You are on page 1of 17

J O U RN A L OF P ROT EO M IC S 8 3 ( 2 01 3 ) 1 9 7 –2 13

Available online at www.sciencedirect.com

www.elsevier.com/locate/jprot

Comparative proteomics, network analysis and


post-translational modification identification reveal
differential profiles of plasma Con A-bound glycoprotein
biomarkers in gastric cancer

Yih-Huei Uena, b, c, d, 1 , Kai-Yuan Linc, e, 1 , Ding-Ping Sunb, 1 , Chen-Chung Liaof, 1 ,


Ming-Song Hsiehg, h, i , Yung-Kai Huangj , Yen-Wei Chenk , Pei-Hsuan Huangf ,
Wei-Jung Chenl , Chih-Chun Taim , Kuan-Wei Leei , You-Chia Cheni , Ching-Yu Lini,⁎
a
Superintendent's Office, Chi-Mei Hospital Chiali, Tainan 722, Taiwan
b
Division of General Surgery, Department of Surgery, Chi-Mei Medical Center, Tainan, 710, Taiwan
c
Department of Medical Research, Chi-Mei Medical Center, Tainan 710, Taiwan
d
Institute of Biomedical Engineering, Southern Taiwan University of Science and Technology, Tainan 710, Taiwan
e
Department of Biotechnology, Chia Nan University of Pharmacy and Science, Tainan 717, Taiwan
f
Proteomics Research Center, National Yang-Ming University, Taipei, Taiwan
g
Department of Infection Control, Microbiological Laboratory, Taipei Medical University-Shuang-Ho Hospital, Taipei Medical University,
New Taipei City, Taiwan
h
Department of Medical Laboratory Science and Biotechnology, Central Taiwan University, Taichung, Taiwan
i
School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical University, Taipei, Taiwan
j
School of Oral Hygiene, College of Oral Medicine, Taipei Medical University, Taiwan
k
Department of Life Science and Institute of Bioinformatics and Structural Biology, College of Life Science, National Tsing Hua University,
Hsin Chu 30013, Taiwan
l
Institute of Biotechnology, National Ilan University, Ilan 26047, Taiwan
m
Department of Laboratory Medicine, Taipei Medical University-Shuang-Ho Hospital, Taipei Medical University, New Taipei City, Taiwan

AR TIC LE I N FO ABS TR ACT

Article history: In the study, we used Con A affinity chromatography, 1-D gel electrophoresis, and
Received 11 May 2012 nano-LC–MS/MS to screen biomarker candidates in plasma samples obtained from 30 patients
Accepted 10 March 2013 with gastric cancer and 30 healthy volunteers. First, we pooled plasma samples matched by age
Available online 26 March 2013 and sex. We identified 17 differentially expressed Con A-bound glycoproteins, including 10
upregulated proteins and 7 downregulated proteins; these differences were significant (Student's
Keywords: t-test, p-value < 0.05). Furthermore, 2 of the upregulated proteins displayed expression levels
Gastric cancer that were increased by 2-fold or more in gastric cancer samples when compared with normal
Glycoproteins control samples. These proteins included leucine-rich alpha-2-glycoprotein (LRG1) and inter-
Concanavalin A alpha-trypsin inhibitor heavy chain H3 (ITIH3), and the expression levels were validated by
Mass spectrometry Western blot analysis. Pathway and network analysis of the differentially expressed proteins by
Post-translational modification Ingenuity Pathway Analysis revealed vital canonical pathways involving acute phase response
Human plasma signaling, the complement system, LXR/RXR activation, hematopoiesis from pluripotent

⁎ Corresponding author at: School of Medical Laboratory Science and Biotechnology, College of Medical Science and Technology, Taipei Medical
University, No. 250, Wuxing Street, Taipei 11031, Taiwan. Tel.: +886 2 2736 1661x3326; fax: +886 2 27324510.
E-mail address: cylin@tmu.edu.tw (C.-Y. Lin).
1
Equal contribution.

1874-3919/$ – see front matter © 2013 Elsevier B.V. All rights reserved.
http://dx.doi.org/10.1016/j.jprot.2013.03.007
198 J O U RN A L OF P ROTE O M IC S 8 3 ( 2 01 3 ) 1 9 7 –21 3

stem cells, and primary immunodeficiency signaling. Our results suggest that Con A-bound LRG1
and ITIH3 may not be practically applicable as a robust biomarker for the early detection of
gastric cancer. Additionally, three novel PTMs in ITIH3 were identified and include hexose-
N-acetyl-hexosamine at asparagine-41, trimethylation at aspartic acid-290, and flavin adenine
dinucleotide at histidine-335.

Biological significance
Our study was to describe a combinatorial approach of Con A affinity chromatography, 1-D
SDS-PAGE, and nano-LC/MS/MS that provides a label-free, comparative glycoproteomic
quantification strategy for the investigation of glycoprotein profiles in plasma from gastric
cancer patients versus healthy volunteers and to identify glycoprotein biomarkers for the
early clinical detection of gastric cancer. Three novel PTMs, HexHexNAc, trimethylation
and FAD, in Con A-bound ITIH3 were identified and built in molecular modeling. The
aspartic acid-290 trimethylation site was located in a metal ion-dependent adhesion site (MIDAS
motif; 290-DXSXS…T…D-313) that may influence important function for binding protein ligands.
© 2013 Elsevier B.V. All rights reserved.

1. Introduction be as low as 50% RSD, but an unlimited number of samples can be


analyzed to independently acquire MS data [17]. However, the
Gastric cancer is one of the most frequent cancers worldwide reproducibility of label-free quantification based on spectral
and shows a wide geographical variation with low incidence counting by 1-D SDS-PAGE coupled to nano-LC–MS/MS ranges
areas in the Americas and high incidence areas in Africa, Europe, from 23.4% to 26.1% RSD [18]. Quantification based on peak
and Asia [1]. Gastric cancer is the fourth most common cancer intensities (extracted ion chromatograms, XIC) is analyzed
and the third leading cause of cancer deaths worldwide [2,3]. In separately in LC–MS/MS runs and can usually provide quantita-
the year 2000, approximately 880,000 people were diagnosed tive accuracy below 30% [19]. The advantages of label-free
with gastric cancer, and approximately 650,000 people died of quantification include its low cost, high proteome coverage,
the disease [1]. In 2008, it was the seventh most prevalent cancer high analytical depth, and high dynamic range. Both label-free
and the sixth leading cause of cancer-related deaths in Taiwan and isotopic labeling quantification require sufficient sample
[4]. Epidemiological studies have indicated that the risks for sizes to overcome individual variability in clinical samples [17]. A
gastric cancer include family history, age, gender, consump- combinatorially pooled sample, 1-D SDS-PAGE and label-free
tion of salt-preserved foods and dietary nitrites, gastrectomy, nano-LC–MS/MS may reduce the complexity of the sample by
Helicobacter pylori (H. pylori) infection, smoking, and alcohol mixing different proteomes.
use [5]. The patients with gastric cancer have a relatively poor Over the past decade, clinical proteomics has successfully
prognosis; the global 5-year survival rate is approximately combined the development of advanced technology and bioin-
20%. However, the rate in Taiwan is less than 40% [4,6]. The formatics to determine the protein/peptide biomarkers responsi-
available serum tumor markers carcinoembryonic antigen ble for disease through altered levels of expression and/or PTMs
(CEA), cancer antigen 19-9 (CA 19-9), and cancer antigen 72-4 (CA and different protein variants [20]. Biomarkers can be divided into
72-4) used in the clinic do not demonstrate sufficient sensitivity three types based on the analytes and assay methods. First,
for the early detection of gastric cancer [7]. Therefore, there is an altered expression of the total protein is measured by competitive
urgent need to discover better biomarkers for gastric cancer. ELISA. Second, single, specific PTMs of proteins are detected with
An important criterion for biomarker candidate discovery is mAb by competitive and sandwich ELISA assays. Third, multiple
the quantitative comparison between disease samples and PTMs of proteins are detected with different specific mAbs by the
control samples. Quantification methods include a labeling sandwich assay [21]. Anderson [22] has analyzed all of the
approach that requires incorporation of the stable isotope protein-based assays cleared or approved by the US Food and
labels 12/13C, 14/15N, and/or 16/18O into proteins or peptides prior to Drug Administration (FDA) (Jan 1993 to Jan 2009), and these
MS analysis or the use of a label-free method. Labeling methods assays have revealed 109 unique protein analytes in the plasma
include methanol (substituted with hydrogen or deuterium) and/or serum, 62 additional tests, and 96 laboratory-developed
esterification reactions [8], tryptic digestion in the presence tests (LDTs), yielding a total of 205 unique proteins. The 62
of H2(16/18)O [9], 12/13C-acrylamide-cysteine labeling [10], 13CNBS additional tests consisted of peptides (7 tests), protein PTMs (18
(2-nitrobenzenesulfenyl) labeling [11], isotope-coded protein tests), protein complexes (3 tests), autoantibodies (20 tests), and
labeling (ICPL) [12], ICAT [13], stable isotope labeling with amino blood cell proteins (14 tests). The 18 protein PTM biomarkers
acids in cell culture (SILAC) [14] and iTRAQ [15]. In general, included fucosylated glycoforms of AFP (AFP-L3%), glycated
labeling methods demonstrate high precision for quantification albumin, bone-specific alkaline phosphatase (BAP), CA 19-9,
(typically 10% RSD), but restrictions due to isotope labels and type I collagen cross-linked deoxypyridinoline, type I collagen
experimental operation limit the number of samples that can be cross-linked N-terminal telopeptide, type I collagen cross-
tested for biomarker discovery [16]. In contrast, the quantitative linked pyridinium, type I collagen C-terminal telopeptide,
precision of label-free techniques based on spectral counting can D-dimer, des-gamma-carboxy prothrombin (DCP), DR-70,
J O U RN A L OF P ROT EO M IC S 8 3 ( 2 01 3 ) 1 9 7 –2 13 199

