Open Access

Upregulation of ECT2 is associated with transcriptional program of cancer stem cells and predicts poor clinical outcome in gastric cancer

  • Authors:
    • Dao-Yin Gong
    • Xian Chen
    • Tian‑Lin Yang
    • Yi Wang
    • Yu Guo
    • Jin‑Hao Zeng
    • Shan‑Ze Chen
  • View Affiliations

  • Published online on: July 27, 2020     https://doi.org/10.3892/ol.2020.11915
  • Article Number: 54
  • Copyright: © Gong 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

Gastric cancer remains the third leading cause of cancer‑associated mortality worldwide. The identification of prognostic indicators that are associated with clinical characteristics is urgently required. The aim of the present study was to determine the involvement of epithelial cell transforming 2 (ECT2) in gastric cancer. The results of the present study demonstrated that ECT2 expression was upregulated in human gastric cancer samples. Furthermore, high ECT2 expression was associated with advanced Tumor‑Node‑Metastasis stage and deeper tumor invasion. ECT2 upregulation was further confirmed in several independent publicly available clinical cohorts from the Gene Expression Omnibus database. In addition, patients with gastric cancer, with high ECT2 expression exhibited a significantly shorter overall survival time than those with low ECT2 expression, and Cox regression analysis demonstrated that ECT2 expression was an independent prognostic marker for overall survival time. Characterization of the transcriptome profiles of ECT2 upregulated gastric tumors indicated that ECT2 upregulation may be associated with transcriptional features of cancer stem cells (CSCs). Additionally, BUB1 mitotic checkpoint serine/threonine kinase and E2F transcription factor 7, two genes previously reported to account for the functionality of CSCs, were strongly enriched in ECT2High gastric cancer samples. Taken together, the results of the present study suggest that ECT2 may serve as a novel marker for CSCs and may be a potential prognostic indicator in gastric cancer.

Introduction

Cancer stem cells (CSCs), also known as tumor-initiating cells, are characterized by stem-like properties, including self-renewal and ability to generate daughter cells. Cancer initiation, dissemination and recurrence are closely associated with CSCs (1). CSCs were first detected in acute myeloid leukemia and have since been identified in several solid tumors, including gastric cancer (1). Furthermore, certain populations of gastric CSCs abilities to self-renewal and undergo multipotent differentiation have been detected in gastric cancer (2). Villin+ and Lgr5+ gastric stem cells have been detected in the antrum, while Troy+ chief cells have been found in the corpus (3). Additionally, Sox2+ gastric stem cells are present in both the antrum and the corpus (3).

Gastric cancer is the seventh most common cancer and the third leading cause of cancer-associated mortality worldwide (4). In 2018, 1,033,701 new gastric cancer cases and 782,685 mortalities were reported worldwide (4). Gastric cancer has been extensively investigated in the biomedical field due to its high morbidity and mortality rates (4). It is speculated that gastric carcinogenesis may be associated with Helicobacter pylori infection, inherited susceptibilities, and environmental and dietary factors (5,6). In recent years, the prevailing hypothesis that the occurrence and progression of gastric cancer is associated with CSCs has been partially proven (7).

Epithelial cell transforming 2 (ECT2) is a proto-oncogene gene encoding a guanine nucleotide exchange factor for the Rho GTPases (8). When expressed in NIH/3T3 fibroblasts, ECT2 promotes their malignant transformation (9). Increased ECT2 expression has been detected in several types of human tumor, including glioma and liver, pancreatic and lung cancer (1013). ECT2 upregulation significantly enhances the activity of RhoGPase, prevents cell apoptosis and induces cancer cell metastasis (10). Conversely, ECT2 downregulation suppresses activation of the ERK signaling pathway and impairs the migration of cancer cells (10). However, whether and how ECT2 contributes to gastric cancer malignancy remains elusive.

The present study aimed to investigate the association between ECT2 expression and the clinicopathological characteristics of patients with gastric cancer. The expression levels of ECT2 were investigated using immunohistochemical analysis, combined with Gene Expression Omnibus database and gene set enrichment analysis, and it was revealed that gastric tumors with elevated ECT2 levels exhibited transcriptional traits of CSCs. In addition, high ECT2 expression predicted poor clinical outcome, suggesting its use as a novel prognostic indicator for gastric carcinoma. Further investigation into the role of ECT2 may provide alternative therapeutic targets for the treatment of gastric cancer.

