Proteomic analysis of TNF-α-activated endothelial cells and endothelial microparticles

  • Authors:
    • Yiyun Liu
    • Wenchang Huang
    • Ruyuan Zhang
    • Jun Wu
    • Lei Li
    • Yaoqing Tang
  • View Affiliations

  • Published online on: October 22, 2012     https://doi.org/10.3892/mmr.2012.1139
  • Pages: 318-326
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Endothelial microparticles (EMPs) are small vesicles released from endothelial cells (ECs) and found circulating in the blood. EMPs are formed by a plasma membrane surrounding a small amount of cytosol and contain a subset of cellular proteins. As the number of EMPs in the blood increases with certain diseases, they may be an attractive biomarker for clinical diagnosis. Proteomic analysis of EMPs has been previously performed by mass spectrometry. However, the proteomic information of the ECs that secrete EMPs is lacking. This study introduces an in vitro model of activated ECs we created for proteomic analyses and reports the changes of the protein content in the ECs and EMPs using proteomic methods. Thus, this study provides valuable information for the analysis of the highly dynamic secretion process of EMPs. There is a direct correlation between the proteins that form EMPs and tumor necrosis factor-α (TNF-α)-activated ECs. The endothelial proteins transferred by EMPs may play important roles in the interaction between EMPs and the target cells, which may lead to endothelial dysfunction.

Introduction

Sepsis and septic shock are considered the primary causes of mortality in the intensive care unit worldwide (1). It has been widely acknowledged that endothelial cells (ECs) play a key role in the pathogenesis of sepsis (2,3). The vascular endothelium is not only the barrier between blood and tissue, but also takes part in the inflammatory and coagulation responses (4). Its dysfunction or damage is a crucial link during the development of sepsis and may lead to multiorgan failure. Thus, it is important and necessary to discover the pathological mechanism of endothelial dysfunction that results in sepsis.

Endothelial microparticles (EMPs) are small vesicles released from activated, apoptotic or injured ECs ranging in size from less than 1 μm (5). EMPs were first discovered by Hamilton et al in 1990 (6). Under normal conditions, there is a low concentration of EMPs in the circulation. Under pathological conditions, particularly in diseases associated with endothelial dysfunction, the level of EMPs is significantly higher (5). Therefore, studying EMPs may be a practicable and effective method of studying ECs.

When ECs are stimulated by proinflammatory, prothrombotic or proapoptotic factors, or are exposed to high shear stress, EMPs will be generated and released. This may also occur in the case of cellular differentiation, senescence or apoptotic cell collapse (7). The phenotype of EMPs is distinct according to the different stimuli. In general, EMPs produced by activation factors include higher levels of endothelial inducible markers, such as CD62E (E-selectin), and EMPs produced by apoptotic factors include higher levels of Annexin V and endothelial constitutive markers, such as CD31 (PECAM) (8). The components of EMPs are also distinct according to the different stimuli. The research of Peterson et al(9) indicated that EMPs induced by PAI-1 and TNF-α have overlapping but distinct protein compositions.

Previous studies merely considered EMPs as inert biomarkers indicating endothelial function, but numerous studies have proven that EMPs are the vectors for intercellular information exchange (10,11). EMPs can alter the function of neighboring cells, as well as cells far away from their original cells. Their effects involve many aspects, for example inflammatory and coagulation responses, angiogenesis, cell proliferation and cell migration.

However, the mechanisms by which EMPs are released from ECs and affect the function of downstream cells remain unclear. In order to investigate the possible mechanisms, it is important to determine the protein composition of EMPs. The majority of the prior research focused on 1 or several proteins of the pathway, but these results were not comprehensive. Other studies, such as the study by Peterson et al(9), used proteomic analysis and discovered more valuable proteins. It is clear that proteomic analysis is an ideal method to investigate the possible mechanisms of EMPs at present.

However, the existing proteomic reports still have some problems. They compared the contents of EMPs generated by different stimuli and found distinct proteins, but ignored the change of protein components that occurred in the corresponding ECs. This may be an important aspect of revealing the mechanisms associated with EMPs.

In this study, we aimed to stimulate ECs to release EMPs using tumor necrosis factor-α (TNF-α), which is a commonly used proinflammatory substance, so as to produce an in vitro model of activated ECs for proteomic research and then comprehensively compare the proteome of unstimulated and TNF-α-stimulated ECs using two-dimensional (2-D) gel electrophoresis followed by mass spectrometry (MS). Simultaneously, we determined the protein composition of EMPs from ECs stimulated by TNF-α using the liquid chromatography-mass spectrometry (LC-MS)/MS method. Finally, using the comparison of proteins as mentioned above, we confirmed that EMPs are capable of transferring biological information, and moreover, gain further insight into the possible functional mechanisms of EMPs generated by TNF-α.

Our results may help to understand the mechanism of endothelial dysfunction and sepsis pathogenesis. Thus we may open up a new field of research for diagnosis and therapy of sepsis.

