Skip to main content
Molecular & Cellular Proteomics : MCP logoLink to Molecular & Cellular Proteomics : MCP
. 2013 Jan 23;12(4):956–978. doi: 10.1074/mcp.M112.024018

Glycoproteomic Analysis of the Secretome of Human Endothelial Cells*

Xiaoke Yin , Marshall Bern §, Qiuru Xing , Jenny Ho , Rosa Viner , Manuel Mayr ‡,**
PMCID: PMC3617342  PMID: 23345538

Abstract

Previous proteomics studies have partially unraveled the complexity of endothelial protein secretion but have not investigated glycosylation, a key modification of secreted and membrane proteins for cell communication. In this study, human umbilical vein endothelial cells were kept in serum-free medium before activation by phorbol-12-myristate-13 acetate, a commonly used secretagogue that induces exocytosis of endothelial vesicles. In addition to 123 secreted proteins, the secretome was particularly rich in membrane proteins. Glycopeptides were enriched by zwitterionic hydrophilic interaction liquid chromatography resins and were either treated with PNGase F and H218O or directly analyzed using a recently developed workflow combining higher-energy C-trap dissociation (HCD) with electron-transfer dissociation (ETD) for a hybrid linear ion trap–orbitrap mass spectrometer. After deglycosylation with PNGase F in the presence of H218O, 123 unique peptides displayed 18O-deamidation of asparagine, corresponding to 86 proteins with a total of 121 glycosylation sites. Direct glycopeptide analysis via HCD-ETD identified 131 glycopeptides from 59 proteins and 118 glycosylation sites, of which 41 were known, 51 were predicted, and 26 were novel. Two methods were compared: alternating HCD-ETD and HCD-product-dependent ETD. The former detected predominantly high-intensity, multiply charged glycopeptides, whereas the latter preferentially selected precursors with complex/hybrid glycans for fragmentation. Validation was performed by means of glycoprotein enrichment and analysis of the input, the flow-through, and the bound fraction. This study represents the most comprehensive characterization of endothelial protein secretion to date and demonstrates the potential of new HCD-ETD workflows for determining the glycosylation status of complex biological samples.


Cardiovascular disease manifests predominantly as myocardial ischemia, heart failure, stroke, aortic aneurysm, and peripheral vascular disease and leads to the majority of deaths and disabilities worldwide. Endothelial cells (ECs) constitute the inner lining of all blood vessels and form the interface between the circulation and the vascular wall (1). The endothelial monolayer is pivotal for maintaining vascular homeostasis through a balance of endothelium-derived factors (2, 3). ECs are preferred targets of cardiovascular risk factors such as hypercholesterolemia, diabetes, hypertension, and smoking (1, 4). Repetitive injury is associated with a varying degree of endothelial dysfunction. Alterations in its anticoagulant and anti-inflammatory properties leave the vasculature susceptible to disease (5) and play a key role in the initiation and progression of cardiovascular disease (6).

Previous proteomics studies (713), including one by our group (8), have investigated the secretome of unstimulated human umbilical vein ECs (HUVECs), the most widely used ECs in cardiovascular research. Only two studies have explored the secretome of HUVECs upon activation by shear stress (10) or with statin treatment (13) thus far. One study used human microvascular ECs (9), which represent a distinct population of ECs from small vessels. Yet many factors secreted by ECs were not identified, probably because of their low abundance. In this study, we used a secretagogue, phorbol ester phorbol-12-myristate-13-acetate (PMA) (14, 15), to induce maximal protein release from serum-starved HUVECs over 45 min. In addition, we applied three different proteomic strategies for the analysis of glycoproteins/glycopeptides to further enrich secreted proteins and characterize their glycosylation sites.

EXPERIMENTAL PROCEDURES

EC Culture

HUVECs (Lonza Group Ltd., Basel, Switzerland) were cultured on 0.1% gelatin-coated flasks in M199 medium supplemented with 1 ng/ml endothelial cell growth factor (Sigma), 3 μg/ml endothelial growth supplement from bovine neural tissue (Sigma), 10 U/ml heparin, 1.25 μg/ml thymidine, 10% fetal bovine serum (A15–108, PAA Laboratories, Velizy-Villacoublay, France), and 100 μg/ml penicillin and streptomycin in a humidified incubator supplemented with 5% CO2 at 37 °C. The cells were subcultured every 2 to 3 days at a ratio of 1:4 (16).

Conditioned Medium Collection

HUVECs were cultured in complete medium until confluent. Then, they were washed and incubated in M199 medium for 30 min twice before stimulation with 50 nm PMA (Sigma) in M199 medium for 45 min. The control group was incubated with M199 medium in the absence of PMA for 45 min. Conditioned media were collected and stored at −80 °C for further analysis.

Immunofluorescence Staining

HUVECs were cultured in Nunc chamber slides (Sigma-Aldrich) for 3 days. HUVECs were stimulated with 50 nm PMA in M199 medium for 45 min or incubated with M199 medium for 45 min. The cells were fixed with 4% formaldehyde in PBS for 10 min, permeabilized with 0.1% Triton X-100 in PBS for 5 min, and blocked in 5% fetal bovine serum in PBS for 30 min at 37 °C. Following 1 h of incubation with the primary antibodies, VE-cadherin (ab33168, Abcam, Cambridge, UK), and von Willebrand factor (vWF) (sc-8068, Santa Cruz Biotechnology, Santa Cruz, CA) at 37 °C, an Alexa Fluor® 594 conjugated donkey anti-rabbit IgG and an Alexa Fluor® 488 conjugated donkey anti-goat IgG, respectively, were added, and the cells were incubated at 37 °C for 30 min. Nuclei were counterstained with 4′,6-diamidino-2-phenylindole (D9542, Sigma) for 5 min. The slide was mounted in fluorescence mounting medium (DAKO, Denmark A/S, Glostrup, Denmark) and examined with an AxioPlan 2 fluorescence microscope (Carl Zeiss, Thornwood, NY) (17).

Proteomics Profiling of the Secretome

Conditioned media were concentrated with an Amicon spin column (3kD MWCO, EDO Millipore Corp., Billerica, MA) and separated via 4%–12% Bis-Tris SDS-PAGE (Invitrogen). Proteins were visualized via silver staining (PlusOne silver staining kit for proteins, GE Healthcare). Gel bands were digested with modified trypsin (Promega Corp., Madison, WI) overnight on a ProGest digestion robot (Digilab Inc., Marlborough, MA) and analyzed via reverse-phase nano-flow HPLC (PepMap C18, 3 μm, 100 Å, 25 cm × 75 μm inner diameter column, Thermo Scientific) interfaced to an LTQ Orbitrap XL MS (Thermo Scientific) (18).

Deglycosylation

Concentrated media were mixed with deglycosylation buffer (150 mm NaCl, 50 mm sodium acetate, 10 mm EDTA, proteinase inhibitors, pH 6.8) supplemented with 0.05U PNGase F (Sigma), chondroitinase ABC (C3667, Sigma), and keratanase (G6920, Sigma) and incubated at 37 °C overnight (19).

Immunoblotting

Concentrated or deglycosylated media were separated via 4%–12% Bis-Tris gel (Invitrogen). Proteins were transferred on a nitrocellulose membrane and blocked with 5% bovine serum albumin in PBS. Membranes were incubated with primary antibody overnight at 4 °C. Secondary antibodies were incubated for 1 h at room temperature. After the addition of ECL (GE Healthcare), the film was developed using a Compact X4 Automatic Processor (Xograph Healthcare Ltd., Stonehouse, UK). The following primary antibodies were used: agrin (sc-25528, Santa Cruz Biotechnology), biglycan (ab54855, Abcam), connective tissue growth factor (sc-25440, Santa Cruz Biotechnology), fibronectin (sc-56391, Santa Cruz Biotechnology), and lymphatic vessel endothelial hyaluronic acid receptor 1 (AF2089, R&D Systems).

Difference Gel Electrophoresis

Conditioned media from HUVECs treated with or without PMA were concentrated using an Amicon spin column (3kD MWCO, Millipore) and the ReadyPrep 2D clean-up kit (Bio-Rad). The pellet was resuspended in difference gel electrophoresis lysis buffer (30 mm Tris, 8 m urea, 4% w/v CHAPS, protease inhibitors, pH 8.5). For each secretome sample, 15 μg of proteins were labeled with Cy3 or Cy5. A dye swap was performed to exclude preferential labeling. Cellular extracts of HUVECs were labeled with Cy2. Cy2-, Cy3-, and Cy5-labeled samples were separated via isoelectric focusing on immobilized pH gradient dry strips (18 cm, pH 3–10 NL, GE Healthcare) with 30 KVH. The strips were equilibrated with 10 mg/ml DTT in equilibration buffer (6 m urea, 2% w/v SDS, 30% v/v glycerol, 50 mm Tris, pH 8.8) for 15 min followed by 48 mg/ml iodoacetamide in equilibration buffer for 15 min before separation via SDS-PAGE at 100 W for 4 h using an Ettan DALTsix vertical electrophoresis system (GE Healthcare) (2022). Gels were scanned on an Ettan difference gel electrophoresis imager (GE Healthcare). Images were overlaid with ImageQuant TL software (GE Healthcare). Common spots present in both the cellular proteome and the secretome were excised, digested with trypsin, and identified using nano-flow HPLC-MS/MS. Detailed protocols are available on our research group's website.

Glycopeptide Enrichment

Conditioned media were desalted via the use of Zeba spin columns (Thermo Scientific). Proteins were then reduced by 5 mm DTT and alkylated with 25 mm iodoacetamide. After acetone precipitation overnight, the pellet was resuspended in 100 mm triethylammonium bicarbonate (pH 8.5, Sigma) and digested with modified trypsin (Promega) at 37 °C overnight. Peptides were labeled at a ratio of 100 μg peptides/0.8 mg Tandem Mass Tag Zero (TMT0) (Thermo Scientific) according to the manufacturer's instruction. Labeled peptides were further enriched for glycopeptides using zwitterionic hydrophilic interaction liquid chromatography resin (Merck) (23).

LC/MS of Intact Glycopeptides

The glycopeptide enriched fraction was separated using the EASY-nLCTM nano-HPLC system (Thermo Scientific) with a Magic C18 spray tip 15 cm × 75 μm inner diameter column (Bruker-Michrom, Auburn, CA). Gradient elution was performed with 4% to 30% acetonitrile in 0.1% formic acid over 60 min at a flow rate of 300 nl/min. The samples were analyzed with an Orbitrap Elite hybrid MS with electron-transfer dissociation (ETD) (Thermo Scientific). The following MS and MS/MS settings were used: Fourier transform: MSn automatic gain control target = 5E4; MS/MS = 1 μscans, max ion time = 200 ms; MS = 300–1800 m/z, resolution = 60,000 at m/z 400, MS target = 1E6; dynamic exclusion = repeat count 1, duration 30 s, exclusion duration 90 s; higher-energy C-trap dissociation (HCD): collision energy = 35%, resolution = 15,000; MSn target ion trap = 1E4, 2 μscans, max ion time = 150 ms; ETD anion automatic gain control target = 2E5, charge-dependent ETD reaction time enabled. For alternating HCD-ETD MS/MS, the top 10 ions were analyzed. For HCD-product-dependent ETD, the top 10 ions were analyzed via HCD, and product-dependent ETD acquisition was triggered by product (oxonium) ions (m/z 163.0812 for Hex; m/z 204.0864 for HexNAc; m/z 138.0554 for HexNAc fragment ion) (24).

Deglycosylation with PNGase F and H218O

Zwitterionic hydrophilic interaction liquid chromatography resin enriched glycopeptides were resuspended in 50 mm ammonium bicarbonate in H218O (97 atom % 18O, Sigma) and deglycosylated with PNGase F (Sigma) for 4 h at 37 °C. The samples were separated via reverse-phase nano-flow HPLC (PepMap C18, 3 μm, 100 Å, 25 cm × 75 μm inner diameter column, Thermo Scientific) before analysis on an LTQ Orbitrap XL MS (Thermo Scientific).

Glycoprotein Enrichment and LC/MS

ConA1 lectin resins (Thermo Scientific) were used to enrich glycoproteins from concentrated conditioned media according to the manufacturer's protocol. The input, glycoprotein-enriched fraction, and flow-through samples were subjected to trypsin digestion. The in-solution digests were separated on a Thermo Scientific Dionex UltiMate 3000 Rapid Separation LC (RSLC) system using a PepMap C18 column (3 μm, 100 Å, 50 cm × 75 μm inner diameter column, Thermo Scientific). The rapid separation LC system was interfaced to a Q Exactive MS (Thermo Scientific), and samples were analyzed using a top-10 HCD method.

Database Search and Data analysis

The following parameters were used for different experiments.

(i) Gel-LC-MS/MS: Peak lists were generated by Mascot daemon (version 2.3.0, Matrix Science Ltd., London, UK) using extract_msn_com.exe and searched against the UniProt/Swiss-Prot mammalian database (version 2012.03, 65,780 entries) using Mascot (version 2.3.01, Matrix Science) with peptide tolerance = 10 ppm, MS/MS tolerance = 0.8 Da, carbamidomethylation of cysteine as a fixed modification, oxidation of methionine as a variable modification, and a maximum of two missed cleavage sites. The search results were loaded into Scaffold software (version 3.6.2, Proteome Software Software, Inc., Portland, OR). A protein probability greater than 99%, a peptide probability greater than 95%, and a minimum number of two peptides per protein were applied as filters to generate the protein list. Bovine contaminant proteins are listed separately.

(ii) PNGase F + H218O experiment: Thermo Scientific Proteome Discoverer software version 1.3 was used to search against the UniProt/Swiss-Prot mammalian database (version 2012.03) using Mascot (version 2.3.01, Matrix Science) with a peptide tolerance of 10 ppm; an MS/MS tolerance of 0.8 Da; carbamidomethylation of cysteine as a fixed modification; oxidation of methionine, TMT0 label on lysine and peptide N-terminus, and deamidation (spontaneous deamidation in ordinary water) and O18-deamidation (deglycosylation by PNGase F in H218O) of asparagine as variable modifications; and a maximum of two missed cleavage sites. Proteome Discoverer produced a custom database containing 136 target proteins based on this search.