Materials and methods

Clinical tissue samples

A total of 130 primary gastric cancer tissues and 108 paired adjacent normal tissues (some paired adjacent normal tissues were not harvested during the operation due to patients clinical conditions) were collected from patients who underwent surgery at the Hospital of Chengdu University of TCM (Chengdu, China) between March 2012 and December 2015, and retrospectively analyzed. Paraffin-embedded tissue samples were stored at room temperature. None of the patients had received anticancer treatment prior to diagnosis and no additional malignancies were present. Pathological staging was based on the Union for International Cancer Control/American Joint Committee on Cancer Tumor-Node-Metastasis (TNM) Classification (8th edition of 2016) (14). The present study was approved by the Institutional Review Board of the Teaching Hospital of Chengdu University of TCM (Chengdu, China) (approval no. 2018KL-023) and written informed consent was provided by all patients prior to the study start.

Immunohistochemistry (IHC)

The tissue samples were fixed in 4% paraformaldehyde >24 h at room temperature, then dehydrated in graded ethanol series (30, 50, 70, 95 and 100%), and embedded in paraffin. For IHC analysis, paraffin-embedded samples were cut into 3-µm-thick sections, dewaxed with xylene at room temperature and rehydrated in a descending ethanol series (100, 95, 85 and 75%). For antigen retrieval, sections were heated at 97°C for 20 min. Following a brief proteolytic digestion with 0.1% trypsin at 37°C for 10 min and peroxidase blocking with 3% hydrogen peroxide solution at room temperature for 15 min, the sections were incubated with primary antibodies against: ECT2 (1:400; cat. no. 07-1364; Sigma-Aldrich, Merck KGaA), BUB1 (1:200; cat. no. DF6698; Affinity Biosciences) and E2F transcription factor 7 (E2F7; 1:200; cat. no. DF2444; Affinity Biosciences) overnight at 4°C. Following the primary antibody incubation, the sections were incubated with a HRP/Fab secondary antibody at room temperature for 20 min (freshly prepared solution from the kit; cat. no. PV-6000-D; Beijing Zhongshan Golden Bridge Biotechnology Co., Ltd.). Tissue sections were stained with diaminobenzidine substrate for 5 min and counterstained with hematoxylin for 20 sec at room temperature. Each slide was analyed using light microscopy (H-7650; Hitachi, Ltd.). The magnification used was ×200.

A total of two independent investigators, without prior knowledge of the clinicopathological data, evaluated the ECT2 staining in a semiquantitative manner. The final immunoreactivity scores (IRS) were determined according to the sum total of the percentage of positive cells (0 points, 0–5% positive cells; 1 point, 6–25%; 2 points, 26–50%; 3 points, 51–75% and 4 points, 76–100%), and staining intensity scores (0 points, no staining; 1 point, weak staining; 2 points, moderate staining and 3 points, strong staining). A final IRS >4 indicated strong positivity, while scores <4 indicated weak positivity.

ECT2 analysis in the Gene Expression Omnibus (GEO) database

ECT2 expression was assessed in several independent gastric cancer clinical datasets (1518) available from the GEO database (https://www.ncbi.nlm.nih.gov/geo/). The GSE13861 dataset (15) included a collection of 65 primary gastric adenocarcinoma and 19 surrounding normal tissues. The GSE29272 (16) dataset included a cohort of 134 gastric adenocarcinoma and paired surrounding normal tissues. The GSE51575 (17) dataset consisted of a cohort of 27 advanced gastric carcinoma and paired surrounding normal tissues. The GSE65801 (18) dataset consisted of a cohort of 32 gastric cancer tissues and paired surrounding noncancerous tissues. The normalization procedures employed for gene expression intensity data are stated in the individual datasets and related publications.

Gene set enrichment analysis (GSEA)

In order to determine how biological processes and signaling pathways are differentially regulated in gastric cancer with low or high ECT2 expression, transcriptomic data of gastric cancer were retrieved from The Cancer Genome Atlas (TCGA) database and analyzed using GSEA. TCGA gastric cancer cohort, consisting of 407 samples and transcriptional profiles, was downloaded from TCGA Data Portal (https://tcga-data.nci.nih.gov/docs/publications/tcga). GSEA was performed using GSEA software (v2.2.2; www.broadinstitute.org/gsea). The median ECT2 expression level (cut-off value=11.13) was used to dichotomize samples into low and high expression groups. A total of 1,000 permutations were used to calculate the P-values. All other parameters were set based on their default values.