fibrin monomers, fibrin(ogen) split/degradation products Therefore, we need an approach to determine potentially novel
(FSP/FDP), fibrinopeptide A, glycated low-density lipopro- glycosylation biomarkers.
tein (LDL), plasmin, prothrombin fragment 1.2 (F1.2), and In this study, our aim was to describe a combinatorial
carbohydrate-deficient transferrin. approach of Con A affinity chromatography, 1-D SDS-PAGE,
PTMs are the chemical modification of proteins after mRNA and nano-LC/MS/MS that provides a label-free, comparative
translation, and they play pivotal roles in cellular physiology and glycoproteomic quantification strategy for the investigation of
disease progression [23–25]. Currently, more than 300 different glycoprotein profiles in plasma from gastric cancer patients
types of PTMs have been identified [26]. These PTMs include the versus healthy volunteers and to identify glycoprotein bio-
addition of chemical groups (e.g., phosphate, citrulline, biotin, markers for the early clinical detection of gastric cancer. We
or acetate) or more complex molecules (e.g., carbohydrates, also used bioinformatic tools to search for protein ontology,
nucleotide, or lipids), the covalent linkage of small proteins category classifications, molecular modeling, PTM peptides, PTM
(ubiquitin or ubiquitin-like proteins), the modification of sites, and pathway analyses. Furthermore, Western blot and Con
side-chain residues of specific amino acids (acetylation, A analyses were performed to confirm the differential expres-
deimination, deamidation, farnesylation, glycosylation, hydrox- sion levels of glycoprotein biomarkers. The potential biological
ylation, methylation, nitration, oxidation, palmitoylation, phos- roles of these altered expression glycoproteins and novel PTM
phorylation, sulfation, SUMOylation, and ubiquitination) and sites in gastric cancer plasma are discussed.
proteolysis [27]. Protein PTMs have led to the alteration of gene
and protein expression levels to activate specific signaling
pathways, enhance cell proliferation, and dysregulate apoptosis 2. Materials and methods
during cancer progression [28–30]. The identification of each type
of PTM may be developed as a unique cancer-specific biomarker, 2.1. Patient samples
and the PTM peptide sequence has also been considered a
neo-epitope. PTM analysis of the plasma proteome has thus Plasma samples from 90 patients with gastric cancer and 90
become an important area for biomarker discovery [27]. age- and sex-matched healthy volunteers (normal controls)
Glycosylated proteins exist as multiple glycoforms that can be were obtained from the Tumor and Serum Bank of Chimei
captured by hydrazide resin and lectin affinity chromatography, Medical Center and the Department of Laboratory Medicine,
among other methods [31]. The tumor markers CEA, CA 19-9, and Shuang-Ho Hospital in Taiwan (Table 1). This study was
CA 72-4 are glycoproteins used for the detection of gastric cancer approved by the hospital ethics committee, and all patients
[7]. Most FDA-approved cancer biomarkers are glycoproteins or and volunteers provided informed consent for the study. The
glycans, and none demonstrate the specificity required for gastric cancer plasma samples were from patients with stage I
diagnosis when used alone [32]. Therefore, protein-based multi- tumors (27 samples), stage II tumors (21 samples), stage III
plex assays, such as the in vitro diagnostic multivariate index tumors (21 samples), and stage IV tumors (21 samples). For
assay (IVDMIA) and the FDA-approved OVA1™ test (Sep 2009) discovery, pooled plasma samples were obtained from 30
[33], will be critical in achieving the high sensitivity and specificity gastric cancer patients and 30 normal controls (the discovery
required for early cancer diagnosis. The panel of 5 biomarkers in set), and the protein panels were further validated against
the OVA1™ test contains of CA125, transthyretin (prealbumin), individual plasma samples from 60 gastric cancer patients
apolipoprotein A1 (APOA1), beta-2 microglobulin (B2M), and and 60 normal controls (the validation set) by Western blot
transferrin (TF). The OVA1 score for serum ranges from 0 to 10 analysis. The validation set of plasma from gastric cancer
and is determined by an algorithm analysis that combines 5 patients and normal controls was separated into three groups
immunoassays into a single numerical result. Different containing plasma from 120 samples [stage I/II tumors (33
cut-off values are used for premenopausal women (5.0) and samples), stage III/IV tumors (27 samples), and normal
postmenopausal women (4.4). The sensitivity of OVA1™ is 89% in controls (60 samples)]. For the validation experiment of the
premenopausal women and 98% in postmenopausal women [34]. discovery set, 30 normal controls (63.3 ± 8.50 years old) and 30

Table 1 – The demographic and clinical characteristics of the individual subjects contributing to the pooled plasma for
gastric cancer patients and normal control samples.
Characteristic Discovery set Validation set
(N = 60) (N = 120)

Cancer, n (%) Control, n (%) Cancer, n (%) Control, n (%)

(n = 30) (n = 30) (n = 60) (n = 60)

Age (years), mean ± SD 65.33 ± 9.03 63.3 ± 8.50 59.92 ± 13.10 58.30 ± 12.80
Gender
Male 16 (53.33%) 16 (53.33%) 36 (60.0%) 36 (60.0%)
Female 14 (46.67%) 14 (46.67%) 24 (40.0%) 24 (40.0%)
Stage of gastric cancer
I 10 (33.33%) 17 (28.33%)
II 5 (16.67%) 16 (26.67%)
III 10 (33.33%) 11 (18.33%)
IV 5 (16.67%) 16 (26.67%)
200 J O U RN A L OF P ROTE O M IC S 8 3 ( 2 01 3 ) 1 9 7 –21 3

gastric cancer patients (65.33 ± 9.03 years old) were selected. 100 ml of 3% acetic acid for 10 min. The gel was then washed
Sixty normal controls (58.30 ± 12.80 years old) and 60 gastric with 25 ml of oxidizing solution for 15 min and washed three
cancer patients (59.92 ± 13.10 years old) participated in the times with 100 ml of 3% acetic acid for 5 min. The gel was next
validation experiment of the validation set. soaked with 25 ml of glycoprotein staining reagent for 15 min
and with 25 ml of reducing solution for 5 min. Finally, the gel was
2.2. Con A affinity chromatography washed with 3% acetic acid followed by deionized water. The
N-deglycosylation reaction of plasma-derived Con A-bound
For glycoprotein enrichment, we used agarose-bound Con A glycoproteins used PNGase F (New England BioLabs, Ipswich,
according to the manufacturer's protocol (Vector Laboratories, MA, USA) treatment and was verified by PAS staining. A duplicate
Burlingame, California, USA). Briefly, 300 μL of agarose-bound gel was run and stained with CBB G-250 as a loading control.
Con A was added to a 1.5 ml centrifuge tube. The agarose-bound
Con A was washed with 3 volumes of binding buffer (25 mM Tris, 2.5. SDS-PAGE and in-gel digestion
1 mM MnCl2, 1 mM CaCl2, and 500 mM NaCl, pH 7.6). Then,
100 μL of pooled plasma was added and mixed with agarose- Plasma protein samples were fractionated by 12% SDS-PAGE
bound Con A at 4 °C overnight. After centrifugation, the (Hoefer®, Inc., Holliston, Massachusetts, USA) according to a
agarose-bound Con A was washed with 8 volumes of binding previously described method [35]. The gels were stained with CBB
buffer to remove nonspecifically bound proteins. Finally, the staining solution (Bio-Rad Laboratories, Hercules, California,
captured glycoproteins were eluted 6 times with 200 μL of elution USA), and 50 μg of each protein sample was loaded onto the gel
buffer (500 mM alpha-methyl mannoside in 20 mM Tris, pH 7.6). in triplicate. The gel lanes were cut into 10 fractions based on
The eluted fractions were pooled and concentrated with a molecular weight, and the gel slices were destained repeatedly in
Microcon YM-10 centrifugal filter unit (Millipore Corporation, a solution of 25 mM NH4HCO3 and 50% (v/v) CAN (1:1) until no
Billerica, Massachusetts, USA), and the Con A-captured glycopro- protein bands were visible. After drying in a Speed-Vac (Thermo
teins were subjected to quantification by the Bradford assay Electron, Waltham, Massachusetts, USA), the slices were
(Bio-Rad Laboratories, Hercules, California, USA). The features of incubated with 2% β-mercaptoethanol and 25 mM NH4HCO3 for
the plasma-derived Con A-bound glycoproteins were confirmed 20 min at room temperature in the dark to reduce the disulfide
using N-deglycosylation strategies, including PNGase F (New bonds in the proteins. For cysteine alkylation, an equal volume of
England BioLabs, Ipswich, MA, USA) treatment, re-Con A affinity 10% 4-vinylpyridine in 25 mM NH4HCO3 and 50% CAN was added
chromatography and analysis by LC–MS/MS. PNGase F treatment for 20 min. After soaking the slices in 1 ml of 25 mM NH4HCO3 for
of the plasma-derived Con A-bound glycoproteins was performed 10 min, the slices were dried in a Speed-Vac for 20 min. Then,
according to the manufacturer's protocol. Briefly, a 10 μL reaction 100 ng of modified trypsin (Promega, Mannheim, Germany) in
including 50 μg of plasma-derived Con A-bound glycoproteins 25 mM NH4HCO3 was added, and tryptic digestion was performed
and 1× glycoprotein denaturing buffer was denatured by heating at 37 °C overnight. The tryptic digest was removed from the gel,
at 100 °C for 10 min. A 20 μL reaction containing denatured dried in a Speed-Vac and stored at −20 °C until further analysis.
glycoproteins, 1× G7 reaction buffer, 1% NP-40 and 1000 U of The tryptic peptides were resuspended in 0.1% formic acid
PNGase F was incubated at 37 °C for 1 h. A parallel reaction immediately before use [18,36].
without PNGase F served as a control.
2.6. Mass spectrometric analysis
2.3. Depletion of high abundance proteins from plasma
Triplicate tryptic peptide mixtures were injected into a nano-flow
Albumin removal using the Albumin and IgG Depletion SpinTrap HPLC system (Agilent Technologies 1200 series, Waldbronn,
column (GE Healthcare Life Sciences, Piscataway, New Jersey, Germany) coupled to an LTQ-Orbitrap Discovery™ hybrid mass
USA) was performed according to the manufacturer's protocol. spectrometer with a nanoelectrospray ionization source (Thermo
Briefly, 30 μL of pooled plasma was diluted with binding buffer Electron, Waltham, Massachusetts, USA). The tryptic peptides
(20 mM sodium phosphate and 150 mM NaCl, pH 7.4) to a final were separated on an Agilent C18 column (100 × 0.075 mm,
volume of 100 μL. Then, 100 μL of the sample was applied to the 3.5 μm in diameter) with mobile phases of 0.1% formic acid in
column, and the column was incubated at 4 °C for 1 h. The water (solvent A) and 0.1% formic acid in CAN (solvent B) at a flow
column was eluted with 5 volumes of binding buffer to collect the rate of 0.5 μL/min using a 30 min linear gradient of 5% to 35%
depleted samples. The depleted samples were pooled and solvent B followed by 95% solvent B for 10 min. Following each
concentrated with a Microcon YM-10 centrifugal filter unit full scan (m/z range of 200–2000), a data-dependent acquired MS/
(Millipore Corporation, Billerica, Massachusetts, USA), and pro- MS scan for a series of precursor ions was selected on the basis of
tein quantification was determined with the Bradford assay the conventional MS spectra (Survey Scan) triggered at high
(Bio-Rad Laboratories, Hercules, California, USA). resolution (M/DM, 60,000 full width half maximum). The former
acquired the spectrum (CID spectrum or MS/MS spectrum) for the
2.4. Periodic acid-Schiff (PAS) staining for glycoproteins fragment ions generated by CID, whereas the latter examined the
accurate mass and the charge state of the selected precursor ion.
The glycoprotein sugar moieties in polyacrylamide gels were
stained using a Pierce Glycoprotein Staining Kit (Thermo 2.7. Database searching
Scientific, Rockford, Illinois, USA) according to the manufacturer's
protocol. Briefly, 50 μg of proteins in the gel was fixed with 100 ml A MS/MS dataset generated by nano-LC/MS/MS was analyzed
of 50% methanol for 30 min. The gel was washed twice with by Xcalibur 2.0 SR1 software (Thermo Electron, Waltham,
J O U RN A L OF P ROT EO M IC S 8 3 ( 2 01 3 ) 1 9 7 –2 13 201

Massachusetts USA) to generate peak lists. The SEQUEST different peptides or one peptide detected by at least two
algorithm (TURBO) was used to identify peptide sequences different fractions or scan times) for protein identification.
against the Universal Protein Resource Knowledgebase, a The individual molecules participating in the protein–protein
human protein database (UniProt; http://www.uniprot.org/) that interactions were searched using Ingenuity Pathway Analysis
contains ion scans obtained from MS/MS data. Information on software (IPA; Ingenuity Systems, USA). Additionally, the mass
category classifications in the protein annotation, including spectra of candidate biomarkers [Con A-bound leucine-rich
cellular components, molecular functions, and biological pro- alpha-2-glycoprotein (LRG1) and inter-alpha-trypsin inhib-
cesses, was recorded. The database searches permitted fixed itor heavy chain H3 (ITIH3)] were searched to allow a wide
modification of the cysteine residue (carboxyamidomethylation, range of modifications using our in-house PTM finder
57 Da), variable modification of the methionine residue (oxida- program that subjects the protein sequence to alignment
tion, 16 Da), peptide mass tolerance of 1.5 Da, and fragment mass to identify PTM peptide sequences, PTM sites, and quanti-
tolerance of 1 Da. These peptides were filtered by Xcorr versus fication [36]. MS spectral counts were normalized on the
charge state with a p-value threshold < 0.05 to determine sum of the spectral counts per biological sample in
significance. Xcorr was used as a match with 1.9 for singly quantitative analyses [18]. Furthermore, all modified MS1
charged ions, 2.2 for doubly charged ions and 3.75 for triply spectra were manually identified in this study, and the
charged ions. Additional filtering was performed for proteins fragmented ions (from the nano-LC/MS/MS) were labeled as
scoring above ten and those having at least two hits (two b, y, y-NH3, and b-H2O ions.

