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): 1465-1478
Published online Apr 15, 2024. doi: 10.4251/wjgo.v16.i4.1465
Human β-defensin-1 affects the mammalian target of rapamycin pathway and autophagy in colon cancer cells through long non-coding RNA TCONS_00014506
Yu-Xin Zhao, Yan Cui, Xin-Hong Li, Wen-Hong Yang, Shi-Xiang An, Jia-Xian Cui, Min-Yu Zhang, Jing-Kun Lu, Xuan Zhang, Xiu-Mei Wang, Li-Li Bao, Peng-Wei Zhao
Yu-Xin Zhao, Department of Anesthesiology, Inner Mongolia Chest Hospital, The Fourth Hospital, Hohhot 010035, Inner Mongolia Autonomous Region, China
Yan Cui, College of Humanities and Education, Inner Mongolia Medical University, Hohhot 010059, Inner Mongolia Autonomous Region, China
Xin-Hong Li, Department of Radiotherapy, The Affiliated Hospital of Inner Mongolia Medical University, Hohhot 010059, Inner Mongolia Autonomous Region, China
Wen-Hong Yang, Shi-Xiang An, Jia-Xian Cui, Min-Yu Zhang, Jing-Kun Lu, Xuan Zhang, Li-Li Bao, Peng-Wei Zhao, School of Basic Medical Science, Inner Mongolia Medical University, Hohhot 010059, Inner Mongolia Autonomous Region, China
Xiu-Mei Wang, Department of Medical Oncology, The Affiliated Cancer Hospital of Inner Mongolia Medical University, Hohhot 010050, Inner Mongolia Autonomous Region, China
Co-first authors: Yu-Xin Zhao and Yan Cui.
Co-corresponding authors: Li-Li Bao and Peng-Wei Zhao.
Author contributions: Zhao YX and Zhao PW wrote this article; Bao LL and Zhao PW designed, organized, and reviewed this article; Li XH, Yang WH, and An SX collected the data; all authors have read and agreed to the published version of the manuscript. Zhao YX and Cui Y contributed equally to this work as co-first authors. The reasons for designating Zhao YX and Cui Y as co-first authors are threefold. First, the research was performed as a collaborative effort, and the designation of co-first authorship accurately reflects the distribution of responsibilities and burdens associated with the time and effort required to complete the study and the resultant paper. Second, the overall research team encompassed authors with a variety of expertise and skills from different fields of the co-first authors best reflects this diversity. They also did many works on the test. Third, Zhao YX and Cui Y contributed efforts of equal substance throughout the research process. The choice of these researchers as co-first authors acknowledges and respects this equal contribution, while recognizing the spirit of teamwork and collaboration of this study. In summary, we believe that designating Zhao YX and Cui Y as co-first authors is fitting for our manuscript as it accurately reflects our team's collaborative spirit, equal contributions, and diversity. Bao LL and Zhao PW contributed equally to this work as co-corresponding authors. The reasons for designating Bao LL and Zhao PW as co-corresponding authors are threefold. First, the research was performed as a collaborative effort, and the designation of co-corresponding authorship accurately reflects the distribution of responsibilities and burdens associated with the time and effort required to complete the study and the resultant paper. This also ensures effective communication and management of post-submission matters, ultimately enhancing the paper's quality and reliability. Second, the overall research team encompassed authors with a variety of expertise and skills from different fields, and the designation of co-corresponding authors best reflects this diversity. This also promotes the most comprehensive and in-depth examination of the research topic, ultimately enriching readers' understanding by offering various expert perspectives. Third, Bao LL and Zhao PW contributed efforts of equal substance throughout the research process. The choice of these researchers as co-corresponding authors acknowledges and respects this equal contribution, while recognizing the spirit of teamwork and collaboration of this study. In summary, we believe that designating Bao LL and Zhao PW as co-corresponding authors is fitting for our manuscript as it accurately reflects our team's collaborative spirit, equal contributions, and diversity.
Supported by National Natural Science Foundation of China, No. 82360329; Inner Mongolia Medical University General Project, No. YKD2023MS047; Inner Mongolia Health Commission Science and Technology Plan Project, No. 202201275.
Institutional review board statement: The study was agreed by Ethics Committee of Inner Mongolia Medical University (Approval No. YKD202201151).
Informed consent statement: Our research was conducted using cell cloning, bioinformatics, and molecular biology techniques. No human samples were involved and informed consent is not required.
Conflict-of-interest statement: The authors declare no conflict of interest for this article.
Data sharing statement: All data generated or analyzes during this study are included in this published article and supplied materials.
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: Peng-Wei Zhao, FEBG, FRS, Academic Research, School of Basic Medical Science, Inner Mongolia Medical University, No. 5 Xinhua Street, Hohhot 010059, Inner Mongolia Autonomous Region, China. pengwzhao@126.com
Received: November 14, 2023
Peer-review started: November 14, 2023
First decision: January 5, 2024
Revised: January 15, 2024
Accepted: February 18, 2024
Article in press: February 18, 2024
Published online: April 15, 2024
ARTICLE HIGHLIGHTS
Research background

Colorectal cancer has a low 5-year survival rate and high mortality. Human β-defensin-1 (hBD-1) may play an integral function in the innate immune system, contributing to the recognition and destruction of cancer cells. Long non-coding RNAs (lncRNAs) are involved in the process of cell differentiation and growth.

Research motivation

There is an urgent need for innovative treatment approaches for colon cancer. Our investigation into hBD-1 has revealed its effect on autophagy in colon cancer cells. During this exploration, we hypothesized that lncRNAs might play a crucial role in influencing autophagy. Consequently, our study delves into understanding the specific functions of lncRNAs in the context of hBD-1-mediated autophagy in colon cancer cells.

Research objectives

To investigate the effect of hBD-1 on the mTOR pathway and autophagy in human colon cancer SW620 cells.

Research methods

CCK8 assay was utilized for the detection of cell proliferation and the determination of the optimal drug concentration. Cell colony formation assay was employed to assess the effect of hBD-1 on SW620 cell proliferation. Bioinformatics was used to identify lncRNAs related to the mTOR pathway, aiming to identify those of potential biological significance. Additionally, p-mTOR (Ser2448), Beclin1, and LC3II/I expression levels in SW620 cells were assessed through Western blot analysis.

Research results

hBD-1 inhibited the proliferative ability of SW620 cells, as evidenced by the reduction in the colony formation capacity of SW620 cells upon exposure to hBD-1. hBD-1 decreased the expression of p-mTOR (Ser2448) protein and increased the expression of Beclin1 and LC3II/I protein. Furthermore, bioinformatics analysis identified seven lncRNAs (2 upregulated and 5 downregulated) related to the mTOR pathway. The lncRNA TCONS_00014506 was ultimately selected. Following the inhibition of the lncRNA TCONS_00014506, exposure to hBD-1 inhibited p-mTOR (Ser2448) and promoted Beclin1 and LC3II/I protein expression.

Research conclusions

HBD-1 inhibits the mTOR pathway and promotes autophagy by upregulating the expression of the lncRNA TCONS_00014506 in SW620 cells.

Research perspectives

We for the first time found that the lncRNA TCONS_00014506 can regulate mTOR during hBD-1-affected autophagy.