9
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Superparamagnetic iron oxide nanoparticles promote ferroptosis of ischemic cardiomyocytes

      letter

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          1 INTRODUCTION AND BACKGROUND Superparamagnetic iron oxide nanoparticles (SPION) have been widely used in the diagnosis and treatment for cardiovascular diseases. 1 , 2 , 3 , 4 , 5 , 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 , 8 , 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 mitochondria, had demonstrated protective effects on I/R myocardium, while deferoxamine (DFO) failed to protect mice against I/R damage due to poor penetrance into mitochondria. Notably, overexpression of ABCB8 in cardiomyocytes in mice reduces mitochondrial iron and protects against I/R damage, 30 suggesting that ABCB8 might play an important role in maintaining iron homeostasis in myocardial mitochondria and regulating ferroptosis after I/R injury. Thus, it is not difficult to speculate that SPION could aggravate mitochondrial iron load in I/R myocardium. SPION applied in ischemic myocardium could be directly degraded by cardiomyocytes, 12 leading to severe mitochondrial iron overload. We detected prominently mitochondrial lipid peroxidation (malondialdehyde, MDA), mitochondrial membrane potential (MMP) loss and ATP depletion at 24 hours and 4 weeks after SPION injected into the peri‐infarcted zones of myocardial ischemia‐reperfusion rats compared with the control group (all P < .01). We found that iron content of mitochondria was significantly higher than that in the control group (P < .001), and the distorted mitochondria were observed by transmission electron microscopy in the SPION group, suggesting that SPION have the potential to destroy mitochondrial structure and function by inducing mitochondria iron overload (data not published). Mitochondria are the major site of iron metabolism and ROS production, thereby cardiomyocytes iron accumulation is especially prone to induce mitochondria iron overload to trigger mitochondrial oxidative damage. Based on the above results, we speculate that SPION might further promote ferroptosis to aggravate left ventricular remodelling and cardiac deterioration by inducing severe mitochondria iron overload to promote lipid peroxidation burst. 2 HYPOTHESIS To summarize, we speculate that SPION applied to ischemic myocardium could exacerbate cardiomyocytes ferroptosis to worsen left ventricular negative remodelling through inducing mitochondria iron overload to catalyse sustained Fenton reaction, eliciting lipid peroxidation burst(as shown in Figure 1). This hypothesis needs to be verified by animal experiments. Firstly, the mitochondrial iron metabolism, lipid peroxidation, morphology and function of mitochondria and ferroptosis should be carefully detected after SPION injected into the peri‐infarcted zones of myocardial ischemia‐reperfusion rats. Secondly, the rat models of myocardial ischemia‐reperfusion were randomly divided into different groups to, respectively, treated with apoptosis inhibitor, necrosis inhibitor, autophagy inhibitor and ferroptosis inhibitor, in order to verify whether ferroptosis play a pivotal role in cardiomyocytes death induced by SPION. Thirdly, SPION were injected into the myocardium of I/R mice model, in which Mlkl −/− or Fadd −/− Mlkl −/− mice were employed to respectively block the pathways of myocardial cell necroptosis or apoptosis, in order to further illustrate whether ferroptosis is the main pathway of SPION‐induced cardiomyocytes death. Fourthly, developing SPION modified by mitochondrial iron chelator or mitochondrial‐targeted antioxidant, the effects of two strategies to improve the myocardial safety of SPION should be comprehensively investigated in vitro and in vivo experiments, potentially promoting the clinical transformation of SPION in cardiovascular field. FIGURE 1 SPION were internalized into cardiomyocytes and further degraded into free ferrous iron in lysosomes. The free ferrous iron entered into mitochondria, resulting in lipid peroxidation of mitochondria to trigger ferroptosis in cardiomyocytes by a large amount of ROS produced via Fenton reaction 3 IMPLICATION The cardiotoxicity of SPION limits its diagnostic or therapeutic application in the cardiovascular field. It is helpful to promote the clinical transformation of SPION in cardiovascular field through rescuing the key target of SPION‐induced cardiomyocyte ferroptosis to improve the myocardial tissue safety. If our hypothesis is true, given that SPION mainly induce mitochondria iron overload of ischemic cardiomyocytes to catalyse lipid peroxidation and exacerbate ferroptosis, and then it is expected to significantly inhibit ferroptosis induced negative remodelling of ischemic myocardium by mitochondrial iron chelator or mitochondrial‐targeted antioxidant peptide modifying SPION to effectively protect mitochondria. CONFLICT OF INTEREST The authors confirm that there are no conflicts of interest.

