Open Access

Overexpression of ELF1 combined with MMP9 is associated with prognosis and tumor microenvironment in gastric cancer

  • Authors:
    • Xiaoxia Zhang
    • Xiaoyan Ren
    • Shu Zhang
    • Yan Wang
  • View Affiliations

  • Published online on: September 27, 2024     https://doi.org/10.3892/etm.2024.12730
  • Article Number: 441
  • Copyright: © Zhang 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 (GC) is a prevalent malignancy of the digestive system. E74‑like factor 1 (ELF1) is a transcription factor that is specific to T cells and belongs to the Ets family. They are typically expressed in numerous tumor cells, such as pancreatic cancer, oral squamous cell, endometrial carcinoma, nasopharyngeal carcinoma and prostate and colorectal cancer, where they can promote cell invasion and migration. MMP9 is an important protease of the MMP family, since it serves a vital role in tumor progression and prognostic evaluation in colorectal cancer, uveal melanoma and clear cell renal cell carcinoma. The present study aimed to investigate the expression, correlation with MMP9 and clinical significance of ELF1 in GC. In addition, it aimed to explore the possible mechanisms. The ELF1 mRNA expression profile was first assessed using the GEPIA database and R4.2.1 software (Limma package). Reverse transcription‑quantitative PCR (RT‑qPCR) was used then to validate ELF1 mRNA expression levels in fresh GC samples from 40 patients. The clinical diagnostic value of ELF1 was also assessed using RT‑qPCR. Tissue microarray immunohistochemistry (TMA‑IHC) was utilized to examine the expression levels of ELF1 and MMP9 proteins in 355 paraffin‑embedded GC samples. Subsequently, the present study further investigated the relationship between ELF1 and MMP9 and their possible effects on the clinicopathological features and prognosis of patients with GC. Gene correlation analysis was conducted using the GEPIA database and complemented with Tumor Immune Estimation Resource (TIMER) and CIBERSORT analyses to explore associations with immune infiltration. A significantly higher expression of ELF1 mRNA was found in GC tissues compared with that in adjacent normal tissues (P<0.05). High ELF1 expression in GC tumor cells was found to distinguish GC tissues from adjacent normal tissues with a sensitivity of 87.5% and specificity of 77.5%. ELF1 and MMP9 proteins also showed higher expression in 355 GC compared with adjacent normal tissues, where they were significantly positively correlated (P<0.001). The two were closely associated with various clinicopathological features, including infiltration depth, lymph node involvement, metastasis, TNM staging, microscopic venous invasion, lymphatic invasion and blood serum carcinoembryonic antigen levels in GC. Furthermore, ELF1 and MMP9 expression levels were negatively associated with the overall survival of patients with GC. Prognostic analysis using the Cox proportional hazards model identified high ELF1 expression [hazards ratio (HR), 2.555; 95% CI, 1.546‑4.224; P=0.002], high MMP9 expression (HR, 3.813; 95% CI, 2.406‑6.041; P<0.001), advanced TNM stage (P=0.001) and advanced N stage (P=0.011) to be independent prognostic factors for patients with GC. Correlation analysis results from the GEPIA database indicated significant associations of ELF1 expression with various GC‑related genes, including MutL homolog 1, erythroblastic leukemia viral oncogene homolog 2, PI3K catalytic subunit α, and tumor suppressor protein 53, MMP‑9, Cadherin 1, TIMP1, growth factor A and kinase insert domain receptor. In addition, immune infiltration correlation analysis on TIMER and CIBERSORT revealed ELF1 positive relationship with specific infiltrating immune cell types, including naive B, memory‑activated CD4+ and gamma delta T cells, and activated NK cells (P<0.05). This observation was further confirmed using immunohistochemistry, showing that ELF1 was associated with CD19 (B‑cells) (P<0.001) and CD4 (CD4+ T cells, P=0.002). In conclusion, results from the present study suggest that ELF1 is overexpressed in GC. ELF1 combined with MMP9 can serve as a predictor of malignant biological behavior in GC and therefore a prognostic indicator for patients, due to its association with the tumor microenvironment.

Introduction

As a prevalent gastrointestinal malignancy, gastric cancer (GC) is distinguished by its high incidence, aggressive progression profile and high mortality rates. There were >968,000 novel cases of stomach cancer in 2022 and close to 660,000 deaths, ranking the disease as fifth in terms of both incidence and mortality worldwide (1). Despite a decline in the global prevalence and death rate of GC, rates remain high in Eastern Asian countries (2). The majority of patients with GC are typically diagnosed already at advanced stages during consultations, resulting in a poor prognosis post-surgery. This scenario poses considerable public health challenges (3). Consequently, the quest for efficacious molecular markers for GC assumes paramount importance in facilitating precise treatment design, extending patient survival and enhancing their overall quality of life.

The primary approach for treating GC continues to involve conventional surgical procedures, followed by postoperative radiotherapy and chemotherapy (4). However, the 5-year survival rate following surgery remains low (<10%) (5). With the rapid advancement of molecular biology research, GC treatment methods have undergone continuous improvements, including the introduction of immunotherapy, such as programmed cell death protein (PD)-1/PD-L1 inhibitors and cytotoxic T lymphocyte antigen 4 inhibitors (6). Additionally, the emergence of chemotherapeutic and molecular-targeted drugs, such as oxaliplatin and herceptin, has instilled optimism among patients with intermediate and advanced stages of GC. Although these interventions have led to improved postoperative survival rates, the prognosis for GC, particularly in cases in, remains poor due to the severe toxic side effects, limited sensitivity and specificity of drugs (7). This is especially the case in patients at intermediate and advanced stages (7). The metastatic propensity of GC represents a formidable obstacle in the treatment paradigm (8).

Therefore, the exploration of effective strategies to inhibit GC's metastatic spread and identification of molecular markers for prognostic assessment is required.

E74-like factor 1 (ELF1), which is highly homologous to the Drosophila E74 factor (9) is a transcription factor inducible by ecdysone in Drosophila and a member of the Ets transcription factor family (9). ELF1 is located on chromosome 13q13 and consists of 619 amino acid residues, possessing the ability of both transcriptional activation and repression of target genes depending on the physiological context. Posttranslational processing determines its subcellular localization, biological activity and metabolic degradation (10). The functional regulation of ELF-1 is complex. ELF1 protein exists in a 80-kDa form in the cytoplasm and enters the nucleus in a 98 kDa form after phosphorylation and glycosylation (11). It is predominantly expressed in lymphocytes, where its posttranslational modifications bestow ELF1 with regulatory functions, enabling it to bind to gene promoters or enhancers critical for the selection, survival and maturation of diverse immune cells (12). The importance of ELF1 extends to its association with the development and metastasis of various malignancies (13), such as glioma, oral squamous cell carcinoma, endometrial carcinoma, nasopharyngeal carcinoma and prostate cancer, and colon cancer. Long non-coding RNAs (lncRNAs) are associated with cancer progression in GC (14). E2F transcription factor 1 has been shown to activate the transcription of terminal differentiation-induced non-coding RNA by binding to its promoter region, thereby promoting the proliferation of GC cells and inhibiting apoptosis (15). lncRNAs NONHSAT057282 and NONHSAG023333 can regulate genes associated with chemoresistance, such as GSTP1, BTG3, SOCS3, and BRAC2, by interacting with the transcription factors ELF1 and E2F1. ELF1 may become a new player in chemoresistance through its interaction with different lncRNA interactions involved in tumor therapy (16). So, it was hypothesized that ELF1 may be associated with GC.

Tumor cells, together with extracellular matrix (ECM), cancer-associated fibroblasts (CAF), vascular-associated smooth muscle cells, pericytes, endothelial cells, mesenchymal stem and immune cells collectively constitute the complex tumor microenvironment (TME) (17). The development, progression and ultimately the prognosis of malignancies, are profoundly influenced by the characteristics of tumor cell invasion and metastasis (18). A pivotal process in this cascade involves ECM degradation, which is tightly regulated by MMPs and tissue inhibitors of metalloproteinases (TIMPs) (19). Among MMPs, MMP-9 is of particular importance as a key protease responsible for ECM degradation, where it serves a crucial role in various types of cancers, such as breast cancer (20,21). During cancer progression, ECM homeostasis is dynamically disrupted by MMP9, enabling cancer invasion and metastasis through the ECM barrier. MMP2 and MMP9 have been previously found to be upregulated in GC tissues (22). MMP9 rs3918242 polymorphism has been associated with the risk of various cancers, including lung, prostate, breast, and colorectal cancers (23). The chromosomal location (20q12-1q13) where MMP9 is located, has been identified as one of the most common regions of genomic gain in GC (24). Therefore, the progression of GC is highly likely to be influenced by altered MMP9 expression.

However, the precise expression pattern, interaction with MMP9 and predictive value of ELF1 in GC remain elusive. Therefore, the present study aimed to explore the expression profile of ELF1 in GC, its relationship with MMP9 and its (combined with MMP9) associations with various clinicopathological parameters, survival and prognosis of patients with GC. Bioinformatics and clinical sample analyses would be performed. In addition, relevant mechanisms, such as the association of ELF1 with epithelial-mesenchymal transition (EMT), angiogenesis and immune infiltration, were explored.

