Next Article in Journal
Rno_circ_0001004 Acts as a miR-709 Molecular Sponge to Regulate the Growth Hormone Synthesis and Cell Proliferation
Next Article in Special Issue
Mitochondrial-Targeted Therapy for Doxorubicin-Induced Cardiotoxicity
Previous Article in Journal
Regulation of Opsin Gene Expression by DNA Methylation and Histone Acetylation
Previous Article in Special Issue
Heart Failure after Cardiac Surgery: The Role of Halogenated Agents, Myocardial Conditioning and Oxidative Stress
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Therapeutic Targets for DOX-Induced Cardiomyopathy: Role of Apoptosis vs. Ferroptosis

Department of Cardiology, Keio University School of Medicine, Tokyo 160-8582, Japan
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2022, 23(3), 1414; https://doi.org/10.3390/ijms23031414
Submission received: 31 December 2021 / Revised: 20 January 2022 / Accepted: 24 January 2022 / Published: 26 January 2022
(This article belongs to the Special Issue Metabolic Therapies for Heart Failure)

Abstract

:
Doxorubicin (DOX) is the most widely used anthracycline anticancer agent; however, its cardiotoxicity limits its clinical efficacy. Numerous studies have elucidated the mechanisms underlying DOX-induced cardiotoxicity, wherein apoptosis has been reported as the most common final step leading to cardiomyocyte death. However, in the past two years, the involvement of ferroptosis, a novel programmed cell death, has been proposed. The purpose of this review is to summarize the historical background that led to each form of cell death, focusing on DOX-induced cardiotoxicity and the molecular mechanisms that trigger each form of cell death. Furthermore, based on this understanding, possible therapeutic strategies to prevent DOX cardiotoxicity are outlined. DNA damage, oxidative stress, intracellular signaling, transcription factors, epigenetic regulators, autophagy, and metabolic inflammation are important factors in the molecular mechanisms of DOX-induced cardiomyocyte apoptosis. Conversely, the accumulation of lipid peroxides, iron ion accumulation, and decreased expression of glutathione and glutathione peroxidase 4 are important in ferroptosis. In both cascades, the mitochondria are an important site of DOX cardiotoxicity. The last part of this review focuses on the significance of the disruption of mitochondrial homeostasis in DOX cardiotoxicity.

1. Introduction

Drug therapy for cancer has made remarkable progress in recent years with the development of molecular-targeted drugs and immune checkpoint inhibitors alongside conventional chemotherapy. As a result, the survival rate of cancer patients has markedly improved [1]. As the prognosis of cancer patients improves, the issue of increased risk of developing cardiovascular disease due to anticancer drug therapy is rapidly gaining attention [2,3,4]. In fact, for breast cancer patients with a history of cardiovascular disease, mortality due to cardiovascular disease is higher than mortality due to breast cancer more than five years after the diagnosis of breast cancer [5]. Conversely, heart failure patients are at an increased risk of cancer [6]. In addition, as the population ages and cancer treatment advances, the number of patients with both conditions has been increasing [7]. In this context, the field of oncocardiology has become increasingly important in recent years [8].
Heart failure associated with anticancer drugs is classified into two categories: whether the mechanism of cardiotoxicity is irreversible (type I) or reversible (type II). As Table 1 shows, irreversible myocardial injury caused by anthracyclines such as doxorubicin (DOX) is defined as type I. In contrast, reversible myocardial injury caused by trastuzumab, a human epidermal growth factor receptor 2 (Her2) inhibitor, and later bevacizumab, sunitinib, and sorafenib, is defined as type II [2,9]. Generally, type II drugs cause cardiomyocyte dysfunction but not myocardial necrosis, and their cardiotoxicity is considered reversible [9]. This review will introduce the molecular mechanisms of DOX cardiotoxicity and anthracyclines that cause type I myocardial injury and will discuss possible countermeasures.
Anthracyclines are a class of drugs used in cancer chemotherapy and are derived from Streptomyces peucetius var. caesius [10]. Anthracyclines include DOX, daunorubicin, epirubicin, idarubicin, pirarubicin, and amrubicin. They were developed in the 1970s and are widely used in clinical practice. They inhibit DNA topoisomerase II (TopII) and intercalate between base pairs of DNA strands, inhibiting DNA synthesis, replication, and transcription and suppressing cell proliferation. Among them, DOX (sold under the brand name Adriamycin) is one of the most widely used anticancer drugs in cancer treatment [11]. It has been used for hematopoietic tumors and solid tumors, such as breast cancer, resulting in improved cure rates and quality of life for cancer patients [12]. However, despite its outstanding efficacy, the risk of DOX cardiomyopathy, which develops in a cumulative dose-dependent manner, is often experienced as a limitation to continued treatment [13].
Many basic studies have attempted to elucidate the molecular mechanisms of DOX cardiotoxicity, and approximately 400 studies have been published since 2000. Although apoptosis has been the focus of attention, as the final process leading to cardiomyocyte death, several recent studies have concluded that the newly proposed programmed death, ferroptosis, is involved in DOX-induced cardiomyocyte death. In this article, we review the reported mechanisms of DOX cardiotoxicity, broadly categorized into apoptosis (Figure 1) and ferroptosis (Figure 2), and classify them according to the molecular mechanism of each cell death.