Survival analysis

The prognostic value of ECT2 in gastric cancer was assessed using the Kaplan-Meier (KM) Plotter database (http://kmplot.com/analysis/index.php?p=service&cancer=gastric), which consists of a pool of gene expression and clinical data (19). The median time to first progression (FP) was 18.3 months and the median overall survival (OS) was 28.9 months. Overall survival time was assessed. The patient samples were divided into two groups according to the median gene expression value (ECT2High/ECT2Low, 437 cases/438 cases). A KM survival plot was used to compare the two groups. The hazard ratio (HR) with 95% confidence intervals (CIs) and log rank P-values were calculated.

Statistical analysis

Data are presented as the mean ± standard error of the mean from three independent experiments and were analyzed with SPSS v.22.0 software (IBM, Corp.). Pearsons χ2 test and Fishers exact test were used to assess the association between ECT2 expression and clinicopathological characteristics of patients with gastric cancer. Unpaired Students t-test was used to assess the differences in ECT2, BUB1 and E2F7 expression levels between gastric cancer and control tissues. Spearmans correlation test was performed between BUB1 and ECT2 expression levels, and E2F7 and ECT2 expression levels, respectively. Survival analysis was performed using the KM method and the log-rank test was used to assess statistical significance between the curves. Univariate and multivariate survival analyses were performed using the Cox proportional hazards regression model. P<0.05 was considered to indicate a statistically significant difference.

Results

ECT2 expression is upregulated in human gastric cancer

Following a routine H&E staining, IHC was performed to detect and grade ECT2 expression in gastric cancer and paired normal tissue sections. ECT2 positive staining was detected mainly in the nucleus and cytoplasm of gastric cancer cells. Weak staining was observed in the adjacent normal tissues. ECT2 expression was identified in 81.5% (106/130) of the gastric carcinoma tissues and 36.1% (39/108) of the adjacent normal tissues. The level of ECT2 protein was significantly higher in the gastric carcinoma tissues compared with the adjacent tissues (IRS, cancer=3.12±2.00 vs. normal=0.98±1.56; P<0.01; Fig. 1A and B).

Association between clinicopathological characteristics and ECT2 positivity in gastric cancer

The clinicopathological significance of ECT2 expression in gastric cancer was investigated. Analysis of the association between ECT2 expression levels (strong positivity vs. weak positivity/absent) and clinicopathological characteristics demonstrated that strong ECT2 positivity was significantly associated with advanced TNM stage (P<0.001) and higher pT stage (deeper tumor invasion; P=0.039). In the 130 gastric cancer cases assessed, high ECT2 expression was not associated with age, sex, tumor localization, pN stage and pM stage (all P>0.05; Table I; Table SI).

Table I.

Correlation between ECT2 positivity and clinicopathological characteristics of patients with gastric cancer (n=130).

Table I.

Correlation between ECT2 positivity and clinicopathological characteristics of patients with gastric cancer (n=130).

CharacteristicsTotal n=130ECT2 strong positivity, n=35ECT2 weak positivity/absent, n=95P-value
Age, years 0.241
  <60672146
  ≥60631449
Sex 0.077
  Male932172
  Female371423
Tumor localization 0.091
  Cardias20713
  Body401525
  Antrum52844
  Whole/Multiple18513
Histology 0.216
  ADC, WD19514
  ADC, MD42735
  ADC, PD481929
  Signet ring cell14311
  Mucinous adenocarcinoma615
  Neuroendocrine carcinoma101
TNM stage <0.001
  I + II67859
  III + IV632736
pT (Tumor invasion) 0.039
  T1 + T222220
  T3 + T41083375
pN (Lymph node metastasis) 0.896
  N0421131
  N1-N3882464
pM (Distant metastasis) 0.473
  M01142985
  M116610

[i] ECT2, epithelial cell transforming 2; ADC, adenocarcinoma; WD, well differentiated; MD, moderately differentiated; PD, poorly differentiated; TNM, tumor-node-metastasis.

