepresence of at least one fragment ion encompassing theglycosite. To ensure inclusion of glycosites containing Lys and/or Arg in the X position (e.g., NKT), which were likely to havebeen cleaved by trypsin, the amino-acid residue following thecarboxy-terminal cleavage site was also considered. Peptidescontaining the motif NGS or NGT were excluded due to thefact that asparagine residues in that sequence are prone tochemical deamidation during overnight trypsin digestion.29 Forall deglycosylated peptides the corresponding MS/MS spectrawere manually examined using an adaptation of previously pub-lished criteria to ensure correct assignment.24,30

Generation of a List of Candidate Triple Negative-SpecificGlycosites

The selection criteria for triple negative-specific glycosites weresubjected to a resampling, nonparametric statistical test inwhich no knowledge about the data’s distribution is necessary,for example, the “bootstrap” technique.31 The basic premise ofthis approach is to consider the null hypothesis that there isstatistically no difference between the luminal and triple nega-tive data sets, for example, that the two are random selectionsfrom the same population. To determine the expected FDRs,we applied 20000 random permutations to the form:Criterion n−m: A Glycosite Satisfies Criterion n−m if it isIdentified in ≤n Luminal Cell Lines and in ≥m TN Cell Lines

The results are shown in Supplementary Table 3 (SupportingInformation).Spectral Viewer: Skyline Spectral Library

An interactive Skyline spectral library file that contains all MS/MS spectra of deglycosylated peptides identified in this studybeen submitted as Supporting Information. Skyline is an opensource program32 available for free download at http://proteome.gs.washington.edu/software/skyline.

Exon Expression Array and RNaseq Experiments

Whole transcriptome shotgun sequencing (RNaseq) was com-pleted on nine of ten breast cancer cell lines (BT549, HCC38,HS578T, MDAMB231, MDAMB175VII, MDAMB361,SKBR3, SUM52PE and UACC812). Expression analysis wasperformed with the ALEXA-seq software package as previouslydescribed.33 On a per sample basis, an average of 58.7 million(76bp paired-end) reads passed quality control, and 37.6 millionmapped to the transcriptome, which resulted in coverage of 40xacross all known genes. Log2 transformed estimates of gene-level expression were extracted for fucosyl- and sialyltransferasegenes, and triple negative candidate biomarker targets thatemerged from the N-glycosite workflow. Corresponding valuesindicating whether expression of a transcript was detectedabove background were also extracted.A 2-sided Student’s t-test was used to compare log2 trans-

formed gene expression levels between the five luminal and thefour triple negative cell lines. This comparison generated rawp-values, which were then adjusted for multiple comparisonsusing the Benjamini-Hochberg method for controlling FDRs.34

The adjustment was achieved with the p.adjust (pvals,”fdr”)function in R version 2.12.1 (2010−12−16). Adjusted FDRp-values lower than 10% (0.1) were considered significant.

■ RESULTS AND DISCUSSIONWorkflow

These experiments utilized a lectin chromatography, MS-basedapproach that we recently optimized and published to identifycandidate cancer biomarkers.24 Initially, we probed nitro-cellulose transfers of electrophoretically separated cell lysates ofbreast cancer lines established from triple negative and luminaltumor subtypes with a panel of nine lectins (SNA, AAL, Viciavillosa, Phaseolus vulgaris leukoagglutinating and erythroaggluti-nating, Galanthus nivalis, Euonymus europaeus, Lycopersiconesculentum, and Arachis hypogaea) that recognized eitherinternal saccharide motifs or terminal sugars. The resultsshowed that SNA (Figure 1a) and AAL (data not shown),which bind motifs with sialic acid and fucose, respectively,reacted with a wide array of glycoproteins. Additionally,some glycoforms were enriched in lines that were derivedfrom the tumors of the same subtype. Staining of intactnonpermeabilized cells with fluorescein-conjugated SNArevealed strong surface labeling (Figure 1b). Together, theseresults suggested that the breast cancer cell lines produceda large repertoire of glycoproteins that reacted with SNA orAAL, including cell-surface molecules poised to be shed orreleased.Next, we used this workflow to compare CM samples from

5 luminal and 5 triple negative breast cancer cell lines toidentify subtype-specific glycosites. The cells, listed in Table 1,are members of a well-annotated collection that have been usedto define the gene expression profiles, drug sensitivities, andprotein splicing patterns of the tumor types from whichthey were derived.20,21,23 Contrary to many other lectin-basedapproaches, the affinity capture step was performed at theglycopeptide, rather than the protein level, which decreasednonspecific binding due to hydrophobic interactions, aphenomenon that we previously observed between lectinsand intact proteins. Thus, the samples were trypsin-digestedprior to HPLC separation on lectin-conjugated POROS. Then,the bound fraction was treated with peptide N-glycosidase F(PNGase F) to remove N-linked glycans prior to LC−MS/MS

Journal of Proteome Research Article

dx.doi.org/10.1021/pr201206w | J. Proteome Res. 2012, 11, 2508−25202511

Page 5: Lectin Chromatography/Mass Spectrometry … Chromatography/Mass Spectrometry Discovery Workflow ... The lines were grown to 75−80% confluence in the appropriate ... (FCS) or phenol

analyses. The results were analyzed using two search engines,ProteinPilot and Mascot, to identify peptides and theircorresponding proteins.28 N-glycosylates were identified asdescribed in the Methods.29 Finally, each MS/MS spectrumwas manually inspected for the presence of at least onefragment ion that encompassed an N-glycosylation site. Thus,this method identified the glycosite that carries an oligosac-charide with a lectin-binding motif and the correspondingprotein. These rigorous criteria were key to making this methodhighly reproducible.24

We know from our participation in the Clinical ProteomicTechnologies for Cancer (CPTAC) network that analysis ofthe same sample at multiple sites on different platforms is oneway to maximize identifications and test the robustness of aworkflow.35,36 The experimental strategy we used, whichexploited this observation, is depicted in Figure 2. CM sampleswere trypsin-digested and aliquoted at a single site (Figure 2A).Lectin enrichment and LC−MS/MS analyses were carried outaccording to a Standard Operating Procedure (SOP, Supple-mental Document 1, Supporting Information) at each ofthree locationsUniversity of California San Francisco,Buck Institute for Research on Aging, and Purdue University(Figure 2B). Prior to initiating the study, each groupevaluated the lectin capture step using a National Institute ofStandards and Technology (NIST) human pooled plasmasample, which we have extensively characterized with respectto the SNA and AAL chromatographic profiles and theglycosite composition of the bound fractions.24 MS analysesyielded glycosite identifications and percent enrichmentvalues (total glycopeptides/total peptides) within theexpected range.24

Two groups, M and X, acquired data using a QSTAR EliteQqTOF (AB Sciex), while the third, S, used an LTQ-Orbitrap(Thermo Fisher Scientific). The data sets were submitted toSite M, where all the searches and bioinformatic analyses werecompleted (Figure 2C). As the work progressed, two changesto the protocol were implemented. First, due to technical prob-lems encountered during the initial analysis, a second prepara-

tion of CM samples was analyzed at two of the three locations(M and S). Second, sites M and S replaced ZipTips with spin-cartridges for the desalting step that followed PNGase Fdigestion. This change was made in response to the fact that, ininitial experiments, Site S routinely identified significantly moreglycosites using this desalting method. All peptides and glyco-peptides observed in these experiments are presented as

Figure 1. Breast cancer cell lines have a complex repertoire of SNA-reactive glycoproteins and exhibit cell surface staining with this lectin. (A)Lysates from a panel of 8 breast cancer cell lines, which included triple negative (1−6) and luminal (7, 8) subtypes, were electrophoreticallyseparated, transferred to nitrocellulose, and probed with SNA. Lane 1, MDAMB468; 2, HCC38; 3, HCC1500; 4, HS578T; 5, MDAMB157;6, MDAMB231; 7, T47D; 8, UCC812. (B) Nonpermeabilized HS578T cells were stained with fluorescein-conjugated SNA and imaged byfluorescence microscopy (magnification 60×).