(iii) Orbitrap Elite MS: Raw files were searched against the 136-protein database (along with reversed proteins as decoys) using ByonicTM (25) with a peptide tolerance of 10 ppm; an MS/MS tolerance of 20 ppm for HCD and 0.6 Da for ETD; the carbamidomethylated cysteine, TMT0 label on lysine and peptide N-terminus as fixed modifications; and oxidation of methionine, deamidation of asparagine and glutamine, and phosphorylation of serine and threonine as variable modifications. ByonicTM allowed one N-glycan modification on the N-X(not P)-S/T consensus motif per peptide, with mass and composition chosen from its “common human” glycan database containing 350 glycan masses up to 6000 Da. Glycan modifications were verified by the presence of corresponding glycan fragment ions, such as the HexNAc oxonium ion at 204.087 Da in HCD spectra. Peptide sequences were identified by ByonicTM from the ETD spectra and verified manually.

(iv) Q Exactive MS: Raw files were searched against the UniProt/Swiss-Prot human database (version 57.13, 20,266 entries) using Proteome Discoverer (version 1.3, Thermo Scientific) with Mascot (version 2.3.0, Matrix Science) and a peptide tolerance of 10 ppm, an MS/MS tolerance of 10 mmu, carbamidomethylation of cysteine as a fixed modification, oxidation of methionine as a variable modification, and a maximum of two missed cleavage sites.

RESULTS

The Secretome of Activated ECs

HUVECs were stimulated with PMA, a commonly used secretagogue that induces exocytosis of endothelial vesicles. As previously reported (26), the morphology of ECs changes from spindle-shaped to round upon PMA activation, and the rod-shaped Weibel-Palade bodies, unique storage vesicles within ECs containing vWF and many other secreted proteins, fuse with the cell membrane (Fig. 1A). In total, the secretomes of 17 primary ECs were analyzed via gel-LC-MS/MS, with or without deglycosylation. Apart from 123 secreted proteins, the conditioned medium of PMA-stimulated ECs was particularly rich in surface antigens and receptors, including many established endothelial markers (Table I). All identified proteins and peptides are listed in supplemental Tables S1 and S2, respectively. The distribution of the frequencies and the cumulated distribution of the number of samples in which proteins were identified are shown in supplemental Fig. S1. MS datasets of three biological replicates have been deposited in PRIDE (accession numbers 26908–27003).

Fig. 1.

Fig. 1.

PMA treatment to stimulate EC secretion. Treatment of HUVECs with PMA, a commonly used secretagogue, resulted in a characteristic morphological change indicative of activation. A, immunofluorescence staining of vWF (green) and VE-cadherin (red) shows the exocytotic effect of PMA. B, PMA increased protein secretion in the conditioned media as confirmed via immunoblotting. C, relative to previous studies, more than twice as many secreted and plasma membrane proteins were identified. D, overlay of intracellular and secreted proteins by means of difference gel electrophoresis. In the left-hand panel, proteins in conditioned media of HUVECs are stained in green (+PMA) and red (−PMA), and cellular proteins are stained in blue. Results were reproduced with different biological replicates using reverse-labeling (right-hand panel: red, +PMA; green, −PMA). The protein corresponding to von Willebrand antigen 2 is highlighted with a box. Common proteins in the secretome and the cellular proteome are numbered in supplemental Fig. S2 and listed in supplemental Table S3.

