Open Access

Expression levels of EPHB4, EFNB2 and caspase‑8 are associated with clinicopathological features and progression of esophageal squamous cell cancer

  • Authors:
    • Qianzhi Ni
    • Pingping Chen
    • Bing Zhu
    • Jingjing Li
    • Dong Xie
    • Xingyuan Ma
  • View Affiliations

  • Published online on: November 28, 2019     https://doi.org/10.3892/ol.2019.11160
  • Pages: 917-929
  • Copyright: © Ni et al. This is an open access article distributed under the terms of Creative Commons Attribution License.

Metrics: Total Views: 0 (Spandidos Publications: | PMC Statistics: )
Total PDF Downloads: 0 (Spandidos Publications: | PMC Statistics: )


Abstract

The upregulation of EPH receptor B4 (EPHB4) results in a survival advantage for tumor cells via the inhibition of the casapse‑8‑mediated apoptotic pathway, which begins from the cell membrane. The present study investigated the expression patterns of EPHB4, ephrin B2 (EFNB2) and caspase‑8 in patients with esophageal squamous cell carcinoma (ESCC). The association between the expression patterns and certain clinicopathological characteristics of the patients was also determined. mRNA levels of EPHB4, EFNB2 and caspase‑8 in paired primary ESCC samples and adjacent esophageal tissues collected from 96 patients with ESCC were quantified using quantitative PCR. Upregulation of EPHB4 and EFNB2 mRNA expression, and downregulation of caspase‑8 mRNA were detected in ESCC samples compared with that in the adjacent esophageal tissues. The expression levels of EPHB4 and EFNB2 were positively correlated with each other, whereas the mRNA levels of both EPHB4 and EFNB2 exhibited a negative correlation with that of caspase‑8. The mRNA levels of both EPHB4 and EFNB2 demonstrated a significant positive association with certain clinicopathological features of patients with ESCC, including family history, tumor size, metastasis and stage. Conversely, a negative association was revealed between the expression level of caspase‑8 and clinicopathological features of patients with ESCC. Moreover, mRNA expression levels of EPHB4 and EFNB2 were negatively associated with survival times of patients with ESCC, whereas the level of caspase‑8 was positively associated with patient outcome. The results from the present study suggested that EPHB4, EFNB2 and caspase‑8 may be implicated in the tumorigenesis and progression of ESCC, and that consequently, they may serve as useful prognostic markers, as well as potential therapeutic targets.

Introduction

The Eph receptor family comprises the largest receptor tyrosine kinase superfamily, and contains 14 distinct members, with 9 molecules identified in its ligand ephrin family (1). According to their sequence homology and binding specificity, both Ephs and ephrins are classified as type A and B. Upon engagement of Eph by the cognate ephrin, the two molecules activate simultaneously and induce intracellular signal transduction, which initiates a number of biological processes, including axon guidance, neural crest cell migration, hindbrain segmentation, somite formation and vasculogenesis (2,3). Moreover, there is accumulating evidence that the Eph/ephrin system also serves a pivotal role in the development and progression of numerous cancer types (4,5).

EPH receptor A2 (EphA2) is the most well characterized of the Eph receptors, particularly when regarding its role in tumorigenesis; it has been revealed to be upregulated in a number of different types of tumor, including prostate, colon and lung cancer, as well as melanomas (4). Furthermore, overexpression of EphA2 is able to induce malignant transformation in mammary epithelial cells (4). The EphB/ephrinB system is also implicated in tumorigenesis (4). The expression level of EphB2 is reported to be upregulated in gastrointestinal, liver, ovarian, lung and renal cancers (4). Although the majority of studies suggest that Ephs and ephrins serve an oncogenic role, EphB2 was reported as a tumor suppressor in prostate and colorectal tumors (68). These findings reflect the complexity of the differential functions of the Eph/ephrin system, which is capable of exerting context-dependent agonistic or antagonistic effects. Caspase-8, a member of the cysteine-aspartic acid protease (caspase) family, is well characterized as an initiator of death receptor-mediated apoptosis, and has been implicated in other similar apoptotic responses (9). Caspase-8 promoter methylation results in the loss of gene expression, which is associated with tumor severity in a variety of different tumor types. The methylation-mediated silencing of key apoptosis-associated genes serves an important role in the pathogenesis and development of therapeutic resistance in human cancer cells (10).

Esophageal cancer represents the sixth most frequent cause of cancer-associated mortality worldwide (11). Esophageal squamous cell carcinoma (ESCC) is the most prevalent histological subtype of esophageal cancer and exhibits high mortality rates and a 5-year overall survival rate of ≤15% (12,13). The most common pathological subtypes of esophageal cancer are ESCC and esophageal adenocarcinoma. Despite the well-characterized pathological progression of ESCC, the underlying molecular mechanisms are predominantly yet to be elucidated. Several studies reported that the expression of EphA2 (and one of its receptors, ephrinA1) were upregulated in ESCC, and correlated with tumor progression and patient survival, revealing their predictive potential for the diagnosis and prognosis of patients with ESCC (14). Previous studies demonstrated that EPHB4 conferred a survival advantage on tumor cells by decreasing apoptosis, whereas knockdown of EPHB4 expression using siRNA induced apoptosis and decreased tumor cell viability via the activation of caspase-8. However, studies focusing on the influence that EphB/ephrin-B and caspase-8 exert on ESCC progression and genesis remain limited. Therefore, the present study investigated the expression levels of EPHB4, its cognate ligand ephrin B2 (EFNB2) (13) and caspase-8 in ESCC. In addition, the association between their relative expression levels and clinical parameters important in the diagnosis and prognosis of ESCC were also investigated in the present study. The results from the present study provide additional understanding, potentially facilitating the development of diagnostic and therapeutic strategies for the treatment of ESCC.

