Cargando…

Iron-Dependent Autophagic Cell Death Induced by Radiation in MDA-MB-231 Breast Cancer Cells

In radiation oncology, ionizing radiation is used to kill cancer cells, in other words, the induction of different types of cell death. To investigate this cellular death and the associated iron accumulation, the transfer, release, and participation of iron after radiation treatment was analyzed. We...

Descripción completa

Detalles Bibliográficos
Autores principales: Ma, Shumei, Fu, Xinxin, Liu, Lin, Liu, Yi, Feng, Hao, Jiang, Heya, Liu, Xiaomei, Liu, Rui, Liang, Zhenzhen, Li, Mengke, Tian, Zhujun, Hu, Boqi, Bai, Yongheng, Liang, Bing, Liu, Xiaodong
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8551752/
https://www.ncbi.nlm.nih.gov/pubmed/34722507
http://dx.doi.org/10.3389/fcell.2021.723801
_version_ 1784591230276993024
author Ma, Shumei
Fu, Xinxin
Liu, Lin
Liu, Yi
Feng, Hao
Jiang, Heya
Liu, Xiaomei
Liu, Rui
Liang, Zhenzhen
Li, Mengke
Tian, Zhujun
Hu, Boqi
Bai, Yongheng
Liang, Bing
Liu, Xiaodong
author_facet Ma, Shumei
Fu, Xinxin
Liu, Lin
Liu, Yi
Feng, Hao
Jiang, Heya
Liu, Xiaomei
Liu, Rui
Liang, Zhenzhen
Li, Mengke
Tian, Zhujun
Hu, Boqi
Bai, Yongheng
Liang, Bing
Liu, Xiaodong
author_sort Ma, Shumei
collection PubMed
description In radiation oncology, ionizing radiation is used to kill cancer cells, in other words, the induction of different types of cell death. To investigate this cellular death and the associated iron accumulation, the transfer, release, and participation of iron after radiation treatment was analyzed. We found that radiation-induced cell death varied in different breast cancer cells and autophagy was induced in MDA-MB-231 and BT549 cells (triple negative breast cancer cell line) rather than in MCF-7 and zr-75 cells. Iron chelator deferoxamine (DFO), the autophagy inhibitor 3MA, silencing of the autophagy-related genes ATG5, and Beclin 1 could decrease radiation induced cell death in MDA-MB-231 cells, while inhibitors of apoptosis such as Z-VAD-FMK, ferroptosis inhibitor ferrostatin-1 (Fer-1), and necroptosis inhibitor Necrostatin-1 showed no change. This suggests the occurrence of autophagic cell death. Furthermore, we found that iron accumulation and iron regulatory proteins, including transferrin (Tf), transferrin receptor (CD71), and Ferritin (FTH), increased after radiation treatment, and the silencing of transferrin decreased radiation-induced cell death. In addition, radiation increased lysosomal membrane permeabilization (LMP) and the release of lysosomal iron and cathepsins, while cathepsins silencing failed to change cell viability. Radiation-induced iron accumulation increased Reactive oxygen species (ROS) generation via the Fenton reaction and increased autophagy in a time-dependent manner. DFO, N-acetylcysteine (NAC), and overexpression of superoxide dismutase 2 (SOD2) decreased ROS generation, autophagy, and cell death. To summarize, for the first time, we found that radiation-induced autophagic cell death was iron-dependent in breast cancer MDA-MB-231 cells. These results provide new insights into the cell death process of cancers and might conduce to the development and application of novel therapeutic strategies for patients with apoptosis-resistant breast cancer.
format Online
Article
Text
id pubmed-8551752
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-85517522021-10-29 Iron-Dependent Autophagic Cell Death Induced by Radiation in MDA-MB-231 Breast Cancer Cells Ma, Shumei Fu, Xinxin Liu, Lin Liu, Yi Feng, Hao Jiang, Heya Liu, Xiaomei Liu, Rui Liang, Zhenzhen Li, Mengke Tian, Zhujun Hu, Boqi Bai, Yongheng Liang, Bing Liu, Xiaodong Front Cell Dev Biol Cell and Developmental Biology In radiation oncology, ionizing radiation is used to kill cancer cells, in other words, the induction of different types of cell death. To investigate this cellular death and the associated iron accumulation, the transfer, release, and participation of iron after radiation treatment was analyzed. We found that radiation-induced cell death varied in different breast cancer cells and autophagy was induced in MDA-MB-231 and BT549 cells (triple negative breast cancer cell line) rather than in MCF-7 and zr-75 cells. Iron chelator