A significant association between strong ECT2 positivity and histological patterns of gastric cancer was not established (Table I; Table SI); however, ECT2 expression was closely associated with the histological differentiation degree (Fig. 1C). ECT2 expression escalated from well differentiated (WD), to moderately differentiated (MD) and to poorly differentiated (PD) variants (IRS, PD=4.06±1.87 vs. WD=2.16±2.06 or MD=2.64±1.83; P<0.01; Fig. 1C).

ECT2 mRNA upregulation in public datasets of gastric cancer

To further investigate the pathological role of ECT2 in the progression of gastric cancer, the ECT2 expression pattern in gastric cancer samples based on transcriptomic data from the GEO database was assessed. Consistent with IHC analysis, ECT2 mRNA expression levels were significantly increased in primary gastric adenocarcinoma tissues compared with surrounding normal tissues in the GSE13861 dataset (P<0.0001; Fig. 2A). Similar trends were observed in comparisons between paired samples of gastric carcinoma and adjacent normal tissues in the GSE29272 dataset (P<0.0001; Fig. 2B), advanced gastric carcinoma and adjacent normal tissues in the GSE51575 dataset (P<0.0001; Fig. 2C), and gastric cancer tissues and adjacent normal tissues in the GSE65801 dataset (P<0.0001; Fig. 2D). Collectively, these results indicate that ECT2 upregulation may play an important role in the malignant progression of human gastric cancer.

ECT2 upregulation predicts poor clinical outcome

The association between ECT2 expression and the prognosis of patients with gastric cancer was assessed using the KM plot database. High ECT2 expression was significantly associated with a shorter survival time in patients with gastric cancer, stratified according to ECT2 expression levels. The median survival time for ECT2High patients was 16 months, which was significantly shorter than the 26 months observed for ECT2Low patients (P=0.0017; Fig. 3A). Survival analysis using KM curves was performed to verify these results. The results demonstrated that patients with gastric cancer, with low ECT2 expression exhibited a significantly longer overall survival time than those with high ECT2 expression (P=0.015; median OS, 74.360 vs. 50.430 months; Fig. 3B).

To determine whether ECT2 is an independent prognostic factor for the survival of patients with gastric cancer, univariate and multivariate Cox regression analyses were performed. As presented in Table II, the univariate analysis suggested that ECT2 was significantly associated with overall survival time in patients with gastric cancer [P=0.017; HR (95% CI), 1.905 (1.122-3.233)]. Tumor location (body, whole and multiple), histology (PD of adenocarcinoma, signet ring cell, mucinous adenocarcinoma) and TNM stage were all associated with overall survival time in patients with gastric cancer (all P<0.05). Multivariate analysis further demonstrated that high ECT2 expression was a significant independent prognostic marker for patients with gastric cancer [P=0.001; HR (95% CI), 3.105 (1.567-6.153)]. Taken together, these results suggest that ECT2 may serve as a prognostic biomarker for patients with gastric cancer.

Table II.

Prognostic factors associated with overall survival as determined by univariate and multivariate analyses.

Table II.

Prognostic factors associated with overall survival as determined by univariate and multivariate analyses.

VariablesHR (95% CI)P-value
Univariate analysis
  ECT21.905 (1.122-3.233)0.017
  Tumor localization (Body)4.842 (1.436-16.328)0.011
  Tumor localization (Whole/Multiple)14.106 (4.057-49.039)<0.001
  Histology (ADC, PD)8.274 (2.538-26.971)<0.001
  Histology (signet ring cell)5.511 (1.403-21.644)0.014
  Histology (mucinous adenocarcinoma)5.319 (1.066-26.531)0.042
  TNM stage4.992 (2.797-8.909)<0.001
    pT stage18.481 (2.557-133.551)0.004
    pN stage2.846 (1.510-5.367)0.001
    pM stage3.013 (1.612-5.632)0.001
Multivariate analysis
  ECT23.105 (1.567-6.153)0.001
  Tumor localization (Whole/Multiple)13.301 (3.468-51.015)<0.001
  Histology (ADC, PD)4.109 (1.246-13.550)0.020
  Histology (Mucinous adenocarcinoma)6.186 (1.049-36.481)0.044
  pT stage12.216 (1.445-103.281)0.022
  pN stage3.967 (1.935-8.132)<0.001

[i] ECT2, epithelial cell transforming 2; ADC, adenocarcinoma; PD, poorly differentiated; TNM, tumor-node-metastasis; HR, hazard ratio; CI, confidence interval.

