Analysis of the gene expression profile in response to human epididymis protein 4 in epithelial ovarian cancer cells

  • Authors:
    • Liancheng Zhu
    • Qian Guo
    • Shan Jin
    • Huilin Feng
    • Huiyu Zhuang
    • Cong Liu
    • Mingzi Tan
    • Juanjuan Liu
    • Xiao Li
    • Bei Lin
  • View Affiliations

  • Published online on: July 11, 2016     https://doi.org/10.3892/or.2016.4926
  • Pages: 1592-1604
Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

Currently, there are emerging multiple studies on human epididymis protein 4 (HE4) in ovarian cancer. HE4 possesses higher sensitivity and specificity than CA125 in the confirmative early diagnosis for ovarian cancer. Although much attention has been given to explore its clinical application, research of the basic mechanisms of HE4 in ovarian cancer are still unclear. In the present study, we provide fundamental data to identify full-scale differentially expressed genes (DEGs) in response to HE4 by use of human whole-genome microarrays in human epithelial ovarian cancer cell line ES-2 following overexpression and silencing of HE4. We found that a total of 717 genes were upregulated and 898 genes were downregulated in the HE4-overexpressing cells vs. the HE4-Mock cells, and 166 genes were upregulated and 285 were downregulated in the HE4-silenced cells vs. the HE4-Mock cells. An overlap of 16 genes consistently upregulated and 8 genes downregulated in response to HE4 were noted. These DEGs were involved in MAPK, steroid biosynthesis, cell cycle, the p53 hypoxia pathway, and focal adhesion pathways. Interaction network analysis predicted that the genes participated in the regulatory connection. Highly differential expression of the FOXA2, SERPIND1, BDKRD1 and IL1A genes was verified by quantitative real-time PCR in 4 cell line samples. Finally, SERPIND1 (HCII) was validated at the protein level by immunohistochemistry in 107 paraffin-embedded ovarian tissues. We found that SERPIND1 may act as a potential oncogene in the development of ovarian cancer. The present study displayed the most fundamental and full-scale data to show DEGs in response to HE4. These identified genes may provide a theoretical basis for investigations of the underlying molecular mechanism of HE4 in ovarian cancer.

Introduction

Ovarian cancer is the most lethal gynecological malignancy and accounts for 25–30% of all malignant tumors in the female reproductive tract. An estimated 22,280 new cases of ovarian cancer and 14,240 related deaths were reported in 2016 in the US (1). Due to its innocuous symptoms, most ovarian cancer patients are diagnosed at the late stage. Despite the development of new antitumor drugs and the improvement of surgical treatment, the majority of patients with advanced disease (stages III–IV) eventually relapse and the 5-year survival rate of late stage patients is only 30% (2). Therefore, early diagnosis of ovarian cancer is crucial to reduce mortality and improve the prognosis for eventual survival. Human epididymis protein 4 (HE4), also known as whey-acidic-protein (WAP) four-disulfide core domain protein 2 (WFDC2), was first found overexpressed in ovarian cancer tissue in 1999 (3). Although it was originally recognized as a small secreted protein that plays a role in sperm maturation in males (4), numerous studies have found that HE4 is a valuable serum biomarker possessing higher sensitivity and specificity than CA125 in the confirmative early diagnosis for epithelial ovarian cancer (EOC) (5,6), and the combined use of HE4 with pelvic ultrasound achieved the best sensitivity for detecting ovarian cancers among the different algorithms tested (7). It seems to be an independent predictive factor for ideal tumor cytoreductive surgery and maintains its prognostic role even after recurrence (8). Moreover, recent investigations have shown that serum HE4 could predict chemotherapy response during first-line chemotherapy (9), and high HE4 levels are correlated with chemoresistance and decreased survival rates in EOC patients (10,11). HE4 shows its potential application as a biomarker for early detection and discrimination of endometrial (12,13) and non-small cell lung cancer (14), and pancreatic adenocarcinoma (15). A high serum HE4 level may be a useful biomarker for the poor prognosis in non-small cell lung cancer patients (16,17).

Over the past decade, much attention has been given to explore the clinical application of HE4 as a biomarker. In recent years, several studies have reported that HE4 acts as an oncogene implicated in various cancer behaviors, such as cell adhesion (18), proliferation, migration, metastasis (1921) and chemoresistance (19,22). The molecular mechanisms may be attributed to the activation of epidermal growth factor (EGF) (18), vascular endothelial growth factor (VEGF) (19), HIF1α (19) or the interaction with Annexin A2 (23), and Lewis y glycosylation (24). Nevertheless, investigation of the role of HE4 in the malignant biological behaviors of ovarian cancer is limited.

Therefore, in the present study, we sought to investigate alteration of the gene expression profile in response to HE4 in ovarian cancer cells. The results obtained from this research may lead to a better understanding of the molecular mechanisms associated with HE4 in ovarian cancer and to facilitate the early diagnosis and therapeutic treatment of ovarian cancer.

Materials and methods

Cell culture, construction of expression vectors and HE4 gene transfection

Human EOC ES-2 cells were purchased from the American Type Culture Collection (ATCC; Manassas, VA, USA) and maintained in RPMI-1640 medium with 10% fetal bovine serum (FBS) and 1% penicillin/streptomycin, at 37°C in a humidified atmosphere with 5% CO2. An HE4 expression construct was generated by subcloning PCR-amplified full-length human HE4 cDNA into the pEGFP-N1 or pCMV6 plasmid. The following primers were used: P1, 5′-TCC GCT CGA GAT GCC TGC TTG TCG CCT AG-3′ and P2, 5′-ATG GGG TAC CGT GAA ATT GGG AGT GAC ACA GG-3′. Two shRNA expression vectors for human HE4 were constructed using the vector pSilencer. The mRNA target sequences chosen for designing HE4-shRNA were: GTC CTG TGT CAC TCC CAA T for HE4-shRNA1 and GAT GAA ATG CTG CCG CAA T for HE4-shRNA2. Transfection was carried out using liposomes with a vector transfection kit according to the instructions. Regarding the stable cell lines, HE4-overexpressing (HE4-H) (O), HE4-shRNA low-expressing (HE4-L) (S) and their respective empty-plasmid transfected cell lines [HE4-H-Mock (OV) and HE4-L-Mock (SV)] were selected for 14 days with G418 (800 μg/ml) (Invitrogen, Carlsbad, CA, USA). All cell lines were labeled and are listed in Table I.

Table I

Cell line sample description and RNA qualification.

Table I

Cell line sample description and RNA qualification.

Sample IDLabelOD260/280OD260/230RINResults
HE4-HO2.022.219.30Pass
HE4-H-vectorOV2.022.358.60Pass
HE4-LS2.022.2410.0Pass
HE4-L-vectorSV2.032.099.00Pass

[i] RIN, RNA Integrity number; HE4, human epididymis protein 4.

