www.fgks.org   »   [go: up one dir, main page]

IMR Press / FBL / Volume 29 / Issue 5 / DOI: 10.31083/j.fbl2905177
Open Access Original Research
F2RL3 Regulates Epithelial-Mesenchymal Transition and Angiogenesis in Gastric Cancer through the Rap1/MAPK Signaling Pathway
Show Less
1 Department of Clinical Medicine, Medical College of Soochow University, 215123 Suzhou, Jiangsu, China
2 General Surgery, Cancer Center, Department of Gastrointestinal and Pancreatic Surgery, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 310014 Hangzhou, Zhejiang, China
3 Key Laboratory of Tumor Molecular Diagnosis and Individualized Medicine of Zhejiang Province, Zhejiang Provincial People's Hospital, Affiliated People's Hospital, Hangzhou Medical College, 310014 Hangzhou, Zhejiang, China
*Correspondence: dshuang126@163.com (Dongsheng Huang)
Front. Biosci. (Landmark Ed) 2024, 29(5), 177; https://doi.org/10.31083/j.fbl2905177
Submitted: 19 July 2023 | Revised: 3 February 2024 | Accepted: 20 March 2024 | Published: 9 May 2024
Copyright: © 2024 The Author(s). Published by IMR Press.
This is an open access article under the CC BY 4.0 license.
Abstract

Background: Gastric cancer (GC) is frequently diagnosed at advanced stages, when cancer cells have already metastasized. Therefore, patients with GC have a low survival rate and poor prognosis even after treatment. Methods: We downloaded GC-related RNA sequencing (RNA-Seq) data, copy number variation (CNV) data, and clinical data for bioinformatics analysis to screen prognostic genes of GC. Single-sample gene set enrichment analysis and survival analyses were performed on the RNA-Seq data, and differential and correlation analyses were conducted on the CNV data to obtain CNV-driven differentially expressed genes (DEGs). Prognostic genes were identified through univariate Cox analyses of the CNV-driven DEGs, combined with the clinical data. F2R like thrombin or trypsin receptor 3 (F2RL3) was finally selected for verification after functional and survival analyses of the prognostic genes. Results: F2RL3 expression was lower in paracancer tissue than in GC tissue, and lower in GES-1 gastric epithelial cells than in GC cells. The cell culture supernatants from F2RL3-knockdown GC cells were collected and used to culture human umbilical vein endothelial cells (HUVECs). It was observed that F2RL3 enhanced the activity, metastasis, invasion, and angiogenesis of GC cells; promoted the epithelial–mesenchymal transition (EMT) of GC cells; and impacted the Ras-associated protein 1 (Rap1)/mitogen-activated protein kinase (MAPK) pathway. To further explore the involvement of the Rap1/MAPK pathway in GC development, a pathway activator was added to GC cells with knockdown of F2RL3 expression. This pathway activator not only enhanced the activity, invasion, and migration of GC cells but also promoted the EMT and blood vessel formation. Conclusions: F2RL3 regulates the angiogenesis and EMT of GC cells through the Rap1/MAPK pathway, thus influencing the onset and progression of GC.

Keywords
gastric cancer
copy number variant driver gene
F2RL3
epithelial–mesenchymal transition
angiogenesis
Rap1/MAPK pathway
1. Introduction

Gastric cancer (GC) is the fifth most common cancer and third leading cause of cancer-related deaths globally, posing a significant and pressing challenge to global health [1]. The primary contributors to GC are as follows: (1) genetic factors [2]; (2) infections, as 90% of GC cases result from Helicobacter pylori (H. pylori) infection, which destroys the gastric mucosa and disrupts the host’s inflammatory response with virulence factors [3], and 10% of GC cases are attributed to Epstein Barr virus (EBV) infection, which interferes with the cell cycle and induces hypermethylation of the tumor suppressor genes [4]; (3) unhealthy eating habits such as long-term consumption of high salt, fried, and overheated food, which increase the risk of cancer [5]; long-term bad habits such as smoking [6] and drinking [7], which stimulate gastric mucosa and accelerate the reproduction of H. pylori and EBV; and (4) uncontrollable factors such as age, sex, and hormones. Men are more likely to suffer from GC than women [8], and the probability of GC significantly increases after menopause, indicating a potential relationship of estrogen with the occurrence of GC [9]. Although radiotherapy, systemic chemotherapy, surgery [10], immunotherapy [11], and targeted therapy [12] are effective treatments for GC, they do little to improve the prognosis of most patients with advanced GC or effectively extend their lifespans [12]. The metastasis of cancer cells is the primary factor contributing to the poor survival rate of patients [13], and GC is generally diagnosed at advanced stages, when the tumor cells have spread and metastasized, so patients with GC often miss the opportunity for optimal treatment. Therefore, to prevent the progression of GC and improve the survival of patients with GC, it is of great research significance to perform early screening and fully investigate the pathogenesis of GC.

The epithelial–mesenchymal transition (EMT) is a critical process in tumor metastasis and invasion, during which time epithelial cells undergo a transition characterized by the loss of intercellular adhesion and acquisition of mesenchymal cell traits, accompanied by enhanced migration and invasion [14, 15]. Tian et al. [16] found that serpin family H member 1 modulates the Wnt/β-catenin pathway and affects the EMT, thus affecting the progression of GC. Homeobox A10 promotes GC metastasis by regulating the transforming growth factor beta-2 (TGFb2)/Smad/methyltransferase-like 3 signal axis to mediate the EMT [17]. Angiogenesis is a crucial event in the development of tumors [18]. Blood vessels play an essential role in delivering nutrients and oxygen to tumor cells, and vascular endothelial growth factor (VEGF) is critical in angiogenesis [19]. Moreover, VEGF is a prognostic biomarker for patients who have undergone GC resection [20, 21, 22].

