Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 2 de 2
Filter
Add more filters










Database
Language
Publication year range
1.
Exp Cell Res ; 431(1): 113733, 2023 10 01.
Article in English | MEDLINE | ID: mdl-37517591

ABSTRACT

IRF1 is a tumor suppressor gene in colon cancer. This study aimed to explore the potential regulation of IRF1 on the ferroptosis of colon cancer and the mechanisms underlying its regulation of GPX4 transcription. IRF1 interacting transcription factors regulating GPX4 transcription were predicted and validated. The role of the IRF1/SPI1-GPX4 axis on the ferroptosis of colon cancer cells was explored. Results showed that IRF1 overexpression reduced GPX4 transcription, increased reactive oxygen species (ROS) and lipid ROS accumulation, and enhanced erastin-induced colon cancer cell growth in vitro and in vivo. SPI1 could directly bind to the GPX4 promoter (-414 to -409) and activate its transcription. IRF1 could bind to SPI1 and suppress its transcriptional activating effects on GPX4 expression. SPI1 overexpression reduced ROS and lipid ROS accumulation and increased colon cancer cell viability and colony formation upon erastin induction. These trends were reversed by IRF1 overexpression. In conclusion, this study revealed a novel oncogenic mechanism of SPI1 by reducing erastin-induced ferroptosis in colon cancer. IRF1 interacts with SPI1 and suppresses its transcriptional activating effect on GPX4 expression. Through this mechanism, IRF1 can enhance erastin-induced ferroptosis of colon cancer. The IRF1/SPI1-GPX4 axis might play a crucial role in modulating ferroptosis in colon cancer and might serve as a potential therapeutic target in the future.


Subject(s)
Colonic Neoplasms , Ferroptosis , Humans , Ferroptosis/genetics , Reactive Oxygen Species , Transcriptional Activation/genetics , Colonic Neoplasms/genetics , Cell Proliferation/genetics , Lipids , Interferon Regulatory Factor-1/genetics
2.
Cell Biol Int ; 44(5): 1224-1236, 2020 May.
Article in English | MEDLINE | ID: mdl-32068322

ABSTRACT

This work aimed to investigate miR-93-5p expression in tumor tissue and its in vitro effects in colorectal cancer (CRC) by targeting programmed death ligand-1 (PD-L1). MiR-93-5p and PD-L1 expression was detected in CRC and adjacent normal tissues by quantitative real-time polymerase chain reaction and immunohistochemistry. The correlation between miR-93-5p and PD-L1 was validated by a dual-luciferase reporter assay. HCT116 and SW480 cells were divided into blank, miR-NC, miR-93-5p mimics, miR-93-5p inhibitor, PD-L1 small interfering RNA (siRNA) and miR-93-5p inhibitor + PD-L1 siRNA groups, and wound-healing and transwell assays were performed to detect cell migration and invasion, respectively. Protein expression was measured by western blotting. The secretion of cytokines was detected in the CRC cell/T coculture models. MiR-93-5p was downregulated in CRC tissues with upregulated PD-L1. In PD-L1-negative patients, miR-93-5p expression was increased compared with that in PD-L1-positive patients. MiR-93-5p and PD-L1 expression levels were associated with the tumor differentiation, lymphatic metastasis, TNM, Duke's stage, and prognosis of CRC. PD-L1 siRNA weakened the migration and invasion abilities via decreased expression of matrix metalloproteinase-1 (MMP-1), -2, and -9, and these effects were abolished by the miR-93-5p inhibitor. Additionally, anti-PD-L1 upregulated the expressions of interleukin-2 (IL-2), tumor necrosis factor-α (TNF-α), and interferon γ (IFN-γ) in the coculture of T cells with CRC cells, but downregulated the expressions of IL-1ß, IL-10, and TGF-ß. However, these changes were partially reversed by miR-93-5p inhibition. miR-93-5p is expected to be a novel target for CRC treatment since it decreases the migration and invasion, as well as the immune evasion, of CRC cells via targeting PD-L1.


Subject(s)
B7-H1 Antigen/metabolism , Colorectal Neoplasms/metabolism , MicroRNAs/metabolism , Adult , Aged , Aged, 80 and over , Cell Movement , Cell Proliferation , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Male , Middle Aged
SELECTION OF CITATIONS
SEARCH DETAIL
...