Transfection identification by quantitative real-time PCR and western blotting
Quantitative real-time polymerase chain reaction (RT-PCR)

Total RNA was extracted with TRIzol reagent and reverse-transcribed to cDNA using SuperScript III (both from Invitrogen). Quantitative real-time PCR was performed on Roche LightCycler 480 (Roche Diagnostics, Mannheim, Germany) sequence detection system. The primers for HE4 were: 5′-AGT GTC CTG GCC AGA TGA AAT G-3′ for forward and 5′-CAG GTG GGC TGG AAC CAG AT-3′ for reverse. GAPDH was used to normalize the quantity of complementary DNA. PCR reactions of each sample were carried out in triplicate. The cycle steps were 95°C for 5 min, 40 cycles of 95°C for 15 sec, 60°C for 1 min and 72°C for 20 sec.

Western blotting

Western blotting was performed as previously described (23). The HE4 antibody (rabbit polyclonal; 1:40; Abcam, Cambridge, MA, USA) was used and the negative control contained only HE4 antibody without protein. The protein bands were visualized by ImageJ 1.31v and normalized relative to the GAPDH protein expression level.

Total RNA extraction and quantity control

Two pairs of cells were prepared for gene chip hybridization analysis: HE4-overexpressing cells vs. HE4-overexpressing vector cells, and HE4-shRNA cells vs. HE4-shRNA vector cells. Total RNA was extracted from all cell lines using RNeasy Mini kit and further purified with RNeasy MinElute Cleanup columns (both from Qiagen, Valencia, CA, USA). RNA quantity and purity were assessed using NanoDrop ND-1000. Pass criteria for absorbance ratios were established as A260/A280 ≥1.8 and A260/A230 ≥1. RNA integrity number (RIN) values were ascertained using Agilent RNA 6000 Nano assay to determine RNA integrity. Pass criterion for RIN value was established at ≥6. The RNA samples in each group are labeled in Table I. The total RNA of each cell line was allocated to 3 chips for further hybridization and analysis to decrease the experimental error.

Gene chip hybridization, data collection and enrichment analysis

Purified RNA samples were submitted to Human Whole Genome OneArray® (HOA6.1) with Phalanx hybridization buffer using Phalanx Hybridization System for microarray analysis. This array contains 30,275 DNA oligonucleotide probes, and each probe is a 60-mer designed in the sense direction. Among the probes, 29,187 probes correspond to the annotated genes in RefSeq v38 and Ensembl v56 database. In addition, 1,088 control probes are also included. After 16 h of hybridization at 50°C, non-specific binding targets were washed away by 3 different washing steps (wash I, 42°C for 5 min; wash II, 42°C for 5 min, 25°C for 5 min; and wash III, rinse 20 times), and the slides were dried by centrifugation and scanned by Axon 4000B scanner (Molecular Devices, Sunnyvale, CA, USA). The intensities of each probe were obtained by GenePix 4.1 software (Molecular Devices). In the present study, 12 chips were used to compare the differentially expressed genes (DEGs). Scatter plots were conducted to show the repeatability of the expression signal between technical repeats. Histogram plots were made to show the fold-change distribution of all probes excluding control and flagged probes. Volcano plots were performed to visually show a distinguishable gene expression profile among samples. Venny-diagram was performed to identify the coordinated DEGs in response to HE4 (http://bioinfogp.cnb.csic.es/tools/venny/). Fold-changes were calculated by Rosetta Resolver 7.2 with error model adjusted by Amersham Pairwise Ration Builder for signal comparison of sample. Standard selection criteria to identify DEGs were as follows: i) log2 |fold-change| ≥1 and P<0.05; ii) log2 ratios ̔NA̓ and the differences in intensity between the two samples ≥1,000.

Validation of gene expression by RT-PCR and immunohistochemical (IHC) staining
RT-PCR

RT-PCR was performed in triplicate with primer sets and probes that were specific for 4 selected genes that were found to be significantly differentially expressed: FOXA2, SERPIND1, BDKRD1 and IL1A. The methods are shown as above.

IHC

To validate the DEGs at the protein level, IHC staining in ovarian samples was conducted. In our previous studies, we established a group of ovarian paraffin-embedded samples (23) including 50 malignant, 27 borderline, 15 benign and 15 normal ovarian tissues. In view of the further investigation, SERPIND1 was selected for IHC staining in these samples and then compared with the expression of HE4. The working dilution for SERPIND1 (#12741-1-AP; ProteinTech, Chicago, IL, USA) was 1:400. The staining of hepatic cancer samples was chosen as the positive control and omission of the primary antibody was designed as the negative control. The staining procedures were performed as previously described (23). Regarding the evaluation method, in brief, the presence of brown-colored granules on the cell membrane or in the cytoplasm was taken as a positive signal, and was classified according to color intensity as follows: not colored, light yellow, brown and tan were recorded as 0, 1, 2 and 3, respectively. A positive cell staining rate of <5, 5–25, 26–50 and 51–75%, and >75% were recorded as 0, 1, 2, 3 and 4. The final score was determined by multiplying the positive cell staining rate and the score values: 0–2 was considered negative (−), 3–4 was (+), 5–8 was (++), and 9–12 was (+++).− and + were considered as low expression; ++ and +++ as high expression. Two observers evaluated the sections to control error. Survival analysis was performed on these patients. The overall survival (OS) time was defined from the date of surgery (earliest was in August 2008) to the date of death or the last follow-up (September, 2015).

Enrichment analysis of DEGs

All the DEGs in each pair group were prepared to run Gene Ontology (GO) and canonical pathways analyses (Biocarta and KEGG). Gene interaction networks were visualized by Cytoscape (25). The property of the network was analyzed with the plug-in network analysis.

Statistical analysis

Statistical analyses were performed using the SPSS program (version 22 for Mac; SPSS, Inc., Chicago, IL, USA) and GraphPad Prism 6 (version 6.0 h for Mac; GraphPad Prism Software Inc., San Diego, CA, USA). Chi-square and one-way ANOVA with LSD or Bonferroni post hoc test was used for comparison between >2 groups. The correlation coefficient R of SERPIND1 with HE4 was calculated by Spearman correlation analysis. Quantitative data are presented as mean ± SD. As to the analysis of quantitative RT-PCR result, the data are expressed as mean ± SEM. Survival analysis was analyzed using Kaplan-Meier curves by log-rank test. A P-value of <0.05 was considered to indicate a statistically significant result.

Results

Identification of HE4 gene transfection

Stable transfected cell lines were established using the ES-2 cells. The gene and protein expression levels of HE4 were obviously increased after HE4 transfection and decreased after shRNA transfection, as detected by quantitative real-time PCR (Fig. 1A) and western blotting (Fig. 1B and C), whereas there was no statistical difference in regards to HE4 in the mock and untreated cells.

