Cargando…

FTH promotes the proliferation and renders the HCC cells specifically resist to ferroptosis by maintaining iron homeostasis

BACKGROUND: Ferroptosis is a newly identified type of programmed cell death, which preferentially targets iron-rich cancer cells such as hepatocellular carcinoma (HCC). Ferritin heavy chain (FTH) is a major iron storing nanocage to store redox-inactive iron, and harbors ferroxidase activity to preve...

Descripción completa

Detalles Bibliográficos
Autores principales: Hu, Wanye, Zhou, Chaoting, Jing, Qiangan, Li, Yancun, Yang, Jing, Yang, Chen, Wang, Luyang, Hu, Jiayu, Li, Huanjuan, Wang, Hairui, Yuan, Chen, Zhou, Yi, Ren, Xueying, Tong, Xiangmin, Du, Jing, Wang, Ying
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8717654/
https://www.ncbi.nlm.nih.gov/pubmed/34965856
http://dx.doi.org/10.1186/s12935-021-02420-x
_version_ 1784624577661370368
author Hu, Wanye
Zhou, Chaoting
Jing, Qiangan
Li, Yancun
Yang, Jing
Yang, Chen
Wang, Luyang
Hu, Jiayu
Li, Huanjuan
Wang, Hairui
Yuan, Chen
Zhou, Yi
Ren, Xueying
Tong, Xiangmin
Du, Jing
Wang, Ying
author_facet Hu, Wanye
Zhou, Chaoting
Jing, Qiangan
Li, Yancun
Yang, Jing
Yang, Chen
Wang, Luyang
Hu, Jiayu
Li, Huanjuan
Wang, Hairui
Yuan, Chen
Zhou, Yi
Ren, Xueying
Tong, Xiangmin
Du, Jing
Wang, Ying
author_sort Hu, Wanye
collection PubMed
description BACKGROUND: Ferroptosis is a newly identified type of programmed cell death, which preferentially targets iron-rich cancer cells such as hepatocellular carcinoma (HCC). Ferritin heavy chain (FTH) is a major iron storing nanocage to store redox-inactive iron, and harbors ferroxidase activity to prevent the iron-mediated production of ROS. Our previous studies have demonstrated that FTH acts as a protective role to increase the cellular resistance to ferroptosis. However, the specific role of FTH in the development of HCC and ferroptosis resistance remains unclear. METHODS: The indicated databases were used for bioinformatics analysis. The abilities of cell proliferation, migration were measured by cell proliferation assay, transwell assay and wound healing assay. The levels of reactive oxygen species (ROS), lipid peroxide, free iron, mitochondrial superoxide, mitochondrial morphology and mitochondrial membrane potential (MMP) were determined by DCF-DA, C11-BODIPY, mitoSOX, mitoTracker, JC-10 and TMRM staining, respectively. The mitochondrial oxygen consumption rate was monitored by the Seahorse XF24 Analyzer. RESULTS: The pan-cancer analysis was performed and showed that FTH expression is upregulated in multiple cancers, such as LIHC, CHOL, HNSC, compared to corresponding normal tissues. In addition, the level of serum ferritin is positively associated with the progression of hepatitis, cirrhosis liver and hepatocellular carcinoma. Further investigation shed light on the strong correlation between FTH expression and tumor grades, cancer stages and prognosis of HCC. Importantly, the proteins interaction network elucidated that FTH is involved in iron homeostasis maintenance and lysosomal-dependent degradation. Enforced expression of FTH accelerates proliferation, migration and endows HCC cells specifically resistant to ferroptosis, but does not protect against cell death caused by cytotoxic compounds like oxaliplatin, irinotecan, and adriamycin. Mechanically, FTH reconstituted cells exhibit diminished peroxides accumulation, reduce mitochondrial ROS level, attenuate the impaired mitochondrial respiratory and rescue the mitochondrial homeostasis. Notably, FTH expression boosts tumorigenic potential in vivo with increased PCNA staining and lesser lipid peroxides generation. CONCLUSION: These results provide new insights that FTH acts as an oncogene in the carcinogenesis and progression of HCC, and is hopeful to be a potential target for therapeutic intervention through ferroptosis. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12935-021-02420-x.