Materials and methods

HUVEC culture and EMP generation

Primary cultures of human umbilical vein endothelial cells (HUVECs) were isolated from fresh human umbilical cord veins using collagenase type I (Sigma, St. Louis, MO, USA) and maintained in medium 200 (Cascade Biologics, Carlsbad, CA, USA) supplemented with low serum growth supplement (LSGS; Cascade Biologics). Cell passages were performed using 0.05% trypsin in 0.02% ethylenediaminetetra-acetic acid. Cells were incubated in a humidified incubator with 5% carbon dioxide at 37°C. The medium was renewed every 48 h until confluence occurred. The HUVECs used for this research were from the second passage.

HUVECs were equally divided into 1 control group and 9 TNF-α-stimulated groups. The control group was incubated in fresh medium 200 with LSGS. The TNF-α-stimulated groups were incubated in serum-free medium for 2 h and then incubated in fresh medium 200 with LSGS containing 10, 100 or 200 ng/ml TNF-α (PeproTech, Rocky Hill, NJ, USA) for 1, 3 or 24 h, respectively.

Following incubation, the cell-conditioned medium of each group was harvested and centrifuged at 200 × g for 5 min at room temperature to remove cell debris. The supernatant was collected and ultracentrifuged at 100,000 × g for 2 h at 4°C. The supernatant was discarded. The sediment was washed once with PBS and ultracentrifuged in the same way. The EMP pellet was used for flow cytometry and LC-MS/MS and it was stored at 4°C for no more than 72 h.

HUVECs from each group were washed 3 times with PBS, digested by trypsin, and then centrifuged at 200 × g for 5 min at room temperature. The supernatant was discarded and cells were collected for 2-D electrophoresis.

EMP detection

Flow cytometry was used for EMP detection. The EMP pellet of each group was resuspended in 200 μl PBS and labeled by CD62E (BD Biosciences, Franklin Lakes, NJ, USA). For counting, a known amount of 1 μm fluorescent latex beads (Sigma) was added to samples as an internal standard. Using these latex beads as gating parameters, EMPs were defined as particles <1 μm in size. EMPs were counted from the gate corresponding to PE-CD62E+ events, so that other small granules in the medium, such as the composition of LSGS, could be excluded. The results are expressed as the number of EMPs/1×106 cells.

Statistical analysis was performed using ANOVA. P<0.05 was considered to indicate a statistically significant difference.

According to the EMP detection results, the group that generated the most EMPs among the 9 TNF-α groups was determined. The EMPs of this group were used for LC-MS/MS. HUVECs of this group and the control group were used for 2-D electrophoresis.

HUVEC 2-D electrophoresis

Two groups of HUVECs were respectively suspended in a solution containing 7 M urea, 2 M thiourea, 4% w/v CHAPS, 1% w/v DTT (all from Sigma), 2% IPG buffer (310), 40 mM Tris-base (both from Bio-Rad, Hercules, CA, USA) and protease inhibitor cocktail EDTA-free (Pierce, Rockford, IL, USA). The suspension was sonicated 5 times for 10 sec at 4°C. The samples were centrifuged at 16,000 × g for 30 min to pellet cell debris. The concentration of sample proteins were detected by RCDC Protein Assay (Bio-Rad). IPG strips (Bio-Rad) used for isoelectric focusing (IEF) were 17 cm with pH 3 to 10. In total, 100 μg of protein from each sample was added into rehydration solution. IPG strips were put into individual grooves in the reswelling tray. IEF was carried out using a horizontal electrophoresis apparatus (Bio-Rad). After the rehydration at 50 V for 12 h at 20°C, focusing was started at 250 V and the voltage was progressively increased to 10,000 V until a maximum of 88,700 Vh. When IEF was finished, strips were equilibrated twice for 15 min in equilibration buffer containing 6 M urea, 30% v/v glycerol (Sigma), 2% w/v SDS (Bio-Rad) and 1.5 M Tris-HCl buffer (pH 8.8) supplemented with 5 mg/ml DTT for the first treatment and 45 mg/ml iodoacetamide (Sigma) for the second treatment. Polyacrylamide gels (12.5%) were used for SDS polyacrylamide gel electrophoresis. The running conditions were 16 mA/gel for 30 min followed by 24 mA/gel for 6 h at 20°C. The gels were stained by silver, according to the method of Westermeier et al(12). 2-D electrophoresis was repeated 3 times. Stained gels were scanned and the images were analyzed by PDQuest Image Analysis software (Bio-Rad) in order to find the differentially expressed protein spots and ratio level of the TNF-α stimulated group to the control group.