deferoxamine (DFO), the autophagy inhibitor 3MA, silencing of the autophagy-related genes ATG5, and Beclin 1 could decrease radiation induced cell death in MDA-MB-231 cells, while inhibitors of apoptosis such as Z-VAD-FMK, ferroptosis inhibitor ferrostatin-1 (Fer-1), and necroptosis inhibitor Necrostatin-1 showed no change. This suggests the occurrence of autophagic cell death. Furthermore, we found that iron accumulation and iron regulatory proteins, including transferrin (Tf), transferrin receptor (CD71), and Ferritin (FTH), increased after radiation treatment, and the silencing of transferrin decreased radiation-induced cell death. In addition, radiation increased lysosomal membrane permeabilization (LMP) and the release of lysosomal iron and cathepsins, while cathepsins silencing failed to change cell viability. Radiation-induced iron accumulation increased Reactive oxygen species (ROS) generation via the Fenton reaction and increased autophagy in a time-dependent manner. DFO, N-acetylcysteine (NAC), and overexpression of superoxide dismutase 2 (SOD2) decreased ROS generation, autophagy, and cell death. To summarize, for the first time, we found that radiation-induced autophagic cell death was iron-dependent in breast cancer MDA-MB-231 cells. These results provide new insights into the cell death process of cancers and might conduce to the development and application of novel therapeutic strategies for patients with apoptosis-resistant breast cancer. Frontiers Media S.A. 2021-10-14 /pmc/articles/PMC8551752/ /pubmed/34722507 http://dx.doi.org/10.3389/fcell.2021.723801 Text en Copyright © 2021 Ma, Fu, Liu, Liu, Feng, Jiang, Liu, Liu, Liang, Li, Tian, Hu, Bai, Liang and Liu. 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 Cell and Developmental Biology
Ma, Shumei
Fu, Xinxin
Liu, Lin
Liu, Yi
Feng, Hao
Jiang, Heya
Liu, Xiaomei
Liu, Rui
Liang, Zhenzhen
Li, Mengke
Tian, Zhujun
Hu, Boqi
Bai, Yongheng
Liang, Bing
Liu, Xiaodong
Iron-Dependent Autophagic Cell Death Induced by Radiation in MDA-MB-231 Breast Cancer Cells
title Iron-Dependent Autophagic Cell Death Induced by Radiation in MDA-MB-231 Breast Cancer Cells
title_full Iron-Dependent Autophagic Cell Death Induced by Radiation in MDA-MB-231 Breast Cancer Cells
title_fullStr Iron-Dependent Autophagic Cell Death Induced by Radiation in MDA-MB-231 Breast Cancer Cells
title_full_unstemmed Iron-Dependent Autophagic Cell Death Induced by Radiation in MDA-MB-231 Breast Cancer Cells
title_short Iron-Dependent Autophagic Cell Death Induced by Radiation in MDA-MB-231 Breast Cancer Cells
title_sort iron-dependent autophagic cell death induced by radiation in mda-mb-231 breast cancer cells
topic Cell and Developmental Biology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8551752/
https://www.ncbi.nlm.nih.gov/pubmed/34722507
http://dx.doi.org/10.3389/fcell.2021.723801
work_keys_str_mv AT mashumei irondependentautophagiccelldeathinducedbyradiationinmdamb231breastcancercells
AT fuxinxin irondependentautophagiccelldeathinducedbyradiationinmdamb231breastcancercells
AT liulin irondependentautophagiccelldeathinducedbyradiationinmdamb231breastcancercells
AT liuyi irondependentautophagiccelldeathinducedbyradiationinmdamb231breastcancercells
AT fenghao irondependentautophagiccelldeathinducedbyradiationinmdamb231breastcancercells
AT jiangheya irondependentautophagiccelldeathinducedbyradiationinmdamb231breastcancercells
AT liuxiaomei irondependentautophagiccelldeathinducedbyradiationinmdamb231breastcancercells
AT liurui irondependentautophagiccelldeathinducedbyradiationinmdamb231breastcancercells
AT liangzhenzhen irondependentautophagiccelldeathinducedbyradiationinmdamb231breastcancercells
AT limengke irondependentautophagiccelldeathinducedbyradiationinmdamb231breastcancercells
AT tianzhujun irondependentautophagiccelldeathinducedbyradiationinmdamb231breastcancercells
AT huboqi irondependentautophagiccelldeathinducedbyradiationinmdamb231breastcancercells
AT baiyongheng irondependentautophagiccelldeathinducedbyradiationinmdamb231breastcancercells
AT liangbing irondependentautophagiccelldeathinducedbyradiationinmdamb231breastcancercells
AT liuxiaodong irondependentautophagiccelldeathinducedbyradiationinmdamb231breastcancercells