Materials and methods

Clinical patient samples

Fresh GC and adjacent normal tissues (located >2 cm from the tumor margin) were randomly collected from 40 patients post-GC surgery at the Affiliated Hospital of Nantong University (Nantong, China) from November to December 2021. This cohort included 28 males and 12 females, with a mean age of 66.25 years (range, 36-92 years; Table SI). The inclusion criteria were as follows: i) Pathological diagnosis of GC; ii) clinical data and overall survival time were complete; iii) no history of antitumor treatment before surgery and iv) no combination of other organic disease and malignant tumors. The exclusion criteria were as follows: i) incomplete clinical data and overall survival time; ii) combined with other organic diseases and malignant tumors; iii) a history of preoperative antitumor treatment; and iv) use of pathological case data without the patient's informed consent. Necrotic tissues were excised and blood contaminants were rinsed with saline before the samples were frozen at -80˚C for further analysis.

Additionally, 355 paraffin-embedded GC specimens and corresponding adjacent normal tissues from the patients undergoing surgery from January 2013 to December 31, 2015, preserved in the Department of Pathology of Affiliated Hospital of Nantong University (Nantong, China, were collected from January 2019 to March 2019 for tissue microarrays for this study. The patients included 241 males and 114 females, with a mean age of 63.63 (range, 26-90) years. The inclusion and exclusion criteria were as aforementioned. The clinical characteristics collected included sex, age, histological type, differentiation, invasive depth (T), lymph node metastasis (N), distant metastasis (M), TNM stage, microvascular invasion (MVI), lymphatic invasion, perineural invasion, carcinoembryonic antigen (CEA), carbohydrate antigen 19-9 and Laurén classification (25). The cases that were processed and stained with H&E were pathologically confirmed on the basis of the latest WHO classification and 8th edition of the TNM classification recommended by the Union for International Cancer Control and American Joint Committee on Cancer (26). None of the patients had undergone any anticancer treatments, such as radiotherapy, chemotherapy or immunotherapy, before surgery. Complete clinical data and postoperative follow-up records (100% completion rate,) were obtained prior to the study. Overall survival (OS) was defined as the time from surgical resection to death or end of follow-up (December 31, 2020).

Reverse transcription-quantitative PCR (RT-qPCR) analysis for ELF1 mRNA expression in GC

The expression of ELF1 mRNA in the 40 fresh human GC tissue samples was validated through RT-qPCR. Total RNA was extracted from tissues by using a TRIzol kit (Invitrogen; Thermo Fisher Scientific, Inc.), which was then treated with DNase I (Cat No. D7073; Beyotime Institute of Biotechnology). The purity and concentration were analyzed using a NanoDrop ND-1000 spectrophotometer (Thermo Fisher Scientific, Inc.), followed by cDNA synthesis with a PowerScript Reverse Transcriptase kit (Bioland Scientific, LLC) according to the manufacturer's protocol. qPCR amplification was then performed on a 7500 real-time PCR system (Thermo Fisher Scientific, Inc.) using a SYBR® Premix ExTaq kit (Takara Bio, Inc.). The following thermocycling conditions were used: 95˚C for 5 min for initial denaturation, followed by 40 cycles of 95˚C for 10 sec (denaturation), 61˚C for 20 sec (annealing) and 70˚C for 40 sec (extension). The specific primers for ELF1 and the internal reference GAPDH were designed based on the NCBI gene sequence and synthesized by Shanghai Yingjun Biotechnology Co., Ltd. The specific forward primer for ELF1 was 5'-TGTCCAACAGAACGACCTAGT-3', whilst the reverse primer was 5'-GGCAGGAAAAATAGCTGGATCAC-3'. The length of the amplified target fragment was 88 bp. The forward primer of the GAPDH gene was 5'-GGAGCGAGATCCCTCCAAAAT-3', whereas the reverse primer was 5'-GGCTGTTGTCATACTTCTCATGG-3'. The length of the amplified target fragment was 197 bp. The results were analyzed by using the 2-ΔΔCq method (27) with each sample assayed in triplicate.

Tissue microarray (TMA)-immunohistochemistry (IHC) for protein expression levels in GC

Postoperative GC and adjacent normal tissues from the 355 patients were fixed in 10% neutral formalin at room temperature for 24 h. Core tissue biopsy samples (0.2 cm in diameter) obtained from the paraffin-embedded blocks were arranged in fresh paraffin blocks using the Quick-Ray Manual Tissue Microarrayer Full Set (cat. no. UT06; Unitma, Co., Ltd.). In total, 12 tissue microarrays comprising 710 samples were ultimately prepared. Sections of 5-µm thickness were analyzed for ELF1, MMP9, CD19, CD3, CD4, CD8 and CD56 protein expression levels by using immunohistochemistry. Sections were deparaffinized by immersion in xylene and rehydrated in gradient ethanol in separate batches, washed in PBS (0.01 M, pH=7.0), boiled (98˚C, 20 min) under pressure in citrate buffer (0.01 M, pH=6.0; antigen recovered) and incubated with 5% goat-blocking serum (cat. no. SL039; Beijing Solarbio Science & Technology Co., Ltd.) in PBS for 30 min at 37˚C to block non-specific binding. Subsequently, mouse antihuman ELF1 polyclonal antibody (dilution 1:400; cat. no. 22565-1-AP; ProteinTech Group, Inc.), MMP9 polyclonal antibody (dilution 1:300; cat. no. 10375-2-AP; ProteinTech Group, Inc.), mouse anti-human CD19 monoclonal (ready to use; cat. no. ZM-0038; ZSGB-BIO; OriGene Technologies, Inc.), mouse anti-human CD3 monoclonal (ready to use; cat. no. ZM-0417; ZSGB-BIO; OriGene Technologies, Inc.), mouse antihuman CD4 monoclonal (ready to use; cat. no. ZM-0418; ZSGB-BIO; OriGene Technologies, Inc.), rabbit anti-human CD8 monoclonal (ready to use; cat. no. ZM-0508 ZSGB-BIO; OriGene Technologies, Inc.) and mouse anti-human CD56 monoclonal (ready to use; cat. no. ZM-0057; ZSGB-BIO; OriGene Technologies, Inc.) were used for staining overnight at 4˚C. HRP-labeled goat anti-mouse secondary antibody (1:1,000; cat. no. ab6728; Abcam) and HRP-labeled goat anti-rabbit IgG (1:1,000; cat. no. Ab6721; Abcam) were used at room temperature for 30 min. Sections were incubated with DAB (cat. no. DA1010; Beijing Solarbio Science & Technology Co., Ltd.) for ~10 min, counterstained with hematoxylin (room temperature, 20-30 sec) and sealed with gelatin glycerol. PBS served as a negative control. Staining results were double-blinded and analyzed by two senior pathologists (XYR and SZ).

Cells were light imaged through an optical microscope (BX51, OLYMPUS) of immunohistochemistry staining were defined as brownish-yellow or brownish-brown nuclei (ELF1) and cytoplasm (MMP9) staining of GC tumor cells, nuclei (ELF1) of the lymphocytes in mesenchyme, and membrane (CD19, CD3, CD4, CD8 and CD56) of infiltrated immune cells. The intensity was scored as follows: i) 0, negative; ii) 1, weak intensity; iii) 2, moderate intensity; and iv) 3, strong intensity. Percentage of positive cells was scored as follows: i) 0, negative; ii) 1, 1-25% positive; iii) 2, 26-50% positive; iv) 3, 51-75% positive; and v) 4, 76-100% positive). The multiplication of the two aforementioned scores was used as the final score, where 0-6 would be deemed no or low expression (-) and 7-12 was considered high expression (+) (28).

Bioinformatics analysis

Data on 375 primary GC tissues and 32 adjacent normal tissues were sourced from The Cancer Genome Atlas (TCGA) database (portal.gdc.cancer.gov/), where ‘STAD’ was searched to download RNA-Seq expression and clinical data for GC patients. Cases with gene expression of ‘0’, lost-visit cases, and cases with incomplete clinical information were excluded by R language (version 4.2.1). Gene Expression Profiling Interactive Analysis (GEPIA; http://gepia.cancer-pku.cn/) was used to compare ELF1 mRNA expression levels in GC and normal tissues. GEPIA is an interactive web server that can analyze RNA sequencing expression data from TCGA and GTEx for tumor and normal samples and can be used for numerous analyses, such as differential analysis, characterization based on cancer type or pathological stage, survival analysis, correlation analysis and downscaling analysis. The specific settings in ‘Expression DIY’ (Boxplot) were as follows: i) Gene symbol, ELF1; ii) Datasets Selection (Cancer name), stomach adenocarcinoma (STAD); iii) |Log2fold change (FC)| Cutoff, 1; iv) P-value cut-off, 0.01; and v) Matched Normal data, Match TCGA normal and GTEx data. The Strawberry Perl software (version 5.38.2, perl.org/get.html) and R language (version 4.2.1) with Limma package (Log2FC filter >1; adjusted P-value filter=0.05; Wilcox test) were used to obtain the ELF1 mRNA expression differentiation. Strawberry Perl software was used to convert probe names from transcriptome files to gene names, and ‘Limma’ was used for analyzing chip data.