In this study, we downloaded RNA sequencing (RNA-Seq) data, copy number variation (CNV) data, and clinical data related to GC from The Cancer Genome Atlas (TCGA). The RNA-Seq data underwent single-sample gene set enrichment analysis (ssGSEA), and the samples were re-grouped according to the EMT and angiogenesis indexes, followed by correlation and survival analyses. Based on the CNV data, we performed screening to identify CNV-driven differentially expressed genes (DEGs). Finally, combined with the clinical data, prognostic genes were obtained by performing univariate Cox analysis of the CNV-driven DEGs. After conducting functional enrichment analysis, protein–protein interaction (PPI) analysis, and Kaplan-Meier (K-M) analysis of the obtained genes, F2R like thrombin or trypsin receptor 3 (F2RL3) was selected for further validation through cell experiments. In conclusion, by combining bioinformatics analysis and experimental investigations, this study lays the groundwork for a better understanding of the potential molecular mechanism of GC metastasis and offers a novel targeted therapy for GC.

2. Materials and Methods
2.1 Data Acquisition

We retrieved RNA-Seq data, CNV data, and clinical data of GC from TCGA database. TCGA provides comprehensive data for more than 20,000 primary cancer samples spanning 33 different cancer types. Gene sets related to the EMT and angiogenesis were obtained from the Molecular Signatures Database (MsigDB), which offers resources of annotated gene sets for use. TCGA data were in count format and standardized as log2 (count+1) values.

2.2 Analyses of the EMT and Angiogenesis Indexes

The gene set variation analysis package in R was used to conduct ssGSEA analysis of the RNA-Seq data obtained from 375 tumor samples and 32 paracancer samples. Subsequently, the EMT and angiogenesis indexes for each sample were calculated. According to the median, the samples were segmented into low/high EMT and low/high angiogenesis index groups. Boxplots illustrating the EMT and angiogenesis indexes were generated using the ggplot2 in R. Correlation analyses between the EMT index and angiogenesis index were carried out using the cor function in R. Survival analyses were conducted using the survival package in R.

2.3 Screening and Functional Analyses of DEGs

The DESeq2 package (Version 4.2) in R was applied for screening DEGs in the high and low angiogenesis index groups. DEGs meeting the criteria of p < 0.05 and |log2FC| >1 were deemed statistically significant. The EnhancedVolcano package (Version 1.11.3) in R was used to generate volcano plots illustrating the observed differences. Kyoto Encyclopedia of Genes and Genomes (KEGG) and Gene Ontology (GO) enrichment analyses were conducted using the clusterProfiler (version 4.2.2, https://yulab-smu.top/biomedical-knowledge-mining-book/ (docs) https://doi.org/10.1016/j.xinn.2021.100141 (paper)) package in R. Enrichment results were analyzed using the ggplot2 (version 3.3.6, https://ggplot2.tidyverse.org, https://github.com/tidyverse/ggplot2) package in R [23].

2.4 Screening for Prognostic Genes

The CNV data and angiogenesis index of the samples were integrated to obtain the segment data of 381 samples. Copy number at the gene level was determined using GISTIC 2.0 (version 7, https://www.genepattern.org/modules/docs/GISTIC_2.0), with a threshold set to 0.3. Fisher’s exact test was employed to assess the hypothesis of gene copy number distribution between samples with high and low angiogenesis, and identify genes with significant differences in copy number between the low and high angiogenesis groups. Then the CNV-driven DEGs were obtained by Pearson’s correlation analysis for copy number and expression of genes. Combined with the clinical data, prognosis-related genes were identified by performing univariate Cox regression analysis of the CNV-driven DEGs using the survival package in R. PPI analysis was carried out using the Search Tool for the Retrieval of Interacting Genes/Proteins (STRING) version 11.5, and K-M analysis was conducted using the Gene Expression Profiling Interactive Analysis database.

2.5 Clinical Sample Collection

During GC resection, specimens of 1 cm3 were obtained from both the GC tissue and paracancer tissue. The surgically resected tissue specimens were fixed in 4% formaldehyde solution, and preserved as a wax block.

2.6 Cell Culture

AGS GC cells (#CRL-1739) were purchased from the American Type Culture Collection (ATCC, Manassas, VA, USA) and cultured in F-12K medium (#30-2004; ATCC) supplemented with 10% fetal bovine serum (FBS). Human umbilical vein endothelial cells (HUVECs) were purchased from ATCC (#PCS-100-013), and cultured in Dulbecco’s Modified Eagle Medium containing 10% FBS and 1% penicillin/streptomycin. GES-1 gastric epithelial cells were purchased from Cobioer Biosciences Co., Ltd. (#CBP60512; Nanjing, China) and cultured in DMEM (Cobioer) supplemented with 10% FBS. HGC-27 GC cells were purchased from Cobioer (#CBP60480) and inoculated in RPMI-1640 medium (Cobioer) supplemented with 10% FBS. All cells used in the experiments were cultured in an incubator at 37 °C with 5% CO2. All cell lines were validated by short tandem repeat profiling and tested negative for mycoplasma. The Ras-associated protein 1 (Rap1)/mitogen-activated protein kinase (MAPK) signaling pathway activator, staurosporine aglycone (SA), was purchased from Sigma (#S3939; St. Louis, MO, USA), and the cells were subjected to pre-treatment with 30 µmol/L SA for 2 h prior to the commencement of subsequent experiments.

2.7 Cell processing

Cells were seeded in 12-well plates (serum-free medium) at a density of 1.2 × 105 per well and transfected with F2RL3 overexpressed plasmid using jetPRIME® Transfection Reagent (Polyplus, Illkirch-Graffenstaden, France). Cells were seeded in 12-well plates (serum-free medium) at a density of 1.2 × 105 per well and transfected with small interfering RNA (siRNA) oligonucleotide targeting F2RL3 (Sangon Biotech, Shanghai, China) using jetPRIME® Transfection Reagent (Polyplus, Illkirch-Graffenstaden, France) for 6 h. Subsequently, medium supplemented with 10% FBS was added for incubation for 24 h, followed by the detection of F2RL3 expression by quantitative polymerase chain reaction (qPCR). After 72 h, F2RL3 protein expression was detected by Western blotting.