Table 2 – Differentially expressed Con A-bound glycoprotein biomarkers in plasma identified by 1-D SDS-PAGE and LC–MS/
MS analysis in gastric cancer and normal samples.
Protein Symbol Protein name Normalized average p-Value Fold Glycosylation N-Deglycosylation
ID spectral counts change a
(mean ± RSD)

Cancer Normal

P02750 LRG1 Leucine-rich 12.97 ± 4.00 ± 0.004 ↑2.24 N-linked/ N.D.


alpha-2-glycoprotein 18.73% 19.41% O-linked
Q06033 ITIH3 Inter-alpha-trypsin 21.54 ± 7.03 ± 0.022 ↑2.06 N-linked N.D.
inhibitor heavy chain H3 32.05% 9.36%
P02790 HPX Hemopexin 41.96 ± 20.78 ± 0.026 ↑1.02 N-linked/ +
7.97% 48.51% O-linked
P05155 SERPING1 Plasma protease 25.43 ± 15.14 ± 0.027 ↑0.68 N-linked/ +
C1 inhibitor 13.49% 26.18% O-linked
P04217 A1BG Alpha-1B-glycoprotein 94.32 ± 63.68 ± 0.022 ↑0.48 N-linked +
11.23% 15.80%
P01876 IGHA1 Ig alpha-1 chain C region 143.84 ± 102.52 ± 0.010 ↑0.40 N-linked/ +
10.55% 3.85% O-linked
P00751 CFB Complement factor B 38.42 ± 27.81 ± 0.004 ↑0.38 N-linked +
2.47% 10.37%
P02751 FN1 Fibronectin 80.09 ± 59.85 ± 0.002 ↑0.34 N-linked/ +
5.33% 4.12% O-linked
P0CG06 IGLC3 Ig lambda-3 chain 5.75 ± 4.37 ± 0.039 ↑0.32 N.A. c N.D.
C regions 10.60% 11.52%
P19827 ITIH1 Inter-alpha-trypsin 72.44 ± 55.75 ± 0.010 ↑0.30 N-linked/ +
inhibitor heavy chain H1 5.76% 8.44% O-linked
P01859 IGHG2 Ig gamma-2 chain C region 11.57 ± 25.50 ± 0.016 ↓1.20 N-linked +
52.07% 1.87%
P01871 IGHM Ig mu chain C region 32.69 ± 54.07 ± 0.024 ↓0.65 N-linked +
18.26% 15.79%
P10909 CLU Clusterin 17.58 ± 27.37 ± 0.038 ↓0.56 N-linked N.D.
22.32% 14.32%
P05090 APOD Apolipoprotein D 15.80 ± 20.89 ± 0.024 ↓0.32 N-linked +
10.60% 8.80%
P00747 PLG Plasminogen N.D. b 5.41 ± 0.002 – N-linked/ N.D.
25.38% O-linked
P01031 C5 Complement C5 N.D. 4.13 ± 0.028 – N-linked N.D.
51.18%
P02746 C1QB Complement C1q N.D. 5.38 ± 0.000 – N.A. N.D.
subcomponent subunit B 9.56%
a
Fold change represent increment (↑) and reduction (↓) fold change compared with gastric cancer versus normal samples.
b
N.D., not detectable.
c
N.A., not available.
202 J O U RN A L OF P ROTE O M IC S 8 3 ( 2 01 3 ) 1 9 7 –21 3

2.8. Molecular modeling Piscataway, New Jersey, USA) and ImageJ software (National
Institutes of Health, Bethesda, Maryland, USA). Western blot
Molecular modeling, figure building, and sequence align- analysis of each protein was performed in duplicate, and a
ment of ITIH3 were performed using SWISS-MODEL (http:// duplicate gel was run and stained with CBB as a loading control.
swissmodel.expasy.org/) [37–39], PyMOL (http://www.pymol.org/)
and multiple sequence alignment (MSA) (http://workbench.sdsc. 2.11. Statistical analyses
edu) software, respectively [36].
Student's t tests were performed to determine the significance of
2.9. Ingenuity Pathway Analysis peptide mass spectra and blot densitometry differences. The
spectral count data are presented as the mean ± RSD. The RSD is
Functional pathway and network analyses of 17 differen- a measure of precision and is usually called the CV. It is often
tially expressed Con A-bound glycoproteins in the plasma of calculated as a percentage. Fold change is defined as (mean of
gastric cancer samples versus normal controls were cancer normalized spectral counts − mean of normal normalized
assessed through the use of IPA analysis. No expression spectral counts) / (mean of normal normalized spectral counts).
value cut-off was selected, and we investigated both The threshold for an upregulated or downregulated protein was a
upregulated and downregulated proteins. The modulated 1.0-fold change in expression. Proteins that were upregulated by
proteins are grouped by known relationships into the 2-fold or more were selected for validation by Western blot
“Functional Analysis” and “Canonical Pathway Analysis” analysis. An ANOVA and Scheffe's post hoc test were used to test
of a network, with significance calculated using Fisher's the LRG1 and ITIH3 protein levels in Con A-captured plasma
exact test (p-value < 0.05) to determine the probability of among normal controls and early- and late-stage gastric cancer
each biological function and/or disease. The canonical samples. SAS version 9.3 (SAS Institute Inc., Cary, North Carolina,
pathway may be explained by chance alone. For the IPA USA) was used for all statistical analyses. The ROC curve was
network analysis, hypothetical protein interaction clusters generated to evaluate the diagnostic performance of Con
between the molecules were analyzed. IPA computes a A-bound LRG1 and ITIH3 using the Stata 9.0 software package
p score to rank networks according to the differentially (StataCrop LP, College Station, TX, USA). For all statistical tests,
expressed proteins. The score is calculated as p score = − log the level of significance was established at 0.05.
10 (p value), which indicates the probability of matching the
input proteins in a protein–protein interaction from the
Ingenuity Knowledgebase by random chance. A score of 3
or higher indicates at least a 99.9% confidence level for 3. Results
excluding random chance [40].
3.1. Detection and enrichment of Con A-captured plasma
2.10. Western blot analyses and Con A blotting
Approximately 88.2% (15/17) and 41.2% (7/17) of the identified
The protein samples (the albumin- and IgG-depleted and differentially expressed proteins were N-linked and O-linked
the plasma-derived Con A-captured glycoprotein samples) glycoproteins, respectively, as annotated by the UniProt
were separated by electrophoresis on a 10% SDS polyacryl- database (Table 2). We utilized an N-deglycosylation strategy to
amide gel (Hoefer®, Inc., Holliston, Massachusetts, USA) confirm the characteristics of plasma-derived Con A-bound
and transferred to a PVDF membrane (GE Healthcare Life glycoproteins. Of the identified glycoproteins, 41.2% (7/17) were
Sciences, Piscataway, New Jersey, USA). Briefly, a total of not detectable by LC–MS/MS (Table 2). The PAS staining approach
50 μg of protein was loaded in each well. The proteins were was used to identify glycoproteins present in the plasma. With
resolved in Tris–glycine buffer (0.1% SDS (w/v), 25 mM Tris, this technique, periodic acid oxidizes the 1,2-glycol functional
192 mM glycine, pH 8.3). After electrophoresis, the separat- groups present in glucose or polysaccharides, resulting in
ed proteins were transferred to PVDF membranes in aldehydes that react with Schiff regent to turn a purple-magenta
transfer buffer (20% methanol, 25 mM Tris base, 192 mM or pink color. After SDS-PAGE, proteins in the molecular weight
glycine, pH 8.0). The blots were blocked with 1 × Carbo-Free (MW) range of 35 to 250 kDa were more highly stained with PAS
™ Blocking Solution (Vector Laboratories, Burlingame, in the Con A-captured plasma samples than in the albumin- and
California, USA) in deionized water with 0.05% Tween-20 IgG-depleted samples as shown in Supplementary Fig. 1A (see
and then incubated with the following antibodies: ITIH3 Supporting Information). This result strongly indicated that the
(dilution 1:200; Santa Cruz Biotechnology, Inc., USA), LRG1 Con A-captured plasma samples contain more glycoproteins
(mAb 2E3, dilution 1:500; Abnova, Taiwan), and biotinylated than the albumin- and IgG-depleted plasma samples. However,
Con A (dilution 1:1000; Vector Laboratories, Burlingame, proteins with a MW less than 35 kDa may be less efficiently
California, USA). After washing, the blots were subsequently stained by PAS. To study the differential profiles of glycoprotein
incubated with HRP-conjugated monoclonal IgG antibody content in gastric cancer versus normal control plasma samples,
(dilution 1:10,000; Millipore Corporation, Billerica, Massachusetts, the albumin- and IgG-depleted samples and the Con A-captured
USA) as the secondary antibody. The reactive protein bands were plasma samples were separated by SDS-PAGE, transferred to
detected using the Immobilon™ Western Chemiluminescent PVDF membranes and visualized with biotin-labeled Con A
HRP Substrate (Millipore Corporation, Billerica, Massachusetts, lectins (Supplementary Fig. 1B). The Con A blotting results
USA). The band intensity was analyzed and estimated using showed differential glycoprotein profiles in both samples
an ImageQuant 400™ Imager (GE Healthcare Life Sciences, depending on the broad glycan specificity and preferential
J O U RN A L OF P ROT EO M IC S 8 3 ( 2 01 3 ) 1 9 7 –2 13 203

