Cargando…

Chemotherapy impairs ovarian function through excessive ROS-induced ferroptosis

Chemotherapy was conventionally applied to kill cancer cells, but regrettably, they also induce damage to normal cells with high-proliferative capacity resulting in cardiotoxicity, nephrotoxicity, peripheral nerve toxicity, and ovarian toxicity. Of these, chemotherapy-induced ovarian damages mainly...

Descripción completa

Detalles Bibliográficos
Autores principales: Zhang, Shenghui, Liu, Qin, Chang, Mengyuan, Pan, Ying, Yahaya, Badrul Hisham, Liu, Yanli, Lin, Juntang
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Nature Publishing Group UK 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10209065/
https://www.ncbi.nlm.nih.gov/pubmed/37225709
http://dx.doi.org/10.1038/s41419-023-05859-0
_version_ 1785046797567131648
author Zhang, Shenghui
Liu, Qin
Chang, Mengyuan
Pan, Ying
Yahaya, Badrul Hisham
Liu, Yanli
Lin, Juntang
author_facet Zhang, Shenghui
Liu, Qin
Chang, Mengyuan
Pan, Ying
Yahaya, Badrul Hisham
Liu, Yanli
Lin, Juntang
author_sort Zhang, Shenghui
collection PubMed
description Chemotherapy was conventionally applied to kill cancer cells, but regrettably, they also induce damage to normal cells with high-proliferative capacity resulting in cardiotoxicity, nephrotoxicity, peripheral nerve toxicity, and ovarian toxicity. Of these, chemotherapy-induced ovarian damages mainly include but are not limited to decreased ovarian reserve, infertility, and ovarian atrophy. Therefore, exploring the underlying mechanism of chemotherapeutic drug-induced ovarian damage will pave the way to develop fertility-protective adjuvants for female patients during conventional cancer treatment. Herein, we firstly confirmed the abnormal gonadal hormone levels in patients who received chemotherapy and further found that conventional chemotherapeutic drugs (cyclophosphamide, CTX; paclitaxel, Tax; doxorubicin, Dox and cisplatin, Cis) treatment significantly decreased both the ovarian volume of mice and the number of primordial and antral follicles and accompanied with the ovarian fibrosis and reduced ovarian reserve in animal models. Subsequently, Tax, Dox, and Cis treatment can induce the apoptosis of ovarian granulosa cells (GCs), likely resulting from excessive reactive oxygen species (ROS) production-induced oxidative damage and impaired cellular anti-oxidative capacity. Thirdly, the following experiments demonstrated that Cis treatment could induce mitochondrial dysfunction through overproducing superoxide in GCs and trigger lipid peroxidation leading to ferroptosis, first reported in chemotherapy-induced ovarian damage. In addition, N-acetylcysteine (NAC) treatment could alleviate the Cis-induced toxicity in GCs by downregulating cellular ROS levels and enhancing the anti-oxidative capacity (promoting the expression of glutathione peroxidase, GPX4; nuclear factor erythroid 2-related factor 2, Nrf2 and heme oxygenase-1, HO-1). Our study confirmed the chemotherapy-induced chaotic hormonal state and ovarian damage in preclinical and clinical examination and indicated that chemotherapeutic drugs initiated ferroptosis in ovarian cells through excessive ROS-induced lipid peroxidation and mitochondrial dysfunction, leading to ovarian cell death. Consequently, developing fertility protectants from the chemotherapy-induced oxidative stress and ferroptosis perspective will ameliorate ovarian damage and further improve the life quality of cancer patients.
format Online
Article
Text
id pubmed-10209065
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Nature Publishing Group UK
record_format MEDLINE/PubMed
