Cargando…

Sulconazole Induces PANoptosis by Triggering Oxidative Stress and Inhibiting Glycolysis to Increase Radiosensitivity in Esophageal Cancer

Esophageal cancer is the seventh most common cancer in the world. Although traditional treatment methods such as radiotherapy and chemotherapy have good effects, their side effects and drug resistance remain problematic. The repositioning of drug function provides new ideas for the research and deve...

Descripción completa

Detalles Bibliográficos
Autores principales: Liu, Lu-Xin, Heng, Jing-Hua, Deng, Dan-Xia, Zhao, Hui, Zheng, Zhen-Yuan, Liao, Lian-Di, Lin, Wan, Xu, Xiu-E., Li, En-Min, Xu, Li-Yan
Formato: Online Artículo Texto
Lenguaje:English
Publicado: American Society for Biochemistry and Molecular Biology 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10205543/
https://www.ncbi.nlm.nih.gov/pubmed/37076047
http://dx.doi.org/10.1016/j.mcpro.2023.100551
_version_ 1785046063458025472
author Liu, Lu-Xin
Heng, Jing-Hua
Deng, Dan-Xia
Zhao, Hui
Zheng, Zhen-Yuan
Liao, Lian-Di
Lin, Wan
Xu, Xiu-E.
Li, En-Min
Xu, Li-Yan
author_facet Liu, Lu-Xin
Heng, Jing-Hua
Deng, Dan-Xia
Zhao, Hui
Zheng, Zhen-Yuan
Liao, Lian-Di
Lin, Wan
Xu, Xiu-E.
Li, En-Min
Xu, Li-Yan
author_sort Liu, Lu-Xin
collection PubMed
description Esophageal cancer is the seventh most common cancer in the world. Although traditional treatment methods such as radiotherapy and chemotherapy have good effects, their side effects and drug resistance remain problematic. The repositioning of drug function provides new ideas for the research and development of anticancer drugs. We previously showed that the Food and Drug Administration–approved drug sulconazole can effectively inhibit the growth of esophageal cancer cells, but its molecular mechanism is not clear. Here, our study demonstrated that sulconazole had a broad spectrum of anticancer effects. It can not only inhibit the proliferation but also inhibit the migration of esophageal cancer cells. Both transcriptomic sequencing and proteomic sequencing showed that sulconazole could promote various types of programmed cell death and inhibit glycolysis and its related pathways. Experimentally, we found that sulconazole induced apoptosis, pyroptosis, necroptosis, and ferroptosis. Mechanistically, sulconazole triggered mitochondrial oxidative stress and inhibited glycolysis. Finally, we showed that low-dose sulconazole can increase radiosensitivity of esophageal cancer cells. Taken together, these new findings provide strong laboratory evidence for the clinical application of sulconazole in esophageal cancer.
format Online
Article
Text
id pubmed-10205543
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher American Society for Biochemistry and Molecular Biology
record_format MEDLINE/PubMed
spelling pubmed-102055432023-05-25 Sulconazole Induces PANoptosis by Triggering Oxidative Stress and Inhibiting Glycolysis to Increase Radiosensitivity in Esophageal Cancer Liu, Lu-Xin Heng, Jing-Hua Deng, Dan-Xia Zhao, Hui Zheng, Zhen-Yuan Liao, Lian-Di Lin, Wan Xu, Xiu-E. Li, En-Min Xu, Li-Yan Mol Cell Proteomics Research Esophageal cancer is the seventh most common cancer in the world. Although traditional treatment methods such as radiotherapy and chemotherapy have good effects, their side effects and drug resistance remain problematic. The repositioning of drug function provides new ideas for the research and development of anticancer drugs. We previously showed that the Food and Drug Administration–approved drug sulconazole can effectively inhibit the growth of esophageal cancer cells, but its molecular mechanism is not clear. Here, our study demonstrated that sulconazole had a broad spectrum of anticancer effects. It can not only inhibit