Clinical and Translational Research
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Oncol. Jun 15, 2024; 16(6): 2555-2570
Published online Jun 15, 2024. doi: 10.4251/wjgo.v16.i6.2555
N6-methyladenosine methylation regulates the tumor microenvironment of Epstein-Barr virus-associated gastric cancer
Yu Zhang, Fang Zhou, Ming-Yu Zhang, Li-Na Feng, Jia-Lun Guan, Ruo-Nan Dong, Yu-Jie Huang, Su-Hong Xia, Jia-Zhi Liao, Kai Zhao
Yu Zhang, Ming-Yu Zhang, Li-Na Feng, Jia-Lun Guan, Ruo-Nan Dong, Yu-Jie Huang, Su-Hong Xia, Jia-Zhi Liao, Kai Zhao, Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
Yu Zhang, Ming-Yu Zhang, Li-Na Feng, Jia-Lun Guan, Ruo-Nan Dong, Yu-Jie Huang, Su-Hong Xia, Jia-Zhi Liao, Kai Zhao, Institute of Liver and Gastrointestinal Diseases, Hubei Key Laboratory of Hepato-Pancreato-Biliary Diseases, Tongji Hospital of Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, Hubei Province, China
Fang Zhou, Department of Pharmacy, Wuhan Fourth Hospital, Wuhan 430030, Hubei Province, China
Co-first authors: Yu Zhang and Fang Zhou.
Co-corresponding authors: Jia-Zhi Liao and Kai Zhao.
Author contributions: Zhang Y and Zhou F contributed equally to this work; Zhang Y, Zhou F, Xia SH, Liao JZ, and Zhao K jointly conceived and designed the study; Zhang Y, Zhou F, and Zhao K designed the research investigations, conducted the experiments, and acquired and analyzed the data; Zhang MY, Feng LN, Guan JL, Dong RN, and Huang YJ conducted the experiments, acquired the data, and provided reagents; Zhang Y and Zhao K drafted the manuscript; Liao JZ revised the manuscript for important intellectual content; Liao JZ and Zhao K took equal responsibility for the study; and all authors read and approved the final manuscript.
Supported by the Sub-Project of the National Key Research and Development Program, No. 2021YFC2600263.
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Open-Access: This article is an open-access article that was selected by an in-house editor and fully peer-reviewed by external reviewers. It is distributed in accordance with the Creative Commons Attribution NonCommercial (CC BY-NC 4.0) license, which permits others to distribute, remix, adapt, build upon this work non-commercially, and license their derivative works on different terms, provided the original work is properly cited and the use is non-commercial. See: https://creativecommons.org/Licenses/by-nc/4.0/
Corresponding author: Kai Zhao, MD, PhD, Doctor, Department of Gastroenterology, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, No. 1095 Jiefang Avenue, Wuhan 430030, Hubei Province, China. zhaokai0227@126.com
Received: December 19, 2023
Revised: February 18, 2024
Accepted: April 8, 2024
Published online: June 15, 2024
Processing time: 178 Days and 11.1 Hours
Abstract
BACKGROUND

N6-methyladenosine (m6A) methylation modification exists in Epstein-Barr virus (EBV) primary infection, latency, and lytic reactivation. It also modifies EBV latent genes and lytic genes. EBV-associated gastric cancer (EBVaGC) is a distinctive molecular subtype of GC. We hypothesized EBV and m6A methylation regulators interact with each other in EBVaGC to differentiate it from other types of GC.

AIM

To investigate the mechanisms of m6A methylation regulators in EBVaGC to determine the differentiating factors from other types of GC.

METHODS

First, The Cancer Gene Atlas and Gene Expression Omnibus databases were used to analyze the expression pattern of m6A methylation regulators between EBVaGC and EBV-negative GC (EBVnGC). Second, we identified Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) functional enrichment of m6A-related differentially expressed genes. We quantified the relative abundance of immune cells and inflammatory factors in the tumor microenvironment (TME). Finally, cell counting kit-8 cell proliferation test, transwell test, and flow cytometry were used to verify the effect of insulin-like growth factor binding protein 1 (IGFBP1) in EBVaGC cell lines.

RESULTS

m6A methylation regulators were involved in the occurrence and development of EBVaGC. Compared with EBVnGC, the expression levels of m6A methylation regulators Wilms tumor 1-associated protein, RNA binding motif protein 15B, CBL proto-oncogene like 1, leucine rich pentatricopeptide repeat containing, heterogeneous nuclear ribonucleoprotein A2B1, IGFBP1, and insulin-like growth factor 2 binding protein 1 were significantly downregulated in EBVaGC (P < 0.05). The overall survival rate of EBVaGC patients with a lower expression level of IGFBP1 was significantly higher (P = 0.046). GO and KEGG functional enrichment analyses showed that the immunity pathways were significantly activated and rich in immune cell infiltration in EBVaGC. Compared with EBVnGC, the infiltration of activated CD4+ T cells, activated CD8+ T cells, monocytes, activated dendritic cells, and plasmacytoid dendritic cells were significantly upregulated in EBVaGC (P < 0.001). In EBVaGC, the expression level of proinflammatory factors interleukin (IL)-17, IL-21, and interferon-γ and immunosuppressive factor IL-10 were significantly increased (P < 0.05). In vitro experiments demonstrated that the expression level of IGFBP1 was significantly lower in an EBVaGC cell line (SNU719) than in an EBVnGC cell line (AGS) (P < 0.05). IGFBP1 overexpression significantly attenuated proliferation and migration and promoted the apoptosis levels in SNU719. Interfering IGFBP1 significantly promoted proliferation and migration and attenuated the apoptosis levels in AGS.

CONCLUSION

m6A regulators could remodel the TME of EBVaGC, which is classified as an immune-inflamed phenotype and referred to as a “hot” tumor. Among these regulators, we demonstrated that IGFBP1 affected proliferation, migration, and apoptosis.

Keywords: N6-methyladenosine methylation, Tumor microenvironment, Epstein-Barr virus, Gastric cancer, Insulin-like growth factor binding protein 1

Core Tip: In this work, we found that N6-methyladenosine methylation regulators activated the expression of immune cells and inflammatory factors in Epstein-Barr virus-associated gastric cancer (EBVaGC), which could remodel the tumor microenvironment. EBVaGC is classified as an immune-inflamed phenotype and referred to as a “hot” tumor. Insulin-like growth factor binding protein 1 affected the proliferation, migration, and apoptosis of EBVaGC. Thus, our study provided a theoretical basis for future research on the pathogenesis and therapeutic targets of EBVaGC.