Basic Study
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Oncol. Mar 15, 2024; 16(3): 1029-1045
Published online Mar 15, 2024. doi: 10.4251/wjgo.v16.i3.1029
CALD1 facilitates epithelial-mesenchymal transition progression in gastric cancer cells by modulating the PI3K-Akt pathway
Wen-Qian Ma, Ming-Chang Miao, Ping-An Ding, Bi-Bo Tan, Wen-Bo Liu, Shuo Guo, Li-Mian Er, Zhi-Dong Zhang, Qun Zhao
Wen-Qian Ma, Shuo Guo, Li-Mian Er, Department of Endoscopy, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei Province, China
Wen-Qian Ma, Ping-An Ding, Bi-Bo Tan, Wen-Bo Liu, Shuo Guo, Li-Mian Er, Zhi-Dong Zhang, Qun Zhao, Hebei Key Laboratory of Precision Diagnosis and Comprehensive Treatment of Gastric Cancer, Shijiazhuang 050011, Hebei Province, China
Ming-Chang Miao, Department of Radiotherapy, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei Province, China
Ping-An Ding, Bi-Bo Tan, Wen-Bo Liu, Zhi-Dong Zhang, Qun Zhao, The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, Shijiazhuang 050011, Hebei Province, China
Author contributions: Ma WQ, Tan BB, and Liu WB designed the study; Ma WQ and Liu WB wrote the manuscript; Ma WQ, Ding PA, Liu WB, and Guo S performed the experiments; Ma WQ, Miao MC, and Er LM analyzed the data; Zhang ZD and Zhao Q reviewed and edited the manuscript; all authors have read and approved the final manuscript.
Supported by The Hebei Provincial Major Science and Technology Special Project, No. 23297701Z; Beijing-Tianjin-Hebei Basic Research Cooperation Special Project, No. 22JCZXJC00140; Hebei Provincial Government-Funded Clinical Talent Project, No. ZF2023047.
Institutional review board statement: The study was approved by the Ethics Committee of the Fourth Hospital of Hebei Medical University.
Conflict-of-interest statement: The authors declare that they have no competing interests to disclose.
Data sharing statement: The datasets used and analyzed during the current study are available from the corresponding author on reasonable request.
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: Qun Zhao, MD, PhD, Chief Doctor, Professor, The Third Department of Surgery, The Fourth Hospital of Hebei Medical University, No. 12 Jiankang Road, Changan District, Shijiazhuang 050011, Hebei Province, China. zhaoqun@hebmu.edu.cn
Received: November 22, 2023
Peer-review started: November 22, 2023
First decision: December 8, 2023
Revised: December 21, 2023
Accepted: January 4, 2024
Article in press: January 4, 2024
Published online: March 15, 2024
Abstract
BACKGROUND

CALD1 has been discovered to be abnormally expressed in a variety of malignant tumors, including gastric cancer (GC), and is associated with tumor progression and immune infiltration; however, the roles and mechanisms of CALD1 in epithelial-mesenchymal transition (EMT) in GC are unknown.

AIM

To investigate the role and mechanism of CALD1 in GC progression, invasion, and migration.

METHODS

In this study, the relationship between CALD1 and GC, as well as the possible network regulatory mechanisms of CALD1, was investigated by bioinformatics and validated by experiments. CALD1-siRNA was synthesized and used to transfect GC cells. Cell activity was measured using the CCK-8 method, cell migration and invasive ability were measured using wound healing assay and Transwell assay, and the expression levels of relevant genes and proteins in each group of cells were measured using qRT-PCR and Western blot. A GC cell xenograft model was established to verify the results of in vitro experiments.

RESULTS

Bioinformatics results showed that CALD1 was highly expressed in GC tissues, and CALD1 was significantly higher in EMT-type GC tissues than in tissues of other types of GC. The prognosis of patients with high expression of CALD1 was worse than that of patients with low expression, and a prognostic model was constructed and evaluated. The experimental results were consistent with the results of the bioinformatics analysis. The expression level of CALD1 in GC cell lines was all higher than that in gastric epithelial cell line GES-1, with the strongest expression found in AGS and MKN45 cells. Cell activity was significantly reduced after CALD1-siRNA transfection of AGS and MKN45 cells. The ability of AGS and MKN45 cells to migrate and invade was reduced after CALD1-siRNA transfection, and the related mRNA and protein expression was altered. According to bioinformatics findings in GC samples, the CALD1 gene was significantly associated with the expression of members of the PI3K-AKT-mTOR signaling pathway as well as the EMT signaling pathway, and was closely related to the PI3K-Akt signaling pathway. Experimental validation revealed that upregulation of CALD1 increased the expression of PI3K, p-AKT, and p-mTOR, members of the PI3K-Akt pathway,while decreasing the expression of PTEN; PI3K-Akt inhibitor treatment decreased the expression of PI3K, p-AKT, and p-mTOR in cells overexpressing CALD1 (still higher than that in the normal group), but increased the expression of PTEN (still lower than that in the normal group). CCK-8 results revealed that the effect of CALD1 on tumor cell activity was decreased by the addition of the inhibitor. Scratch and Transwell experiments showed that the effect of CALD1 on tumor cell migration and invasion was weakened by the addition of the PI3K-Akt inhibitor. The mRNA and protein levels of EMT-related genes in AGS and MKN45 cells were greatly altered by the overexpression of CALD1, whereas the effect of overexpression of CALD1 was significantly weakened by the addition of the PI3K-Akt inhibitor. Animal experiments showed that tumour growth was slow after inhibition of CALD1, and the expression of some PI3K-Akt and EMT pathway proteins was altered.

CONCLUSION

Increased expression of CALD1 is a key factor in the progression, invasion, and metastasis of GC, which may be associated with regulating the PI3K-Akt pathway to promote EMT.

Keywords: Gastric tumor, CALD1, Epithelial-mesenchymal transition, Gene disruption, Invasion, Migration, Bioinformatics

Core Tip: In this study, the relationship between CALD1 and gastric cancer (GC) and the possible network regulatory mechanisms of CALD1 were explored by bioinformatics methods and validated by experiments. We conducted functional analysis and verification through tissue and cell experiments, delving into possible pathways and mechanisms involved. It was showed that CALD1 may participate in the proliferation, invasion, and migration, and epithelial-mesenchymal transition (EMT)-related gene and protein expression in GC cells. Our study suggested that CALD1, through PI3K-Akt signaling pathway activation, may regulate EMT in GC cells, representing a potentially novel target for GC treatment.