GEPIA was also utilized for various correlation analyses, including the analysis of correlations between ELF1 and expressions of GC molecular typing-related genes, including MutL homolog 1 (MLH1), erythroblastic leukemia viral oncogene homolog 2 (ERBB2), PI3K subunit α (PIK3CA) and p53(29). EMT-related molecules, including MMP9, Cadherin 1 (CDH1) and TIMP1; and angiogenic indices, including vascular endothelial growth factor A (VEGFA) and kinase insert domain receptor (KDR). ‘Correlation Analysis’ was chosen on the database. Then ELF1 was designated as ‘Gene A’ for the x-axis, whilst other genes to be studied served as ‘Gene B’ for the y-axis. ‘Pearson’ was selected as ‘Correlation Coefficient’. STAD tumor was selected from ‘TCGA Tumor’ dialog box to ‘Used Expression Datasets’.

Tumor Immune Estimation Resource 2.0 (TIMER) (https://cistrome.shinyapps.io/timer/), a web server for the comprehensive analysis of infiltrated immune cells, is a resource for the systematic analysis of immune infiltrates among the different cancer types. The abundance of six infiltrated immune cell types (B cells, CD4+ T cells, CD8+ T cells, neutrophils, macrophages and dendritic cells) can be estimated using the TIMER algorithm (30). Users can conveniently access tumor immunological, clinical and genomic features by inputting function-specific parameters. ‘Gene’ module was chosen to visualize the scatter plots visualizing the correlation between ELF1 expression and different levels of infiltration by immune cell types in GC. ‘Spearman’ was selected as the ‘Correlation Coefficient’.

Cell-type Identification By Estimating Relative Subsets Of RNA Transcript (CIBERSORT; version 2023.1.2, cibersort.stanford.edu/), a software for the deconvolution of transcriptome expression matrices to estimate the composition and abundance of immune components in mixed cells on the basis of linear support vector regression principles, was further applied to study the correlation of 22 immune cell scores with ELF1 expression levels (31). CIBERSORT was used to analyze immune cell content of each sample, those with a CIBERSORT output of P<0.05 were considered accurate and enrolled for further construction of the immune landscape, otherwise, samples were eliminated. Subsequently the correlation of ELF1 with these immune cells was analyzed.

Statistical analysis

Epidata 3.1 software (epidata.dk; Epidata Association from Denmark, was utilized to manage clinical and laboratory data. SPSS 21.0 (IBM Corp.) and GraphPad 5.0 software (Dotmatics) were used for data analysis. Wilcoxon signed-rank non-parametric test was used to analyze the experimental data from RT-qPCR, as it was non-normally distributed. MedCalc software (version 18.2.1, MedCalc Software Ltd.) was used for receiver operating characteristic (ROC) curve analysis to estimate the diagnostic value of ELF1. Categorical variables in IHC were compared by the χ2 or Fisher exact tests, as appropriate. Survival curves were plotted by using the Kaplan-Meier method and tested through the log-rank test. Cox regression provided the hazard ratio (HR) and 95% CI in all prognostic analyses. P<0.05 was considered to indicate a statistically significant difference, association or correlation.

Results

Expression level of ELF1 mRNA in GC tissues

Examination in GEPIA revealed increased ELF1 mRNA expression in GC compared with normal gastric tissue (P<0.05; Fig. 1A). This trend was supported by results reported by the R4.2.1 software based on data from TCGA database (Fig. 1B and C). RT-qPCR assay was next performed to measure ELF1 mRNA expression in GC tissues, with an amplification efficiency of 99.33%. ELF1 mRNA expression in GC tissues was found to be significantly higher compared with that in adjacent normal tissues (P<0.05; Fig. 1D). Increased ELF1 mRNA expression was also revealed to be a reliable indicator for distinguishing GC tissues from normal adjacent tissues, as evidenced by an area under the curve of 0.8078 and a 95% CI of 0.7015-0.9141. The sensitivity and specificity of this differentiation were 87.5 and 77.5%, respectively (P<0.05; Fig. 1E).

Expression levels of ELF1 and MMP9 proteins in GC tissues

TMA-IHC revealed that the incidence of ELF1 (mainly localized to the nucleus) and MMP9 (mainly localized to the cytoplasm) positivity were statistically significantly higher in GC tissues compared with that in adjacent normal tissues (both P<0.001; Fig. 2 and Table I). However, no significant difference in the incidence of positive ELF1 expression in lymphocytes in the mesenchyme of GC tissues and adjacent normal tissues could be found (Fig. 2; Table I).

Table I

Expression of ELF1 and MMP9 by immunohistochemistry in GC and adjacent normal tissues (N=355).

Table I

Expression of ELF1 and MMP9 by immunohistochemistry in GC and adjacent normal tissues (N=355).

CharacteristicNegativePositiveP-value
GC103 (29.0)252 (71.0)<0.001
Adjacent normal245 (69.0)110 (31.0) 
CharacteristicNegativePositiveP-value
GC129 (36.3)226 (63.7)<0.001
Adjacent normal257 (72.4)98 (27.6) 
CharacteristicNegativePositiveP-value
GC241 (67.9)114 (32.1)0.099
Adjacent normal140 (62.0)215 (38.0) 

[i] ELF1, E74-like factor 1; GC, gastric cancer.

Correlations of ELF1 and MMP9 protein expression levels, their associations and clinical characteristics in GC

TMA-IHC revealed a positive association between the incidence of ELF1 and MMP9 positivity in GC tissues (P<0.001; Table II). ELF1 and MMP9 protein expression levels were also found to associate with T (both P<0.001), N (both P<0.001), M (ELF1, P=0.047; MMP9, P=0.001), TNM staging (both P<0.001), MVI (ELF1, P=0.040; MMP9, P=0.008), lymphatic invasion (ELF1, P=0.015; MMP9, P=0.017) and blood serum CEA levels (ELF1, P=0.002; MMP9, P<0.001; Table III). However, ELF1 expression in lymphocytes in the GC mesenchyme was not associated found to be with any of the clinicopathological parameters of patients with GC (Table III).

Table II

Association between ELF1 and MMP9 protein expression positivity in tissue microarray according to immunohistochemistry in patients with gastric cancer.

Table II

Association between ELF1 and MMP9 protein expression positivity in tissue microarray according to immunohistochemistry in patients with gastric cancer.

 MMP9 
ELF1-+P-value
-9013<0.001
+39213 

[i] ELF1, E74-like factor 1.

Table III

Association of ELF1 and MMP9 expression by tissue microarray-immunohistochemistry with clinical characteristics in patients with GC.

Table III

Association of ELF1 and MMP9 expression by tissue microarray-immunohistochemistry with clinical characteristics in patients with GC.