Figure 2. Experimental workflow. (A) CM samples from breastcancer cell lines established from five luminal and 5 triple nega-tive tumors were prepared in one laboratory, then distributed to3 sites. (B) Each group separated the 10 CM samples, in duplicate,by AAL or SNA chromatography, which generated 40 fractions.The samples were deglycosylated using PNGaseF and analyzedin duplicate by LC−MS/MS, yielding a total of 80 MS/MS data setsper site. (C) Files were transferred to a central location for bio-informatic analyses.

Journal of Proteome Research Article

dx.doi.org/10.1021/pr201206w | J. Proteome Res. 2012, 11, 2508−25202512

Page 6: Lectin Chromatography/Mass Spectrometry … Chromatography/Mass Spectrometry Discovery Workflow ... The lines were grown to 75−80% confluence in the appropriate ... (FCS) or phenol

supplemental data (Supplementary Table 1, SupportingInformation).Identification of >500 Cell-Surface or SecretedGlycoproteins

We tabulated the MS identifications according to the CMsamples in which they were detected. Summaries of the data,including the number of glycoproteins, glycopeptides andN-glycosites observed in each CM sample, and the percentglycopeptide enrichment, are shown in Figures 3 and 4, and in

Supplementary Table 2 (Supporting Information). Overall thethree groups identified a total of 1011 distinct N-glycositesfrom 533 glycoproteins. Of these, 945 and 641 were observedfollowing AAL and SNA chromatography, respectively. Interest-ingly, the same workflow applied to pooled healthy human plasmaresulted in many fewer identifications. Approximately half thespecies captured from CM bound to both lectins; the remainderpreferentially interacted either with AAL or SNA. (Figure 3A).A similar phenomenon was observed when the N-glycosites weregrouped according to tumor subtype (Figure 3B and C). Thus,it was clear that employing multiple lectins in our workflowresulted in a greater number of identifications. Furthermore, thedata showed that the luminal and triple negative samplescontained substantially different lectin-reactive species.An overall comparison of the data obtained for luminal and

triple negative samples across the three sites showed relativelyhigh levels of enrichment in both cases (Figure 4). Importantly,very few intracellular proteins were identified, additionalevidence that the cells were not undergoing apoptosis. Approxi-mately 90% of the glycoproteins observed reside either at thecell surface (59%) or in the extracellular matrix (29%), suggest-ing that our strategy of using CM as a source of secreted and/orshed glycoproteins was successful (Figure 5). Since we wantedto identify candidate cancer biomarkers, we were interested tofind that a number of the identified species have functions that

are relevant to tumor biology. For example, we observed protein-ases, including cathepsins and ADAM family members; adhe-sion molecules, including cadherins and integrins; extracellularmatrix components, including decorin and SPARC; and cyto-kines, including leukemia inhibitory factor and vascularendothelial growth factor C. Furthermore, some of the glyco-proteins had been previously identified as putative breastcancer biomarkers, including CD44, galectin-3 bindingprotein, insulin-like growth factor binding protein 3, andtissue inhibitor of metalloproteinase 1.37−39 We alsoidentified clinically useful markers, such as HER2/ErbB2,and the CA-125 antigen, MUC16, which is commonly usedto screen for ovarian cancer but can be also be up-regulatedin breast tumors.40,41

Identification of Putative Glycosite Biomarkers of TripleNegative Breast Cancers

Next, we used statistical analyses to generate a list of putativetriple negative-specific glycosites. Specifically, we performed astatistical analysis using resampling methods that tested 20,000random permutations of the data. This process generated atable (Supplementary Table 3, Supporting Information) withthe number of “triple negative-specific” glycosites expected atrandom for any given set of selection criteria (e.g., observed in“≥1 triple negative and 0 luminal” or “≥4 triple negative and1 luminal”). This analysis allowed us to select parameters thatmaximized the identification of putative triple negative specificglycosites while controlling the FDR. In this context, werequired that a glycosite be identified at least once in CMsamples from ≥3 triple negative cell lines and not observed inluminal CMs. Using these criteria, the computed FDR for bothlectin capture steps was ∼15%. This yielded 49 candidates thatbound to SNA and 76 that bound to AAL (Figure 6). Of these,we removed glycosites from highly polymorphic HLA class Ihistocompatibility antigens, which are variably expressed in thepopulation. The final list of 100 glycosites, from 83 glyco-proteins, that were putative triple negative-specific candidates isshown in Table 2.Next, we asked whether the glycosites we identified could

have been predicted from transcriptome analyses. To answerthis question, we used existing exon expression array profiles forall of the cell lines and RNaseq data for 9 of the 10. Since thetwo platforms identified similar sets of differentially expressedgenes, we performed statistical analyses using values from theRNaseq experiments, which are better able to differentiate signalfrom noise (Supplementary Table 4, Supporting Information).These analyses showed that 46 of the 83 mRNAs encoding theprotein scaffolds that carried biomarker glycosites were up-regulated ≥2-fold in triple negative vs luminal cells. Thissuggested that the differential detection of these glycosites intriple negative CM samples may have been attributable todifferences in relative protein abundances. In contrast, morethan half of the triple negative-specific candidates could nothave been predicted from the mRNA expression data, as therewas no difference in mRNA abundances between the luminaland triple negative subsets. The identification of theseglycosites may have been driven by alterations in the proteinglycosylation machinery of triple negative cell lines. To addressthis possibility, we looked for differences in mRNA levels of thetransferases that add fucose (recognized by AAL), and sialicacid (recognized by SNA). The results are shown in Supple-mentary Table 5 (Supporting Information). Two fucosyltrans-ferases and 8 sialyltransferases were differentially expressed,

Figure 3. Diagrammatic summary of the glycosite (glycoprotein)enrichment data according to lectin type (AAL vs SNA) and CMsamples (luminal vs triple negative) showed distinct and overlappingspecificities. (A) Intersecting circles depict the total number ofN-glycosites (glycoproteins) captured by each lectin. (B and C) Venndiagrams illustrating the chromatographic separation of luminal(LUM) and triple negative (TN) CM samples.

Journal of Proteome Research Article

dx.doi.org/10.1021/pr201206w | J. Proteome Res. 2012, 11, 2508−25202513

Page 7: Lectin Chromatography/Mass Spectrometry … Chromatography/Mass Spectrometry Discovery Workflow ... The lines were grown to 75−80% confluence in the appropriate ... (FCS) or phenol

either up or down-regulated, in triple negative vs luminal celllines. Given that we observed both gains and losses ofenzymatic activity, it is difficult to predict, in structural terms,the net consequences of these changes. However, our glycosite

data are empirical evidence of subtype-specific glycosylationpatterns in breast cancer.

Disease Relevance of Biomarker Scaffolds

Initial inspection of the 100 triple negative-specific candidatesshowed that many targets were derived from glycoproteins thatare involved in cancer-relevant processes. To more fully explorethis correlation, we performed pathway analyses using two

Figure 4. Lectin capture resulted in significant glycopeptide enrichment. The percent enrichment for the separations performed using AAL (left) orSNA (right) at Sites M (top), X (middle), and S (bottom). The dark line indicates the median; the box depicts the first and third quartiles; thewhiskers show the minimum and maximum values observed. Sites M and X acquired data using QSTAR Elite instruments, while Site S used anOrbitrap mass spectrometer.

Figure 5. Nearly 90% of identified glycoproteins resided in the plasmamembrane or extracellular compartments. A portion (241/560) of theidentified glycoproteins were annotated in the cellular component ofGene Ontology. Of these, the great majority were cell surface orsecreted molecules.

Figure 6. Putative triple negative-specific glycosites (glycoproteins)enriched by AAL or SNA. The criteria applied were detection in ≥3triple negative and 0 luminal cell line CMs.

