超顺磁性氧化铁纳米颗粒促进缺血心肌细胞铁下垂。

IF 5.3 2区 医学 Q1 Biochemistry, Genetics and Molecular Biology
Journal of Cellular and Molecular Medicine Pub Date : 2020-09-01 Epub Date: 2020-08-11 DOI:10.1111/jcmm.15722
Hao Zheng, Jieyun You, Xiaobo Yao, Qizheng Lu, Wei Guo, Yunli Shen
{"title":"超顺磁性氧化铁纳米颗粒促进缺血心肌细胞铁下垂。","authors":"Hao Zheng, Jieyun You, Xiaobo Yao, Qizheng Lu, Wei Guo, Yunli Shen","doi":"10.1111/jcmm.15722","DOIUrl":null,"url":null,"abstract":"Superparamagnetic iron oxide nanoparticles (SPION) have been widely used in the diagnosis and treatment for cardiovascular diseases.1-6 Correspondingly, the myocardial tissue safety of SPION is becoming a bottleneck to seriously restrict its clinical translation. In recent years, in vitro and in vivo experiments have confirmed that SPION-induced oxidative stress of normal myocardium in mice, leading to myocardial cell injury, apoptosis or necrosis.7-9 More alarmingly, SPION applied to ischemic myocardium could accumulate in the target sites for a long time with high concentration,5,6,10 thereby probably further aggravating oxidative stress injury and cardiomyocytes death.11,12 So far, however, the specific molecular mechanism of cardiotoxicity of SPION remains unclear. Previous studies have reported that SPION-induced apoptosis of murine macrophage (J774) cells 13 and necrosis of human endothelial cells.14 SPION can selectively induce autophagy-mediated cell death of human cancer cells (A549).15 After SPION pre-treatment, H9C2 cardiomyocytes were exposed to acrolein or H2O2, leading to reactive oxygen species (ROS) dependent cell necrosis.7 Our in vitro experiment showed that SPION significantly increased oxidative stress damage to overactivate autophagy and endoplasmic reticulum stress, eventually resulting in cardiomyocyte apoptosis.12 Furthermore, SPION could elicit IL-1βrelease and pyroptosis in macrophages, especially with the octapod and plate morphology.16 Notably, it has been recently reported that sorafenib or cisplatin assembled into nano-devices containing SPION, which are phagocytized by tumour cells and degraded into free divalent iron to accelerate Fenton reaction, leading to the lipid peroxidation burst to promote ferroptosis of tumour cells.17,18 Taken together, SPION can induce apoptosis, necrosis, autophagy, pyroptosis or ferroptosis in vitro and in vivo studies. The discrepancy may be attributed to distinct cell types and experiments design. It has already been well documented that the toxicity of SPION is mainly due to its degradation and release of free iron to catalyse Fenton reaction, leading to oxidative stress by a large number of ROS generation.19,20 Then, what is the downstream molecular mechanism of SPION mediated cardiotoxicity? Ferroptosis is a novel form of regulated cell death characterized by the iron-dependent accumulation of lipid peroxides to lethal levels, which is morphologically, biochemically, and genetically distinct from apoptosis, necroptosis and autophagy.21 Recent studies found that ferroptosis is not only an important pathological mechanism in the case of circulating iron overload of hemochromatosis,22 but also a key molecular mechanism of cellular iron overload in doxorubicin (DOX) induced cardiomyopathy.23 DOX induced mitochondria iron overload by down-regulating ABCB8,24 a mitochondrial protein that facilitates iron export, to elicit lipid peroxidation and mitochondria dysfunction, eventually causing cardiomyocytes ferroptosis.23 Mice that were subjected to 30 minutes of myocardial ischemia followed by 24 hours of reperfusion had significantly higher levels of cardiac non-heme iron, cardiac ferritin H, ferritin L and Ptgs2 mRNA. Both ferroptosis inhibitor Ferrostatin-1 (Fer-1) and iron chelator Dexrazoxane (DXZ) pre-treatment significantly reduced I/R-induced cardiac remodelling and fibrosis, indicating that ischemia-reperfusion could also induce cardiomyocytes iron overload to cause ferroptosis and subsequent left ventricular remodelling.23 Myocardial haemorrhage is a frequent complication after successful myocardial reperfusion,25,26 