A, ELF1 expression
CharacteristicNNegativePositiveP-value
Total355103 (29.0)252 (71.0) 
Sex   0.204
     Male24175 (31.1)166 (68.9) 
     Female11428 (24.6)86 (75.4) 
Age   0.680
     ≤6010930 (27.5)79 (72.5) 
     >6024673 (29.7)173 (70.3) 
Histological type   0.212
     Tubular24671 (28.9)175 (71.1) 
     Papillary72 (28.6)5 (71.4) 
     Mucinous253 (12.0)22 (88.0) 
     Mixed (tubular and mucinous)51 (20.0)4 (80.0) 
     Signet ring cell7226 (36.1)46 (63.9) 
Differentiation   0.158
     Well5514 (25.5)41 (74.5) 
     Middle16650 (30.1)116 (69.9) 
     Poor10435 (33.7)69 (66.3) 
     Others304 (13.3)26 (86.7) 
T   <0.001
     Tis65 (83.3)31 (16.7) 
     13119 (61.3)12 (38.7) 
     26023 (38.3)37 (61.7) 
     323855 (23.1)183 (76.9) 
     4201 (5.0)19 (95.0) 
N   <0.001
     014558 (40.0)87 (60.0) 
     15516 (29.1)39 (70.9) 
     28419 (22.6)65 (77.4) 
     37110 (14.1)61 (85.9) 
M   0.047
     0339102 (30.1)237 (69.9) 
     1161 (6.3)15 (93.8) 
TNM stage   <0.001
     0+14222 (52.4)20 (47.6) 
     211552 (45.2)63 (54.8) 
     318228 (15.4)154 (84.6) 
     4161 (6.3)15 (93.7) 
MVI   0.040
     No20167 (33.3)134 (66.7) 
     Yes15436 (23.4)118 (76.6) 
Lymphatic invasion   0.015
     No26987 (32.3)182 (67.7) 
     Yes8616 (18.6)70 (81.4) 
Perineural invasion   0.628
     No31891 (28.6)277 (71.4) 
     Yes3712 (32.4)25 (67.6) 
CEA (ng/ml)   0.002
     ≤512248 (39.3)74 (60.7) 
     >521952 (23.7)167 (76.3) 
     Unknown143 (21.4)11 (78.6) 
CA 19-9 (U/ml)   0.232
     ≤3719161 (31.9)130 (68.1) 
     >3715039 (26.0)111 (74.0) 
     Unknown143 (21.4)11 (78.6) 
Laurén classification   0.704
     Intestinal type26074 (28.5)186 (71.5) 
     Diffuse type9529 (30.5)66 (69.5) 
B, MMP9 expression
CharacteristicNNegative, N (%)Positive, N (%)P-value
Total355129 (36.3)226 (63.7) 
Sex   0.418
     Male24191 (37.8)150 (62.2) 
     Female11438 (33.3)76 (66.7) 
Age   0.884
     ≤6010939 (35.8)70 (64.2) 
     >6024690 (36.6)156 (63.4) 
Histological type   0.097
     Tubular24685 (34.6)161 (65.4) 
     Papillary74 (57.1)3 (42.9) 
     Mucinous255 (20.0)20 (80.0) 
     Mixed (tubular and mucinous)52 (40.0)3 (60.0) 
     Signet ring cell7233 (45.8)39 (54.2) 
Differentiation   0.200
     Well5520 (36.4)35 (63.6) 
     Middle16657 (34.3)109 (65.7) 
     Poor10445 (43.3)59 (56.7) 
     Others307 (23.3)23 (76.7) 
T   <0.001
     Tis65 (83.3)1 (16.7) 
     13123 (74.2)8 (25.8) 
     26033 (55.0)27 (45.0) 
     323866 (27.7)172 (72.3) 
     4202 (10.0)18 (90.0) 
N   <0.001
     014573 (50.3)72 (49.7) 
     15519 (34.5)36 (65.5) 
     28422 (26.2)62 (73.8) 
     37115 (21.1)56 (78.9) 
M   0.001
     0339129 (38.1)210 (61.9) 
     1160 (0.0)16 (100.0) 
TNM stage   <0.001
     0+14227 (64.3)15 (35.7) 
     211561 (53.0)54 (47.0) 
     318241 (22.5)141 (77.5) 
     4160 (0.0)16 (100.0) 
MVI   0.008
     No20185 (42.3)116 (57.7) 
     Yes15444 (28.6)110 (71.4) 
Lymphatic invasion   0.017
     No269107 (39.8)162 (60.2) 
     Yes8622 (25.6)64 (74.4) 
Perineural invasion   0.108
     No318120 (37.7)198 (62.3) 
     Yes379 (24.3)28 (75.7) 
CEA (ng/ml)   <0.001
     ≤512260 (49.2)62 (50.8) 
     >521964 (29.2)155 (70.8) 
     Unknown145 (35.7)9 (64.3) 
CA 19-9 (U/ml)   0.017
     ≤3719180 (41.9)111 (58.1) 
     >3715044 (29.3)106 (70.7) 
     Unknown145 (35.7)9 (64.3) 
Laurén classification   0.386
     Intestinal type26091 (35.0)169 (65.0) 
     Diffuse type9538 (40.0)57 (60.0) 
C, ELF1 expression in lymphocytes
CharacteristicNNegativePositiveP-value
Total355241 (67.9)114 (32.1) 
Sex   0.525
     Male241161 (66.8)80 (33.2) 
     Female11480 (70.2)34 (29.8) 
Age   0.459
     ≤6010977 (70.6)32 (29.4) 
     >60246164 (66.7)82 (33.3) 
Histological type   718
     Tubular246165 (67.1)81 (32.9) 
     Papillary75 (71.4)2 (28.6) 
     Mucinous2520 (80.0)5 (20.0) 
     Mixed (tubular and mucinous)54 (80.0)1 (20.0) 
     Signet ring cell7247 (65.3)25 (34.7) 
Differentiation   0.305
     Well5540 (72.7)15 (27.3) 
     Middle166111 (66.9)55 (33.1) 
     Poor10466 (63.5)38 (36.5) 
     Others3024 (80.0)6 (20.0) 
T   0.988
     Tis64 (66.7)2 (33.3) 
     13122 (71.0)9 (29.0) 
     26040 (66.7)20 (33.3) 
     3238162 (68.1)76 (31.9) 
     42013 (65.0)7 (35.0) 
N   0.699
     0145102 (70.3)43 (29.7) 
     15534 (61.8)21 (38.2) 
     28456 (66.7)28 (33.3) 
     37149 (69.0)22 (31.0) 
M   0.104
     0339227 (67.0)112 (33.0) 
     11614 (87.5)2 (12.5) 
TNM stage   0.296
     0+14229 (69.0)13 (31.0) 
     211580 (69.6)35 (30.4) 
     3182118 (64.8)64 (35.2) 
     41614 (87.5)2 (12.5) 
MVI   0.723
     No201138 (68.7)63 (31.3) 
     Yes154103 (66.9)51 (33.1) 
Lymphatic invasion   0.487
     No269180 (66.9)89 (33.1) 
     Yes8661 (70.9)25 (29.1) 
Perineural invasion   0.743
     No318215 (67.6)103 (32.4) 
     Yes3726 (70.3)11 (29.7) 
CEA (ng/ml)   0.756
     ≤512281 (66.4)41 (33.6) 
     >5219149 (68.0)70 (32.0) 
     Unknown1411 (78.6)3 (21.4) 
CA 19-9 (U/ml)   0.175
     ≤37191123 (64.4)68 (35.6) 
     >37150107 (71.3)43 (28.7) 
     Unknown1411 (78.6)3 (21.4) 
Laurén classification   0.896
     Intestinal type260176 (67.7)84 (32.3) 
     Diffuse type9565 (68.4)30 (31.6) 

[i] ELF1, E74-like factor 1; Tis, tumor in situ; MVI, microvascular invasion; CEA, carcinoembryonic antigen; CA19-9, carbohydrate antigen 19-9.

Association of ELF1 and MMP9 expression with the prognosis of patients with GC

Kaplan-Meier survival analysis revealed that patients with GC and elevated levels of ELF1 and MMP9 protein expression both faced significantly inferior overall survival (OS) compared with that in patients with lower expression levels of ELF1 and MMP9 proteins (P<0.001 for both; Fig. 3A and B). Specifically, OS stood at 17.1 and 11.5% in patients with high ELF1 and MMP9 expression levels, respectively. This is in contrast to that in patients with lower expression levels of ELF1 and MMP9 proteins, where the OS was observed to be 72.8% for ELF1 and 71.3% for MMP9 (Fig. 3A and B). The lowest OS, which was 10.8%, was observed in patients with simultaneously high expression levels of ELF1 and MMP9 (Fig. 3C). In addition, OS decreased with increasing TNM stage (P<0.001; Fig. 3D). Further prognostic analysis using the Cox regression model showed that high ELF1 expression (HR, 2.555; 95% CI, 1.546-4.224; P=0.002), high MMP9 expression (HR, 3.813; 95% CI, 2.406-6.041; P<0.001), advanced TNM stage (P=0.001) and advanced N stage (P=0.011) were independent prognostic factors for patients with GC (Table IV). However, there was no significant association between ELF1 expression in lymphocytes in the GC mesenchyme and the prognosis of patients with GC (Table IV).

Table IV

Cox regression analysis of prognostic factors for 5-year survival from gastric cancer.

Table IV

Cox regression analysis of prognostic factors for 5-year survival from gastric cancer.

 Univariate analysisMultivariate analysis
ParameterHR (95% CI)P-valueHR (95% CI)P-value
ELF1 expression, high vs. low + none5.519 (3.707-8.217)<0.0012.555 (1.546-4.224)0.002
MMP9 expression, high vs. low + none6.125 (4.291-8.744)<0.0013.813 (2.406-6.041)<0.001
Age, ≤60 vs. >60 years0.868 (0.662-1.138)0.306  
Sex, male vs. female0.978 (0.745-1.283)0.870  
Histological type 0.043 0.803
     Tubular vs. papillary0.533 (0.170-1.669)0.2801.030 (0.315-3.365)0.961
     Tubular vs. mucinous1.365 (0.849-2.194)0.1991.108 (0.672-1.828)0.687
     Tubular vs. mixed (tubular + mucinous)2.023 (0.829-4.936)0.1210.900 (0.322-2.517)0.841
     Tubular vs. signet ring cells0.716 (0.509-1.007)0.0550.811 (0.561-1.172)0.266
Differentiation 0.064  
     Well vs. middle1.075 (0.744-1.553)0.699  
     Well vs. poor0.828 (0.553-1.241)0.361  
     Well vs. others1.557 (0.934-2.596)0.090  
TNM stage <0.001 0.001
     0+1 vs. 22.992 (1.579-5.667)0.0014.440 (1.966-10.029)<0.001
     0+1 vs. 35.380 (2.908-9.956)<0.0014.983 (2.022-12.276<0.001
     0+1 vs. 412.030 (5.494-26.345)<0.0018.796 (3.031-25.521)<0.001
T trend <0.001 0.179
     Tis vs. 10.550 (0.146-2.072)0.3770.290 (0.070-1.197)0.087
     Tis vs. 21.406 (0.431-4.585)0.5720.365 (0.102-1.302)0.120
     Tis vs. 32.403 (0.767-7.525)0.1320.255 (0.069-0.935)0.039
     Tis vs. 43.243 (0.944-11.143)0.0620.219 (0.053-0.914)0.037
N trend <0.001 0.011
     0 vs. 12.200 (1.512-3.201)<0.0011.452 (0.958-2.203)0.079
     0 vs. 22.750 (1.968-3.842)<0.0011.909 (1.275-2.859)0.002
     0 vs. 32.236 (1.562-3.200)<0.0011.273 (0.821-1.971)0.280
M, 0 vs. 13.159 (1.866-5.348)<0.0010.919 (0.481-1.757)0.799
MVI, no vs. yes1.346 (1.043-1.737)0.0230.946 (0.718-1.246)0.692
Lymphatic invasion, no vs. yes1.275 (0.953-1.707)0.102  
Perineural invasion, no vs. yes1.316 (0.875-1.978)0.187  
Carcinoembryonic antigen level, ≤5 vs. >5 ng/ml1.645 (1.234-2.193)0.0010.690 (0.460-1.037)0.074
Carbohydrate antigen 19-9 level, ≤37 vs. >37 U/ml1.268 (0.977-1.646)0.074  
Laurén classification, intestinal type vs. diffuse type0.836 (0.622-1.125)0.238  
ELF1 expression in lymphocytes, high vs. low and none1.015 (0.772-1.334)0.918  

[i] HR, hazard ratio; ELF1, E74-like factor 1; Tis, Tumor in situ; MVI, microvascular invasion.