Journal of Proteome Research Article

dx.doi.org/10.1021/pr201206w | J. Proteome Res. 2012, 11, 2508−25202514

Page 8: Lectin Chromatography/Mass Spectrometry … Chromatography/Mass Spectrometry Discovery Workflow ... The lines were grown to 75−80% confluence in the appropriate ... (FCS) or phenol

Table 2. Putative Triple Negative-Specific Glycosites Captured by AAL and SNA

gene name entry nameaccessionnumbera glycoprotein glycosite(s)

observedTNAALb

observedTNSNAb functionc

knownfucosylation

orsialylationd

NT5E 5NTD_HUMAN P21589 5′-nucleotidase N333 3 2 NucleotidaseAPP A4_HUMAN P05067 Amyloid beta A4 protein N571 3 1 Adhesion molecule Yes (58, 59)ANPEP AMPN_HUMAN P15144 Aminopeptidase N N265 0 3 ProteaseANTXR1 ANTR1_HUMAN Q96EC6 Anthrax toxin receptor 1 N184 4 2 Adhesion moleculeB3GNT2 B3GN2_HUMAN B3GNT2 UDP-GlcNAc:betaGal beta-1,3-

N-acetylglucosaminyltransferase 2

N173 3 1 Glycosyltransferase

BMP1 BMP1_HUMAN P13497 Bone morphogenetic protein 1 N91 4 1 Protease Yes (60)BMP1 BMP1_HUMAN P13497 Bone morphogenetic protein 1 N142 3 1 Protease Yes (60)BTD BTD_HUMAN P43251 Biotinidase N150 4 3 Metabolism Yes (61)CDH2 CADH2_HUMAN P19022 Cadherin-2 N325 3 0 Adhesion molecule Yes (62)CDH2 CADH2_HUMAN P19022 Cadherin-2 N402 3 0 Adhesion molecule Yes (62)CDH2 CADH2_HUMAN P19022 Cadherin-2 N692 3 3 Adhesion molecule Yes (62)CTSB CATB_HUMAN P07858 Cathepsin B N38 5 4 Protease Yes (63)CTSL1 CATL1_HUMAN P07711 Cathepsin L1 N221 4 3 ProteaseCTSL2 CATL2_HUMAN O60911 Cathepsin L2 N221 3 2 ProteaseCCDC80 CCD80_HUMAN Q8R2G6 Coiled-coil domain-containing

protein 80N667 3 1 Adhesion molecule

CCDC80 CCD80_HUMAN Q8R2G6 Coiled-coil domain-containingprotein 80

N668 4 3 Adhesion molecule

CD109 CD109_HUMAN Q6YHK3 CD109 antigene N68 3 1 TGF-beta pathwayCD109 CD109_HUMAN Q6YHK3 CD109 antigene N397 4 0 TGF-beta pathwayCD44 CD44_HUMAN P16070 CD44 antigene N25 3 5 Adhesion molecule Yes (64)CGB1 CGB1_HUMAN A6NKQ9 Choriogonadotropin subunit

beta variant 1N63 3 0 Hormone Yes (65)

CLIC1 CLIC1_HUMAN O00299 Chloride intracellular channelprotein 1

N42 4 2 Ion channel

CLU CLUS_HUMAN P10909 Clusterine N354 4 3 Receptor Yes (66)COL1A1 CO1A1_HUMAN P02452 Collagen alpha-1 (I) chain N1365 2 3 ECMCOL5A1 CO5A1_HUMAN P20908 Collagen alpha-1 (V) chain N176 3 4 ECMCOL6A1 CO6A1_HUMAN P12109 Collagen alpha-1 (VI) chain N804 3 3 ECMCOL6A2 CO6A2_HUMAN P12110 Collagen alpha-2 (VI) chain N140 2 3 ECMCOL6A2 CO6A2_HUMAN P12110 Collagen alpha-2 (VI) chain N785 3 3 ECMCOL12A1 COCA1_HUMAN Q99715 Collagen alpha-1 (XII) chain N2679 4 4 ECMCOL18A1 COIA1_HUMAN P39060 Collagen alpha-1 (XVIII) chain N926 3 0 ECMCPVL CPVL_HUMAN Q9H3G5 Probable serine

carboxypeptidase CPVLN346 3 1 Protease

CRIM1 CRIM1_HUMAN Q9NZV1 Cysteine-rich motor neuron 1protein

N71 3 3 Receptor

CRTAP CRTAP_HUMAN O75718 Cartilage-associated protein N87 3 2 ECMDCBLD1 DCBD1_HUMAN Q8N8Z6 Discoidin, CUB and LCCL

domain-containing protein 1N124 4 2 Unknown

DKK3 DKK3_HUMAN Q9UBP4 Dickkopf-related protein 3 N96 3 1 Wnt signalingpathway

ECE1 ECE1_HUMAN P42892 Endothelin-converting enzyme 1 N166 3 0 ProteaseECM1 ECM1_HUMAN Q16610 Extracellular matrix protein 1e N444 3 3 AngiogenesisEXT1 EXT1_HUMAN Q16394 Exostosin-1 N330 4 2 GAG synthesisEXT2 EXT2_HUMAN Q93063 Exostosin-2 N288 3 0 GAG synthesisFAT1 FAT1_HUMAN Q14517 Protocadherin Fat 1 N2328 3 0 Adhesion molecule Yes (67)FBN1 FBN1_HUMAN P35555 Fibrillin-1 N1581 4 3 TGF-beta pathwayFBN1 FBN1_HUMAN P35555 Fibrillin-1 N448 and

N27674 4 TGF-beta pathway

FN1 FINC_HUMAN P02751 Fibronectin N430 3 3 ECM Yes (68)FSTL1 FSTL1_HUMAN Q12841 Follistatin-related protein 1 N175 4 5 Cell growth,

DifferentiationYes (69)

FSTL1 FSTL1_HUMAN Q12841 Follistatin-related protein 1 N180 3 5 Cell growth,Differentiation

Yes (69)

FSTL3 FSTL3_HUMAN O95633 Follistatin-related protein 3 N215 4 3 TGF-beta pathwayFST FST_HUMAN P19883 Follistatin N288 3 2 Hormonal regulation Yes (70)SERPINE2 GDN_HUMAN P07093 Glia-derived nexin N118 4 3 Protease inhibitorGCNT2 GNT2A_HUMAN Q8N0V5 N-Acetyllactosaminide beta-1,6-

N-acetylglucosaminyl-transferase, isoform A

N41 3 2 Glycosyltransferase

Journal of Proteome Research Article

dx.doi.org/10.1021/pr201206w | J. Proteome Res. 2012, 11, 2508−25202515

Page 9: Lectin Chromatography/Mass Spectrometry … Chromatography/Mass Spectrometry Discovery Workflow ... The lines were grown to 75−80% confluence in the appropriate ... (FCS) or phenol

Table 2. continued

gene name entry nameaccessionnumbera glycoprotein glycosite(s)

observedTNAALb

observedTNSNAb functionc

knownfucosylation

orsialylationd

GPC1 GPC1_HUMAN P35052 Glypican-1 N79 5 4 GAGGRN GRN_HUMAN P28799 Granulins N236 5 4 CytokineHSPA13 HSP13_HUMAN P48723 Heat shock 70 kDa protein 13 N184 4 3 ATPaseIGFBP3 IBP3_HUMAN P17936 Insulin-like growth factor-

binding protein 3N199 5 4 Cell growth,

DifferentiationICAM5 ICAM5_HUMAN Q9UMF0 Intercellular adhesion

molecule 5N646 3 2 Adhesion molecule Yes (71)

ITGA3 ITA3_HUMAN P26006 Integrin alpha-3 N265 3 0 Adhesion molecule Yes (72)ITGA5 ITA5_HUMAN P08648 Integrin alpha-5 N868 3 1 Adhesion moleculeITGB1 ITB1_HUMAN P05556 Integrin beta-1 N520 4 3 Adhesion molecule Yes (72)ITGB1 ITB1_HUMAN P05556 Integrin beta-1 N669 4 3 Adhesion molecule Yes (72)JAG1 JAG1_HUMAN P78504 Protein jagged-1 N217 4 0 Cell growth,