HUVEC matrix-assisted laser desorption/ionization-time-of-flight (MALDI-TOF)/TOF-MS

Protein spots were excised from the preparative gels, and destained with 100 mM NH4HCO3 in 30% ACN. After removing the destaining buffer, the gel pieces were lyophilized and rehydrated in 30 μl of 50 mM NH4HCO3 containing 50 ng trypsin (Promega, Madison, WI, USA). Following overnight digestion at 37°C, the peptides were extracted 3 times with 0.1% TFA in 60% ACN. Extracts were pooled together and lyophilized. The resulting lyophilized tryptic peptides were maintained at −80°C until mass spectrometric analysis. A protein-free gel piece was treated as above and used as a control to identify autoproteolysis products derived from trypsin.

MS and MS/MS spectra were obtained using the ABI 4800 Proteomics Analyzer MALDI-TOF/TOF (Applied Biosystems, Carlsbad, CA, USA) operating in a result-dependent acquisition mode. Peptide mass maps were acquired in positive ion reflector mode (20 kV accelerating voltage) with 1000 laser shots per spectrum. Monoisotopic peak masses were automatically determined within the mass range 800–4000 Da with a signal-to-noise ratio minimum set to 10 and a local noise window width of 250 m/z. Up to 5 of the most intense ions with minimum signal-to-noise ratio of 50 were selected as precursors for MS/MS acquisition, excluding common trypsin autolysis peaks and matrix ion signals. In MS/MS-positive ion mode, spectra were averaged, collision energy was 2 kV and default calibration was set. Monoisotopic peak masses were automatically determined with a signal-to-noise ratio minimum set to 5 and a local noise window width of 250 m/z. The MS together with MS/MS spectra were searched against the UniprotKB/Swiss-Prot database using the software GPS Explorer (Applied Biosystems) and MASCOT (Matrix Science, Boston, MA, USA) with the following parameter settings: trypsin cleavage, one missed cleavage allowed, carbamidomethylation set as fixed modification, oxidation of methionines allowed as variable modification, peptide mass tolerance set to 100 ppm, fragment tolerance set to ±0.3 Da and minimum ion score confidence interval for MS/MS data set to 95%.

EMP LC-MS/MS

EMPs were disrupted by lysis buffer with protease inhibitor cocktail EDTA-free (Pierce). The suspension was sonicated 5 times for 10 sec at 4°C. The sample was then centrifuged at 16,000 × g for 30 min to pellet the insoluble protein fraction. The supernatant containing soluble proteins was used for LC-MS/MS.

Ettan™ MDLC system (GE Healthcare, Piscataway, NJ, USA) was applied for desalting and separation of tryptic peptide mixtures. In this system, samples were desalted on RP trap columns (Zorbax 300SB-C18, Agilent Technologies, Santa Clara, CA, USA), and then separated on a RP column (150 μm i.d., 100 mm length, Column Technologies Inc., Downers Grove, IL, USA). The mobile phase A (0.1% formic acid in HPLC-grade water) and mobile phase B (0.1% formic acid in acetonitrile) were selected. A total of 20 μg of tryptic peptide mixtures of EMPs were loaded onto the columns, and separation was performed at a flow rate of 2 μl/min by using a linear gradient of 4–50% B for 120 min. A Finnigan™ LTQ™ linear ion trap MS (Thermo Electron, Waltham, MA, USA) equipped with an electrospray interface was connected to the LC setup for detection of eluted peptides. Data-dependent MS/MS spectra were obtained simultaneously. Each scan cycle consisted of 1 full MS scan in profile mode followed by 5 MS/MS scans in centroid mode with the following Dynamic Exclusion™ settings: repeat count 2, repeat duration 30 sec, exclusion duration 90 sec. Each sample was analyzed in triplicate.

The MS/MS spectra were automatically searched against the non-redundant International Protein Index (IPI) human protein database (version 3.26, 67687 entries) using the BioworksBrowser (Thermo Electron). Protein identification results were extracted from the SEQUEST files with BuildSummary (13).

The peptides were constrained to be tryptic and up to 2 missed cleavages were allowed. Carbamidomethylation of cysteines was treated as a fixed modification, whereas oxidation of methionine residues was considered as a variable modification. The mass tolerance allowed for the precursor ions was 2.0 Da and fragment ions was 0.2 Da, respectively. The protein identification criteria were based on Delta CN (≥0.1) and cross-correlation scores (Xcorr, 1 charge ≥1.9, 2 charges ≥2.2, 3 charges ≥3.75).

Protein analysis

All accession IDs of the proteins identified in HUVECs via MALDI-TOF/TOF-MS and in EMPs via LC-MS/MS were unified to Swiss-Prot IDs at http://www.uniprot.org/ and the protein names were also discovered. The endothelial differentially expressed proteins were compared with the proteome of EMPs in order to determine any existing correlations. The Swiss-Prot IDs of the proteins we were interested in were used to annotate these proteins with their corresponding Gene Ontology (GO) annotations including the cellular components, molecular functions and biological processes involved. In addition, KEGG pathway annotations of these proteins were also found at http://www.genome.jp/kegg/.