Gastric tumors with higher ECT2 expression levels possess transcriptional traits of CSCs

To determine the ECT2-associated cellular processes and signaling pathways in gastric cancer, GSEA was performed using transcriptome data from TCGA. GSEA demonstrated highly significant enrichment of breast-cancer-progenitor-related genes in gastric cancer samples with higher ECT2 expression (Fig. 4A). Notably, BUB1 mitotic checkpoint serine/threonine kinase and E2F7, two genes previously reported to account for CSC functionality (2022), were strongly enriched in ECT2High gastric cancer samples (Fig. 4B). A significant correlation was identified between ECT2 and BUB1 mRNA expression levels (r=0.63; P<0.0001; Fig. 5A), and between ECT2 and E2F7 mRNA expression levels in gastric cancer tissues (r=0.45; P<0.0001; Fig. 5B).

BUB1 and E2F7 IHC staining was performed in gastric cancer samples to verify these findings. The results demonstrated that BUB1 protein expression was significantly higher in gastric cancer tissues compared with adjacent normal tissues (IRS, cancer=4.61±1.89 vs. normal=1.94±0.89; P<0.01; Fig. 6A and B). Similarly, E2F7 protein expression was significantly higher in gastric cancer tissues compared with adjacent normal tissues (IRS, cancer=4.01±1.75 vs. normal=1.72±0.86; P<0.01; Fig. 6A and C). Collectively, these results suggest that gastric tumors with high ECT2 expression levels may possess transcriptional traits of CSCs.

Discussion

Gastric cancer remains the third leading cause of cancer-associated mortality worldwide (4). CSCs are implicated in different types of cancer, including gastric cancer (1). The identification of gastric CSCs has improved understanding of the molecular and cellular etiology of gastric cancer, and may aid the development of effective treatments. Experimentally, CSCs are characterized by their capacity for tumor propagation (2). CSCs are resistant to chemotherapy and radiotherapy and possess a quiescent nature (23). Thus, these cells play an important role in cancer recurrence (24). As a result, identification of specific gastric CSCs and the detection of their expression level will lead to the development of novel methods for the diagnosis and treatment of gastric cancer, which can further improve the survival rate of patients with the disease.

The results of the present study demonstrated that ECT2 expression was upregulated in gastric cancer tissues compared with adjacent normal tissues. This result was further verified based on the transcriptomic data from several independent clinical datasets. Consistent with the results of IHC analysis, ECT2 mRNA expression levels were significantly increased in gastric cancer tissues compared with adjacent normal tissues, suggesting that ECT2 upregulation may serve an important role in the malignant progression of human gastric cancer.

The present study also investigated the biological implications of ECT2 upregulation using GSEA. BUB1 and E2F7 upregulation have previously been demonstrated to play important roles in essential cellular processes, such as cell proliferation (2527). It is speculated that BUB1 and E2F7 may be associated with transcriptional features of CSCs (2022). A previous study revealed that BUB1 depletion using shRNAs reduces cancer stem cell potential of the MDA-MB-231 breast cancer cell line, resulting in inhibited formation of xenografts in immunocompromised mice (20). In addition, overexpression of E2F7 significantly enhanced the spheroid formation and growth rate of HepG2 and Huh7 cells (hepatocellular carcinoma cell lines), and also decreased their apoptosis (28). GSEA analysis indicated that ECT2 expression was notably associated with the transcriptional program of CSCs, with co-staining of BUB1 and E2F7 in gastric cancer tissues confirmed by IHC analysis.

The present study is not without limitations. First, only IHC analysis was performed to determine ECT2 protein expression, additional methods such as western blotting should be considered in future studies. However, IHC can simultaneously evaluate tissue expression localization, as well as morphology in cancer tissues, and thus is the preferred approach in analysis of clinical samples. Furthermore, future studies will focus on in vitro experiments to better understand the molecular mechanisms underlying ECT2 function in gastric cancer.