1.1. Apoptosis

1.1.1. DNA Damage

TopII has attracted attention as a classical cellular target of DOX [14]. There are two forms of the TopII enzyme: TopIIα and TopIIβ [15]. TopIIα is highly expressed in tumor cells, whereas TopIIβ is highly expressed in cardiomyocytes. TopIIβ-DOX-DNA ternary complex formation is important for DOX cardiotoxicity and induces DNA double-strand breaks in cardiomyocytes, resulting in cell death [16]. In the presence of TopIIβ, DOX decreases the transcription of genes, such as PPARG coactivator 1α (PGC1-α), involved in mitochondrial biogenesis and function [17]. Cancer patients with high expression levels of TopIIβ in cardiomyocytes are more sensitive to DOX cardiotoxicity, while mice lacking TopIIβ are resistant to DOX cardiotoxicity [17]. One reason for the increased cardiotoxicity risk with the combination of anthracyclines and trastuzumab is that trastuzumab changes the expression of the TopIIβ gene and protein in cardiomyocytes, leading to apoptosis [18]. Selective inhibitors of TopIIβ are a promising strategy to reduce toxicity to cardiomyocytes without compromising the potent anticancer effects of DOX. Dexrazoxane functions as a catalytic inhibitor of TopIIβ and reduces anthracycline-induced cardiotoxicity by suppressing DOX-induced DNA double-strand breaks [19]. Although concerns about second malignant neoplasms have been reported [20], dexrazoxane has been approved and clinically used in more than 30 countries to treat extravascular leakage of anthracycline antineoplastic agents.

1.1.2. Oxidative Stress

Oxidative stress is one of the major mechanisms of DOX cardiotoxicity [21,22], and attenuating reactive oxygen species (ROS) by N-acetylcysteine and resveratrol is reportedly an efficient strategy for alleviating DOX-induced cardiotoxicity [23,24,25]. In the heart, after DOX administration, the major enzymes of the glycolytic system, triosephosphate isomerase, β-enolase, and ubiquinone oxidoreductase, which function as a transporter of electrons to the mitochondrial respiratory chain, were oxidized, and their activities decreased, suggesting that the bioenergetic pathway is an important target of DOX-induced oxidative stress [26]. There are many reports on the importance of the mitochondria as a source and target of oxidative stress [27,28,29]. The hearts of glutathione peroxidase (GPX) 1 knockout mice, scavenging H2O2 in the mitochondria and cytoplasm, showed significantly more severe DOX-induced cardiac dysfunction, mitochondrial injury, protein nitration, and apoptosis than the hearts of wild-type mice [30]. Although it is possible to enhance antioxidant stress mechanisms in cardiomyocytes by preconditioning with non-lethal oxidative stress stimuli, clinical application is difficult because similar cellular responses are expected to occur in cancer cells.
Research is also underway to establish iPS cells from patients and to differentiate them into cardiomyocytes to elucidate individual differences in sensitivity to DOX and the mechanisms involved. Human-induced pluripotent stem cell-derived cardiomyocytes (hiPSC-CMs) from breast cancer patients who experienced DOX-induced cardiotoxicity were consistently more sensitive to DOX toxicity, with decreased cell survival, impaired mitochondrial and metabolic function, impaired calcium handling, decreased antioxidant pathway activity, and increased production of ROS, compared to patients who did not experience DOX-induced cardiotoxicity [23]. These results suggest that individual differences in antioxidant mechanisms may define susceptibility to DOX. In fact, most recently, Chi Keung Lam et al. highlighted the importance of the use of patient-specific hiPSC-CMs in identifying populations who are at risk of drug-induced cardiotoxicity [31]. This strategy has potential applications in future precision medicine practice, especially in regard to the overlap cases between type I and type II drugs, such as sequential treatment with DOX and Her2 inhibitors, because the incidence of cardiotoxicity in this setting varies according to patient-related factors.