Results

TNF-α dose and time-dependency of EMP generation

All 9 TNF-α groups generated more EMPs than the control group, which generated 5123 EMPs/1×106 cells. Fig. 1 indicates clearly that the group treated with 100 ng/ml TNF-α for 24 h had the most EMPs (54588 EMPs/1×106 cells). Differences among 9 groups under different TNF-α doses and time conditions were statistically analyzed (Fig. 1).

2-D patterns of unstimulated HUVECs and TNF-α-stimulated HUVECs

2-D gel electrophoresis was used to respectively separate all proteins from unstimulated HUVECs and TNF-α-stimulated HUVECs and repeated 3 times. Fig. 2 shows these 2 patterns of silver stained 2-D polyacrylamide gels based on protein molecular weight (MW) and isoelectric point (pI), each of which contained 100 μg of proteins. pH ranged from 3 to 10 and MW ranged from 14.4 to 97.4 kDa. The patterns were generally consistent with the pattern reported previously (14).

The protein spots were analyzed by PDQuest Image Analysis software in order to detect the differentially expressed proteins. We found 47 differentially expressed proteins. Each protein level ratio of TNF-α-stimulated HUVECs to unstimulated HUVECs was calculated. Among these 47 proteins, the expression of 11 proteins was upregulated, 21 proteins were downregulated, 13 proteins were no longer expressed, and 2 proteins were freshly generated after the stimulation.

Identification of differentially expressed proteins in HUVECs by MALDI-TOF/TOF-MS

In total, 29 of these 47 differentially expressed proteins were successfully identified through MALDI-TOF/TOF-MS and every protein score C.I.% was higher than 95%. Among these 29 proteins, the expression of 8 proteins was upregulated, 12 proteins were downregulated, 8 proteins were no longer expressed, and 1 protein was freshly generated (Fig. 2). Protein name, protein level ratio and the other information are shown in Table I. The highest ratio of upregulation was approximately 3.5-fold and the highest ratio of downregulation was approximately 3.18-fold.

Table I

Differentially expressed proteins between unstimulated HUVECs and TNF-α-stimulated HUVECs identified by MALDI-TOF/TOF-MS.

Table I

Differentially expressed proteins between unstimulated HUVECs and TNF-α-stimulated HUVECs identified by MALDI-TOF/TOF-MS.

Spot no.Protein nameSwiss-ProtPredicted MW (Da)Predicted pIProtein scoreProtein score CI (%)Ratioa
1Far upstream element-binding protein 1Q96AE467689.57.18309100
2Vimentinb,cP0867053738.15.036998.646
3Four and a half LIM domains protein 2Q1419234166.47.807699.717
4LDLR chaperone MESDQ1469626231.47.60402100
53-Hydroxyacyl-CoA dehydrogenase type-2Q9971420738.87.939299.992
6Galectin-1P0938214868.35.34171100
7Superoxide dismutase [Cu-Zn]P0044116023.05.707299.221
840S ribosomal protein S12P2539814904.66.81200100
9Annexin A2b,cP0735538779.97.57423100−3.17897
10VinculinP18206124292.05.50719100−2.31222
11Septin-2Q1501947068.86.95380100−2.10470
12Four and a half LIM domains protein 3Q1364333210.35.67191100−1.98458
13Protein canopy homolog 2Q9Y2B020981.34.81246100−1.94463
14Heterogeneous nuclear ribonucleoproteins C1/C2P0791032003.75.10176100−1.87902
15Heat shock cognate 71 kDa proteinP1114271082.35.377799.759−1.80698
16T-complex protein 1 subunit βP7837122924.25.88146100−1.80438
17Actin, cytoplasmic 1bP6070940536.25.556596.271−1.67332
18Protein disulfide-isomeraseb,cP0723757479.84.76141100−1.59406
19Tubulin β chainb,cP0743748135.14.70411100−1.59367
20Elongation factor 1-δP2969231216.84.90445100−1.55537
21Profilin-1P0773715084.68.48221100
22Heat shock protein β-1bP0479222825.55.98371100+3.50088
23 NucleophosminbP0674831090.34.71633100+2.09525
24Cathepsin ZQ9UBR227758.95.48168100+1.96644
25β-hexosaminidase subunit βP076866829.46.819499.996+1.86827
26Ran-specific GTPase-activating proteinP4348723395.65.198999.986+1.84756
27Putative nucleoside diphosphate kinaseO6036115690.08.76359100+1.81783
28Myosin light polypeptide 6b,cP6066017090.24.56276100+1.61712
29Clathrin light chain BP0949723281.24.63244100+1.52998

a Protein level ratio of TNF-α-stimulated HUVECs to unstimulated HUVECs.

b Proteins also included in EMPs.

c Proteins also reported in the study by Sander et al(21).

{ label (or @symbol) needed for fn[@id='tfn4-mmr-07-01-0318'] } Nos. 1–8, proteins are no longer expressed; nos. 9–20, protein expression is downregulated; no. 21, protein is freshly generated; and nos. 22–29, protein expression is upregulated. +, Upregulation; −, downregulation; EMPs, endothelial microparticles; HUVECs, human umbilical vein endothelial cells.