Gene expression analysis and clustering

The expression profiles of all the samples passed the microarray quality control (Table I). Histogram plots of fold-change distribution of all probes excluding control and flagged probes were conducted for all signals collected (Fig. 2A), and the volcano plots revealed the DEGs for each pair of gene chips (Fig. 2B). A total of 717 genes were upregulated and 898 genes were downregulated in O vs. OV, 166 genes were upregulated and 285 were downregulated in S vs. SV. The top 20 DEGs in each group are shown in Table II. Venn diagrams showed that an overlap of 16 genes was consistently upregulated and 8 genes downregulated in response to HE4 (Fig. 3).

Table II

Top 20 differentially expressed genes in each group.

Table II

Top 20 differentially expressed genes in each group.

Gene symbolRefSeqDescriptionLog2 (fold-change)P-value
O vs. OV upregulated
 EPB41L3NM 012307.2Erythrocyte membrane protein band 4.1-like 35.2462320
 FOXA2NM 153675.2Forkhead box A24.6199213.02E-32
 GDF15NM 004864.2Growth differentiation factor 154.6120660
 TAGLN3NM 001008273.1Transgelin 34.6039760
 C12orf39NM 030572.2Chromosome 12 open reading frame 394.1973290
 MMP3NM 002422.3Matrix metallopeptidase 33.9617720
 IL11NM 000641.3Interleukin 113.8875011.55E-14
 IL24NM 001185158.1Interleukin 243.6740280
 FAM24BNM 152644.2Family with sequence similarity 24, member B3.3901642.4E-30
 PPP1R15ANM 014330.3Protein phosphatase 1, regulatory subunit 15A3.317365.28E-31
 EGR1NM 001964.2Early growth response 13.2172775.29E-40
 PHLDA1NM 007350.3Pleckstrin homology-like domain, family A, member 12.9922938.23E-34
 LIFNM 001257135.1Leukemia inhibitory factor2.9733420
 DUSP5NM 004419.3Dual specificity phosphatase 52.9544816.36E-16
 SESN2NM 031459.4Sestrin 22.9206113.56E-39
 BCL2A1NM 001114735.1BCL2-related protein A12.9111518.87039E-09
 HECW2NM 020760.1HECT, C2 and WW domain containing 2 E3 ubiquitin protein ligase2.903431.25E-37
 ALOX5APNM 001204406.1Arachidonate 5-lipoxygenase-activating protein2.8013880
 SAT1NM 002970.2Spermidine/spermine N1-acetyltransferase 12.7859467.82E-15
 FESNM 001143785.1Feline sarcoma oncogene2.755075.74E-23
O vs. OV downregulated
 NNMTNM 006169.2Nicotinamide N-methyltransferase−6.643856190
 MAGED4BNM 177535.2Melanoma antigen family D, 4B−5.4417194090
 CCL2NM 002982.3Chemokine (C-C motif) ligand 2−4.9868292250
 IGFBP7NM 001253835.1Insulin-like growth factor binding protein 7−4.1300758663.86E-42
 HSPA1ANM 005345.5Heat shock 70 kDa protein 1A−4.109383410
 EMR1NM 001256255.1EGF-like module containing, mucin-like, hormone receptor-like 1−4.0469576370
 SKP2NM 001243120.1S-phase kinase-associated protein 2, E3 ubiquitin protein ligase−3.8989116930
 PDE5ANM 001083.3Phosphodiesterase 5A, cGMP-specific−3.8554905710
 CD70NM 001252.3CD70 molecule−3.8198338920
 CFINM 000204.3Complement factor I−3.7664592115.14012E-09
 LIMS3NM 033514.4LIM and senescent cell antigen-like domains 3−3.6510452310
 SSX3NM 021014.2Synovial sarcoma, X breakpoint 3−3.6187557910
 IFITM1NM 003641.3Interferon-induced transmembrane protein 1−3.5256792523.07E-22
 KIF20ANM 005733.2Kinesin family member 20A−3.4197990730
 SSX1NM 005635.2Synovial sarcoma, X breakpoint 1−3.4197297772.21E-35
 HLA-DRB1NM 002124.3Major histocompatibility complex, class II, DR β 1−3.4052158088.44E-18
 BTN3A2NM 001197247.1Butyrophilin, subfamily 3, member A2−3.3974699362.41E-38
 SERPINA5NM 000624.5Serpin peptidase inhibitor, clade A, member 5−3.391633647.73E-36
 CA12NM 206925.1Carbonic anhydrase XII−3.3891823563.73E-24
 TCAM1PNR 002947.2Testicular cell adhesion molecule 1, pseudogene−3.3733897111.8E-36
S vs. SV upregulated
 NOP56NR 027700.2NOP56 ribonucleoprotein3.8263580
 BST1NM 004334.2Bone marrow stromal cell antigen 13.2845460
 MMP3NM 002422.3Matrix metallopeptidase 32.4361510
 PTPRBNM 001206972.1Protein tyrosine phosphatase, receptor type, B2.4256144.99E-35
 PHYHD1NM 001100877.1Phytanoyl-CoA dioxygenase domain containing 12.2014667.67E-31
 HBDNM 000519.3Hemoglobin, δ2.166155.78E-35
 PTPRRNM 001207016.1Protein tyrosine phosphatase, receptor type, R1.8959082.74E-32
 MAVSNM 001206491.1Mitochondrial antiviral signaling protein1.8649018.41E-45
 NGFNM 002506.2Nerve growth factor (β polypeptide)1.8646249.83E-28
 CDH13NM 001220490.1Cadherin 13, H-cadherin (heart)1.8477372.15E-30
 HSPA12ANM 025015.2Heat shock 70 kDa protein 12A1.7904511.94E-23
 CTAG1BNM 001327.2Cancer/testis antigen 1B1.7395195.56E-18
 AK5NM 174858.2Adenylate kinase 51.6917674.62E-28
 OR6F1NM 001005286.1Olfactory receptor, family 6, subfamily F, member 11.684942.46E-15
 CDH13NM 001220490.1Cadherin 13, H-cadherin (heart)1.6570279.68E-28
 PHYHD1NM 001100877.1Phytanoyl-CoA dioxygenase domain containing 11.6502185.27E-23
 KRT15NM 002275.3Keratin 151.6459171.26E-24
 RFX8NM 001145664.1RFX family member 8, lacking RFX DNA binding domain1.6331697.81E-13
 SIGLEC15NM 213602.2Sialic acid binding Ig-like lectin 151.6200681.14E-23
 QKINM 206855.2QKI, KH domain containing, RNA binding1.6132195436.66E-16
S vs. SV Downregulated
 NNMTNM 006169.2Nicotinamide N-methyltransferase−6.3703828780
 HLA-DRB1NM 002124.3Major histocompatibility complex, class II, DR β 1−4.6213763080
 MAGEC2NM 016249.3Melanoma antigen family C, 2−4.3253425860
 CSAG1NM 001102576.1 Chondrosarcoma-associated gene 1−3.5839553057.57E-44
 SERPINA5NM 000624.5Serpin peptidase inhibitor, clade A, member 5−3.570769510
 EMR1NM 001256255.1EGF-like module containing, mucin-like, hormone receptor-like 1−3.4566730390
 PSMB9NM 002800.4Proteasome (prosome, macropain) subunit, β type, 9−3.1659761010
 CFINM 000204.3Complement factor I−3.1348113260
 PAGE2NM 207339.2P-antigen family, member 2 (prostate-associated)−3.0722861450
 CSAG1NM 001102576.1 Chondrosarcoma-associated gene 1−2.8489476215.84154E-07
 SSX3NM 021014.2Synovial sarcoma, X breakpoint 3−2.7530832011.24441E-09
 HLA-FNM 001098478.1Major histocompatibility complex, class I, F−2.7152182230
 BEX1NM 018476.3Brain expressed, X-linked 1−2.7075276140
 IL1ANM 000575.3Interleukin 1, α−2.6916343610
 PDGFRBNM 002609.3Platelet-derived growth factor receptor, β polypeptide−2.6736870336.05E-19
 NUPR1NM 001042483.1Nuclear protein, transcriptional regulator, 1−2.6512040643.01E-41
 QPRTNM 014298.3Quinolinate phosphoribosyltransferase−2.599899280
 PDE5ANM 001083.3Phosphodiesterase 5A, cGMP-specific−2.5680048672.56E-33
 NDRG2NM 201541.1NDRG family member 2−2.5257927452.63E-28
 CD70NM 001252.3CD70 molecule−2.4897396860
Validation of gene expression results by RT-PCR