Materials and methods

Patients and samples

In the present study, 96 ESCC samples, and their paired paracancerous esophageal tissues, were obtained from patients with ESCC treated at the First Affiliated Hospital of Zhengzhou University (Henan, China), between July 2002 and August 2006, following the provision of written informed consent. The tumor stage was classified according to the 8th edition of the TNM classification (15). Cancerous tissues were surgically resected from patients who had not received any neo-adjuvant therapy, and the corresponding non-cancerous ‘normal’ tissues, located at least 3 cm away from the tumor site, were obtained in the same manner. Each specimen was divided into 2 pieces, one of which was fixed in 4% formalin at 4°C overnight, sectioned and examined using immunohistochemical (IHC) staining, and the other of which was stored at −80°C. The present study was approved by the Institutional Review Board of the Institute for Nutritional Sciences, Chinese Academy of Sciences.

Quantitative (q)PCR

RNA extraction, DNA template synthesis and amplification reactions were performed as previously described (16). The primers for the qPCR are listed as follows: EPHB4 forward, 5′-TCCTTCCTGCGGCTAAAC-3′ and reverse, 5′-CTTTGCAGACGAGGTTGCT-3′; EFNB2 forward, 5′-TCTTTGGAGGGCCTGGATAA-3′ and reverse, 5′-CGTCTGTGCTAGAACCTGGATT-3′; caspase-8 forward, 5′-CTGCAGAGGAACCTGGTACATCC-3′ and reverse, 5′-TCTTACTCCAAGGTGGCCATG-3′; and β-actin forward, 5′-GATCATTGCTCCTCCTGAGC-3′ and reverse, 5′-ACTCCTGCTTGCTGATCCAC-3′. All primers were designed using PRIMER5 software (version 5.00; Premier Biosoft International) and purchased from Shanghai Sangong Pharmaceutical Co., Ltd. Reactions were characterized at the point during cycling when amplification of the PCR product was first detected after a fixed number of cycles. Quantification was performed by measuring the quantitation cycle (Cq) value. The levels of target genes in each sample were normalized to the housekeeping gene β-actin via the following formula: Normalized level (NL)=level(target)/level (β-actin)=2Cq(target)/2Cq(β−actin)=2Cq(target)−Cq(β−actin)=2∆Cq. Furthermore, the relative levels (RL) of target genes in cancer tissues vs. corresponding normal samples were calculated according to the formula: RL=NL(cancer)/NL(normal)=2∆Cq(cancer)/2∆Cq(normal)=2[∆Cq(cancer)−∆Cq(normal)]=2∆∆Cq. As both NL and RL are represented as 2Cq, the present study used ∆Cq and ∆∆Cq to represent NL and RL, respectively, when performing statistical analysis.

IHC

The specimens were fixed in 4% formalin at 4°C overnight. The paraffin-embedded tissues were cut into 5-µm thick sections, deparaffinized, rehydrated in graded dimethylbenzene and ethanol solutions, and subjected to antigen retrieval. Subsequently, the sections were blocked using 5% normal goat serum (Invitrogen; Thermo Fisher Scientific, Inc.) at room temperature for 1 h. The tissue sections were then incubated with the following primary antibodies at 4°C overnight: Rabbit anti-human EPHB4 (1:500; cat. no. sc-365510), rabbit anti-human EFNB2 (1:500, cat. no. sc-398735) (both Santa Cruz Biotechnology, Inc.) and rabbit anti-human caspase-8 (1:100; cat. no. 552143; BD Pharmingen; BD Biosciences). Following primary incubation, the sections were incubated at 37°C for 1 h with horseradish peroxidase-conjugated goat anti-rabbit IgG secondary antibody (1:1,000; cat. no. A-10194; Chemicon International; Thermo Fisher Scientific Inc.). Finally, all sections were counterstained with hematoxylin at room temperature for 5–8 min.

Scoring of IHC staining was simultaneously performed by three independent pathologists. Tumor cells positive and negative for staining were counted separately under a light microscope (magnification, ×200). For each slide, 7–10 microscopic fields with ≥300 cells/microscopic field were randomly selected. The ratio of positive cells was calculated as the number of positively stained tumor cells divided by the total tumor cells, in each high-power field area. The level of protein expression was quantified by calculating the percentage ratio of positively stained cells in the esophageal cancer sample compared with that in the matched paracancerous esophageal tissues. Patients with high expression of EPHB4, EFNB2 and caspase-8 had protein levels of ≥1.89, ≥1.57 and ≥0.56, respectively; whereas patients with low expression had protein levels of <1.89, <1.57 and <0.56, respectively.

Statistical analysis

The χ2 test was used to determine the association between the expression levels of EPHB4, EFNB2 and caspase-8 in ESCC samples and the clinical characteristics, respectively. Pearson's correlation analysis was used to estimate the relative degree. Kaplan-Meier survival curves comparing patients with high and low expression at the mRNA and protein levels were plotted and univariate survival analysis was performed using log-rank test.

Multivariate analyses were performed to estimate the effects of certain clinicopathological characteristics, and the expression levels of the two genes, on survival. The data were analyzed using Student's t-test. P<0.05 were considered to indicate a statistically significant difference. All statistical analyses were performed using SPSS version 22 (IBM Corporation).

Results

Expression of EPHB4, EFNB2 and caspase-8 genes in ESCC and matched normal esophageal tissues

In order to investigate the expression pattern of EPHB4, EFNB2 and caspase-8 in ESCC, the mRNA levels of the three genes were quantified in 96 pairs of tumor samples and matched normal esophageal tissue samples using qPCR. Expression levels were presented as a ratio between EPHB4, EFNB2 or caspase-8 and the reference gene β-actin. Upregulation of EPHB4 and EFNB2 occurred in 63 out of 96 (66%) ESCC samples, and 53 out of 96 (55%) paired normal esophageal tissues, respectively. By contrast, downregulation of caspase-8 was observed in 55 out of 96 (57%) ESCC samples, when compared with that in normal tissues (Fig. 1). Univariate analysis revealed that the mRNA level of EPHB4 was significantly increased in tumor tissues, compared with paired normal tissues (P=0.001), while the expression level of caspase-8 was significantly lower in the ESCC samples (P=0.001). However, there was no significant difference in the mRNA level of EFNB2 between the ESCC and paired normal tissues (P=0.172) (Table I). Pearson's correlation analysis demonstrated that the mRNA expression of EPHB4 was positively correlated with that of EFNB2 (R2=0.620; P<0.001). Notably, EPHB4 (R2=−0.428; P=0.001) and EFNB2 (R2=−0.267, P=0.028) were both negatively correlated with caspase-8 (Table II).