Proteome identification of EMPs generated from TNF-α-stimulated HUVECs by LC-MS/MS and protein analysis

A total of 83 proteins were identified in EMPs generated from TNF-α-stimulated HUVECs. Comparing these proteins with 29 differentially expressed proteins as noted previously, we observed 8 common proteins within EMPs, of which 1 protein was no longer expressed, the expression of 4 proteins was downregulated, and the expression of 3 proteins was upregulated in TNF-α-stimulated HUVECs. GO annotation and KEGG pathway analysis data of these 8 proteins is shown in Table II.

Table II

GO and KEGG pathway analysis of common proteins between the proteome of EMPs and differential proteins of HUVECs.

Table II

GO and KEGG pathway analysis of common proteins between the proteome of EMPs and differential proteins of HUVECs.

Gene Ontology

Protein nameCellular componentMolecular functionBiological processKEGG pathway
VimentinCytosol
Intermediate filament
Protein C-terminus binding
Structural constituent of cytoskeleton
Cellular component disassembly involved in apoptosis
Annexin A2Soluble fractionCalcium ion binding
Calcium-dependent phospholipid binding
Cytoskeletal protein binding
Phospholipase inhibitor activity
Cellular component movement
Positive regulation of vesicle fusion
Actin, cytoplasmic 1Nucleus
Cytoskeleton
Cytosol
Ribonucleoprotein complex
ATP binding
Kinesin binding
Nitric-oxide synthase binding
Structural constituent of cytoskeleton
De novo’ posttranslational protein folding
Adherens junction organization
Blood coagulation
Cell junction assembly
Cellular component movement
Phagosome
Focal adhesion
Adherens junction
Tight junction
Leukocyte transendothelial migration
Regulation of actin cytoskeleton
Vibrio cholerae infection
Pathogenic Escherichia coli infection
Shigellosis
Protein disulfide-isomeraseER-Golgi intermediate compartment
Cell surface
Endoplasmic reticulum lumen
Extracellular region
Plasma membrane
Procollagen-proline 4-dioxygenase activity
Protein binding
Protein disulfide isomerase activity
Cell redox homeostasis
Lipid metabolic process
Lipoprotein metabolic process
Peptidyl-proline hydroxylation to 4-hydroxy-L-proline
Protein processing in endoplasmic reticulum
Tubulin β chainCytosol
Microtubule
GTP binding
GTPase activity
MHC class I protein binding
G2/M transition of mitotic cell cycle
Cellular component movement
Microtubule-based movement
Natural killer cell mediated cytotoxicity
Protein polymerization
Phagosome
Gap junction
Pathogenic Escherichia coli infection
Heat shock protein β-1Cell surface
Cytosol
Nucleus
Spindle
Identical protein binding Anti-apoptosis
Cell death
Cellular component movement
mRNA metabolic process
Regulation of translational initiation
Response to heat
Response to unfolded protein
Response to virus
MAPK signaling pathway
VEGF signaling pathway
Amoebiasis
NucleophosminSpindle pole centrosomeNF-κB binding
Tat protein binding
Histone binding
Protein binding
Protein heterodimerization activity
Protein homodimerization activity
Ribosomal large subunit binding
Ribosomal small subunit binding
Transcription coactivator activity
Unfolded protein binding
CenH3-containing nucleosome assembly at centromere
DNA repair
Anti-apoptosis
Centrosome cycle
Intracellular protein transport
Negative regulation of cell proliferation
Negative regulation of centrosome duplication
Positive regulation of NF-κB transcription factor activity
Protein oligomerization
Regulation of endodeoxyribonuclease activity
Regulation of endoribonuclease activity
Ribosome assembly
Signal transduction
Myosin light polypeptide 6Cytosol
Unconventional myosin complex
Actin-dependent ATPase activity
Calcium ion binding
Motor activity
Muscle filament slidingVascular smooth muscle contraction

[i] EMPs, endothelial microparticles; HUVECs, human umbilical vein endothelial cells.

Discussion

In EMP research, TNF-α is widely used to activate ECs to produce EMPs and mimic acute inflammation. However, there are scarce details about the TNF-α dose and time-dependency of the EMP generation. Our results showed that TNF-α enhanced EMP release compared with unstimulated HUVECs. When treated with 100 ng/ml TNF-α for 24 h, HUVECs generated the most EMPs. According to these findings, the in vitro model of activated ECs could be used in order to generate enough EMPs for proteomic research.