To validate the gene expression profile results, 4 DEGs (FOXA2, SERPIND1, BDKRD1 and IL1A) were selected for RT-PCR analysis verification (Fig. 4A). Generally, the trends for upregulation or downregulation of the DEGs by real-time PCR analysis were consistent with those of the DEG expression profiling analysis, confirming the reliability of the microarray results.

Validation of protein expression by IHC

Similar to HE4, the expression of SERPIND1 was mainly located on the membrane and in the cytoplasm (Fig. 4B). The positive expression rates of SERPIND1 in malignant, borderline, benign and normal ovarian tissues were 88, 62.96, 20 and 13.33%, respectively (Table III). Malignant groups displayed the highest positive expression and was significantly higher than the rate of the borderline (P=0.010), whereas the positive expression rate in the borderline groups was markedly higher than that in the benign groups (P=0.008). In general, the expression pattern of SERPIND1 was similar to that of HE4 (up to 74.8%). Among the 50 cases of ovarian cancer samples, a total of 37 cases simultaneously showed positive expression of both HE4 and SERPIND1 and 4 cases showed both negative expression (Table IV). Spearman correlation analysis revealed that the expression of SERPIND1 and HE4 was positively correlated (R=0.402, P=0.003). Survival analysis revealed that high expression levels of both HE4 and SERPIND1 were significantly correlated with poor prognosis (Kaplan-Meier analysis, log-rank, P=0.0145 and P=0.0009 as to OS; Fig. 4C, respectively).

Table III

Expression of HE4 and HCII in different ovarian tissues.

Table III

Expression of HE4 and HCII in different ovarian tissues.

GroupsCasesHE4
HCII
++++++Positive casesPositive rate (%)++++++Positive casesPositive rate (%)
Malignant5011616173978a61312194488c
Borderline271256415 55.56b1066517 62.96c
Benign151230032012111320
Normal15150000013200213.33

a Compared with the borderline group, P=0.040;

b compared with the benign group, P=0.026;

c compared with the borderline group, P=0.010; dcompared with the benign group, P=0.008. HE4, human epididymis protein 4; HCII, heparin co-factor II (SERPIND1).

Table IV

Relevance of HE4 and HCII expression in ovarian cancer samples.

Table IV

Relevance of HE4 and HCII expression in ovarian cancer samples.

HE4HCII
Total
NegativePositive
Negative4711
Positive23739
Total64450

[i] HE4, human epididymis protein 4; HCII, heparin co-factor II (SERPIND1).

GO function analysis and canonical pathway result of DEGs

GO analysis showed that all the DEGs in the different groups were predominantly involved in molecular function (MF), biological process (BP) and cellular component (CC), as shown in Table V. Canonical pathway analysis demonstrated that a total of 39 pathways were enriched in O vs. OV, and 50 pathways in S vs. SV. The top 20 pathways are shown in Table VI, such as MAPK signaling pathway, pathway in cancer and P53 signaling pathway in O vs. OV and focal adhesion, pathway in cancer and cell adhesion molecules (CAMs) in S vs. SV.

Table V

GO enrichment analysis for differentially expressed genes (DEGs).

Table V

GO enrichment analysis for differentially expressed genes (DEGs).

GO termsGenes in gene setGenes in overlapP-valueFDR q-value
Molecular function
(Top 10 in O vs. OV)
 Enzyme binding178151.12E-092.62E-07
 Transcription factor binding307191.32E-092.62E-07
 DNA binding602262.92E-093.86E-07
 Transcription repressor activity152121.07E-071.06E-05
 Transcription cofactor activity228142.16E-071.71E-05
 Receptor binding377178.97E-075.92E-05
 Receptor activity583206.62E-063.62E-04
 Transcription corepressor activity9488.23E-063.62E-04
 Transcription factor activity354158.24E-063.62E-04
 Transmembrane receptor activity418161.47E-055.81E-04
(Top 10 in S vs. SV)
 Receptor activity583206.29E-082.49E-05
 Phosphoric ester hydrolase activity153104.64E-079.18E-05
 Transmembrane receptor activity418151.63E-061.57E-04
 Peptidase activity176101.67E-061.57E-04
 Hydrolase activity acting on ester bonds269121.98E-061.57E-04
 Endopeptidase activity11784.71E-063.02E-04
 Enzyme inhibitor activity11985.35E-063.02E-04
 Protein kinase binding6261.02E-055.04E-04
 Protein kinase regulator activity3951.42E-056.24E-04
 Kinase binding7062.06E-058.15E-04
 Cell cycle GO 0007049315226.29E-126.49E-10
 Cell proliferation GO 0008283513271.85E-111.53E-09
 Protein metabolic process1,231431.85E-111.53E-09
Cellular component
(Top 10 in O vs. OV)
 Nucleus1,430546.66E-151.55E-12
 Cytoplasm2,131624.27E-124.97E-10
 Extracellular region447239.06E-107.04E-08
 Membrane1,994531.65E-099.64E-08
 Extracellular region part338196.40E-092.84E-07
 Membrane part1,670467.31E-092.84E-07
 Extracellular space245161.25E-084.15E-07
 Intracellular non membrane bound organelle631253.19E-088.25E-07
 Non membrane bound organelle631253.19E-088.25E-07
 Nuclear part579224.33E-079.83E-06
(Top 10 in S vs. SV)
 Membrane1,994531.55E-143.62E-12
 Membrane part1,670431.65E-111.30E-09
 Extracellular region447221.67E-111.30E-09
 Cytoplasm2,131493.02E-111.76E-09
 Intrinsic to membrane1,348363.14E-101.46E-08
 Plasma membrane1,426373.87E-101.50E-08
 Integral to membrane1,330358.24E-102.74E-08
 Extracellular region part338151.12E-073.27E-06
 Plasma membrane part1,158282.44E-076.32E-06
 Nucleus1,430294.89E-061.05E-04
Biological process
(Top 10 in O vs. OV)
 Biopolymer metabolic process1,684630.00E+000.00E+00
 Programmed cell death432311.11E-163.05E-14
 Apoptosis GO431311.11E-163.05E-14
 Cell development577341.67E-153.43E-13
 Nucleobase nucleoside nucleotide and nucleic acid metabolic process1,244497.66E-151.26E-12
 Multicellular organismal development1,049411.74E-122.39E-10
 Response to stress508282.62E-123.08E-10
 Cell cycle GO 0007049315226.29E-126.49E-10
 Cell proliferation GO 0008283513271.85E-111.53E-09
 Protein metabolic process1,231431.85E-111.53E-09
(Top 10 in S vs. SV)
 Signal transduction1,634455.53E-134.57E-10
 Multicellular organismal development1,049346.25E-122.58E-09
 Response to chemical stimulus314178.24E-101.99E-07
 Anatomical structure development1,013309.64E-101.99E-07
 System development861272.05E-092.91E-07
 Nucleobase nucleoside nucleotide and nucleic acid metabolic process1,244332.12E-092.91E-07
 Biopolymer metabolic process1,684393.07E-093.62E-07
 Negative regulation of biological process677237.63E-097.87E-07
 Negative regulation of cellular process646221.56E-081.43E-06
 Immune response235136.34E-085.23E-06