spelling pubmed-102090652023-05-26 Chemotherapy impairs ovarian function through excessive ROS-induced ferroptosis Zhang, Shenghui Liu, Qin Chang, Mengyuan Pan, Ying Yahaya, Badrul Hisham Liu, Yanli Lin, Juntang Cell Death Dis Article Chemotherapy was conventionally applied to kill cancer cells, but regrettably, they also induce damage to normal cells with high-proliferative capacity resulting in cardiotoxicity, nephrotoxicity, peripheral nerve toxicity, and ovarian toxicity. Of these, chemotherapy-induced ovarian damages mainly include but are not limited to decreased ovarian reserve, infertility, and ovarian atrophy. Therefore, exploring the underlying mechanism of chemotherapeutic drug-induced ovarian damage will pave the way to develop fertility-protective adjuvants for female patients during conventional cancer treatment. Herein, we firstly confirmed the abnormal gonadal hormone levels in patients who received chemotherapy and further found that conventional chemotherapeutic drugs (cyclophosphamide, CTX; paclitaxel, Tax; doxorubicin, Dox and cisplatin, Cis) treatment significantly decreased both the ovarian volume of mice and the number of primordial and antral follicles and accompanied with the ovarian fibrosis and reduced ovarian reserve in animal models. Subsequently, Tax, Dox, and Cis treatment can induce the apoptosis of ovarian granulosa cells (GCs), likely resulting from excessive reactive oxygen species (ROS) production-induced oxidative damage and impaired cellular anti-oxidative capacity. Thirdly, the following experiments demonstrated that Cis treatment could induce mitochondrial dysfunction through overproducing superoxide in GCs and trigger lipid peroxidation leading to ferroptosis, first reported in chemotherapy-induced ovarian damage. In addition, N-acetylcysteine (NAC) treatment could alleviate the Cis-induced toxicity in GCs by downregulating cellular ROS levels and enhancing the anti-oxidative capacity (promoting the expression of glutathione peroxidase, GPX4; nuclear factor erythroid 2-related factor 2, Nrf2 and heme oxygenase-1, HO-1). Our study confirmed the chemotherapy-induced chaotic hormonal state and ovarian damage in preclinical and clinical examination and indicated that chemotherapeutic drugs initiated ferroptosis in ovarian cells through excessive ROS-induced lipid peroxidation and mitochondrial dysfunction, leading to ovarian cell death. Consequently, developing fertility protectants from the chemotherapy-induced oxidative stress and ferroptosis perspective will ameliorate ovarian damage and further improve the life quality of cancer patients. Nature Publishing Group UK 2023-05-24 /pmc/articles/PMC10209065/ /pubmed/37225709 http://dx.doi.org/10.1038/s41419-023-05859-0 Text en © The Author(s) 2023 https://creativecommons.org/licenses/by/4.0/Open Access This 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 license, and indicate if changes were made. The images or other third party material in this article are included in the article’s Creative Commons license, unless indicated otherwise in a credit line to the material. If material is not included in the article’s Creative Commons license 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 license, visit http://creativecommons.org/licenses/by/4.0/ (https://creativecommons.org/licenses/by/4.0/) .
spellingShingle Article
Zhang, Shenghui
Liu, Qin
Chang, Mengyuan
Pan, Ying
Yahaya, Badrul Hisham
Liu, Yanli
Lin, Juntang
Chemotherapy impairs ovarian function through excessive ROS-induced ferroptosis
title Chemotherapy impairs ovarian function through excessive ROS-induced ferroptosis
title_full Chemotherapy impairs ovarian function through excessive ROS-induced ferroptosis
title_fullStr Chemotherapy impairs ovarian function through excessive ROS-induced ferroptosis
title_full_unstemmed Chemotherapy impairs ovarian function through excessive ROS-induced ferroptosis
title_short Chemotherapy impairs ovarian function through excessive ROS-induced ferroptosis
title_sort chemotherapy impairs ovarian function through excessive ros-induced ferroptosis
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10209065/
https://www.ncbi.nlm.nih.gov/pubmed/37225709
http://dx.doi.org/10.1038/s41419-023-05859-0
work_keys_str_mv AT zhangshenghui chemotherapyimpairsovarianfunctionthroughexcessiverosinducedferroptosis
AT liuqin chemotherapyimpairsovarianfunctionthroughexcessiverosinducedferroptosis
AT changmengyuan chemotherapyimpairsovarianfunctionthroughexcessiverosinducedferroptosis
AT panying chemotherapyimpairsovarianfunctionthroughexcessiverosinducedferroptosis
AT yahayabadrulhisham chemotherapyimpairsovarianfunctionthroughexcessiverosinducedferroptosis
AT liuyanli chemotherapyimpairsovarianfunctionthroughexcessiverosinducedferroptosis
AT linjuntang chemotherapyimpairsovarianfunctionthroughexcessiverosinducedferroptosis