Table I. Extracellular and plasma membrane proteins identified in the HUVEC-conditioned media after PMA stimulation.
Protein name UniProt ID UniProt accession number Gene name Cellular component Glycoprotein EC marker
Calcium ion-binding proteins
    Annexin A1 ANXA1_HUMAN P04083 ANXA1 Plasma membrane
    Annexin A2a ANXA2_HUMAN P07355 ANXA2 Extracellular Plasma membrane
    Annexin A3 ANXA3_HUMAN P12429 ANXA3 Plasma membrane
    Calreticulin CALR_HUMAN P27797 CALR Extracellular Glycoprotein
    Calumenin CALU_HUMAN O43852 CALU Extracellular Glycoprotein
    Calpain-1 catalytic subunit CAN1_HUMAN P07384 CAPN1 Plasma membrane
    Calpain-2 catalytic subunit CAN2_HUMAN P17655 CAPN2 Plasma membrane
    Calpain small subunit 1 CPNS1_HUMAN P04632 CAPNS1 Plasma membrane
    Calsyntenin-1 CSTN1_HUMAN O94985 CLSTN1 Plasma membrane Glycoprotein
    Calsyntenin-3 CSTN3_HUMAN Q9BQT9 CLSTN3 Plasma membrane Glycoprotein
    Desmoglein-1 DSG1_HUMAN Q02413 DSG1 Plasma membrane Glycoprotein
    Nucleobindin-2 NUCB2_HUMAN P80303 NUCB2 Extracellular
Carbohydrate and glycan metabolism
    Alpha-amylase 1 AMY1_HUMAN P04745 AMY1C Extracellular Glycoprotein
    Exostosin-like 2 EXTL2_HUMAN Q9UBQ6 EXTL2 Extracellular Glycoprotein
    Polypeptide N-acetylgalactosaminyltransferase 1 GALT1_HUMAN Q10472 GALNT1 Extracellular Glycoprotein
    Sialate O-acetylesterase SIAE_HUMAN Q9HAT2 SIAE Extracellular Glycoprotein
    UDP-N-acetylhexosamine pyrophosphorylase UAP1_HUMAN Q16222 UAP1 Plasma membrane
Coagulation and related proteins
    Amyloid-like protein 2 APLP2_HUMAN Q06481 APLP2 Plasma membrane Glycoprotein
    Multimerin-1 MMRN1_HUMAN Q13201 MMRN1 Extracellular Glycoprotein
    Plasminogen activator inhibitor 1 PAI1_HUMAN P05121 SERPINE1 Extracellular Glycoprotein
    Plasminogen activator inhibitor 2 PAI2_HUMAN P05120 SERPINB2 Extracellular Glycoprotein
    Tissue factor pathway inhibitor TFPI1_HUMAN P10646 TFPI Extracellular Glycoprotein
    Tissue factor pathway inhibitor 2 TFPI2_HUMAN P48307 TFPI2 Extracellular Glycoprotein
    Tissue-type plasminogen activator TPA_HUMAN P00750 PLAT Extracellular Glycoprotein
    von Willebrand factor VWF_HUMAN P04275 VWF Extracellular Glycoprotein EC marker
Extracellular matrix components and associated proteins
    Agrin AGRIN_HUMAN O00468 AGRN Extracellular Glycoprotein
    Collagen alpha-2(IV) chain CO4A2_HUMAN P08572 COL4A2 Extracellular Glycoprotein
    Collagen alpha-1(VI) chain CO6A1_HUMAN P12109 COL6A1 Extracellular Glycoprotein
    Collagen alpha-1(XII) chain COCA1_HUMAN Q99715 COL12A1 Extracellular Glycoprotein
    Collagen alpha-1(XVIII) chain COIA1_HUMAN P39060 COL18A1 Extracellular Glycoprotein
    EGF-containing fibulin-like extracellular matrix protein 1 FBLN3_HUMAN Q12805 EFEMP1 Extracellular Glycoprotein
    Fibrillin-1 FBN1_HUMAN P35555 FBN1 Extracellular Glycoprotein
    Fibrillin-2 FBN2_HUMAN P35556 FBN2 Extracellular Glycoprotein
    Fibronectin FINC_HUMAN P02751 FN1 Extracellular Glycoprotein
    Hyaluronan and proteoglycan link protein 3 HPLN3_HUMAN Q96S86 HAPLN3 Extracellular
    Laminin subunit alpha-4 LAMA4_HUMAN Q16363 LAMA4 Extracellular Glycoprotein
    Laminin subunit beta-1 LAMB1_HUMAN P07942 LAMB1 Extracellular Glycoprotein
    Laminin subunit gamma-1 LAMC1_HUMAN P11047 LAMC1 Extracellular Glycoprotein
    Lysyl oxidase homolog 2 LOXL2_HUMAN Q9Y4K0 LOXL2 Extracellular Glycoprotein
    Multimerin-2 MMRN2_HUMAN Q9H8L6 MMRN2 Extracellular Glycoprotein
    Nidogen-1 NID1_HUMAN P14543 NID1 Extracellular Glycoprotein
    Nidogen-2 NID2_HUMAN Q14112 NID2 Extracellular Glycoprotein
    Prolyl 3-hydroxylase 1 P3H1_HUMAN Q32P28 LEPRE1 Extracellular Glycoprotein
    Basement membrane-specific heparan sulfate proteoglycan core protein PGBM_HUMAN P98160 HSPG2 Extracellular Glycoprotein
    Biglycan PGS1_HUMAN P21810 BGN Extracellular Glycoprotein
    Peroxidasin homolog PXDN_HUMAN Q92626 PXDN Extracellular Glycoprotein
    SPARC SPRC_HUMAN P09486 SPARC Extracellular Glycoprotein
    Target of Nesh-SH3 TARSH_HUMAN Q7Z7G0 ABI3BP Extracellular Glycoprotein
    Testican-1 TICN1_HUMAN Q08629 SPOCK1 Extracellular Glycoprotein
    Thrombospondin-1 TSP1_HUMAN P07996 THBS1 Extracellular Plasma membrane Glycoprotein
Growth factors and related proteins
    C-type lectin domain family 11 member A CLC11_HUMAN Q9Y240 CLEC11A Extracellular Glycoprotein
    Cysteine-rich motor neuron 1 protein CRIM1_HUMAN Q9NZV1 CRIM1 Extracellular Plasma membrane Glycoprotein
    Connective tissue growth factor CTGF_HUMAN P29279 CTGF Extracellular Glycoprotein
    Protein CYR61, insulin-like growth factor-binding protein 10 CYR61_HUMAN O00622 CYR61 Extracellular
    Dickkopf-related protein 3 DKK3_HUMAN Q9UBP4 DKK3 Extracellular Glycoprotein
    Follistatin-related protein 1 FSTL1_HUMAN Q12841 FSTL1 Extracellular Glycoprotein
    Hepatoma-derived growth factor HDGF_HUMAN P51858 HDGF Extracellular
    Insulin-like growth factor-binding protein 2 IBP2_HUMAN P18065 IGFBP2 Extracellular Glycoprotein
    Insulin-like growth factor-binding protein 7 IBP7_HUMAN Q16270 IGFBP7 Extracellular Glycoprotein
    Latent-transforming growth factor beta-binding protein 1 LTBP1_HUMAN Q14766 LTBP1 Extracellular Glycoprotein
    Latent-transforming growth factor beta-binding protein 2 LTBP2_HUMAN Q14767 LTBP2 Extracellular Glycoprotein
    Neuronal growth regulator 1 NEGR1_HUMAN Q7Z3B1 NEGR1 Plasma membrane Glycoprotein
Immunity- and inflammation-related proteins
    Amyloid beta A4 protein A4_HUMAN P05067 APP Extracellular Plasma membrane Glycoprotein
    Beta-2-microglobulin B2MG_HUMAN P61769 B2M Extracellular Glycoprotein
    Complement C1q tumor necrosis factor-related protein 5 C1QT5_HUMAN Q9BXJ0 C1QTNF5 Extracellular
    Complement factor H CFAH_HUMAN P08603 CFH Extracellular Glycoprotein
    Interleukin-25, UPF0556 protein C19orf10 CS010_HUMAN Q969H8 C19orf10 Extracellular
    Granulins GRN_HUMAN P28799 GRN Extracellular Glycoprotein
    Interferon-induced transmembrane protein 1 IFM1_HUMAN P13164 IFITM1 Plasma membrane
    Galectin-1a LEG1_HUMAN P09382 LGALS1 Extracellular
    Galectin-3 LEG3_HUMAN P17931 LGALS3 Extracellular
    Macrophage migration inhibitory factora MIF_HUMAN P14174 MIF Extracellular
    NKG2D ligand 2 N2DL2_HUMAN Q9BZM5 ULBP2 Extracellular Plasma membrane Glycoprotein
    Pentraxin-related protein PTX3 PTX3_HUMAN P26022 PTX3 Extracellular Glycoprotein
    Protein S100-A7 S10A7_HUMAN P31151 S100A7 Extracellular
    Protein S100-A8 S10A8_HUMAN P05109 S100A8 Extracellular Plasma membrane
    Tubulointerstitial nephritis antigen-like TINAL_HUMAN Q9GZM7 TINAGL1 Extracellular Glycoprotein
    Nuclease-sensitive element-binding protein 1 YBOX1_HUMAN P67809 YBX1 Extracellular
    Zinc-alpha-2-glycoprotein ZA2G_HUMAN P25311 AZGP1 Extracellular Glycoprotein
Membrane antigens and receptors
    HLA class I histocompatibility antigen, A-24 alpha chain 1A24_HUMAN P05534 HLA-A Plasma membrane Glycoprotein
    HLA class I histocompatibility antigen, A-30 alpha chain 1A30_HUMAN P16188 HLA-A Plasma membrane Glycoprotein
    HLA class I histocompatibility antigen, Cw-12 alpha chain 1C12_HUMAN P30508 HLA-C Plasma membrane Glycoprotein
    Alpha-2-macroglobulin receptor-associated protein AMRP_HUMAN P30533 LRPAP1 Extracellular Plasma membrane Glycoprotein
    Basal cell adhesion molecule BCAM_HUMAN P50895 BCAM Plasma membrane Glycoprotein
    Complement component C1q receptor C1QR1_HUMAN Q9NPY3 CD93 Plasma membrane Glycoprotein EC marker
    Cadherin-13 CAD13_HUMAN P55290 CDH13 Plasma membrane Glycoprotein
    Cadherin-2 CADH2_HUMAN P19022 CDH2 Plasma membrane Glycoprotein
    Cadherin-5 CADH5_HUMAN P33151 CDH5 Plasma membrane Glycoprotein EC marker
    CD109 antigen CD109_HUMAN Q6YHK3 CD109 Plasma membrane Glycoprotein
    CD166 antigen CD166_HUMAN Q13740 ALCAM Plasma membrane Glycoprotein
    CD44 antigen CD44_HUMAN P16070 CD44 Plasma membrane Glycoprotein
    CD59 glycoprotein CD59_HUMAN P13987 CD59 Extracellular Plasma membrane Glycoprotein
    CD9 antigen CD9_HUMAN P21926 CD9 Plasma membrane Glycoprotein
    C-type lectin domain family 14 member A CLC14_HUMAN Q86T13 CLEC14A Plasma membrane Glycoprotein
    Dystroglycan DAG1_HUMAN Q14118 DAG1 Extracellular Plasma membrane Glycoprotein
    Endoglin EGLN_HUMAN P17813 ENG Plasma membrane Glycoprotein EC marker
    Endothelial protein C receptor EPCR_HUMAN Q9UNN8 PROCR Plasma membrane Glycoprotein EC marker
    Ephrin type-B receptor 4 EPHB4_HUMAN P54760 EPHB4 Plasma membrane Glycoprotein
    Endothelial cell-selective adhesion molecule ESAM_HUMAN Q96AP7 ESAM Plasma membrane Glycoprotein EC marker
    Leucine-rich repeat transmembrane protein FLRT2 FLRT2_HUMAN O43155 FLRT2 Plasma membrane Glycoprotein
    Guanine nucleotide-binding protein subunit beta-2-like 1a GBLP_HUMAN P63244 GNB2L1 Plasma membrane
    HLA class I histocompatibility antigen, alpha chain E HLAE_HUMAN P13747 HLA-E Plasma membrane Glycoprotein
    Intercellular adhesion molecule 1 ICAM1_HUMAN P05362 ICAM1 Extracellular Plasma membrane Glycoprotein EC marker
    Intercellular adhesion molecule 2 ICAM2_HUMAN P13598 ICAM2 Plasma membrane Glycoprotein EC marker
    Integrin alpha-2 ITA2_HUMAN P17301 ITGA2 Plasma membrane Glycoprotein
    Integrin alpha-5 ITA5_HUMAN P08648 ITGA5 Plasma membrane Glycoprotein
    Integrin alpha-6 ITA6_HUMAN P23229 ITGA6 Plasma membrane Glycoprotein
    Integrin beta-1 ITB1_HUMAN P05556 ITGB1 Plasma membrane Glycoprotein EC marker
    Protein jagged-1 JAG1_HUMAN P78504 JAG1 Plasma membrane Glycoprotein
    Protein jagged-2 JAG2_HUMAN Q9Y219 JAG2 Plasma membrane Glycoprotein
    Junctional adhesion molecule A JAM1_HUMAN Q9Y624 F11R Plasma membrane Glycoprotein
    BTB/POZ domain-containing protein KCTD12 KCD12_HUMAN Q96CX2 KCTD12 Plasma membrane
    Kinectin KTN1_HUMAN Q86UP2 KTN1 Plasma membrane Glycoprotein
    Lysosome-associated membrane glycoprotein 1 LAMP1_HUMAN P11279 LAMP1 Plasma membrane Glycoprotein
    Low-density lipoprotein receptor LDLR_HUMAN P01130 LDLR Plasma membrane Glycoprotein
    Low-density lipoprotein receptor-related protein 5 LRP5_HUMAN O75197 LRP5 Plasma membrane Glycoprotein
    Lymphatic vessel endothelial hyaluronic acid receptor 1 LYVE1_HUMAN Q9Y5Y7 LYVE1 Plasma membrane Glycoprotein EC marker
    Hepatocyte growth factor receptor MET_HUMAN P08581 MET Extracellular Plasma membrane Glycoprotein
    Cation-independent mannose-6-phosphate receptor MPRI_HUMAN P11717 IGF2R Plasma membrane Glycoprotein
    C-type mannose receptor 2 MRC2_HUMAN Q9UBG0 MRC2 Plasma membrane Glycoprotein
    Cell surface glycoprotein MUC18 MUC18_HUMAN P43121 MCAM Plasma membrane Glycoprotein EC marker
    Neuroligin-1 NLGN1_HUMAN Q8N2Q7 NLGN1 Plasma membrane Glycoprotein
    Neuronal cell adhesion molecule NRCAM_HUMAN Q92823 NRCAM Plasma membrane Glycoprotein
    Neuropilin-1 NRP1_HUMAN O14786 NRP1 Extracellular Plasma membrane Glycoprotein
    Neuropilin-2 NRP2_HUMAN O60462 NRP2 Plasma membrane Glycoprotein
    Neurotrimin NTRI_HUMAN Q9P121 NTM Plasma membrane Glycoprotein
    Protocadherin-10 PCD10_HUMAN Q9P2E7 PCDH10 Plasma membrane Glycoprotein
    Protocadherin-12 PCD12_HUMAN Q9NPG4 PCDH12 Plasma membrane Glycoprotein
    Protocadherin gamma-A11 PCDGB_HUMAN Q9Y5H2 PCDHGA11 Plasma membrane Glycoprotein
    Protocadherin gamma-A12 PCDGC_HUMAN O60330 PCDHGA12 Plasma membrane Glycoprotein
    Protocadherin gamma-B7 PCDGJ_HUMAN Q9Y5F8 PCDHGB7 Plasma membrane Glycoprotein
    Protocadherin-1 PCDH1_HUMAN Q08174 PCDH1 Plasma membrane Glycoprotein
    Protocadherin-9 PCDH9_HUMAN Q9HC56 PCDH9 Plasma membrane Glycoprotein
    Programmed cell death 1 ligand 2 PD1L2_HUMAN Q9BQ51 PDCD1LG2 Extracellular Plasma membrane Glycoprotein
    Platelet endothelial cell adhesion molecule PECA1_HUMAN P16284 PECAM1 Plasma membrane Glycoprotein EC marker
    Plexin-D1 PLXD1_HUMAN Q9Y4D7 PLXND1 Plasma membrane Glycoprotein
    Inactive tyrosine-protein kinase 7 PTK7_HUMAN Q13308 PTK7 Plasma membrane Glycoprotein
    Receptor-type tyrosine-protein phosphatase delta PTPRD_HUMAN P23468 PTPRD Plasma membrane Glycoprotein
    Receptor-type tyrosine-protein phosphatase F PTPRF_HUMAN P10586 PTPRF Plasma membrane Glycoprotein
    Receptor-type tyrosine-protein phosphatase kappa PTPRK_HUMAN Q15262 PTPRK Plasma membrane Glycoprotein
    Poliovirus receptor PVR_HUMAN P15151 PVR Extracellular Plasma membrane Glycoprotein
    Poliovirus receptor-related protein 2 PVRL2_HUMAN Q92692 PVRL2 Plasma membrane Glycoprotein EC marker
    Roundabout homolog 1 ROBO1_HUMAN Q9Y6N7 ROBO1 Plasma membrane Glycoprotein
    Roundabout homolog 4 ROBO4_HUMAN Q8WZ75 ROBO4 Plasma membrane Glycoprotein
    Syndecan-4 SDC4_HUMAN P31431 SDC4 Extracellular Plasma membrane Glycoprotein
    Semaphorin-4D SEM4D_HUMAN Q92854 SEMA4D Plasma membrane Glycoprotein
    Semaphorin-6B SEM6B_HUMAN Q9H3T3 SEMA6B Plasma membrane Glycoprotein
    Tyrosine-protein phosphatase non-receptor type substrate 1 SHPS1_HUMAN P78324 SIRPA Plasma membrane Glycoprotein
    Stabilin-1 STAB1_HUMAN Q9NY15 STAB1 Plasma membrane Glycoprotein EC marker
    Transferrin receptor protein 1 TFR1_HUMAN P02786 TFRC Extracellular Plasma membrane Glycoprotein
    Tyrosine-protein kinase receptor Tie-1 TIE1_HUMAN P35590 TIE1 Plasma membrane Glycoprotein
    Tyrosine-protein kinase receptor UFO UFO_HUMAN P30530 AXL Extracellular Plasma membrane Glycoprotein
    Vascular endothelial growth factor receptor 2 VGFR2_HUMAN P35968 KDR Extracellular Plasma membrane Glycoprotein EC marker
    Vascular endothelial growth factor receptor 3 VGFR3_HUMAN P35916 FLT4 Extracellular Plasma membrane Glycoprotein EC marker
    Very low-density lipoprotein receptor VLDLR_HUMAN P98155 VLDLR Plasma membrane Glycoprotein
Miscellaneous membrane proteins
    Brain acid soluble protein 1 BASP1_HUMAN P80723 BASP1 Plasma membrane
    DnaJ homolog subfamily B member 4 DNJB4_HUMAN Q9UDY4 DNAJB4 Plasma membrane
    RNA-binding protein EWS EWS_HUMAN Q01844 EWSR1 Plasma membrane
    Nck-associated protein 1 NCKP1_HUMAN Q9Y2A7 NCKAP1 Plasma membrane
    Na(+)/H(+) exchange regulatory cofactor NHE-RF2 NHRF2_HUMAN Q15599 SLC9A3R2 Plasma membrane
    Polymerase I and transcript release factor PTRF_HUMAN Q6NZI2 PTRF Plasma membrane
    Serum deprivation-response protein SDPR_HUMAN O95810 SDPR Plasma membrane
    Sushi repeat-containing protein SRPX2 SRPX2_HUMAN O60687 SRPX2 Extracellular
    Erythrocyte band 7 integral membrane protein STOM_HUMAN P27105 STOM Plasma membrane
Miscellaneous secreted proteins
    Peptidyl-glycine alpha-amidating monooxygenase AMD_HUMAN P19021 PAM Extracellular Glycoprotein
    Angiopoietin-2 ANGP2_HUMAN O15123 ANGPT2 Extracellular Glycoprotein
    Endothelin-1 EDN1_HUMAN P05305 EDN1 Extracellular
    Endothelial cell-specific molecule 1 ESM1_HUMAN Q9NQ30 ESM1 Extracellular Glycoprotein
    Protein FAM3C FAM3C_HUMAN Q92520 WNT16 Extracellular
    Epididymal secretory protein E1 NPC2_HUMAN P61916 NPC2 Extracellular Glycoprotein
    Programmed cell death protein 10 PDC10_HUMAN Q9BUL8 PDCD10 Plasma membrane
    Prolactin-inducible protein PIP_HUMAN P12273 PIP Extracellular Glycoprotein
    Sulfhydryl oxidase 1 QSOX1_HUMAN O00391 QSOX1 Extracellular Glycoprotein
    Secretoglobin family 1D member 2 SG1D2_HUMAN O95969 SCGB1D2 Extracellular
    Thioredoxina THIO_HUMAN P10599 TXN Extracellular
    Thymosin beta-4 TYB4_HUMAN P62328 TMSB4X Extracellular
Protease inhibitors
    Cystatin-C CYTC_HUMAN P01034 CST3 Extracellular Glycoprotein
    Leukocyte elastase inhibitor ILEU_HUMAN P30740 SERPINB1 Extracellular
    Inter-alpha-trypsin inhibitor heavy chain H2 ITIH2_HUMAN P19823 ITIH2 Extracellular Glycoprotein
    Serpin B9 SPB9_HUMAN P50453 SERPINB9 Extracellular
    Metalloproteinase inhibitor 1 TIMP1_HUMAN P01033 TIMP1 Extracellular Glycoprotein
    Metalloproteinase inhibitor 2 TIMP2_HUMAN P16035 TIMP2 Extracellular
Proteases
    Angiotensin-converting enzyme ACE_HUMAN P12821 ACE Extracellular Plasma membrane Glycoprotein EC marker
    Disintegrin and metalloproteinase domain-containing protein 10 ADA10_HUMAN O14672 ADAM10 Plasma membrane Glycoprotein
    Aminopeptidase B AMPB_HUMAN Q9H4A4 RNPEP Extracellular
    Aminopeptidase N AMPN_HUMAN P15144 ANPEP Plasma membrane Glycoprotein
    Bone morphogenetic protein 1 BMP1_HUMAN P13497 BMP1 Extracellular Glycoprotein
    Cathepsin B CATB_HUMAN P07858 CTSB Extracellular Glycoprotein
    Cathepsin D CATD_HUMAN P07339 CTSD Extracellular Glycoprotein
    Cathepsin Z CATZ_HUMAN Q9UBR2 CTSZ Extracellular Glycoprotein
    Carboxypeptidase Q CBPQ_HUMAN Q9Y646 CPQ Extracellular Glycoprotein
    Dipeptidyl peptidase 2 DPP2_HUMAN Q9UHL4 DPP7 Extracellular Glycoprotein
    Dipeptidyl peptidase 3 DPP3_HUMAN Q9NY33 DPP3 Plasma membrane
    Endoplasmic reticulum aminopeptidase 1 ERAP1_HUMAN Q9NZ08 ERAP1 Extracellular Glycoprotein
    Furin FURIN_HUMAN P09958 FURIN Plasma membrane Glycoprotein
    Gamma-glutamyl hydrolase GGH_HUMAN Q92820 GGH Extracellular Glycoprotein
    Serine protease HTRA1 HTRA1_HUMAN Q92743 HTRA1 Extracellular
    Insulin-degrading enzyme IDE_HUMAN P14735 IDE Extracellular Plasma membrane
    Interstitial collagenase MMP1_HUMAN P03956 MMP1 Extracellular Glycoprotein
    Stromelysin-2 MMP10_HUMAN P09238 MMP10 Extracellular
    Matrix metalloproteinase-14 MMP14_HUMAN P50281 MMP14 Plasma membrane
    72 kDa type IV collagenase MMP2_HUMAN P08253 MMP2 Extracellular Glycoprotein
    Lysosomal Pro-X carboxypeptidase PCP_HUMAN P42785 PRCP Plasma membrane Glycoprotein
    Serine protease 23 PRS23_HUMAN O95084 PRSS23 Extracellular Glycoprotein
    Ubiquitin carboxyl-terminal hydrolase 14 UBP14_HUMAN P54578 USP14 Plasma membrane
Signal transduction proteins
    Adenylyl cyclase-associated protein 1 CAP1_HUMAN Q01518 CAP1 Plasma membrane
    Cell division control protein 42 homolog CDC42_HUMAN P60953 CDC42 Plasma membrane
    Contactin-associated protein-like 3 CNTP3_HUMAN Q9BZ76 CNTNAP3 Extracellular Plasma membrane Glycoprotein
    Adapter molecule crk CRK_HUMAN P46108 CRK Plasma membrane
    Ras GTPase-activating protein-binding protein 1 G3BP1_HUMAN Q13283 G3BP1 Plasma membrane
    Growth arrest-specific protein 6 GAS6_HUMAN Q14393 GAS6 Extracellular Glycoprotein
    Interferon-induced guanylate-binding protein 1 GBP1_HUMAN P32455 GBP1 Extracellular
    Guanine nucleotide-binding protein G(i) subunit alpha-2 GNAI2_HUMAN P04899 GNAI2 Plasma membrane
    Glypican-1 GPC1_HUMAN P35052 GPC1 Extracellular Plasma membrane Glycoprotein
    Hedgehog-interacting protein HHIP_HUMAN Q96QV1 HHIP Extracellular Plasma membrane Glycoprotein
    Histidine triad nucleotide-binding protein 1a HINT1_HUMAN P49773 HINT1 Plasma membrane
    Integrin-linked protein kinase ILK_HUMAN Q13418 ILK Plasma membrane
    Ras GTPase-activating-like protein IQGAP1 IQGA1_HUMAN P46940 IQGAP1 Plasma membrane
    cAMP-dependent protein kinase type II-alpha regulatory subunit KAP2_HUMAN P13861 PRKAR2A Plasma membrane
    Ras-related protein Rab-18 RAB18_HUMAN Q9NP72 RAB18 Plasma membrane
    Ras-related protein Rab-5C RAB5C_HUMAN P51148 RAB5C Plasma membrane
    Ras-related C3 botulinum toxin substrate 1 RAC1_HUMAN P63000 RAC1 Plasma membrane
    Ras-related protein Ral-A RALA_HUMAN P11233 RALA Plasma membrane
    Ras-related protein Rap-1b RAP1B_HUMAN P61224 RAP1B Plasma membrane
    GTPase NRas RASN_HUMAN P01111 NRAS Plasma membrane
    Ras-related protein Rab-11A RB11A_HUMAN P62491 RAB11A Plasma membrane
    Rho-related GTP-binding protein RhoC RHOC_HUMAN P08134 RHOC Plasma membrane
    Rho-associated protein kinase 2 ROCK2_HUMAN O75116 ROCK2 Plasma membrane
    Ras-related protein R-Ras2 RRAS2_HUMAN P62070 RRAS2 Plasma membrane
    Protein S100-A6a S10A6_HUMAN P06703 S100A6 Plasma membrane
    Protein S100-A10a S10AA_HUMAN P60903 S100A10 Plasma membrane
    Switch-associated protein 70 SWP70_HUMAN Q9UH65 SWAP70 Plasma membrane
    NEDD8-activating enzyme E1 regulatory subunit ULA1_HUMAN Q13564 NAE1 Plasma membrane
Transport-related proteins
    AP-2 complex subunit alpha-1 AP2A1_HUMAN O95782 AP2A1 Plasma membrane
    AP-2 complex subunit alpha-2 AP2A2_HUMAN O94973 AP2A2 Plasma membrane
    ADP-ribosylation factor 1 ARF1_HUMAN P84077 ARF1 Plasma membrane
    ADP-ribosylation factor 6 ARF6_HUMAN P62330 ARF6 Plasma membrane
    ADP-ribosylation factor-like protein 3 ARL3_HUMAN P36405 ARL3 Plasma membrane
    Beta-arrestin-1 ARRB1_HUMAN P49407 ARRB1 Plasma membrane
    Chloride intracellular channel protein 1 CLIC1_HUMAN O00299 CLIC1 Plasma membrane
    Chloride intracellular channel protein 4 CLIC4_HUMAN Q9Y696 CLIC4 Plasma membrane
    Clusterin CLUS_HUMAN P10909 CLU Extracellular Glycoprotein
    Coatomer subunit beta COPB_HUMAN P53618 COPB1 Plasma membrane
    EH domain-containing protein 1 EHD1_HUMAN Q9H4M9 EHD1 Plasma membrane
    EH domain-containing protein 2 EHD2_HUMAN Q9NZN4 EHD2 Plasma membrane
    Palmitoyl-protein thioesterase 1 PPT1_HUMAN P50897 PPT1 Extracellular Glycoprotein
    Protein S100-A13 S10AD_HUMAN Q99584 S100A13 Extracellular
    Solute carrier family 12 member 2 S12A2_HUMAN P55011 SLC12A2 Plasma membrane
    Proactivator polypeptide SAP_HUMAN P07602 PSAP Extracellular Glycoprotein
    Syntaxin-binding protein 1 STXB1_HUMAN P61764 STXBP1 Plasma membrane
    Syntaxin-binding protein 3 STXB3_HUMAN O00186 STXBP3 Plasma membrane
    Transmembrane emp24 domain-containing protein 10 TMEDA_HUMAN P49755 TMED10 Plasma membrane Glycoprotein
    Vesicle-associated membrane protein-associated protein A VAPA_HUMAN Q9P0L0 VAPA Plasma membrane