the proliferation but also inhibit the migration of esophageal cancer cells. Both transcriptomic sequencing and proteomic sequencing showed that sulconazole could promote various types of programmed cell death and inhibit glycolysis and its related pathways. Experimentally, we found that sulconazole induced apoptosis, pyroptosis, necroptosis, and ferroptosis. Mechanistically, sulconazole triggered mitochondrial oxidative stress and inhibited glycolysis. Finally, we showed that low-dose sulconazole can increase radiosensitivity of esophageal cancer cells. Taken together, these new findings provide strong laboratory evidence for the clinical application of sulconazole in esophageal cancer. American Society for Biochemistry and Molecular Biology 2023-04-17 /pmc/articles/PMC10205543/ /pubmed/37076047 http://dx.doi.org/10.1016/j.mcpro.2023.100551 Text en © 2023 The Authors https://creativecommons.org/licenses/by/4.0/This is an open access article under the CC BY license (http://creativecommons.org/licenses/by/4.0/).
spellingShingle Research
Liu, Lu-Xin
Heng, Jing-Hua
Deng, Dan-Xia
Zhao, Hui
Zheng, Zhen-Yuan
Liao, Lian-Di
Lin, Wan
Xu, Xiu-E.
Li, En-Min
Xu, Li-Yan
Sulconazole Induces PANoptosis by Triggering Oxidative Stress and Inhibiting Glycolysis to Increase Radiosensitivity in Esophageal Cancer
title Sulconazole Induces PANoptosis by Triggering Oxidative Stress and Inhibiting Glycolysis to Increase Radiosensitivity in Esophageal Cancer
title_full Sulconazole Induces PANoptosis by Triggering Oxidative Stress and Inhibiting Glycolysis to Increase Radiosensitivity in Esophageal Cancer
title_fullStr Sulconazole Induces PANoptosis by Triggering Oxidative Stress and Inhibiting Glycolysis to Increase Radiosensitivity in Esophageal Cancer
title_full_unstemmed Sulconazole Induces PANoptosis by Triggering Oxidative Stress and Inhibiting Glycolysis to Increase Radiosensitivity in Esophageal Cancer
title_short Sulconazole Induces PANoptosis by Triggering Oxidative Stress and Inhibiting Glycolysis to Increase Radiosensitivity in Esophageal Cancer
title_sort sulconazole induces panoptosis by triggering oxidative stress and inhibiting glycolysis to increase radiosensitivity in esophageal cancer
topic Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10205543/
https://www.ncbi.nlm.nih.gov/pubmed/37076047
http://dx.doi.org/10.1016/j.mcpro.2023.100551
work_keys_str_mv AT liuluxin sulconazoleinducespanoptosisbytriggeringoxidativestressandinhibitingglycolysistoincreaseradiosensitivityinesophagealcancer
AT hengjinghua sulconazoleinducespanoptosisbytriggeringoxidativestressandinhibitingglycolysistoincreaseradiosensitivityinesophagealcancer
AT dengdanxia sulconazoleinducespanoptosisbytriggeringoxidativestressandinhibitingglycolysistoincreaseradiosensitivityinesophagealcancer
AT zhaohui sulconazoleinducespanoptosisbytriggeringoxidativestressandinhibitingglycolysistoincreaseradiosensitivityinesophagealcancer
AT zhengzhenyuan sulconazoleinducespanoptosisbytriggeringoxidativestressandinhibitingglycolysistoincreaseradiosensitivityinesophagealcancer
AT liaoliandi sulconazoleinducespanoptosisbytriggeringoxidativestressandinhibitingglycolysistoincreaseradiosensitivityinesophagealcancer
AT linwan sulconazoleinducespanoptosisbytriggeringoxidativestressandinhibitingglycolysistoincreaseradiosensitivityinesophagealcancer
AT xuxiue sulconazoleinducespanoptosisbytriggeringoxidativestressandinhibitingglycolysistoincreaseradiosensitivityinesophagealcancer
AT lienmin sulconazoleinducespanoptosisbytriggeringoxidativestressandinhibitingglycolysistoincreaseradiosensitivityinesophagealcancer
AT xuliyan sulconazoleinducespanoptosisbytriggeringoxidativestressandinhibitingglycolysistoincreaseradiosensitivityinesophagealcancer