[i] GO, Gene Ontology; DEGs, differentially expressed genes; FDR, false discovery rate; OV, HE4-H-vector; O, HE4-H; SV, HE4-L-vector; S, HE4-L cells.

Table VI

Canonical pathway analysis in the differentially expressed genes (DEGs).

Table VI

Canonical pathway analysis in the differentially expressed genes (DEGs).

PathwaysGenes in gene setGenes in overlapP-valueFDR q-valueGene symbols
O vs. OV overlap pathways
 KEGG MAPK signaling pathway267211.99E-127.46E-10TP53, FGF2, FGF1, FGF5, NFKB2, GADD45A, HSPA1A, RPS6KA1, IL1A, IL1R1, HSPA1B, HSPA2, HSPA8, DUSP1, MAP3K6, DDIT3, JUND, DUSP4, DUSP5, RELB, STMN1
 KEGG steroid biosynthesis1783.70E-127.46E-10CYP27B1, DHCR24, DHCR7, EBP, FDFT1, LSS, SQLE, TM7SF2
 KEGG pathways in cancer328219.60E-111.29E-08TP53, FGF2, FGF1, FGF5, NFKB2, CDKN1A, SKP2, CASP9, HIF1A, BMP2, BMP4, SMO, FZD4, FZD7, RAD51, PTGS2, LAMC2, RARB, DAPK2, MSH6, BCR
 KEGG cell cycle128121.57E-081.58E-06TP53, GADD45A, CDKN1A, SKP2, PCNA, CDC25C, MCM2, MCM3, MCM5, CDKN2C, SMC1A, SMC3
 Biocarta caspase pathway2361.28E-078.59E-06CASP9, DFFB, LMNA, LMNB1, CASP2, CASP6
 Biocarta p53 hypoxia pathway2361.28E-078.59E-06TP53, GADD45A, HSPA1A, CDKN1A, HIF1A, NFKBIB
 Biocarta p53 pathway1655.52E-073.18E-05TP53, GADD45A, CDKN1A, PCNA, TIMP3
 KEGG p53 signaling pathway6987.81E-073.94E-05TP53, GADD45A, CDKN1A, CASP9, RRM2, GTSE1, PMAIP1, SESN2
 Biocarta G2 pathway2455.00E-062.24E-04TP53, GADD45A, RPS6KA1, CDKN1A, CDC25C
 KEGG basal cell carcinoma5562.75E-051.11E-03TP53, BMP2, BMP4, SMO, FZD4, FZD7
 Biocarta MCM pathway1843.55E-051.15E-03MCM2, MCM3, MCM5, CDT1
 KEGG cytokine cytokine receptor interaction267123.70E-051.15E-03IL1A, IL1R1, BMP2, IL12A, INHBA, IL11, CCL2, CXCL2, CXCL3, CXCL5, LIF, IL24
 Biocarta HIVNEF pathway5863.73E-051.15E-03CASP9, DFFB, LMNA, LMNB1, CASP2, CASP6
 KEGG DNA replication3653.99E-051.15E-03PCNA, MCM2, MCM3, MCM5, POLE2
 Biocarta ATM pathway2045.53E-051.49E-03TP53, GADD45A, CDKN1A, RAD51
 KEGG complement andcoagulation cascades6961.00E-042.52E-03PLAU, BDKRB1, CFI, PLAUR, SERPINA1, SERPINA5
 Biocarta TNFR1 pathway2942.51E-045.95E-03DFFB, LMNA, LMNB1, CASP2
 Biocarta FAS pathway3042.87E-046.29E-03DFFB, LMNA, LMNB1, CASP6
 KEGG small cell lung cancer8462.96E-046.29E-03TP53, SKP2, CASP9, PTGS2, LAMC2, RARB
 KEGG apoptosis8863.81E-047.66E-03TP53, IL1A, IL1R1, CASP9, DFFB, CASP6
S vs. SV overlap pathways
 KEGG focal adhesion201159.86E-113.97E-08PDGFRB, PIK3CD, PIK3R3, AKT3, LAMA3, LAMA1, PDGFD, ITGB5, THBS1, COL1A1, COL5A1, COL6A1, COL6A3, VASP, FYN
 KEGG pathways in cancer328157.61E-081.02E-05PDGFRB, PIK3CD, PIK3R3, AKT3, LAMA3, LAMA1, E2F2, FGF2, FGF13, KIT, KITLG, MMP2, DAPK2, ARNT2, FZD7
 KEGG complement and coagulation cascades6988.04E-081.02E-05BDKRB1, PLAU, F3, CD46, CFH, CFI, SERPINA5, SERPIND1
 KEGG melanoma7181.01E-071.02E-05PDGFRB, PIK3CD, PIK3R3, AKT3, PDGFD, E2F2, FGF2, FGF13
 KEGG cytokine cytokine receptor interaction267132.77E-072.23E-05PDGFRB, KIT, KITLG, IL1A, IL7R, IL4R, IL12A, IFNAR1, CCL5, CCL20, CXCL5, TNFSF12, TNFSF4
 KEGG ECM receptor interaction8483.78E-072.54E-05LAMA3, LAMA1, ITGB5, THBS1, COL1A1, COL5A1, COL6A1, COL6A3
 KEGG hematopoietic cell lineage8876.88E-063.74E-04KIT, KITLG, IL1A, IL7R, IL4R, MME, CD9
 KEGG prostate cancer8977.42E-063.74E-04PDGFRB, PIK3CD, PIK3R3, AKT3, PDGFD, E2F2, CREB3L1
 KEGG regulation of actin cytoskeleton216101.03E-054.59E-04PDGFRB, PIK3CD, PIK3R3, PDGFD, ITGB5, FGF2, FGF13, BDKRB1, ITGB2, GSN
 KEGG cell adhesion CAMs molecules13481.28E-055.16E-04ITGB2, HLA-C, HLA-DPA1, HLA-F, JAM2, NCAM1, ALCAM, PVR
 KEGG type I diabetes mellitus4452.59E-059.49E-04IL1A, IL12A, HLA-C, HLA-DPA1, HLA-F
 KEGG leukocytetransendothelial migration11874.66E-051.57E-03PIK3CD, PIK3R3, VASP, MMP2, ITGB2, JAM2, NCF2
 KEGG small cell lung cancer8465.81E-051.80E-03PIK3CD, PIK3R3, AKT3, LAMA3, LAMA1, E2F2
 KEGG natural killer cell-mediated cytotoxicity13771.20E-043.44E-03PIK3CD, PIK3R3, FYN, IFNAR1, ITGB2, HLA-C, SH2D1B
 KEGG prion diseases3541.69E-044.07E-03FYN, IL1A, CCL5, NCAM1
 KEGG TOLL-like receptor signaling pathway10261.71E-044.07E-03PIK3CD, PIK3R3, AKT3, IL12A, IFNAR1, CCL5
 KEGG glioma6551.72E-044.07E-03PDGFRB, PIK3CD, PIK3R3, AKT3, E2F2
 KEGG allograft rejection3842.33E-045.23E-03IL12A, HLA-C, HLA-DPA1, HLA-F
 KEGG JAK-STAT signaling pathway15572.56E-045.43E-03PIK3CD, PIK3R3, AKT3, IL7R, IL4R, IL12A, IFNAR1
 KEGG leishmania infection7252.78E-045.60E-03IL1A, IL12A, ITGB2, HLA-DPA1, NCF2