2.8 Western Blotting

Proteins were extracted from the cell lysate supplemented with 1% protease inhibitor. The protein content in the sample was quantified using the bicinchoninic acid assay, followed by electrophoresis, electrotransfer of proteins to a membrane, and incubation with the following primary antibodies: F2RL3 (#ab137927; Abcam, Cambridge, MA, USA), E-cadherin (#ab40772; Abcam), vimentin (#ab92547; Abcam), Rap1 GTPase-activating-protein (Rap1GAP) (#ab32373; Abcam), Rap1 (#ab175329; Abcam), phosphorylated extracellular signal-regulated kinase 1/2 (p-ERK1/2) (#4370T; Cell Signaling Technology [CST], Danvers, MA, USA), ERK1/2 (#9194S; CST), p-p38 MAPK (#4511S; CST), p38 MAPK (#8690S; CST), and Snail (#3879S; CST). β-actin (#93473SF; CST) was selected as the internal reference gene. After the proteins were developed, their expression levels were quantified with ImageJ software. Three biological replicates were performed for all experiments.

2.9 Quantitative Polymerase Chain Reaction (qPCR)

Total RNA extraction was performed using the TRIzol Kit (Thermo Fisher Scientific, Waltham, MA, USA), and the extracted RNA was treated with DNase. The PrimeScript RT Reagent Kit (TaKaRa, Shiga, Japan) was used to obtain the cDNA, and the cDNA template was diluted with RNase-free water. As per the instructions of the SYBR Green PCR Kit (Thermo Fisher Scientific), qPCR was performed in a total reaction volume of 10 µL. The qPCR conditions included pre-denaturation at 95 °C for 15 min, followed by 40 cycles of denaturation at 94 °C for 15 s, annealing at 55 °C for 30 s, and annealing at 72 °C for 30 s. The primers sequences were as follows: F2RL3 forward primer, AGAAGAGGAGAGGACACAGAGACAC; F2RL3 reverse primer, CTTGGCATCGTGGCATCCCTTAG; β-actin forward primer, CAGATGTGGATCAGCAAGCAGGAG; and β-actin reverse primer, CGCAACTAAGTCATAGTCCGCCTAG. The 2-ΔΔCt method was used to calculate the obtained results. Three biological replicates were performed for all experiments.

2.10 Immunohistochemistry

Tissue sections were prepared through slicing, dewaxing, and hydration processes. Antigen retrieval and inactivation were performed, followed by serum blocking. Then the sections were incubated with F2RL3 primary antibody (#25306-1-AP; Proteintech, Rosemont, IL, USA), followed by incubation with secondary antibody. Diaminobenzidine (DAB) color development was carried out, and cell nuclei were counterstained with hematoxylin. Finally, the samples were dehydrated, cleared, and observed, with images captured under a microscope. Three biological replicates were performed for all experiments.

2.11 Evaluation of Cell Activity, Invasion, and Metastasis

Cells in logarithmic growth phase were used to prepare a cell suspension, cultured for 24 h, and then treated with Cell Counting Kit-8 (CCK-8) reagent (C0038; Beyotime, Beijing, China) for an additional 2 h. The absorbance at 450 nm of each cell suspension was detected using an enzymoleter. The upper compartment of the membrane at the bottom of the Transwell chamber was coated with Matrigel. After solidification, 100 µL of the cell suspension was added to the lower compartment, followed by the addition of medium to the lower compartment. Following 24 h of routine culture in the cell incubator and staining with crystal violet, cell invasion was assessed by observing the number of cells in the visual field under a microscope. For the scratch experiment, a trace gun was used to mark the central region of cell growth in single-layer adherent cells cultured in vitro dishes. Cells in the central part were removed, and culture continued with serum-free medium. Cell migration was evaluated by observing whether the surrounding cells migrated to the central scratch area under a microscope. Three biological replicates were performed for all experiments.

2.12 Tube Formation Test

The supernatant of GC cells with F2RL3 knockdown was collected and used to culture HUVECs. Precooled Matrigel matrix was added to 24-well plates, followed by the addition of 500 µL cell suspension of HUVECs co-cultured with GC cells into each respective well. Tube formation outcomes were assessed following routine culture in a cell incubator for 2 to 6 h. Tube formation and number were recorded using NIS-Elements BR software (Nikon, Melville, NY, USA). Three biological replicates were performed for all experiments.

2.13 Statistical Analyses

Statistical analyses of the bioinformatics analysis results and graphing were completed using R, and between-group comparisons were performed using the Wilcoxon rank-sum test. For the experimental results, statistical analyses and graphing were completed using Graphpad Prism (version 9.4.0, Boston, MA, USA), and between-group comparisons were conducted using t-tests. All experiments at the cellular level were performed with three biological replicates. p < 0.05 was considered statistically significant.

3. Results
3.1 Analyses of the EMT and Angiogenesis Indexes of GC

The EMT and angiogenesis indexes of 375 tumor samples and 32 paracancer samples of GC were analyzed and calculated. Results indicated a higher EMT index (Fig. 1A) and angiogenesis index (Fig. 1B) of the tumor samples compared to the paracancer samples. Further, a notable positive correlation was identified between the EMT index and the angiogenesis index (p = 2.2 × 10-16, R = 0.67; Fig. 1C). In addition, according to survival analyses, a statistically significant difference was revealed in the survival time between low and high angiogenesis indexes, with a poorer prognosis noted for those with a high angiogenesis index (Fig. 1D).

Fig. 1.

Analyses of the epithelial-mesenchymal transition (EMT) and angiogenesis indexes of the gastric cancer (GC) samples. (A) The EMT index between tumor and paracancer samples. (B) Angiogenesis index between tumor and paracancer samples. (C) Correlation analyses of the EMT and angiogenesis index. (D) Survival analyses between the low and high angiogenesis groups.