a These proteins were also identified in the cellular proteome according to the difference gel electrophoresis analysis presented in the supplemental data.

Immunoblots confirmed that proteins such as fibronectin and biglycan were constitutively secreted (Fig. 1B). Others such as agrin and lymphatic vessel endothelial hyaluronic acid receptor 1 were released upon PMA stimulation, providing an explanation for why previously unidentified proteins (8, 10) were found in the present analysis (Fig. 1C). An overlay between secreted (Cy3 and Cy 5; green and red color) and cellular (Cy 2; blue color) proteins is shown in Fig. 1D. Common spots were numbered (supplemental Fig. S2) and identified via LC-MS/MS (supplemental Table S3). Certain proteins, such as von Willebrand antigen 2 (a propeptide of vWF, AA 23–763), were clearly more abundant in the secretome of PMA-treated HUVECs.

The Endothelial Glycoproteome

Among the 1252 identified proteins were 253 extracellular or plasma membrane proteins (approximately 20%) related to cell adhesion, blood coagulation, hemostasis, signaling transduction, and protein transportation, of which 166 were known glycoproteins (Table I). To further characterize this subproteome, we employed a glycoproteomics approach. Secreted proteins were precipitated and digested with trypsin, and tryptic peptides were labeled with TMT0 to increase their charge state prior to enrichment by means of zwitterionic hydrophilic interaction liquid chromatography purification (24). For glycosite identification, an indirect and a direct strategy were pursued (Fig. 2A): (i) digestion with PNGase F in the presence of 18O water to label the conversion of asparagine to aspartic acid upon the removal of N-glycans, and (ii) alternating HCD and ETD (HCD-alt-ETD) or HCD-product-dependent ETD (HCD-pd-ETD) fragmentation on an Orbitrap Elite MS (24).

Fig. 2.

Fig. 2.

Glycoproteomics. A, glycopeptide identification workflow. Comparison of direct and indirect glycopeptide detection using HCD-ETD and 18O-deamidation after PNGase F + H218O treatment, respectively: identified unique glycopeptides (B), unique glycosylation sites (C), and unique glycoproteins (D).

There was little overlap in the numbers of glycopeptides (Fig. 2B) and glycosylation sites (Fig. 2C) identified via the direct (HCD-ETD) and the indirect (PNGase F + H218O) methods. Better agreement was observed at the protein level (Fig. 2D). With the indirect (PNGase F + H218O) method, 27 peptides were identified with N[+2.99] modification at non-consensus sequence, out of 1139 total identified peptides with N[+2.99]. This anomaly rate of 2.4% (27/1139) combines the rate of false identifications and the rate of chance deamidations in 18O water that were not in the consensus sequence of glycosylation (i.e. N-X(not P)-S/T). All glycopeptides identified are listed in Table II and supplemental Table S4. Three spectra (full MS, HCD, and ETD) from a neuronal cell adhesion molecule (UniProt accession number Q92823) (AA - 222FNHTQTIQQK231) are presented in Fig. 3.

