Highly Metastatic Subpopulation of TNBC Cells Has Limited Iron Metabolism and Is a Target of Iron Chelators

Excess iron is known to be a risk factor of carcinogenesis. Although iron chelators show anti-cancer effects, they have not been used successfully to treat cancer patients. Triple-negative breast cancer (TNBC) is a disease with poor prognosis without effective treatments. Thus, we aimed to evaluate...

Full description

Bibliographic Details
Main Authors: Yuze Wang, Toshiaki Ohara, Yuehua Chen, Yusuke Hamada, Chunning Li, Masayoshi Fujisawa, Teizo Yoshimura, Akihiro Matsukawa
Format: Article
Language:English
Published: MDPI AG 2023-01-01
Series:Cancers
Subjects:
Online Access:https://www.mdpi.com/2072-6694/15/2/468
_version_ 1797444768113885184
author Yuze Wang
Toshiaki Ohara
Yuehua Chen
Yusuke Hamada
Chunning Li
Masayoshi Fujisawa
Teizo Yoshimura
Akihiro Matsukawa
author_facet Yuze Wang
Toshiaki Ohara
Yuehua Chen
Yusuke Hamada
Chunning Li
Masayoshi Fujisawa
Teizo Yoshimura
Akihiro Matsukawa
author_sort Yuze Wang
collection DOAJ
description Excess iron is known to be a risk factor of carcinogenesis. Although iron chelators show anti-cancer effects, they have not been used successfully to treat cancer patients. Triple-negative breast cancer (TNBC) is a disease with poor prognosis without effective treatments. Thus, we aimed to evaluate a possibility of iron chelators as a therapy for TNBC. Deferasirox (DFX), an iron chelator, suppressed the growth of 4T1 murine TNBC cell line cells in vitro and in vivo. Lung metastasis was further significantly reduced, leading to the hypothesis that iron metabolism between metastatic and non-metastatic cells may be different. An analysis of existing database demonstrated that the expression of iron-uptake genes was significantly suppressed in TNBC cells that metastasized to lymph nodes or lungs compared to those in primary tumors. A highly metastatic clone of the murine 4T1 TNBC cells (4T1-HM) did not proliferate well under iron-rich or iron-depleted conditions by iron chelators compared to a low-metastatic clone (4T1-LM). Bulk RNA-seq analysis of RNA from 4T1-HM and 4T1-LM cells suggested that the PI3K-AKT pathway might be responsible for this difference. Indeed, DFX suppressed the proliferation via the AKT-mTOR pathway in 4T1-HM and the human MDA-MB-231 cells, a human mesenchymal-like TNBC cell line. DFX also suppressed the growth of 4T1-HM tumors in comparison to 4T1-LM tumors, and reduced lung metastases after surgical resection of primary 4T1 tumors. These results indicated, for the first time, that highly metastatic TNBC cells have limited iron metabolism, and they can be more effectively targeted by iron chelators.
first_indexed 2024-03-09T13:17:20Z
format Article
id doaj.art-4ab128025b4641d790f93d477fae144c
institution Directory Open Access Journal
issn 2072-6694
language English
last_indexed 2024-03-09T13:17:20Z
publishDate 2023-01-01
publisher MDPI AG
record_format Article
series Cancers
spelling doaj.art-4ab128025b4641d790f93d477fae144c2023-11-30T21:34:18ZengMDPI AGCancers2072-66942023-01-0115246810.3390/cancers15020468Highly Metastatic Subpopulation of TNBC Cells Has Limited Iron Metabolism and Is a Target of Iron ChelatorsYuze Wang0Toshiaki Ohara1Yuehua Chen2Yusuke Hamada3Chunning Li4Masayoshi Fujisawa5Teizo Yoshimura6Akihiro Matsukawa7Department of Pathology and Experimental Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, JapanDepartment of Pathology and Experimental Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, JapanDepartment of Pathology and Experimental Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, JapanDepartment of Pathology and Experimental Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, JapanDepartment of Pathology and Experimental Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, JapanDepartment of Pathology and Experimental Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, JapanDepartment of Pathology and Experimental Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, JapanDepartment of Pathology and