Carcinoembryonic antigen and cancer antigen 19-9 serve as the standard biomarkers for the diagnosis of gastric cancer; however, their use in clinical practice is limited due to low diagnostic sensitivity (29). Although emerging candidate biomarkers, such as microRNA and DNA methylation products have been extensively studied, several challenges hinder their application in a clinical setting (30). The results of the present study demonstrated that strong ECT2 positivity was significantly associated with advanced TNM stage and deeper tumor invasion. Furthermore, high ECT2 expression levels were associated with a shorter overall survival time. Thus, ECT2 expression may serve as an independent prognostic marker for the overall survival time of patients with gastric cancer. Similar results were reported by previous studies that demonstrated the prognostic value of ECT2 for gastric cancer (31,32). Taken together, the results of the present study suggest that upregulation of ECT2 predicts unfavorable clinical outcomes of patients with gastric cancer. Thus, ECT2 may serve as a potential prognostic marker and therapeutic target for the management of gastric cancer.

Supplementary Material

Supporting Data

Acknowledgements

Not applicable.

Funding

The present study was funded by the National Natural Science Foundation of China (grant nos. 81803183, 81804066, 81904178 and 81873073), the Key Scientific Research Foundation of Department of Science and Technology of Sichuan Province (grant no. 19ZX0161Z090116002), the Project of Sichuan Provincial Administration of TCM (grant no. 2018QN022), the Science and Technology Developmental Foundation of the Hospital of Chengdu University of TCM (grant nos. 19TS03, 19LW05 and 19LW06), the ‘Xing-lin Scholars’ Project of Chengdu University of Traditional Chinese Medicine (grant no. QNXZ2019017) and ‘Hundred Talents Program’ of the Hospital of Chengdu University of Traditional Chinese Medicine (grant nos. 20-Q03, 20-Q05 and 20-Q18).

Availability of data and materials

The datasets used during the present study are available from the corresponding author upon reasonable request.

Authors contributions

JHZ and SZC conceived and designed the present study. TLY and YG collected and prepared the clinical samples. XC and YW performed H&E staining and confirmed pathological diagnosis. JHZ and DYG performed IHC and clinicopathological characteristics analyses. SZC acquired, interpreted and analyzed the GEO and GSEA data. DYG and SZC drafted the initial manuscript and critically revised it for important intellectual content. All authors have read and approved the final manuscript to be published.

Ethics approval and consent to participate

This retrospective study was approved by the Institutional Review Board of the Teaching Hospital of Chengdu University of TCM (Chengdu, China) (approval no. 2018KL-023) and written informed consent was provided by all patients prior to the study start.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Shackleton M, Quintana E, Fearon ER and Morrison SJ: Heterogeneity in cancer: Cancer stem cells versus clonal evolution. Cell. 138:822–829. 2009. View Article : Google Scholar : PubMed/NCBI

2 

McCracken KW, Catá EM, Crawford CM, Sinagoga KL, Schumacher M, Rockich BE, Tsai YH, Mayhew CN, Spence JR, Zavros Y, et al: Modelling human development and disease in pluripotent stem-cell-derived gastric organoids. Nature. 516:400–404. 2014. View Article : Google Scholar : PubMed/NCBI

3 

Zhao Y, Feng F and Zhou YN: Stem cells in gastric cancer. World J Gastroenterol. 21:112–123. 2015. View Article : Google Scholar : PubMed/NCBI

4 

Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA and Jemal A: Global cancer statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 68:394–424. 2018. View Article : Google Scholar : PubMed/NCBI

5 

Maeda M, Yamashita S, Shimazu T, Iida N, Takeshima H, Nakajima T, Oda I, Nanjo S, Kusano C, Mori A, et al: Novel epigenetic markers for gastric cancer risk stratification in individuals after Helicobacter pylori eradication. Gastric Cancer. 21:745–755. 2018. View Article : Google Scholar : PubMed/NCBI

6 

Yaghoobi M, Bijarchi R and Narod SA: Family history and the risk of gastric cancer. Br J Cancer. 102:237–242. 2010. View Article : Google Scholar : PubMed/NCBI

7 

Matsuo J, Kimura S, Yamamura A, Koh CP, Hossain MZ, Heng DL, Kohu K, Voon DC, Hiai H, Unno M, et al: Identification of stem cells in the epithelium of the stomach corpus and antrum of mice. Gastroenterology. 152:218–231.e14. 2017. View Article : Google Scholar : PubMed/NCBI

8 

Tatsumoto T, Xie X, Blumenthal R, Okamoto I and Miki T: Human ECT2 is an exchange factor for Rho GTPases, phosphorylated in G2/M phases, and involved in cytokinesis. J Cell Biol. 147:921–928. 1999. View Article : Google Scholar : PubMed/NCBI