1.1.3. Intracellular Signaling

Among the intracellular signaling molecules, mitogen-activated protein kinase (MAPK) is one of the most studied target molecules for triggering DOX-induced cardiotoxicity. The MAPK cascade forms a signaling network that regulates cellular processes, including cell proliferation, growth, differentiation, transformation, and apoptosis. There are four major branching pathways of the MAPK pathway [32], and the p38 cascade is the main contributor to DOX-induced cardiotoxicity. Activated p38 induces apoptosis by acting on BAX, Bcl-2, and p53 [32,33,34,35,36]. A study examining the role of individual p38 isoforms, particularly the alternative isoforms p38γ and p38δ, in DOX-induced cardiotoxicity reported that p38δ plays an important role in promoting DOX-induced cardiotoxicity in females by inhibiting autophagy [37]. Conversely, many reports suggest that PI3k/Akt signaling, one of the most important kinases in the regulation of cell survival, has a significant effect on the inhibition of DOX-induced cardiotoxicity [38,39,40,41]. Indeed, the upregulation of the PI3k/Akt expression can inhibit apoptosis and improve DOX-induced cardiotoxicity [41,42,43]. Specifically, the hormone irisin, the membrane protein fibronectin type III domain-containing 5, and its cleavage substance, activate AKT/mTOR signaling and attenuate DOX cardiotoxicity-induced apoptosis [43]. In addition, some reports have suggested that JNK and ERK mediate DOX-induced cardiotoxicity [44,45]. Mechanistically, DOX treatment for rat myoblastic H9c2 cells shows increased phosphorylation levels of JNK and ERK, leading to the upregulation of NFkB, which further enhances cardiomyocyte apoptosis. Future studies to clarify the differences in the intracellular signaling mechanisms activated after treatment with DOX between cardiomyocytes and cancer cells will be useful for identifying target molecules that can mitigate cardiomyocyte toxicity while maintaining the anticancer effects of DOX.

1.1.4. Transcription Factors

In response to DNA damage, p53 induces cell cycle arrest and promotes DNA repair and apoptosis [46]. In DOX-induced cardiotoxicity, p53, activated by DNA double-strand breaks, triggers apoptosis. Dox-treated H9c2 cells showed increased phosphorylation of Ser15 in p53, the release of cytochrome c, and increased apoptosis [47]. In mouse hearts, DOX treatment promoted the acetylation of p53 protein and binding of p53 to the BAX promoter, upregulated BAX expression, and increased cytochrome c release from the mitochondria [48]. Using hiPSC-CMs, p53 was identified by RNA-seq as an upstream regulator of DOX-induced transcriptome changes [49]. Clustering and pathway analyses revealed that an increased expression of the death receptor and the exogenous apoptotic pathway were significantly associated with DOX-induced cardiotoxicity [49]. Suppression of the p53-mediated PGC1-α and APLNR (apelin receptor) signaling pathways limits energy production through fatty acid oxidation in the myocardium of DOX-treated mice [50]. DOX also inhibits histone deacetylases (HDACs) activity in a ROS-independent manner and promotes p53 acetylation [51]. Studies in p53-deficient mice have shown that the inhibition of p53 is cardioprotective during DOX treatment, but paradoxically leads to increased cardiotoxicity after the cessation of DOX treatment [52,53,54]. Other studies have shown that selective deletion of p53 in cardiomyocytes is insufficient to prevent the DOX-induced myocardial generation of reactive oxygen and nitrogen species, apoptosis, interstitial fibrosis, and perivascular fibrosis [55]. Thus, it is necessary to consider the possibility that the role of p53 in DOX-induced cardiotoxicity may differ between the acute and chronic phases. It has also been reported that DOX cardiotoxicity is associated with the reduced function of GATA binding protein 4 (GATA4), a transcription factor that regulates fetal cardiac development and stress response in adult cardiomyocytes [56]. By decreasing GATA4 levels, DOX downregulates Bcl2 expression and induces autophagy-related genes, leading to apoptosis and autophagy [57,58], and inhibits myofilament gene transcription, causing cardiac dysfunction due to extensive sarcomere structural abnormalities [59]. The establishment of animal models that separately investigate the mechanisms of acute and delayed cardiotoxicity after DOX treatment is warranted.

