Basic Study
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Oncol. Apr 15, 2024; 16(4): 1500-1513
Published online Apr 15, 2024. doi: 10.4251/wjgo.v16.i4.1500
Transcriptome sequencing reveals novel biomarkers and immune cell infiltration in esophageal tumorigenesis
Jian-Rong Sun, Dong-Mei Chen, Rong Huang, Rui-Tao Wang, Li-Qun Jia
Jian-Rong Sun, Rong Huang, Rui-Tao Wang, School of Clinical Medicine, Beijing University of Chinese Medicine, Beijing 100029, China
Dong-Mei Chen, Li-Qun Jia, Integrated Chinese and Western Medicine Oncology, China-Japan Friendship Hospital, Beijing 100029, China
Author contributions: Sun JR and Jia LQ designed the study and modified the manuscript; Sun JR and Chen DM carried out the data extraction and statistical analysis; Sun JR, Chen DM, Huang R, and Wang RT drafted the manuscript; and all authors contributed to the article and approved the submitted version.
Supported by National Natural Foundation of China, No. 82174223; and 2019 Chinese and Western Medicine Clinical Collaborative Capacity Building Project for Major Difficult Diseases, No. 2019-ZX-005.
Institutional review board statement: The research involving human tissue samples was approved by the Institutional Ethics Committee of Cixian Cancer Hospital (2019-CXCH-19) and China-Japan Friendship Hospital (2019-190-K131).
Institutional animal care and use committee statement: All the animal procedures and experiments conducted in the current study were approved by the Animal Ethics Committee of the China-Japan Friendship Hospital (Permit No. ZRDWLL20230210).
Conflict-of-interest statement: All the authors report no relevant conflicts of interest for this article.
Data sharing statement: The data used to support the findings of this study are available from the corresponding author upon request.
ARRIVE guidelines statement: The authors have read the ARRIVE guidelines, and the manuscript was prepared and revised according to the ARRIVE guidelines.
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: Li-Qun Jia, MD, Chief Doctor, Professor, Integrated Chinese and Western Medicine Oncology, China-Japan Friendship Hospital, No. 2 Yinghua East Street, Chaoyang District, Beijing 100029, China. liqun_jia@hotmail.com
Received: November 21, 2023
Peer-review started: November 21, 2023
First decision: December 27, 2023
Revised: January 7, 2024
Accepted: February 4, 2024
Article in press: February 4, 2024
Published online: April 15, 2024
ARTICLE HIGHLIGHTS
Research background

Precancerous lesions including low-grade intraepithelial neoplasia (LGIN) and high-grade IN (HGIN) are a mandatory stage in the development of esophageal squamous cell carcinoma (ESCC). The regulatory molecules driving esophageal carcinogenesis remain unclear, which largely hampers early esophageal cancer diagnosis and treatment.

Research motivation

We aimed to establish a mouse model mimicking the multi-stage development of ESCC and then to examine the key genes involved in the development of ESCC through transcriptome sequencing. Besides, we aimed to explore the immune cell infiltration in the microenvironment associated with the multi-stage progression of ESCC. Additionally, we hope to validate our results using human samples, which may provide a novel perspective for clinical application.

Research objectives

To examine the genes and immune cell infiltration in the microenvironment associated with the multi-stage progression of ESCC to facilitate diagnosis and early intervention.

Research methods

The mouse model of esophageal tumorigenesis was established by providing 4-nitroquinoline 1-oxide (4NQO) containing water to C57BL/6 mice. The transcriptome sequencing was performed for esophageal tissues with different pathological statuses. Differentially expressed gene analysis (DEGA) and Venn diagram were used to identify key genes. Gene Ontology (GO) and Kyoto Encyclopedia of Genes and Genomes (KEGG) enrichment analysis were used to explore the biological function of genes. The CIBERSORT algorithm was used to detect the pattern of immune cell infiltration. The immunohistochemistry (IHC) was conducted to validate our results. Furthermore, the Luminex multiplex cytokines analysis was utilized to measure the serum level of cytokines in mice.

Research results

A total of 86 genes were identified as key genes in the development of ESCC. Enrichment analysis showed that these genes were mainly enriched in keratinization, epidermal cell differentiation, and interleukin (IL)-17 signaling pathways. CIBERSORT analysis showed that the esophageal carcinogenesis group had more infiltration of M0 and M1 macrophages which was then validated by IHC. Serum cytokines analysis showed that IL-1β and IL-6 were upregulated, while IL-10 was downregulated in LGIN, HGIN, and carcinoma groups. Moreover, the expression of the representative key genes, like S100a8 and Krt6b, was verified in external human samples, and the results of immunohistochemical staining were consistent with the findings in mice.

Research conclusions

We identified a set of key genes represented by S100a8 and Krt6b and investigated their potential biological functions. In addition, we found that macrophage infiltration and abnormal alterations of inflammation-associated cytokines, such as IL-1β, IL-6, and IL-10, in the peripheral blood may be closely associated with the development of ESCC.

Research perspectives

We have identified a set of key genes that are closely associated with esophageal carcinogenesis and may serve as novel biomarkers for early detection of ESCC. Additionally, the aberrant infiltration of inflammatory macrophages and dysregulation of cytokines may contribute to esophageal carcinogenesis.