DifferentiationLAMC1 LAMC1_HUMAN P11047 Laminin subunit gamma-1 N1205 4 3 ECMLAMC1 LAMC1_HUMAN P11047 Laminin subunit gamma-1 N1395 5 3 ECMLIF LIF_HUMAN P09056 Leukemia inhibitory factor N85 3 0 Cell growth,

DifferentiationLOXL2 LOXL2_HUMAN Q9Y4K0 Lysyl oxidase homologue 2 N288 4 3 ECM cross-linkingLOX LYOX_HUMAN P28300 Protein-lysine 6-oxidase N81 4 2 ECM cross-linkingLOX LYOX_HUMAN P28300 Protein-lysine 6-oxidase N144 4 1 ECM cross-linkingMET MET_HUMAN P08581 Hepatocyte growth factor

receptorN106 3 0 Cell growth,

DifferentiationMFGE8 MFGM_HUMAN Q08431 Lactadherin N325 3 4 Tissue homeostasis Yes (73)MICA MICA_HUMAN Q29983 MHC class I polypeptide-related

sequence AN79 4 3 Immune regulator

MRC2 MRC2_HUMAN Q9UBG0 C-type mannose receptor 2 N497 4 1 ECM remodelingOLFML3 OLFL3_HUMAN Q9NRN5 Olfactomedin-like protein 3 N177 4 3 DevelopmentLEPRE1 P3H1_HUMAN Q32P28 Prolyl 3-hydroxylase 1 N540 4 4 GAGSERPINF1 PEDF_HUMAN P36955 Pigment epithelium-derived

factoreN285 1 4 Cell growth,

DifferentiationPLOD2 PLOD2_HUMAN O00469 Procollagen-lysine,2-

oxoglutarate 5-dioxygenase 2N63 4 1 ECM cross-linking

PLOD3 PLOD3_HUMAN O60568 Procollagen-lysine,2-oxoglutarate 5-dioxygenase 3

N548 3 0 ECM cross-linking

PLTP PLTP_HUMAN P55058 Phospholipid transfer protein N143 3 3 Lipid metabolismPLTP PLTP_HUMAN P55058 Phospholipid transfer protein N398 3 3 Lipid metabolismPOSTN POSTN_HUMAN Q15063 Periostin N599 4 3 Adhesion moleculePPGB PPGB_HUMAN P10619 Lysosomal protective protein N333 1 4 Glycan degradationPRNP PRIO_HUMAN P04156 Major prion protein N181 5 4 Unknown Yes (74)PTK7 PTK7_HUMAN Q13308 Tyrosine-protein kinase-like 7 N405 5 1 Adhesion moleculePTK7 PTK7_HUMAN Q13308 Tyrosine-protein kinase-like 7 N567 5 3 Adhesion moleculePVR PVR_HUMAN P15151 Poliovirus receptor N120 4 3 Immune regulatorSEZ6L2 SE6L2_HUMAN Q6UXD5 Seizure 6-like protein 2 N247 3 2 UnknownSEZ6L2 SE6L2_HUMAN Q6UXD5 Seizure 6-like protein 2 N373 3 1 UnknownSPARC SPRC_HUMAN P09486 SPARC (Osteonectin) N116 5 3 Cell growth,

DifferentiationYes (75)

SUSD5 SUSD5_HUMAN O60279 Sushi domain-containingprotein 5

N354 4 4 Unknown

ABI3BP TARSH_HUMAN Q7Z7G0 Target of Nesh-SH3 N44 3 2 Cell migrationTFPI TFPI1_HUMAN P10646 Tissue factor pathway inhibitor N145 5 4 Complement

cascadeYes (76)

TGFB1 TGFB1_HUMAN P01137 Transforming growth factorbeta-1

N82 4 1 TGF-beta pathway Yes (77)

TGFB2 TGFB2_HUMAN P61812 Transforming growth factorbeta-2

N241 5 4 TGF-beta pathway

THBS3 TSP3_HUMAN P49746 Thrombospondin-3 N407 3 2 Adhesion moleculeTWSG1 TWSG1_HUMAN Q9GZX9 Twisted gastrulation protein

homologue 1N52 3 2 Cell growth,

DifferentiationTXNDC15 TXD15_HUMAN Q96J42 Thioredoxin domain-containing

protein 15N293 3 2 Unknown

AXL UFO_HUMAN P30530 Tyrosine-protein kinase receptorUFO

N43 4 4 Receptor

AXL UFO_HUMAN P30530 Tyrosine-protein kinase receptorUFO

N157 4 2 Receptor

Journal of Proteome Research Article

dx.doi.org/10.1021/pr201206w | J. Proteome Res. 2012, 11, 2508−25202516

Page 10: Lectin Chromatography/Mass Spectrometry … Chromatography/Mass Spectrometry Discovery Workflow ... The lines were grown to 75−80% confluence in the appropriate ... (FCS) or phenol

bioinformatics resources: Kyoto Encyclopedia of Genes andGenomes (KEGG) and Ingenuity (IPA). However, theprograms recognized only small portions of the data set,together matching 38% of the total proteins (SupplementaryTables 6 and 7), and most of the results were driven by only afew molecules, for example, integrins. As an alternative,literature searches enabled assignment of biological functionsto 90% of the putative triple negative-specific glycoproteins.Three prominent, interrelated themes emerged38% of thetargets were up- or downstream components of the TGFβpathway; 21% were involved in ECM remodeling; and at least18% were proteinases or proteolytic targets. Minor recurringassociations included the epithelial to mesenchymal transition(EMT, 9%) and bone morphogenic protein signaling (6%).TGFβ signaling governs important aspects of ECM re-

modeling and proteinase activities. Through the synthesis,cross-linking, and degradation of a variety of protein and carbo-hydrate matrix components, the composition and tensilestrength of the ECM are modulated, both of which dramaticallyinfluence the behavior of surrounding cells.42,43 With respect tocancer, these activities are strongly associated with increasedmigration and invasion. TGFβ is also considered to be a centralmediator of EMT, through both canonical (i.e., Smad-dependent) and noncanonical (e.g., PI3K and MAPK)pathways.44 Cells undergoing EMT lose apical-basal polarityand stabilizing adhesive epithelial interactions in exchange forthe acquisition of a more migratory mesenchymal phenotype.These changes can lead to cell invasion and metastasis, functionsthat have been linked to TGFβ activity.45,46 Thus, as a group, theputative triple negative-specific targets we identified were derivedfrom proteins with striking functional similarities and diseaserelevance.47 It is possible that these biomarker candidates may alsosuggest subtype-specific clinical targets, which currently do notexist for triple negative breast cancer.18,19

Clinical Relevance of Putative Biomarker Targets

The heterogeneous nature of breast cancer is widely accepted.13

Tumor subtyping is commonly based on immunohistochemicalanalyses of tissue sections cut from biopsies to profile expres-sion of a marker panelER, PR, HER2, cytokeratin 5/6 andepidermal growth factor receptor. Increasingly, clinicians areusing this information to determine prognoses and optimizetreatment.48 For example, the risk prediction tool Adjuvant!Online (www.adjuvantonline.com) can be used to identify thepatients who will benefit most from postoperative treatment(s).