Table I.

Expression of EPHB4, EFNB2 and caspase-8 genes in esophageal cancer and paired paracancerous esophageal tissues (n=96 pairs).

Table I.

Expression of EPHB4, EFNB2 and caspase-8 genes in esophageal cancer and paired paracancerous esophageal tissues (n=96 pairs).

mRNA/proteinnCancerous Matched-paracancerous(N-C)/(C/N)t-testP-value
EPHB4 mRNA9612.89±10.0815.45±10.262.56±3.926.411 <0.01a
EFNB2 mRNA968.05±5.888.86±5.690.81±5.771.3750.172
caspase-8 mRNA967.38±2.475.46±1.87−1.92±2.677.307 <0.001a
EPHB4 protein9621.35±8.2962.80±0.9478.74±5.65−21.603 <0.001a
EFNB2 protein9611.67±2.4781.71±0.5977.83±3.44−38.322 <0.001a
Caspase-8 protein962.51±2.3848.85±7.8790.28±0.15−22.761 <0.001a

a P<0.05. N-C, Cq value of normal esophageal minus that of cancerous esophageal tissues; C/N, percentage of positively stained cells in esophageal cancer tissues compared with the matched normal samples; EPHB4, EPH receptor B4; EFNB2, ephrin B2.

Table II.

Pearson's correlation analysis of mRNA and protein expression of EPHB4, EFNB2 and caspase-8 in esophageal cancer and matched normal esophageal tissues (n=96 pairs).

Table II.

Pearson's correlation analysis of mRNA and protein expression of EPHB4, EFNB2 and caspase-8 in esophageal cancer and matched normal esophageal tissues (n=96 pairs).

mRNAProtein


GenesR-valueP-valueR-valueP-value
EPHB4 and EFNB2   0.620 <0.001a0.2020.049a
EPHB4 and caspase-8−0.428 <0.001a−0.3400.001a
EFNB2 and caspase-8−0.2670.028a−0.1980.041a

a P<0.05. EPHB4, EPH receptor B4; EFNB2, ephrin B2.

Subsequently, IHC was performed to investigate the protein expression levels of EPHB4, EFNB2 and caspase-8 proteins in 96 pairs of esophageal tissues. As presented in Fig. 2A and C, EPHB4 and EFNB2 proteins were not apparent in the majority of normal esophageal epithelial cells, while they were highly expressed in the majority tumor cells in corresponding ESCC tissues (Fig. 2B and D), synonymous with previous reports (14,17). As presented in Fig. 2E, strong staining of caspase-8 was observed in the superficial layer of normal esophageal epithelia, but was almost undetectable in the ESCC samples (Fig. 2F), which is also consistent with other studies (18,19). The IHC scoring analysis revealed that the ratio of EPHB4/EFNB2-positive to -negative cells in ESCC tissues was significantly higher in comparison with that in the corresponding normal tissue, whereas the ratio of caspase-8-positive to -negative cells was lower in ESCC tissues compared with that in their normal counterparts (Table I).

Association between the expression of EPHB4, EFNB2 and caspase-8, and the clinicopathological features of patients with ESCC

The univariate analysis revealed a significant association between the expression of EPHB4 and family history, metastasis, and tumor size, position and stage. The expression level of EPHB4 was significantly higher in patients with a family history of cancer (P<0.001). A significant association also existed between increased levels of EPHB4 and metastasis (P=0.001), larger tumors (P=0.001), ESCC located in the lower segment of the esophagus (P=0.010) and a higher stage (P=0.043), indicating that the upregulation of EPHB4 expression was associated with ESCC progression. Sex and age were not significantly associated with the expression level of EPHB4 (Table III).

Table III.

Association between the levels of EPHB4/EFNB2 and caspase-8 mRNA expression and the clinical and pathological features of individuals with esophageal cancer (n=96 pairs).

Table III.

Association between the levels of EPHB4/EFNB2 and caspase-8 mRNA expression and the clinical and pathological features of individuals with esophageal cancer (n=96 pairs).

EPHB4 EFNB2 Caspase-8



FactorsHigh, nLow, nχ2P-valueHigh, nLow, nχ2P-valueHigh, nLow, nχ2P-value
Sex 0.2330.629 1.1490.284 0.5260.506
  Male4522 3631 2641
  Female1811 1910 1514
Age, years 2.4330.303 2.0010.368 1.7330.494
  ≤3520 20 11
  35–501210 1111 1210
  ≥504923 4230 2844
Metastasis 11.0390.001a 0.9700.325 41.552 <0.001a
  Yes4512 2019 948
  No1821 3522 327
Family history 53.940 <0.001a 15.668 <0.001a 7.3270.012a
  Yes532 4114 1738
  No1031 1427 2417
Tumor size, cm3 14.4150.001a 5.0600.080 32.714 <0.001a
  ≤1001420 1519 277
  100–2003210 2517 1329
  >200173 155 119
TNM stage 6.2940.043a 1.9420.379 16.816 <0.001a
  I1414 1315 208
  II2112 2013 1419
  III287 2213 728
Tumor position 9.1670.010a 6.0750.048a 7.8870.019a
  Upper1216 1117 1810
  Middle3713 3416 1832
  Lower144 108 513

a P<0.05. EPHB4, EPH receptor B4; EFNB2, ephrin B2.

Statistical analysis also demonstrated that the expression level of EFNB2 was significantly associated with several clinical features, including tumor position and family history. The mRNA level of EFNB2 was significantly higher in the patients with a family history of cancer (P<0.001). ESCCs located in the lower segment of the esophagus exhibited higher EFNB2 expression than those in the upper segment (P=0.048). However, no associations were observed between the expression level of EFNB2 and sex, age, metastasis or tumor size and stage (Table III).

