Identification of integrin β1 as a prognostic biomarker for human lung adenocarcinoma using 2D-LC‑MS/MS combined with iTRAQ technology

  • Authors:
    • Peng-Fei Zhang
    • Gu-Qing Zeng
    • Lun-Zhao Yi
    • Jian-Ping Liu
    • Xun‑Xun Wan
    • Jia-Quan Qu
    • Jian-Huang Li
    • Cui Li
    • Can-E Tang
    • Rong Hu
    • Xu Ye
    • Yu Chen
    • Zhu-Chu Chen
    • Zhi-Qiang Xiao
  • View Affiliations

  • Published online on: May 15, 2013     https://doi.org/10.3892/or.2013.2477
  • Pages: 341-349
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

To discover novel lung adenocarcinoma (AdC) biomarkers, isobaric tags for relative and absolute quantitation (iTRAQ)-tagging combined with 2D-LC-MS/MS analysis was used to identify differentially expressed plasma membrane proteins in lung AdC and paired paraneoplastic normal lung tissues (PNLTs) adjacent to tumors. In this study, significant caveolin-1 downregulation and integrin β1 upregulation was observed in primary lung AdC vs. PNLT. As there has been no report on the association of integrin β1 with lung AdC, immunohistochemical staining was performed to detect the expression of integrin β1 in an independent set of archival tissue specimens including 42 cases of PLNT, 46 cases of without lymph node metastasis primary AdC (non-LNM AdC) and 62 cases of LNM AdC; the correlation of their expression levels with clinicopathological characteristics and clinical outcomes were evaluated. Based on the data, upregulation of integrin β1 was significantly correlated with advanced clinical stage and lymph node metastasis. Integrin β1 overexpression was significantly associated with advanced clinical stage (P<0.05), lymph node metastasis (P<0.05), increased relapse rate (P<0.05) and decreased overall survival (P<0.05) in AdCs. Cox regression analysis indicated that integrin β1 overexpression is an independent prognostic factor. The data suggest that integrin β1 is a potential biomarker for LNM and prognosis of AdC and integrin β1 upregulation may play an important role in the pathogenesis of AdC.

Introduction

Lung cancer remains the leading cause of cancer-related mortality in several countries, including China (1,2). Although significant improvements have been made in the diagnosis and treatment of lung cancer, the prognosis for lung cancer is poor, with an overall five-year survival rate of approximately 16% (3). Lung adenocarcinoma (AdC) is a common histological type of lung cancer. In recent years, the frequency of lung AdC has increased and its prognosis remains poor (4,5). This high mortality rate is often attributed to the presence of advanced-stage metastasis, with more than two thirds of patients showing lymph node involvement and metastasis at the initial diagnosis. Therefore, to develop effective new strategies for the prediction, diagnosis and treatment of lung cancer metastasis, molecular mechanisms controlling metastasis must be identified.

Several groups have successfully used gene expression profiling techniques and model systems with different invasive or metastatic ability to identify genes that correlate with invasiveness or metastatic potential (68). Proteomics, particularly quantitative proteomics, has introduced a new approach to cancer research which aims at identifying differential expression proteins associated with carcinogenesis, providing new opportunities to reveal the molecular mechanism underlying this disease. Identification of differentially expressed proteins in lung AdC using proteomics revealed that expression levels of proteins may have some predictive power for metastasis and prognosis (912). In our lab, comparative proteomic studies of primary lung AdC with and without lymph node metastasis suggest that Annexin A3 and Annexin A1 are potential biomarkers for lymph node metastasis and prognosis in lung AdC (13,14).

However, in the past years, lung AdC proteomic studies have focused on whole cellular proteomic analysis. Subcellular proteomic analysis has advantages over whole cellular proteomic analysis, including its ability to identify low-abundance proteins that may play a crucial role in tumors and provides a deeper insight into cellular events as protein abundances can be revealed on the level of different subcellular compartments and also protein translocations between different cell parts can be detected (15,16). The cell membrane possesses a number of important biological functions, such as signaling transduction into and out of the cells, ion transport and cell-cell and cell-matrix interactions and communications (17,18). Plasma membrane (PM) proteins are known to have implications in cell proliferation, cell adhesion, cell motility and tumor cell invasion (1921) and account for more than two thirds of currently known drug targets (22,23). Therefore, the cell membrane is of substantial interest with regard to various aspects of tumor, from carcinogenic and metastatic mechanisms to molecular diagnosis and therapeutics. A membrane proteomic analysis offers unprecedented possibilities for identification of tumor biomarkers and therapeutic targets and for understanding carcinogenic mechanisms.

In the present study, isobaric tags for relative and absolute quantitation (iTRAQ) labeling followed by 2D-LC-MS/MS was performed to identify differential PM proteins in AdC tissues and paired normal lung tissues adjacent to tumors. Two different proteins, caveolin-1 and integrin β1, identified by the quantitative proteomics, were selected for validation by western blotting. Furthermore, the clinicopathological significance of integrin β1 was further evaluated using immunohistochemistry of paraffin-embedded archival tissue specimens and statistical analyses. Our data facilitate an understanding of AdC carcinogenesis and mining biomarkers for the diagnosis and treatment of this disease.