which is associated with residual myocardial iron in post-myocardial infarction (MI) patients received reperfusion therapy.27 It is reasonable to infer that this iron accumulation has a potential to generate excessive ROS and trigger pathological events such as ferroptosis. A previous study also confirmed that ferroptosis is a significant type of cell death in cardiomyocytes; moreover, mechanistic target of rapamycin (mTOR) was found to play an important role in protecting cardiomyocytes against excess iron and ferroptosis by regulating ROS production.28 In addition, glutathione peroxidase 4 (GPX4), which protects cells from ferroptosis, was down-regulated in the early and middle stages of MI mouse model, suggesting that ferroptosis during MI was in part due to a reduction in GPX4 protein.29 Even though signalling pathways of ferroptosis in cardiovascular diseases is not yet well characterized, it has been confirmed that ischemia-reperfusion (I/R) could induce mitochondrial iron overload in cardiomyocytes rather than the increase of iron content in cytoplasm.30 In this study, mice treated with 2,2′-bipyridyl (BPD), which has high membrane permeability and thus is able to access","PeriodicalId":15215,"journal":{"name":"Journal of Cellular and Molecular Medicine","volume":null,"pages":null},"PeriodicalIF":5.3000,"publicationDate":"2020-09-01","publicationTypes":"Journal Article","fieldsOfStudy":null,"isOpenAccess":false,"openAccessPdf":"https://sci-hub-pdf.com/10.1111/jcmm.15722","citationCount":"17","resultStr":"{\"title\":\"Superparamagnetic iron oxide nanoparticles promote ferroptosis of ischemic cardiomyocytes.\",\"authors\":\"Hao Zheng, Jieyun You, Xiaobo Yao, Qizheng Lu, Wei Guo, Yunli Shen\",\"doi\":\"10.1111/jcmm.15722\",\"DOIUrl\":null,\"url\":null,\"abstract\":\"Superparamagnetic iron oxide nanoparticles (SPION) have been widely used in the diagnosis and treatment for cardiovascular diseases.1-6 Correspondingly, the myocardial tissue safety of SPION is becoming a bottleneck to seriously restrict its clinical translation. In recent years, in vitro and in vivo experiments have confirmed that SPION-induced oxidative stress of normal myocardium in mice, leading to myocardial cell injury, apoptosis or necrosis.7-9 More alarmingly, SPION applied to ischemic myocardium could accumulate in the target sites for a long time with high concentration,5,6,10 thereby probably further aggravating oxidative stress injury and cardiomyocytes death.11,12 So far, however, the specific molecular mechanism of cardiotoxicity of SPION remains unclear. Previous studies have reported that SPION-induced apoptosis of murine macrophage (J774) cells 13 and necrosis of human endothelial cells.14 SPION can selectively induce autophagy-mediated cell death of human cancer cells (A549).15 After SPION pre-treatment, H9C2 cardiomyocytes were exposed to acrolein or H2O2, leading to reactive oxygen species (ROS) dependent cell necrosis.7 Our in vitro experiment showed that SPION significantly increased oxidative stress damage to overactivate autophagy and endoplasmic reticulum stress, eventually resulting in cardiomyocyte apoptosis.12 Furthermore, SPION could elicit IL-1βrelease and pyroptosis in macrophages, especially with the octapod and plate morphology.16 Notably, it has been recently reported that sorafenib or cisplatin assembled into nano-devices containing SPION, which are phagocytized by tumour cells and degraded into free divalent iron to accelerate Fenton reaction, leading to the lipid peroxidation burst to promote ferroptosis of tumour cells.17,18 Taken together, SPION can induce apoptosis, necrosis, autophagy, pyroptosis or ferroptosis in vitro and in vivo studies. The discrepancy may be attributed to distinct cell types and experiments design. It has already been well documented that the toxicity of SPION is mainly due to its degradation and release of free iron to catalyse Fenton reaction, leading to oxidative stress by a large number of ROS generation.19,20 Then, what is the downstream molecular mechanism of SPION