9 

Chan AM, McGovern ES, Catalano G, Fleming TP and Miki T: Expression cDNA cloning of a novel oncogene with sequence similarity to regulators of small GTP-binding proteins. Oncogene. 9:1057–1063. 1994.PubMed/NCBI

10 

Chen J, Xia H, Zhang X, Karthik S, Pratap SV, Ooi LL, Hong W and Hui KM: ECT2 regulates the Rho/ERK signalling axis to promote early recurrence in human hepatocellular carcinoma. J Hepatol. 62:1287–1295. 2015. View Article : Google Scholar : PubMed/NCBI

11 

Liu B, Yang H, Taher L, Denz A, Grützmann R, Pilarsky C and Weber GF: Identification of prognostic biomarkers by combined mRNA and miRNA expression microarray analysis in pancreatic cancer. Transl Oncol. 11:700–714. 2018. View Article : Google Scholar : PubMed/NCBI

12 

Zhou S, Wang P, Su X, Chen J, Chen H, Yang H, Fang A, Xie L, Yao Y and Yang J: High ECT2 expression is an independent prognostic factor for poor overall survival and recurrence-free survival in non-small cell lung adenocarcinoma. PLoS One. 12:e01873562017. View Article : Google Scholar : PubMed/NCBI

13 

Sano M, Genkai N, Yajima N, Tsuchiya N, Homma J, Tanaka R, Miki T and Yamanaka R: Expression level of ECT2 proto-oncogene correlates with prognosis in glioma patients. Oncol Rep. 16:1093–1098. 2006.PubMed/NCBI

14 

Amin MB, Edge S, Greene F, Byrd DR, Brookland RK, Washington MK, Gershenwald JE, Compton CC, Hess KR, Sullivan DC, Jessup JM, Brierley JD, Gaspar LE, Schilsky RL, Balch CM, Winchester DP, Asare EA, Madera M, Gress DM and Meyer LR: AJCC Cancer Staging Manual. 8th. Springer; New York, NY: 2016

15 

Cho JY, Lim JY, Cheong JH, Park YY, Yoon SL, Kim SM, Kim SB, Kim H, Hong SW, Park YN, et al: Gene expression signature-based prognostic risk score in gastric cancer. Clin Cancer Res. 17:1850–1857. 2011. View Article : Google Scholar : PubMed/NCBI

16 

Wang G, Hu N, Yang HH, Wang L, Su H, Wang C, Clifford R, Dawsey EM, Li JM, Ding T, et al: Comparison of global gene expression of gastric cardia and noncardia cancers from a high-risk population in china. PLoS One. 8:e638262013. View Article : Google Scholar : PubMed/NCBI

17 

Kim SY, Park C, Kim HJ, Park J, Hwang J, Kim JI, Choi MG, Kim S, Kim KM and Kang MS: Deregulation of immune response genes in patients with Epstein-Barr virus-associated gastric cancer and outcomes. Gastroenterology. 148:137–147.e9. 2015. View Article : Google Scholar : PubMed/NCBI

18 

Li H, Yu B, Li J, Su L, Yan M, Zhang J, Li C, Zhu Z and Liu B: Characterization of differentially expressed genes involved in pathways associated with gastric cancer. PLoS One. 10:e01250132015. View Article : Google Scholar : PubMed/NCBI

19 

Szász AM, Lánczky A, Nagy Á, Förster S, Hark K, Green JE, Boussioutas A, Busuttil R, Szabó A and Győrffy B: Cross-validation of survival associated biomarkers in gastric cancer using transcriptomic data of 1,065 patients. Oncotarget. 7:49322–49333. 2016. View Article : Google Scholar : PubMed/NCBI

20 

Han JY, Han YK, Park GY, Kim SD and Lee CG, Jo WS and Lee CG: Bub1 is required for maintaining cancer stem cells in breast cancer cell lines. Sci Rep. 5:159932015. View Article : Google Scholar : PubMed/NCBI

21 

Venere M, Miller TE and Rich JN: Mitotic control of cancer stem cells. Cancer Discov. 3:141–144. 2013. View Article : Google Scholar : PubMed/NCBI

22 

Mariani SA, Minieri V, De Dominici M, Iacobucci I, Peterson LF and Calabretta B: CDKN2A-independent role of BMI1 in promoting growth and survival of Ph+ acute lymphoblastic leukemia. Leukemia. 30:1682–1690. 2016. View Article : Google Scholar : PubMed/NCBI