format Online
Article
Text
id pubmed-8717654
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-87176542022-01-05 FTH promotes the proliferation and renders the HCC cells specifically resist to ferroptosis by maintaining iron homeostasis Hu, Wanye Zhou, Chaoting Jing, Qiangan Li, Yancun Yang, Jing Yang, Chen Wang, Luyang Hu, Jiayu Li, Huanjuan Wang, Hairui Yuan, Chen Zhou, Yi Ren, Xueying Tong, Xiangmin Du, Jing Wang, Ying Cancer Cell Int Primary Research BACKGROUND: Ferroptosis is a newly identified type of programmed cell death, which preferentially targets iron-rich cancer cells such as hepatocellular carcinoma (HCC). Ferritin heavy chain (FTH) is a major iron storing nanocage to store redox-inactive iron, and harbors ferroxidase activity to prevent the iron-mediated production of ROS. Our previous studies have demonstrated that FTH acts as a protective role to increase the cellular resistance to ferroptosis. However, the specific role of FTH in the development of HCC and ferroptosis resistance remains unclear. METHODS: The indicated databases were used for bioinformatics analysis. The abilities of cell proliferation, migration were measured by cell proliferation assay, transwell assay and wound healing assay. The levels of reactive oxygen species (ROS), lipid peroxide, free iron, mitochondrial superoxide, mitochondrial morphology and mitochondrial membrane potential (MMP) were determined by DCF-DA, C11-BODIPY, mitoSOX, mitoTracker, JC-10 and TMRM staining, respectively. The mitochondrial oxygen consumption rate was monitored by the Seahorse XF24 Analyzer. RESULTS: The pan-cancer analysis was performed and showed that FTH expression is upregulated in multiple cancers, such as LIHC, CHOL, HNSC, compared to corresponding normal tissues. In addition, the level of serum ferritin is positively associated with the progression of hepatitis, cirrhosis liver and hepatocellular carcinoma. Further investigation shed light on the strong correlation between FTH expression and tumor grades, cancer stages and prognosis of HCC. Importantly, the proteins interaction network elucidated that FTH is involved in iron homeostasis maintenance and lysosomal-dependent degradation. Enforced expression of FTH accelerates proliferation, migration and endows HCC cells specifically resistant to ferroptosis, but does not protect against cell death caused by cytotoxic compounds like oxaliplatin, irinotecan, and adriamycin. Mechanically, FTH reconstituted cells exhibit diminished peroxides accumulation, reduce mitochondrial ROS level, attenuate the impaired mitochondrial respiratory and rescue the mitochondrial homeostasis. Notably, FTH expression boosts tumorigenic potential in vivo with increased PCNA staining and lesser lipid peroxides generation. CONCLUSION: These results provide new insights that FTH acts as an oncogene in the carcinogenesis and progression of HCC, and is hopeful to be a potential target for therapeutic intervention through ferroptosis. SUPPLEMENTARY INFORMATION: The online version contains supplementary material available at 10.1186/s12935-021-02420-x. BioMed Central 2021-12-29 /pmc/articles/PMC8717654/ /pubmed/34965856 http://dx.doi.org/10.1186/s12935-021-02420-x Text en © The Author(s) 2021 https://creativecommons.org/licenses/by/4.0/Open AccessThis article is licensed under a Creative Commons Attribution 4.0 International License, which permits use, sharing, adaptation, distribution and reproduction in any medium or format, as long as you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons licence, and indicate if changes were made. The images or other third party material in this article are included in the article's Creative Commons licence, unless indicated otherwise in a credit line to the material. If material is not included in the article's Creative Commons licence and your intended use is not permitted by statutory regulation or exceeds the permitted use, you will need to obtain permission directly from the copyright holder. To view a copy of this licence, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) . The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/ (https://creativecommons.org/publicdomain/zero/1.0/) ) applies to the data made available in this article, unless otherwise stated in a credit line to the data.