Materials and methods

Tissue specimens

Twenty cases of fresh primary lung AdCs and paired paraneoplastic normal lung tissues (PNLTs) adjacent to tumors from the lung AdC patients undergoing curative surgery were obtained from the Department of Cardiothoracic Surgery, Xiangya Hospital of Central South University, China, and stored at −80°C until use. The patients signed an informed consent form for the study which was approved by the local Ethics Committee. Two pairs of matched tumor and normal tissues were used for iTRAQ labeling and eighteen pairs of matched tumor and normal tissues were used for western blotting. An independent set of formalin-fixed and paraffin embedded archival tissue specimens used for immunohistochemistry were obtained from the Department of Pathology, Xiangya Hospital of Central South University and included 42 cases of PNLT, 46 cases of without lymph node metastasis primary AdC (non-LNM AdC) and 62 cases of with lymph node metastatic (LNM) AdC between January 2004 and May 2006 from the AdC patients undergoing curative surgery. The patients recruited in this study had not received chemotherapy, radiotherapy prior to the surgery. The clinicopathological characteristics of the patients used in the present study are noted in Table I.

Table I

Relationship between integrin β1 expression and clinicopathological factors in lung adenocarcinoma.

Table I

Relationship between integrin β1 expression and clinicopathological factors in lung adenocarcinoma.

Integrin β1

NLow (0–2)Moderate (34)High (56)P-value
Age0.87
 <5020767
 ≥5088312928
Gender0.908
 Male60252015
 Female48131520
Differentiation0.893
 Well22877
 Moderate23887
 Poor63222021
pT stage0.072
 T12211101
 T26329331
 T3–4234613
pN stage0.024a
 pN0342761
 pN1329212
 pN2421833
Clinical stage0.016a
 I4230111
 II246180
 III–IV422634
Recurrence0.000a
 Negative261673
 Positive82122832

a P<0.05 by Mann-Whitney U test.

Purification of PM

PM was purified using sucrose density centrifugation in combination with aqueous two-phase partition as described by Cao et al(24). Ten samples were pooled to purified PM for each AdC and PNLT. The purified PM fractions were pelleted by centrifugation and frozen at −80°C until used for protein extraction.

Protein extraction, digestion and labeling with iTRAQ reagents

The PMs were dissolved in lysis buffer (7 M urea, 2 M thiourea, 65 mM DTT, 0.1 mM PMSF) at 4°C for 1 h and then centrifuged at 12,000 rpm for 30 min at 4°C. The supernatant was collected and desalted using 2D Cleanup Kit (Amersham Biosciences). The protein concentration was determined by 2D Quantification Kit (Amersham Biosciences). Trypsin digestion and iTRAQ labeling were performed according to the manufacturer’s protocol (Applied Biosystems). Briefly, 100 μg protein sample was reduced and alkylated and then digested overnight at 37°C with 1 mg/ml trypsin solution and labeled with iTRAQ™ Reagents (Applied Biosystems). The iTRAQ labeling was labeled with 114 and 116 iTRAQ tags for lung AdC samples and 115 and 117 iTRAQs for normal lung tissue samples. Four labeled digests were then mixed and dried using a rotary vacuum concentrator.

LC-MS/MS

The mixed peptides were fractionated by strong cation exchange (SCX) chromatography on an LC-20AD HPLC system (Shimadzu) using a polySulfoethyl column (2.1×100 mm, 5 μm, 300 Å; The Nest Group, Inc.) as previously described by us (25). Briefly, the mixed peptides were desalted with Sep-Pak Cartridge (Waters), diluted with the loading buffer [10 mM KH2PO4 in 25% acetonitrile (ACN), pH 2.8] and loaded onto the column. Buffer A was identical in composition to the loading buffer and buffer B was the same as buffer A except that it contained 350 mM KCl. Separation was performed using a linear binary gradient of 0–80% buffer B in buffer A at a flow rate of 200 μl/min for 60 min. The absorbance at 214 and 280 nm was monitored and a total of 30 SCX fractions were collected along the gradient.

Each SCX fraction was dried down by the rotary vacuum concentrator, dissolved in buffer C (5% ACN, 0.1% FA) and analyzed on Qstar XL (Applied Biosystems) as previously described by us (25). Briefly, peptides were separated on a reverse-phase (RB) column (ZORBAX 300SB-C18 column, 5 μm, 300 Å, 0.1×15 mm; Micromass) using an LC-20AD HPLC system. The HPLC gradient was 5–35% buffer D (95% ACN, 0.1% FA) in buffer C at a flow rate of 0.2 μl/min for 65 min. Survey scans were acquired from 400–1,800 with up to 4 precursors selected for MS/MS from m/z 100–2,000 using a dynamic exclusion of 30 S. The iTRAQ labeled peptides fragmented under CID conditions to give reporter ions at 114.1, 115.1, 116.1 and 117.1 Th. The ratios of peak areas of the iTRAQ reporter ions reflect the relative abundances of the peptides and, consequently, the proteins in the samples. Larger sequence-information-rich fragment ions were also produced under these MS/MS conditions and gave the identity of the protein from which the peptide originated.