Table II. Glycopeptides identified via the HCD-ETD method.
Protein name UniProt ID Peptide Glycosite Type Glycans Observed m/z Z ΔMass (ppm)
Afamin AFAM_HUMAN $DIENFN(+1702.582)^STQk N33 Known Hex8HexNAc2 843.359 4 −5.8
Aminopeptidase N AMPN_HUMAN $kLN(+892.318)YTLSQGHRVVLR N128 Known Hex3HexNAc2 781.916 4 −2.8
$NAN(+2042.720)^SSPVAsTTPSASATTNPASATTLDQSk N42 Novel Hex4HexNAc4NeuAc2 1095.276 5 −0.3
Alpha-N-acetylglucosaminidase ANAG_HUMAN $SVYN(+1257.450)cSGEAcRGhNRSPLVR N503 Potential Hex4HexNAc3 954.927 4 1.1
Angiopoietin-2 ANGP2_HUMAN $kIVTATVN(+568.212)NSVLQk N240 Potential Hex1HexNAc2 689.645 4 −0.3
$SGhTTNGIYTLTFPN(+1038.375)STEEIk N304 Potential Hex3HexNAc2dHex1 763.565 5 −1.4
Attractin ATRN_HUMAN $DLDMFIN(+1241.455)ASk N1198 Known Hex3HexNAc3dHex1 948.450 3 6.7
$GcScFSDWQGPGcSVPVPAN(+1095.397)QSFWTR N325 Potential Hex3HexNAc3 1077.708 4 −2.1
$GcScFSDWQGPGcSVPVPAN(+892.317)QSFWTREEYSnLk N325 Potential Hex3HexNAc2 1040.061 5 −2.4
Cadherin-2 CADH2_HUMAN $EQIARFHLRAHAVDInGNQVENPIDIVINVIDMNDNRPEFLHQVWN(+1751.624)GTVPEGSk N273 Known Hex4HexNAc4NeuAc1 930.011 9 2.8
Cadherin-5 CADH5_HUMAN $EN(+1054.370)^ISEYHLTAVIVDk N112 Known Hex4HexNAc2 815.146 4 0.7
$ELDREVYPWYN(+1241.455)LTVEAk N442 Known Hex3HexNAc3dHex1 954.972 4 10.0
CD109 antigen CD109_HUMAN $LNLYLDSVN(+1038.375)^ETQFcVNIPAVR N1355 Novel Hex3HexNAc2dHex1 938.447 4 1.5
$kkN(+1540.529)ITk N279 Potential Hex7HexNAc2 793.410 4 −0.9
$QN(+1848.640)^STMFSLTPENSWTPk N513 Novel Hex8HexNAc2dHex1 1072.719 4 1.0
CD59 glycoprotein CD59_HUMAN $TAVN(+1954.704)^csSDFDAcLITk N43 Known Hex4HexNAc5NeuAc1 1077.708 4 9.1
Complement factor I CFAI_HUMAN $FLNN(+1054.370)GTcTAEGk N103 Known Hex4HexNAc2 939.094 3 −4.0
$LISN(+3534.244)^cSk N494 Potential Hex8HexNAc6dHex1NeuAc3 1207.501 4 5.7
CAP-Gly domain-containing linker protein 1 CLIP1_HUMAN $GEN(+1257.450)^ASAk N1263 Novel Hex4HexNAc3 802.359 3 −1.4
$GEN(+1784.635)ASAk N1263 Novel Hex6HexNAc4 970.437 3 10.7
$EPSATPPISN(+2188.741)LTk N187 Novel Hex11HexNAc2 999.192 4 −6.9
$ANEN(+1200.428)^ASFLQkSIEDMTVk N971 Novel Hex4HexNAc2dHex1 980.724 4 2.6
$ANEN(+1216.423)^ASFLQkSIEDMTVk N971 Novel Hex5HexNAc2 985.232 4 11.0
$ANEN(+1257.449)ASFLQkSIEDMTVk N971 Novel Hex4HexNAc3 989.489 4 8.2
$ANEN(+1444.534)ASFLQkSIEDMTVk N971 Novel Hex3HexNAc4dHex1 1036.764 4 9.3
Ephrin type-A receptor 2 EPHA2_HUMAN $TASVSIN(+892.317)QTEPPkVRLEGR N435 Known Hex3HexNAc2 856.694 4 −0.4
Fibrous sheath-interacting protein 2 FSIP2_HUMAN $IGWEYESTN(+1751.624)ISR N1423 Novel Hex4HexNAc4NeuAc1 858.373 4 0.8
$TITFSAN(+1362.481)VSSHEhTYk N1675 Novel Hex5HexNAc2dHex1 912.923 4 3.0
$GGIN(+892.318)ISGQGSIISAQVSPTR N215 Novel Hex3HexNAc2 1020.509 3 1.5
$ENSN(+1200.428)FSQLALSNEILLGHkEk N2216 Novel Hex4HexNAc2dHex1 708.363 6 6.3
$mPIEN(+1444.534)LSSIQQk N2824 Novel Hex3HexNAc4dHex1 825.398 4 0.8
$YN(+2204.772)k N427 Novel Hex5HexNAc4NeuAc2 1026.779 3 7.7
N-acetylglucosamine-6-sulfatase GNS_HUMAN $LVkRLEFTGELN(+2018.708)^NTYIFYTSDnGYHTGQFSLPIDkR N317 Known Hex6HexNAc3dHex1NeuAc1 696.027 10 −9.6
$GPGIkPN(+1540.529)QTSk N362 Potential Hex7HexNAc2 835.657 4 −0.8
Golgin subfamily A member 4 GOGA4_HUMAN $ELEhVN(+1735.630)^LSVk N1612 Novel Hex3HexNAc4dHex1NeuAc1 848.139 4 −3.9
$kELEHVN(+1038.375)LSVk N1612 Novel Hex3HexNAc2dHex1 752.648 4 0.8
$kELEHVN(+1524.534)LSVk N1612 Novel Hex6HexNAc2dHex1 874.186 4 −1.6
$SLQENkN(+1257.450)QSk N585 Novel Hex4HexNAc3 777.391 4 10.2
$TRILELESSLEkSLQENkN(+1216.423)QSk N585 Novel Hex5HexNAc2 938.686 5 −2.3
Intercellular adhesion molecule 2 ICAM2_HUMAN $GSLEVN(+2028.741)^cSTTcNQPEVGGLETSLDk N47 Known Hex5HexNAc6 1039.861 5 5.1
$HYLVSN(+568.212)ISHDTVLQcHFTcSGk N82 Known Hex1HexNAc2 905.687 4 −0.6
ICOS ligand ICOSL_HUMAN $IARTPSVNIGccIENVLLQQN(+2457.877)LTVGSQTGNDIGER N225 Potential Hex8HexNAc5dHex1 724.219 9 −5.4
$IARTPSVNIGccIENVLLQQN(+2594.937)^LTVGSQTGNDIGER N225 Potential Hex4HexNAc6dHex1NeuAc2 667.906 10 0.1
Interleukin-6 receptor subunit beta IL6RB_HUMAN $SHLQN(+568.212)YTVNATkLTVNLTNDRYLATLTVRNLVGk N379 Known Hex1HexNAc2 725.691 7 6.5
$SHLQN(+568.212)YTVNATkLTVNLTNDRYLATLTVRNLVGk N379 Known Hex1HexNAc2 1015.568 5 9.4
$LTVN(+1460.529)LTNDRYLATLTVRNLVGk N390 Known Hex4HexNAc4 877.848 5 −8.4
$QN(+568.212)cSQHESSPDIsHFER N818 Novel Hex1HexNAc2 733.814 4 8.8
Integrin alpha-3 ITA3_HUMAN $DVRkLLLSIN(+892.317)VTNTR N656 Potential Hex3HexNAc2 1028.554 3 −2.2
$AHcVWLEcPIPDAPVVTN(+1362.481)VTVk N926 Potential Hex5HexNAc2dHex1 864.223 5 8.3
Integrin beta-1 ITB1_HUMAN $kNkNVTN(+1444.534)^RSk N97 Potential Hex3HexNAc4dHex1 888.710 4 2.8
Junctional adhesion molecule C JAM3_HUMAN $IWN(+1581.555)VTRRDSALYRcEVVARNDR N104 Potential Hex6HexNAc3 1139.528 4 −4.8
$IWN(+892.318)^VTR N104 Potential Hex3HexNAc2 643.303 3 −0.7
Lysosome-associated membrane glycoprotein 1 LAMP1_HUMAN $GHTLTLN(+1378.476)FTR N103 Known Hex6HexNAc2 921.421 3 −0.4
$GHTLTLN(+1540.529)FTR N103 Known Hex7HexNAc2 975.439 3 −0.4
$YNVSGTNGTcLLASMGLQLN(+2042.720)^LTYERkDNTTVTRLLnINPNk N241 Known Hex4HexNAc4NeuAc2 734.661 10 6.4
$kDN(+2059.735)TTVTR N249 Known Hex5HexNAc4dHex1NeuAc1 861.641 4 1.0
$kDN(+2204.772)TTVTR N249 Known Hex5HexNAc4NeuAc2 898.156 4 5.8
$kDN(+2350.830)TTVTR N249 Known Hex5HexNAc4dHex1NeuAc2 934.414 4 −0.2
$kDN(+2569.905)TTVTR N249 Known Hex6HexNAc5NeuAc2 989.440 4 6.2
$kDN(+2570.925)TTVTR N249 Known Hex6HexNAc5dHex2NeuAc1 989.690 4 1.0
$kDN(+2571.946)TTVTR N249 Known Hex6HexNAc5dHex4 989.690 4 −2.4
$kDN(+2715.963)^TTVTR N249 Known Hex6HexNAc5dHex1NeuAc2 1031.190 4 −2.0
$kDN(+2715.963)TTVTR N249 Known Hex6HexNAc5dHex1NeuAc2 1025.696 4 −0.9
$kDN(+2717.978)TTVTR N249 Known Hex8HexNAc7 1025.949 4 −1.2
$kDN(+2861.000)TTVTR N249 Known Hex6HexNAc5NeuAc3 1062.211 4 3.1
$kDN(+2864.036)TTVTR N249 Known Hex8HexNAc7dHex1 1062.466 4 1.2
$kDN(+3007.058)TTVTR N249 Known Hex6HexNAc5dHex1NeuAc3 1098.476 4 4.4
$AAN(+1702.581)GSLR N322 Known Hex8HexNAc2 872.711 3 2.6
$AAN(+1864.634)GSLR N322 Known Hex9HexNAc2 926.395 3 3.5
Lysosome-associated membrane glycoprotein 2 LAMP2_HUMAN $EkPEAGTYSVNNGN(+1054.370)DTcLLATmGLQLNITQDk N229 Known Hex4HexNAc2 1048.506 5 0.8
Lysosomal alpha-glucosidase LYAG_HUMAN $QVVEN(+1095.397)^MTRAHFPLDVQWNDLDYMDSR N390 Known Hex3HexNAc3 905.408 5 0.9
$GAYTQVIFLARN(+1054.370)NTIVNELVR N882 Known Hex4HexNAc2 918.718 4 1.4
Protein-lysine 6-oxidase LYOX_HUMAN $AEN(+1054.370)QTAPGEVPALSNLR N144 Potential Hex4HexNAc2 762.369 4 8.1
$DPGAAVPGAANASAQQPRTPILLIRDN(+2432.884)R N97 Potential Hex3HexNAc6dHex1NeuAc2 1106.316 5 −5.0
Lymphatic vessel endothelial hyaluronic acid receptor 1 LYVE1_HUMAN $AN(+1362.481)DSNPNEESkkTDk N289 Novel Hex5HexNAc2dHex1 984.727 4 5.8
Lysosomal alpha-mannosidase MA2B1_HUMAN $GFkDHFTFcQQLN(+892.317)ISIcPLSQTAARFQVIVYnPLGRk N497 Potential Hex3HexNAc2 987.504 6 −1.2
Hepatocyte growth factor receptor MET_HUMAN $TLLRN(+1751.624)^SsGcEARRDEYR N405 Potential Hex4HexNAc4NeuAc1 1040.946 4 6.4
$VIVQPDQN(+2042.720)^^FTGLIAGVVSISTALLLLLGFFLWLkkRk N930 Potential Hex4HexNAc4NeuAc2 886.225 8 3.5
$VIVQPDQN(+2174.799)^FTGLIAGVVSISTALLLLLGFFLWLkkR N930 Potential Hex5HexNAc6dHex1 1140.772 6 4.0
Interstitial collagenase MMP1_HUMAN $AFQLWSN(+2034.703)^VTPLTFTk N143 Novel Hex7HexNAc3NeuAc1 1065.992 4 3.1
$AFQLWSN(+2059.735)VTPLTFTk N143 Novel Hex5HexNAc4dHex1NeuAc1 1065.999 4 0.3
$AFQLWSN(+2075.730)^VTPLTFTk N143 Novel Hex6HexNAc4NeuAc1 1076.245 4 0.2
$AFQLWSN(+2075.730)VTPLTFTk N143 Novel Hex5HexNAc4dHex2NeuGc1 1070.250 4 1.4
$AFQLWSN(+2076.750)VTPLTFTk N143 Novel Hex6HexNAc4dHex2 1070.251 4 −1.0
$AFQLWSN(+2092.745)VTPLTFTk N143 Novel Hex7HexNAc4dHex2 1074.251 4 0.2
$AFQLWSN(+2350.830)^VTPLTFTk N143 Novel Hex5HexNAc4dHex1NeuAc2 1144.769 4 −0.3
$AFQLWSN(+2350.830)VTPLTFTk N143 Novel Hex5HexNAc4dHex1NeuAc2 1139.021 4 −1.9
$AFQLWSN(+2351.851)VTPLTFTk N143 Novel Hex5HexNAc4dHex3NeuAc1 1139.277 4 −0.9
Multimerin-1 MMRN1_HUMAN $INALkkPTVN(+1200.428)LTTVLIGR N1020 Known Hex4HexNAc2dHex1 957.031 4 1.5
$GARLFVLLSSLWSGGIGLN(+1241.455)^NSk N21 Potential Hex3HexNAc3dHex1 1001.006 4 −3.0
$LFVLLSSLWSGGIGLN(+568.212)NSk N21 Potential Hex1HexNAc2 756.162 4 −0.8
$IDN(+1095.397)^ISLTVNDVRNTYSSLEGk N344 Known Hex3HexNAc3 976.973 4 −1.1
$InN(+568.212)LTVSLEMEk N576 Potential Hex1HexNAc2 803.746 3 −1.8
$kIEN(+892.317)LTSAVNSLNFIIk N680 Potential Hex3HexNAc2 868.469 4 −3.4
$LNQSNFQkmYQMFN(+2262.815)^ETTSQVR N828 Potential Hex5HexNAc5dHex1NeuAc1 1069.470 5 2.0
$ALEAkSIHLSInFFSLN(+568.212)k N921 Potential Hex1HexNAc2 819.195 4 −5.2
$SIHLSINFFSLN(+892.318)^k N921 Potential Hex3HexNAc2 721.609 4 −6.7
C-type mannose receptor 2 MRC2_HUMAN $VTPAcN(+2059.735)TSLPAQR N69 Known Hex5HexNAc4dHex1NeuAc1 925.654 4 −2.1
$SN(+2059.735)VTk N954 Potential Hex5HexNAc4dHex1NeuAc1 1020.122 3 0.2
Nck-associated protein 5 NCKP5_HUMAN $ERGPQGQGHGRMALNLQLSDTDDN(+2018.708)^ETFDELHIESSDEk N585 Novel Hex6HexNAc3dHex1NeuAc1 1127.492 6 −3.7
Lysosomal protein NCU-G1 NCUG1_HUMAN $LLHTADTcQLEVALIGAsPRGN(+1241.454)R N230 Potential Hex3HexNAc3dHex1 1010.223 4 −1.4
Natural cytotoxicity triggering receptor 3 ligand 1 NR3L1_HUMAN $LN(+2172.745)SSQEDPGTVYQcVVRHASLHTPLR N216 Potential Hex10HexNAc2dHex1 1073.477 5 −4.3
Neuronal cell adhesion molecule NRCAM_HUMAN $IPAN(+2715.963)k N1009 Potential Hex6HexNAc5dHex1NeuAc2 927.404 4 −1.4
$IPAN(+2960.068)^^k N1009 Potential Hex5HexNAc7dHex1NeuAc2 1000.181 4 6.5
