Basic Study
Copyright ©The Author(s) 2024. Published by Baishideng Publishing Group Inc. All rights reserved.
World J Gastrointest Oncol. Feb 15, 2024; 16(2): 398-413
Published online Feb 15, 2024. doi: 10.4251/wjgo.v16.i2.398
Prohibitin 1 inhibits cell proliferation and induces apoptosis via the p53-mediated mitochondrial pathway in vitro
Juan-Juan Shi, Yi-Kai Wang, Mu-Qi Wang, Jiang Deng, Ning Gao, Mei Li, Ya-Ping Li, Xin Zhang, Xiao-Li Jia, Xiong-Tao Liu, Shuang-Suo Dang, Wen-Jun Wang
Juan-Juan Shi, Yi-Kai Wang, Mu-Qi Wang, Jiang Deng, Ning Gao, Mei Li, Ya-Ping Li, Xin Zhang, Xiao-Li Jia, Shuang-Suo Dang, Wen-Jun Wang, Department of Infectious Diseases, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi'an 710004, Shaanxi Province, China
Xiong-Tao Liu, Department of Operating Room, The Second Affiliated Hospital of Xi’an Jiaotong University, Xi'an 710004, Shaanxi Province, China
Co-corresponding authors: Shuang-Suo Dang and Wen-Jun Wang.
Author contributions: Dang SS, Wang WJ, and Shi JJ designed and coordinated the study; Shi JJ, Wang YK, Wang MQ, Gao N, Li M, Zhang X, and Liu XT performed the study; Shi JJ, Wang WJ, Li YP, and Jia XL acquired and analysed the data; Shi JJ wrote the manuscript; all authors were involved in the critical review of the results and have contributed to, read, and approved the final manuscript. Dang SS and Wang WJ contributed equally to this study as co-corresponding authors. There were two reasons for designating Dang SS and Wang WJ as co-corresponding authors. 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, Dang SS and Wang WJ 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.
Supported by the Key Research and Development Program of Shaanxi, No. 2021SF-227 and No. 2020SF-297; and the Natural Science Basic Research Program of Shaanxi, No. 2023-JC-YB-770.
Institutional review board statement: This study was approved by the Institutional Review Board of The Second Affiliated Hospital of Xi’an Jiaotong University and by the Institutional Review Board of the Second Affiliated Hospital of Xi’an Jiaotong University.
Conflict-of-interest statement: The authors have no conflicts of interest to declare.
Data sharing statement: No additional data are available.
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: Shuang-Suo Dang, MD, PHD, Professor, Department of Infectious Diseases, The Second Affiliated Hospital of Xi’an Jiaotong University, No. 157 Xiwu Road, Xincheng District, Xi'an 710004, Shaanxi Province, China. dang212@126.com
Received: October 27, 2023
Peer-review started: October 27, 2023
First decision: November 6, 2023
Revised: November 23, 2023
Accepted: January 9, 2024
Article in press: January 9, 2024
Published online: February 15, 2024
Abstract
BACKGROUND

Prohibitin 1 (PHB1) has been identified as an antiproliferative protein that is highly conserved and ubiquitously expressed, and it participates in a variety of essential cellular functions, including apoptosis, cell cycle regulation, proliferation, and survival. Emerging evidence indicates that PHB1 may play an important role in the progression of hepatocellular carcinoma (HCC). However, the role of PHB1 in HCC is controversial.

AIM

To investigate the effects of PHB1 on the proliferation and apoptosis of human HCC cells and the relevant mechanisms in vitro.

METHODS

HCC patients and healthy individuals were enrolled in this study according to the inclusion and exclusion criteria; then, PHB1 levels in the sera and liver tissues of these participates were determined using ELISA, RT-PCR, and immunohistochemistry. Human HepG2 and SMMC-7721 cells were transfected with the pEGFP-PHB1 plasmid and PHB1-specific shRNA (shRNA-PHB1) for 24-72 h. Cell proliferation was analysed with an MTT assay. Cell cycle progression and apoptosis were analysed using flow cytometry (FACS). The mRNA and protein expression levels of the cell cycle-related molecules p21, Cyclin A2, Cyclin E1, and CDK2 and the cell apoptosis-related molecules cytochrome C (Cyt C), p53, Bcl-2, Bax, caspase 3, and caspase 9 were measured by real-time PCR and Western blot, respectively.

RESULTS

Decreased levels of PHB1 were found in the sera and liver tissues of HCC patients compared to those of healthy individuals, and decreased PHB1 was positively correlated with low differentiation, TNM stage III-IV, and alpha-fetoprotein ≥ 400 μg/L. Overexpression of PHB1 significantly inhibited human HCC cell proliferation in a time-dependent manner. FACS revealed that the overexpression of PHB1 arrested HCC cells in the G0/G1 phase of the cell cycle and induced apoptosis. The proportion of cells in the G0/G1 phase was significantly increased and the proportion of cells in the S phase was decreased in HepG2 cells that were transfected with pEGFP-PHB1 compared with untreated control and empty vector-transfected cells. The percentage of apoptotic HepG2 cells that were transfected with pEGFP-PHB1 was 15.41% ± 1.06%, which was significantly greater than that of apoptotic control cells (3.65% ± 0.85%, P < 0.01) and empty vector-transfected cells (4.21% ± 0.52%, P < 0.01). Similar results were obtained with SMMC-7721 cells. Furthermore, the mRNA and protein expression levels of p53, p21, Bax, caspase 3, and caspase 9 were increased while the mRNA and protein expression levels of Cyclin A2, Cyclin E1, CDK2, and Bcl-2 were decreased when PHB1 was overexpressed in human HCC cells. However, when PHB1 was upregulated in human HCC cells, Cyt C expression levels were increased in the cytosol and decreased in the mitochondria, which indicated that Cyt C had been released into the cytosol. Conversely, these effects were reversed when PHB1 was knocked down.

CONCLUSION

PHB1 inhibits human HCC cell viability by arresting the cell cycle and inducing cell apoptosis via activation of the p53-mediated mitochondrial pathway.

Keywords: Prohibitin 1, Hepatocellular carcinoma cells, Apoptosis, Cell cycle, Mitochondrial pathway

Core Tip: Prohibitin 1 (PHB1) has been identified as an antiproliferative protein that may play an important role in the progression of hepatocellular carcinoma (HCC). However, the role of PHB1 in HCC is controversial. Here, we investigated the effect of PHB1 on HCC cells and the underlying molecular mechanisms. Our findings demonstrated that PHB1 is a unique tumour suppressor and that its overexpression inhibits human HCC cell viability by arresting the cell cycle and inducing cell apoptosis via activation of the p53-mediated mitochondrial pathway.