binding to oligomannosidic/glucosidic, hybrid, and biantennary normal controls according to peptide identification and/or
N-glycans. A few Con A-bound proteins were detected in the quantification values by LC–MS/MS. Therefore, we further
MW range of 55 to 110 kDa in the albumin- and IgG-depleted investigated the Con A-bound LRG1 and ITIH3 expression levels
plasma samples. Most Con A-bound proteins were present as in human plasma to determine whether these proteins might be
strong protein bands in the MW range of 35 to 250 kDa in the valuable biomarker candidates for gastric cancer diagnosis. We
Con A-captured plasma samples. The comparative results of used the albumin- and IgG-depleted samples and the Con A-
the PSA staining and Con A blotting were highly consistent for captured plasma pools for the initial validation, as the presence
the Con A-captured plasma samples. The detection range of the of high-abundance plasma proteins may interfere with the
Con A blotting is smaller than that of the PSA staining as shown detection of low-abundance proteins by Western blot analysis.
in the albumin- and IgG-depleted plasma samples. The gel band In a previous study, the mAb against LRG1 recognized one
patterns of plasma-derived Con A-bound glycoproteins after protein band at ~47 kDa in human plasma [41]; however, we
the N-deglycosylation reaction were obviously changed, and determined that the major molecular weight of LRG1 in plasma
most glycoproteins were not stained by the PAS reaction was ~55 kDa. Likewise, the predicted molecular weight of ITIH3
(Supplementary Fig. 1C–D). is approximately 80 or 125 kDa [64], but ITIH3 was detected as a
major band at ~125 kDa in this study. In the discovery set, the
3.2. Identification of the differentially expressed Con A-bound densitometry of the Western blot data showed 1.20- (average
glycoprotein biomarkers by nano-LC–MS/MS densitometry value: 14,248 versus 11,866; p = 0.58) and 1.40-fold
(average densitometry value: 33,835 versus 24,249; p = 0.001)
The enrichment of Con A-captured plasma samples from a overexpression of LRG1, respectively, in the albumin- and
single pair of each of the 30 pooled plasma samples (gastric IgG-depleted samples and the Con A-captured plasma pools of
cancer versus normal control samples) was analyzed in triplicate gastric cancer compared with normal controls. Similarly, ITIH3
by 1-D SDS-PAGE coupled to nano-LC–MS/MS. A list of 82 expression levels were 1.35- (average densitometry value: 28,315
proteins and peptides was identified at a false discovery rate versus 21,003; p = 0.005) and 1.56-fold (average densitometry
(FDR) of 0.3% (Supplementary Table 1). Seventeen of the value: 21,881 versus 14,022; p = 0.013) higher in gastric cancer
differentially expressed proteins varied significantly (Student's samples than in normal controls. Equal loadings of LRG1 and
t-test, p-value < 0.05), as shown in Table 2. There were 10 ITIH3 amounts in these experiments were also examined
upregulated proteins and 7 downregulated proteins. Relative to (Fig. 1A–C). The validation set was further examined by
the control plasma pools, expression of 2 of the identified subjecting individual Con A-captured plasma to immunoblotting
proteins was increased or decreased by more than 2-fold in the with the LRG1 and ITIH3 antibodies. The LRG1 expression levels
gastric cancer plasma pools, and expression of 12 proteins was in early- (stage I/II) and late-stage (stage III/IV) gastric cancers
increased or decreased by 0.30- to 1.20-fold. Three proteins were were increased 1.30- (average densitometry value: 87,759 versus
undetectable in the gastric cancer plasma pools. The vast 67,715; p > 0.05) and 1.59-fold (average densitometry value:
majority of proteins are tumor-associated proteins (88.2%), 107,411 versus 67,715; p < 0.05), respectively, compared with
including LRG1, ITIH3, hemopexin (HPX), plasma protease C1 normal controls as shown in Fig. 1D. Additionally, the levels of
inhibitor (SERPING1), alpha-1B-glycoprotein (A1BG), comple- ITIH3 in early- and late-stage gastric cancer samples were higher
ment factor B (CFB), fibronectin (FN1), inter-alpha-trypsin than normal controls by 1.80- (average densitometry value:
inhibitor heavy chain H1 (ITIH1), Ig gamma-2 chain c region 101,740 versus 56,345; p < 0.05) and 1.73-fold (average densitom-
(IGHG2), Ig mu chain c region (IGHM), clusterin (CLU), apolipo- etry value: 97,723 versus 56,345; p < 0.05), respectively, as shown
protein D (APOD), plasminogen (PLG), complement C5 (C5), and in Fig. 1D. The box plot generated by ANOVA statistical analysis
complement C1q subcomponent subunit B (C1QB). These pro- revealed that the difference in LRG1 (p = 0.0004) and ITIH3
teins have been found to be differentially expressed in cancers (p < 0.0001) expression levels was significant among normal
such as ovarian [41,42], lung [43–45], liver [46], pancreatic [47,48], controls and early- and late-stage gastric cancer patients
bladder [49], biliary tract [50], breast [51–55], nasopharyngeal [56], (Fig. 1E). These experiments were performed in duplicate.
renal [57], papillary thyroid [58], colon [59], prostate [60,61], and The difference in the levels of Con A-bound LRG1 and ITIH3
cutaneous malignant melanoma [62,63]. The summary of between early- and late-stage gastric cancer patients was not
the category classification of the differentially expressed Con statistically significant. The average densitometry signals of
A-bound glycoproteins according to cellular component, molec- LRG1 and ITIH3 expression for individual Con A-captured
ular function, biological process, and their predicted functional plasma were calculated, and an asterisk (*) indicates a
partners is shown in Supplementary Table 2. This category p-value < 0.05 (Fig. 1). A ROC curve was plotted based on
classification provided useful information regarding protein these results to calculate the sensitivity and specificity of Con
ontology in response to gastric cancer. A-bound LRG1 and ITIH3 in gastric cancer detection. As shown
in Fig. 1F, the AUC value of the ROC curve was found to be 0.65
3.3. Validation of differential expression by Western blot (95% CI: 0.55 - 0.75) at an average densitometry cut-off of 75,000,
giving Con A-bound LRG1 a sensitivity and specificity of 56.7%
To validate the LC–MS/MS data, we analyzed the levels of Con and 66.7%, respectively. Furthermore, at an average densitom-
A-bound LRG1 and ITIH3 by Western blot analysis in the etry cut-off of 75,000, the AUC value of the ROC curve for Con
discovery and validation sets (Fig. 1 and Supplementary Fig. 2). A-bound ITIH3 was found to be 0.75 (95% CI: 0.66–0.84), yielding
These are potential tumor-specific proteins. Plasma-derived Con a sensitivity and specificity of 73.3% and 76.7%, respectively
A-bound LRG1 and ITIH3 were identified as being upregulated by (Fig. 1F). The sensitivity and specificity were estimated at 95%
2-fold or more in the gastric cancer samples compared with confidence for gastric cancer detection.
204 J O U RN A L OF P ROTE O M IC S 8 3 ( 2 01 3 ) 1 9 7 –21 3

A LRG1 B C
*
kDa 1 2 3 4 40000
kDa 1 2 3 4
70
55 30000 250

Intensity
20000 130
Albumin-and IgG-depleted plasma 100
Normal control average: 11,866 10000
Gastric cancer average: 14,248 70
Con A-captured plasma 55
0
Normal control average: 24,249 1 2 3 4
Gastric cancer average: 33,835

35

ITIH3 *
35000 25
kDa 1 2 3 4 *
30000

25000
130

Intensity
20000 15
100
15000
Albumin-and IgG-depleted plasma
Normal control average: 21,003 10000
Gastric cancer average: 28,315 5000
Con A-captured plasma
Normal control average: 14,022 0
Gastric cancer average: 21,881
1 2 3 4

D E

F
J O U RN A L OF P ROT EO M IC S 8 3 ( 2 01 3 ) 1 9 7 –2 13 205

3.4. Networks and functional pathway analysis Additionally, the b7 ion was shifted by the 43.0548 Da, the
mass of a trimethyl group. We also observed a novel FAD
Supplementary Table 3 lists the top 5 canonical pathways that modification in ITIH3. The modified peptide was identified
involve the 17 differentially expressed Con A-bound glycopro- from the unmodified y16 ion and modified b2 and b3 ions with a
tein biomarkers in plasma from patients with gastric cancer. mass shift of 783.1415 Da. The sequence of the peptide in the
The analysis shows the canonical pathways of acute phase spectrum showed a peptide fragmentation pattern that demon-
response signaling, the complement system, LXR/RXR activa- strated the b and y ion series. These data show that the modified
tion, hematopoiesis from pluripotent stem cells, and primary peptide 334-EFHLVQATPENLQEARTFVK-352 contained a FAD
immunodeficiency signaling. The interactive relationship be- covalently bound at histidine- 335, which was confirmed by
tween the proteins in the top canonical pathway is shown in the b and y ion series as shown in Fig. 3C and Table 3.
Fig. 2.
3.6. Molecular modeling
3.5. A novel type of PTM identified in ITIH3
When searched against the UniProt database, alignment of
LRG1 and ITIH3 are known glycoproteins. There are 4N- and 1 the human plasma ITIH3 sequence showed similarity to the
O-linked glycosylation sites in LRG1 at positions asparagine-79, known sequences of ITIH3 from other species, including
asparagine-186, asparagine-269, asparagine-325 [65,66], and Rattus norvegicus (Q63416), Mus musculus (Q61704), Mesocricetus
threonine-37, respectively, as indicated by the UniProt database. auratus (P97280), Pongo abelii (Q5RB37), Bos taurus (P56652), and
No novel PTMs to LRG1 were found in this study. The three Sus scrofa (F1SH94). In the alignment shown in Supplementary
types of PTMs found on ITIH3 were N-linked glycosylation Fig. 3, two domains, the vault protein inter-alpha-trypsin
(asparagine-91 and asparagine-580) [65], 1-(chondroitin (VIT) domain and a von Willebrand factor (vWF) type A
4-sulfate)-ester modification (aspartic acid-651) [67], and phos- (VWFA) domain, are conserved among all ITIH3 members,
phorylation (threonine-705 and threonine-727) [Human Protein and the three novel modifications documented in the present
Reference database, http://www.hprd.org/], and they are shown study were identified inside these domains. The glycation
in Supplementary Fig. 3. Three novel types of PTMs on ITIH3 residue (HexHexNAc) was found in the VIT domain (amino
were identified by manual examination of the modified spectra acid sequence 29 to 158), and the other two modifications (the
using our in-house PTM finder program. The identified peptides trimethyl group and the FAD) were observed in the VWFA
and the corresponding modified peptides are listed in Table 3. domain (amino acid sequence 284 to 467). Taking these
The peptide 33-RSLPEGVAHNGIEVYSTK-49 was used to identify identified modification sites into consideration, we performed
ITIH3 and was found to be glycated at asparagine-41, and molecular modeling of human plasma ITIH3 in the SWISS-
identification of the peptide moiety of the glycopeptide was MODEL Repository. Two templates, bacterial micronemal pro-
based on the presence of b- and y-series ions derived from tein 2 (PDB ID: 2xggB; modeled residue range 280 to 428; 22.8%
the peptide. The glycation residue is a hexose-N-acetyl- identity; E value 4.6E-13) and human anthrax toxin receptor 2
hexosamine (HexHexNAc), and this residue was confirmed by (PDB ID: 1shuX; modeled residue range 282 to 468; 19.1%
an unmodified y7 ion followed by a modified b9 ion, corre- identity; E value 4E-18), were better matched. We selected
sponding to a mass increase of 365.1322 Da (Fig. 3A). In addition human anthrax toxin receptor 2 (the VWFA domain of human
to glycation, we were able to identify a series of other novel capillary morphogenesis protein 2) as a template to model the
modifications by manual examination. Two other modifica- three-dimensional (3-D) structure of the VWFA domain in ITIH3
tions in ITIH3 were found and include trimethylation at aspartic using PyMol. The PTM sites are shown on Fig. 4; the aspartic
acid-290 and flavin adenine dinucleotide (FAD) covalently bound acid-290 trimethylation site was shown in red and the
at histidine-335. Trimethylation of aspartic acid-290 in the histidine-335 FAD site is shown in blue. However, the SWISS-
modified peptide 284-NVAFVITDISGSMAGR-298 was shown in MODEL did not predict the asparagine-41, most likely due to its
Fig. 3B. The spacing of the b and y ions was used to decipher the low identity or reduced conservation. The conserved amino
sequence of the modified peptide. The novel trimethylation site acid residues of a metal ion-dependent adhesion site (MIDAS
was discovered by detection of the unmodified b6 ion. motif; 290-DXSXS…T…D-313) are labeled (Fig. 4).