Data processing

The software used for data acquisition was Analyst QS 1.1 (Applied Biosystems). The software used for protein identification and quantitation was ProteinPilot™ 3.0 software (Applied Biosystems). The software compares relative intensity of proteins present in samples based on the intensity of reporter ions released from each labeled peptide and automatically calculates protein ratios and P-values for each protein. The data from LC-MS/MS analyses were merged and searched against combined human Swiss-Prot protein sequence database. The following search parameters were used: iTRAQ 4-plex as the sample type, digestion with trypsin and cystein alkylation with methyl methane thiosulfate. The precursor tolerance was set to 150 ppm and the iTRAQ fragment tolerance was set to 0.2 Da, one of missed cleavages permitted, fixed and variable modifications as well as the peak list generating parameters are built-in functions of ProteinPilot. Identified proteins were grouped by the software to minimize redundancy. All peptides used for the calculation of protein ratios were unique to the given protein or proteins within the group and peptides that were common to other isoforms or proteins of the same family were ignored. The protein confidence threshold cutoff was set to 1.3 (unused) with at least one peptide above the 95% confidence level. The average iTRAQ ratios from the two experiments were calculated for each protein. In addition, false discovery rate for the protein identification was calculated by searching against a reversed database.

Bioinformatics analysis

Predictions for putative transmembrane domains (TMDs) in all identified proteins were carried out using the transmembrane hidden Markov model (TMHMM) algorithm available at http://www.cbs.dtu.dk/services/TMHMM (26). The average hydropathy for identified proteins and peptides was calculated using the ProtParam software available at http://www.expasy.org (27). Proteins with positive grand average of hydropathicity (GRAVY) values were considered to be hydrophobic and those with negative values, hydrophilic.

Differential protein validation

Eighteen pairs of matched lung AdC and normal lung tissues were used for western blotting. Briefly, 50 μg of lysates were separated by 10% sodium dodecyl sulfate polyacrylamide gel electrophoresis (SDS-PAGE) and transferred to a PVDF membrane. Blots were blocked with 5% nonfat dry milk for 2 h at room temperature and then incubated with primary anti-caveolin-1, or anti-integrin β1 antibody overnight at 4°C, followed by incubation with a horseradish peroxidase-conjugated secondary antibody (1:3,000; Amersham Biosciences) for 1 h at room temperature. The signal was visualized with an ECL detection reagent and quantities by densitometry using Image J software (http://rsb.info.nih.gov/ij). β-actin was detected simultaneously as a loading control.

Immunohistochemical analysis

Immunohistochemistry was performed on formalin-fixed and paraffin-embedded tissue sections using a standard immunohistochemical technique. Briefly, 4 μm of tissue sections were deparaffinized, rehydrated and treated with an antigen retrieval solution (10 mmol/l sodium citrate buffer, pH 6.0). The sections were incubated with anti-integrin β1 antibody (1:40) overnight at 4°C and were then incubated with 1:1,000 dilution of biotinylated secondary antibody followed by avidin-biotin peroxidase complex (DAKO) according to the manufacturer’s instructions. Finally, tissue sections were incubated with 3′,3′-diaminobenzidine (Sigma-Aldrich) until a brown color developed and counterstained with Harris modified hematoxylin. In negative controls, primary antibodies were omitted.

Immunostaining was blindly evaluated by two investigators in an effort to provide a consensus on staining patterns by light microscopy. A quantitative score was performed by adding the score of staining area and the score of staining intensity for each case to assess the expression levels of the proteins as previously described by us (28). A combined staining score of ≤2 was considered to be low staining (no expression), a score between 3 and 4 was considered to be moderate staining (expression), and a score between 5 and 6 was considered to be strong staining (high expression).

Statistical analysis

All statistical analyses were performed using SPSS 13.0 Software. The significant difference integrin β1 expression between the tumor and normal tissues and primary and metastatic tumors was determined by using the Mann-Whitney U test. Significant differences between the expression of those two factors and clinical variables, including age, gender, histologic type/grade, primary tumor (T) stage, regional lymph node (N) metastasis, clinical stage and recurrence, were compared by the Mann-Whitney U test or ANOVA test. All patients underwent postoperative chemotherapy and were followed-up by telephone to obtain the information of patient outcome. The follow-up period lasted up to 60 months. Relapse-free duration was calculated from the time of surgery to the time of first recurrence after surgery. Overall survival was calculated from the time of surgery to the time of death. Mortality due to lung AdC was considered as outcome; mortality due to other causes was censored and the missing values were replaced by the series mean method. Relapse-free probability curves and overall survival curves were obtained by the Kaplan-Meier method and log-rank testing was used to evaluate the statistical significance of differences. Cox regression analysis was used to evaluate the prognostic significance of clinicopathological factors. A difference of P<0.05 was considered statistically significant.

Results

Identification of differentially expressed proteins in lung AdC and normal lung tissue using iTRAQ-2D-LC-MS/MS

The 2D-LC-MS/MS analysis resulted in the identification of 353 proteins (data not shown) in iTRAQ labeling experiment, with ≥95% confidence, using one or more peptides. We also analyzed identified proteins on the basis of subcellular location and predicted TMDs. Among the proteins with their location annotated, proteins located on PM are known as PM proteins. Of the 353 identified proteins by MS/MS analysis, 224 (63.5%) proteins are PM or PM-related proteins. Of these, 91 (25.8%) are predicted to have one or more TMDs. The GRAVY values of identified PM proteins range from −1.165 to 1.027.

In the present study, the proteins that met the following criteria were considered as differential proteins between the two types of tissues: i) proteins were identified based on ≥2 peptides in the iTRAQ labeling experiments; ii) proteins were quantified with at least two peptides; iii) P-value of identified proteins <0.05; and iv) proteins showed an averaged ratio-fold-change ≥1.5 or ≤0.66. According to these criteria, a total of 45 differentially expressed proteins were found in the two types of tissues, 21 proteins upregulated and 24 downregulated (Table II). MS/MS spectra used for the identification and quantitation of caveolin-1 and integrin β1 are shown in Fig. 1.