mediated cardiotoxicity? Ferroptosis is a novel form of regulated cell death characterized by the iron-dependent accumulation of lipid peroxides to lethal levels, which is morphologically, biochemically, and genetically distinct from apoptosis, necroptosis and autophagy.21 Recent studies found that ferroptosis is not only an important pathological mechanism in the case of circulating iron overload of hemochromatosis,22 but also a key molecular mechanism of cellular iron overload in doxorubicin (DOX) induced cardiomyopathy.23 DOX induced mitochondria iron overload by down-regulating ABCB8,24 a mitochondrial protein that facilitates iron export, to elicit lipid peroxidation and mitochondria dysfunction, eventually causing cardiomyocytes ferroptosis.23 Mice that were subjected to 30 minutes of myocardial ischemia followed by 24 hours of reperfusion had significantly higher levels of cardiac non-heme iron, cardiac ferritin H, ferritin L and Ptgs2 mRNA. Both ferroptosis inhibitor Ferrostatin-1 (Fer-1) and iron chelator Dexrazoxane (DXZ) pre-treatment significantly reduced I/R-induced cardiac remodelling and fibrosis, indicating that ischemia-reperfusion could also induce cardiomyocytes iron overload to cause ferroptosis and subsequent left ventricular remodelling.23 Myocardial haemorrhage is a frequent complication after successful myocardial reperfusion,25,26 which is associated with residual myocardial iron in post-myocardial infarction (MI) patients received reperfusion therapy.27 It is reasonable to infer that this iron accumulation has a potential to generate excessive ROS and trigger pathological events such as ferroptosis. A previous study also confirmed that ferroptosis is a significant type of cell death in cardiomyocytes; moreover, mechanistic target of rapamycin (mTOR) was found to play an important role in protecting cardiomyocytes against excess iron and ferroptosis by regulating ROS production.28 In addition, glutathione peroxidase 4 (GPX4), which protects cells from ferroptosis, was down-regulated in the early and middle stages of MI mouse model, suggesting that ferroptosis during MI was in part due to a reduction in GPX4 protein.29 Even though signalling pathways of ferroptosis in cardiovascular diseases is not yet well characterized, it has been confirmed that ischemia-reperfusion (I/R) could induce mitochondrial iron overload in cardiomyocytes rather than the increase of iron content in cytoplasm.30 In this study, mice treated with 2,2′-bipyridyl (BPD), which has high membrane permeability and thus is able to access\",\"PeriodicalId\":15215,\"journal\":{\"name\":\"Journal of Cellular and Molecular Medicine\",\"volume\":null,\"pages\":null},\"PeriodicalIF\":5.3000,\"publicationDate\":\"2020-09-01\",\"publicationTypes\":\"Journal Article\",\"fieldsOfStudy\":null,\"isOpenAccess\":false,\"openAccessPdf\":\"https://sci-hub-pdf.com/10.1111/jcmm.15722\",\"citationCount\":\"17\",\"resultStr\":null,\"platform\":\"Semanticscholar\",\"paperid\":null,\"PeriodicalName\":\"Journal of Cellular and Molecular Medicine\",\"FirstCategoryId\":\"3\",\"ListUrlMain\":\"https://doi.org/10.1111/jcmm.15722\",\"RegionNum\":2,\"RegionCategory\":\"医学\",\"ArticlePicture\":[],\"TitleCN\":null,\"AbstractTextCN\":null,\"PMCID\":null,\"EPubDate\":\"2020/8/11 0:00:00\",\"PubModel\":\"Epub\",\"JCR\":\"Q1\",\"JCRName\":\"Biochemistry, Genetics and Molecular Biology\",\"Score\":null,\"Total\":0}","platform":"Semanticscholar","paperid":null,"PeriodicalName":"Journal of Cellular and Molecular Medicine","FirstCategoryId":"3","ListUrlMain":"https://doi.org/10.1111/jcmm.15722","RegionNum":2,"RegionCategory":"医学","ArticlePicture":[],"TitleCN":null,"AbstractTextCN":null,"PMCID":null,"EPubDate":"2020/8/11 0:00:00","PubModel":"Epub","JCR":"Q1","JCRName":"Biochemistry, Genetics and Molecular Biology","Score":null,"Total":0}
引用次数: 17

摘要

本文章由计算机程序翻译,如有差异,请以英文原文为准。

Superparamagnetic iron oxide nanoparticles promote ferroptosis of ischemic cardiomyocytes.

Superparamagnetic iron oxide nanoparticles promote ferroptosis of ischemic cardiomyocytes.