23 

Hur W and Yoon SK: Molecular pathogenesis of radiation-induced cell toxicity in stem cells. Int J Mol Sci. 18:27492017. View Article : Google Scholar

24 

Moore N and Lyle S: Quiescent, slow-cycling stem cell populations in cancer: A review of the evidence and discussion of significance. J Oncol. 2011:3960762011. View Article : Google Scholar : PubMed/NCBI

25 

Gjoerup OV, Wu J, Chandler-Militello D, Williams GL, Zhao J, Schaffhausen B, Jat PS and Roberts TM: Surveillance mechanism linking Bub1 loss to the p53 pathway. Proc Natl Acad Sci USA. 104:8334–8339. 2007. View Article : Google Scholar : PubMed/NCBI

26 

Xu B, Xu T, Liu H, Min Q, Wang S and Song Q: miR-490-5p suppresses cell proliferation and invasion by targeting BUB1 in hepatocellular carcinoma cells. Pharmacology. 100:269–282. 2017. View Article : Google Scholar : PubMed/NCBI

27 

Mitxelena J, Apraiz A, Vallejo-Rodríguez J, Malumbres M and Zubiaga AM: E2F7 regulates transcription and maturation of multiple microRNAs to restrain cell proliferation. Nucleic Acids Res. 44:5557–5570. 2016. View Article : Google Scholar : PubMed/NCBI

28 

Ma YS, Lv ZW, Yu F, Chang ZY, Cong XL, Zhong XM, Lu GX, Zhu J and Fu D: MicroRNA-302a/d inhibits the self-renewal capability and cell cycle entry of liver cancer stem cells by targeting the E2F7/AKT axis. J Exp Clin Cancer Res. 37:2522018. View Article : Google Scholar : PubMed/NCBI

29 

Pan YQ, Ruan YY, Peng JB, Han QY, Zhang X, Lin A and Yan WH: Diagnostic significance of soluble human leukocyte antigen-G for gastric cancer. Hum Immunol. 77:317–324. 2016. View Article : Google Scholar : PubMed/NCBI

30 

Toiyama Y, Okugawa Y and Goel A: DNA methylation and microRNA biomarkers for noninvasive detection of gastric and colorectal cancer. Biochem Biophys Res Commun. 455:43–57. 2014. View Article : Google Scholar : PubMed/NCBI

31 

Jin Y, Yu Y, Shao Q, Ma Y, Zhang R, Yao H and Xu Y: Up-regulation of ECT2 is associated with poor prognosis in gastric cancer patients. Int J Clin Exp Pathol. 7:8724–8731. 2014.PubMed/NCBI

32 

Wang HB, Yan HC and Liu Y: Clinical significance of ECT2 expression in tissue and serum of gastric cancer patients. Clin Transl Oncol. 18:735–742. 2016. View Article : Google Scholar : PubMed/NCBIPubMed/NCBIPubMed/NCBIPubMed/NCBI

Related Articles

Journal Cover

October-2020
Volume 20 Issue 4

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
Gong D, Chen X, Yang TL, Wang Y, Guo Y, Zeng JH and Chen SZ: Upregulation of ECT2 is associated with transcriptional program of cancer stem cells and predicts poor clinical outcome in gastric cancer. Oncol Lett 20: 54, 2020
APA
Gong, D., Chen, X., Yang, T., Wang, Y., Guo, Y., Zeng, J., & Chen, S. (2020). Upregulation of ECT2 is associated with transcriptional program of cancer stem cells and predicts poor clinical outcome in gastric cancer. Oncology Letters, 20, 54. https://doi.org/10.3892/ol.2020.11915
MLA
Gong, D., Chen, X., Yang, T., Wang, Y., Guo, Y., Zeng, J., Chen, S."Upregulation of ECT2 is associated with transcriptional program of cancer stem cells and predicts poor clinical outcome in gastric cancer". Oncology Letters 20.4 (2020): 54.
Chicago
Gong, D., Chen, X., Yang, T., Wang, Y., Guo, Y., Zeng, J., Chen, S."Upregulation of ECT2 is associated with transcriptional program of cancer stem cells and predicts poor clinical outcome in gastric cancer". Oncology Letters 20, no. 4 (2020): 54. https://doi.org/10.3892/ol.2020.11915