Gene correlation analysis

Investigations using the GEPIA database revealed notable positive associations between ELF1 expression and several crucial genes involved in GC occurrence (32). ELF1 expression showed a mild positive association with MLH1 (r=0.35; P<0.001), PIK3CA (r=0.4; P<0.001) and CDH1 (r=0.39; P<0.001; Fig. 4A-C), but not with ERBB2 (r=0.16), TP53 (r=0.1), MMP9 (r=0.12), TIMP1 (r=-0.13), VEGFA (r=0.19) and KDR (r=0.16; Fig. 4D-I). These findings suggest an association between ELF1 expression and the GC molecular subtypes, implying roles in various processes, such as EMT and angiogenesis.

Association between ELF1 expression and immune cell infiltration into GC

Infiltrating immune cell types into tumor tissues mainly include B-cells (CD19+), T-cells (CD3+, CD4+ and CD8+) and natural killer cells (NKs, CD56+) (33,34). The association between ELF1 expression and the infiltrating immune cells in GC was examined. TIMER database was used as an initial analysis to assess the correlation between the degree of infiltration by various immune cell types and ELF1 expression levels. This examination revealed no correlations between ELF1 expression and the levels of infiltration by B cells (r=0.187), CD8+ T cells (r=-0.017), CD4+ T cells (r=0.169), Macrophage (r=0.058), neutrophil (r=0.039) or dendritic cells (r=0.067; Fig. 5A). Additionally, by using the CIBERSORT software, the relationship between ELF1 expression and infiltration by 24 subsets of different immune cell types integral to tumor immunity was evaluated. The outcomes revealed marked variations in several immune cell types across groups with differing ELF1 expression levels. In particular, ELF1 had a positive association with naive B cells (P=0.028), memory-activated CD4+ T cells (P=0.009), γδ T cells (P=0.029) and activated NK cells (P=0.001), but no relationship with others. (Fig. 5B).

IHC was next used to further validate the relationship between ELF1 expression and infiltrating immune cells in GC. The results revealed that ELF1 had associations with CD19 (B-cells; P<0.001) and CD4 (CD4+ T cells; P=0.002), but not with CD3, CD8 or CD56 (Table V; Fig. 6).

Table V

Association between ELF1 expression by tissue microarray-immunohistochemistry and that of different immune cell markers in GC.

Table V

Association between ELF1 expression by tissue microarray-immunohistochemistry and that of different immune cell markers in GC.

 CD19CD3CD4CD8CD56
ELF1-+-+-+-+-+
-93107033693462417132
+113139167851241281609216983
P-value<0.001 0.759 0.002 0.560 0.733 

[i] ELF1, E74-like factor 1.

Discussion

GC is a common malignant tumor worldwide and is characterized by high incidence and mortality rates, with multifactorial associations encompassing dietary habits (including high-fat intake, elevated salt consumption, and smoking), infections (such as Helicobacter pylori and Epstein-Barr virus infections) and genetic predispositions (35,36). Familial clustering is discernible in GC development, with ~25% of autosomal dominant diffuse GC-prone families harboring E-cadherin mutations (37). The aggressive and metastatic attributes of GC are closely associated with the poor patient prognosis, with a median survival of <12 months (38). Although significant improvements have been made in identifying biomarkers and molecular targeted agents (trastuzumab, Bevacizumab, Panitumumab, Everolimus, etc.) for improving patient prognosis, such as epidermal growth factor receptor (EGFR), vascular endothelial growth factor (VEGF) (39), mTOR and PI3K (40), predictive biomarkers and corresponding targeted agents specifically addressing GC invasion and metastasis remain scarce.

Ets genes, known for their erythroblast transformation specificity, constitute a class of highly conserved oncogenes that serve important roles in regulating tumor infiltration and metastasis (41). Among the genes in the Ets transcription factor family, ELF1 is one of the most prominent members (42). ELF1 is a T cell-specific transcription factor that was originally cloned from a human T-cell library through hybridization to a probe encoding the DNA-binding structural domain (Ets structural domain) of human Ets-1 cDNA. This transcription factor mainly exerts its influence by binding to gene promoters or enhancers (42) and can function to either activate and repress the expression of target genes. In addition, it is unique in being subject to glycosylation and phosphorylation (10). Elevated levels of ELF1 expression have been previously associated with the initiation and progression of various malignancies, such as pancreatic and colon cancer (13,43). The high expression of ELF1 has been associated with poor prognosis in patients with endometrial (44) and ovarian cancer (45). ELF1 can also promote the malignant progression of glioma (46). In addition, ELF1 has been found to promote the proliferation of oral squamous cell carcinoma cells by increasing the expression of β-catenin mRNA (47). By contrast, ELF1 can activate the doublecortin-like kinase 1/Janus kinase/STAT signaling pathway, thereby promoting the malignant progression of pancreatic cancer (13). Another recent study has elucidated ELF1 effect on the biological behavior of colon cancer, which was by modulating the transcriptional activation of serine peptidase inhibitor kazal type 4, thereby promoting colon cancer progression (43). However, conflicting reports exist that also suggest ELF1 to serve an inhibitory role in certain cancers, such as prostate cancer (48) and Hodgkin's lymphoma (49).

The extracellular matrix (ECM) is divided into basement membrane and interstitial matrix, and is composed of various proteins such as type IV collagen, laminin, elastin, and hyaluronic acid, which provide structural support for cells and are also involved in the development of epithelial cells. The composition and stability of the ECM is closely related to the protein-protein and polysaccharide-protein binding in the extracellular matrix, which is one of the main barriers to prevent tumor metastasis. Degradation of ECM facilitates tumor cell invasion and metastasis and is a key trigger of EMT, where MMPs serve a key role because they can degrade almost all ECM components (50). The MMP family includes 26 different members, which can then be divided into various subtypes, including collagenases, gelatinases, matrix proteases and membrane-type MMPs (22). Previous studies have shown that MMP9 (a member of the gelatinase family), which can degrade the ECM and basement membranes, in addition to being associated with cancer cell adhesion and migration, is an important marker for predicting poor tumor prognosis in colorectal cancer (51) and endometrial carcinoma (44). In addition, MMP9 has been reported to positively correlate with the infiltration of a diverse range of immune cell types, including Th1 cells, neutrophils and macrophages, and regulates their transport in uveal melanoma and clear Cell Renal Cell Carcinoma, which is essential for establishing and coordinating the tumor immune environment (52,53). The influence of MMP9 has been documented to extend to angiogenic and lymphangiogenic factors, such as VEGF, TGF-β, tumor necrosis factor-α, ΙL-8 and EGFR (54). Elevated levels of MMP9 mRNA and protein expression have been frequently found in various cancer types, including ovarian (55), colorectal (22), breast (56) and lung cancers (57). High MMP9 expression is also consistently associated with advanced tumor stages and adverse clinical outcomes, underscoring its potential as a prognostic indicator for patients with cancer of uveal melanoma and breast cancer (54,56).

Ets family proteins enhance the expression of the urokinase-type plasminogen activator (uPA) gene by binding to its inducible enhancer region. uPA in turn activates a variety of MMPs (MMP1, MMP2 and MMP9) whilst promoting protein hydrolysis in the ECM (44). ELF1 serves a pivotal role in tumor angiogenesis, ECM remodeling and metastasis, by regulating various stages of neovascularization (which encompasses the generation of proangiogenic factors, MMPs and protease inhibitors) (58). In addition, ELF1 overexpression has been documented to contribute to a critical facet of tumor infiltration (pancreatic and colon cancer), namely ECM penetration. This was mediated through the transcriptional control of genes encoding enzymes involved in ECM degradation, such as MMP9(59). The aforementioned findings suggest that ELF1 may regulate tumorigenesis and progression through the TME, which is associated with EMT, angiogenesis and immune infiltration. The present study revealed the expression profiles of ELF1 in GC, the possible mechanistic interplay between ELF1 and MMP9, in addition to their collective effect on patient survival and prognosis.