Although immunohistology-based diagnoses are changing theclinical oncology landscape and improving patient outcomes,there remains much room for advancement. Currently, subtypediagnoses require identification of a lesion, and an invasive pro-cedure to obtain a biopsy. Therefore, the need for circulatingbiomarkers that serve as sentinels of breast cancer and enablesubtyping remains great.In this context, our biomarker discovery method used cancer

cell line CM, that is, the secretome, as the starting material toidentify candidate glycoproteins that carried putative subtype-specific N-glycosites. For the enrichment step, we used lectincapture at the glycopeptide, rather than glycoprotein level. Thisapproach gives more information, in terms of glycancomposition and location along the peptide backbone, thanother commonly used related methods (e.g., lectin chromatog-raphy at the glycoprotein level, and hydrazide- or boronic acid-mediated chemical capture of glycoproteins/glycopeptides).24

Accordingly, we interrogated a largely unexplored biomarkerdiscovery space. This theory is substantiated by the fact thatonly four of the targets that we identified were among the 150most abundant plasma proteins as described by Hortin et al.49

Furthermore, only 52 of the targets were among the recentlypublished high-confidence human plasma proteome thatincluded estimated protein concentrations.50 Of those foundin this data set, 73% were predicted to be <50 ng/mL, while40% were likely to be <10 ng/mL, reasonably low backgroundlevels against which to observe circulating disease-derivedsignals. As additional support for this concept, only six of theputative triple negative-specific N-glycosites from five glyco-proteins were found in a previous study in which we used thesame workflows and AAL or SNA chromatography to analyze asample of NIST pooled human plasma from 100 healthyindividuals.24 These included glycosites from CD109, CD44,clusterin, extracellular matrix protein 1, and pigment epithelium-derived factor.In summary, the workflow that we developed could serve as a

blueprint for biomarker discovery. In this paradigm, an initialcandidate list is developed using an easily obtained renewablematerial, such as cell line CM, rather than valuable, and oftendifficult to obtain, clinical samples such as plasma or serum. Asstudies that employ targeted enrichment strategies are con-siderably more sensitive than shotgun proteomics methods, theability to generate a candidate biomarker list from a biologicallyrelevant source significantly improves the chances of success

Table 2. continued

gene name entry nameaccessionnumbera glycoprotein glycosite(s)

observedTNAALb

observedTNSNAb functionc

knownfucosylation

orsialylationd

AXL UFO_HUMAN P30530 Tyrosine-protein kinase receptorUFO

N198 4 2 Receptor

AXL UFO_HUMAN P30530 Tyrosine-protein kinase receptorUFO

N339 4 3 Receptor

PLAUR UPAR_HUMAN Q03405 Urokinase plasminogen activatorsurface receptor

N222 2 3 ECM remodeling Yes (78)

PLAU UROK_HUMAN P00749 Urokinase-type plasminogenactivator

N322 4 4 ECM remodeling

Not available YK047_HUMAN Q68D85 Putative Ig-like domain-containing proteinDKFZp686O24166/DKFZp686I21167

N242 3 0 Unknown

aUniprot accession number. bNumber of triple-negative (TN) cell lines in which a glycosite was observed with this lectin. cUniprot databaseannotation. dReferences in parentheses. eDenotes glycoproteins observed in healthy plasma following AAL or SNA enrichment.24

Journal of Proteome Research Article

dx.doi.org/10.1021/pr201206w | J. Proteome Res. 2012, 11, 2508−25202517

Page 11: Lectin Chromatography/Mass Spectrometry … Chromatography/Mass Spectrometry Discovery Workflow ... The lines were grown to 75−80% confluence in the appropriate ... (FCS) or phenol

during the subsequent verification stage.51 This method may beespecially useful for diseases, such as ovarian cancer, for whichthe cell type of origin is uncertain and, consequently, it is dif-ficult to choose control samples.52,53 A limitation of the methodis that O-linked and intact N-linked glycopeptides are notanalyzed due to the absence of universal enzymes to removecarbohydrates and the lack of sufficiently powerful software forrapid identifications, respectively. However, we do not view thisas a liability. This workflow was designed as a high-throughputplatform to generate biomarker candidates for subsequentverification by MRM. In general, due to heterogeneity, endo-genous glycopeptides make poor MRM targets. By contrast, ourmethod yielded a list of putative biomarker targets for directfollow up in clinical samples, and is easily accessible to anylaboratory performing proteomics. Indeed, several groups haverecently employed similar methods to identify candidatebiomarkers of various cancers including prostate, colon, thyroidand breast.54−57 Interestingly, a few of the biomarkers that weidentified were also observed in the latter study, suggesting thatthis general approach is reproducible and robust.54 Finally, thisworkflow is well suited to the development of a multiplexedclinical assay, analogous to a reverse protein array approach,with antibody capture as the first step and lectin binding as thesecond.

■ ASSOCIATED CONTENT

*S Supporting Information

Supplementary materials as described in text. This material isavailable free of charge via the Internet at http://pubs.acs.org.

■ AUTHOR INFORMATION

Corresponding Author

*Susan Fisher, phone (415) 476-5297, fax 415-476-5623,e-mail [email protected]. Bradford W. Gibson, phone (415)209-2032, fax (415) 209-2231, [email protected].

Notes

The authors declare no competing financial interest.

■ ACKNOWLEDGMENTSWe thank Ms. Tiffany Sham for excellent assistance formattingtables. This work was supported by an NCRR sharedinstrumentation grant S10 RR024615 (BWG) and by grantsfrom the National Cancer Institute, U24 CA126477 (S.J.F.)and a U24 Subcontract (B.W.G.) that are part of the NCIClinical Proteomic Technologies for Cancer initiative (http://proteomics.cancer.gov). Additional support was provided bythe Director, Office of Science, Office of Biological &Environmental Research, of the U.S. Department of Energyunder Contract No. DE-AC02-05CH11231, by the NationalInstitutes of Health, National Cancer Institute grants P50 CA58207, the U54 CA 112970, the U24 CA 126477 and the NIHNHGRI U24 CA 126551 for J.W.G. A portion of the massspectrometric analyses was performed in the UCSF Sandler-Moore Mass Spectrometry Core Facility, which acknowledgessupport from the Sandler Family Foundation, the Gordon andBetty Moore Foundation, and NIH/NCI Cancer CenterSupport Grant P30 CA082103. O.L.G. is supported by theCanadian Institutes of Health Research and the Stand Up ToCancer-American Association for Cancer Research Dream

Team Translational Cancer Research Grant SU2C-AACR-DT0409.