Table II

Forty-five differentially expressed proteins in AdC vs. matched PNLT identified by iTRAQ labeling combined with 2D-LC-MS/MS.

Table II

Forty-five differentially expressed proteins in AdC vs. matched PNLT identified by iTRAQ labeling combined with 2D-LC-MS/MS.

Accession no.Protein nameAdC vs. PNLTLocatedTMDsGRAVY
Q9UGM3Deleted in malignant brain tumor 1 protein40.005Secreted0−0.346
P06731Carcinoembryonic antigen-related cell adhesion molecule 513.094PM0−0.411
P15941Mucin-19.713PM1−0.501
P05362Intercellular adhesion molecule 19.034PM1−0.326
P01833Polymeric immunoglobulin receptor6.916PM1−0.405
Q99828Calcium and integrin-binding protein 15.191PM0−0.329
Q14764Major vault protein3.965Cytoplasm0−0.361
P04406 Glyceraldehyde-3-phosphate dehydrogenase3.837Cytoplasm1−0.114
O96009Napsin-A3.768Secreted00.193
P08575Leukocyte common antigen3.375PM2−0.628
A7Y9J9Mucin-5AC3.150Secreted0−0.379
P98088Mucin-5AC (fragments)2.959Secreted0−0.379
O00299Chloride intracellular channel protein 12.619PM0−0.293
Q00610Clathrin heavy chain 12.606M0−0.244
P08311Cathepsin G2.503M0−0.562
P05023 Sodium/potassium-transporting ATPase subunit α-12.456PM100.010
P04083Annexin A12.453M Related0−0.426
O75955Flotillin-12.281PM0−0.338
P01871Ig mu chain C region2.240PM0−0.326
P06733α-enolase1.720M0−0.226
P05556Integrin β11.583PM1−0.455
P27105Erythrocyte band 7 integral membrane protein0.496PM10.037
Q99536Synaptic vesicle membrane protein VAT-1 homolog0.490Cytoplasm0−0.043
Q9BVC6Transmembrane protein 1090.442PM50.411
P11233Ras-related protein Rap-1A0.436PM0−0.683
Q969L2Protein MAL20.436PM40.728
Q99758ATP-binding cassette sub-family A member 30.393PM110.089
P07099Epoxide hydrolase 10.387PM0−0.261
P21333Filamin-A0.341PM0−0.318
P62158Calmodulin0.285PM0−0.671
O60437Periplakin0.284M0−0.982
P21964Catechol O-methyl-transferase0.236PM10.16
Q9UGT4Sushi domain-containing protein 20.229PM1−0.275
P00387NADH-cytochrome b5 reductase 30.223PM0−0.187
P08758Annexin A50.223M related0−0.337
P02730Band 3 anion transport protein0.160PM110.213
O00159Myosin-Ic0.148PM0−0.387
Q16853Membrane primary amine oxidase0.147PM1−0.135
P50148Guanine nucleotide-binding protein G(q) subunit α0.123PM0−0.440
Q03135Caveolin-10.115PM10.054
P10301Ras-related protein R-Ras0.086PM0−0.408
Q9NZN4EH domain-containing protein 20.057PM0−0.309
Q09666Neuroblast differentiation-associated protein AHNAK0.057Nucleus0−0.499
Q6NZI2Polymerase I and transcript release factor0.053M0−0.272
P22748Carbonic anhydrase 40.024PM1−0.576

[i] AdC, lung adenocarcinoma; PM, plasma membrane; PNLT, paraneoplastic normal lung tissue; TMDs, transmembrane domains; GRAVY, grand average of hydropathicity.

Validation of differentially expressed proteins identified by quantitative proteomics

To confirm the expression levels of the differential proteins identified by a proteomics approach, expressions of caveolin-1 and integrin β1 in 18 pairs of AdC and matched PNLT adjacent to tumors were detected by western blotting. As shown in Fig. 2, caveolin-1 was downregulated, whereas integrin β1 was upregulated in AdC compared with PNLT, which is consistent with the findings in MS/MS analysis.

Expression of integrin β1 in PNLT, primary lung AdC and lymph node metastases

We detected the expression of integrin β1 using immunohistochemical staining in 46 cases of non-LNM AdC, 62 cases of LNM AdC and 42 cases of PNLT. As shown in Fig. 3 and Table III, integrin β1 was significantly upregulated in non-LNM AdC vs. PNLT and in LNM AdC vs. non-LNM AdC.

Table III

Difference of integrin β1 expression in PNLT, non-LNM AdC and LNM AdC.

Table III

Difference of integrin β1 expression in PNLT, non-LNM AdC and LNM AdC.

Score

nLow (0–2)Moderate (34)High (56)P-value
Integrin β1
 PNLT4234620.001a,b
 Non-LNM AdC4621205
 LNM AdC621715300.001a,c

a P<0.05 by Mann-Whitney U test.

b PNLT vs. non-LNM AdC;

c non-LNM AdC vs. LNM AdC.

{ label (or @symbol) needed for fn[@id='tfn6-or-30-01-0341'] } PNLT, paraneoplastic normal lung tissue; non-LNM AdC, lung AdC without lymph node metastasis; LNM AdC, lung AdC with lymph node metastasis.