Initial analysis using the GEPIA database and R 4.2.1 software indicated elevated ELF1 expression in GC. Subsequently, RT-qPCR analysis corroborated these findings, suggesting the diagnostic value of ELF1 expression in distinguishing GC from adjacent normal tissues. Therefore, it became possible to initially distinguish GC tissues from normal tissues based on ELF1 expression. Recent in vitro experiments showed that ELF1 overexpression directly activated MMP9 promoter activity, which identified an ELF1-triggered transcriptional mechanism by which neurotrauma upregulated MMP9 expression in the dorsal root ganglion (59). ELF1 has also been shown to activate forkhead box D3-antisense 1 to promote the migration, invasion and EMT of osteosarcoma cells by sponging microRNA-296-5p, preventing the inhibition of zinc finger CCHC-type containing 3(60). Transcription factor ELF1 activates MEIS1 transcription and promotes MEIS1 expression. Overexpression of MEIS1 increases growth factor independent protein 1) expression by activating the GFI1 enhancer, but decreases FBW7 expression and thus promoting glioma cell proliferation (decreased PCNA), migration and invasive ability (decreased MMP-9), and reducing apoptosis (increased capase-3) promotes glioma development (46). ELF1 is involved in prostate cancer tumor cell migration and EMT by interfering with the oncogenic ETS function of ETS/Activator protein-1 cis-regulatory motifs (48). To elucidate the association between ELF1 and EMT in GC, TMA-IHC was performed. The results revealed increased levels of ELF1 and MMP9 proteins in GC tissues compared with adjacent normal tissues, where both exhibited positive associations with key clinical parameters, including T, N and M stages, TNM staging, MVI, lymphatic invasion and blood CEA levels. Survival and prognostic analyses also revealed that patients with high expression levels of ELF1 and MMP9 proteins and advanced TNM staging had shorter survival and poorer prognoses. The findings of IHC revealed a positive correlation between the expression levels of ELF1 and MMP9. These findings suggest that high ELF1 expression may serve an important role in the pathogenesis and malignant progression of GC, which can be exploited to predict the survival and prognosis of patients with GC. However, the specific mechanistic interactions between ELF1 and MMP9 remain unclear, which require further study.

Based on heterogeneity of gastric cancer and the rapid development of molecular biology, TCGA proposed a molecular classification system for gastric cancer by analyzing data from multiple platforms, consisting of four distinct subtypes, including Epstein-Barr virus+ (accompanied by PIK3CA and ERBB2 amplification), microsatellite instability (usually accompanied by MLH1 silence, high-frequency mutation of ERBB2 and PIK3CA), genomically stable (accompanied by CDH1 gene mutation) and chromosome instability (accompanied by high mutation rates of TP53 and ERBB2 amplification (30). TIMP1, an endogenous inhibitor of MMP-9, executes a role not only in impeding the matrix degradation activity of MMP-9 whilst also activating pivotal cytokines, including VEGF, TGF-α,TGF-β (transforming growth factor-β) and CAM (cell adhesion molecule), binding cell surface protein CD63, activating FAK-PI3K/AKT (focal adhesion kinase-phosphatidylinositol 3 kinase/protein kinase B) signaling pathway and MAPK) signaling pathway, leading to tumor cell proliferation (61). VEGFA and KDR are involved in tumor angiogenesis in addition to molecular subtype identification of GC (62,63). The CDH1 gene encodes the vital cell adhesion molecule E-cadherin, which is crucial for maintaining epithelial tissue integrity. E-CAD reduction is associated with EMT), generation of stem-cell, and metastasis (64,65). Analysis of the GEPIA database in the present study revealed associations of ELF1 with MLH1, PIK3CA and CDH1. ELF1 has been previously reported to serve an indispensable role in the survival, differentiation and maturation of T and B lymphocyte cells, especially NK/T cells, where the absence of which results in T cell apoptosis (66). Findings from the TIMER database, CIBERSORT and IHC in the present study unveiled associations between ELF1 expression and B cells and CD4+ T cells. CD4+ and CD8+ T cells contribute to tumor growth and became effective targets for cancer survival, prognosis and treatment in lung cancer (67). B cells can produce cytokines (e.g., IL-10) that inhibit the antitumor response of T cells (inhibit CTL-mediated tumor clearance) and promote the production of immune complexes (generated by antitumor antibodies), inducing tumorigenesis. Breg (regulatory B) cells exhibit pro-tumorigenic activity due to a lack of response to CTLA4, resulting in a shortened overall survival in cutaneous melanoma, and produce adenosine, which is involved in the inhibition of T cell activation and/or inactivation influencing prognosis (such as urothelial bladder and gastric cancer) (68,69). CD4+ T cells can produce tumor cytokines (such as IL-4), associate with other cell types (myeloid-derived suppressor cells and tumor-associated macrophages) and transform into regulatory T cells, which ultimately exert pro-tumorigenic functions (69,70). Based on the aforementioned results, we hypothesized that ELF1 appeared to be associated with GC development, which are intricately intertwined with the TME including EMT, angiogenesis and immune infiltration.

However, the present study has several limitations. All the specimens used for this study were selected randomly, and the size and quality of specimens could not be controlled. The detection of protein expression using IHC may be affected by tumor heterogeneity and subjective scoring system analysis. The selected tumor location may not accurately represent the entire tumor due to intratumor heterogeneity. In addition, the r-value of the correlation analysis based on data from the GEPIA database was low, which require confirmation through further detailed exploration in subsequent studies. The majority of the studies on the involvement of ELF1 in EMT and angiogenesis in GC were based on bioinformatics, where the exact mechanism and related signaling pathways requires further experimental verification. However, the results may provide guidance for future prospective clinical trials. Any future studies should conduct comprehensive and in-depth studies at the cytological and molecular levels to provide a solid theoretical foundation for the study of GC molecular targets.

Supplementary Material

Clinical characteristics for the 40 patients with gastric cancer.

Acknowledgements

Not applicable.

Funding

Funding: The present study was supported by the Nantong Science and Technology Program (grant no. JCZ19093).

Availability of data and materials

The data generated in the present study may be requested from the corresponding author.

Authors' contributions

YW designed the study. Data analysis was performed by XZ and XR. XZ drafted the article. XZ performed the experiments. Critical revision of article was done by SZ and YW. All authors approved the final version of the article. YW and XZ confirm the authenticity of all the raw data.

Ethics approval and consent to participate

The present study complies with the ethical standards of the Declaration of Helsinki (as revised in 2013) and was approved by the Ethics Committee of Affiliated Hospital of Nantong University (approval no. 2018-L042). Written informed consent was obtained by each patient or his or her family prior to this study.

Patient consent for publication

Not applicable.

Competing interests

The authors declare that they have no competing interests.

References

1 

Bray F, Laversanne M, Sung H, Ferlay J, Siegel RL, Soerjomataram I and Jemal A: Global cancer statistics 2022: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 74:229–263. 2024.PubMed/NCBI View Article : Google Scholar

2 

Lei ZN, Teng QX, Tian Q, Chen W, Xie Y, Wu K, Zeng Q, Zeng L, Pan Y, Chen ZS and He Y: Signaling pathways and therapeutic interventions in gastric cancer. Signal Transduct Target Ther. 7(358)2022.PubMed/NCBI View Article : Google Scholar

3 

Yeoh KG and Tan P: Mapping the genomic diaspora of gastric cancer. Nat Rev Cancer. 22:71–84. 2022.PubMed/NCBI View Article : Google Scholar

4 

Sexton RE, Al Hallak MN, Diab M and Azmi AS: Gastric cancer: A comprehensive review of current and future treatment strategies. Cancer Metastasis Rev. 39:1179–1203. 2020.PubMed/NCBI View Article : Google Scholar

5 

Sung H, Ferlay J, Siegel RL, Laversanne M, Soerjomataram I, Jemal A and Bray F: Global cancer statistics 2020: GLOBOCAN estimates of incidence and mortality worldwide for 36 cancers in 185 countries. CA Cancer J Clin. 71:209–249. 2021.PubMed/NCBI View Article : Google Scholar

6 

Cai H, Li M, Deng R, Wang M and Shi Y: Advances in molecular biomarkers research and clinical application progress for gastric cancer immunotherapy. Biomark Res. 10(67)2022.PubMed/NCBI View Article : Google Scholar

7 

Zang YS, Dai C, Xu X, Cai X, Wang G, Wei J, Wu A, Sun W, Jiao S and Xu Q: Comprehensive analysis of potential immunotherapy genomic biomarkers in 1000 Chinese patients with cancer. Cancer Med. 8:4699–4708. 2019.PubMed/NCBI View Article : Google Scholar

8 

Ajani JA, D'Amico TA, Bentrem DJ, Chao J, Cooke D, Corvera C, Das P, Enzinger PC, Enzler T, Fanta P, et al: Gastric cancer, version 2.2022, NCCN clinical practice guidelines in oncology. J Natl Compr Canc Netw. 20:167–192. 2022.PubMed/NCBI View Article : Google Scholar

9 

Thompson CB, Wang CY, Ho IC, Bohjanen PR, Petryniak B, June CH, Miesfeldt S, Zhang L, Nabel GJ, Karpinski B, et al: cis-acting sequences required for inducible interleukin-2 enhancer function bind a novel Ets-related protein, Elf-1. Mol Cell Biol. 12:1043–1053. 1992.PubMed/NCBI View Article : Google Scholar

10 

Tsokos GC, Nambiar MP and Juang YT: Activation of the Ets transcription factor Elf-1 requires phosphorylation and glycosylation: Defective expression of activated Elf-1 is involved in the decreased TCR zeta chain gene expression in patients with systemic lupus erythematosus. Ann NY Acad Sci. 987:240–245. 2003.PubMed/NCBI View Article : Google Scholar

11 

Andrews PGP, Kennedy MW, Popadiuk CM and Kao KR: Oncogenic activation of the human Pygopus2 promoter by E74-like factor-1. Mol Cancer Res. 6:259–266. 2008.PubMed/NCBI View Article : Google Scholar

12 

Hu M, Li H, Xie H, Fan M, Wang J, Zhang N, Ma J and Che S: ELF1 transcription factor enhances the progression of glioma via ATF5 promoter. ACS Chem Neurosci. 12:1252–1261. 2021.PubMed/NCBI View Article : Google Scholar