■ REFERENCES(1) Drake, P. M.; Cho, W.; Li, B.; Prakobphol, A.; Johansen, E.;Anderson, N. L.; Regnier, F. E.; Gibson, B. W.; Fisher, S. J. Sweeteningthe Pot: Adding Glycosylation to the Biomarker Discovery Equation.Clin. Chem. 2010, 56, 223−36.(2) Hart, G. W.; Copeland, R. J. Glycomics hits the big time. Cell2010, 143 (5), 672−6.(3) Clowers, B. H.; Dodds, E. D.; Seipert, R. R.; Lebrilla, C. B. Sitedetermination of protein glycosylation based on digestion withimmobilized nonspecific proteases and Fourier transform ion cyclo-tron resonance mass spectrometry. J. Proteome Res. 2007, 6 (10),4032−40.(4) Duffy, M. J.; Evoy, D.; McDermott, E. W. CA 15−3: uses andlimitation as a biomarker for breast cancer. Clin. Chim. Acta 2010, 411(23−24), 1869−74.(5) Orntoft, T. F.; Vestergaard, E. M. Clinical aspects of alteredglycosylation of glycoproteins in cancer. Electrophoresis 1999, 20 (2),362−71.(6) Hammarstrom, S. The carcinoembryonic antigen (CEA) family:structures, suggested functions and expression in normal andmalignant tissues. Semin. Cancer Biol. 1999, 9 (2), 67−81.(7) Meany, D. L.; Zhang, Z.; Sokoll, L. J.; Zhang, H.; Chan, D. W.Glycoproteomics for prostate cancer detection: changes in serum PSAglycosylation patterns. J. Proteome Res. 2009, 8 (2), 613−9.(8) Moss, E. L.; Hollingworth, J.; Reynolds, T. M. The role of CA125in clinical practice. J. Clin. Pathol. 2005, 58 (3), 308−12.(9) Witz, I. P. The selectin-selectin ligand axis in tumor progression.Cancer Metastasis Rev. 2008, 27 (1), 19−30.(10) Rosen, S. D. Ligands for L-selectin: homing, inflammation, andbeyond. Annu. Rev. Immunol. 2004, 22, 129−56.(11) Perou, C. M.; Borresen-Dale, A. L. Systems Biology andGenomics of Breast Cancer. Cold Spring Harb. Perspect. Biol. 2011, 3(2, Feb 1), pii: a003293, DOI: 10.1101/cshperspect.a003293.(12) O’Brien, K. M.; Cole, S. R.; Tse, C. K.; Perou, C. M.; Carey,L. A.; Foulkes, W. D.; Dressler, L. G.; Geradts, J.; Millikan, R. C. Intrinsicbreast tumor subtypes, race, and long-term survival in the Carolina BreastCancer Study. Clin. Cancer Res. 2010, 16 (24), 6100−10.(13) Espinosa, E.; Vara, J. A.; Navarro, I. S.; Gamez-Pozo, A.; Pinto,A.; Zamora, P.; Redondo, A.; Feliu, J. Gene profiling in breast cancer:Time to move forward. Cancer Treat. Rev. 2011, 37 (6, Oct), 416−21.(14) Prat, A.; Parker, J. S.; Karginova, O.; Fan, C.; Livasy, C.;Herschkowitz, J. I.; He, X.; Perou, C. M. Phenotypic and molecularcharacterization of the claudin-low intrinsic subtype of breast cancer.Breast Cancer Res. 2010, 12 (5), R68.(15) Toft, D. J.; Cryns, V. L. Minireview: Basal-like breast cancer:from molecular profiles to targeted therapies. Mol. Endocrinol. 2011, 25(2), 199−211.(16) Abramson, V.; Arteaga, C. L. New strategies in HER2-overexpressing breast cancer: Many combinations of targeted drugsavailable. Clin. Cancer Res. 2011, 17 (5, Mar 1), 952−8.(17) McDermott, U.; Settleman, J. Personalized cancer therapy withselective kinase inhibitors: an emerging paradigm in medical oncology.J. Clin. Oncol. 2009, 27 (33), 5650−9.(18) Yagata, H.; Kajiura, Y.; Yamauchi, H. Current strategy for triple-negative breast cancer: appropriate combination of surgery, radiation,and chemotherapy. Breast Cancer 2011, 18 (3, Jul), 165−73.(19) Pal, S. K.; Childs, B. H.; Pegram, M. Triple negative breastcancer: unmet medical needs. Breast Cancer Res. Treat. 2011, 125 (3),627−36.(20) Lapuk, A.; Marr, H.; Jakkula, L.; Pedro, H.; Bhattacharya, S.;Purdom, E.; Hu, Z.; Simpson, K.; Pachter, L.; Durinck, S.; Wang, N.;Parvin, B.; Fontenay, G.; Speed, T.; Garbe, J.; Stampfer, M.;Bayandorian, H.; Dorton, S.; Clark, T. A.; Schweitzer, A.; Wyrobek,A.; Feiler, H.; Spellman, P.; Conboy, J.; Gray, J. W. Exon-levelmicroarray analyses identify alternative splicing programs in breastcancer. Mol. Cancer Res. 2010, 8 (7), 961−74.

Journal of Proteome Research Article

dx.doi.org/10.1021/pr201206w | J. Proteome Res. 2012, 11, 2508−25202518

Page 12: Lectin Chromatography/Mass Spectrometry … Chromatography/Mass Spectrometry Discovery Workflow ... The lines were grown to 75−80% confluence in the appropriate ... (FCS) or phenol

(21) Neve, R. M.; Chin, K.; Fridlyand, J.; Yeh, J.; Baehner, F. L.; Fevr,T.; Clark, L.; Bayani, N.; Coppe, J. P.; Tong, F.; Speed, T.; Spellman,P. T.; DeVries, S.; Lapuk, A.; Wang, N. J.; Kuo, W. L.; Stilwell, J. L.;Pinkel, D.; Albertson, D. G.; Waldman, F. M.; McCormick, F.;Dickson, R. B.; Johnson, M. D.; Lippman, M.; Ethier, S.; Gazdar, A.;Gray, J. W. A collection of breast cancer cell lines for the studyof functionally distinct cancer subtypes. Cancer Cell 2006, 10 (6),515−27.(22) Korkola, J.; Gray, J. W. Breast cancer genomes--form andfunction. Curr. Opin. Genet. Dev. 2010, 20 (1), 4−14.(23) Kuo, W. L.; Das, D.; Ziyad, S.; Bhattacharya, S.; Gibb, W. J.;Heiser, L. M.; Sadanandam, A.; Fontenay, G. V.; Hu, Z.; Wang, N. J.;Bayani, N.; Feiler, H. S.; Neve, R. M.; Wyrobek, A. J.; Spellman, P. T.;Marton, L. J.; Gray, J. W. A systems analysis of the chemosensitivity ofbreast cancer cells to the polyamine analogue PG-11047. BMC Med.2009, 7, 77.(24) Drake, P. M.; Schilling, B.; Niles, R. K.; Braten, M.; Johansen, E.;Liu, H.; Lerch, M.; Sorensen, D. J.; Li, B.; Allen, S.; Hall, S. C.;Witkowska, H. E.; Regnier, F. E.; Gibson, B. W.; Fisher, S. J. A lectinaffinity workflow targeting glycosite-specific, cancer-related carbo-hydrate structures in trypsin-digested human plasma. Anal. Biochem.2011, 408 (1), 71−85.(25) Yingling, J. M.; Blanchard, K. L.; Sawyer, J. S. Development ofTGF-beta signalling inhibitors for cancer therapy. Nat. Rev. DrugDiscovery 2004, 3 (12), 1011−22.(26) Janatpour, M. J.; McMaster, M. T.; Genbacev, O.; Zhou, Y.;Dong, J.; Cross, J. C.; Israel, M. A.; Fisher, S. J. Id-2 regulates criticalaspects of human cytotrophoblast differentiation, invasion andmigration. Development 2000, 127 (3), 549−58.(27) Keshishian, H.; Addona, T.; Burgess, M.; Kuhn, E.; Carr, S. A.Quantitative, multiplexed assays for low abundance proteins in plasmaby targeted mass spectrometry and stable isotope dilution. Mol. Cell.Proteomics 2007, 6 (12), 2212−29.(28) Shilov, I. V.; Seymour, S. L.; Patel, A. A.; Loboda, A.; Tang,W. H.; Keating, S. P.; Hunter, C. L.; Nuwaysir, L. M.; Schaeffer, D. A. TheParagon Algorithm, a next generation search engine that uses sequencetemperature values and feature probabilities to identify peptides fromtandem mass spectra. Mol. Cell. Proteomics 2007, 6 (9), 1638−55.(29) Krokhin, O. V.; Antonovici, M.; Ens, W.; Wilkins, J. A.;Standing, K. G. Deamidation of -Asn-Gly- sequences during samplepreparation for proteomics: Consequences for MALDI and HPLC-MALDI analysis. Anal. Chem. 2006, 78 (18), 6645−50.(30) Link, A. J.; Eng, J.; Schieltz, D. M.; Carmack, E.; Mize, G. J.;Morris, D. R.; Garvik, B. M.; Yates, J. R. 3rd Direct analysis of proteincomplexes using mass spectrometry. Nat. Biotechnol. 1999, 17 (7),676−82.(31) Edgington, E. S. Randomization tests, 3rd ed.; Marcel-Dekker:New York, 1995.(32) MacLean, B.; Tomazela, D. M.; Shulman, N.; Chambers, M.;Finney, G. L.; Frewen, B.; Kern, R.; Tabb, D. L.; Liebler, D. C.;MacCoss, M. J. Skyline: an open source document editor for creatingand analyzing targeted proteomics experiments. Bioinformatics 2010,26 (7), 966−8.(33) Griffith, M.; Griffith, O. L.; Mwenifumbo, J.; Goya, R.; Morrissy,A. S.; Morin, R. D.; Corbett, R.; Tang, M. J.; Hou, Y. C.; Pugh, T. J.;Robertson, G.; Chittaranjan, S.; Ally, A.; Asano, J. K.; Chan, S. Y.; Li,H. I.; McDonald, H.; Teague, K.; Zhao, Y.; Zeng, T.; Delaney, A.;Hirst, M.; Morin, G. B.; Jones, S. J.; Tai, I. T.; Marra, M. A. Alternativeexpression analysis by RNA sequencing. Nat. Methods 2010, 7 (10),843−7.(34) Benjamini, Y.; Hochberg, Y. Controlling the false discovery rate:a practical and powerful approach to multiple testing. J. R. Stat. Soc.,Ser. B 1995, 57 (1), 289−300.(35) Addona, T. A.; Abbatiello, S. E.; Schilling, B.; Skates, S. J.; Mani,D. R.; Bunk, D. M.; Spiegelman, C. H.; Zimmerman, L. J.; Ham, A. J.;Keshishian, H.; Hall, S. C.; Allen, S.; Blackman, R. K.; Borchers, C. H.;Buck, C.; Cardasis, H. L.; Cusack, M. P.; Dodder, N. G.; Gibson,B. W.; Held, J. M.; Hiltke, T.; Jackson, A.; Johansen, E. B.; Kinsinger,C. R.; Li, J.; Mesri, M.; Neubert, T. A.; Niles, R. K.; Pulsipher, T. C.;