1.1.5. Epigenetic Regulators

Epigenetic modifications, including DNA methylation, histone modifications, and noncoding RNA expression, play an important role in regulating gene expression and contribute to DOX cardiotoxicity [60]. For instance, Ferreira et al. showed that pretreatment with DOX ameliorated H9c2 cells resistance against subsequent exposure to DOX [61]. They demonstrated that DOX pretreatment resulted in upregulation of mitochondrial DNA transcripts accompanied by a decrease in DNA methyltransferase 1 and global methylation levels. These results suggest that DOX pretreatment induces a beneficial and possibly epigenetic-based mitochondrial adaptation. Besides DNA methylation, histone modifications are also involved in DOX-induced cardiotoxicity. The expressions of several HDACs, such as HDAC6 and HDAC2, are affected by DOX treatment in cardiomyocytes [62,63,64]. For example, HDAC6 is upregulated in DOX-treated primary rat cardiomyocytes, resulting in deacetylation of α-tubulin [62]. On the other hand, HDAC2 is downregulated by DOX treatment, as well as decreased histone 3 acetylation [64]. Moreover, as mentioned above, another report showed that DOX cardiotoxicity leads to the suppression of the HDACs activity, which results in upregulation of p53 [51]. These data suggest that regulating these HDACs may also have a therapeutic potential for DOX cardiotoxicity. Furthermore, recent studies have demonstrated that noncoding RNAs, including long noncoding RNAs and microRNAs (miRNAs), are also involved in DOX cardiotoxicity [65,66,67,68]. Several miRNAs, such as miR-15 [65], miR-23a [68], and miR-34a [66,67], have been reportedly affected either in vitro or in vivo models of DOX-induced cardiomyopathy. In fact, overexpression of particular miRNAs, miR-232/132, showed substantial efficacy for DOX cardiotoxicity prevention in a mouse model [69]. Because epigenetic modification of the gene expression is a key early event in the transcriptional reprogramming of cardiac structural remodeling and metabolic flux in failing hearts [70], elucidating therapies to control this early step may be an important strategy in the future.

1.1.6. Autophagy

DOX is also known to exert toxic effects by affecting macroautophagy, chaperone-mediated autophagy, and the lysosome itself, the site where molecules transported by autophagy are degraded, thereby impairing proteolysis [71]. While some reports suggest that activating autophagy can inhibit DOX toxicity [72], others suggest that inhibiting dysregulated (or excessive) autophagy (or mitophagy) is necessary to reduce DOX toxicity [73,74]. Activating the Toll-Like Receptor 9/PI3Kγ pathway by DOX-damaged mitochondrial DNA in autolysosomes inhibits autophagy, thereby causing cardiac dysfunction [75]. Ghrelin inhibits DOX-induced autophagy and suppresses cardiomyocyte apoptosis by inducing mTOR via AMPK and p38-MAPK. [76] Resveratrol exerts its cardioprotective effect by inhibiting DOX-induced autophagy via S6K inhibition [77]. Bnip3, an autophagy-related molecule, interacts with LC3 on the outer mitochondrial membrane to sequester and degrade the mitochondria into autophagosomes [78]. In DOX-treated cardiomyocytes, Bnip3 expression was upregulated, inducing the opening of the mitochondrial permeability pore (mPTP) and loss of ∆Ψm, excessive mitochondrial fragmentation, mitophagy, and necrosis. Knockdown of Bnip3 prevents mitochondrial fission and DOX-induced cell death, suggesting that Bnip3 is involved in the cardiotoxicity of DOX [79,80]. Bnip3 is known to induce various forms of cell death depending on the cell situation, and there is a report that Bnip3 contributes to DOX-induced cardiomyocyte pyroptosis [79].