3.2 DEG Screening and Functional Analysis

Analysis of the differential expression of genes showed 573 DEGs between the high and low angiogenesis samples, including 496 upregulated and 77 downregulated genes (Fig. 2A). These DEGs were mainly enriched in the phosphoinositide 3-kinase/protein kinase B pathway, integrin binding, phagocytosis, signaling receptor activator activity, humoral immune response mediated by circulating immunoglobulin, and extracellular matrix–receptor interaction as demonstrated by GO and KEGG enrichment analyses (Fig. 2B–E).

Fig. 2.

Enrichment analyses of differentially expressed genes (DEGs). (A) Volcanic map. (B) Gene Ontology (GO)-Biological Process. (C) GO-Cellular Component. (D) GO-Molecular Function. (E) Kyoto Encyclopedia of Genes and Genomes (KEGG). PI3K-Akt, phosphoinositide 3-kinase-protein kinase B; ECM, Extracellular matrix; IL-17, interleukin-17; AGE-RAGE, advanced glycation end products-receptor for advanced glycation end products.

3.3 Acquisition of Prognostic Genes Associated with Angiogenesis

The gene-level copy number was calculated, and the distribution of gene copy number between samples with low and high angiogenesis was tested. Genes with significant differences in copy number between the high and low angiogenesis groups were identified. Then correlation analysis of gene copy number and expression level was conducted, leading to the identification of 37 CNV-driven DEGs (copy number was significantly different and positively correlated with the expression level). Combined with the clinical data, univariate Cox analyses of these 37 genes revealed 16 prognosis-related genes. Axis inhibition protein 2 (AXIN2) demonstrated a hazard ratio (HR) less than 1, indicating its role as a protective factor; that is, higher AXIN2 expression was correlated with a higher survival rate. Conversely, the HRs of other genes exceeded 1, indicating that these genes were risk factors, where higher gene expression was associated with lower survival rate (Fig. 3).

Fig. 3.

Forest map of the prognosis-related genes. HR, hazard ratio; AXIN2, Axis inhibition protein 2; TENM3, teneurin transmembrane protein 3; DUSP1, dual specificity phosphatase 1; FBN1, fibrillin 1; THBS1, thrombospondin-1; LBP, lipopolysaccharide binding protein; MRAP, Melanocortin 2 Receptor Accessory Protein; ERG, E-twenty six transcription factor; GREM1, Gremlin 1; ADAMTS1, A disintegrin and metalloproteinase with thrombospondin motifs 1; PTGFR, prostaglandin F receptor; ADH1B, alcohol dehydrogenase 1B (class I), beta polypeptide; F2RL3, F2R like thrombin or trypsin receptor 3; CCDC178, coiled-coil domain containing 178; CYTL1, cytokine like 1; PCDHGA3, protocadherin gamma subfamily A, 3.

3.4 Screening of Key Prognostic Genes

The 16 identified genes underwent PPI analysis using the STRING database (Fig. 4A), and KEGG analysis was performed using clusterProfiler (Fig. 4B and Table 1). KEGG analyses revealed the enrichment of fibrillin-1/thrombospondin 1/gremlin 1 in the TGFb signaling pathway, AXIN2/melanocortin 2 receptor accessory protein in Cushing’s syndrome, and thrombospondin-1/F2RL3 in the Rap1 pathway (p < 0.05). Currently, there is a lack of substantial advancements in studies on the mechanism of the Rap1 pathway in GC, and the role of F2RL3 participating in this pathway in GC remains unexplored. Therefore, an in-depth investigation on F2RL3 was conducted in this study. PPI analysis revealed an interaction between F2RL3 and prostaglandin F receptor (Fig. 4A). While F2RL3 expression did not significantly differ between the tumor and normal tissues (p > 0.05; Fig. 4C), it was notably upregulated in advanced GC (stages I, III, and IV) compared to early-stage GC (stage I) (Fig. 4D). As shown by K-M analyses, higher expression of F2RL3 was associated with lower survival rates (Fig. 4E).

Fig. 4.

Screening of key prognostic genes. (A) Protein–protein interaction (PPI) analysis. (B) KEGG analysis. (C) Expression of F2RL3 between tumor and normal tissues. (D) Expression of F2RL3 in GC stages. (E) Kaplan-Meier (K-M) analysis of F2RL3.

Table 1.KEGG enrichment analysis.
ID Description p Gene ID
hsa04350 TGFb signaling pathway 0.000233 FBN1/THBS1/GREM1
hsa04934 Cushing syndrome 0.017834 AXIN2/MRAP
hsa04015 Rap1 signaling pathway 0.031484 THBS1/F2RL3
hsa05165 Human papillomavirus infection 0.071648 THBS1/AXIN2
hsa04080 Neuroactive ligand–receptor interaction 0.07549 PTGFR/F2RL3

TGFb, Transforming growth factor beta.

3.5 F2RL3 Expression in GC Tissue

Western blotting, qPCR, and immunohistochemistry were used for the detection of F2RL3 expression in GC and paracancer tissues, with the results showing significantly lower F2RL3 expression in paracancer tissue compared to GC tissue. At the same time, according to the TNM stage, we divided the clinical samples into pre-tumor (stage I) and pro-tumor (stage II, III, IV) samples, and found that the expression of F2RL3 was significantly increased in the pro-tumor samples compared with the pre-tumor samples (Fig. 5A–D). Therefore, we speculated that the expression of F2RL3 can promote the malignant progression of tumors.

Fig. 5.

F2RL3 expression level in human GC tissue. (A) F2RL3 expression detected by Western blotting (n = 6). (B) Bar chart showing F2RL3 expression (n = 6). (C) F2RL3 expression detected by quantitative polymerase chain reaction (qPCR) (n = 6). (D) F2RL3 expression detected by immunohistochemistry (n = 6), Scale: 100 µm. **p < 0.01.

