Cargando…

Ferroptosis induced by iron overload promotes fibrosis in ovarian endometriosis and is related to subpopulations of endometrial stromal cells

Endometriosis (EMs) is defined as the presence of tissue somewhat resembling endometrial glands and stroma outside the uterus; the retrograded endometrium grows in the peritoneal cavity and elicits fibrosis. Ferroptosis is a recently discovered form of programmed cell death, which is iron-dependent....

Descripción completa

Detalles Bibliográficos
Autores principales: Zhang, Yanqin, Liu, Xinyu, Deng, Mengqi, Xu, Chunyu, Zhang, Yubo, Wu, Di, Tang, Fan, Yang, Ruiye, Miao, Jinwei
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9478936/
https://www.ncbi.nlm.nih.gov/pubmed/36120348
http://dx.doi.org/10.3389/fphar.2022.930614
_version_ 1784790683690729472
author Zhang, Yanqin
Liu, Xinyu
Deng, Mengqi
Xu, Chunyu
Zhang, Yubo
Wu, Di
Tang, Fan
Yang, Ruiye
Miao, Jinwei
author_facet Zhang, Yanqin
Liu, Xinyu
Deng, Mengqi
Xu, Chunyu
Zhang, Yubo
Wu, Di
Tang, Fan
Yang, Ruiye
Miao, Jinwei
author_sort Zhang, Yanqin
collection PubMed
description Endometriosis (EMs) is defined as the presence of tissue somewhat resembling endometrial glands and stroma outside the uterus; the retrograded endometrium grows in the peritoneal cavity and elicits fibrosis. Ferroptosis is a recently discovered form of programmed cell death, which is iron-dependent. The induction of ferroptosis has been found to participate in fibrosis. However, the relationship between EMs fibrosis and ferroptosis remains unknown. In this study, we confirmed that the iron content in ectopic stromal tissue in ovarian EMs is significantly increased. We explored the role of iron-induced ferroptosis in the pathogenesis of ovarian EMs fibrosis for the first time. We found that ferroptosis in ectopic tissues was significantly enhanced than that in eutopic tissues. Furthermore, we performed in vivo drug screening and found that ferroptosis induced by ferric ammonium citrate (FAC) could aggravate fibrosis. To clarify the mechanism of this process, the stromal composition of human uterine endometrium and endometrial tissue was characterized. Fibroblast-specific protein-1 was used for fibroblasts, smooth muscle actin alpha for myofibroblasts, and platelet-derived growth factor receptor beta (CD140b) for mesenchymal stromal cells (MSCs). The results demonstrated that the percentage of myofibroblasts was higher and the portion of MSCs was lower in ectopic endometrial stroma than those in eutopic endometrium. Moreover, the proportion of MSCs decreased significantly and the percentage of myofibroblasts increased considerably after FAC treatment in vitro. However, disruption of intracellular iron levels or ferroptosis via chelation of intracellular iron deferoxamine mesylate or ferroptosis inhibitor ferrostatin-1 could reverse this process, indicating that iron-induced ferroptosis plays a vital role in ovarian EMs fibrosis. Considering that iron accumulation can feed the Fenton reaction to generate unquenchable amounts of free radicals, causing ferroptosis and tissue damage and thereby contributing to fibrosis, we validated the underlying mechanism that excess iron can facilitate fibrotic responses. Collectively, these data provide evidence that supernumerary iron is a key regulator in promoting MSCs ferroptosis and inducing ovarian EMs fibrosis.
format Online
Article
Text
id pubmed-9478936
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-94789362022-09-17 Ferroptosis induced by iron overload promotes fibrosis in ovarian endometriosis and is related to subpopulations of endometrial stromal cells Zhang, Yanqin Liu, Xinyu Deng, Mengqi Xu, Chunyu Zhang, Yubo Wu, Di Tang, Fan Yang, Ruiye Miao, Jinwei Front Pharmacol Pharmacology Endometriosis (EMs) is defined as the presence of tissue somewhat resembling endometrial glands and stroma outside the uterus; the retrograded endometrium grows in the peritoneal cavity and elicits fibrosis. Ferroptosis is a recently discovered form of programmed cell death, which is iron-dependent. The induction of ferroptosis has been found to participate in fibrosis. However, the relationship between EMs fibrosis and ferroptosis remains unknown. In this study, we confirmed that the iron content in ectopic stromal tissue in ovarian EMs is significantly increased. We explored the role of iron-induced ferroptosis in the pathogenesis of ovarian EMs fibrosis for the