Cargando…

MLN2238 exerts its anti-tumor effects via regulating ROS/JNK/mitochondrial signaling pathways in intrahepatic cholangiocarcinoma

Background: Intrahepatic Cholangiocarcinoma (iCCA) is a highly malignant tumor with limited treatment options that contributes largely to cancer-related deaths worldwide. Compared with traditional transcriptomic analysis, single-cell RNA sequencing (scRNA-seq) is emerging as a more advanced and popu...

Descripción completa

Detalles Bibliográficos
Autores principales: Xu, Hao, Xu, Guangyu, Xu, Qianhui, Xu, Chang, Zhou, Xiaohu, Bai, Yang, Yin, Lu, Ding, Yuan, Wang, Weilin
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9659592/
https://www.ncbi.nlm.nih.gov/pubmed/36386204
http://dx.doi.org/10.3389/fphar.2022.1040847
_version_ 1784830233324552192
author Xu, Hao
Xu, Guangyu
Xu, Qianhui
Xu, Chang
Zhou, Xiaohu
Bai, Yang
Yin, Lu
Ding, Yuan
Wang, Weilin
author_facet Xu, Hao
Xu, Guangyu
Xu, Qianhui
Xu, Chang
Zhou, Xiaohu
Bai, Yang
Yin, Lu
Ding, Yuan
Wang, Weilin
author_sort Xu, Hao
collection PubMed
description Background: Intrahepatic Cholangiocarcinoma (iCCA) is a highly malignant tumor with limited treatment options that contributes largely to cancer-related deaths worldwide. Compared with traditional transcriptomic analysis, single-cell RNA sequencing (scRNA-seq) is emerging as a more advanced and popular tool for the in-depth exploration of cellular diversity and molecular complexity. As a next-generation proteasome inhibitor, MLN2238 presents better pharmacodynamics, pharmacokinetics, and therapeutic responses in various cancers. However, its effects and mechanisms of action in iCCA remain unknown. Methods: iCCA tumor heterogeneity was determined based on 4,239 qualified scRNA-seq data from 10 iCCA samples. The potential biological roles of proteasome-related genes in iCCA were investigated using a pseudo-trajectory reconstruction. The effect of MLN2238 on iCCA cell proliferation was estimated using the CCK-8, EdU, and clone formation assays. Flow cytometry was used to examine the effect of added MLN2238 on cell cycle and apoptosis levels. Autophagic flux was detected using AdPlus-mCherry-GFP-LC3B cells. ROS levels and mitochondrial membrane potential were determined using DCFH-DA probing and JC-1 staining. JNK activation and mitochondrial apoptosis were observed using western blotting and immunofluorescence microscopy, respectively. Finally, we used a tumor-bearing mouse model to validate its efficacy in vivo for iCCA treatment. Results: Proteasome-related genes were dysregulated in iCCA progression and expressed at higher levels in tumor tissues. MLN2238 suppressed cell proliferation, blocked the cell cycle in the G2/M phase, promoted apoptosis, and induced cytoprotective autophagy in iCCA cells. Furthermore, MLN2238 increased ROS levels and activated the JNK signaling pathway. Inhibition of ROS and JNK activation by NAC and SP600125 significantly reversed MLN2238-induced apoptosis. MLN2238 also suppressed the growth of iCCA tumors in vivo. Conclusion: Proteasome-related genes play pivotal roles in iCCA development. MLN2238, as a proteasome inhibitor, induces apoptosis in iCCA cells through ROS/JNK/mitochondrial signaling pathways, and hence, making MLN2238 a potential therapeutic choice for iCCA.
format Online
Article
Text
id pubmed-9659592
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-96595922022-11-15 MLN2238 exerts its anti-tumor effects via regulating ROS/JNK/mitochondrial signaling pathways in intrahepatic cholangiocarcinoma Xu, Hao Xu, Guangyu Xu, Qianhui Xu, Chang Zhou, Xiaohu Bai, Yang Yin, Lu Ding, Yuan Wang, Weilin Front Pharmacol Pharmacology Background: Intrahepatic Cholangiocarcinoma (iCCA) is a highly malignant tumor with limited treatment options that contributes largely to cancer-related deaths worldwide. Compared with traditional transcriptomic analysis, single-cell RNA sequencing (scRNA-seq) is emerging as a more advanced and popular tool for the in-depth exploration of cellular diversity and molecular complexity. As a next-generation proteasome inhibitor, MLN2238 presents better pharmacodynamics, pharmacokinetics, and therapeutic responses in various cancers. However, its effects and mechanisms of action in iCCA remain unknown. Methods: iCCA tumor heterogeneity was determined based on 4,239 qualified scRNA-seq data from 10 iCCA samples. The potential biological roles of proteasome-related genes in iCCA were investigated using a pseudo-trajectory reconstruction. The effect of MLN2238 on iCCA cell proliferation was estimated using the CCK-8, EdU, and clone formation assays. Flow cytometry was used to examine the effect of added MLN2238 on cell cycle and apoptosis levels. Autophagic flux was detected using AdPlus-mCherry-GFP-LC3B cells. ROS levels and mitochondrial membrane potential were determined using DCFH-DA probing and JC-1 staining. JNK activation and mitochondrial apoptosis were observed using western blotting and immunofluorescence microscopy, respectively. Finally, we used a tumor-bearing mouse model to validate its efficacy in vivo for iCCA treatment. Results: Proteasome-related genes were dysregulated in iCCA progression and expressed at higher levels in tumor tissues. MLN2238 suppressed cell proliferation, blocked the cell cycle in the G2/M phase, promoted apoptosis, and induced cytoprotective autophagy in iCCA cells. Furthermore, MLN2238 increased ROS levels and activated the JNK signaling pathway. Inhibition of ROS and JNK activation by NAC and SP600125 significantly reversed MLN2238-induced apoptosis. MLN2238 also suppressed the growth of iCCA tumors in vivo. Conclusion: Proteasome-related genes play pivotal roles in iCCA development. MLN2238, as a proteasome inhibitor, induces apoptosis in iCCA cells through ROS/JNK/mitochondrial signaling pathways, and hence, making MLN2238 a potential therapeutic choice for iCCA. Frontiers Media S.A. 2022-10-31 /pmc/articles/PMC9659592/ /pubmed/36386204 http://dx.doi.org/10.3389/fphar.2022.1040847 Text en Copyright © 2022 Xu, Xu, Xu, Xu, Zhou, Bai, Yin, Ding and Wang. https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Pharmacology
Xu, Hao
Xu, Guangyu
Xu, Qianhui
Xu, Chang
Zhou, Xiaohu
Bai, Yang
Yin, Lu
Ding, Yuan
Wang, Weilin
MLN2238 exerts its anti-tumor effects via regulating ROS/JNK/mitochondrial signaling pathways in intrahepatic cholangiocarcinoma
title MLN2238 exerts its anti-tumor effects via regulating ROS/JNK/mitochondrial signaling pathways in intrahepatic cholangiocarcinoma
title_full MLN2238 exerts its anti-tumor effects via regulating ROS/JNK/mitochondrial signaling pathways in intrahepatic cholangiocarcinoma
title_fullStr MLN2238 exerts its anti-tumor effects via regulating ROS/JNK/mitochondrial signaling pathways in intrahepatic cholangiocarcinoma
title_full_unstemmed MLN2238 exerts its anti-tumor effects via regulating ROS/JNK/mitochondrial signaling pathways in intrahepatic cholangiocarcinoma
title_short MLN2238 exerts its anti-tumor effects via regulating ROS/JNK/mitochondrial signaling pathways in intrahepatic cholangiocarcinoma
title_sort mln2238 exerts its anti-tumor effects via regulating ros/jnk/mitochondrial signaling pathways in intrahepatic cholangiocarcinoma
topic Pharmacology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9659592/
https://www.ncbi.nlm.nih.gov/pubmed/36386204
http://dx.doi.org/10.3389/fphar.2022.1040847
work_keys_str_mv AT xuhao mln2238exertsitsantitumoreffectsviaregulatingrosjnkmitochondrialsignalingpathwaysinintrahepaticcholangiocarcinoma
AT xuguangyu mln2238exertsitsantitumoreffectsviaregulatingrosjnkmitochondrialsignalingpathwaysinintrahepaticcholangiocarcinoma
AT xuqianhui mln2238exertsitsantitumoreffectsviaregulatingrosjnkmitochondrialsignalingpathwaysinintrahepaticcholangiocarcinoma
AT xuchang mln2238exertsitsantitumoreffectsviaregulatingrosjnkmitochondrialsignalingpathwaysinintrahepaticcholangiocarcinoma
AT zhouxiaohu mln2238exertsitsantitumoreffectsviaregulatingrosjnkmitochondrialsignalingpathwaysinintrahepaticcholangiocarcinoma
AT baiyang mln2238exertsitsantitumoreffectsviaregulatingrosjnkmitochondrialsignalingpathwaysinintrahepaticcholangiocarcinoma
AT yinlu mln2238exertsitsantitumoreffectsviaregulatingrosjnkmitochondrialsignalingpathwaysinintrahepaticcholangiocarcinoma
AT dingyuan mln2238exertsitsantitumoreffectsviaregulatingrosjnkmitochondrialsignalingpathwaysinintrahepaticcholangiocarcinoma
AT wangweilin mln2238exertsitsantitumoreffectsviaregulatingrosjnkmitochondrialsignalingpathwaysinintrahepaticcholangiocarcinoma