spellingShingle Primary Research
Hu, Wanye
Zhou, Chaoting
Jing, Qiangan
Li, Yancun
Yang, Jing
Yang, Chen
Wang, Luyang
Hu, Jiayu
Li, Huanjuan
Wang, Hairui
Yuan, Chen
Zhou, Yi
Ren, Xueying
Tong, Xiangmin
Du, Jing
Wang, Ying
FTH promotes the proliferation and renders the HCC cells specifically resist to ferroptosis by maintaining iron homeostasis
title FTH promotes the proliferation and renders the HCC cells specifically resist to ferroptosis by maintaining iron homeostasis
title_full FTH promotes the proliferation and renders the HCC cells specifically resist to ferroptosis by maintaining iron homeostasis
title_fullStr FTH promotes the proliferation and renders the HCC cells specifically resist to ferroptosis by maintaining iron homeostasis
title_full_unstemmed FTH promotes the proliferation and renders the HCC cells specifically resist to ferroptosis by maintaining iron homeostasis
title_short FTH promotes the proliferation and renders the HCC cells specifically resist to ferroptosis by maintaining iron homeostasis
title_sort fth promotes the proliferation and renders the hcc cells specifically resist to ferroptosis by maintaining iron homeostasis
topic Primary Research
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC8717654/
https://www.ncbi.nlm.nih.gov/pubmed/34965856
http://dx.doi.org/10.1186/s12935-021-02420-x
work_keys_str_mv AT huwanye fthpromotestheproliferationandrendersthehcccellsspecificallyresisttoferroptosisbymaintainingironhomeostasis
AT zhouchaoting fthpromotestheproliferationandrendersthehcccellsspecificallyresisttoferroptosisbymaintainingironhomeostasis
AT jingqiangan fthpromotestheproliferationandrendersthehcccellsspecificallyresisttoferroptosisbymaintainingironhomeostasis
AT liyancun fthpromotestheproliferationandrendersthehcccellsspecificallyresisttoferroptosisbymaintainingironhomeostasis
AT yangjing fthpromotestheproliferationandrendersthehcccellsspecificallyresisttoferroptosisbymaintainingironhomeostasis
AT yangchen fthpromotestheproliferationandrendersthehcccellsspecificallyresisttoferroptosisbymaintainingironhomeostasis
AT wangluyang fthpromotestheproliferationandrendersthehcccellsspecificallyresisttoferroptosisbymaintainingironhomeostasis
AT hujiayu fthpromotestheproliferationandrendersthehcccellsspecificallyresisttoferroptosisbymaintainingironhomeostasis
AT lihuanjuan fthpromotestheproliferationandrendersthehcccellsspecificallyresisttoferroptosisbymaintainingironhomeostasis
AT wanghairui fthpromotestheproliferationandrendersthehcccellsspecificallyresisttoferroptosisbymaintainingironhomeostasis
AT yuanchen fthpromotestheproliferationandrendersthehcccellsspecificallyresisttoferroptosisbymaintainingironhomeostasis
AT zhouyi fthpromotestheproliferationandrendersthehcccellsspecificallyresisttoferroptosisbymaintainingironhomeostasis
AT renxueying fthpromotestheproliferationandrendersthehcccellsspecificallyresisttoferroptosisbymaintainingironhomeostasis
AT tongxiangmin fthpromotestheproliferationandrendersthehcccellsspecificallyresisttoferroptosisbymaintainingironhomeostasis
AT dujing fthpromotestheproliferationandrendersthehcccellsspecificallyresisttoferroptosisbymaintainingironhomeostasis
AT wangying fthpromotestheproliferationandrendersthehcccellsspecificallyresisttoferroptosisbymaintainingironhomeostasis