[i] DEGs, differentially expressed genes; FDR, false discovery rate; OV, HE4-H-vector; O, HE4-H; SV, HE4-L-vector; S, HE4-L cells.

Interaction network for the DEGs

Three aforementioned pathways for the DEGs were selected to conduct interaction network analysis in 2 pairs, respectively (Fig. 5). Among the interaction networks, various interaction genes were predicted, such as MAPK3 and MAPK8 in MAPK signaling pathway, UBB and EP300 in pathways in cancer, SRC in cell adhesion molecules (CAMs) pathway, which appeared to be the connected predicted hub genes.

Discussion

The mortality rate of ovarian cancer patients ranks first among all gynecological malignant tumors, and up to 75% of diagnosed patients are already at an advanced stage. The early detection of ovarian cancer is difficult due to its indefinite symptoms in the early stage and lack of a specific marker; meanwhile, a poor understanding of the mechanisms of oncogenesis also impedes the development of new treatment modalities. In recent years, HE4 has drawn extensive attention in the study of ovarian cancer, due to its potential clinical application benefits for early detection (5), discrimination (6), better tumor cytoreductive surgery (8), chemoresistance and prognosis (9,11). Moreover, it was also reported to be a reliable marker for early diagnosis of endometrial (12), lung (14) and pancreatic cancer (15). Thus, increasing research on the clinical application of HE4 stimulated elucidation of the basic mechanisms of its function. Unfortunately, relevant research is far from enough. In the present study, by the use of human whole genome microarray detection, we investigated the gene expression profile in response to HE4 in epithelial ovarian cancer (EOC) cells. We identify a total of 717 genes that were upregulated and 898 genes that were downregulated in HE4-overexpressing cells vs. HE4-Mock and 166 genes were upregulated and 285 were downregulated in the HE4-silenced cells vs. HE4-Mock cells. Furthermore, an overlap of 16 genes was consistently upregulated and 8 genes downregulated in response to HE4. The result was validated at the mRNA and protein levels. GO and pathway enrichment analysis were applied. Multiple pathways are involved, including KEGG MAPK signaling pathway, KEGG steroid biosynthesis, and KEGG pathways in cancer. Finally, interaction network plots were constructed and interactive genes were predicted. The known functions of these genes can provide novel ideas and breakthrough points for further research.

SERPIND1, also known as heparin co-factor II (HCII or HC2), attracted our attention. HCII belongs to the serpin superfamily and it is a serum glycoprotein that acts as a thrombin inhibitor through interactions with heparin and other endogenous glycosaminoglycans (26). Over the past 3 decades, HCII has been intermittently studied as a protease inhibitor. It can inhibit thrombin in atherosclerotic lesions where thrombin can exert a proatherogenic inflammatory response (27). It has also been proposed to promote angiogenesis in response to ischaemia (28). However, to date, the biological effect of HCII on cancer occurrence and development is still largely unknown, particularly in ovarian cancer. One group reported that high HCII expression in tumor tissues was associated with increased cancer recurrence and shorter OS in non-small cell lung cancer (NSCLC) patients. HCII promoted cell motility, invasion ability and filopodium dynamics in NSCLC cells partly through the PI3K pathway (29). Recently, HCII was found to be upregulated in the serum samples of B-cell acute lymphoblastic leukemia (B-ALL) patients and it was identified as a candidate biomarker for early diagnosis of B-ALL (30). In the present study, we found that the positive expression rates of HCII in malignant, borderline, benign and normal ovarian tissues were 88, 62.96, 20 and 13.33%, respectively. The protein expression pattern of HCII was positively related to that of HE4 (Spearman coefficient ratio, R=0.402, P=0.003) and high expression of HCII was correlated with poor OS of ovarian cancer patients. HCII may act as a potential oncogene in the development of ovarian cancer. It seems to be the first study on its potential function in the tumorigenesis of ovarian cancer and sheds light on the possible application as a tumor biomarker or therapeutic target along with HE4. However, further investigations are needed to elucidate the underlying mechanism.