3.6 F2RL3 Expression in GC Cells

qPCR and western blotting were used to detect F2RL3 expression in normal human gastric epithelial cells (GES-1) and human GC cells (HGC-27 and AGS) (Fig. 6A–C). The results showed significantly lower F2RL3 expression in GES-1 cells than in AGS and HGC-27 cells. Additionally, AGS cells exhibited higher expression of F2RL3 compared to HGC-27 cells.

Fig. 6.

F2RL3 expression level in human GC cells. (A) F2RL3 expression detected by Western blotting. (B) Bar chart showing F2RL3 expression. (C) F2RL3 expression detected by qPCR (n = 3). *p < 0.05, **p < 0.01.

3.7 Effects of F2RL3 on the EMT

To determine the effect of F2RL3 in GC cells, the siF2RL3 plasmid was transfected into AGS and HGC-27 cells to knockdown F2RL3 expression (Fig. 7A–C). The siRNA with the highest knockdown efficiency (AGS-siF2RL3 b and HGC-27-siF2RL3 b) was selected for subsequent experiments. The CCK-8 assay demonstrated a notable reduction in the activity of GC cells after knocking down F2RL3 expression (Fig. 7D). The Transwell (Fig. 7E,F) and scratch (Fig. 7G,H) assays demonstrated a notable reduction in the metastasis and invasion of GC cells upon knockdown of F2RL3 expression. At the same time, the regulation of F2RL3 expression led to upregulation of the epithelial marker E-cadherin and downregulation of the interstitial markers Snail and vimentin (Fig. 7I,J), indicating that F2RL3 can regulate the EMT of GC cells.

Fig. 7.

Effects of F2RL3 knockdown on the EMT of GC cells. (A) F2RL3 expression detected by Western blotting. (B) Bar chart showing F2RL3 expression. (C) F2RL3 expression detected by qPCR. (D) GC cell activity detected by the CCK-8. (E) GC cell invasion detected by the Transwell, Scale: 100 µm. (F) Bar graph showing the invasion of GC cells. (G) GC cell migration detected by the scratch assay, Scale: 100 µm. (H) Bar graph showing GC cell migration. (I) Expression of E-cadherin, Snail, and vimentin detected by Western blotting. (J) Bar chart showing E-cadherin, Snail, and vimentin expression (n = 3). *p < 0.05, **p < 0.01.

3.8 Effects of F2RL3 on Angiogenesis

To investigate the role of F2RL3 in angiogenesis, we knocked down F2RL3 expression in AGS and HGC-27 cells, cultured the cells, and then collected the cell culture supernatants. HUVECs were cultured with GC cell culture supernatants. The activity of HUVECs in the AGS-siF2RL3 and HGC27-siF2RL3 groups was significantly decreased (Fig. 8A), and its metastasis and invasion abilities (Fig. 8B–E) as well as angiogenesis (Fig. 8F,G) were significantly attenuated (p < 0.05).

Fig. 8.

Effects of F2RL3 expression knockdown on angiogenesis. (A) The activity of Human Umbilical Vein Endothelial Cells (HUVECs) detected by the CCK-8. (B) The invasion of HUVECs detected by the Transwell, Scale: 100 µm. (C) Bar graph showing the invasion of HUVECs. (D) The migration of HUVECs detected by the scratch assay, Scale: 100 µm. (E) Bar graph showing the migration of HUVECs. (F) Tube-forming ability of HUVECs detected by the tube formation assay, Scale: 100 µm. (G) Bar graph showing the tube-forming ability of HUVECs (n = 3). *p < 0.05, **p < 0.01.

3.9 F2RL3 Regulates the Expression of Rap1/MAPK Pathway in GC Cells

F2RL3 is known to regulate the EMT and angiogenesis in GC, but its molecular mechanism is not fully understood. Thus, we investigated the impact of F2RL3 on the Rap1/MAPK signaling pathway. Western blot analysis of the expression of Rap1/MAPK pathway-related proteins (Rap1GAP, Rap1, p-ERK1/2, ERK1/2, p-p38 MAPK, and p38 MAPK) revealed the upregulated expression of Rap1GAP in the AGS-siF2RL3 group, accompanied by the downregulated expression of Rap1, p-p38 MAPK, and p-ERK1/2. In the AGS-oeF2RL3 group, the expression of Rap1GAP was downregulated, while that of Rap1, p-p38 MAPK, and p-ERK1/2 was upregulated (Fig. 9A,B). These results indicate that F2RL3 can regulate the Rap1/MAPK pathway.

Fig. 9.

F2RL3 regulates the expression of Rap1/MAPK signaling pathway in GC cells. (A) Expression of proteins related to F2RL3 and the Rap1/MAPK signaling pathway detected by Western blotting. (B) Bar graph showing the expression of proteins related to F2RL3 and the Rap1/MAPK pathway (n = 3). **p < 0.01.

3.10 F2RL3 Regulates the EMT of GC Cells through the Rap1/MAPK Signaling Pathway

F2RL3 was identified as a regulator of the Rap1/MAPK signaling pathway, prompting further investigation into its influence on the EMT process of GC cells. Groups were set as AGS, AGS-siControl, AGS-siF2RL3, and AGS-siF2RL3-Rap1/MAPK signaling pathway activator (SA). Following knockdown of F2RL3 expression in AGS cells, E-cadherin expression increased, while Snail and vimentin expression decrease (Fig. 10A,B). Meanwhile, the activity (Fig. 10C), metastasis, and invasion of GC cells (Fig. 10D–G) was significantly decreased (p < 0.05). Further treatment with SA showed weakened effect of siF2RL3, along with the downregulated expression of E-cadherin and upregulated expression of Snail and vimentin (Fig. 10A,B). Meanwhile, there was increased cell activity (Fig. 10C), and enhanced metastasis and invasion (Fig. 10D–G). These results collectively indicate that F2RL3 can regulate the EMT of GC cells through the Rap1/MAPK signaling pathway.