13 

Yang B, Shen F, Zhu Y, Lu W and Cai H: E74-like ETS transcription factor 1 promotes the progression of pancreatic cancer by regulating doublecortin-like kinase 1/Janus kinase/signal transducer and activator of transcription pathway. Am J Cancer Res. 14:616–629. 2024.PubMed/NCBI View Article : Google Scholar

14 

Zhang J, Chen L, Wei W and Mao F: Long non-coding RNA signature for predicting gastric cancer survival based on genomic instability. Aging (Albany NY). 15:15114–15133. 2023.PubMed/NCBI View Article : Google Scholar

15 

Xu TP, Wang YF, Xiong WL, Ma P, Wang WY, Chen WM, Huang MD, Xia R, Wang R, Zhang EB, et al: E2F1 induces TINCR transcriptional activity and accelerates gastric cancer progression via activation of TINCR/STAU1/CDKN2B signaling axis. Cell Death Dis. 8(e2837)2017.PubMed/NCBI View Article : Google Scholar

16 

He DX, Zhang GY, Gu XT, Mao AQ, Lu CX, Jin J, Liu DQ and Ma X: Genome-wide profiling of long non-coding RNA expression patterns in anthracycline-resistant breast cancer cells. Int J Oncol. 49:1695–1703. 2016.PubMed/NCBI View Article : Google Scholar

17 

Zeltz C, Primac I, Erusappan P, Alam J, Noel A and Gullberg D: Cancer-associated fibroblasts in desmoplastic tumors: Emerging role of integrins. Semin Cancer Biol. 62:166–181. 2020.PubMed/NCBI View Article : Google Scholar

18 

Harrison JD and Fielding JW: Prognostic factors for gastric cancer influencing clinical practice. World J Surg. 19:496–500. 1995.PubMed/NCBI View Article : Google Scholar

19 

Grzesiak M, Kaminska K, Knapczyk-Stwora K and Hrabia A: The expression and localization of selected matrix metalloproteinases (MMP-2, -7 and -9) and their tissue inhibitors (TIMP-2 and -3) in follicular cysts of sows. Theriogenology. 185:109–120. 2022.PubMed/NCBI View Article : Google Scholar

20 

Wang Y, Chuang CY, Hawkins CL and Davies MJ: Activation and inhibition of human matrix metalloproteinase-9 (MMP9) by HOCl, myeloperoxidase and chloramines. Antioxidants (Basel). 11(1616)2022.PubMed/NCBI View Article : Google Scholar

21 

Farina AR and Mackay AR: Gelatinase B/MMP-9 in tumour pathogenesis and progression. Cancers (Basel). 6:240–296. 2014.PubMed/NCBI View Article : Google Scholar

22 

Buttacavoli M, Di Cara G, Roz E, Pucci-Minafra I, Feo S and Cancemi P: Integrated multi-omics investigations of metalloproteinases in colon cancer: Focus on MMP2 and MMP9. Int J Mol Sci. 22(12389)2021.PubMed/NCBI View Article : Google Scholar

23 

Teng Z, Wang S, Yuan H, Wang H, Li J, Chang X, Zhang Y, Han Z and Wang Y: MMP-9 gene polymorphisms on cancer risk: An updated systematic review and meta-analysis. Nucleosides Nucleotides Nucleic Acids. 3:1–24. 2024.PubMed/NCBI View Article : Google Scholar

24 

Fu CK, Chang WS, Tsai CW, Wang YC, Yang MD, Hsu HS, Chao CY, Yu CC, Chen JC, Pei JS and Bau DT: The association of MMP9 promoter Rs3918242 genotype with gastric cancer. Anticancer Res. 41:3309–3315. 2021.PubMed/NCBI View Article : Google Scholar

25 

Costache S, Sajin M, Wedden S and D'Arrigo C: A consolidated working classification of gastric cancer for histopathologists (review). Biomed Rep. 19(58)2023.PubMed/NCBI View Article : Google Scholar

26 

Liu JY, Peng CW, Yang XJ, Huang CQ and Li Y: The prognosis role of AJCC/UICC 8th edition staging system in gastric cancer, a retrospective analysis. Am J Transl Res. 10:292–303. 2018.PubMed/NCBI

27 

Livak KJ and Schmittgen TD: Analysis of relative gene expression data using real-time quantitative PCR and the 2(-Delta Delta C(T)) method. Methods. 25:402–408. 2001.PubMed/NCBI View Article : Google Scholar

28 

Liao Q and Xiong J: YTHDF1 regulates immune cell infiltration in gastric cancer via interaction with p53. Exp Ther Med. 27(255)2024.PubMed/NCBI View Article : Google Scholar

29 

Cancer Genome Atlas Research Network. Comprehensive molecular characterization of gastric adenocarcinoma. Nature. 513:202–209. 2014.PubMed/NCBI View Article : Google Scholar

30 

Li T, Fu J, Zeng Z, Cohen D, Li J, Chen Q, Li B and Liu XS: TIMER2.0 for analysis of tumor-infiltrating immune cells. Nucleic Acids Res. 48 (W1):W509–W514. 2020.PubMed/NCBI View Article : Google Scholar

31 

Newman AM, Liu CL, Green MR, Gentles AJ, Feng W, Xu Y, Hoang CD, Diehn M and Alizadeh AA: Robust enumeration of cell subsets from tissue expression profiles. Nat Methods. 12:453–457. 2015.PubMed/NCBI View Article : Google Scholar

32 

Usui G, Matsusaka K, Mano Y, Urabe M, Funata S, Fukayama M, Ushiku T and Kaneda A: DNA methylation and genetic aberrations in gastric cancer. Digestion. 102:25–32. 2021.PubMed/NCBI View Article : Google Scholar

33 

Quan Q, Guo L, Huang L, Liu Z, Guo T, Shen Y, Ding S, Liu C and Cao L: Expression and clinical significance of PD-L1 and infiltrated immune cells in the gastric adenocarcinoma microenvironment. Medicine (Baltimore). 102(e36323)2023.PubMed/NCBI View Article : Google Scholar

34 

Liu D, Heij LR, Czigany Z, Dahl E, Lang SA, Ulmer TF, Luedde T, Neumann UP and Bednarsch J: The role of tumor-infiltrating lymphocytes in cholangiocarcinoma. J Exp Clin Cancer Res. 41(127)2022.PubMed/NCBI View Article : Google Scholar

35 

Alhalabi MM, Alsayd SA and Albattah ME: Advanced diffuse gastric adenocarcinoma in young Syrian woman. A case report. Ann Med Surg (Lond). 78(103728)2022.PubMed/NCBI View Article : Google Scholar

36 

Tuei VC, Maiyoh GK and Ndombera FT: The role of infections in the causation of cancer in Kenya. Cancer Causes Control. 33:1391–1400. 2022.PubMed/NCBI View Article : Google Scholar

37 

Chang ZW, Dong L, Qin YR, Song M, Guo HY and Zhu QL: Correlations between gastric cancer family history and ROBO2 and RASSF2A gene methylations. J Cancer Res Ther. 12:597–600. 2016.PubMed/NCBI View Article : Google Scholar

38 

Tan AC, Chan DL, Faisal W and Pavlakis N: New drug developments in metastatic gastric cancer. Therap Adv Gastroenterol. 11(1756284818808072)2018.PubMed/NCBI View Article : Google Scholar

39 

Li Z, Zhao Z, Wang C, Wang D, Mao H, Liu F, Yang Y, Tao F and Lu Z: Association between DCE-MRI perfusion histogram parameters and EGFR and VEGF expressions in different lauren classifications of advanced gastric cancer. Pathol Oncol Res. 27(1610001)2021.PubMed/NCBI View Article : Google Scholar

40 

He Y and Wang X: Identification of molecular features correlating with tumor immunity in gastric cancer by multi-omics data analysis. Ann Transl Med. 8(1050)2020.PubMed/NCBI View Article : Google Scholar

41 

Hsing M, Wang Y, Rennie PS, Cox ME and Cherkasov A: ETS transcription factors as emerging drug targets in cancer. Med Res Rev. 40:413–430. 2020.PubMed/NCBI View Article : Google Scholar

42 

Oettgen P, Akbarali Y, Boltax J, Best J, Kunsch C and Libermann TA: Characterization of NERF, a novel transcription factor related to the Ets factor ELF-1. Mol Cell Biol. 16:5091–5106. 1996.PubMed/NCBI View Article : Google Scholar

43 

Li T, Jia Z, Liu J, Xu X, Wang H, Li D and Qiu Z: Transcription activation of SPINK4 by ELF-1 augments progression of colon cancer by regulating biological behaviors. Tissue Cell. 84(102190)2023.PubMed/NCBI View Article : Google Scholar

44 

Takai N, Miyazaki T, Nishida M, Shang S, Nasu K and Miyakawa I: Clinical relevance of Elf-1 overexpression in endometrial carcinoma. Gynecol Oncol. 89:408–413. 2003.PubMed/NCBI View Article : Google Scholar

45 

Takai N, Miyazaki T, Nishida M, Nasu K and Miyakawa I: The significance of Elf-1 expression in epithelial ovarian carcinoma. Int J Mol Med. 12:349–354. 2003.PubMed/NCBI