first time. We found that ferroptosis in ectopic tissues was significantly enhanced than that in eutopic tissues. Furthermore, we performed in vivo drug screening and found that ferroptosis induced by ferric ammonium citrate (FAC) could aggravate fibrosis. To clarify the mechanism of this process, the stromal composition of human uterine endometrium and endometrial tissue was characterized. Fibroblast-specific protein-1 was used for fibroblasts, smooth muscle actin alpha for myofibroblasts, and platelet-derived growth factor receptor beta (CD140b) for mesenchymal stromal cells (MSCs). The results demonstrated that the percentage of myofibroblasts was higher and the portion of MSCs was lower in ectopic endometrial stroma than those in eutopic endometrium. Moreover, the proportion of MSCs decreased significantly and the percentage of myofibroblasts increased considerably after FAC treatment in vitro. However, disruption of intracellular iron levels or ferroptosis via chelation of intracellular iron deferoxamine mesylate or ferroptosis inhibitor ferrostatin-1 could reverse this process, indicating that iron-induced ferroptosis plays a vital role in ovarian EMs fibrosis. Considering that iron accumulation can feed the Fenton reaction to generate unquenchable amounts of free radicals, causing ferroptosis and tissue damage and thereby contributing to fibrosis, we validated the underlying mechanism that excess iron can facilitate fibrotic responses. Collectively, these data provide evidence that supernumerary iron is a key regulator in promoting MSCs ferroptosis and inducing ovarian EMs fibrosis. Frontiers Media S.A. 2022-09-02 /pmc/articles/PMC9478936/ /pubmed/36120348 http://dx.doi.org/10.3389/fphar.2022.930614 Text en Copyright © 2022 Zhang, Liu, Deng, Xu, Zhang, Wu, Tang, Yang and Miao. 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
Zhang, Yanqin
Liu, Xinyu
Deng, Mengqi
Xu, Chunyu
Zhang, Yubo
Wu, Di
Tang, Fan
Yang, Ruiye
Miao, Jinwei
Ferroptosis induced by iron overload promotes fibrosis in ovarian endometriosis and is related to subpopulations of endometrial stromal cells
title Ferroptosis induced by iron overload promotes fibrosis in ovarian endometriosis and is related to subpopulations of endometrial stromal cells
title_full Ferroptosis induced by iron overload promotes fibrosis in ovarian endometriosis and is related to subpopulations of endometrial stromal cells
title_fullStr Ferroptosis induced by iron overload promotes fibrosis in ovarian endometriosis and is related to subpopulations of endometrial stromal cells
title_full_unstemmed Ferroptosis induced by iron overload promotes fibrosis in ovarian endometriosis and is related to subpopulations of endometrial stromal cells
title_short Ferroptosis induced by iron overload promotes fibrosis in ovarian endometriosis and is related to subpopulations of endometrial stromal cells
title_sort ferroptosis induced by iron overload promotes fibrosis in ovarian endometriosis and is related to subpopulations of endometrial stromal cells
topic Pharmacology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9478936/
https://www.ncbi.nlm.nih.gov/pubmed/36120348
http://dx.doi.org/10.3389/fphar.2022.930614
work_keys_str_mv AT zhangyanqin ferroptosisinducedbyironoverloadpromotesfibrosisinovarianendometriosisandisrelatedtosubpopulationsofendometrialstromalcells
AT liuxinyu ferroptosisinducedbyironoverloadpromotesfibrosisinovarianendometriosisandisrelatedtosubpopulationsofendometrialstromalcells
AT dengmengqi ferroptosisinducedbyironoverloadpromotesfibrosisinovarianendometriosisandisrelatedtosubpopulationsofendometrialstromalcells
AT xuchunyu ferroptosisinducedbyironoverloadpromotesfibrosisinovarianendometriosisandisrelatedtosubpopulationsofendometrialstromalcells
AT zhangyubo ferroptosisinducedbyironoverloadpromotesfibrosisinovarianendometriosisandisrelatedtosubpopulationsofendometrialstromalcells
AT wudi ferroptosisinducedbyironoverloadpromotesfibrosisinovarianendometriosisandisrelatedtosubpopulationsofendometrialstromalcells
AT tangfan ferroptosisinducedbyironoverloadpromotesfibrosisinovarianendometriosisandisrelatedtosubpopulationsofendometrialstromalcells
AT yangruiye ferroptosisinducedbyironoverloadpromotesfibrosisinovarianendometriosisandisrelatedtosubpopulationsofendometrialstromalcells
AT miaojinwei ferroptosisinducedbyironoverloadpromotesfibrosisinovarianendometriosisandisrelatedtosubpopulationsofendometrialstromalcells