Fig. 10.

Effects of the Rap1/MAPK signaling pathway on the EMT of GC cells. (A) Expression of F2RL3, E-cadherin, vimentin, and Snail detected by Western blotting. (B) Bar chart showing the expression of F2RL3, vimentin, E-cadherin, and Snail. (C) Activity of GC cells detected by the CCK-8. (D) Invasion of GC cells detected by the Transwell, Scale: 100 µm. (E) Bar graph showing the invasion of GC cells. (F) Migration of GC cells detected by the scratch assay, Scale: 100 µm. (G) Bar graph showing the migration of GC cells (n = 3) (*p < 0.05, **p < 0.01).

3.11 F2RL3 Regulates Angiogenesis through the Rap1/MAPK Pathway

HUVECs were cultured with GC cell culture supernatants collected from the AGS, AGS-siControl, AGS-siF2RL3, AGS-siF2RL3-SA groups, and its activity (Fig. 11A), metastasis, invasion (Fig. 11B–E), and angiogenesis (Fig. 11F,G) were assessed. The results showed that the activity of HUVECs in the AGS-siF2RL3 group was decreased (p < 0.05), accompanied by attenuated metastasis and invasion, along with significantly reduced angiogenesis. However, HUVECs in the AGS-siF2RL3-SA group showed significantly enhanced cell activity, metastasis, invasion, and angiogenesis (p < 0.05). Together, these results underscore the regulatory role of F2RL3 in angiogenesis through the Rap1/MAPK signaling pathway.

Fig. 11.

Impact of the Rap1/MAPK signaling pathway on angiogenesis. (A) Activity of HUVECs detected by the CCK-8. (B) Invasion of HUVECs detected by the Transwell, Scale: 100 µm. (C) Bar graph showing the invasion of HUVECs. (D) Migration of HUVECs detected by the scratch assay, Scale: 100 µm. (E) Bar graph showing the migration of HUVECs. (F) Tube forming ability of HUVECs detected by the tube formation assay, Scale: 100 µm. (G) Number of HUVEC tubules (n = 3). **p < 0.01.

4. Discussion

As one of the most common malignancies in China, GC originates from mucosal epithelial cells on the surface layer of the gastric wall, and its occurrence is a multistep and multifactor process [24]. Although current therapeutic methods for GC are relatively effective, there is still low survival rate for patients after treatment, due to tumor metastasis [11]. Early clinical trials, including those on ramucirumab, paclitaxel, and anti-netrin-1 antibody, have demonstrated that inhibition of the EMT and angiogenesis show promising antitumor activities [25, 26]. In addition, dextran sulfate has been identified as a treatment for GC by inhibiting angiogenesis through interference with the polarization of M2-type macrophages [27]. Furthermore, resveratrol exhibits potential in inhibiting the EMT by weakening the Hippo/Yes-associated protein 1 signaling pathway to delay the progression of GC [28]. The EMT [29] and angiogenesis [18] distinctly contribute to tumor growth and progression during tumor metastasis, yet the precise regulatory molecular mechanism of tumor metastasis after treatment still remains to be clarified.

To investigate the mechanism that affects the metastasis of advanced GC, GC-related data were collected from TCGA and MsigDB. A strong positive correlation was found between the EMT and angiogenesis indexes of the samples within the dataset. Notably, patients with a high angiogenesis index exhibited a significantly poorer prognosis compared to those with a low angiogenesis index. Subsequent hypothesis test and correlation analyses conducted based on gene-level copy number of samples led to the identification of 37 CNV-driven DEGs. By conducting survival analyses, 16 prognosis-related genes were finally obtained. According to the results of KEGG functional analyses, the prognosis-related genes were mostly enriched in the TGFb and Rap1 signaling pathways. In the early stage of cancer, the TGFb signaling pathway inhibits cancer development by promoting apoptosis and cell cycle progression, while in the late stage of cancer, it contributes to cancer development by promoting tumor invasion and metastasis [30]. Finally, F2RL3 was selected for further experimental verification. No significant difference was found in the expression of F2RL3 between normal and tumor tissues, and the expression of F2RL3 in the tumor tissues of patients in the late stages of GC is significantly upregulated compared with that in the early stages. There was significantly lower levels of F2RL3 expression in GES-1 cells than in GC cells. Data obtained by high-throughput sequencing were used for bioinformatics analysis, and the expression of all exon regions was taken into account when calculating the gene expression level, which explained why there was no difference in expression. F2RL3 is essential for platelet activation [31] and is involved in the invasion and migration of colon tumors [32]. Zhang et al. [33] found that F2RL3 hypomethylation is strongly associated with the incidence and mortality of lung cancer and is also recognized as a biomarker of smoking-related diseases [34].

After knockdown of F2RL3 expression in GC cells, the activity, invasion, and migration of GC cells were weakened, which restrained the angiogenesis and EMT of GC cells. KEGG analyses revealed the involvement of F2RL3 in the Rap1 signaling pathway, and the expression of Rap1/MAPK pathway-related proteins was also significantly changed after knockdown of F2RL3 expression. Rap1, a member of the Ras superfamily of small G proteins, promotes not only the invasion, migration and metastasis of various types of cancers [35] but also angiogenesis and endothelial barrier function [36]. Rap1GAP can interact with Rap1-GTP, converting the latter into inactive Rap1-GDP, thereby inhibiting activation of the RAS/ERK/MAPK mitosis pathway [37]. Rap1GAP is a tumor suppressor gene that inhibits tumor growth [38]. Yang et al. [39] found that Rap1GAP is an tumor metastasis and EMT suppressor in GC, and low expression of Rap1GAP is associated with a poor prognosis in patients with GC. The Rap1/MAPK pathway is related to the occurrence and development of colon cancer [40]. To verify the impact of the Rap1/MAPK signaling pathway on tumor metastasis, a Rap1/MAPK signaling pathway activator was added to F2RL3-knockdown GC cells, after which the activity, invasion, and migration of GC cells were enhanced, and the EMT and angiogenesis of GC cells were promoted. Therefore, F2RL3 can regulate the EMT and angiogenesis of GC by affecting the Rap1/MAPK signaling pathway.