Correlation of integrin β1 expression in primary AdC with clinicopathological factors

Table I shows the correlation of several clinical pathological factors with integrin β1 expression status in 108 cases of primary AdC (including 46 cases of non-LNM AdC and 62 cases of LNM AdC). Integrin β1 expression levels were significantly correlated with clinical stage, recurrence and lymph node metastasis. Tumors with the upregulation of integrin β1 tended to have a more advanced clinical stage and more frequent recurrence and lymph node metastasis. No significant correlations were found between the expression of integrin β1 and other characteristics, including gender, age, tumor size and tumor differentiation.

Correlation of postoperative relapse and survival with integrin β1 expression

By the end of the study, 82 of the 108 patients had died, 24 patients were still alive and 2 patients had been lost to follow-up. The relapse-free times of the patients with low, moderate and high expression of integrin β1 were 51.6±10.1, 47.1±13.3 and 16.5±10 months, respectively. The relapse-free probability curve showed that the relapse rate was significantly increased along with increasing integrin β1 expression (Fig. 4, left). The mean survival rates of the patients with low, moderate and high expression of integrin β1 were 53±8.7, 38.4±12.9 and 17.8±9.5 months, respectively. The survival curves showed that the overall survival rate was significantly decreased along with increasing expressions of integrin β1 (Fig. 4, right). In univariate analysis (Table IV), increased postoperative relapse and decreased survival were correlated with advanced clinical stage, lymph node metastasis, increasing expressions of integrin β1. In multivariate analysis (Table V), advanced clinical stage, lymph node metastasis and increasing expressions of integrin β1 remained the significant independent prognostic factors of increased relapse rate and decreased overall survival rate.

Table IV

Univariate Cox regression analysis of relapse-free and overall survival for integrin β1 expression.

Table IV

Univariate Cox regression analysis of relapse-free and overall survival for integrin β1 expression.

Relapse-free probabilityOverall survival


VariablesHR (95% CI)P-valueHR (95% CI)P-value
Age 0.501(0.308–0.813)0.0940.465 (0.287–0.753)0.120
Gender 0.926(0.596–1.438)0.7310.929 (0.596–1.4520.747
 Male (reference)1.0001.000
 Female 0.926(0.596–1.438)0.7310.929 (0.595–1.452)0.747
Tumor differentiation0.7000.502
 Well (reference)1.0001.000
 Moderate0.871 (0.486–1.56)0.4261.384 (0.681–2.811)0.369
 Poor1.504 (0.618–1.796)0.7501.210 (0.769–1.904)0.410
pT stage0.5130.779
 T1 (reference)1.0001.000
 T20.675 (0.341–1.336)0.6111.009 (0.352–2.895)0.861
 T3–40.781 (0.449–1.360)0.2571.288 (0.560–2.962)0.551
pN stage0.000a0.000a
 N0 (reference)1.0001.000
 N114.812 (6.024–36.421)0.00016.639 (6.513–42.507)0.000
 N2–311.357 (6.244–20.660)0.00011.988 (6.518–22.045)0.006
Clinical stage0.000a0.000a
 I (reference)1.0001.000
 II19.744 (7.291–53.464)0.00018.973 (7.073–50.899)0.000
 III–IV21.445 (10.799–42.585)0.00020.501 (13.340–44648)0.000
Integrin β1 expression0.000a0.000a
 Low1.0001.000
 Moderate2.202 (1.231–3.935)0.0082.626 (1.442–4.780)0.002
 High6.592 (4.106–10.584)0.0006.183 (3.851–9.929)0.000

a Overall P<0.05.

{ label (or @symbol) needed for fn[@id='tfn8-or-30-01-0341'] } HR, hazard ratio; CI, confidence interval.

Table V

Multivariate Cox regression analysis of relapse-free and overall survival for integrin β1 expression.

Table V

Multivariate Cox regression analysis of relapse-free and overall survival for integrin β1 expression.

Relapse-free probabilityOverall survival


VariablesHR (95% CI)P-valueHR (95% CI)P-value
pN stage0.011a0.006a
 N0 (reference)1.0001.000
 N1 5.354(1.537–18.644)0.008 7.484(2.040–27.463)0.002
 N2–3 3.508(1.493–8.240)0.004 3.551(1.477–8.537)0.005
Clinical stage0.003a0.005a
 I (reference)1.0001.000
 II 4.440(1.186–16.619)0.027 4.165(1.130–15.348)0.032
 III–IV 5.167(2.028–13.163)0.001 4.745(1.862–12.094)0.001
Integrin β1 expression0.000a0.000a
 Low1.0001.000
 Moderate 2.086(1.010–4.310)0.047 2.866(1.311–6.265)0.008
 High 4.045(2.169–7.543)0.000 3.557(1.879–6.734)0.000

a Overall P<0.05.

{ label (or @symbol) needed for fn[@id='tfn10-or-30-01-0341'] } HR, hazard ratio; CI, confidence interval.

Discussion

In the present study, iTRAQ labeling combined with 2D-LC-MS/MS was performed to identify differential PM proteins in AdC and PNLT. As a result, 45 differentially expressed proteins were identified and differential PM proteins, caveolin-1 and integrin β1, were selectively validated. Next, the clinicopathological significance of integrin β1 was further evaluated using immunohistochemistry of paraffin-embedded archival tissue specimens and statistical analysis. Results show that integrin β1 is a potential biomarker for LNM and prognosis of AdC.