1.1.7. Metabolic and Inflammation

SIRT1 protects cells from DNA damage and p53-dependent apoptosis [81] and also inhibits apoptosis by suppressing ROS production, p38 MAPK phosphorylation, and caspase-3 activation [82]. Many reports suggest that SIRT1 activation has a protective effect against DOX cardiotoxicity. Resveratrol activates SIRT1 and improves DOX cardiotoxicity [82,83,84]. AMPK also attenuates cardiotoxicity by activating SIRT1 [85,86,87]. In fact, calorie restriction and resveratrol exert protective effects against DOX cardiotoxicity via the activation of AMPK and SIRT1 and the induction of autophagy [88]. In addition, many reports show the inhibition of cardiotoxicity by activating SIRT1 through C1q/tumor necrosis factor-related protein-3 [89] and erythropoietin [90]. The p38- and p53-mediated pathways lead to apoptosis, but the pathway mediated by NF-κB is also important [91]. NF-κB activity inside and outside the mitochondria may affect mitochondrial dynamics, apoptosis, respiratory regulation, and gene expression [91]. Regarding DOX cardiotoxicity, several studies have shown that DOX activates NF-κB and causes apoptosis in cardiomyocytes, leading to cardiotoxicity [92,93,94]. As a therapeutic means of suppressing NF-κB and improving DOX cardiotoxicity, DHA [95], N-acetylcysteine [96], and berberine [97] alkaloids extracted from various herbs have been proposed. In contrast, it has been reported that DOX suppresses NF-κB signaling. Although DOX-induced upregulation of Bnip3 disrupts the homeostasis of mitochondrial dynamics and induces cell death, it has been reported that NF-κB signaling, which transcriptionally suppresses Bnip3 activation, is dramatically reduced after DOX treatment [98]. This report demonstrated that Bnip3 forms a protein complex with cyclophilin D and is involved in mPTP opening, leading to cell death.

1.2. Ferroptosis

Several studies have recently demonstrated that DOX-induced cardiotoxicity is caused by ferroptosis, a novel form of cell death [99,100,101,102,103,104,105]. Ferroptosis is a type of programmed cell death dependent on iron, and is characterized by the accumulation of lipid peroxides. The concept of ferroptosis was first reported by Dixon et al. in 2012 [106]. Since then, this programmed cell death has been implicated in various diseases [107,108,109]. In 2019, Fang et al. identified ferroptosis as the primary cause of cell death in DOX-induced cardiomyopathy [99]. They showed that only the ferroptosis inhibitor Ferrostatin-1 (Fer-1) significantly reduced DOX-induced mortality; in contrast, in mice treated with apoptosis, necroptosis, or autophagy inhibitors, survival was not significantly improved. DOX increases the nuclear accumulation of nuclear factor erythroid 2 -related factor 2 (Nrf2) and the expression of Hmox1 in cardiomyocytes, leading to the degradation of heme, resulting in the release of free iron and the formation of oxidized lipids in the mitochondrial membrane. Based on this report, the involvement of ferroptosis in the pathogenesis of DOX cardiomyopathy has attracted attention [100,101], and inhibiting this type of cell death with ferroptosis inhibitors, such as Fer-1, is expected to be a new therapeutic strategy [99,100,101]. Alpha-tocopherol, a well-known antioxidant that inhibits lipid peroxidation [110] also reduces DOX cardiotoxicity [111,112,113].