Our study indicates that a direct correlation exists between the proteins comprising EMPs and the proteins expressed by ECs induced by TNF-α. We confirmed that endothelial proteins of various pathways will change when TNF-α, as an important proinflammatory factor during sepsis, stimulates HUVECs. These changes take place not only in quantity but also in type of proteins. According to our results, there were 29 differentially expressed EC proteins activated by TNF-α compared to the unstimulated ECs, 8 of which were upregulated, 12 of which were downregulated, 8 of which were no longer expressed, and 1 of which was freshly generated. The probable causes of the modification of proteins are as follows. The upregulation and new generation occur due to the augmentation of protein synthesis or the conversion from other proteins. The downregulation and the lack of expression occur due to the decrease of protein synthesis, transforming to other proteins, protein degradation or the release out of the cells, which may include the direct release to the extracellular space and the release via EMPs. Further study revealed that EMPs contained 8 of the 29 proteins, of which the expression of 3 were upregulated, 4 were downregulated, and 1 was no longer expressed. We consider that the shedding of EMPs is an important cause of endothelial protein modification. It should be noted that the shedding of EMPs did not only occur during protein downregulation and lack of expression, but also occurred during upregulation, as we found 3 upregulated proteins in EMPs. The fact that the quantity of these proteins still increases is perhaps due to the protein synthesis exceeding the release, or the release having a negative feedback effect, which makes the synthesis augment.

Due to this correlation, we proved that EMPs are not only markers reflecting the condition of ECs but also vectors exchanging intercellular information. The cellular component of these 8 common proteins tells us that EMPs could carry proteins from any component out of the ECs (Table II). Among these 8 proteins, there were 2 upregulated proteins associated with anti-apoptosis [heat shock protein β-1 (HspB1) and nucleophosmin] and 3 proteins associated with cytoskeletal structure (vimentin, actin cytoplasmic 1 and tubulin β chain). TNF-α is capable of causing endothelial apoptosis by activating ECs (15). Therefore the generation of anti-apoptotic proteins rises to a higher level. The knowledge of EMP production and release comes from research on platelet microparticles (16). Briefly, the mechanism concerns the alteration of the cytoskeleton. Thus the related endothelial proteins change. The proteome of EMPs may provide a detailed statement about the situation of the original ECs, such as what stimulated the cells and how the cells responded. However, EMPs as vesicles transferring biological signals and information should contain proteins playing a role in essential biological processes. Among these 8 proteins, for example, there were proteins (actin cytoplasmic 1 and tubulin β chain) involved in cell adhesion, cell junction or leukocyte transendothelial migration, which are key proteins in the processes of endothelial high permeability and capillary leak.

Furthermore, when these 8 proteins were investigated, we found that both the protein with the highest increase in expression and that with the lowest among the 29 differentially expressed endothelial proteins examined, were within the EMPs. This does not mean that the greatest change is the most important change, but it is evident that these 2 proteins, HspB1 and annexin A2, are significant proteins.

HspB1 was 3.50-fold more abundant in the TNF-α-induced ECs than in resting ECs. According to GO analysis, HspB1 is involved in anti-apoptosis, cellular component movement, mRNA metabolic processes, regulation of translational initiation and other biological processes. A recent report clarified that ECs with upregulated HspB1 could survive apoptosis (17). Another report revealed that HspB1 could have a protective effect on the cytoskeleton and preserve EC integrity, which could protect against acute kidney injury after hepatic ischemia and reperfusion (18). Moreover, KEGG pathway analysis showed that HspB1 is associated with the MAPK and VEGF signaling pathways. Thus, once activated by a proinflammatory factor such as TNF-α, ECs will upregulate HspB1 to protect themselves. This condition could be detected through EMPs and EMPs may carry HspB1 to affect other cells.

Annexin A2 was 3.18-fold less abundant in TNF-α-induced ECs than in resting ECs. The endothelial-related information of GO analysis shows that annexin A2 positively regulates the vesicle fusion. We know that ECs liberate EMPs so they need to downregulate proteins leading to vesicle fusion, such as annexin A2. Reflecting this condition, EMPs with annexin A2 tend to fuse with other cells and then play biological roles. In addition, it was confirmed that annexin A2 is capable of assembling plasminogen and has a positive effect on vascular fibrinolysis (19,20).

We consider that perhaps the proteins within EMPs shedding from ECs have 2 different effects. Firstly, they are useful to regulate the function of other cells, no matter whether the cells are neighboring or remote, or whether the cells are the same type or a different type, for example HspB1. Secondly, they are harmful to the original ECs, so they are released to protect the cells themselves. However, these proteins, for example annexin A2, may have a positive effect on downstream cells. Therefore, EMPs are not inert cellular debris, but vital vectors.