Fig. 1 – Validation of Con A-bound LRG1 and ITIH3 expression levels by Western blot analysis. A representative Western blot of the
albumin- and IgG-depleted samples and the Con A-captured plasma pools for the discovery set (A–C). The average densitometry
values were calculated from the validation set and used to construct box plots from the duplicate data (D–E). A total of 50 μg of
plasma proteins was resolved by SDS-PAGE and detected using anti-LRG1 and ITIH3 antibodies and Western blot analysis
(A and D). In the discovery set: lanes 1–2, albumin- and IgG-depleted plasma protein; lanes 3–4, Con A-captured plasma protein;
lanes 1 and 3, plasma pools from 30 normal controls; lanes 2 and 4, plasma pools from 30 gastric cancer patients. A duplicate gel
was stained with CBB as a loading control (C). In the validation set: lane 1, individual plasma sample selected from a set of 60 controls;
lane 2, individual plasma sample selected from 33 early-stage (stage I/II) gastric cancers; lane 3, individual plasma sample selected
from 27 late-stage (stage III/IV) gastric cancers. The average densitometry reading of the protein expression level for each
glycoprotein biomarker was calculated. Significant differences in expression levels (indicated with an asterisk *, p-value < 0.05)
were found between normal controls and gastric cancers. The ROC curve was generated using the average densitometry readings
of Con A-bound LRG1 and ITIH3 for all individual plasma samples (F).
206 J O U RN A L OF P ROTE O M IC S 8 3 ( 2 01 3 ) 1 9 7 –21 3

Fig. 2 – The top 1 canonical pathway was assigned by the Ingenuity Pathway Analysis for differentially expressed Con A-bound
glycoproteins in plasma with gastric cancer. The red and green colors in each shape indicate the up- or down-regulation of
pathway-associated protein expression when gastric cancer samples were compared with normal control samples. The proteins
in a gray color are not identified as differentially expressed in the list but are associated with the possible computationally
generated networks based on the Ingenuity database. The node (protein) and edge (protein relationship) symbols are described in
the bottom part of the figure. Each line and arrow between proteins represents known functional and physical interactions, with
solid lines indicating direct relationships (the two molecules have physical contact) and dashed lines representing indirect
or hypothetical interactions (does not require physical contact). The node shapes denote enzymes ( ), peptidases ( ), translation
regulator ( ), transmembrane receptor ( ), cytokines (□), growth factor (□), transporter ( ), Complex / Group (Ⓞ) and other
(○).

tissue polypeptide antigen (TPA; cytokeratins 8, 18, and 19),


4. Discussion tissue polypeptide-specific antigen (TPS; cytokeratin 18)], and
the free beta-subunit of HCG. The early detection rate of
The most common serum-based tumor markers for gastric tumor markers in gastric cancer diagnosis is approximately
cancer in clinical use include CEA, CA 19-9, CA 72-4, 15 to 35% [68]; the majority of these markers are glycopro-
cytokeratins [CYFRA 21-1 (soluble cytokeratin 19 fragment), teins. Therefore, there is an urgent need for early detection
J O U RN A L OF P ROT EO M IC S 8 3 ( 2 01 3 ) 1 9 7 –2 13 207

biomarkers to screen for gastric cancer and to enhance the and physiological function of LRG1 are unknown. Increment
survival rate. Previous reports on the glycoprotein biomarkers levels of LRG1 in the serum and/or plasma have been reported
discovered in gastric cancer plasma and/or serum include in various diseases, including gastric cancer [76], ovarian cancer
mucin-1 (MUC1), mucin-5AC (MUC5AC), IgG, haptoglobin (HP), [77], biliary tract cancer [50], pancreatic cancer [47], lung cancer
TF, alpha-1-acid glycoprotein (AGP), A1BG, vitamin D binding [78], uterine leiomyomas [79], microbial infections [80], rheu-
protein (GC), alpha-1-antitrypsin (SERPINA1), antithrombin matoid arthritis [81], heart failure [82], and graft-versus-host
(SERPINC1), angiotensin (AGT), albumin (ALB), CFB, kininogen-1 disease [83]. LRG1 has also been detected as a urinary biomarker
(KNG1), serpin peptidase inhibitor, Clade A (SERPINA3), IGHM, in lung cancer [84] and bladder cancer [85]. The expression level
LRG1, C4A variant protein (C4A), Zn-alpha-2-glycoprotein of LRG1 in the serum was increased in gastric cancer samples
(AZGP1), CLU, alpha-2-HS-glycoprotein (AHSG), ITIH2, comple- compared with normal controls, and the levels increase in
ment factor H (CFH), ceruloplasmin (CP), interalpha-trypsin relation to the increase in cancer progression stage (stage I,
inhibitor HCRP, and SERPING1 [69–72]. Nine of these proteins 1.2-fold; stage II, 1.6-fold; stage III, 1.7-fold; stage IV, 1.9-fold)
have been previously identified as Con A-bound glycoproteins [76]. Upregulated expression of LRG1 was identified in other
and include ITIH2, CFH, CP, ITIH4, SERPING1, C4A, A1BG, cancer versus normal controls by proteomic tools, including a
SERPINA3, and HP. These glycoproteins were enriched from 1.20 to 1.50-fold increase in ovarian cancer (2-D-DIGE) [77], a
depleted serum using the Pierce Con A Glycoprotein Kit and 4.49-fold increase in biliary tract cancer (2-DE) [50], a 6.0-fold
were identified by LC-MS/MS as 12 protein bands labeled in gel increase in lung cancer (2-D-DIGE) [78], and a 2.05 to 2.84-fold
[72]. Here, we used the Con A-captured plasma samples as increase in pancreatic cancer (2-D-DIGE) [47]. Similar to our study,
starting materials; these samples were purified directly from the trend of upregulated expression of plasma Con A-bound LRG1
plasma by Con A affinity chromatography and identified by 1-D in gastric cancer samples versus normal controls was consistent
SDS-PAGE and nano-LC–MS/MS. In the present study, a total of in the study by Kim et al. (Fig. 1) [76]. LRG1 contains 8 repetitive
82 proteins were identified, and 17 proteins were differentially consensus sequences known as the leucine-rich repeat (LRR)
expressed at a significant level. In this study, the 17 domain, and its primary function may be to provide a curved
differentially expressed proteins identified in triplicate from solenoid structural framework that is particularly suitable for
the 10 gel bands from 1-D SDS-PAGE coupled to LC-MS/MS protein–protein interactions [86]. LRG1 binds cytochrome c,
exhibited an average RSD of 16.32% (ranges from 1.87% to which may play a role in cell survival [80]. Shirai et al. found
52.07%) for the spectral counts (Table 2). Piersma et al. suggested that LRG1 was a secretory type 1 acute-phase protein, and its
an average RSD (ranges from 23.4% to 26.1%) for the spectral expression was induced by IL-6, IL-1β, and TNFα [87].
counts for proteins detected in triplicate experiments from 10 ITIH3 is one of the five heavy chains (ITIH1-5) belonging to
gel bands from 1-D SDS-PAGE coupled to LC–MS/MS [18]. Gautier the family of inter-alpha-trypsin inhibitors (ITIs). The ITI family
et al. indicated a RSD (median of 7.19%, maximum of 145.40%) in is built from one light chain, bikunin, and a variable set of heavy
label-free quantification after 3 LC-MS/MS analyses of the 12 gel chains. ITIs act as carriers of hyaluronan in the serum or as
bands from 1-D SDS-PAGE shotgun analysis [73]. These studies binding proteins between hyaluronan and other matrix pro-
used protein sample fractionation by SDS-PAGE followed by LC– teins to stabilize the extracellular matrix (ECM) and prevent
MS/MS analysis and yielded similar results. These Con A-bound tumor metastasis; they are also involved in plasma protease
proteins revealed reactivity with the α-D-mannose/ α-D- inhibition [88]. Paris et al. have shown that overexpression of
glucosyl-specific Con A lectin and reduced the complexity of the ITI family chains causes inhibition of tumor growth and
the plasma sample. Of the identified Con A-bound proteins, metastatic spreading [89]. ITIH3 contains the two domains
78.0% (64/82) were N-linked glycoproteins as annotated by the conserved in all known ITIHs, the VIT and VWFA domains
UniProt database (data not shown). This result was confirmed [89,90]. The function of the VIT domain in ITIH3 is unknown.
by the data from protein samples comparing CBB staining, PAS The VWFA domain has the capacity to bind integrins, collagens,
staining, and Con A blotting (Fig. 1 and Supplementary Fig. 1). proteoglycans, and heparin and mediates adhesion via the
After N-deglycosylation, 58.8% (10/17) of glycoproteins were still metal ion-dependent adhesion site [91,92]. ITIH3 has also been
detected (Table 2). PNGase F (N-glycosidase F) is an amidase that shown to be upregulated in the serum and/or plasma of lung
cleaves the bond between the innermost GlcNAc and all cancer and gastric cancer samples versus normal controls
asparagine-linked high mannose, hybrid, and complex oligo- [44,90]. Chong et al. found that the plasma ITIH3 expression
saccharides unless the core contains an α(1 → 3)-fucose from level in gastric cancer samples was 1.9-fold higher when
the N-linked glycoprotein [74]. It is likely that the incubation time compared with normal controls by Western blot analysis [90].
with PNGase F must be increased as steric constraints in N-glycan Furthermore, the expression level of Con A-bound ITIH3 in
may prevent enzyme activity or additional exoglycosidases may gastric cancer samples versus normal controls in our study was
be needed to remove other N- and O-glycans. Albumin and IgG, similar to that observed in a study by Chong et al. (Fig. 1) [90].
the most abundant proteins in human serum, must be removed However, the sensitivity (96.0% versus 73.7%) and specificity
by immunodepletion in the study of proteomics. Koutroukides (66.0% versus 76.7%) of ITIH3 versus Con A-bound ITIH3 in
et al. indicated that depleted serum had been co-depleted of 147 gastric cancer detection calculated from the ROC curve were
non-target serum proteins using an immunoaffinity kit [75]. inconsistent [90]. Our preliminary experiment for PTM peptide
This depleted serum is known to provide new insights into quantification compared gastric cancer samples with normal
clinical proteomics. The use of lectin-captured plasma and/or controls. The relative modified peptide ratios of the three novel
serum could prevent the loss of important information. modifications of ITIH3 were identified and included HexHexNAc
Two proteins with a potential role in carcinogenesis were (↓ 1.90-fold, p = 0.0071), trimethylation (↑ 1.78-fold, p = 0.00001),
identified, Con A-bound LRG1 and ITIH3 (Table 2). The isoforms and FAD (↓ 2.9-fold, p = 0.02). There was a newly found
208 J O U RN A L OF P ROTE O M IC S 8 3 ( 2 01 3 ) 1 9 7 –21 3

Table 3 – The identification of novel types of post-translational modifications in ITIH3 using mass spectrometry.
Protein a PTM b Site Modified peptides c Position d Obs. Calc. dM (Obs.– dM
m/z e m/z f calc.) g (ppm) h

ITIH3 Hexose-N-acetyl-hexosamine Asparagine-41 RSLPEGVA*NGIEVYSTK 33–49 1093.058 1093.046 0.013 11.497


(HexHexNAc)
Trimethylation Aspartic NVAFVI*DISGSMAGR 284–298 532.611 532.615 -0.004 -7.923
acid-290
Flavin adenine dinucleotide Histidine-335 E*HLVQATPENLQEARTFVK 334–352 998.445 998.438 0.007 6.573
(FAD)
a
Name of the modified protein.
b
Type of modification.
c
Site of modified peptide is marked with an asterisk (*) and boldface.
d
Amino acid positions of the first and the last residues in accession number.
e
Obs: observed m/z of the modified peptides.
f
Cal: calculated m/z of the modified peptides.
g
dM (Obs.–calc.): mass accuracy.
h
Modified peptide mass accuracy (ppm).