This study had some limitations. In clinical practice, the conditions of patients with GC are more complex. According to previous studies, the onset of GC correlates with uncontrollable factors such as age, sex, and lifestyle, and there are individual differences. Therefore, it is unknown whether the results of our study will be applicable in clinical practice in the future to help develop treatments that will delay the progression of GC. In the coming years, we will conduct further in-depth investigations into the mechanism of action of F2RL3 in tumors in vivo and try to collect more clinical samples to verify the function of F2RL3 in the tumor tissues of GC patients.

5. Conclusions

During this study, bioinformatics analysis of GC-related data was conducted based on TCGA database, and F2RL3 was substantiated as a potential key contributor to the metastasis of GC. Through experimental verification, it was observed that decreased expression of F2RL3 reduced the activity, attenuated the invasion and migration of GC cells, and inhibited the angiogenesis and EMT of GC. However, the addition of a Rap1/MAPK pathway activator reversed the decreased expression of F2RL3 to some extent.

To summarize, we uncovered the novel role of F2RL3, a previously unexplored gene in GC-related mechanisms, and confirmed that F2RL3 can affect the EMT and angiogenesis. Our findings not only provide an in-depth understanding of the potential molecular mechanism underlying the metastasis of GC but also offers innovative avenues for targeted therapies in patients with GC.

Availability of Data and Materials

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

Author Contributions

JM contributed substantially to the conception and design of the work, and drafted the manuscript. YS and QL contributed to the acquisition, analysis, and interpretation of data for the work. DH made supporting efforts in the conception and design of the work, as well as reviewed the manuscript and revised it critically. All authors agreed the final version of the manuscript to be published. All authors have participated sufficiently in the work to take public responsibility for appropriate portions of the content and agreed to be accountable for all aspects of the work in ensuring that questions related to its accuracy or integrity. All authors contributed to editorial changes in the manuscript.

Ethics Approval and Consent to Participate

The protocols and experiments involved in this study were approved by Zhejiang Provincial People’s Hospital, and all participants signed informed consent (Ethics Number: QT2023006).

Acknowledgment

Not applicable.

Funding

This research was funded by the Medical and Health Science and Technology Program of Zhejiang Province (grant number: 2021KY523).

Conflict of Interest

The authors declare no conflict of interest.