Compared with the previous proteomic reports of EMPs, 5 of these 8 proteins included in the TNF-α-derived EMPs in our study were the same as the proteins in the plasminogen activator inhibitor type 1 (PAI-1)-derived EMPs in the study by Sander et al(21) (indicated in Table I). However, all of these 8 proteins in our study were not found in proteins identified as unique to control EMPs, PAI-1 EMPs, or TNF-α EMPs in the study by Peterson et al(9). This is an interesting finding. The various proteomic methods used in these 3 studies is a possible cause for these differences since Sander et al used 2-D/MALDI-TOF/TOF-MS, Peterson et al used LC-MS/MS, and in our study, we used both 2-D/MALDI-TOF/TOF-MS and LC-MS/MS. 2-D/MALDI-TOF/TOF-MS is the classical method for finding differentially expressed proteins and LC-MS/MS is suitable for detecting substances with lower quantities of proteins, so we used the former to find differentially expressed proteins of HUVECs and the latter to detect the proteome of EMPs. Another possible cause is that the previous studies used commercially available HUVECs and our study used cells from fresh human umbilical cord veins. The HUVECs we used are more in accord with the actual situation found in the human body. Putting aside the factors of sensitivity and heterogeneity, it appears that there are proteins in all types of EMPs and these common proteins could account for the mechanisms of formation and function of EMPs.

It should be noted that the 8 no-longer expressed and 1 freshly generated endothelial proteins we found could be the consequence of the relatively low sensitivity of the 2-D electrophoresis method. It means that these 9 proteins do exist in ECs but they could not be detected in the 2-D gels. In other words, the no-longer expressed proteins could be downregulated proteins and the freshly generated protein could be an upregulated one. Even so, the conclusion of our study is meaningful as the changes of these endothelial proteins did occur. In addition, the number of proteins found within EMPs was less than that reported in previous proteomic research. This is probably due to fewer EMPs released by HUVECs from the fresh human umbilical cord veins we used than by commercially available HUVECs. However, if we consider the proteome of EMPs as a whole, this study of the proteins with relatively great changes will clearly indicate the important effect of EMPs.

In conclusion, we created an in vitro model of activated ECs for proteomic research with cultured HUVECs treated with 100 ng/ml TNF-α for 24 h. We found the differentially expressed proteins between the control ECs and the ECs stimulated by TNF-α and confirmed that EMPs could carry endothelial proteins out of the cells. GO and KEGG pathway analysis indicated that the common proteins included in EMPs have effects on vital biological processes, which may lead to endothelial dysfunction. Furthermore, we concluded that EMPs play roles not only in protecting their original ECs but also in affecting other cells. In ongoing studies in our laboratory, we are focusing on each important protein of ECs or EMPs according to this study to make clear the whole pathway through which the protein alters downstream cell function. We believe that EMPs could be diagnostic biomarkers and therapeutic targets in diseases associated with endothelial dysfunction, particularly in sepsis.

Acknowledgements

We thank Jianwen Hu, Yuan Cao and the other staff of Shanghai Applied Protein Technology Co. Ltd. for their technical assistance.

Abbreviations:

EMPs

endothelial microparticles

ECs

endothelial cells

HUVECs

human umbilical vein endothelial cells

2-D

two-dimensional, MS, mass spectrometry

GO

gene ontology

References

1 

Dellinger RP, Levy MM, Carlet JM, Bion J, Parker MM, Jaeschke R, Reinhart K, Angus DC, Brun-Buisson C, Beale R, Calandra T, Dhainaut JF, Gerlach H, Harvey M, Marini JJ, Marshall J, Ranieri M, Ramsay G, Sevransky J, Thompson BT, Townsend S, Vender JS, Zimmerman JL and Vincent JL: Surviving Sepsis Campaign: international guidelines for management of severe sepsis and septic shock: 2008. Crit Care Med. 36:296–327. 2008. View Article : Google Scholar

2 

Schouten M, Wiersinga WJ, Levi M and van der Poll T: Inflammation, endothelium, and coagulation in sepsis. J Leukoc Biol. 83:536–545. 2008. View Article : Google Scholar : PubMed/NCBI

3 

Sriskandan S and Altmann DM: The immunology of sepsis. J Pathol. 214:211–223. 2008. View Article : Google Scholar

4 

Rizvi AA: Cytokine biomarkers, endothelial inflammation, and atherosclerosis in the metabolic syndrome: emerging concepts. Am J Med Sci. 338:310–318. 2009. View Article : Google Scholar : PubMed/NCBI

5 

Chironi GN, Boulanger CM, Simon A, Dignat-George F, Freyssinet JM and Tedgui A: Endothelial microparticles in diseases. Cell Tissue Res. 335:143–151. 2009. View Article : Google Scholar : PubMed/NCBI

6 

Hamilton KK, Hattori R, Esmon CT and Sims PJ: Complement proteins C5b-9 induce vesiculation of the endothelial plasma membrane and expose catalytic surface for assembly of the prothrombinase enzyme complex. J Biol Chem. 265:3809–3814. 1990.PubMed/NCBI

7 

Mause SF and Weber C: Microparticles: protagonists of a novel communication network for intercellular information exchange. Circ Res. 107:1047–1057. 2010. View Article : Google Scholar : PubMed/NCBI