Experimental Medicine, Faculty of Medicine, Dentistry and Pharmaceutical Sciences, Okayama University, Okayama 700-8558, JapanExcess iron is known to be a risk factor of carcinogenesis. Although iron chelators show anti-cancer effects, they have not been used successfully to treat cancer patients. Triple-negative breast cancer (TNBC) is a disease with poor prognosis without effective treatments. Thus, we aimed to evaluate a possibility of iron chelators as a therapy for TNBC. Deferasirox (DFX), an iron chelator, suppressed the growth of 4T1 murine TNBC cell line cells in vitro and in vivo. Lung metastasis was further significantly reduced, leading to the hypothesis that iron metabolism between metastatic and non-metastatic cells may be different. An analysis of existing database demonstrated that the expression of iron-uptake genes was significantly suppressed in TNBC cells that metastasized to lymph nodes or lungs compared to those in primary tumors. A highly metastatic clone of the murine 4T1 TNBC cells (4T1-HM) did not proliferate well under iron-rich or iron-depleted conditions by iron chelators compared to a low-metastatic clone (4T1-LM). Bulk RNA-seq analysis of RNA from 4T1-HM and 4T1-LM cells suggested that the PI3K-AKT pathway might be responsible for this difference. Indeed, DFX suppressed the proliferation via the AKT-mTOR pathway in 4T1-HM and the human MDA-MB-231 cells, a human mesenchymal-like TNBC cell line. DFX also suppressed the growth of 4T1-HM tumors in comparison to 4T1-LM tumors, and reduced lung metastases after surgical resection of primary 4T1 tumors. These results indicated, for the first time, that highly metastatic TNBC cells have limited iron metabolism, and they can be more effectively targeted by iron chelators.https://www.mdpi.com/2072-6694/15/2/468triple-negative breast canceriron metabolismiron chelatorphosphoinositide-3-kinase-protein kinaseheterogeneitymetastasis
spellingShingle Yuze Wang
Toshiaki Ohara
Yuehua Chen
Yusuke Hamada
Chunning Li
Masayoshi Fujisawa
Teizo Yoshimura
Akihiro Matsukawa
Highly Metastatic Subpopulation of TNBC Cells Has Limited Iron Metabolism and Is a Target of Iron Chelators
Cancers
triple-negative breast cancer
iron metabolism
iron chelator
phosphoinositide-3-kinase-protein kinase
heterogeneity
metastasis
title Highly Metastatic Subpopulation of TNBC Cells Has Limited Iron Metabolism and Is a Target of Iron Chelators
title_full Highly Metastatic Subpopulation of TNBC Cells Has Limited Iron Metabolism and Is a Target of Iron Chelators
title_fullStr Highly Metastatic Subpopulation of TNBC Cells Has Limited Iron Metabolism and Is a Target of Iron Chelators
title_full_unstemmed Highly Metastatic Subpopulation of TNBC Cells Has Limited Iron Metabolism and Is a Target of Iron Chelators
title_short Highly Metastatic Subpopulation of TNBC Cells Has Limited Iron Metabolism and Is a Target of Iron Chelators
title_sort highly metastatic subpopulation of tnbc cells has limited iron metabolism and is a target of iron chelators
topic triple-negative breast cancer
iron metabolism
iron chelator
phosphoinositide-3-kinase-protein kinase
heterogeneity
metastasis
url https://www.mdpi.com/2072-6694/15/2/468
work_keys_str_mv AT yuzewang highlymetastaticsubpopulationoftnbccellshaslimitedironmetabolismandisatargetofironchelators
AT toshiakiohara highlymetastaticsubpopulationoftnbccellshaslimitedironmetabolismandisatargetofironchelators
AT yuehuachen highlymetastaticsubpopulationoftnbccellshaslimitedironmetabolismandisatargetofironchelators
AT yusukehamada highlymetastaticsubpopulationoftnbccellshaslimitedironmetabolismandisatargetofironchelators
AT chunningli highlymetastaticsubpopulationoftnbccellshaslimitedironmetabolismandisatargetofironchelators
AT masayoshifujisawa highlymetastaticsubpopulationoftnbccellshaslimitedironmetabolismandisatargetofironchelators
AT teizoyoshimura highlymetastaticsubpopulationoftnbccellshaslimitedironmetabolismandisatargetofironchelators
AT akihiromatsukawa highlymetastaticsubpopulationoftnbccellshaslimitedironmetabolismandisatargetofironchelators