Integrins, a large family of membrane receptors, are α/β-heterodimeric transmembrane adhesion molecules which bind to specific ECM ligands (29,30). Integrin β1 is an important subunit of integrins and recognizes the sequence R-G-D in a wide array of ligands. Integrin β1 comprises at least eight isoform members and plays a role in cell signaling and thereby defines cellular shape, mobility and regulates the cell cycle (31). Integrin β1 has been shown to regulate signaling through transmembrane growth factor receptors such as epidermal growth factor receptor and transforming growth factor-β receptor (32,33). Integrin β1 has been reported as critical for TGF-β1-mediated transcription and epithelial cell plasticity in vitro(34). Integrin β1 was involved in the development and progression of carcinogenesis in several types of cancer, including kidney cancer, breast cancer and fibrosarcoma, bladder and colon carcinoma (34,35). Bredin et al(36) found that integrin β1 was involved in lung cancer cell migration in vitro towards fibronectin, laminin and type IV collagen. Upregulation of integrin β1 is an important factor for gefitinib resistance in the NSCLC cell line (37) and overexpression of integrin β1 is correlated with the invasion and metastasis events of HCC in patients (38). Integrin β1 is correlated with highly invasive and metastatic behavior and is a poor prognostic factor in patients with SCLC (39,40). Herein, an increase in integrin β1 expression level was associated with advanced clinical stage and lymph node metastases, suggesting that integrin β1 is associated with the progression and LNM of lung AdC. In addition, a univariate analysis indicated that high integrin β1 expression is strongly associated with increased tumor relapse and a multivariate analysis further indicated that high integrin β1 expression is an independent relapse factor for increased tumor relapse in lung AdC.

In conclusion, the present study not only confirmed expression of caveolin-1 and integrin β1 by proteomic approaches in primary AdC and paired normal lung tissues adjacent to tumors, but also showed that primary AdC with higher integrin β1 expression tended to have later clinical stage, more frequent recurrence and LNM. Furthermore, survival curves showed that the AdC patients with integrin β1 upregulation had a poor prognosis. Multivariate analysis confirmed that integrin β1 expression was an independent prognostic indicator. Findings of the present study may have clinical value in predicting the prognosis of AdC and identifying AdC patients that are at high risk of metastasis and recurrence.

Acknowledgements

The authors acknowledge the grants from the Major New Drug Discovery Science and Technology of China (2012ZX09303013-006) and the National Natural Science Foundation of China (81272609, 21105129, 81102046). We also acknowledge the Institutes of Biomedical Sciences of Fudan University Xiao-Hui Liu for her assistance with iTRAQ analysis.

Abbreviations:

AdC

lung adenocarcinoma

PM

plasma membrane

PNLT

paraneoplastic normal lung tissue

non-LNM AdC

without lymph node metastasis primary AdC

LNM AdC

with lymph node metastatic primary AdC

SDS-PAGE

sodium dodecyl sulfate-polyacrylamide gel electrophoresis

SCX

strong cation exchange

ACN

acetonitrile

GRAVY

grand average of hydropathicity

iTRAQ

isobaric tags for relative and absolute quantitation

References

1 

Parkin DM, Bray F, Ferlay J and Pisani P: Global cancer statistics 2002. CA Cancer J Clin. 55:74–108. 2005. View Article : Google Scholar

2 

Yang L, Parkin DM, Li LD, Chen YD and Bray F: Estimation and projection of the national profile of cancer mortality in China: 1991–2005. Br J Cancer. 90:2157–2166. 2004.PubMed/NCBI

3 

Howe HL, Wingo PA, Thun MJ, Ries LA, Rosenberg HM, Feigal EG and Edwards BK: Annual report to the nation on the status of cancer (1973 through 1998), featuring cancers with recent increasing trends. J Natl Cancer Inst. 93:824–842. 2001. View Article : Google Scholar : PubMed/NCBI

4 

Little AG, Gay EG, Gaspar LE and Stewart AK: National survey of non-small cell lung cancer in the United States: epidemiology, pathology and patterns of care. Lung Cancer. 57:253–260. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Jemal A, Siegel R, Ward E, Murray T, Xu J, Smigal C and Thun MJ: Cancer statistics, 2006. CA Cancer J Clin. 56:106–130. 2006. View Article : Google Scholar

6 

Steeg PS, Bevilacqua G, Kopper L, Thorgeirsson UP, Talmadge JE, Liotta LA and Sobel ME: Evidence for a novel gene associated with low tumor metastatic potential. J Natl Cancer Inst. 80:200–204. 1988. View Article : Google Scholar : PubMed/NCBI

7 

Chen JJ, Peck K, Hong TM, Yang SC, Sher YP, Shih JY, Wu R, et al: Global analysis of gene expression in invasion by a lung cancer model. Cancer Res. 61:5223–5230. 2001.PubMed/NCBI

8 

Nakamura N, Kobayashi K, Nakamoto M, Kohno T, Sasaki H, Matsuno Y and Yokota J: Identification of tumor markers and differentiation markers for molecular diagnosis of lung adenocarcinoma. Oncogene. 25:4245–4255. 2006. View Article : Google Scholar : PubMed/NCBI

9 

Tian T, Hao J, Xu A, Hao J, Luo C, Liu C, Huang L, Xiao X and He D: Determination of metastasis-associated proteins in non-small cell lung cancer by comparative proteomic analysis. Cancer Sci. 98:1265–1274. 2007. View Article : Google Scholar : PubMed/NCBI