The expression level of caspase-8 was significantly downregulated in patients with family history (P=0.012). Downregulated expression levels of caspase-8 were significantly associated with metastasis (P<0.000), increased tumor size (P<0.000), ESCC at the lower segment of the esophagus (P=0.019) and a higher stage (P<0.000), indicating that low caspase-8 expression is associated with the progression of ESCC (Table III). However, there was no significant association observed between caspase-8 expression and sex or age.

IHC scoring analysis revealed that the ratio of EPHB4-positive to -negative cells in tissue samples was higher in patients with a family history of cancer (P=0.002). There was also a significant association between a higher positive-staining ratio and metastasis (P=0.005), larger tumors (P<0.001) and higher tumor stages (P=0.004). The increased ratio of EFNB2-positive to -negative cells, as well as a decreased ratio of caspase-8 was identified in patients with metastasis or greater tumors, respectively (EFNB2, P=0.004 and P=0.018, respectively; and caspase-8, P=0.000 and P=0.000, respectively) (Table IV). Taken together, the associations between protein levels of EPHB4, EFNB2 or caspase-8 and certain clinicopathological features of patients with ESCC (according to IHC), were consistent with the results concerning the mRNA levels.

Table IV.

Associations between EPHB4/Ephrinb2 and caspase-8 protein expression and the clinical and pathological features of individuals with esophageal cancer (n=96 pairs).

Table IV.

Associations between EPHB4/Ephrinb2 and caspase-8 protein expression and the clinical and pathological features of individuals with esophageal cancer (n=96 pairs).

EPHB4EFNB2Caspase-8



FactorsnC/Nt/FP-valueC/Nt/FP-valueC/Nt/FP-value
Sex −1.0050.318 2.5890.011a 1.5920.115
  Male678.36±5.95 8.41±3.67 0.27±0.54
  Female299.62±4.86 6.48±2.41 0.30±0.35
Age, years 2.0440.135 2.0440.135 0.8320.438
  ≤35210.33±0.94 10.33±0.94 0.28±0.54
  35–50226.65±3.43 6.65±3.43 0.27±0.40
  ≥50729.34±6.11 9.34±6.11 0.28±0.44
Metastasis −2.8620.005a −2.9870.004a 5.072 <0.001a
  Yes57 10.06±6.05a 8.66±3.38 0.21±0.31
  No396.82±4.41 6.61±3.20 0.38±0.47
Family history −3.1810.002a −0.1360.892 1.7030.092
  Yes5510.26±6.03 7.78±3.40 0.29±0.43
  No416.72±4.40 7.88±3.55 0.28±0.33
Tumor size 13.129 <0.001a 4.1740.018a 16.534 <0.001a
  I345.71±2.60 6.50±2.59 0.36±0.41
  II429.23±4.33 8.47±3.23 0.25±0.31
  III2012.89±8.49 8.73±4.50 0.21±0.23
  I vs. II 0.003a 0.012a <0.001a
  I vs. III <0.001a 0.020a <0.001a
  II vs. III 0.009 0.777 0.108
Tumor stage 5.8820.004a 0.8200.443 4.0830.020a
  I285.91±2.50 7.73±3.13 0.32±0.22
  II339.33±4.56 7.32±3.33 0.28±0.26
  III3510.46±5.65 8.38±3.79 0.25±0.49
  I vs. II 0.015a 0.642 0.308
  I vs. III 0.001a 0.460 0.006a
  II vs. III 0.385 0.208 0.070
Tumor position 2.9860.055 0.2780.758 1.3370.268
  Upper286.65±3.12 0.017a7.44±3.23 0.28±0.43
  Middle509.83±6.66 0.1697.92±3.59 0.27±0.25
  Lower188.97±4.90 0.5708.17±3.49 0.31±0.31
  Upper vs. middle 0.560 0.114
  Upper vs. lower 0.490 0.272
  Middle vs. lower 0.797 0.877

{ label (or @symbol) needed for fn[@id='tfn4-ol-0-0-11160'] } Values expressed as mean ±SD. t values are provided for comparisons of 2 groups, while F values are provided for comparisons between ≥3 groups.

a P<0.05. EPHB4, EPH receptor B4; EFNB2, ephrin B2; C/N, the ratio of percentage of positively immunostaining cells in (C) esophagus cancer sample compared with (N) matched normal esophagus sample.

Expression of EphB4, EFNB2 or caspase-8 and clinical outcomes of ESCC

The univariate survival analysis demonstrated that patient age, family history and tumor metastasis were all significantly associated with survival time. The mRNA expression levels of EPHB4 and EFNB2 but not caspase-8, was associated with survival time and the protein expression levels of EPHB4 and caspase-8, but not EFNB2, was associated with survival time (Table V). Kaplan-Meier curves indicated that patients with higher mRNA (P<0.0001) and protein (P<0.0001) expression levels of EPHB4 exhibited a significantly shortened median survival time, compared with patients with lower expression levels (Fig. 3A and B; Table V). Similarly, patients with higher mRNA level of EFNB2 expression (P=0.041; Fig. 3C), or patients with lower protein level expression of caspase-8 expression (P=0.045; Fig. 3F) also exhibited a significantly shortened median survival time (Table V). In addition, as presented in Table V, patients with a family history of cancer exhibited a significantly decreased survival time compared with those without a family history of cancer (P<0.001). Furthermore, older patients also had a shortened survival time compared with those patients that were younger (P<0.001), and patients with metastatic tumors exhibited a markedly decreased survival time compared with those without tumor metastasis (P<0.001) (Table V). However, there were no significant associations observed between survival and sex, tumor size, stage or position. The multivariate analysis results revealed that the mRNA expression level of EPHB4 and EFNB2, the protein expression level of EPHB4 and caspase-8, metastasis and family history were all significant independent risk factors for ESCC, with hazard ratios of 5.290, 3.146, 1.394, 2.784, 1.885 and 1.786, respectively (Table VI).

Table V.

Univariate survival analysis of the association between expression levels of EPHB4, EFNB2 and caspase-8 and certain clinicopathological characteristics in patients with esophageal cancer.

Table V.

Univariate survival analysis of the association between expression levels of EPHB4, EFNB2 and caspase-8 and certain clinicopathological characteristics in patients with esophageal cancer.