$FN(+1378.476)HTQTIQQk N223 Potential Hex6HexNAc2 768.611 4 0.9
$FN(+1378.476)HTQTIQQk N223 Potential Hex6HexNAc2 1024.480 3 2.2
$SSRERPPTFLTPEGN(+1710.598)ASNk N276 Known Hex5HexNAc3NeuAc1 1062.994 4 −0.4
Nuclear receptor-interacting protein 3 NRIP3_HUMAN $LMETN(+568.212)LSk N72 Novel Hex1HexNAc2 651.345 3 8.7
Plasminogen activator inhibitor 1 PAI1_HUMAN $GN(+1694.603)^MTRLPRLLVLPkFSLETEVDLRk N288 Known Hex4HexNAc3dHex1NeuAc1 1063.953 5 3.4
$GN(+2059.735)MTR N288 Known Hex5HexNAc4dHex1NeuAc1 955.394 3 0.5
$GN(+2350.830)^mTR N288 Known Hex5HexNAc4dHex1NeuAc2 799.313 4 −3.7
$GN(+2351.851)^mTR N288 Known Hex5HexNAc4dHex3NeuAc1 798.814 4 −5.6
Platelet-derived growth factor subunit B PDGFB_HUMAN $LLHGDPGEEDGAELDLN(+1864.634)mTRSHSGGELESLARGRR N63 Novel Hex9HexNAc2 1176.921 5 −5.5
Secretory phospholipase A2 receptor PLA2R_HUMAN $MQDTSGHGVNTsDMYPMPNTLEYGN(+2204.772)^^RTYk N1123 Novel Hex5HexNAc4NeuAc2 1014.913 6 −1.9
Procollagen-lysine,2-oxoglutarate 5-dioxygenase 2 PLOD2_HUMAN $YFN(+1856.656)YTVkVLGQGEEWR N63 Potential Hex5HexNAc3dHex1NeuAc1 1074.744 4 0.2
Phospholipid transfer protein PLTP_HUMAN $qLLYWFFYDGGYIN(+2157.783)^ASAEGVSIRTGLELSR N117 Potential Hex4HexNAc6NeuAc1 1118.898 5 0.2
$IYSN(+1403.507)HSALESLALIPLQAPLk N398 Known Hex4HexNAc3dHex1 1033.782 4 1.5
Plexin-C1 PLXC1_HUMAN $IAN(+1524.534)^FTSDVEYSDDHchLILPDSEAFQDVQGkRHR N1308 Novel Hex6HexNAc2dHex1 1203.342 5 −1.8
$IAN(+2204.773)FTSDVEYSDDhcHLILPDSEAFQDVQGkR N1308 Novel Hex5HexNAc4NeuAc2 638.689 10 5.9
$VILGEN(+1298.476)LTSNcPEVIYEIk N407 Known Hex3HexNAc4 985.483 4 −1.8
$ELcQN(+1378.476)k N548 Potential Hex6HexNAc2 873.730 3 8.0
$DVcIQFDGGNcSSVGSLSYIALPHcSLIFPATTWISGGQN(+1403.508)^ITMMGR N771 Potential Hex4HexNAc3dHex1 607.279 11 0.5
$ENDNFN(+1054.370)ISk N871 Novel Hex4HexNAc2 861.725 3 −4.1
$ENDNFN(+2522.916)^ISk N871 Novel Hex5HexNAc7NeuAc1 1019.438 4 7.5
Tyrosine-protein kinase-like 7 PTK7_HUMAN $SAN(+3007.058)ASFNIk N116 Known Hex6HexNAc5dHex1NeuAc3 1102.722 4 2.1
$SAN(+3026.090)ASFNIk N116 Known Hex9HexNAc7dHex1 1107.479 4 1.3
$ATVFAN(+1710.598)GSLLLTQVRPR N283 Known Hex5HexNAc3NeuAc1 945.458 4 0.0
$RQDVN(+1686.586)ITVATVPSWLk N405 Potential Hex7HexNAc2dHex1 991.485 4 3.2
Pentraxin-related protein PTX3 PTX3_HUMAN $ATDVLN(+1419.502)kTILFsYGTk N220 Potential Hex5HexNAc3 986.474 4 −4.0
Proactivator polypeptide SAP_HUMAN $TN(+1038.375)STFVQALVEHVk N215 Known Hex3HexNAc2dHex1 1020.847 3 1.2
$LIDNN(+1216.423)kTEk N332 Known Hex5HexNAc2 988.825 3 −0.7
$LIDNN(+1378.476)kTEk N332 Known Hex6HexNAc2 1042.510 3 0.7
$LIDNN(+1694.603)kTEk N332 Known Hex4HexNAc3dHex1NeuAc1 861.415 4 −0.8
$N(+2245.800)^STk N426 Known Hex4HexNAc5NeuAc2 1056.113 3 −0.8
$NLEkN(+1378.476)STk N426 Known Hex6HexNAc2 995.485 3 2.5
Histone deacetylase complex subunit SAP30 SAP30_HUMAN $N(+1095.397)kSDLk N209 Novel Hex3HexNAc3 824.751 3 −3.7
$GGDAAAAVAAVVAAAAAAAsAGN(+1589.571)GTGAGTGAEVPGAGAVSAAGPPGAAGPGPGQLccLR N34 Novel Hex3HexNAc4NeuAc1 682.815 10 −0.7
Serpin H1 SERPH_HUMAN $SLSN(+1378.476)^STAR N120 Potential Hex6HexNAc2 821.017 3 −1.4
$SLSN(+1378.476)STAR N120 Potential Hex6HexNAc2 813.358 3 1.9
$SLSN(+1540.529)STAR N120 Potential Hex7HexNAc2 867.371 3 −3.6
$SLSN(+1702.581)STAR N120 Potential Hex8HexNAc2 922.062 3 1.6
$SLSN(+1864.634)STAR N120 Potential Hex9HexNAc2 975.414 3 4.5
$N(+1540.529)VTWk N125 Potential Hex7HexNAc2 879.399 3 −0.5
$N(+1702.581)VTWk N125 Potential Hex8HexNAc2 933.418 3 0.6
Tyrosine-protein phosphatase non-receptor type substrate 1 SHPS1_HUMAN $LQLTWLEnGN(+1200.428)VSR N292 Known Hex4HexNAc2dHex1 739.847 4 −3.5
$LQLTWLENGN(+1872.651)^VSR N292 Known Hex6HexNAc3NeuAc1 912.893 4 −11.5
SPARC SPRC_HUMAN $VcSNDN(+1622.582)k N116 Known Hex5HexNAc4 970.425 3 3.5
$VcSNDN(+1767.619)k N116 Known Hex4HexNAc4NeuGc1 1019.107 3 4.9
$VcSNDN(+1768.640)k N116 Known Hex5HexNAc4dHex1 1018.439 3 0.0
$VcSNDN(+1864.634)k N116 Known Hex9HexNAc2 1051.108 3 2.2
$VcSNDN(+1913.677)^k N116 Known Hex5HexNAc4NeuAc1 806.336 4 −1.4
$VcSNDN(+1913.677)k N116 Known Hex5HexNAc4NeuAc1 800.591 4 −0.3
$VcSNDN(+1913.677)k N116 Known Hex5HexNAc4NeuAc1 1066.783 3 −1.7
$VcSNDN(+1914.697)k N116 Known Hex5HexNAc4dHex2 800.844 4 −2.5
$VcSNDN(+1914.697)k N116 Known Hex5HexNAc4dHex2 1067.125 3 0.6
$VcSNDN(+2059.735)^k N116 Known Hex5HexNAc4dHex1NeuAc1 842.598 4 −3.0
$VcSNDN(+2059.735)k N116 Known Hex5HexNAc4dHex1NeuAc1 836.857 4 3.1
$VcSNDN(+2059.735)k N116 Known Hex5HexNAc4dHex1NeuAc1 1115.472 3 1.6
$VcSNDN(+2350.830)k N116 Known Hex5HexNAc4dHex1NeuAc2 909.629 4 0.3
Stabilin-1 STAB1_HUMAN $ELLQHHGLVPQIEAATAYTIFVPTnRSLEAQGN(+2366.825)^SSHLDADTVR N1178 Potential Hex6HexNAc4NeuAc2 732.443 10 3.2
$ELkGDGPFTIFVPHADLMSN(+568.212)LSQDELARIR N1626 Known Hex1HexNAc2 1097.310 4 −5.4
$LLPAHsGLSLIISDAGPDN(+892.317)SSWAPVAPGTVVVSR N2424 Known Hex3HexNAc2 655.464 7 −7.8
$ILTMAnQVLAVN(+1095.397)ISEEGR N606 Potential Hex3HexNAc3 820.390 4 −9.4
$GN(+1216.423)cSDGIQGNGAcLcFPDYk N745 Potential Hex5HexNAc2 975.155 4 −10.2
Suppressor of G2 allele of SKP1 homolog SUGT1_HUMAN $RAMN(+568.212)kSFMESGGTVLSTNWsDVGk N329 Novel Hex1HexNAc2 981.480 4 6.2
Angiopoietin-1 receptor TIE2_HUMAN $ISN(+568.212)ITHSSAVIsWTILDGYSISSITIR N649 Potential Hex1HexNAc2 762.382 5 −3.1
Tetraspanin-3 TSN3_HUMAN $TYN(+1825.661)GTNPDAASRAIDYVQR N127 Potential Hex5HexNAc5 1041.454 4 −7.9
Thrombospondin-1 TSP1_HUMAN $VVN(+1913.677)STTGPGEHLR N1067 Known Hex5HexNAc4NeuAc1 877.141 4 0.2
Vascular endothelial growth factor receptor 1 VGFR1_HUMAN $RIIWDSRkGFIISN(+892.317)ATYk N196 Potential Hex3HexNAc2 934.508 4 8.1
$SVN(+892.318)TSVhIYDkAFITVk N323 Potential Hex3HexNAc2 876.466 4 10.2
$WFWHPcNHN(+1419.502)HSEARcDFcSNNEESFILDADSnMGNR N474 Potential Hex5HexNAc3 1017.415 6 1.3
$mAITkEhSITLNLTIMN(+1257.450)^VSLQDSGTYAcRAR N625 Potential Hex4HexNAc3 1051.702 5 −0.2
von Willebrand factor VWF_HUMAN $YFN(+2059.735)^k N156 Potential Hex5HexNAc4dHex1NeuAc1 776.330 4 −4.7
$YFN(+2059.735)^k N156 Potential Hex5HexNAc4dHex1NeuAc1 1034.440 3 −1.2
$YFN(+2059.735)k N156 Potential Hex5HexNAc4dHex1NeuAc1 1027.114 3 −0.1
$YFN(+2059.736)k N156 Potential Hex5HexNAc4dHex1NeuAc1 770.587 4 −0.6
$YFN(+2204.772)k N156 Potential Hex5HexNAc4NeuAc2 1075.798 3 3.0
$YFN(+2221.789)k N156 Potential Hex6HexNAc4dHex1NeuAc1 1081.467 3 0.8
$YFN(+2350.830)^^k N156 Potential Hex5HexNAc4dHex1NeuAc2 854.851 4 −3.2
$YFN(+2350.830)^k N156 Potential Hex5HexNAc4dHex1NeuAc2 848.856 4 −0.9
$YFN(+2350.830)^k N156 Potential Hex5HexNAc4dHex1NeuAc2 1131.473 3 −0.2
$YFN(+2350.830)k N156 Potential Hex5HexNAc4dHex1NeuAc2 843.362 4 0.5
$YFN(+2350.830)k N156 Potential Hex5HexNAc4dHex1NeuAc2 1124.145 3 −0.7
$YFN(+2350.831)^k N156 Potential Hex5HexNAc4dHex1NeuAc2 848.857 4 0.0
$YFN(+2350.831)k N156 Potential Hex5HexNAc4dHex1NeuAc2 843.362 4 1.2
$YFN(+2351.851)^k N156 Potential Hex5HexNAc4dHex3NeuAc1 849.613 4 −0.2
$YFN(+2366.825)^k N156 Potential Hex5HexNAc4dHex1NeuAc1NeuGc1 853.098 4 −9.4
$YFN(+2366.825)k N156 Potential Hex5HexNAc4dHex1NeuAc1NeuGc1 1129.813 3 0.5
$YFN(+2715.963)^k N156 Potential Hex6HexNAc5dHex1NeuAc2 940.387 4 −3.7
$YFN(+2715.963)k N156 Potential Hex6HexNAc5dHex1NeuAc2 934.643 4 −0.8
$YFN(+2716.983)k N156 Potential Hex6HexNAc5dHex3NeuAc1 934.898 4 −1.5
$YFN(+2861.001)k N156 Potential Hex6HexNAc5NeuAc3 971.409 4 3.7
$YFN(+3007.058)k N156 Potential Hex6HexNAc5dHex1NeuAc3 1007.667 4 −2.2
$YFN(+3009.074)k N156 Potential Hex8HexNAc7 1007.923 4 0.6
$YFN(+3026.089)k N156 Potential Hex9HexNAc7dHex1 1012.676 4 −2.0
$YFN(+3372.190)k N156 Potential Hex7HexNAc6dHex1NeuAc3 1098.951 4 −0.9
$GDILQRVREIRYQGGN(+1378.476)R N1574 Known Hex6HexNAc2 909.439 4 −0.2
$GDILQRVREIRYQGGN(+568.212)RTNTGLALR N1574 Known Hex1HexNAc2 912.984 4 −4.0
$YQGGN(+2059.735)R N1574 Known Hex5HexNAc4dHex1NeuAc1 993.411 3 1.2
$ASPPSSScN(+2059.735)ISSGEmQk N211 Potential Hex5HexNAc4dHex1NeuAc1 1073.955 4 −2.4
$ASPPSSScN(+2350.830)^ISSGEmQk N211 Potential Hex5HexNAc4dHex1NeuAc2 1151.736 4 8.9
$ASPPSSScN(+2350.830)ISSGEmQk N211 Potential Hex5HexNAc4dHex1NeuAc2 1146.231 4 0.3
$N(+2059.735)VScPQLEVPVcPSGFQLSck N2546 Known Hex5HexNAc4dHex1NeuAc1 984.037 5 −3.6
$GQVYLQcGTPcN(+2059.735)LTcR N666 Potential Hex5HexNAc4dHex1NeuAc1 1053.444 4 0.9
$GQVYLQcGTPcN(+2075.730)LTcR N666 Potential Hex6HexNAc4NeuAc1 1057.695 4 2.2
$GQVYLQcGTPcN(+2076.750)LTcR N666 Potential Hex6HexNAc4dHex2 1058.193 4 −0.6
$GQVYLQcGTPcN(+2350.830)LTcR N666 Potential Hex5HexNAc4dHex1NeuAc2 1126.222 4 4.5
Bovine proteins
    Alpha-1-acid glycoprotein A1AG_BOVIN $QN(+2861.000)^GTLSk N104 Potential Hex6HexNAc5NeuAc3 1020.428 4 0.4
$QN(+2861.000)GTLSk N104 Potential Hex6HexNAc5NeuAc3 1014.934 4 1.8
$QN(+2880.031)GTLSk N104 Potential Hex9HexNAc7 1019.690 4 0.5
$NPEYN(+2814.010)^^k N57 Potential Hex5HexNAc7NeuAc2 1018.923 4 6.2
    Alpha-fetoprotein FETA_BOVIN $AEN(+1200.428)ATEcFETk N197 Potential Hex4HexNAc2dHex1 983.428 3 −5.8
$AEN(+2204.772)ATEcFETk N197 Potential Hex5HexNAc4NeuAc2 988.913 4 0.1
$AN(+2204.772)FTEIQk N251 Potential Hex5HexNAc4NeuAc2 901.651 4 2.5
    Alpha-2-HS-glycoprotein FETUA_BOVIN $kLcPDcPLLAPLN(+2204.772)DSR N156 Known Hex5HexNAc4NeuAc2 905.409 5 0.3
$kLcPDcPLLAPLN(+2861.000)DSR N156 Known Hex6HexNAc5NeuAc3 1036.654 5 −1.0
$LcPDcPLLAPLN(+2204.772)DSR N156 Known Hex5HexNAc4NeuAc2 1043.195 4 −1.7