HexHexNAc residue at asparagine-41, and two glycation residues carcinogenesis (Supplementary Fig. 3). In the molecular modeling
were located within the VIT domain. The ratio of N-linked of ITIH3, no suitable template structure of the VIT domain
glycosylation modification was decreased in the process of was selected because of the reduced conservation between

Fig. 3 – (A) A representative MS/MS spectrum of the peptide sequence of 33-RSLPEGVAHNGIEVYSTK-49 and the modified peptide
bearing the asparagine glycation. The glycation residue is a HexHexNAc. (B) The MS/MS spectrum of 284-NVAFVITDISGSMAGR-298
and the modified peptide bearing the aspartic acid trimethylation. (C) The MS/MS spectrum of the peptide 334-EFHLVQATPENLQEARTF
VK-352 with a FAD covalently bound at a histidine residue.
J O U RN A L OF P ROT EO M IC S 8 3 ( 2 01 3 ) 1 9 7 –2 13 209

Fig. 4 – Novel PTM patterns of Con A-bound ITIH3 VWFA domain and the 3-D structure developed with PyMol. The highlighted amino
acid residues include D290 (aspartic acid- 290; red), S292 (serine- 292; red), S294 (serine- 294; red), T303 (threonine- 303; green),
D313 (aspartic acid-313; green) and H335 (histidine-335; blue). The conserved amino acids of the MIDAS motif are 290-DXSXS…T…D-313.
The PTM sites include trimethylation at D290 and FAD at H335. The A and B view with a different orientation.

species. In the alignment shown in Supplementary Fig. 3, 4 interaction of RARalpha with its modulators by site-specific
of the 8 amino acid residues at modification sites (N-linked hydrophobicity [100]. Whether trimethylation of the VWFA
glycosylation at asparagine- 91, trimethylation at aspartic domain in ITIH3 enhances its binding capacity with ITI, ITIH1,
acid- 290, 1-(chondroitin 4-sulfate)-ester modification at ITIH2, TGFB, and HNF4A requires further study (Supplementary
aspartic acid-651, and phosphorylation at threonine-727) are Table 2). FAD is an essential coenzyme in cellular oxidation-
conserved in all ITIH3 members. These highly conserved reduction reactions. Werner et al. hypothesized that riboflavin
regions included asparagine-91 in 74-AKEVSFDVELPKTAFIT deficiency leads to hepatocarcinoma cell stress by rapidly
NFTLTIDGV-99, aspartic acid-290 in 287-FVIDISGSM-295, aspartic inducing cell cycle arrest [101]. An animal model by Pinto et al.
acid-651 in 644-PYYYVDGDPHFIIQ-657, and threonine-727 in supported the hypothesis that riboflavin deprivation causes a
725
-EVTTE-729. The heavy chains of ITI are linked to bikunin disturbance in the formation of FAD and covalently bound flavins
through chondroitin 4-sulfate esterification to the alpha-carboxyl in hepatomas [102]. Koturbash et al. indicated that FAD may be a
of the C-terminal at aspartic acid-651 after propeptide cleavage significant regulator of the cellular epigenome [103]. The relative
[93]. The VWFA domain adopts a classic alpha/beta secondary modified peptide ratio of FAD in ITIH3 may be affected by
structure (Rossmann fold; order beta-alpha-beta-alpha-beta) and metabolic changes in the carcinogenesis process. Our MS/MS
contains a MIDAS motif for binding protein ligands [94]. Bajt and spectra findings provide the first evidence of trimethylation and
Loftus demonstrated that mutation of the DXSXS sequence FAD modification in the VWFA domain (Fig. 3). In this study, the
abolished ligand binding [95]. It is interesting that aspartic PTM analysis has revealed a Con A-bound biomarker in addition
acid-290 in 290-DISGSMAGRKLEQTKEALLRILED-313 was modified to glycosylation in gastric cancer samples versus normal controls.
by trimethylation in the present study. Many previous studies Some protein PTMs associated with gastric cancer carcinogenesis
have indicated that the trimethylation of histones is a tumor- have been studied and include glycosylation of mucin [70] and
associated PTM [96–99]. Zhang et al. found that significant IgG [71], phosphorylation of H. pylori CagA [104], citrullination of
alterations in the trimethylation of histone H3 at lysine 27 peptidylarginine deiminase type 4 (PAD4) [105], methylglyoxal
(H3K27me3) in gastric cancer tissues were involved in the modification of heat-shock protein 27 [106], polyubiquitylation of
pathogenesis of gastric cancer [98]. Li et al. indicated that the tripartite motif 31 (TRIM31) [107], trimethylation of histone H3
transcription factor CBX7 could initiate trimethylation of histone [98,99], and others.
H3 at lysine 9 (H3K9me3) at the p16 promoter to cause epigenetic Consistent with the crucial role of the inflammatory
silencing of the p16 gene in gastric cancer [99]. Huq et al. found response in carcinogenesis [108], the upregulation of plas-
trimethylation at lysine-347 in retinoic acid receptor-alpha ma Con A-bound LRG1 and ITIH3 participated in the
(RARalpha) protein; this trimethylation enhanced the molecular inflammatory response, and both are mediated by interleukin-
210 J O U RN A L OF P ROTE O M IC S 8 3 ( 2 01 3 ) 1 9 7 –21 3

6 (IL-6) signaling [72,87]. Ikeguchi et al. reported that high serum and grant NSC101-2622-B-038-001-CC3 from the National
IL-6 levels may be associated with gastric cancer progression Science Council in Taiwan.
[109]. Unfortunately, IL-6 was undetectable, which indicates
that IL-6 may not react with Con A lectin in this study. In Fig. 2
and Supplementary Table 3, the expression levels of proteins REFERENCES
participating in acute phase response signaling are shown,
including SERPING1, HPX, ITIH3, FN1, and CFB. These data are
consistent with the study by Bones et al., apart from their [1] Tsugane S, Sasazuki S. Diet and the risk of gastric cancer: review
of epidemiological evidence. Gastric Cancer 2007;10:75–83.
additional findings of C5 and PLG [72].
[2] Parkin DM. The global health burden of infection-associated
The ROC curve of Con A-bound LRG1 and ITIH3 in early- and
cancers in the year 2002. Int J Cancer 2006;118:3030–44.
late-stage gastric cancer was estimated (data not shown). The [3] Stewart BWKP, editor. World Cancer Report. Lyon: IARC Press;
cut-off for Con A-bound LRG1 and ITIH3 in early- or late-stage 2003.
gastric cancer was set at 75,000. Con A-bound LRG1 sensitivity [4] Department of Health. Cancer registry annual report, Taiwan;
and specificity in detecting early-stage gastric cancer were 51.5% 2004–2008.
and 66.7%, respectively; the AUC value of the ROC curve was 0.57 [5] Crew KD, Neugut AI. Epidemiology of gastric cancer. World J
Gastroenterol 2006;12:354–62.
(95% CI: 0.44–0.71). Con A-bound LRG1 sensitivity and specificity
[6] Kamangar F, Dores GM, Anderson WF. Patterns of cancer
in detecting late-stage gastric cancer were 63.0% and 66.7%,
incidence, mortality, and prevalence across five continents:
respectively; the AUC value of the ROC curve was 0.73 (95% CI: defining priorities to reduce cancer disparities in different
0.61–0.85). Con A-bound ITIH3 sensitivity and specificity in geographic regions of the world. J Clin Oncol 2006;24:2137–50.
detecting early-stage gastric cancer were 57.6% and 76.7%, [7] Kim DH, Oh SJ, Oh CA, Choi MG, Noh JH, Sohn TS, et al. The
respectively; the AUC value of the ROC curve was 0.67 (95% CI: relationships between perioperative CEA, CA 19-9, and CA
0.56–0.81). Con A-bound ITIH3 sensitivity and specificity in 72-4 and recurrence in gastric cancer patients after curative
radical gastrectomy. J Surg Oncol 2011;104:585–91.
detecting late-stage gastric cancer were 92.6% and 76.7%,
[8] Goodlett DR, Keller A, Watts JD, Newitt R, Yi EC, Purvine S,
respectively; the AUC value of the ROC curve was 0.82 (95% CI: et al. Differential stable isotope labeling of peptides for
0.73–0.91). Such small differences (<2-fold) in the protein levels of quantitation and de novo sequence derivation. Rapid
Con A-bound LRG1 and ITIH3 may not be of practical use as a Commun Mass Spectrom 2001;15:1214–21.
clinical biomarker for the early detection of gastric cancer (Fig. 1). [9] Heller M, Mattou H, Menzel C, Yao X. Trypsin catalyzed
In conclusion, we have performed a comparative glyco- 16O-to-18O exchange for comparative proteomics: tandem
mass spectrometry comparison using MALDI-TOF,
proteomic analysis to discover and investigate specific, differ-
ESI-QTOF, and ESI-ion trap mass spectrometers. J Am Soc
entially expressed Con A-bound biomarkers in plasma from
Mass Spectrom 2003;14:704–18.
gastric cancer samples and normal controls. Plasma Con [10] Sechi S. A method to identify and simultaneously determine
A-bound LRG1 and ITIH3 were validated as biomarkers by the relative quantities of proteins isolated by gel
Western blot analysis against a discovery set by pooled plasma electrophoresis. Rapid Commun Mass Spectrom 2002;16:
and a validation set of individual plasma. We further identified 1416–24.
novel types of PTMs by using an in-house program to analyze [11] Kuyama H, Watanabe M, Toda C, Ando E, Tanaka K,
Nishimura O. An approach to quantitative proteome
the mass spectra of both candidate biomarkers. The use of
analysis by labeling tryptophan residues. Rapid Commun
molecular modeling and multiple sequence alignments will
Mass Spectrom 2003;17:1642–50.
assist in the discussion of the biological consequences of novel [12] Schmidt A, Kellermann J, Lottspeich F. A novel strategy for
PTMs of Con A-bound ITIH3. IPA also assisted in the pathway quantitative proteomics using isotope-coded protein labels.
analysis of 17 differentially expressed proteins. It supports the Proteomics 2005;5:4–15.
important biological roles of acute phase response signals in [13] Gygi SP, Rist B, Gerber SA, Turecek F, Gelb MH, Aebersold R.
gastric cancer and reveals differential physiological responses Quantitative analysis of complex protein mixtures using
isotope-coded affinity tags. Nat Biotechnol 1999;17:994–9.
related to gastric cancer carcinogenesis. This strategy may serve
[14] Ong SE, Blagoev B, Kratchmarova I, Kristensen DB, Steen H,
as the basis for a glycoproteomic platform for additional cancer Pandey A, et al. Stable isotope labeling by amino acids in cell
diagnostic and therapeutic applications. culture, SILAC, as a simple and accurate approach to
Supplementary data to this article can be found online at expression proteomics. Mol Cell Proteomics 2002;1:376–86.
http://dx.doi.org/10.1016/j.jprot.2013.03.007. [15] Ross PL, Huang YN, Marchese JN, Williamson B, Parker K,
Hattan S, et al. Multiplexed protein quantitation in
Saccharomyces cerevisiae using amine-reactive isobaric
tagging reagents. Mol Cell Proteomics 2004;3:1154–69.
Competing interests [16] Schulze WX, Usadel B. Quantitation in mass-
spectrometry-based proteomics. Annu Rev Plant Biol 2010;61:
491–516.
The authors declare that there are no competing interests.
[17] Old WM, Meyer-Arendt K, Aveline-Wolf L, Pierce KG,
Mendoza A, Sevinsky JR, et al. Comparison of label-free
methods for quantifying human proteins by shotgun
Acknowledgments proteomics. Mol Cell Proteomics 2005;4:1487–502.
[18] Piersma SR, Fiedler U, Span S, Lingnau A, Pham TV,
Hoffmann S, et al. Workflow comparison for label-free,
We thank Professor Yuan-Chii Lee for the discussions quantitative secretome proteomics for cancer biomarker
regarding the IPA analysis. This study was supported by grant discovery: method evaluation, differential analysis, and
99CM-TMU-12 from the Foundation of Chi Mei Medical Center verification in serum. J Proteome Res 2010;9:1913–22.
J O U RN A L OF P ROT EO M IC S 8 3 ( 2 01 3 ) 1 9 7 –2 13 211