46 

Cheng M, Zeng Y, Zhang T, Xu M, Li Z and Wu Y: Transcription factor ELF1 activates MEIS1 transcription and then regulates the GFI1/FBW7 axis to promote the development of glioma. Mol Ther Nucleic Acids. 23:418–430. 2020.PubMed/NCBI View Article : Google Scholar

47 

Qiao C, Qiao T, Yang S, Liu L and Zheng M: SNHG17/miR-384/ELF1 axis promotes cell growth by transcriptional regulation of CTNNB1 to activate Wnt/β-catenin pathway in oral squamous cell carcinoma. Cancer Gene Ther. 29:122–132. 2022.PubMed/NCBI View Article : Google Scholar

48 

Budka JA, Ferris MW, Capone MJ and Hollenhorst PC: Common ELF1 deletion in prostate cancer bolsters oncogenic ETS function, inhibits senescence and promotes docetaxel resistance. Genes Cancer. 9:198–214. 2018.PubMed/NCBI View Article : Google Scholar

49 

Paczkowska J, Soloch N, Bodnar M, Kiwerska K, Janiszewska J, Vogt J, Domanowska E, Martin-Subero JI, Ammerpohl O, Klapper W, et al: Expression of ELF1, a lymphoid ETS domain-containing transcription factor, is recurrently lost in classical Hodgkin lymphoma. Br J Haematol. 185:79–88. 2019.PubMed/NCBI View Article : Google Scholar

50 

Karamanos NK, Theocharis AD, Piperigkou Z, Manou D, Passi A, Skandalis SS, Vynios DH, Orian-Rousseau V, Ricard-Blum S, Schmelzer CEH, et al: A guide to the composition and functions of the extracellular matrix. FEBS J. 288:6850–6912. 2021.PubMed/NCBI View Article : Google Scholar

51 

Peltonen R, Hagström J, Tervahartiala T, Sorsa T, Haglund C and Isoniemi H: High expression of MMP-9 in primary tumors and high preoperative MPO in serum predict improved prognosis in colorectal cancer with operable liver metastases. Oncology. 99:144–160. 2021.PubMed/NCBI View Article : Google Scholar

52 

Wang T, Zhang Y, Bai J, Xue Y and Peng Q: MMP1 and MMP9 are potential prognostic biomarkers and targets for uveal melanoma. BMC Cancer. 21(1068)2021.PubMed/NCBI View Article : Google Scholar

53 

Xu T, Gao S, Liu J, Huang Y, Chen K and Zhang X: MMP9 and IGFBP1 regulate tumor immune and drive tumor progression in clear cell renal cell carcinoma. J Cancer. 12:2243–2257. 2021.PubMed/NCBI View Article : Google Scholar

54 

Quintero-Fabián S, Arreola R, Becerril-Villanueva E, Torres Romero JC, Arana-Argáez V, Lara-Riegos J, Ramírez Camacho MA and Alvarez-Sánchez ME: Role of matrix metalloproteinases in angiogenesis and cancer. Front Oncol. 9(1370)2019.PubMed/NCBI View Article : Google Scholar

55 

Liu C, Shen Y and Tan Q: Diagnostic and prognostic values of MMP-9 expression in ovarian cancer: A study based on bioinformatics analysis and meta-analysis. Int J Biol Markers. 38:15–24. 2023.PubMed/NCBI View Article : Google Scholar

56 

Jiang H and Li H: Prognostic values of tumoral MMP2 and MMP9 overexpression in breast cancer: A systematic review and meta-analysis. BMC Cancer. 21(149)2021.PubMed/NCBI View Article : Google Scholar

57 

Shao W, Wang W, Xiong XG, Cao C, Yan TD, Chen G, Chen H, Yin W, Liu J, Gu Y, et al: Prognostic impact of MMP-2 and MMP-9 expression in pathologic stage IA non-small cell lung cancer. J Surg Oncol. 104:841–846. 2011.PubMed/NCBI View Article : Google Scholar

58 

Lin Z, Liu Y, Sun Y and He X: Expression of Ets-1, Ang-2 and maspin in ovarian cancer and their role in tumor angiogenesis. J Exp Clin Cancer Res. 30(31)2011.PubMed/NCBI View Article : Google Scholar

59 

Zhang L, Li X, Feng X, Berkman T, Ma R, Du S, Wu S, Huang C, Amponsah A, Bekker A and Tao YX: E74-like factor 1 contributes to nerve trauma-induced nociceptive hypersensitivity through transcriptionally activating matrix metalloprotein-9 in dorsal root ganglion neurons. Pain. 164:119–131. 2023.PubMed/NCBI View Article : Google Scholar

60 

Wang L: ELF1-activated FOXD3-AS1 promotes the migration, invasion and EMT of osteosarcoma cells via sponging miR-296-5p to upregulate ZCCHC3. J Bone Oncol. 26(100335)2020.PubMed/NCBI View Article : Google Scholar

61 

Liu Y, Ma R, Juan D, Yuan Z, Sun J, Wang M, Li Y, Bao Y and Jin H: Adipose-derived mesenchymal stem cell-loaded β-chitin nanofiber hydrogel activates the AldoA/HIF-1α pathway to promote diabetic wound healing. Am J Stem Cells. 12:1–11. 2023.PubMed/NCBI

62 

Wang Y, Hu C, Kwok T, Bain CA, Xue X, Gasser RB, Webb GI, Boussioutas A, Shen X, Daly RJ and Song J: DEMoS: a deep learning-based ensemble approach for predicting the molecular subtypes of gastric adenocarcinomas from histopathological images. Bioinformatics. 38:4206–4213. 2022.PubMed/NCBI View Article : Google Scholar

63 

Bonsang B, Maksimovic L, Maille P, Martin N, Laurendeau I, Pasmant E, Bièche I, Deschamps J, Wolkenstein P and Ortonne N: VEGF and VEGFR family members are expressed by neoplastic cells of NF1-associated tumors and may play an oncogenic role in malignant peripheral nerve sheath tumor growth through an autocrine loop. Ann Diagn Pathol. 60(151997)2022.PubMed/NCBI View Article : Google Scholar

64 

De Re V, Alessandrini L, Brisotto G, Caggiari L, De Zorzi M, Casarotto M, Miolo G, Puglisi F, Garattini SK, Lonardi S, et al: HER2-CDH1 interaction via Wnt/B-catenin is associated with patients' survival in HER2-positive metastatic gastric adenocarcinoma. Cancers (Basel). 14(1266)2022.PubMed/NCBI View Article : Google Scholar

65 

Qin X, Chen Y, Ma S, Shen L and Ju S: Immune-related gene TM4SF18 could promote the metastasis of gastric cancer cells and predict the prognosis of gastric cancer patients. Mol Oncol. 16:4043–4059. 2022.PubMed/NCBI View Article : Google Scholar

66 

Liu C, Omilusik K, Toma C, Kurd NS, Chang JT, Goldrath AW and Wang W: Systems-level identification of key transcription factors in immune cell specification. PLoS Comput Biol. 18(e1010116)2022.PubMed/NCBI View Article : Google Scholar

67 

Klugman M, Fazzari M, Xue X, Ginsberg M, Rohan TE, Halmos B, Hanna DB, Shuter J and Hosgood HD III: The associations of CD4 count, CD4/CD8 ratio, and HIV viral load with survival from non-small cell lung cancer in persons living with HIV. AIDS Care. 34:1014–1021. 2022.PubMed/NCBI View Article : Google Scholar

68 

Fridman WH, Petitprez F, Meylan M, Chen TWW, Sun CM, Roumenina LT and Sautès-Fridman C: B cells and cancer: To B or not to B? J Exp Me. 218(e20200851)2021.PubMed/NCBI View Article : Google Scholar

69 

Peña-Romero AC and Orenes-Piñero E: Dual effect of immune cells within tumour microenvironment: Pro- and anti-tumour effects and their triggers. Cancers (Basel). 14(1681)2022.PubMed/NCBI View Article : Google Scholar

70 

Dou A and Fang J: Heterogeneous myeloid cells in tumors. Cancers (Basel). 13(3772)2021.PubMed/NCBI View Article : Google Scholar

Related Articles

Journal Cover

December-2024
Volume 28 Issue 6

Print ISSN: 1792-0981
Online ISSN:1792-1015

Sign up for eToc alerts

Recommend to Library

Copy and paste a formatted citation
x
Spandidos Publications style
Zhang X, Ren X, Zhang S and Wang Y: Overexpression of ELF1 combined with MMP9 is associated with prognosis and tumor microenvironment in gastric cancer. Exp Ther Med 28: 441, 2024.
APA
Zhang, X., Ren, X., Zhang, S., & Wang, Y. (2024). Overexpression of ELF1 combined with MMP9 is associated with prognosis and tumor microenvironment in gastric cancer. Experimental and Therapeutic Medicine, 28, 441. https://doi.org/10.3892/etm.2024.12730
MLA
Zhang, X., Ren, X., Zhang, S., Wang, Y."Overexpression of ELF1 combined with MMP9 is associated with prognosis and tumor microenvironment in gastric cancer". Experimental and Therapeutic Medicine 28.6 (2024): 441.
Chicago
Zhang, X., Ren, X., Zhang, S., Wang, Y."Overexpression of ELF1 combined with MMP9 is associated with prognosis and tumor microenvironment in gastric cancer". Experimental and Therapeutic Medicine 28, no. 6 (2024): 441. https://doi.org/10.3892/etm.2024.12730