Superparamagnetic iron oxide nanoparticles (SPION) have been widely used in the diagnosis and treatment for cardiovascular diseases.1-6 Correspondingly, the myocardial tissue safety of SPION is becoming a bottleneck to seriously restrict its clinical translation. In recent years, in vitro and in vivo experiments have confirmed that SPION-induced oxidative stress of normal myocardium in mice, leading to myocardial cell injury, apoptosis or necrosis.7-9 More alarmingly, SPION applied to ischemic myocardium could accumulate in the target sites for a long time with high concentration,5,6,10 thereby probably further aggravating oxidative stress injury and cardiomyocytes death.11,12 So far, however, the specific molecular mechanism of cardiotoxicity of SPION remains unclear. Previous studies have reported that SPION-induced apoptosis of murine macrophage (J774) cells 13 and necrosis of human endothelial cells.14 SPION can selectively induce autophagy-mediated cell death of human cancer cells (A549).15 After SPION pre-treatment, H9C2 cardiomyocytes were exposed to acrolein or H2O2, leading to reactive oxygen species (ROS) dependent cell necrosis.7 Our in vitro experiment showed that SPION significantly increased oxidative stress damage to overactivate autophagy and endoplasmic reticulum stress, eventually resulting in cardiomyocyte apoptosis.12 Furthermore, SPION could elicit IL-1βrelease and pyroptosis in macrophages, especially with the octapod and plate morphology.16 Notably, it has been recently reported that sorafenib or cisplatin assembled into nano-devices containing SPION, which are phagocytized by tumour cells and degraded into free divalent iron to accelerate Fenton reaction, leading to the lipid peroxidation burst to promote ferroptosis of tumour cells.17,18 Taken together, SPION can induce apoptosis, necrosis, autophagy, pyroptosis or ferroptosis in vitro and in vivo studies. The discrepancy may be attributed to distinct cell types and experiments design. It has already been well documented that the toxicity of SPION is mainly due to its degradation and release of free iron to catalyse Fenton reaction, leading to oxidative stress by a large number of ROS generation.19,20 Then, what is the downstream molecular mechanism of SPION mediated cardiotoxicity? Ferroptosis is a novel form of regulated cell death characterized by the iron-dependent accumulation of lipid peroxides to lethal levels, which is morphologically, biochemically, and genetically distinct from apoptosis, necroptosis and autophagy.21 Recent studies found that ferroptosis is not only an important pathological mechanism in the case of circulating iron overload of hemochromatosis,22 but also a key molecular mechanism of cellular iron overload in doxorubicin (DOX) induced cardiomyopathy.23 DOX induced mitochondria iron overload by down-regulating ABCB8,24 a mitochondrial protein that facilitates iron export, to elicit lipid peroxidation and mitochondria dysfunction, eventually causing cardiomyocytes ferroptosis.23 Mice that were subjected to 30 minutes of myocardial ischemia followed by 24 hours of reperfusion had significantly higher levels of cardiac non-heme iron, cardiac ferritin H, ferritin L and Ptgs2 mRNA. Both ferroptosis inhibitor Ferrostatin-1 (Fer-1) and iron chelator Dexrazoxane (DXZ) pre-treatment significantly reduced I/R-induced cardiac remodelling and fibrosis, indicating that ischemia-reperfusion could also induce cardiomyocytes iron overload to cause ferroptosis and subsequent left ventricular remodelling.23 Myocardial haemorrhage is a frequent complication after successful myocardial reperfusion,25,26 which is associated with residual myocardial iron in post-myocardial infarction (MI) patients received reperfusion therapy.27 It is reasonable to infer that this iron accumulation has a potential to generate excessive ROS and trigger pathological events such as ferroptosis. A previous study also confirmed that ferroptosis is a significant type of cell death in cardiomyocytes; moreover, mechanistic target of rapamycin (mTOR) was found to play an important role in protecting cardiomyocytes against excess iron and ferroptosis by regulating ROS production.28 In addition, glutathione peroxidase 4 (GPX4), which protects cells from ferroptosis, was down-regulated in the early and middle stages of MI mouse model, suggesting that ferroptosis during MI was in part due to a reduction in GPX4 protein.29 Even though signalling pathways of ferroptosis in cardiovascular diseases is not yet well characterized, it has been confirmed that ischemia-reperfusion (I/R) could induce mitochondrial iron overload in cardiomyocytes rather than the increase of iron content in cytoplasm.30 In this study, mice treated with 2,2′-bipyridyl (BPD), which has high membrane permeability and thus is able to access
求助全文
通过发布文献求助,成功后即可免费获取论文全文。 去求助
来源期刊
CiteScore
10.00
自引率
1.90%
发文量
496
审稿时长
28 weeks
期刊介绍: Bridging physiology and cellular medicine, and molecular biology and molecular therapeutics, Journal of Cellular and Molecular Medicine publishes basic research that furthers our understanding of the cellular and molecular mechanisms of disease and translational studies that convert this knowledge into therapeutic approaches.
×
引用
GB/T 7714-2015
复制
MLA
复制
APA
复制
导出至
BibTeX EndNote RefMan NoteFirst NoteExpress
×
提示
您的信息不完整,为了账户安全,请先补充。
现在去补充
×
提示
您因"违规操作"
具体请查看互助需知
我知道了
×
提示
确定
请完成安全验证×
copy
已复制链接
快去分享给好友吧!
我知道了
右上角分享
点击右上角分享
0
联系我们:info@booksci.cn Book学术提供免费学术资源搜索服务,方便国内外学者检索中英文文献。致力于提供最便捷和优质的服务体验。 Copyright © 2023 布克学术 All rights reserved.
京ICP备2023020795号-1
ghs 京公网安备 11010802042870号
Book学术文献互助
Book学术文献互助群
群 号:481959085
Book学术官方微信