Ransohoff, D.; Rodriguez, H.; Rudnick, P. A.; Smith, D.; Tabb, D. L.;Tegeler, T. J.; Variyath, A. M.; Vega-Montoto, L. J.; Wahlander, A.;Waldemarson, S.; Wang, M.; Whiteaker, J. R.; Zhao, L.; Anderson,N. L.; Fisher, S. J.; Liebler, D. C.; Paulovich, A. G.; Regnier, F. E.;Tempst, P.; Carr, S. A. Multi-site assessment of the precision andreproducibility of multiple reaction monitoring-based measurements ofproteins in plasma. Nat. Biotechnol. 2009, 27 (7), 633−41.(36) Tabb, D. L.; Vega-Montoto, L.; Rudnick, P. A.; Variyath, A. M.;Ham, A. J.; Bunk, D. M.; Kilpatrick, L. E.; Billheimer, D. D.; Blackman,R. K.; Cardasis, H. L.; Carr, S. A.; Clauser, K. R.; Jaffe, J. D.; Kowalski,K. A.; Neubert, T. A.; Regnier, F. E.; Schilling, B.; Tegeler, T. J.; Wang,M.; Wang, P.; Whiteaker, J. R.; Zimmerman, L. J.; Fisher, S. J.; Gibson,B. W.; Kinsinger, C. R.; Mesri, M.; Rodriguez, H.; Stein, S. E.; Tempst,P.; Paulovich, A. G.; Liebler, D. C.; Spiegelman, C. Repeatability andreproducibility in proteomic identifications by liquid chromatography-tandem mass spectrometry. J. Proteome Res. 2010, 9 (2), 761−76.(37) Wu, Z. S.; Wu, Q.; Yang, J. H.; Wang, H. Q.; Ding, X. D.; Yang,F.; Xu, X. C. Prognostic significance of MMP-9 and TIMP-1 serumand tissue expression in breast cancer. Int. J. Cancer 2008, 122 (9),2050−6.(38) Wang, Y.; Ao, X.; Vuong, H.; Konanur, M.; Miller, F. R.;Goodison, S.; Lubman, D. M. Membrane glycoproteins associated withbreast tumor cell progression identified by a lectin affinity approach.J. Proteome Res. 2008, 7 (10), 4313−25.(39) Baricevic, I.; Masnikosa, R.; Lagundzin, D.; Golubovic, V.;Nedic, O. Alterations of insulin-like growth factor binding protein 3(IGFBP-3) glycosylation in patients with breast tumours. Clin.Biochem. 2010, 43 (9), 725−31.(40) Bast, R. C. Jr.; Xu, F. J.; Yu, Y. H.; Barnhill, S.; Zhang, Z.; Mills,G. B. CA 125: the past and the future. Int. J. Biol. Markers 1998, 13(4), 179−87.(41) Yin, B. W.; Lloyd, K. O. Molecular cloning of the CA125ovarian cancer antigen: identification as a new mucin, MUC16. J. Biol.Chem. 2001, 276 (29), 27371−5.(42) Yu, H.; Mouw, J. K.; Weaver, V. M. Forcing form and function:biomechanical regulation of tumor evolution. Trends Cell Biol. 2011,21 (1), 47−56.(43) Rowe, R. G.; Weiss, S. J. Navigating ECM barriers at the invasivefront: the cancer cell-stroma interface. Annu. Rev. Cell Dev. Biol. 2009,25, 567−95.(44) Viloria-Petit, A. M.; Wrana, J. L. The TGFbeta-Par6 polaritypathway: linking the Par complex to EMT and breast cancerprogression. Cell Cycle 2010, 9 (4), 623−4.(45) Barcellos-Hoff, M. H.; Akhurst, R. J. Transforming growthfactor-beta in breast cancer: too much, too late. Breast Cancer Res.2009, 11 (1), 202.(46) Bergers, G.; Javaherian, K.; Lo, K. M.; Folkman, J.; Hanahan, D.Effects of angiogenesis inhibitors on multistage carcinogenesis in mice.Science 1999, 284 (5415), 808−12.(47) Hanahan, D.; Weinberg, R. A. Hallmarks of cancer: the nextgeneration. Cell 2011, 144 (5), 646−74.(48) Blows, F. M.; Driver, K. E.; Schmidt, M. K.; Broeks, A.; vanLeeuwen, F. E.; Wesseling, J.; Cheang, M. C.; Gelmon, K.; Nielsen,T. O.; Blomqvist, C.; Heikkila, P.; Heikkinen, T.; Nevanlinna, H.; Akslen,L. A.; Begin, L. R.; Foulkes, W. D.; Couch, F. J.; Wang, X.; Cafourek,V.; Olson, J. E.; Baglietto, L.; Giles, G. G.; Severi, G.; McLean, C. A.;Southey, M. C.; Rakha, E.; Green, A. R.; Ellis, I. O.; Sherman, M. E.;Lissowska, J.; Anderson, W. F.; Cox, A.; Cross, S. S.; Reed, M. W.;Provenzano, E.; Dawson, S. J.; Dunning, A. M.; Humphreys, M.;Easton, D. F.; Garcia-Closas, M.; Caldas, C.; Pharoah, P. D.Huntsman, D., Subtyping of breast cancer by immunohistochemistryto investigate a relationship between subtype and short and long termsurvival: a collaborative analysis of data for 10,159 cases from 12studies. PLoS Med. 2010, 7 (5), e1000279.(49) Hortin, G. L.; Sviridov, D.; Anderson, N. L. High-abundancepolypeptides of the human plasma proteome comprising the top 4 logsof polypeptide abundance. Clin. Chem. 2008, 54 (10), 1608−16.(50) Farrah, T.; Deutsch, E. W.; Omenn, G. S.; Campbell, D. S.; Sun,Z.; Bletz, J. A.; Mallick, P.; Katz, J. E.; Malmstrom, J.; Ossola, R.;

Journal of Proteome Research Article

dx.doi.org/10.1021/pr201206w | J. Proteome Res. 2012, 11, 2508−25202519

Page 13: Lectin Chromatography/Mass Spectrometry … Chromatography/Mass Spectrometry Discovery Workflow ... The lines were grown to 75−80% confluence in the appropriate ... (FCS) or phenol