[19] Andersen JS, Wilkinson CJ, Mayor T, Mortensen P, Nigg EA, [39] Guex N, Peitsch MC. SWISS-MODEL and the
Mann M. Proteomic characterization of the human centrosome Swiss-PdbViewer: an environment for comparative protein
by protein correlation profiling. Nature 2003;426:570–4. modeling. Electrophoresis 1997;18:2714–23.
[20] Matta A, Ralhan R, DeSouza LV, Siu KW. Mass [40] Balasenthil S, Chen N, Lott ST, Chen J, Carter J, Grizzle WE, et al.
spectrometry-based clinical proteomics: head-and-neck A migration signature and plasma biomarker panel for
cancer biomarkers and drug-targets discovery. Mass pancreatic adenocarcinoma. Cancer Prev Res (Phila) 2011;4:
Spectrom Rev 2010;29:945–61. 137–49.
[21] Karsdal MA, Henriksen K, Leeming DJ, Woodworth T, [41] Andersen JD, Boylan KL, Jemmerson R, Geller MA, Misemer
Vassiliadis E, Bay-Jensen AC. Novel combinations of B, Harrington KM, et al. Leucine-rich alpha-2-glycoprotein-1
Post-Translational Modification (PTM) neo-epitopes provide is upregulated in sera and tumors of ovarian cancer
tissue-specific biochemical markers—are they the cause or patients. J Ovarian Res 2010;3:21.
the consequence of the disease? Clin Biochem 2010;43: [42] He P, He HZ, Dai J, Wang Y, Sheng QH, Zhou LP, et al. The human
793–804. plasma proteome: analysis of Chinese serum using shotgun
[22] Anderson NL. The clinical plasma proteome: a survey of strategy. Proteomics 2005;5:3442–53.
clinical assays for proteins in plasma and serum. Clin Chem [43] Li Y, Zhang Y, Qiu F, Qiu Z. Proteomic identification of
2010;56:177–85. exosomal LRG1: a potential urinary biomarker for detecting
[23] Choudhary C, Kumar C, Gnad F, Nielsen ML, Rehman M, NSCLC. Electrophoresis 2011;32:1976–83.
Walther TC, et al. Lysine acetylation targets protein complexes [44] Heo SH, Lee SJ, Ryoo HM, Park JY, Cho JY. Identification of
and co-regulates major cellular functions. Science 2009;325: putative serum glycoprotein biomarkers for human lung
834–40. adenocarcinoma by multilectin affinity chromatography
[24] Kovacech B, Novak M. Tau truncation is a productive and LC–MS/MS. Proteomics 2007;7:4292–302.
posttranslational modification of neurofibrillary degeneration [45] Bourguignon J, Borghi H, Sesboue R, Diarra-Mehrpour M,
in Alzheimer's disease. Curr Alzheimer Res 2010;7:708–16. Bernaudin JF, Metayer J, et al. Immunohistochemical
[25] Guo HB, Johnson H, Randolph M, Nagy T, Blalock R, Pierce M. distribution of inter-alpha-trypsin inhibitor chains in
Specific posttranslational modification regulates early normal and malignant human lung tissue. J Histochem
events in mammary carcinoma formation. Proc Natl Acad Cytochem 1999;47:1625–32.
Sci U S A 2010;107:21116–21. [46] Jha RK, Ma Q, Chen S, Sha H, Ding S. Relationship of fibronectin
[26] Chen Y, Chen W, Cobb MH, Zhao Y. PTMap—a sequence and CD44v6 expression with invasive growth and metastasis of
alignment software for unrestricted, accurate, and liver cancer. Cancer Invest 2009;27:324–8.
full-spectrum identification of post-translational modification [47] Kakisaka T, Kondo T, Okano T, Fujii K, Honda K, Endo M,
sites. Proc Natl Acad Sci U S A 2009;106:761–6. et al. Plasma proteomics of pancreatic cancer patients by
[27] Krueger KE, Srivastava S. Posttranslational protein multi-dimensional liquid chromatography and
modifications: current implications for cancer detection, two-dimensional difference gel electrophoresis (2D-DIGE):
prevention, and therapeutics. Mol Cell Proteomics 2006;5: up-regulation of leucine-rich alpha-2-glycoprotein in
1799–810. pancreatic cancer. J Chromatogr B Analyt Technol Biomed Life
[28] Zhang XW, Yan XJ, Zhou ZR, Yang FF, Wu ZY, Sun HB, et al. Sci 2007;852:257–67.
Arsenic trioxide controls the fate of the PML-RARalpha [48] Zhao J, Patwa TH, Qiu W, Shedden K, Hinderer R, Misek DE,
oncoprotein by directly binding PML. Science 2010;328:240–3. et al. Glycoprotein microarrays with multi-lectin detection:
[29] Klose RJ, Yamane K, Bae Y, Zhang D, Erdjument-Bromage H, unique lectin binding patterns as a tool for classifying normal,
Tempst P, et al. The transcriptional repressor JHDM3A chronic pancreatitis and pancreatic cancer sera. J Proteome
demethylates trimethyl histone H3 lysine 9 and lysine 36. Res 2007;6:1864–74.
Nature 2006;442:312–6. [49] Linden M, Lind SB, Mayrhofer C, Segersten U, Wester K,
[30] Vucic D, Dixit VM, Wertz IE. Ubiquitylation in apoptosis: a Lyutvinskiy Y, et al. Proteomic analysis of urinary biomarker
post-translational modification at the edge of life and death. candidates for nonmuscle invasive bladder cancer. Proteomics
Nat Rev Mol Cell Biol 2011;12:439–52. 2012;12:135–44.
[31] McDonald CA, Yang JY, Marathe V, Yen TY, Macher BA. [50] Sandanayake NS, Sinclair J, Andreola F, Chapman MH, Xue A,
Combining results from lectin affinity chromatography and Webster GJ, et al. A combination of serum leucine-rich
glycocapture approaches substantially improves the coverage alpha-2-glycoprotein 1, CA19-9 and interleukin-6 differentiate
of the glycoproteome. Mol Cell Proteomics 2009;8:287–301. biliary tract cancer from benign biliary strictures. Br J Cancer
[32] Polanski M, Anderson NL. A list of candidate cancer biomarkers 2011;105:1370–8.
for targeted proteomics. Biomark Insights 2007;1:1–48. [51] Mazouni C, Baggerly K, Hawke D, Tsavachidis S, Andre F,
[33] Fung ET. A recipe for proteomics diagnostic test development: Buzdar AU, et al. Evaluation of changes in serum protein
the OVA1 test, from biomarker discovery to FDA clearance. profiles during neoadjuvant chemotherapy in HER2-positive
Clin Chem 2010;56:327–9. breast cancer using an LC–MALDI-TOF/MS procedure.
[34] OVA1™. Package Insert; 2009. Proteomics 2010;10:3525–32.
[35] Laemmli UK. Cleavage of structural proteins during the [52] Zeng Z, Hincapie M, Pitteri SJ, Hanash S, Schalkwijk J, Hogan JM,
assembly of the head of bacteriophage T4. Nature et al. A proteomics platform combining depletion, multi-lectin
1970;227:680–5. affinity chromatography (M-LAC), and isoelectric focusing to
[36] Rajkumar R, Karthikeyan K, Archunan G, Huang PH, Chen YW, study the breast cancer proteome. Anal Chem 2011;83:4845–54.
Ng WV, et al. Using mass spectrometry to detect buffalo [53] Liang S, Singh M, Dharmaraj S, Gam LH. The PCA and LDA
salivary odorant-binding protein and its post-translational analysis on the differential expression of proteins in breast
modifications. Rapid Commun Mass Spectrom 2010;24: cancer. Dis Markers 2010;29:231–42.
3248–54. [54] Hamm A, Veeck J, Bektas N, Wild PJ, Hartmann A, Heindrichs U,
[37] Arnold K, Bordoli L, Kopp J, Schwede T. The SWISS-MODEL et al. Frequent expression loss of inter-alpha-trypsin inhibitor
workspace: a web-based environment for protein structure heavy chain (ITIH) genes in multiple human solid tumors: a
homology modelling. Bioinformatics 2006;22:195–201. systematic expression analysis. BMC Cancer 2008;8:25.
[38] Schwede T, Kopp J, Guex N, Peitsch MC. SWISS-MODEL: an [55] Cho W, Jung K, Regnier FE. Sialylated Lewis × antigen
automated protein homology-modeling server. Nucleic bearing glycoproteins in human plasma. J Proteome Res
Acids Res 2003;31:3381–5. 2010;9:5960–8.
212 J O U RN A L OF P ROTE O M IC S 8 3 ( 2 01 3 ) 1 9 7 –21 3