FactorsCases (n=96)Events, nMedian survival, monthsSELog-rankP-value
Sex 1.220.259
  Male674735.5851.179
  Female292237.1771.765
Age, years 47.37 <0.001a
  <302237.6610.100
  30–50221636.0402.179
  >50725120.5001.077
Metastasis 25.30 <0.001a
  Yes573628.8681.624
  No393340.5791.079
Family history 15.95 <0.001a
  Yes553131.1261.152
  No413840.1501.125
Tumor size, cm3 2.130.334
  ≤100342938.2911.325
  100–200423034.5931.609
  >200201033.6072.517
TNM stage 0.440.809
  I282337.2331.662
  II332135.0721.660
  III352534.8351.761
Tumor position 3.480.175
  Upper282437.9371.779
  Middle503435.0721.424
  Lower181134.8351.740
EPHB4 (mRNA) 20.77 <0.001a
  Low333341.4001.154
  High633631.3580.960
EFNB2 (mRNA) 3.030.041a
  Low413837.8631.391
  High553133.8981.245
Caspase-8 (mRNA) 0.5320.466
  Low373348.5003.911
  High593652.8153.307
EPHB4 (protein) 7.4200.006a
  Low483639.5251.274
  High483330.1681.342
EFNB2 (protein) 2.715   0.095
  Low463738.5730.641
  High503232.4172.169
Caspase-8 (protein) 4.016   0.045a
  Low362634.8981.245
  High604339.8631.391

a P<0.05. EPHB4, EPH receptor B4; EFNB2, ephrin B2; TNM, Tumor-Node-Metastasis.

Table VI.

Multivariate Cox proportional hazards regression analysis (n=96 pairs).

Table VI.

Multivariate Cox proportional hazards regression analysis (n=96 pairs).

VariablesHazard ratio95% CIP-value
Sex, male vs. female1.2530.533–2.9460.606
Age, years
  35–50 vs. ≤350.7410.341–1.6410.451
  ≥50 vs. ≤350.7620.355–1.6930.493
Metastasis, yes vs. no1.8851.545–2.5170.037a
Family history, yes vs. no1.7861.217–2.3890.026a
Tumor size, cm3
  100–200 vs. ≤1001.4720.668–2.1150.107
  >200 vs. ≤1001.6620.715–2.2620.227
Tumor stage
  II vs. I1.0010.441–1.3790.110
  III vs. I1.0090.449–1.3820.172
Tumor position
  Middle vs. upper0.9150.473–1.7710.752
  Lower vs. upper0.9360.484–1.8170.912
High vs. low expression
  EPHB4 (mRNA)5.2903.723–7.7060.012a
  EFNB2 (mRNA)3.1462.070–5.2480.037a
  Caspase-8 (mRNA)0.9360.323–2.7130.903
  EPHB4 (protein)1.3941.011–1.9680.035a
  EFNB2 (protein)1.3500.596–3.0580.472
  Caspase-8 (protein)2.7841.888–5.7270.031a

a P<0.05. EPHB4, EPH receptor B4; EFNB2, ephrin B2; CI, confidence interval.

Discussion

A number of studies have reported that EPHB4 and/or EFNB2 expression is upregulated in multiple malignancies, including gastric (20), colon (21), uterine endometrial (22,23), breast (24), cervical (25) and ovarian cancer (26), melanoma (27), esophageal squamous cell carcinoma (14,16) and squamous cell carcinoma of the head and neck (28), which suggests that EPHB4 and EFNB2 may serve an oncogenic role in these tumor types. In the present study, it was observed that the expression of either EPHB4 or EPNB2 was increased in ESCC samples compared with that in corresponding normal esophageal tissues. It has been previously demonstrated that the upregulation of EPHB4 or EPNB2 is associated with metastasis and decreased survival in patients with ESCC (14,16); however, the present study revealed that it is also associated with tumor size and position, and family history, as well as confirming its association with decreased survival. Therefore, EPHB4 and EFNB2 may also serve oncogenic roles in the development and progression of ESCC.

The present study revealed that EPHB4 expression exhibited a positive correlation with EFNB2 expression, at both the mRNA and protein level; furthermore, IHC demonstrated that both molecules were expressed in the majority of ESCC cells. Considering they are cognate receptors and ligands, the aforementioned results suggested their potential ligation and the activation of downstream pathways in ESCC. Previous studies demonstrated that the activation of EPHB4 and/or EFNB2 triggered ‘forward’ and ‘reverse’ bidirectional signaling (2,3), which may stimulate angiogenesis in vivo (25,2932), and stimulated the growth of primary and metastatic tumor cells (33,34). The EPHB4 ‘forward’ signaling was able to promote the proliferation and migration of endothelial cells via the Pl-3 kinase pathway, which increased the formation of new cancer vasculature (35). The EFNB2 ‘reverse’ signaling, upon activation by EPHB4, not only induced an angiogenic response in cultured endothelial cells, but also promoted angiogenesis in breast cancer xenografts in vivo (35). In addition, EPHB4 and EFNB2 were also revealed to promote angiogenesis-independent tumor formation, in which the EFNB2-dependent EPHB4 ‘forward’ signaling enhanced the migration and invasion of melanoma cells (36), via the activation of RhoA GTPase. The present study determined that EPHB4 expression was associated with tumor size, metastasis and stage, indicating that EPHB4 may influence ESCC cell proliferation and migration. EPHB4 has been reported to promote the proliferation and migration of tumor cells in a variety of different cancer types (22,23,28,3642), which supports the results of the present study. In addition, the present study demonstrated that the upregulation of EPHB4 and EFNB2 was associated with poor outcome, and there have been similar reports in squamous cell carcinoma of the head and neck (28), as well as in endometrial (22) and ovarian cancer (26,43).