10 

Chen G, Gharib TG, Huang CC, Thomas DG, Shedden KA, Taylor JM, Kardia SL, et al: Proteomic analysis of lung adenocarcinoma: identification of a highly expressed set of proteins in tumors. Clin Cancer Res. 8:2298–2305. 2002.PubMed/NCBI

11 

Chen G, Gharib TG, Thomas DG, Huang CC, Misek DE, Kuick RD, Giordano TJ, et al: Proteomic analysis of eIF-5A in lung adenocarcinomas. Proteomics. 3:496–504. 2003. View Article : Google Scholar : PubMed/NCBI

12 

Rho JH, Roehrl MH and Wang JY: Tissue proteomics reveals differential and compartment-specific expression of the homologs transgelin and transgelin-2 in lung adenocarcinoma and its stroma. J Proteome Res. 8:5610–5618. 2009. View Article : Google Scholar : PubMed/NCBI

13 

Liu YF, Xiao ZQ, Li MX, Li MY, Zhang PF, Li C, Li F, et al: Quantitative proteome analysis reveals annexin A3 as a novel biomarker in lung adenocarcinoma. J Pathol. 217:54–64. 2009. View Article : Google Scholar : PubMed/NCBI

14 

Liu YF, Zhang PF, Li MY, Li QQ and Chen ZC: Identification of annexin A1 as a proinvasive and prognostic factor for lung adenocarcinoma. Clin Exp Metastasis. 28:413–425. 2011. View Article : Google Scholar : PubMed/NCBI

15 

Emmott E, Wise H, Loucaides EM, Matthews DA, Digard P and Hiscox JA: Quantitative proteomics using SILAC coupled to LC-MS/MS reveals changes in the nucleolar proteome in influenza A virus-infected cells. J Proteome Res. 9:5335–5345. 2010. View Article : Google Scholar : PubMed/NCBI

16 

Qattan AT, Mulvey C, Crawford M, Natale DA and Godovac-Zimmermann J: Quantitative organelle proteomics of MCF-7 breast cancer cells reveals multiple subcellular locations for proteins in cellular functional processes. J Proteome Res. 9:495–508. 2010. View Article : Google Scholar

17 

Wu CC, MacCoss MJ, Howell KE and Yates JR III: A method for the comprehensive proteomic analysis of membrane proteins. Nat Biotechnol. 21:532–538. 2003. View Article : Google Scholar : PubMed/NCBI

18 

Wu CC and Yates JR III: The application of mass spectrometry to membrane proteomics. Nat Biotechnol. 21:262–267. 2003. View Article : Google Scholar : PubMed/NCBI

19 

Dowling P, Meleady P, Dowd A, Henry M, Glynn S and Clynes M: Proteomic analysis of isolated membrane fractions from superinvasive cancer cells. Biochim Biophys Acta. 1774:93–101. 2007. View Article : Google Scholar : PubMed/NCBI

20 

Liang X, Zhao J, Hajivandi M, Wu R, Tao J, Amshey JW and Pope RM: Quantification of membrane and membrane-bound proteins in normal and malignant breast cancer cells isolated from the same patient with primary breast carcinoma. J Proteome Res. 5:2632–2641. 2006. View Article : Google Scholar : PubMed/NCBI

21 

Stockwin LH, Blonder J, Bumke MA, Lucas DA, Chan KC, Conrads TP, Issaq HJ, et al: Proteomic analysis of plasma membrane from hypoxia-adapted malignant melanoma. J Proteome Res. 5:2996–3007. 2006. View Article : Google Scholar : PubMed/NCBI

22 

Slamon DJ, Leyland-Jones B, Shak S, Fuchs H, Paton V, Bajamonde A, Fleming T, et al: Use of chemotherapy plus a monoclonal antibody against HER2 for metastatic breast cancer that overexpresses HER2. N Engl J Med. 344:783–792. 2001. View Article : Google Scholar : PubMed/NCBI

23 

Oh P, Li Y, Yu J, Durr E, Krasinska KM, Carver LA, Testa JE and Schnitzer JE: Subtractive proteomic mapping of the endothelial surface in lung and solid tumors for tissue-specific therapy. Nature. 429:629–635. 2004. View Article : Google Scholar : PubMed/NCBI

24 

Cao R, Li X, Liu Z, Peng X, Hu W, Wang X, Chen P, et al: Integration of a two-phase partition method into proteomics research on rat liver plasma membrane proteins. J Proteome Res. 5:634–642. 2006. View Article : Google Scholar : PubMed/NCBI

25 

Xiao Z, Li G, Chen Y, Li M, Peng F, Li C, Li F, et al: Quantitative proteomic analysis of formalin-fixed and paraffin-embedded nasopharyngeal carcinoma using iTRAQ labeling, two-dimensional liquid chromatography, and tandem mass spectrometry. J Histochem Cytochem. 58:517–527. 2010. View Article : Google Scholar

26 

Krogh A, Larsson B, von Heijne G and Sonnhammer EL: Predicting transmembrane protein topology with a hidden Markov model: application to complete genomes. J Mol Biol. 305:567–580. 2001. View Article : Google Scholar : PubMed/NCBI

27 

Kyte J and Doolittle RF: A simple method for displaying the hydropathic character of a protein. J Mol Biol. 157:105–132. 1982. View Article : Google Scholar : PubMed/NCBI