Peptide modification symbols: $, N-terminal TMT0 labelling (+224.152); ^, Na adduct on glycan (+21.982); ^^, 2 Na adduct on glycan (+43.964); c, carbamidomethylation of cysteine (+57.021); h, oxidation of histidine (+15.995); k, TMT0 labeling of lysine (+224.152); m, oxidation of methionine (+15.995); n, deamidation of asparagine (+0.984); q, Gln->pyro-Glu and loss of TMT0 (−241.179); s, phosphorylation of serine (+79.966).

Underlined glycopeptides were also detected via the PNGase F + H218O method (supplemental Table S4).

Fig. 3.

Fig. 3.

HCD-pd-ETD fragmentation. Full MS showing the different glycoforms of the same peptide sequence (A). Characteristic oxonium ion detected by HCD at m/z = 204.09 (B). This HexNAc signature triggered an ETD scan to identify the peptide sequence and confirm the glycosylation site (C).

For the same samples, HCD-pd-ETD revealed 28 known, 25 potential, and 16 novel glycosylation sites based on 209 identified spectra; HCD-alt-ETD revealed 20 known, 32 potential, and 14 novel glycosylation sites from 110 identified spectra. The HCD-alt-ETD method selected mostly precursors with higher intensities, higher charge, and smaller m/z (Fig. 4A). Several large glycopeptides were detected via only HCD-alt-ETD, and more low-abundant glycopeptides were detected via HCD-pd-ETD. There was limited overlap in the identified glycopeptides but better agreement in the protein level (Fig. 4B). Among the 319 total glycopeptides identified in the conditioned media, 31 were attached with a trimannosyl core (-HexNAc2Hex3) or truncated core (-HexNAc2Hex), 50 with high mannose (-HexNAc2Hex4–9), and 238 with complex/hybrid glycans. Notably, HCD-pd-ETD detected almost twice as many complex/hybrid glycoforms as HCD-alt-ETD (Fig. 4C).

Fig. 4.

Fig. 4.

Comparison of HCD-pd-ETD and HCD-alt-ETD. The two methods, HCD-pd-ETD (blue) and HCD-alt-ETD (red), displayed distinct distributions of the observed m/z, charge state, mass of identified peptides (M+H), and glycan mass, as well as the intensity of the precursor ions and the ByonicsTM score (all y-axes). The x-axes represent index numbers after proteins were sorted by their corresponding y-axis value from lower to higher (A). There was limited overlap in the identified glycopeptides (B). C, the HCD-pd-ETD method preferentially identified complex/hybrid glycans.

Validation of Glycoproteins

To validate the glycosylation status, we performed additional analysis before and after glycoprotein enrichment with affinity resins of ConA lectin (n = 4) using a Q Exactive MS (Thermo Scientific). We then compared the number of identified spectra in the glycoprotein-enriched fraction, the flow-through, and the input (supplemental Table S5). For most glycoproteins, a higher spectral count was observed in the glycoprotein-enriched fraction than in the original input and/or the flow-through. Representative examples (fibronectin, neuronal cell adhesion molecule, tyrosine-protein-kinase-like 7, and vWF) are shown in Fig. 5A. Non-glycosylated proteins, such as annexin A2 and alpha-enolase, were more abundant in the flow-through. Glycoproteins identified in all three methods are highlighted in Fig. 5B.

Fig. 5.

Fig. 5.

Glycoprotein enrichment for validation. A, spectral count of input, glycoprotein-enriched fraction (GP), and flow-through fraction (FT) from representative glycoproteins and non-glycoproteins. B, complementarity of the different methods (HCD-ETD, PNGase F + H218O treatment, and glycoprotein enrichment). Only 18 glycoproteins were consistently identified.

Confirmation of Predicted Glycosylation Sites

The hemostatic protein vWF is the main protein stored within Weibel-Palade bodies (27). After secretagogue stimulation, Weibel-Palade bodies undergo exocytosis, releasing vWF filaments. vWF is one of the few known proteins containing the ABO blood group signature, which is formed by different glycans. Although the released glycan composition of this protein has been investigated extensively (28, 29), experimental evidence for many putative glycosylation sites is still missing. The coverage obtained for vWF in our proteomics analysis is shown in Fig. 6A. The precursor protein consists of homologous units such as the VWF type A, C, and D domains and a C-terminal cystine know (CTCK). The vWF propeptide (D1-D2, AA 23–763) is separated from the remaining domains of mature vWF (AA 764–2813) via furin-mediated proteolytic cleavage. We confirmed 6 N-glycosylation sites. Notably, three N-glycosylation sites were located within the propeptide (AA 23–763). Examples of ETD spectra are shown in Fig. 6B.

Fig. 6.

Fig. 6.

Sequence coverage for vWF. A, schematic illustration of vWF sequence. Coverage is highlighted in green, and potential glycosylation sites are shown in red. A large hexagon indicates a glycosylation site with a reference in the Uniprot database. By using the HCD-ETD (H) or PNGase F (P) method, we confirmed six N-glycosylation sites on vWF. B, ETD spectra of glycopeptides identified via HCD-ETD (N156, N211, N666, N1574). The following abbreviations are used: a, y, g, k = TMT modified Ala, Tyr, Gly, and Lys, respectively; c = carboxyamidomethylation of Cys; m = oxidation of Met.

DISCUSSION

This study represents a significant advance over the existing proteomics literature on ECs. Unlike other cell types, ECs do not tolerate prolonged serum starvation, and their susceptibility to cell death upon serum withdrawal poses a major challenge for proteomic workflows targeting their secretome. We performed secretome analysis after 45 min of PMA stimulation combined with enrichment strategies for glycoproteins and glycopeptides. Glycopeptides were analyzed via three complementary MS techniques: the detection of 18O asparagine deamidation after digestion with PNGase F in H218O, HCD-alt-ETD, and HCD-pd-ETD using an Orbitrap Elite MS.

The Endothelial Secretome

The secretagogue PMA minimized EC death by allowing a shorter incubation period under serum-free conditions while increasing coverage in the proteomic analysis by inducing the exocytosis of intracellular storage vesicles (14) such as Weibel-Palade bodies. These unique storage vesicles in ECs play a major role in hemostasis and cell-to-cell communication. Using this approach, many more proteins were identified than in any previous proteomics study on ECs, including known endothelial surface markers such as endoglin (CD105), integrin beta-1 (CD29), tyrosine-protein kinase receptor Tie-1, and junctional adhesion molecule A; secreted growth factors (i.e. C-type lectin domain family 11 member A); co-receptors (i.e. neuropilin-1 (co-receptor for VEGF-A)); proteases(i.e. furin); and inflammatory mediators (i.e. macrophage migration inhibitory factor), to name just a few. Short-term PMA treatment does not release microparticles (30), as shedding events make it difficult to discern intracellular from secreted/membrane proteins. In a direct comparison of the cellular proteome and the secretome utilizing difference gel electrophoresis, 70 out of 96 proteins analyzed were present in both samples, representing <10% of the visible protein spots in the secretome.

Biological Importance of Glycosylation

Glycosylation is key for the stability and solubility of secreted and membrane proteins. It is the most complex post-translational modification (31) and mediates extracellular matrix network assembly, cell–cell interactions, and cell–matrix interactions. Unlike polynucleotides and polypeptides, which have a linear structure, sugars tend to be arranged in branched polymers, resulting in an exponential increase of possible polysaccharide combinations. Theoretically, just six monosaccharides can give rise to 1012 different glycan structures. This high diversity of protein-bound glycans requires a combination of different techniques. For example, new MS-based methods were developed to profile the cell surface N-glycoproteome as a differentiation marker for stem cells (32). We applied a combination of different glycoproteomics techniques to further enrich for secreted and shed membrane proteins and reveal potential glycosylation sites within the endothelial secretome. Glycoproteins play important roles in many biological processes related to ECs, such as angiogenesis, in which the structural change of the glycans will determine the attachment property of cells and influence cell-to-cell interactions (33). Interestingly, vWF is a glycoprotein produced uniquely by ECs and megakaryocytes. Previous publications investigating vWF isolated from plasma failed to identify glycosylation sites within the propeptide (29). In plasma, the concentration of the propeptide is about one-tenth of the concentration of mature vWF (34, 35). In the conditioned medium of ECs, however, we observed several glycopeptides of the propeptide. Thus, the endothelial secretome allowed us to interrogate the glycosylation sites of von Willebrand antigen 2, the N-terminal cleavage product of vWF that aids N-terminal multimerization and protein compartmentalization of mature vWF in storage granules.

Conventional Methods for Glycoproteomics

As reviewed elsewhere (36), conventional glycoproteomic methods involve the enrichment of glycoproteins (typically with lectins like ConA and wheat germ agglutinin), cleavage of the glycans, and identification of the remaining peptide sequence. The most widely used method for detecting N-glycopeptides is digestion by PNGase F. PNGase F cleaves the GlcNAc molecule closest to the peptide (37). After PNGase F treatment, formerly N-linked glycosylated peptides are identified based on the conversion of Asn to Asp (deamidation) in the consensus motif for N-linked glycosylation (sequence N-X(not P)-S/T). This method has two major caveats. The first of these is a high false positive rate due to spontaneous deamidation. Asn-Gly sites, in particular, are prone to spontaneous deamidation (3840). To reduce false positives, PNGase F treatment is performed in 18O water, adding a larger tag of 2.99 Da. Importantly, all known glycosyltransferases that mediate N-linked glycosylation are supposed to recognize a consensus motif, and this consensus sequence for N-linked glycosylation must be taken into consideration (41). 2) The second caveat is that after PNGase F cleavage, the released sugars can be analyzed separately, but the link to the identified peptides with deamidated amino acids is lost (42, 43). Ideally, intact glycopeptides are analyzed directly via MS/MS even in complex biological samples.

Novel HCD-ETD Method

HCD fragmentation mostly breaks glycosidic bonds, whereas ETD preserves the glycan attachment and fragments the peptide backbone, providing more complete peptide sequence information. Current MS/MS acquisition strategies for glycopeptide analysis rely on the acquisition of MS/MS spectra for all precursor ions. In this study, HCD was employed to generate glycan oxonium ions and trigger an ETD spectrum in a data-dependent manner. HCD presents the sugar signatures within the low m/z range, which are otherwise lost as a result of the one-third rule of ion trap fragmentation (44). Glycopeptides with terminal HexNAc generate typically an m/z 204.0864 oxonium ion and its fragments at m/z 168.0653 and 138.0550. The oxonium ion and its fragments are measured with the high mass accuracy of the Orbitrap analyzer, and the unambiguous identification of the glycan oxonium ion generated by the HCD scan serves as a diagnostic marker for glycopeptides. This approach was compared against conventional HCD-alt-ETD scans using a complex biological sample. The HCD-alt-ETD preferentially detects higher charged and higher intensity precursor ions than HCD-pd-ETD. This might be because (i) a higher charge increases ETD fragmentation efficiency, resulting in more identified glycopeptides; (ii) high-charged precursors did not produce HCD spectra of sufficient quality to trigger ETD based on the diagnostic oxonium ions; or (iii) more abundant peptides were selected in HCD-alt-ETD because the instrument duty cycle is less efficient than in HCD-pd-ETD. Overall, the combination of multiple MS methods used in our study provides greater confidence in the identification of glycopeptides than studies relying on a single approach and offers complementary advantages in the assessment of the glycoproteome, notably, the simultaneous identification of the peptide sequence, the glycosylation site, and the glycan composition.

Study Limitations

N-linked and O-linked glycosylation are the two most common forms of glycosylation in mammals (45). Only N-linked glycosylation was analyzed in the present study. Unlike N-linked glycosylation, O-linked glycosylation has no consensus site (46). This makes the analysis of O-linked glycopeptides a more daunting task (47). Lectins are widely used for glycoprotein enrichment. There are many types of lectins binding to different sugars, such as ConA (binds to α-d-mannosyl and α-d-glucosyl residues) and wheat germ agglutinin (binds to GlcNAcβ1–4GlcNAcβ1–4GlcNAc- and N-acetylneuraminic acid). Here we used only ConA as a proof of principle to demonstrate the complementary results of multiple glycoprotein identification methods. ConA is known to display nonspecific avidity for hydrophobic ligands such as certain domains of tropomyosin (48). Furthermore, the standard protocol for the ConA glycoprotein enrichment kit is not optimized for cleanliness, and several known non-glycoproteins were also detected in the eluate samples. Sequential washes with low- and high-ionic-strength buffers before elution might have reduced this contamination (49). Also, mixing different lectins would increase the coverage of the glycoproteome in biological samples (39). Additional efforts are needed for a complete structural characterization of protein glycosylation; in particular, the quantitation of the occupancy rates and the identification of the glycan structure as complex/hybrid glycans cannot be discerned via our current MS approach.

CONCLUSIONS

Cardiovascular diseases arise from exposure to risk factors that induce complex pathophysiological perturbations of endothelial protein secretion. The recent advent of new proteomic technologies has enabled us to obtain information on the dynamic regulation of endothelial protein secretion. We present results from an extensive glycoproteomic analysis with information on glycan composition obtained via a direct MS method. Future proteomics studies linking endothelial secretory processes to cardiovascular risk factors and endothelial dysfunction will provide valuable insights about the mechanisms contributing to cardiovascular disease.

Supplementary Material

Supplemental Table 1

Acknowledgments

We thank Dr. Sarah Langley for assistance with the gene ontology annotation.

Footnotes

* This work was funded by the Department of Health via a National Institute for Health Research (NIHR) Biomedical Research Centre award to Guy's and St. Thomas' NHS Foundation Trust in partnership with King's College London and King's College Hospital NHS Foundation Trust. Dr. M. Mayr is supported by a Senior Research Fellowship of the British Heart Foundation.

Inline graphic This article contains supplemental material.

1 The abbreviations used are:

ConA
concanavalin A
EC
endothelial cell
ETD
electron-transfer dissociation
GlcNAc
N-acetylglucosamine
HCD
higher-energy C-trap dissociation
Hex
hexose
HexNAc
N-acetylhexosamine
HUVEC
human umbilical vein endothelial cell
PMA
phorbol-12-myristate-13-acetate
PNGase F
peptide: N-glycosidase F
TMT0
Tandem Mass Tag Zero
vWF
von Willebrand factor.