Resistance to apoptosis is required for tumor growth, and is a hallmark of cancer cells (44). Apoptosis resistance contributes to tumorigenesis, and results in the failure of cytotoxic therapies and a poor prognosis in patients, suggesting that targeting apoptotic pathways may represent a promising therapeutic approach for anticancer treatment. Accumulating evidence has demonstrated that apoptosis resistance, caused by downregulation of proapoptotic signaling molecules (such as caspase-8), frequently occurs in tumors of various origins. The present study demonstrated that the mRNA and protein level of caspase-8 was significantly downregulated in ESCC tissues compared with that in paracancerous tissues, indicating that this molecule may influence escape from endogenous growth control in the development and progression of ESCCs, which was similar to the findings previously reported (19). However, the present study also revealed that the expression of caspase-8 was associated with certain clinicopathological characteristics, including metastasis, tumor size, position and stage, and patient prognosis, in contrast to certain previously reported results (18). In conclusion, the downregulation of caspase-8 expression in ESCC suggested that it may serve as a useful predictor of prognosis in this type of cancer. Furthermore, the present study analyzed the associations between EPHB4, EFNB2 and caspase-8 in ESCC. The results revealed that, in ESCC tissues, the expression levels of EPHB4 and EFNB2 were negatively correlated with caspase-8 at both the mRNA and protein levels, which, to the best of our knowledge, has not been yet reported elsewhere.

The present study indicates that the upregulation of EPHB4 and EFNB2 expression in tumor cells promotes growth (via the inhibition of apoptotic pathways), which may be facilitated by a decrease in caspase-8 expression, resulting from regulation of the downstream effectors of EPHB4/EFNB2. A diagram representing the underlying molecular mechanism concerning the role of EPHB4, EFNB2 and caspase-8 in ESCC cells is exhibited in Fig. 4. The negative association between caspase-8 activation and EPHB4 expression has been previously reported in ovarian carcinoma (26), and is consistent with the results of the present study. The Ras/MAPK/ERK and Akt signaling pathways, downstream of EPHB4, could confer anti-apoptotic characteristics. However, the molecular mechanism underlying the negative correlation between EPHB4/EFNB2 and caspase-8 expression requires further investigation. Overall, the upregulation of EPHB4 and EFNB2 in tumor cells may disrupt caspase-8-mediated apoptosis and confer a survival advantage in tumor cells.

In summary, the present study reported that both EPHB4 and EFNB2 were upregulated, while caspase-8 was downregulated, in ESCC tissues compared with that in matched normal tissues. Expression levels were closely associated with a number of clinicopathological features, as well as patient survival. The current findings indicate the importance of the three molecules studied with regard to the genesis and progression of ESCC. Consequently, the expression levels of EPHB4, EFNB2 and caspase-8 may serve as biological signatures and useful prognostic indicators in ESCC, as well as potentially representing novel therapeutic targets in this type of cancer.

Acknowledgements

Not applicable.

Funding

The present study was supported by the National Key R&D Program of China (grant no. 2018YFC1603002 and 2018YFC1604404); the ‘Personalized Medicines-Molecular Signature-based Drug Discovery and Development’, Strategic Priority Research Program of the Chinese Academy of Sciences (grant no. XDA12010316); the National Natural Science Foundation of China (grant nos. 31520103907, 81730083) to Dong Xie; the National Natural Science Foundation of China (grant nos. 31771538, 81972757); the Youth Innovation Promotion Association of the Chinese Academy of Sciences Fund and the Sanofi-SIBS 2018 Young Faculty Award; and Postdoctoral Science Foundation of China (grant no. 2017M622677).

Availability of data and materials

The datasets used and/or analyzed during the present study are available from the corresponding author on reasonable request.

Authors' contributions

XM, DX and JL conceived and designed the experiments. QN, BZ and PC performed the experiments. QN wrote the manuscript. QN collected and analyzed the data. PC assisted with revising the manuscript. All authors read and approved the final manuscript.

Ethical approval and consent to participate

All procedures performed in studies involving human participants were in accordance with the ethical standards of the institutional and/or national research committee and with the 1964 Helsinki declaration and its later amendments or comparable ethical standards. The present study was approved by the Institutional Review Board of the Institute for Nutritional Sciences, Chinese Academy of Sciences (project number 30930023). Written informed consent was obtained from all patients.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Unified nomenclature for Eph family receptors and their ligands, the ephrins. Eph Nomenclature Committee. Cell. 90:403–404. 1997. View Article : Google Scholar : PubMed/NCBI

2 

Wilkinson DG: Multiple roles of EPH receptors and ephrins in neural development. Nat Rev Neurosci. 2:155–164. 2001. View Article : Google Scholar : PubMed/NCBI

3 

Palmer A and Klein R: Multiple roles of ephrins in morphogenesis, neuronal networking, and brain function. Genes Dev. 17:1429–1450. 2003. View Article : Google Scholar : PubMed/NCBI

4 

Heroult M, Schaffner F and Augustin HG: Eph receptor and ephrin ligand-mediated interactions during angiogenesis and tumor progression. Exp Cell Res. 312:642–650. 2006. View Article : Google Scholar : PubMed/NCBI

5 

Kd gsbrun M and Eichmann A: A role for axon guidance receptors and ligands in blood vessel development and tumor angiogenesis. Cytokine Growth Factor Rev. 16:535–548. 2005. View Article : Google Scholar : PubMed/NCBI

6 

Batlle E, Bacani J, Begthel H, Jonkheer S, Gregorieff A, van de Born M, Malats N, Sancho E, Boon E, Pawson T, et al: EphB receptor activity suppresses colorectal cancer progression. Naturec. 435:1126–1130. 2005. View Article : Google Scholar

7 

Huusko P, Ponciano-Jackson D, Wolf M, Kiefer JA, Azorsa DO, Tuzmen S, Weaver D, Robbins C, Moses T, Allinen M, et al: Nonsense-mediated decay microarray analysis identifies mutations of EPHB2 in human prostate cancer. Nat Genet. 36:979–983. 2004. View Article : Google Scholar : PubMed/NCBI

8 

Okumura F, Joo-Okumura A, Obara K, Petersen A, Nishikimi A, Fukui Y, Nakatsukasa K and Kamura T: Ubiquitin ligase SPSB4 diminishes cell repulsive responses mediated by EphB2. Mol Biol Cell. 8:3532–3541. 2017. View Article : Google Scholar

9 

Teitz T, Lahti JM and Kidd VJ: Aggressive childhood neuroblastomas do not express caspase-8: An important component of programmed cell death. J Mol Med (Berlin, Germany). 79:428–436. 2001. View Article : Google Scholar