          Related collections

          Most cited references21

          • Record: found
          • Abstract: found
          • Article: not found

          Fenton-Reaction-Accelerable Magnetic Nanoparticles for Ferroptosis Therapy of Orthotopic Brain Tumors

          Cancer is one of the leading causes of morbidity and mortality in the world, but more cancer therapies are needed to complement existing regimens due to problems of existing cancer therapies. Herein, we term ferroptosis therapy (FT) as a form of cancer therapy and hypothesize that the FT efficacy can be significantly improved via accelerating the Fenton reaction by simultaneously increasing the local concentrations of all reactants (Fe2+, Fe3+, and H2O2) in cancer cells. Thus, Fenton-reaction-acceleratable magnetic nanoparticles, i.e., cisplatin (CDDP)-loaded Fe3O4/Gd2O3 hybrid nanoparticles with conjugation of lactoferrin (LF) and RGD dimer (RGD2) (FeGd-HN@Pt@LF/RGD2), were exploited in this study for FT of orthotopic brain tumors. FeGd-HN@Pt@LF/RGD2 nanoparticles were able to cross the blood-brain barrier because of its small size (6.6 nm) and LF-receptor-mediated transcytosis. FeGd-HN@Pt@LF/RGD2 can be internalized into cancer cells by integrin αvβ3-mediated endocytosis and then release Fe2+, Fe3+, and CDDP upon endosomal uptake and degradation. Fe2+ and Fe3+ can directly participate in the Fenton reaction, whereas the CDDP can indirectly produce H2O2 to further accelerate the Fenton reaction. The acceleration of Fenton reaction generates reactive oxygen species to induce cancer cell death. FeGd-HN@Pt@LF/RGD2 successfully delivered reactants involved in the Fenton reaction to the tumor site and led to significant inhibition of tumor growth. Finally, the intrinsic magnetic resonance imaging (MRI) capability of the nanoparticles was used to assess and monitor tumor response to FT (self-MRI monitoring).
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            Induction of ROS, mitochondrial damage and autophagy in lung epithelial cancer cells by iron oxide nanoparticles.

            Autophagy has attracted a great deal of research interest in tumor therapy in recent years. An attempt was made in this direction and now we report that iron oxide NPs synthesized by us selectively induce autophagy in cancer cells (A549) and not in normal cells (IMR-90). It was also noteworthy that autophagy correlated with ROS production as well as mitochondrial damage. Protection of NAC against ROS clearly suggested the implication of ROS in hyper-activation of autophagy and cell death. Pre-treatment of cancer cells with 3-MA also exhibited protection against autophagy and promote cellular viability. Results also showed involvement of classical mTOR pathway in autophagy induction by iron oxide NPs in A549 cells. Our results had shown that bare iron oxide NPs are significantly cytotoxic to human cancer cells (A549) but not to the normal human lung fibroblast cells (IMR-90).In other words our nanoparticles selectively kill cancerous cells. It is encouraging to conclude that iron oxide NPs bear the potential of its applications in biomedicine, such as tumor therapy specifically by inducing autophagy mediated cell death of cancer cells. Copyright © 2011 Elsevier Ltd. All rights reserved.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: found
              Is Open Access

              Concentration-dependent toxicity of iron oxide nanoparticles mediated by increased oxidative stress

