Cargando…

Caseinolytic protease P (CLPP) activated by ONC201 inhibits proliferation and promotes apoptosis in human epithelial ovarian cancer cells by inducing mitochondrial dysfunction

BACKGROUND: Caseinolytic protease P (CLPP) is a mitochondrial specific protein which has been reported to be related to tumor cell apoptosis. This study aims to explore the roles of CLPP in human epithelial ovarian cancer (EOC). METHODS: We determined CLPP expression in paracancerous tissues and can...

Descripción completa

Detalles Bibliográficos
Autores principales: Kou, Xinxin, Ding, Hui, Li, Lei, Chao, Hongtu
Formato: Online Artículo Texto
Lenguaje:English
Publicado: AME Publishing Company 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8506775/
https://www.ncbi.nlm.nih.gov/pubmed/34734015
http://dx.doi.org/10.21037/atm-21-4321
_version_ 1784581758422876160
author Kou, Xinxin
Ding, Hui
Li, Lei
Chao, Hongtu
author_facet Kou, Xinxin
Ding, Hui
Li, Lei
Chao, Hongtu
author_sort Kou, Xinxin
collection PubMed
description BACKGROUND: Caseinolytic protease P (CLPP) is a mitochondrial specific protein which has been reported to be related to tumor cell apoptosis. This study aims to explore the roles of CLPP in human epithelial ovarian cancer (EOC). METHODS: We determined CLPP expression in paracancerous tissues and cancer tissues obtained from 20 EOC patients, and also in 4 EOC cell lines, and one normal ovarian cell line (IOSE-80). We knocked CLPP expression down in SK-OV-3 and A2780 cells and overexpressed it in SW626 and OVcar3 cells. The effect of CLPP expression on cell proliferation, mitochondrial membrane potential, and apoptosis was then assessed by flow cytometry assay. Furthermore, the effect of ONC201 (agonist of CLPP) on the EOC cell lines was also investigated. RESULTS: The CLPP expression was markedly down-regulated in EOC cancer tissues, and the Kaplan-Meier Plotter database revealed its low expression was linked to poor prognosis in EOC patients. Low expression of CLPP up-regulated the expression of NADH: ubiquinone oxidoreductase subunit A12 (NDUFA12), succinate dehydrogenase complex flavoprotein subunit A (SDHA), and succinate dehydrogenase complex iron sulfur subunit B (SDHB), which are key members of the mitochondrial respiratory chain, and these up-regulated proteins further led to the increase of mitochondrial membrane potential, cell proliferation promotion and neoplasm metastasis. Conversely, while overexpression of CLPP led to the opposite results, including inducing the decrease of mitochondrial membrane potential and apoptosis. In addition, stimulation with ONC201 enhanced the function of CLPP in SW626 and OVcar3 cells, and silencing of CLPP could neutralize the effect of ONC201. CONCLUSIONS: Our findings suggest that CLPP mediated mitochondrial dysfunction inhibits the proliferation and migration of EOC cells.
format Online
Article
Text
id pubmed-8506775
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher AME Publishing Company
record_format MEDLINE/PubMed
spelling pubmed-85067752021-11-02 Caseinolytic protease P (CLPP) activated by ONC201 inhibits proliferation and promotes apoptosis in human epithelial ovarian cancer cells by inducing mitochondrial dysfunction Kou, Xinxin Ding, Hui Li, Lei Chao, Hongtu Ann Transl Med Original Article BACKGROUND: Caseinolytic protease P (CLPP) is a mitochondrial specific protein which has been reported to be related to tumor cell apoptosis. This study aims to explore the roles of CLPP in human epithelial ovarian cancer (EOC). METHODS: We determined CLPP expression in paracancerous tissues and cancer tissues obtained from 20 EOC patients, and also in 4 EOC cell lines, and one normal ovarian cell line (IOSE-80). We knocked CLPP expression down in SK-OV-3 and A2780 cells and overexpressed it in SW626 and OVcar3 cells. The effect of CLPP expression on cell proliferation, mitochondrial membrane potential, and apoptosis was then assessed by flow cytometry assay. Furthermore, the effect of ONC201 (agonist of CLPP) on the EOC cell lines was also investigated. RESULTS: The CLPP expression was markedly down-regulated in EOC cancer tissues, and the Kaplan-Meier Plotter database revealed its low expression was linked to poor prognosis in EOC patients. Low expression of CLPP up-regulated the expression of NADH: ubiquinone oxidoreductase subunit A12 (NDUFA12), succinate dehydrogenase complex flavoprotein subunit A (SDHA), and succinate dehydrogenase complex iron sulfur subunit B (SDHB), which are key members of the mitochondrial respiratory chain, and these up-regulated proteins further led to the increase of mitochondrial membrane potential, cell proliferation promotion and neoplasm metastasis. Conversely, while overexpression of CLPP led to the opposite results, including inducing the decrease of mitochondrial membrane potential and apoptosis. In addition, stimulation with ONC201 enhanced the function of CLPP in SW626 and OVcar3 cells, and silencing of CLPP could neutralize the effect of ONC201. CONCLUSIONS: Our findings suggest that CLPP mediated mitochondrial dysfunction inhibits the proliferation and migration of EOC cells. AME Publishing Company 2021-09 /pmc/articles/PMC8506775/ /pubmed/34734015 http://dx.doi.org/10.21037/atm-21-4321 Text en 2021 Annals of Translational Medicine. All rights reserved. https://creativecommons.org/licenses/by-nc-nd/4.0/Open Access Statement: This is an Open Access article distributed in accordance with the Creative Commons Attribution-NonCommercial-NoDerivs 4.0 International License (CC BY-NC-ND 4.0), which permits the non-commercial replication and distribution of the article with the strict proviso that no changes or edits are made and the original work is properly cited (including links to both the formal publication through the relevant DOI and the license). See: https://creativecommons.org/licenses/by-nc-nd/4.0 (https://creativecommons.org/licenses/by-nc-nd/4.0/) .
spellingShingle Original Article
Kou, Xinxin
Ding, Hui
Li, Lei
Chao, Hongtu
Caseinolytic protease P (CLPP) activated by ONC201 inhibits proliferation and promotes apoptosis in human epithelial ovarian cancer cells by inducing mitochondrial dysfunction
title Caseinolytic protease P (CLPP) activated by ONC201 inhibits proliferation and promotes apoptosis in human epithelial ovarian cancer cells by inducing mitochondrial dysfunction
title_full Caseinolytic protease P (CLPP) activated by ONC201 inhibits proliferation and promotes apoptosis in human epithelial ovarian cancer cells by inducing mitochondrial dysfunction
title_fullStr Caseinolytic protease P (CLPP) activated by ONC201 inhibits proliferation and promotes apoptosis in human epithelial ovarian cancer cells by inducing mitochondrial dysfunction
title_full_unstemmed Caseinolytic protease P (CLPP) activated by ONC201 inhibits proliferation and promotes apoptosis in human epithelial ovarian cancer cells by inducing mitochondrial dysfunction
title_short Caseinolytic protease P (CLPP) activated by ONC201 inhibits proliferation and promotes apoptosis in human epithelial ovarian cancer cells by inducing mitochondrial dysfunction
title_sort caseinolytic protease p (clpp) activated by onc201 inhibits proliferation and promotes apoptosis in human epithelial ovarian cancer cells by inducing mitochondrial dysfunction
topic Original Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8506775/
https://www.ncbi.nlm.nih.gov/pubmed/34734015
http://dx.doi.org/10.21037/atm-21-4321
work_keys_str_mv AT kouxinxin caseinolyticproteasepclppactivatedbyonc201inhibitsproliferationandpromotesapoptosisinhumanepithelialovariancancercellsbyinducingmitochondrialdysfunction
AT dinghui caseinolyticproteasepclppactivatedbyonc201inhibitsproliferationandpromotesapoptosisinhumanepithelialovariancancercellsbyinducingmitochondrialdysfunction
AT lilei caseinolyticproteasepclppactivatedbyonc201inhibitsproliferationandpromotesapoptosisinhumanepithelialovariancancercellsbyinducingmitochondrialdysfunction
AT chaohongtu caseinolyticproteasepclppactivatedbyonc201inhibitsproliferationandpromotesapoptosisinhumanepithelialovariancancercellsbyinducingmitochondrialdysfunction