28 

Cheng AL, Huang WG, Chen ZC, Peng F, Zhang PF, Li MY, Li F, et al: Identification of novel nasopharyngeal carcinoma biomarkers by laser capture microdissection and proteomic analysis. Clin Cancer Res. 14:435–445. 2008. View Article : Google Scholar : PubMed/NCBI

29 

Tarone G, Hirsch E, Brancaccio M, De Acetis M, Barberis L, Balzac F, Retta SF, et al: Integrin function and regulation in development. Int J Dev Biol. 44:725–731. 2000.PubMed/NCBI

30 

Hollenbeck ST, Itoh H, Louie O, Faries PL, Liu B and Kent KC: Type I collagen synergistically enhances PDGF-induced smooth muscle cell proliferation through pp60src-dependent crosstalk between the α2β1 integrin and PDGFβ receptor. Biochem Biophys Res Commun. 325:328–337. 2004. View Article : Google Scholar : PubMed/NCBI

31 

Moro L, Venturino M, Bozzo C, Silengo L, Altruda F, Beguinot L, Tarone G and Defilippi P: Integrins induce activation of EGF receptor: role in MAP kinase induction and adhesion-dependent cell survival. EMBO J. 17:6622–6632. 1998. View Article : Google Scholar : PubMed/NCBI

32 

Bhowmick NA, Zent R, Ghiassi M, McDonnell M and Moses HL: Integrin β1 signaling is necessary for transforming growth factor-β activation of p38MAPK and epithelial plasticity. J Biol Chem. 276:46707–46713. 2001.

33 

Frish SM and Francis H: Disruption of epithelial cell-matrix interaction induces apoptosis. J Cell Biol. 124:619–626. 1994. View Article : Google Scholar : PubMed/NCBI

34 

Aoudjit F and Vuori K: Integrin signaling inhibits paclitaxel-induced apoptosis in breast cancer cells. Oncogene. 20:4995–5004. 2001. View Article : Google Scholar : PubMed/NCBI

35 

Yamada KM, Kennedy DW, Yamada SS, Gralnick H, Chen WT and Akiyama SK: Monoclonal antibody and synthetic peptide inhibitors of human tumor cell migration. Cancer Res. 50:4485–4496. 1990.PubMed/NCBI

36 

Bredin CG, Sundqvist KG, Hauzenberger D and Klominek J: Integrin dependent migration of lung cancer cells to extracellular matrix components. Eur Respir J. 11:400–407. 1998. View Article : Google Scholar : PubMed/NCBI

37 

Ju LX, Zhou CC, Li W and Yan LH: Integrin beta1 over-expression associates with resistance to tyrosine kinase inhibitor gefitinib in non-small cell lungcancer. J Cell Biochem. 111:1565–1574. 2010. View Article : Google Scholar : PubMed/NCBI

38 

Zhao G, Cui J, Qin Q, Zhang J, Liu L, Deng S, Wu C, et al: Mechanical stiffness of liver tissues in relation to integrin β1 expression may influence the development of hepatic cirrhosis and hepatocellular carcinoma. J Surg Oncol. 102:482–489. 2010.PubMed/NCBI

39 

Oshita F, Kameda Y, Ikehara M, Tanaka G, Yamada K, et al: Increased expression of integrin beta1 is a poor prognostic factor in small-cell lung cancer. Anticancer Res. 22:1065–1070. 2002.PubMed/NCBI

40 

Chang MH, Lee K, Lee KY, Kim YS, Kim YK and Kang JH: Prognostic role of integrin β1, E-cadherin, and rac1 expression in small cell lung cancer. APMIS. 120:28–38. 2012.

Related Articles

Journal Cover

July 2013
Volume 30 Issue 1

Print ISSN: 1021-335X
Online ISSN:1791-2431

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang P, Zeng G, Yi L, Liu J, Wan XX, Qu J, Li J, Li C, Tang C, Hu R, Hu R, et al: Identification of integrin β1 as a prognostic biomarker for human lung adenocarcinoma using 2D-LC‑MS/MS combined with iTRAQ technology. Oncol Rep 30: 341-349, 2013
APA
Zhang, P., Zeng, G., Yi, L., Liu, J., Wan, X., Qu, J. ... Xiao, Z. (2013). Identification of integrin β1 as a prognostic biomarker for human lung adenocarcinoma using 2D-LC‑MS/MS combined with iTRAQ technology. Oncology Reports, 30, 341-349. https://doi.org/10.3892/or.2013.2477
MLA
Zhang, P., Zeng, G., Yi, L., Liu, J., Wan, X., Qu, J., Li, J., Li, C., Tang, C., Hu, R., Ye, X., Chen, Y., Chen, Z., Xiao, Z."Identification of integrin β1 as a prognostic biomarker for human lung adenocarcinoma using 2D-LC‑MS/MS combined with iTRAQ technology". Oncology Reports 30.1 (2013): 341-349.
Chicago
Zhang, P., Zeng, G., Yi, L., Liu, J., Wan, X., Qu, J., Li, J., Li, C., Tang, C., Hu, R., Ye, X., Chen, Y., Chen, Z., Xiao, Z."Identification of integrin β1 as a prognostic biomarker for human lung adenocarcinoma using 2D-LC‑MS/MS combined with iTRAQ technology". Oncology Reports 30, no. 1 (2013): 341-349. https://doi.org/10.3892/or.2013.2477