REFERENCES

  • 1. Mayr M., Mayr U., Chung Y. L., Yin X., Griffiths J. R., Xu Q. (2004) Vascular proteomics: linking proteomic and metabolomic changes. Proteomics 4, 3751–3761 [DOI] [PubMed] [Google Scholar]
  • 2. Pula G., Perera S., Prokopi M., Sidibe A., Boulanger C. M., Mayr M. (2008) Proteomic analysis of secretory proteins and vesicles in vascular research. Proteomics Clin. Appl. 2, 882–891 [DOI] [PubMed] [Google Scholar]
  • 3. Pober J. S., Sessa W. C. (2007) Evolving functions of endothelial cells in inflammation. Nat. Rev. Immunol. 7, 803–815 [DOI] [PubMed] [Google Scholar]
  • 4. Cai H., Harrison D. G. (2000) Endothelial dysfunction in cardiovascular diseases: the role of oxidant stress. Circ. Res. 87, 840–844 [DOI] [PubMed] [Google Scholar]
  • 5. Libby P., Simon D. I. (2001) Inflammation and thrombosis: the clot thickens. Circulation 103, 1718–1720 [DOI] [PubMed] [Google Scholar]
  • 6. Ross R. (1999) Atherosclerosis—an inflammatory disease. N. Engl. J. Med. 340, 115–126 [DOI] [PubMed] [Google Scholar]
  • 7. Pellitteri-Hahn M. C., Warren M. C., Didier D. N., Winkler E. L., Mirza S. P., Greene A. S., Olivier M. (2006) Improved mass spectrometric proteomic profiling of the secretome of rat vascular endothelial cells. J. Proteome Res. 5, 2861–2864 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Tunica D. G., Yin X., Sidibe A., Stegemann C., Nissum M., Zeng L., Brunet M., Mayr M. (2009) Proteomic analysis of the secretome of human umbilical vein endothelial cells using a combination of free-flow electrophoresis and nanoflow LC-MS/MS. Proteomics 9, 4991–4996 [DOI] [PubMed] [Google Scholar]
  • 9. Flora J. W., Edmiston J., Secrist R., Li G., Rana G. S., Langston T. B., McKinney W. (2008) Identification of in vitro differential cell secretions due to cigarette smoke condensate exposure using nanoflow capillary liquid chromatography and high-resolution mass spectrometry. Anal. Bioanal. Chem. 391, 2845–2856 [DOI] [PubMed] [Google Scholar]
  • 10. Burghoff S., Schrader J. (2011) Secretome of human endothelial cells under shear stress. J. Proteome Res. 10, 1160–1169 [DOI] [PubMed] [Google Scholar]
  • 11. Hocking S. L., Wu L. E., Guilhaus M., Chisholm D. J., James D. E. (2010) Intrinsic depot-specific differences in the secretome of adipose tissue, preadipocytes, and adipose tissue-derived microvascular endothelial cells. Diabetes 59, 3008–3016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Griffoni C., Di Molfetta S., Fantozzi L., Zanetti C., Pippia P., Tomasi V., Spisni E. (2011) Modification of proteins secreted by endothelial cells during modeled low gravity exposure. J. Cell. Biochem. 112, 265–272 [DOI] [PubMed] [Google Scholar]
  • 13. Brioschi M., Lento S., Tremoli E., Banfi C. (2013) Proteomic analysis of endothelial cell secretome: a means of studying the pleiotropic effects of Hmg-CoA reductase inhibitors. J. Proteomics 78, 346–361 [DOI] [PubMed] [Google Scholar]
  • 14. Castagna M., Takai Y., Kaibuchi K., Sano K., Kikkawa U., Nishizuka Y. (1982) Direct activation of calcium-activated, phospholipid-dependent protein kinase by tumor-promoting phorbol esters. J. Biol. Chem. 257, 7847–7851 [PubMed] [Google Scholar]
  • 15. Jacobs J. M., Waters K. M., Kathmann L. E., Camp D. G., 2nd, Wiley H. S., Smith R. D., Thrall B. D. (2008) The mammary epithelial cell secretome and its regulation by signal transduction pathways. J. Proteome Res. 7, 558–569 [DOI] [PubMed] [Google Scholar]
  • 16. Zampetaki A., Kiechl S., Drozdov I., Willeit P., Mayr U., Prokopi M., Mayr A., Weger S., Oberhollenzer F., Bonora E., Shah A., Willeit J., Mayr M. (2010) Plasma microRNA profiling reveals loss of endothelial miR-126 and other microRNAs in type 2 diabetes. Circ. Res. 107, 810–817 [DOI] [PubMed] [Google Scholar]
  • 17. Margariti A., Zampetaki A., Xiao Q., Zhou B., Karamariti E., Martin D., Yin X., Mayr M., Li H., Zhang Z., De Falco E., Hu Y., Cockerill G., Xu Q., Zeng L. (2010) Histone deacetylase 7 controls endothelial cell growth through modulation of beta-catenin. Circ. Res. 106, 1202–1211 [DOI] [PubMed] [Google Scholar]
  • 18. Yin X., Cuello F., Mayr U., Hao Z., Hornshaw M., Ehler E., Avkiran M., Mayr M. (2010) Proteomics analysis of the cardiac myofilament subproteome reveals dynamic alterations in phosphatase subunit distribution. Mol. Cell. Proteomics 9, 497–509 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Didangelos A., Yin X., Mandal K., Baumert M., Jahangiri M., Mayr M. (2010) Proteomics characterization of extracellular space components in the human aorta. Mol Cell. Proteomics 9, 2048–2062 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20. Mayr M., Grainger D., Mayr U., Leroyer A. S., Leseche G., Sidibe A., Herbin O., Yin X., Gomes A., Madhu B., Griffiths J. R., Xu Q., Tedgui A., Boulanger C. M. (2009) Proteomics, metabolomics, and immunomics on microparticles derived from human atherosclerotic plaques. Circ. Cardiovasc. Genet. 2, 379–388 [DOI] [PubMed] [Google Scholar]
  • 21. Pula G., Mayr U., Evans C., Prokopi M., Vara D. S., Yin X., Astroulakis Z., Xiao Q., Hill J., Xu Q., Mayr M. (2009) Proteomics identifies thymidine phosphorylase as a key regulator of the angiogenic potential of colony-forming units and endothelial progenitor cell cultures. Circ. Res. 104, 32–40 [DOI] [PubMed] [Google Scholar]
  • 22. Urbich C., De Souza A. I., Rossig L., Yin X., Xing Q., Prokopi M., Drozdov I., Steiner M., Breuss J., Xu Q., Dimmeler S., Mayr M. (2011) Proteomic characterization of human early pro-angiogenic cells. J. Mol. Cell. Cardiol. 50, 333–336 [DOI] [PubMed] [Google Scholar]
  • 23. Hagglund P., Bunkenborg J., Elortza F., Jensen O. N., Roepstorff P. (2004) A new strategy for identification of N-glycosylated proteins and unambiguous assignment of their glycosylation sites using HILIC enrichment and partial deglycosylation. J. Proteome Res. 3, 556–566 [DOI] [PubMed] [Google Scholar]
  • 24. Saba J., Dutta S., Hemenway E., Viner R. (2012) Increasing the productivity of glycopeptides analysis by using higher-energy collision dissociation-accurate mass-product-dependent electron transfer dissociation. Int. J. Proteomics 2012, 560391. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Bern M., Cai Y., Goldberg D. (2007) Lookup peaks: a hybrid of de novo sequencing and database search for protein identification by tandem mass spectrometry. Anal Chem. 79, 1393–1400 [DOI] [PubMed] [Google Scholar]
  • 26. Antonov A. S., Lukashev M. E., Romanov Y. A., Tkachuk V. A., Repin V. S., Smirnov V. N. (1986) Morphological alterations in endothelial cells from human aorta and umbilical vein induced by forskolin and phorbol 12-myristate 13-acetate: a synergistic action of adenylate cyclase and protein kinase C activators. Proc. Natl. Acad. Sci. U.S.A. 83, 9704–9708 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27. Sadler J. E. (1998) Biochemistry and genetics of von Willebrand factor. Annu. Rev. Biochem. 67, 395–424 [DOI] [PubMed] [Google Scholar]
  • 28. Matsui T., Titani K., Mizuochi T. (1992) Structures of the asparagine-linked oligosaccharide chains of human von Willebrand factor. Occurrence of blood group A, B, and H(O) structures. J. Biol. Chem. 267, 8723–8731 [PubMed] [Google Scholar]
  • 29. Canis K., McKinnon T. A., Nowak A., Haslam S. M., Panico M., Morris H. R., Laffan M. A., Dell A. (2012) Mapping the N-glycome of human von Willebrand factor. Biochem. J. 447, 217–228 [DOI] [PubMed] [Google Scholar]
  • 30. Simak J., Holada K., Vostal J. G. (2002) Release of annexin V-binding membrane microparticles from cultured human umbilical vein endothelial cells after treatment with camptothecin. BMC Cell Biol. 3, 11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31. Hart G. W., Copeland R. J. (2010) Glycomics hits the big time. Cell 143, 672–676 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Wollscheid B., Bausch-Fluck D., Henderson C., O'Brien R., Bibel M., Schiess R., Aebersold R., Watts J. D. (2009) Mass-spectrometric identification and relative quantification of N-linked cell surface glycoproteins. Nat. Biotechnol. 27, 378–386 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33. Leth-Larsen R., Lund R. R., Ditzel H. J. (2010) Plasma membrane proteomics and its application in clinical cancer biomarker discovery. Mol. Cell. Proteomics 9, 1369–1382 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34. Madabhushi S. R., Shang C., Dayananda K. M., Rittenhouse-Olson K., Murphy M., Ryan T. E., Montgomery R. R., Neelamegham S. (2012) von Willebrand factor (VWF) propeptide binding to VWF D'D3 domain attenuates platelet activation and adhesion. Blood 119, 4769–4778 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. van Mourik J. A., Boertjes R., Huisveld I. A., Fijnvandraat K., Pajkrt D., van Genderen P. J., Fijnheer R. (1999) von Willebrand factor propeptide in vascular disorders: a tool to distinguish between acute and chronic endothelial cell perturbation. Blood 94, 179–185 [PubMed] [Google Scholar]
  • 36. Pan S., Chen R., Aebersold R., Brentnall T. A. (2011) Mass spectrometry based glycoproteomics—from a proteomics perspective. Mol. Cell. Proteomics 10, R110.003251. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37. Maley F., Trimble R. B., Tarentino A. L., Plummer T. H., Jr. (1989) Characterization of glycoproteins and their associated oligosaccharides through the use of endoglycosidases. Anal. Biochem. 180, 195–204 [DOI] [PubMed] [Google Scholar]
  • 38. Flatmark T. (1967) Multiple molecular forms of bovine heart cytochrome c. V. A comparative study of their physicochemical properties and their reactions in biological systems. J. Biol. Chem. 242, 2454–2459 [PubMed] [Google Scholar]
  • 39. Zielinska D. F., Gnad F., Wisniewski J. R., Mann M. (2010) Precision mapping of an in vivo N-glycoproteome reveals rigid topological and sequence constraints. Cell 141, 897–907 [DOI] [PubMed] [Google Scholar]
  • 40. Angel P. M., Lim J. M., Wells L., Bergmann C., Orlando R. (2007) A potential pitfall in 18O-based N-linked glycosylation site mapping. Rapid Commun. Mass Spectrom. 21, 674–682 [DOI] [PubMed] [Google Scholar]
  • 41. Palmisano G., Melo-Braga M. N., Engholm-Keller K., Parker B. L., Larsen M. R. (2012) Chemical deamidation: a common pitfall in large-scale N-linked glycoproteomic mass spectrometry-based analyses. J. Proteome Res. 11, 1949–1957 [DOI] [PubMed] [Google Scholar]
  • 42. Wuhrer M., Deelder A. M., Hokke C. H. (2005) Protein glycosylation analysis by liquid chromatography-mass spectrometry. J. Chromatogr. B Analyt. Technol. Biomed. Life Sci. 825, 124–133 [DOI] [PubMed] [Google Scholar]
  • 43. Morelle W., Michalski J. C. (2007) Analysis of protein glycosylation by mass spectrometry. Nat. Protoc. 2, 1585–1602 [DOI] [PubMed] [Google Scholar]
  • 44. Zhang M. Y., Pace N., Kerns E. H., Kleintop T., Kagan N., Sakuma T. (2005) Hybrid triple quadrupole-linear ion trap mass spectrometry in fragmentation mechanism studies: application to structure elucidation of buspirone and one of its metabolites. J. Mass Spectrom. 40, 1017–1029 [DOI] [PubMed] [Google Scholar]
  • 45. Tissot B., North S. J., Ceroni A., Pang P. C., Panico M., Rosati F., Capone A., Haslam S. M., Dell A., Morris H. R. (2009) Glycoproteomics: past, present and future. FEBS Lett. 583, 1728–1735 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Hang H. C., Yu C., Kato D. L., Bertozzi C. R. (2003) A metabolic labeling approach toward proteomic analysis of mucin-type O-linked glycosylation. Proc. Natl. Acad. Sci. U.S.A. 100, 14846–14851 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47. Zhao P., Viner R., Teo C. F., Boons G. J., Horn D., Wells L. (2011) Combining high-energy C-trap dissociation and electron transfer dissociation for protein O-GlcNAc modification site assignment. J. Proteome Res. 10, 4088–4104 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48. Owen J. B., Di Domenico F., Sultana R., Perluigi M., Cini C., Pierce W. M., Butterfield D. A. (2009) Proteomics-determined differences in the concanavalin-A-fractionated proteome of hippocampus and inferior parietal lobule in subjects with Alzheimer's disease and mild cognitive impairment: implications for progression of AD. J. Proteome Res. 8, 471–482 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49. Ghosh D., Krokhin O., Antonovici M., Ens W., Standing K. G., Beavis R. C., Wilkins J. A. (2004) Lectin affinity as an approach to the proteomic analysis of membrane glycoproteins. J. Proteome Res. 3, 841–850 [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Supplementary Materials

Supplemental Table 1

Articles from Molecular & Cellular Proteomics : MCP are provided here courtesy of American Society for Biochemistry and Molecular Biology

RESOURCES