              Iron oxide nanoparticles with unique magnetic properties have a high potential for use in several biomedical, bioengineering and in vivo applications, including tissue repair, magnetic resonance imaging, immunoassay, drug delivery, detoxification of biologic fluids, cell sorting, and hyperthermia. Although various surface modifications are being done for making these nonbiodegradable nanoparticles more biocompatible, their toxic potential is still a major concern. The current in vitro study of the interaction of superparamagnetic iron oxide nanoparticles of mean diameter 30 nm coated with Tween 80 and murine macrophage (J774) cells was undertaken to evaluate the dose- and time-dependent toxic potential, as well as investigate the role of oxidative stress in the toxicity. A 15–30 nm size range of spherical nanoparticles were characterized by transmission electron microscopy and zeta sizer. MTT assay showed >95% viability of cells in lower concentrations (25–200 μg/mL) and up to three hours of exposure, whereas at higher concentrations (300–500 μg/mL) and prolonged (six hours) exposure viability reduced to 55%–65%. Necrosis-apoptosis assay by propidium iodide and Hoechst-33342 staining revealed loss of the majority of the cells by apoptosis. H2DCFDDA assay to quantify generation of intracellular reactive oxygen species (ROS) indicated that exposure to a higher concentration of nanoparticles resulted in enhanced ROS generation, leading to cell injury and death. The cell membrane injury induced by nanoparticles studied using the lactate dehydrogenase assay, showed both concentration- and time-dependent damage. Thus, this study concluded that use of a low optimum concentration of superparamagnetic iron oxide nanoparticles is important for avoidance of oxidative stress-induced cell injury and death.
                Bookmark

                Author and article information

                Contributors
                shenyunli2011@163.com
                Journal
                J Cell Mol Med
                J. Cell. Mol. Med
                10.1111/(ISSN)1582-4934
                JCMM
                Journal of Cellular and Molecular Medicine
                John Wiley and Sons Inc. (Hoboken )
                1582-1838
                1582-4934
                11 August 2020
                September 2020
                : 24
                : 18 ( doiID: 10.1111/jcmm.v24.18 )
                : 11030-11033
                Affiliations
                [ 1 ] Department of Cardiology Shanghai East Hospital Tongji University School of Medicine Shanghai China
                Author notes
                [*] [* ] Correspondence

                Yunli Shen, Department of Cardiology, Shanghai East Hospital, Tongji University School of Medicine, Jimo Road No, 150, Shanghai, China.

                Email: shenyunli2011@ 123456163.com

                Author information
                https://orcid.org/0000-0002-3204-6594
                Article
                JCMM15722
                10.1111/jcmm.15722
                7521151
                32780538
                19492304-ac4c-4f72-9a24-f5dfa74f7618
                © 2020 The Authors. Journal of Cellular and Molecular Medicine published by Foundation for Cellular and Molecular Medicine and John Wiley & Sons Ltd

                This is an open access article under the terms of the http://creativecommons.org/licenses/by/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited.

                History
                : 31 March 2020
                : 05 July 2020
                : 18 July 2020
                Page count
                Figures: 1, Tables: 0, Pages: 4, Words: 2403
                Funding
                Funded by: Top‐level Clinical Discipline Project of Shanghai Pudong District
                Award ID: PWYgf2018‐02
                Funded by: the National Key Research and Development Program of China
                Award ID: 2016YFC1301200
                Funded by: the Natural Science Foundation of Jiangxi Province of China
                Award ID: 20192BAB205006
                Funded by: National Natural Science Foundation of China , open-funder-registry 10.13039/501100001809;
                Award ID: 81500201
                Categories
                Letter to the Editor
                Letter to the Editor
                Custom metadata
                2.0
                September 2020
                Converter:WILEY_ML3GV2_TO_JATSPMC version:5.9.1 mode:remove_FC converted:28.09.2020

                Molecular medicine
                cardiotoxicity,ferroptosis,mitochondria,superparamagnetic iron oxide nanoparticles

                Comments

                Comment on this article