[56] Seriramalu R, Pang WW, Jayapalan JJ, Mohamed E, of complex proteomes by one-dimensional
Abdul-Rahman PS, Bustam AZ, et al. Application of SDS-PAGE/NanoLC–MS: evaluation for the large scale analysis
champedak mannose-binding lectin in the glycoproteomic of inflammatory human endothelial cells. Mol Cell Proteomics
profiling of serum samples unmasks reduced expression of 2012;11:527–39.
alpha-2 macroglobulin and complement factor B in patients [74] Maley F, Trimble RB, Tarentino AL, Plummer Jr TH.
with nasopharyngeal carcinoma. Electrophoresis 2010;31: Characterization of glycoproteins and their associated
2388–95. oligosaccharides through the use of endoglycosidases. Anal
[57] Waalkes S, Atschekzei F, Kramer MW, Hennenlotter J, Vetter G, Biochem 1989;180:195–204.
Becker JU, et al. Fibronectin 1 mRNA expression correlates with [75] Koutroukides TA, Guest PC, Leweke FM, Bailey DM, Rahmoune
advanced disease in renal cancer. BMC Cancer 2010;10:503. H, Bahn S, et al. Characterization of the human serum
[58] Wilson C. Cancer: oncofetal fibronectin mRNA and papillary depletome by label-free shotgun proteomics. J Sep Sci 2011;34:
thyroid cancer. Nat Rev Endocrinol 2010;6:475. 1621–6.
[59] Wei PL, Kuo LJ, Huang MT, Ting WC, Ho YS, Wang W, et al. [76] Kim CW OS, Han MY, Shin YS. Protein markers for
Nicotine enhances colon cancer cell migration by induction diagnosing stomach cancer and the diagnostic kit using
of fibronectin. Ann Surg Oncol 2011;18:1782–90. them. USPTO Issued patent, US7892761. 2011.
[60] Beecken WD, Bentas W, Engels K, Glienke W, Urbschat A, [77] Andersen JD, Boylan KL, Xue FS, Anderson LB, Witthuhn BA,
Jonas D, et al. Reduced plasma levels of coagulation Markowski TW, et al. Identification of candidate biomarkers in
factors in relation to prostate cancer. Prostate 2002;53: ovarian cancer serum by depletion of highly abundant proteins
160–7. and differential in-gel electrophoresis. Electrophoresis 2010;31:
[61] Seal US, Doe RP, Byar DP, Corle DK. Response of plasma 599–610.
fibrinogen and plasminogen to hormone treatment and the [78] Okano T, Kondo T, Kakisaka T, Fujii K, Yamada M, Kato H,
relation of pretreatment values to mortality in patients with et al. Plasma proteomics of lung cancer by a linkage of
prostatic cancer. Cancer 1976;38:1108–17. multi-dimensional liquid chromatography and
[62] Miranda E, Vizoso F, Martin A, Quintela I, Corte MD, Segui two-dimensional difference gel electrophoresis. Proteomics
ME, et al. Apolipoprotein D expression in cutaneous 2006;6:3938–48.
malignant melanoma. J Surg Oncol 2003;83:99–105. [79] Lin CP, Chen YW, Liu WH, Chou HC, Chang YP, Lin ST, et al.
[63] Luo Y, Robinson S, Fujita J, Siconolfi L, Magidson J, Edwards Proteomic identification of plasma biomarkers in uterine
CK, et al. Transcriptome profiling of whole blood cells leiomyoma. Mol BioSyst 2012;8:1136–45.
identifies PLEK2 and C1QB in human melanoma. PLoS One [80] Weivoda S, Andersen JD, Skogen A, Schlievert PM, Fontana
2011;6:e20971. D, Schacker T, et al. ELISA for human serum leucine-rich
[64] Josic D, Brown MK, Huang F, Lim YP, Rucevic M, Clifton JG, alpha-2-glycoprotein-1 employing cytochrome c as the
et al. Proteomic characterization of inter-alpha inhibitor capturing ligand. J Immunol Methods 2008;336:22–9.
proteins from human plasma. Proteomics 2006;6:2874–85. [81] Serada S, Fujimoto M, Ogata A, Terabe F, Hirano T, Iijima
[65] Liu T, Qian WJ, Gritsenko MA, Camp 2nd DG, Monroe ME, Moore H, et al. iTRAQ-based proteomic identification of
RJ, et al. Human plasma N-glycoproteome analysis by leucine-rich alpha-2 glycoprotein as a novel inflammatory
immunoaffinity subtraction, hydrazide chemistry, and mass biomarker in autoimmune diseases. Ann Rheum Dis 2010;69:
spectrometry. J Proteome Res 2005;4:2070–80. 770–4.
[66] Nilsson J, Ruetschi U, Halim A, Hesse C, Carlsohn E, [82] Watson CJ, Ledwidge MT, Phelan D, Collier P, Byrne JC, Dunn
Brinkmalm G, et al. Enrichment of glycopeptides for glycan MJ, et al. Proteomic analysis of coronary sinus serum reveals
structure and attachment site identification. Nat Methods leucine-rich alpha2-glycoprotein as a novel biomarker of
2009;6:809–11. ventricular dysfunction and heart failure. Circ Heart Fail
[67] Enghild JJ, Salvesen G, Hefta SA, Thogersen IB, Rutherfurd S, 2011;4:188–97.
Pizzo SV. Chondroitin 4-sulfate covalently cross-links the [83] Wang H, Clouthier SG, Galchev V, Misek DE, Duffner U, Min
chains of the human blood protein pre-alpha-inhibitor. J Biol CK, et al. Intact-protein-based high-resolution
Chem 1991;266:747–51. three-dimensional quantitative analysis system for proteome
[68] Sturgeon CM, Duffy MJ, Hofmann BR, Lamerz R, Fritsche HA, profiling of biological fluids. Mol Cell Proteomics 2005;4:
Gaarenstroom K, et al. National Academy of Clinical 618–25.
Biochemistry Laboratory Medicine Practice Guidelines for [84] Dai L, Liu Y, He J, Flack CG, Talsma CE, Crowley JG, et al.
use of tumor markers in liver, bladder, cervical, and gastric Differential profiling studies of N-linked glycoproteins in
cancers. Clin Chem 2010;56:e1-48. glioblastoma cancer stem cells upon treatment with
[69] Bones J, Mittermayr S, O'Donoghue N, Guttman A, Rudd PM. gamma-secretase inhibitor. Proteomics 2011;11:4021–8.
Ultra performance liquid chromatographic profiling of serum [85] Linden M, Lind SB, Mayrhofer C, Segersten U, Wester K,
N-glycans for fast and efficient identification of cancer Lyutvinskiy Y, et al. Proteomic analysis of urinary biomarker
associated alterations in glycosylation. Anal Chem 2010;82: candidates for nonmuscle invasive bladder cancer. Proteomics
10208–15. 2012;12:135–44.
[70] Xu Y, Zhang L, Hu G. Potential application of alternatively [86] Bella J, Hindle KL, McEwan PA, Lovell SC. The leucine-rich
glycosylated serum MUC1 and MUC5AC in gastric cancer repeat structure. Cell Mol Life Sci 2008;65:2307–33.
diagnosis. Biologicals 2009;37:18–25. [87] Shirai R, Hirano F, Ohkura N, Ikeda K, Inoue S. Up-regulation
[71] Klaamas K, Kodar K, Kurtenkov O. An increased level of the of the expression of leucine-rich alpha(2)-glycoprotein in
Concanavalin A-positive IgG in the serum of patients with hepatocytes by the mediators of acute-phase response.
gastric cancer as evaluated by a lectin enzyme-linked Biochem Biophys Res Commun 2009;382:776–9.
immunosorbent assay (LELISA). Neoplasma 2008;55:143–50. [88] Himmelfarb M, Klopocki E, Grube S, Staub E, Klaman I,
[72] Bones J, Byrne JC, O'Donoghue N, McManus C, Scaife C, Boissin Hinzmann B, et al. ITIH5, a novel member of the
H, et al. Glycomic and glycoproteomic analysis of serum from inter-alpha-trypsin inhibitor heavy chain family is
patients with stomach cancer reveals potential markers downregulated in breast cancer. Cancer Lett 2004;204:69–77.
arising from host defense response mechanisms. J Proteome [89] Paris S, Sesboue R, Delpech B, Chauzy C, Thiberville L,
Res 2011;10:1246–65. Martin JP, et al. Inhibition of tumor growth and metastatic
[73] Gautier V, Mouton-Barbosa E, Bouyssie D, Delcourt N, Beau M, spreading by overexpression of inter-alpha-trypsin inhibitor
Girard JP, et al. Label-free quantification and shotgun analysis family chains. Int J Cancer 2002;97:615–20.
J O U RN A L OF P ROT EO M IC S 8 3 ( 2 01 3 ) 1 9 7 –2 13 213

[90] Chong PK, Lee H, Zhou J, Liu SC, Loh MC, Wang TT, et al. [100] Huq MD, Tsai NP, Khan SA, Wei LN. Lysine trimethylation of
ITIH3 is a potential biomarker for early detection of gastric retinoic acid receptor-alpha: a novel means to regulate
cancer. J Proteome Res 2010;9:3671–9. receptor function. Mol Cell Proteomics 2007;6:677–88.
[91] Baldwin ET, Sarver RW, Bryant Jr GL, Curry KA, Fairbanks MB, [101] Werner R, Manthey KC, Griffin JB, Zempleni J. HepG2 cells
Finzel BC, et al. Cation binding to the integrin CD11b I domain develop signs of riboflavin deficiency within 4 days of
and activation model assessment. Structure 1998;6:923–35. culture in riboflavin-deficient medium. J Nutr Biochem
[92] Colombatti A, Bonaldo P. The superfamily of proteins with 2005;16:617–24.
von Willebrand factor type A-like domains: one theme [102] Pinto J, Huang YP, Chaudhuri R, Rivlin RS. Disturbances in
common to components of extracellular matrix, hemostasis, the formation of FAD and covalently bound flavins in
cellular adhesion, and defense mechanisms. Blood 1991;77: Novikoff hepatoma from riboflavin-deficient rats. Nutr
2305–15. Cancer 1987;10:95–102.
[93] Enghild JJ, Salvesen G, Thogersen IB, Valnickova Z, Pizzo SV, [103] Koturbash I, Simpson NE, Beland FA, Pogribny IP. Alterations
Hefta SA. Presence of the in histone H4 lysine 20 methylation: implications for cancer
protein-glycosaminoglycan-protein covalent cross-link in detection and prevention. Antioxid Redox Signal 2012;17:
the inter-alpha-inhibitor-related proteinase inhibitor heavy 365–74.
chain 2/bikunin. J Biol Chem 1993;268:8711–6. [104] Wada Y, Ito M, Takata S, Tanaka S, Yoshihara M, Chayama
[94] Lee JO, Rieu P, Arnaout MA, Liddington R. Crystal structure K. Relationship between Helicobacter pylori
of the A domain from the alpha subunit of integrin CR3 tyrosine-phosphorylated CagA-related markers and the
(CD11b/CD18). Cell 1995;80:631–8. development of diffuse-type gastric cancers: a case–control
[95] Bajt ML, Loftus JC. Mutation of a ligand binding domain of study. Digestion 2010;82:10–7.
beta 3 integrin. Integral role of oxygenated residues in alpha [105] Chang X, Han J, Pang L, Zhao Y, Yang Y, Shen Z. Increased
IIb beta 3 (GPIIb-IIIa) receptor function. J Biol Chem 1994;269: PADI4 expression in blood and tissues of patients with
20913–9. malignant tumors. BMC Cancer 2009;9:40.
[96] Kondo Y, Shen L, Cheng AS, Ahmed S, Boumber Y, Charo C, [106] Oya-Ito T, Naito Y, Takagi T, Handa O, Matsui H, Yamada M,
et al. Gene silencing in cancer by histone H3 lysine 27 et al. Heat-shock protein 27 (Hsp27) as a target of
trimethylation independent of promoter DNA methylation. methylglyoxal in gastrointestinal cancer. Biochim Biophys
Nat Genet 2008;40:741–50. Acta 1812;2011:769–81.
[97] Schneider AC, Heukamp LC, Rogenhofer S, Fechner G, [107] Sugiura T. The cellular level of TRIM31, an RBCC protein
Bastian PJ, von Ruecker A, et al. Global histone H4K20 overexpressed in gastric cancer, is regulated by multiple
trimethylation predicts cancer-specific survival in patients mechanisms including the ubiquitin–proteasome system.
with muscle-invasive bladder cancer. BJU Int 2011;108: Cell Biol Int 2011;35:657–61.
E290–6. [108] Grivennikov SI, Greten FR, Karin M. Immunity, inflammation,
[98] Zhang L, Zhong K, Dai Y, Zhou H. Genome-wide analysis of and cancer. Cell 2010;140:883–99.
histone H3 lysine 27 trimethylation by ChIP-chip in gastric [109] Ikeguchi M, Hatada T, Yamamoto M, Miyake T, Matsunaga T,
cancer patients. J Gastroenterol 2009;44:305–12. Fukumoto Y, et al. Serum interleukin-6 and -10 levels in
[99] Li Q, Wang X, Lu Z, Zhang B, Guan Z, Liu Z, et al. Polycomb patients with gastric cancer. Gastric Cancer 2009;12:95–100.
CBX7 directly controls trimethylation of histone H3 at lysine
9 at the p16 locus. PLoS One 2010;5:e13732.

You might also like