8 

Jimenez JJ, Jy W, Mauro LM, Soderland C, Horstman LL and Ahn YS: Endothelial cells release phenotypically and quantitatively distinct microparticles in activation and apoptosis. Thromb Res. 109:175–180. 2003. View Article : Google Scholar : PubMed/NCBI

9 

Peterson DB, Sander T, Kaul S, Wakim BT, Halligan B, Twigger S, Pritchard KA Jr, Oldham KT and Ou JS: Comparative proteomic analysis of PAI-1 and TNF-alpha-derived endothelial microparticles. Proteomics. 8:2430–2446. 2008. View Article : Google Scholar : PubMed/NCBI

10 

Ratajczak J, Wysoczynski M, Hayek F, Janowska-Wieczorek A and Ratajczak MZ: Membrane-derived microvesicles: important and underappreciated mediators of cell-to-cell communication. Leukemia. 20:1487–1495. 2006. View Article : Google Scholar : PubMed/NCBI

11 

Thery C, Ostrowski M and Segura E: Membrane vesicles as conveyors of immune responses. Nat Rev Immunol. 9:581–593. 2009. View Article : Google Scholar : PubMed/NCBI

12 

Westermeier R, Naven T and Höpker HR: Proteomics in Practice: A Guide to Successful Experimental Design. John Wiley & Sons; Weinheim: pp. 363–366. 2008

13 

Dai J, Shieh CH, Sheng QH, Zhou H and Zeng R: Proteomic analysis with integrated multiple dimensional liquid chromatography/mass spectrometry based on elution of ion exchange column using pH steps. Anal Chem. 77:5793–5799. 2003. View Article : Google Scholar : PubMed/NCBI

14 

Bruneel A, Labas V, Mailloux A, Sharma S, Vinh J, Vaubourdolle M and Baudin B: Proteomic study of human umbilical vein endothelial cells in culture. Proteomics. 3:714–723. 2003. View Article : Google Scholar : PubMed/NCBI

15 

Bannerman DD and Goldblum SE: Mechanisms of bacterial lipopolysaccharide-induced endothelial apoptosis. Am J Physiol Lung Cell Mol Physiol. 284:L899–914. 2003. View Article : Google Scholar : PubMed/NCBI

16 

Hugel B, Martínez MC, Kunzelmann C and Freyssinet JM: Membrane microparticles: two sides of the coin. Physiology (Bethesda). 20:22–27. 2005. View Article : Google Scholar : PubMed/NCBI

17 

Gawad A, Ptak-Belowska A, Brzozowski T and Pawlik WW: Monocytes and vascular endothelial cells apoptosis. Role of p-HSP27. J Physiol Pharmacol. 60:55–61. 2009.PubMed/NCBI

18 

Park SW, Chen SW, Kim M, D’Agati VD and Lee HT: Human heat shock protein 27-overexpressing mice are protected against acute kidney injury after hepatic ischemia and reperfusion. Am J Physiol Renal Physiol. 297:F885–894. 2009. View Article : Google Scholar : PubMed/NCBI

19 

Dassah M, Deora AB, He K and Hajjar KA: The endothelial cell annexin A2 system and vascular fibrinolysis. Gen Physiol Biophys. 28:F20–28. 2009.PubMed/NCBI

20 

He KL, Sui G, Xiong H, Broekman MJ, Huang B, Marcus AJ and Hajjar KA: Feedback regulation of endothelial cell surface plasmin generation by PKC dependent phosphorylation of annexin A2. J Biol Chem. 286:15428–15439. 2010.PubMed/NCBI

21 

Sander TL, Ou JS, Densmore JC, Kaul S, Matus I, Twigger S, Halligan B, Greene AS, Pritchard KA Jr and Oldham KT: Protein composition of plasminogen activator inhibitor type 1-derived endothelial microparticles. Shock. 29:504–511. 2008. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January 2013
Volume 7 Issue 1

Print ISSN: 1791-2997
Online ISSN:1791-3004

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Liu Y, Huang W, Zhang R, Wu J, Li L and Tang Y: Proteomic analysis of TNF-α-activated endothelial cells and endothelial microparticles. Mol Med Rep 7: 318-326, 2013
APA
Liu, Y., Huang, W., Zhang, R., Wu, J., Li, L., & Tang, Y. (2013). Proteomic analysis of TNF-α-activated endothelial cells and endothelial microparticles. Molecular Medicine Reports, 7, 318-326. https://doi.org/10.3892/mmr.2012.1139
MLA
Liu, Y., Huang, W., Zhang, R., Wu, J., Li, L., Tang, Y."Proteomic analysis of TNF-α-activated endothelial cells and endothelial microparticles". Molecular Medicine Reports 7.1 (2013): 318-326.
Chicago
Liu, Y., Huang, W., Zhang, R., Wu, J., Li, L., Tang, Y."Proteomic analysis of TNF-α-activated endothelial cells and endothelial microparticles". Molecular Medicine Reports 7, no. 1 (2013): 318-326. https://doi.org/10.3892/mmr.2012.1139