10 

Teng Y, Dong YC, Liu Z, Zou Y, Xie H, Zhao Y, Su J, Cao F, Jin H and Ren H: DNA methylation-mediated caspase-8 downregulation is associated with anti-apoptotic activity and human malignant glioma grade. Int J Mol Med. 39:725–733. 2017. View Article : Google Scholar : PubMed/NCBI

11 

Zhang Y: Epidemiology of esophageal cancer. World J Gastroenterol. 19:5598–5606. 2013. View Article : Google Scholar : PubMed/NCBI

12 

Siegel RL, Miller KD and Jemal A: Cancer Statistics, 2017. CA Cancer J Clin. 67:7–30. 2017. View Article : Google Scholar : PubMed/NCBI

13 

Domper Arnal MJ, Ferrandez Arenas A and Lanas Arbeloa A: Esophageal cancer: Risk factors, screening and endoscopic treatment in Western and Eastern countries. World J Gastroenterol. 21:7933–7943. 2015. View Article : Google Scholar : PubMed/NCBI

14 

Tachibana M, Tonomoto Y, Hyakudomi R, Hyakudomi M, Hattori S, Ueda S, Kinugasa S and Yoshimura H: Expression and prognostic significance of EFNB2 and EPHB4 genes in patients with oesophageal squamous cell carcinoma. Dig Liver Dis. 39:725–732. 2007. View Article : Google Scholar : PubMed/NCBI

15 

Rice TW, Ishwaran H, Ferguson MK, Blackstone EH and Goldstraw P: Cancer of the esophagus and esophagogastric junction: An eighth edition staging primer. J Thorac Oncol. 12:36–42. 2017. View Article : Google Scholar : PubMed/NCBI

16 

Wang Y, Liu DP, Chen PP, Koeffler HP, Tong XJ and Xie D: Involvement of IFN regulatory factor (IRF)-1 and IRF-2 in the formation and progression of human esophageal cancers. Cancer Res. 67:2535–2543. 2007. View Article : Google Scholar : PubMed/NCBI

17 

Hasina R, Mollberg N, Kawada I, Mutreja K, Kanade G, Yala S, Surati M, Liu R, Li X, Zhou Y, et al: Critical role for the receptor tyrosine kinase EPHB4 in esophageal cancers. Cancer Res. 73:184–194. 2013. View Article : Google Scholar : PubMed/NCBI

18 

Takikita M, Hu N, Shou JZ, Wang QH, Giffen C, Taylor PR and Hewitt SM: Biomarkers of apoptosis and survival in esophageal squamous cell carcinoma. BMC Cancer. 9:3102009. View Article : Google Scholar : PubMed/NCBI

19 

Xue LY, Hu N, Song YM, Zou SM, Shou JZ, Qian LX, Ren LQ, Lin DM, Tong T, He ZG, et al: Tissue microarray analysis reveals a tight correlation between protein expression pattern and progression of esophageal squamous cell carcinoma. BMC Cancer. 6:2962006. View Article : Google Scholar : PubMed/NCBI

20 

Yin J, Cui Y, Li L, Ji J and Jiang WG: Overexpression of EPHB4 is associated with poor survival of patients with gastric cancer. Anticancer Res. 37:4489–4497. 2017.PubMed/NCBI

21 

Liu W, Ahmad SA, Jung YD, Reinmuth N, Fan F, Bucana CD and Ellis LM: Coexpression of ephrin-Bs and their receptors in colon carcinoma. Cancer. 94:934–939. 2002. View Article : Google Scholar : PubMed/NCBI

22 

Alam S, Fujimoto J, Jahan I, Sato E and Tamaya T: Overexpression of ephrinB2 and EPHB4 in tumor advancement of uterine endometrial cancers. Ann Oncol. 18:485–490. 2007. View Article : Google Scholar : PubMed/NCBI

23 

Takai N, Miyazaki T, Fujisawa K, Nasu K and Miyakawa I: Expression of receptor tyrosine kinase EPHB4 and its ligand ephrin-B2 is associated with malignant potential in endometrial cancer. Oncol Rep. 8:567–573. 2001.PubMed/NCBI

24 

Li X, Song C, Huang G, Sun S, Qiao J, Zhao J, Zhao Z and Li M: The coexpression of EPHB4 and EphrinB2 is associated with poor prognosis in HER2-positive breast cancer. OncoTargets Ther. 10:1735–1742. 2017. View Article : Google Scholar

25 

Zhang S, Jiang T and Liang M: Expression of Eph B4 and Ephrin B2 in cervical cancer tissues and angiogenesis. Int J Gynaecol Obstet. 96:46–47. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Kumar SR, Masood R, Spannuth WA, Singh J, Scehnet J, Kleiber G, Jennings N, Deavers M, Krasnoperov V, Dubeau L, et al: The receptor tyrosine kinase EphB4 is overexpressed in ovarian cancer, provides survival signals and predicts poor outcome. Br J Cancer. 96:1083–1091. 2007. View Article : Google Scholar : PubMed/NCBI

27 

Neuber C, Belter B, Meister S, Hofheinz F, Bergmann R, Pietzsch HJ and Pietzsch J: Overexpression of receptor tyrosine kinase EPHB4 triggers tumor growth and hypoxia in A375 melanoma xenografts: Insights from multitracer small animal imaging experiments. Molecules. 23(pii): E4442018. View Article : Google Scholar : PubMed/NCBI

28 

Masood R, Kumar SR, Sinha UK, Crowe DL, Krasnoperov V, Reddy RK, Zozulya S, Singh J, Xia G, Broek D, et al: EPHB4 provides survival advantage to squamous cell carcinoma of the head and neck. Int J Cancer. 119:1236–1248. 2006. View Article : Google Scholar : PubMed/NCBI

29 

Erber R, Eichelsbacher U, Powajbo V, Korn T, Djonov V, Lin J, Hammes HP, Grobholz R, Ullrich A and Vajkoczy P: EphB4 controls blood vascular morphogenesis during postnatal angiogenesis. EMBO J. 25:628–641. 2006. View Article : Google Scholar : PubMed/NCBI