Watts, J. D.; Lin, B.; Zhang, H.; Moritz, R. L.; Aebersold, R. H. A high-confidence human plasma proteome reference set with estimatedconcentrations in PeptideAtlas. Mol. Cell. Proteomics 2011, 10 (9, Sep),M110.006353.(51) Rifai, N.; Gillette, M. A.; Carr, S. A. Protein biomarker discoveryand validation: the long and uncertain path to clinical utility. Nat.Biotechnol. 2006, 24 (8), 971−83.(52) Lengyel, E. Ovarian cancer development and metastasis. Am.J. Pathol. 2010, 177 (3), 1053−64.(53) Vang, R.; Shih, Ie, M.; Kurman, R. J. Ovarian low-grade andhigh-grade serous carcinoma: pathogenesis, clinicopathologic andmolecular biologic features, and diagnostic problems. Adv. Anat.Pathol. 2009, 16 (5), 267−82.(54) Ahn, Y.; Kang, U. B.; Kim, J.; Lee, C. Mining of serumglycoproteins by an indirect approach using cell line secretome. Mol.Cells 2010, 29 (2), 123−30.(55) Arcinas, A.; Yen, T. Y.; Kebebew, E.; Macher, B. A. Cell surfaceand secreted protein profiles of human thyroid cancer cell lines revealdistinct glycoprotein patterns. J. Proteome Res. 2009, 8 (8), 3958−68.(56) Rangiah, K.; Tippornwong, M.; Sangar, V.; Austin, D.; Tetreault,M. P.; Rustgi, A. K.; Blair, I. A.; Yu, K. H. Differential secretedproteome approach in murine model for candidate biomarkerdiscovery in colon cancer. J. Proteome Res. 2009, 8 (11), 5153−64.(57) Sardana, G.; Jung, K.; Stephan, C.; Diamandis, E. P. Proteomicanalysis of conditioned media from the PC3, LNCaP, and 22Rv1prostate cancer cell lines: discovery and validation of candidateprostate cancer biomarkers. J. Proteome Res. 2008, 7 (8), 3329−38.(58) Akasaka-Manya, K.; Manya, H.; Sakurai, Y.; Wojczyk, B. S.;Spitalnik, S. L.; Endo, T. Increased bisecting and core-fucosylatedN-glycans on mutant human amyloid precursor proteins. Glycoconj. J.2008, 25 (8), 775−86.(59) Nakagawa, K.; Kitazume, S.; Oka, R.; Maruyama, K.; Saido,T. C.; Sato, Y.; Endo, T.; Hashimoto, Y. Sialylation enhances thesecretion of neurotoxic amyloid-beta peptides. J. Neurochem. 2006, 96(4), 924−33.(60) Garrigue-Antar, L.; Hartigan, N.; Kadler, K. E. Post-translationalmodification of bone morphogenetic protein-1 is required forsecretion and stability of the protein. J. Biol. Chem. 2002, 277 (45),43327−34.(61) Wolf, B. Biotinidase Deficiency: New Directions and PracticalConcerns. Curr. Treat. Options Neurol. 2003, 5 (4), 321−28.(62) Ciolczyk-Wierzbicka, D.; Amoresano, A.; Casbarra, A.; Hoja-Lukowicz, D.; Litynska, A.; Laidler, P. The structure of theoligosaccharides of N-cadherin from human melanoma cell lines.Glycoconj. J. 2004, 20 (7−8), 483−92.(63) Takahashi, T.; Schmidt, P. G.; Tang, J. Novel carbohydratestructures of cathepsin B from porcine spleen. J. Biol. Chem. 1984, 259(10), 6059−62.(64) Dimitroff, C. J.; Lee, J. Y.; Rafii, S.; Fuhlbrigge, R. C.; Sackstein,R. CD44 is a major E-selectin ligand on human hematopoieticprogenitor cells. J. Cell Biol. 2001, 153 (6), 1277−86.(65) Elliott, M. M.; Kardana, A.; Lustbader, J. W.; Cole, L. A.Carbohydrate and peptide structure of the alpha- and beta-subunits ofhuman chorionic gonadotropin from normal and aberrant pregnancyand choriocarcinoma. Endocrine 1997, 7 (1), 15−32.(66) Kapron, J. T.; Hilliard, G. M.; Lakins, J. N.; Tenniswood, M. P.;West, K. A.; Carr, S. A.; Crabb, J. W. Identification and character-ization of glycosylation sites in human serum clusterin. Protein Sci.1997, 6 (10), 2120−33.(67) Goldberg, M.; Peshkovsky, C.; Shifteh, A.; Al-Awqati, Q.mu-Protocadherin, a novel developmentally regulated protocadherinwith mucin-like domains. J. Biol. Chem. 2000, 275 (32), 24622−9.(68) Hirnle, L.; Katnik-Prastowska, I. Amniotic fibronectinfragmentation and expression of its domains, sialyl and fucosylglycotopes associated with pregnancy complicated by intrauterineinfection. Clin. Chem. Lab. Med. 2007, 45 (2), 208−14.(69) Miyamae, T.; Marinov, A. D.; Sowders, D.; Wilson, D. C.;Devlin, J.; Boudreau, R.; Robbins, P.; Hirsch, R. Follistatin-like

protein-1 is a novel proinflammatory molecule. J. Immunol. 2006, 177(7), 4758−62.(70) Hyuga, M.; Itoh, S.; Kawasaki, N.; Ohta, M.; Ishii, A.; Hyuga, S.;Hayakawa, T. Analysis of site-specific glycosylation in recombinanthuman follistatin expressed in Chinese hamster ovary cells. Biologicals2004, 32 (2), 70−7.(71) Ohgomori, T.; Funatsu, O.; Nakaya, S.; Morita, A.; Ikekita, M.Structural study of the N-glycans of intercellular adhesion molecule-5(telencephalin). Biochim. Biophys. Acta 2009, 1790 (12), 1611−23.(72) Pochec, E.; Litynska, A.; Amoresano, A.; Casbarra, A.Glycosylation profile of integrin alpha 3 beta 1 changes withmelanoma progression. Biochim. Biophys. Acta 2003, 1643 (1−3),113−23.(73) Newburg, D. S.; Peterson, J. A.; Ruiz-Palacios, G. M.; Matson,D. O.; Morrow, A. L.; Shults, J.; Guerrero, M. L.; Chaturvedi, P.;Newburg, S. O.; Scallan, C. D.; Taylor, M. R.; Ceriani, R. L.; Pickering,L. K. Role of human-milk lactadherin in protection againstsymptomatic rotavirus infection. Lancet 1998, 351 (9110), 1160−4.(74) Stimson, E.; Hope, J.; Chong, A.; Burlingame, A. L. Site-specificcharacterization of the N-linked glycans of murine prion protein byhigh-performance liquid chromatography/electrospray mass spectrometryand exoglycosidase digestions. Biochemistry 1999, 38 (15), 4885−95.(75) Sato, S.; Rahemtulla, F.; Prince, C. W.; Tomana, M.; Butler, W.T. Acidic glycoproteins from bovine compact bone. Connect. Tissue Res.1985, 14 (1), 51−64.(76) Nakahara, Y.; Miyata, T.; Hamuro, T.; Funatsu, A.; Miyagi, M.;Tsunasawa, S.; Kato, H. Amino acid sequence and carbohydratestructure of a recombinant human tissue factor pathway inhibitorexpressed in Chinese hamster ovary cells: one N-and two O-linkedcarbohydrate chains are located between Kunitz domains 2 and 3 andone N-linked carbohydrate chain is in Kunitz domain 2. Biochemistry1996, 35 (20), 6450−9.(77) Brunner, A. M.; Gentry, L. E.; Cooper, J. A.; Purchio, A. F.Recombinant type 1 transforming growth factor beta precursorproduced in Chinese hamster ovary cells is glycosylated andphosphorylated. Mol. Cell. Biol. 1988, 8 (5), 2229−32.(78) Ploug, M.; Rahbek-Nielsen, H.; Nielsen, P. F.; Roepstorff, P.;Dano, K. Glycosylation profile of a recombinant urokinase-typeplasminogen activator receptor expressed in Chinese hamster ovarycells. J. Biol. Chem. 1998, 273 (22), 13933−43.

Journal of Proteome Research Article

dx.doi.org/10.1021/pr201206w | J. Proteome Res. 2012, 11, 2508−25202520


Top Related