In our previous studies, we demonstrated that HE4 enhanced the proliferation, invasion and metastasis of ovarian cancer (20) partially via the interaction of Annexin A2 (23), and fucoglycosylation may increase this effect (24,31). A similar phenomenon was also noted in others studies (18,19,21), even in endometrial (32,33) and pancreatic cancer (33). Nevertheless, the underlying mechanisms of these phenomena warrant more discussion. One group reported that the expression of HE4 is associated with cell adhesion, migration and tumor growth via the activation of the EGFR-MAPK signaling pathway (18) in ovarian cancer cells. However, another group reported a controversial finding that HE4 may play a protective role in the progression of ovarian cancer by inhibiting cell proliferation, whereas they also speculated that this effect may be regulated by MAPK and PI3K/Akt signaling pathway. As to the influence of HE4 on the chemotherapeutic resistance in ovarian cancer, our previous study noted that the recombinant HE4 protein could repress carboplatin-induced apoptosis in ovarian cancer cells (22). Another group reported that HE4 overexpression promoted chemoresistance against cisplatin in an animal model leading to reduced survival rates (19). They further demonstrated that tumor microenvironment constituents participated in the modulation of HE4, in which HE4 could interact with EGFR, IGF1R and transcription factor HIF1α, inducing the nuclear translocation of HE4 to promote aggressive and chemoresistant disease and denote poor prognosis for ovarian cancer patients (19). Recently, a group reported that recombinant HE4 protein increased the mRNA and protein levels of cell cycle marker PCNA and cell cycle inhibitor p21 in endometrial and pancreatic cancer cell lines, indicating that HE4 function may be mediated by the p21-CDK-Rb pathway (33). In the present study, we presented the results for the possible pathways HE4 may enrich, including MAPK, steroid biosynthesis, cell cycle, p53 hypoxia pathway, focal adhesion, ECM receptor interaction, and cell adhesion molecules (CAMs). To the best of our knowledge, this is the first study to provide the most detailed, full-scale and fundamental data for the DEGs in response to HE4, thus laying the basis for further investigation on the underlying mechanisms of HE4 in ovarian cancer. More comprehensive and in-depth studies are needed, to provide more evidence for the development of novel drugs and therapeutic strategies selectively targeting HE4 in ovarian cancer.

Collectively, the present study analyzed the gene expression profile in response to HE4 in EOC cells. The identified DEGs are valuable to determine the underlying mechanism of HE4 in cancer, providing new views for the comprehensive study of ovarian cancer treatment. Prospective investigations using the identified DEGs are required to further elucidate the mechanisms of the tumorgenesis and development of ovarian cancer.

Acknowledgments

The present study was supported by the National Natural Science Foundation of China (grant nos. 81172491, 81101527, 81472437 and 81402129), the Education Department Doctor Project Fund (grant nos. 20112104110016 and 20112104120019), and the Outstanding Scientific Fund of Shengjing Hospital (grant no. 201303).

References

1 

Siegel RL, Miller KD and Jemal A: Cancer statistics, 2016. CA Cancer J Clin. 66:7–30. 2016. View Article : Google Scholar : PubMed/NCBI

2 

Pliarchopoulou K and Pectasides D: Epithelial ovarian cancer: Focus on targeted therapy. Crit Rev Oncol Hematol. 79:17–23. 2011. View Article : Google Scholar

3 

Schummer M, Ng WV, Bumgarner RE, Nelson PS, Schummer B, Bednarski DW, Hassell L, Baldwin RL, Karlan BY and Hood L: Comparative hybridization of an array of 21,500 ovarian cDNAs for the discovery of genes overexpressed in ovarian carcinomas. Gene. 238:375–385. 1999. View Article : Google Scholar : PubMed/NCBI

4 

Cornwall GA, von Horsten HH, Swartz D, Johnson S, Chau K and Whelly S: Extracellular quality control in the epididymis. Asian J Androl. 9:500–507. 2007. View Article : Google Scholar : PubMed/NCBI

5 

Karlsen MA, Høgdall EV, Christensen IJ, Borgfeldt C, Kalapotharakos G, Zdrazilova-Dubska L, Chovanec J, Lok CA, Stiekema A, Mutz-Dehbalaie I, et al: A novel diagnostic index combining HE4, CA125 and age may improve triage of women with suspected ovarian cancer - An international multicenter study in women with an ovarian mass. Gynecol Oncol. 138:640–646. 2015. View Article : Google Scholar : PubMed/NCBI

6 

Romagnolo C, Leon AE, Fabricio AS, Taborelli M, Polesel J, Del Pup L, Steffan A, Cervo S, Ravaggi A, Zanotti L, et al: HE4, CA125 and risk of ovarian malignancy algorithm (ROMA) as diagnostic tools for ovarian cancer in patients with a pelvic mass: An Italian multicenter study. Gynecol Oncol. 141:303–311. 2016. View Article : Google Scholar : PubMed/NCBI

7 

Wilailak S, Chan KK, Chen CA, Nam JH, Ochiai K, Aw TC, Sabaratnam S, Hebbar S, Sickan J, Schodin BA, et al: Distinguishing benign from malignant pelvic mass utilizing an algorithm with HE4, menopausal status, and ultrasound findings. J Gynecol Oncol. 26:46–53. 2015. View Article : Google Scholar :

8 

Chen WT, Gao X, Han XD, Zheng H, Guo L and Lu RQ: HE4 as a serum biomarker for ROMA prediction and prognosis of epithelial ovarian cancer. Asian Pac J Cancer Prev. 15:101–105. 2014. View Article : Google Scholar : PubMed/NCBI

9 

Angioli R, Capriglione S, Aloisi A, Guzzo F, Luvero D, Miranda A, Damiani P, Montera R, Terranova C and Plotti F: Can HE4 predict platinum response during first-line chemotherapy in ovarian cancer? Tumour Biol. 35:7009–7015. 2014. View Article : Google Scholar : PubMed/NCBI

10 

Vallius T, Hynninen J, Auranen A, Carpén O, Matomäki J, Oksa S, Virtanen J and Grénman S: Serum HE4 and CA125 as predictors of response and outcome during neoadjuvant chemotherapy of advanced high-grade serous ovarian cancer. Tumour Biol. 35:12389–12395. 2014. View Article : Google Scholar : PubMed/NCBI

11 

Zhu LC, Gao J, Hu ZH, Schwab CL, Zhuang HY, Tan MZ, Yan LM, Liu JJ, Zhang DY and Lin B: Membranous expressions of Lewis y and CAM-DR-related markers are independent factors of chemotherapy resistance and poor prognosis in epithelial ovarian cancer. Am J Cancer Res. 5:830–843. 2015.PubMed/NCBI

12 

Kemik P, Saatli B, Yıldırım N, Kemik VD, Deveci B, Terek MC, Koçtürk S, Koyuncuoğlu M and Saygılı U: Diagnostic and prognostic values of preoperative serum levels of YKL-40, HE-4 and DKK-3 in endometrial cancer. Gynecol Oncol. 140:64–69. 2016. View Article : Google Scholar

13 

Li X, Gao Y, Tan M, Zhuang H, Gao J, Hu Z, Wang H, Zhu L, Liu J and Lin B: Expression of HE4 in endometrial cancer and its clinical significance. Biomed Res Int. 2015:4374682015.PubMed/NCBI

14 

Jiang Y, Wang C, Lv B, Ma G and Wang L: Expression level of serum human epididymis 4 and its prognostic significance in human non-small cell lung cancer. Int J Clin Exp Med. 7:5568–5572. 2014.