References
[1]
Smyth EC, Nilsson M, Grabsch HI, van Grieken NC, Lordick F. Gastric cancer. Lancet (London, England). 2020; 396: 635–648.
[2]
Dong LM, Potter JD, White E, Ulrich CM, Cardon LR, Peters U. Genetic susceptibility to cancer: the role of polymorphisms in candidate genes. JAMA. 2008; 299: 2423–2436.
[3]
Sukri A, Hanafiah A, Mohamad Zin N, Kosai NR. Epidemiology and role of Helicobacter pylori virulence factors in gastric cancer carcinogenesis. APMIS: Acta Pathologica, Microbiologica, et Immunologica Scandinavica. 2020; 128: 150–161.
[4]
Naseem M, Barzi A, Brezden-Masley C, Puccini A, Berger MD, Tokunaga R, et al. Outlooks on Epstein-Barr virus associated gastric cancer. Cancer Treatment Reviews. 2018; 66: 15–22.
[5]
Maddineni G, Xie JJ, Brahmbhatt B, Mutha P. Diet and carcinogenesis of gastric cancer. Current Opinion in Gastroenterology. 2022; 38: 588–591.
[6]
Scherübl H. Smoking tobacco and cancer risk. Deutsche Medizinische Wochenschrift (1946). 2021; 146: 412–417. (In German)
[7]
Deng W, Jin L, Zhuo H, Vasiliou V, Zhang Y. Alcohol consumption and risk of stomach cancer: A meta-analysis. Chemico-biological Interactions. 2021; 336: 109365.
[8]
Sukanya JS, Raj PV, Thanka J. Role of HER2neu expression in gastric cancer. Indian Journal of Pathology & Microbiology. 2021; 64: 58–64.
[9]
Zhang GQ, Chen JL, Luo Y, Mathur MB, Anagnostis P, Nurmatov U, et al. Menopausal hormone therapy and women’s health: An umbrella review. PLoS Medicine. 2021; 18: e1003731.
[10]
Tan Z. Recent Advances in the Surgical Treatment of Advanced Gastric Cancer: A Review. Medical Science Monitor: International Medical Journal of Experimental and Clinical Research. 2019; 25: 3537–3541.
[11]
Song Z, Wu Y, Yang J, Yang D, Fang X. Progress in the treatment of advanced gastric cancer. Tumour Biology: the Journal of the International Society for Oncodevelopmental Biology and Medicine. 2017; 39: 1010428317714626.
[12]
Patel TH, Cecchini M. Targeted Therapies in Advanced Gastric Cancer. Current Treatment Options in Oncology. 2020; 21: 70.
[13]
Xia C, Dong X, Li H, Cao M, Sun D, He S, et al. Cancer statistics in China and United States, 2022: profiles, trends, and determinants. Chinese Medical Journal. 2022; 135: 584–590.
[14]
Dongre A, Weinberg RA. New insights into the mechanisms of epithelial-mesenchymal transition and implications for cancer. Nature Reviews. Molecular Cell Biology. 2019; 20: 69–84.
[15]
Taki M, Abiko K, Ukita M, Murakami R, Yamanoi K, Yamaguchi K, et al. Tumor Immune Microenvironment during Epithelial-Mesenchymal Transition. Clinical Cancer Research. 2021; 27: 4669–4679.
[16]
Tian S, Peng P, Li J, Deng H, Zhan N, Zeng Z, et al. SERPINH1 regulates EMT and gastric cancer metastasis via the Wnt/β-catenin signaling pathway. Aging. 2020; 12: 3574–3593.
[17]
Song C, Zhou C. HOXA10 mediates epithelial-mesenchymal transition to promote gastric cancer metastasis partly via modulation of TGFB2/Smad/METTL3 signaling axis. Journal of Experimental & Clinical Cancer Research. 2021; 40: 62.
[18]
Pinto MP, Owen GI, Retamal I, Garrido M. Angiogenesis inhibitors in early development for gastric cancer. Expert Opinion on Investigational Drugs. 2017; 26: 1007–1017.
[19]
Park DJ, Thomas NJ, Yoon C, Yoon SS. Vascular endothelial growth factor a inhibition in gastric cancer. Gastric Cancer. 2015; 18: 33–42.
[20]
Liu L, Ma XL, Xiao ZL, Li M, Cheng SH, Wei YQ. Prognostic value of vascular endothelial growth factor expression in resected gastric cancer. Asian Pacific Journal of Cancer Prevention. 2012; 13: 3089–3097.
[21]
Liu M, Xu W, Su M, Fan P. REC8 suppresses tumor angiogenesis by inhibition of NF-κB-mediated vascular endothelial growth factor expression in gastric cancer cells. Biological Research. 2020; 53: 41.
[22]
Chen G, Zhou Z, Jin J, Zhou Y, Liu Y, Wang W. CXCR4 is a prognostic marker that inhibits the invasion and migration of gastric cancer by regulating VEGF expression. Oncology Letters. 2021; 22: 587.
[23]
Liberzon A, Birger C, Thorvaldsdóttir H, Ghandi M, Mesirov JP, Tamayo P. The Molecular Signatures Database (MSigDB) hallmark gene set collection. Cell Systems. 2015; 1: 417–425.
[24]
Thrift AP, El-Serag HB. Burden of Gastric Cancer. Clinical Gastroenterology and Hepatology. 2020; 18: 534–542.
[25]
Nakayama I, Takahari D. The Role of Angiogenesis Targeted Therapies in Metastatic Advanced Gastric Cancer: A Narrative Review. Journal of Clinical Medicine. 2023; 12: 3226.
[26]
Cassier PA, Navaridas R, Bellina M, Rama N, Ducarouge B, Hernandez-Vargas H, et al. Netrin-1 blockade inhibits tumour growth and EMT features in endometrial cancer. Nature. 2023; 620: 409–416.
[27]
Guo J, Li Z, Ma Q, Li M, Zhao Y, Li B, et al. Dextran Sulfate Inhibits Angiogenesis and Invasion of Gastric Cancer by Interfering with M2-type Macrophages Polarization. Current Cancer Drug Targets. 2022; 22: 904–918.
[28]
Deng L, Zou J, Su Y, Wang M, Zhao L. Resveratrol inhibits TGF-β1-induced EMT in gastric cancer cells through Hippo-YAP signaling pathway. Clinical & Translational Oncology. 2022; 24: 2210–2221.
[29]
Li H, Xu L, Li C, Zhao L, Ma Y, Zheng H, et al. Ubiquitin ligase Cbl-b represses IGF-I-induced epithelial mesenchymal transition via ZEB2 and microRNA-200c regulation in gastric cancer cells. Molecular Cancer. 2014; 13: 136.
[30]
Syed V. TGF-β Signaling in Cancer. Journal of Cellular Biochemistry. 2016; 117: 1279–1287.
[31]
Rwibasira Rudinga G, Khan GJ, Kong Y. Protease-Activated Receptor 4 (PAR4): A Promising Target for Antiplatelet Therapy. International Journal of Molecular Sciences. 2018; 19: 573.
[32]
Gratio V, Walker F, Lehy T, Laburthe M, Darmoul D. Aberrant expression of proteinase-activated receptor 4 promotes colon cancer cell proliferation through a persistent signaling that involves Src and ErbB-2 kinase. International Journal of Cancer. 2009; 124: 1517–1525.
[33]
Zhang Y, Schöttker B, Ordóñez-Mena J, Holleczek B, Yang R, Burwinkel B, et al. F2RL3 methylation, lung cancer incidence and mortality. International Journal of Cancer. 2015; 137: 1739–1748.
[34]
Tantoh DM, Wu MC, Chuang CC, Chen PH, Tyan YS, Nfor ON, et al. AHRR cg05575921 methylation in relation to smoking and PM2.5 exposure among Taiwanese men and women. Clinical Epigenetics. 2020; 12: 117.
[35]
Shah S, Brock EJ, Ji K, Mattingly RR. Ras and Rap1: A tale of two GTPases. Seminars in Cancer Biology. 2019; 54: 29–39.
[36]
Chrzanowska-Wodnicka M. Rap1 in endothelial biology. Current Opinion in Hematology. 2017; 24: 248–255.
[37]
Mochizuki N, Ohba Y, Kiyokawa E, Kurata T, Murakami T, Ozaki T, et al. Activation of the ERK/MAPK pathway by an isoform of rap1GAP associated with G alpha(i). Nature. 1999; 400: 891–894.
[38]
Hattori M, Minato N. Rap1 GTPase: functions, regulation, and malignancy. Journal of Biochemistry. 2003; 134: 479–484.
[39]
Yang Y, Zhang J, Yan Y, Cai H, Li M, Sun K, et al. Low expression of Rap1GAP is associated with epithelial-mesenchymal transition (EMT) and poor prognosis in gastric cancer. Oncotarget. 2017; 8: 8057–8068.
[40]
Li H, Liang J, Wang J, Han J, Li S, Huang K, et al. Mex3a promotes oncogenesis through the RAP1/MAPK signaling pathway in colorectal cancer and is inhibited by hsa-miR-6887-3p. Cancer Communications. 2021; 41: 472–491.

Publisher’s Note: IMR Press stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share
Back to top