30 

Kertesz N, Krasnoperov V, Reddy R, Leshanski L, Kumar SR, Zozulya S and Gill PS: The soluble extracellular domain of EPHB4 (sEphB4) antagonizes EPHB4-EphrinB2 interaction, modulates angiogenesis, and inhibits tumor growth. Blood. 107:2330–2338. 2006. View Article : Google Scholar : PubMed/NCBI

31 

He S, Ding Y, Zhou J, Krasnoperov V, Zozulya S, Kumar SR, Ryan SJ, Gill PS and Hinton DR: Soluble EPHB4 regulates choroidal endothelial cell function and inhibits laser-induced choroidal neovascularization. Invest Ophthalmol Vis Sci. 46:4772–4779. 2005. View Article : Google Scholar : PubMed/NCBI

32 

Noren NK, Lu M, Freeman AL, Koolpe M and Pasquale EB: Interplay between EPHB4 on tumor cells and vascular ephrin-B2 regulates tumor growth. Proc Natl Acad Sci USA. 101:5583–5588. 2004. View Article : Google Scholar : PubMed/NCBI

33 

Folkman J: Angiogenesis in cancer, vascular, rheumatoid and other disease. Nat Med. 1:27–31. 1995. View Article : Google Scholar : PubMed/NCBI

34 

Bouck N: Angiogenesis: A mechanism by which oncogenes and tumor suppressor genes regulate tumorigenesis. Cancer Treat Res. 63:359–371. 1992. View Article : Google Scholar : PubMed/NCBI

35 

Steinle JJ, Meininger CJ, Forough R, Wu G, Wu MH and Granger HJ: Eph B4 receptor signaling mediates endothelial cell migration and proliferation via the phosphatidylinositol 3-kinase pathway. J Biol Chem. 277:43830–43835. 2002. View Article : Google Scholar : PubMed/NCBI

36 

Yang NY, Pasquale EB, Owen LB and Ethell IM: The EPHB4 receptor-tyrosine kinase promotes the migration of melanoma cells through Rho-mediated actin cytoskeleton reorganization. J Biol Chem. 281:32574–32586. 2006. View Article : Google Scholar : PubMed/NCBI

37 

Meyer S, Hafner C, Guba M, Flegel S, Geissler EK, Becker B, Koehl GE, Orso E, Landthaler M and Vogt T: Ephrin-B2 overexpression enhances integrin-mediated ECM-attachment and migration of B16 melanoma cells. Int J Oncol. 27:1197–1206. 2005.PubMed/NCBI

38 

Xia G, Kumar SR, Masood R, Koss M, Templeman C, Quinn D, Zhu S, Reddy R, Krasnoperov V and Gill PS: Up-regulation of EPHB4 in mesothelioma and its biological significance. Clin Cancer Res. 11:4305–4315. 2005. View Article : Google Scholar : PubMed/NCBI

39 

Xia G, Kumar SR, Masood R, Zhu S, Reddy R, Krasnoperov V, Quinn DI, Henshall SM, Sutherland RL, Pinski JK, et al: EPHB4 expression and biological significance in prostate cancer. Cancer Res. 65:4623–4632. 2005. View Article : Google Scholar : PubMed/NCBI

40 

Xia G, Kumar SR, Stein JP, Singh J, Krasnoperov V, Zhu S, Hassanieh L, Smith DL, Buscarini M, Broek D, et al: EPHB4 receptor tyrosine kinase is expressed in bladder cancer and provides signals for cell survival. Oncogene. 25:769–780. 2006. View Article : Google Scholar : PubMed/NCBI

41 

Lian H, Jia X, Shi N, Xie S, Wang J, Wang W, Ma F, Liu H, Wang A, Cheng X and Liu C: Notch signaling promotes serrated neoplasia pathway in colorectal cancer through epigenetic modification of EPHB2 and EPHB4. Cancer Manag Res. 10:6129–6141. 2018. View Article : Google Scholar : PubMed/NCBI

42 

Lv J, Xia Q, Wang J, Shen Q, Zhang J and Zhou X: EPHB4 promotes the proliferation, invasion, and angiogenesis of human colorectal cancer. Exp Mol Pathol. 100:402–408. 2016. View Article : Google Scholar : PubMed/NCBI

43 

Wu Q, Suo Z, Kristensen GB, Baekelandt M and Nesland JM: The prognostic impact of EphB2/B4 expression on patients with advanced ovarian carcinoma. Gynecol Oncol. 102:15–21. 2006. View Article : Google Scholar : PubMed/NCBI

44 

Hanahan D and Weinberg RA: The hallmarks of cancer. Cell. 100:57–70. 2000. View Article : Google Scholar : PubMed/NCBI

Related Articles

Journal Cover

January-2020
Volume 19 Issue 1

Print ISSN: 1792-1074
Online ISSN:1792-1082

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Ni Q, Chen P, Zhu B, Li J, Xie D and Ma X: Expression levels of EPHB4, EFNB2 and caspase‑8 are associated with clinicopathological features and progression of esophageal squamous cell cancer. Oncol Lett 19: 917-929, 2020
APA
Ni, Q., Chen, P., Zhu, B., Li, J., Xie, D., & Ma, X. (2020). Expression levels of EPHB4, EFNB2 and caspase‑8 are associated with clinicopathological features and progression of esophageal squamous cell cancer. Oncology Letters, 19, 917-929. https://doi.org/10.3892/ol.2019.11160
MLA
Ni, Q., Chen, P., Zhu, B., Li, J., Xie, D., Ma, X."Expression levels of EPHB4, EFNB2 and caspase‑8 are associated with clinicopathological features and progression of esophageal squamous cell cancer". Oncology Letters 19.1 (2020): 917-929.
Chicago
Ni, Q., Chen, P., Zhu, B., Li, J., Xie, D., Ma, X."Expression levels of EPHB4, EFNB2 and caspase‑8 are associated with clinicopathological features and progression of esophageal squamous cell cancer". Oncology Letters 19, no. 1 (2020): 917-929. https://doi.org/10.3892/ol.2019.11160