15 

Huang T, Jiang SW, Qin L, Senkowski C, Lyle C, Terry K, Brower S, Chen H, Glasgow W, Wei Y, et al: Expression and diagnostic value of HE4 in pancreatic adenocarcinoma. Int J Mol Sci. 16:2956–2970. 2015. View Article : Google Scholar : PubMed/NCBI

16 

Lan WG, Hao YZ, Xu DH, Wang P, Zhou YL and Ma LB: Serum human epididymis protein 4 is associated with the treatment response of concurrent chemoradiotherapy and prognosis in patients with locally advanced non-small cell lung cancer. Clin Transl Oncol. 18:375–380. 2016. View Article : Google Scholar

17 

Lamy PJ, Plassot C and Pujol JL: Serum HE4: An independent prognostic factor in non-small cell lung cancer. PLoS One. 10:e01288362015. View Article : Google Scholar : PubMed/NCBI

18 

Lu R, Sun X, Xiao R, Zhou L, Gao X and Guo L: Human epididymis protein 4 (HE4) plays a key role in ovarian cancer cell adhesion and motility. Biochem Biophys Res Commun. 419:274–280. 2012. View Article : Google Scholar : PubMed/NCBI

19 

Moore RG, Hill EK, Horan T, Yano N, Kim K, MacLaughlan S, Lambert-Messerlian G, Tseng YD, Padbury JF, Miller MC, et al: HE4 (WFDC2) gene overexpression promotes ovarian tumor growth. Sci Rep. 4:35742014. View Article : Google Scholar : PubMed/NCBI

20 

Zhu L, Zhuang H, Wang H, Tan M, Schwab CL, Deng L, Gao J, Hao Y, Li X, Gao S, et al: Overexpression of HE4 (human epididymis protein 4) enhances proliferation, invasion and metastasis of ovarian cancer. Oncotarget. 7:729–744. 2016.

21 

Chen Y, Mu X, Wang S, Zhao L, Wu Y, Li J and Li M: WAP four-disulfide core domain protein 2 mediates the proliferation of human ovarian cancer cells through the regulation of growth-and apoptosis-associated genes. Oncol Rep. 29:288–296. 2013.

22 

Wang H, Zhu L, Gao J, Hu Z and Lin B: Promotive role of recombinant HE4 protein in proliferation and carboplatin resistance in ovarian cancer cells. Oncol Rep. 33:403–412. 2015.

23 

Zhuang H, Tan M, Liu J, Hu Z, Liu D, Gao J, Zhu L and Lin B: Human epididymis protein 4 in association with Annexin II promotes invasion and metastasis of ovarian cancer cells. Mol Cancer. 13:2432014. View Article : Google Scholar : PubMed/NCBI

24 

Zhuang H, Hu Z, Tan M, Zhu L, Liu J, Liu D, Yan L and Lin B: Overexpression of Lewis y antigen promotes human epididymis protein 4-mediated invasion and metastasis of ovarian cancer cells. Biochimie. 105:91–98. 2014. View Article : Google Scholar : PubMed/NCBI

25 

Diaz-Montana JJ and Diaz-Diaz N: Development and use of the Cytoscape app GFD-Net for measuring semantic dissimilarity of gene networks. F1000Res. 3:1422014.PubMed/NCBI

26 

Tollefsen DM: Heparin cofactor II modulates the response to vascular injury. Arterioscler Thromb Vasc Biol. 27:454–460. 2007. View Article : Google Scholar

27 

Rau JC, Deans C, Hoffman MR, Thomas DB, Malcom GT, Zieske AW, Strong JP, Koch GG and Church FC: Heparin cofactor II in atherosclerotic lesions from the Pathobiological Determinants of Atherosclerosis in Youth (PDAY) study. Exp Mol Pathol. 87:178–183. 2009. View Article : Google Scholar : PubMed/NCBI

28 

Ikeda Y, Aihara K, Yoshida S, Iwase T, Tajima S, Izawa-Ishizawa Y, Kihira Y, Ishizawa K, Tomita S, Tsuchiya K, et al: Heparin cofactor II, a serine protease inhibitor, promotes angiogenesis via activation of the AMP-activated protein kinase-endothelial nitric-oxide synthase signaling pathway. J Biol Chem. 287:34256–34263. 2012. View Article : Google Scholar : PubMed/NCBI

29 

Liao WY, Ho CC, Hou HH, Hsu TH, Tsai MF, Chen KY, Chen HY, Lee YC, Yu CJ, Lee CH, et al: Heparin co-factor II enhances cell motility and promotes metastasis in non-small cell lung cancer. J Pathol. 235:50–64. 2015. View Article : Google Scholar

30 

Cavalcante MS, Torres-Romero JC, Lobo MD, Moreno FB, Bezerra LP, Lima DS, Matos JC, Moreira RA and Monteiro-Moreira AC: A panel of glycoproteins as candidate biomarkers for early diagnosis and treatment evaluation of B-cell acute lymphoblastic leukemia. Biomark Res. 4:12016. View Article : Google Scholar :

31 

Zhuang H, Gao J, Hu Z, Liu J, Liu D and Lin B: Co-expression of Lewis y antigen with human epididymis protein 4 in ovarian epithelial carcinoma. PLoS One. 8:e689942013. View Article : Google Scholar : PubMed/NCBI

32 

Li J, Chen H, Mariani A, Chen D, Klatt E, Podratz K, Drapkin R, Broaddus R, Dowdy S and Jiang SW: HE4 (WFDC2) promotes tumor growth in endometrial cancer cell lines. Int J Mol Sci. 14:6026–6043. 2013. View Article : Google Scholar : PubMed/NCBI

33 

Lu Q, Chen H, Senkowski C, Wang J, Wang X, Brower S, Glasgow W, Byck D, Jiang SW and Li J: Recombinant HE4 protein promotes proliferation of pancreatic and endometrial cancer cell lines. Oncol Rep. 35:163–170. 2016.

Related Articles

Journal Cover

September-2016
Volume 36 Issue 3

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
Zhu L, Guo Q, Jin S, Feng H, Zhuang H, Liu C, Tan M, Liu J, Li X, Lin B, Lin B, et al: Analysis of the gene expression profile in response to human epididymis protein 4 in epithelial ovarian cancer cells. Oncol Rep 36: 1592-1604, 2016
APA
Zhu, L., Guo, Q., Jin, S., Feng, H., Zhuang, H., Liu, C. ... Lin, B. (2016). Analysis of the gene expression profile in response to human epididymis protein 4 in epithelial ovarian cancer cells. Oncology Reports, 36, 1592-1604. https://doi.org/10.3892/or.2016.4926
MLA
Zhu, L., Guo, Q., Jin, S., Feng, H., Zhuang, H., Liu, C., Tan, M., Liu, J., Li, X., Lin, B."Analysis of the gene expression profile in response to human epididymis protein 4 in epithelial ovarian cancer cells". Oncology Reports 36.3 (2016): 1592-1604.
Chicago
Zhu, L., Guo, Q., Jin, S., Feng, H., Zhuang, H., Liu, C., Tan, M., Liu, J., Li, X., Lin, B."Analysis of the gene expression profile in response to human epididymis protein 4 in epithelial ovarian cancer cells". Oncology Reports 36, no. 3 (2016): 1592-1604. https://doi.org/10.3892/or.2016.4926