1.2.1. Apoptosis or Ferroptosis?

A new form of cell death called ferroptosis was identified after looking back on previous reports, and it was concluded that apoptosis is the central mechanism of DOX-induced cardiotoxicity. However, even among studies concluding that the final form of cell death is apoptosis, many reports are suggesting the involvement of iron in the process of cell death. For example, one trial of patients with advanced breast cancer revealed that dexrazoxane, which is the only drug that is FDA approved to prevent DOX cardiotoxicity, has a significant cardioprotective effect [114]. Mechanistically, dexrazoxane is a chelator of intracellular iron, which blocks iron-assisted oxidative radical production, suggesting the importance of iron overload in the heart as a mechanism caused by DOX [115,116]. In bovine aortic endothelial cells, iron uptake into the cells is markedly increased during DOX-induced cell death, and iron regulatory protein-1, a central cytosolic regulator of cellular iron metabolism, is activated [117]. This DOX-induced iron uptake occurs through a transferrin receptor-dependent mechanism, and the administration of anti-transferrin receptor antibodies dramatically suppress DOX-induced iron uptake, intracellular oxidant production, and cell death. In clinical practice, serum transferrin levels in patients receiving DOX chemotherapy correlate with left ventricular dysfunction severity [118]. Treatment of the HL-1 cell line with DOX caused a time-dependent increase in cytoplasmic and mitochondrial free iron pools, resulting in a loss of mitochondrial membrane potential [102]. Ferritin sequesters iron, consequently protecting cells against iron-mediated free radical damage. Mice lacking mitochondrial ferritin localized in the mitochondrial matrix exhibited enhanced DOX-induced cardiotoxicity [119]. In addition, another study demonstrated that DOX becomes concentrated in the mitochondria and increases both mitochondrial iron and cellular ROS levels [120]. The ATP-binding cassette (ABC) transporter ABCB8, localized in the mitochondrial inner membrane, is responsible for mitochondrial iron export and plays an essential role in maintaining mitochondrial iron homeostasis and maturation cytosolic Fe/S proteins [121]. An overexpression of ABCB8 in the heart reduces mitochondrial iron and intracellular ROS levels and suppresses DOX-induced cardiomyopathy [120].
Understanding the difference between these two programmed cell death pathways is important to determine whether the underlying mechanism of DOX-induced cardiotoxicity is apoptosis or ferroptosis (Table 2). In fact, the morphological changes and cell death cascade induced by apoptosis are completely different from those of ferroptosis [106]. In ferroptosis, chromatin aggregation is not observed as in apoptosis. Moreover, caspase activation was not observed during ferroptosis [122,123]. In DOX, cardiotoxicity, apoptosis, and cell death due to ferroptosis may occur simultaneously [100,101,124]. In our study, when primary cultured rat myocardial cells were treated with 5 μM DOX for 24 h, cell death caused by low-dose DOX treatment could be rescued by either the apoptosis inhibitors ZVAD or Fer-1. However, the inhibitory effect of Fer-1 on cell death was stronger than that of ZVAD [100]. Ferroptosis inhibitors do not alter caspase activation or the amount of cytochrome c in the cytosol, representing mitochondria-dependent apoptosis. In contrast, the apoptosis inhibitor ZVAD does not reduce the number of lipid peroxides that cause ferroptosis [101,124]. This result might indicate that both apoptosis and ferroptosis present DOX toxicity as independent cell death that does not affect each other. As Table 2 demonstrates, there are several apoptosis or ferroptosis inhibitors other than ZVAD and Fer-1. For instance, Emricasan, Q-VD-Oph, IDN-6556, and DEVD-CHO have been used as apoptosis inhibitors in several studies [103,125]. While Emricasan failed to show protective effects for DOX-induced cardiotoxicity [99], other apoptosis inhibitors, including Q-VD-Oph and DEVD-CHO, have not been investigated well in DOX cardiotoxicity. On the other hand, as ferroptosis inhibitors, vitamin E (α-tocopherol) and liproxstatin-1 are well-known radical trapping agents. In fact, vitamin E had been reported to play an effective antioxidant role to prevent apoptosis before ferroptosis was first reported. Indeed, as mentioned above, previous reports showed that vitamin E decreased doxorubicin-induced cardiotoxicity [113]. Although there is a scarcity of data that directly show the liproxstatin-1 efficacy for DOX cardiotoxicity, these radical trapping agents, including Fer-1, may be potential targets for DOX cardiotoxicity prevention.
In addition, DOX activates necroptosis, which, together with necrosis, causes more cell death than apoptosis [102,128]. DOX upregulates RIPK3, which binds to and phosphorylates calmodulin kinase II, which in turn regulates the opening of mPTP, leading to necroptosis and apoptosis [102]. Thus, various types of cell death are involved in DOX-induced cardiotoxicity, and the mechanisms of each type of cell death at least partially overlap.

1.2.2. Ferroptosis Regulatory Pathway

Glutathione Peroxidase 4 (GPX4)

GPX4 is a selenium-containing enzyme that uses reduced glutathione (GSH) as a co-factor to reduce lipid peroxide accumulation [129]. GPX4 is considered to be the primary enzyme that prevents ferroptosis. The ferroptosis activator RSL3 (1S,3R) induces ferroptosis by inhibiting GPX, which contains the essential microelement selenium, particularly GPX4 [130]. Tadokoro et al. demonstrated that DOX downregulates GPX4 expression and induces excessive lipid peroxidation via the mitochondrial DOX-Fe2+ complex, leading to mitochondria-dependent ferroptosis [101].

MITOL/MARCH5

As mentioned above, DOX downregulates the GPX4 expression and causes ferroptosis. However, the reason DOX decreases the expression of GPX4 is unclear. We found that the depletion of GSH in cardiomyocytes caused a decrease in the expression of GPX4 in the mitochondria [100]. MITOL/MARCH5 is an E3 ubiquitin ligase that plays an important role in regulating mitochondrial quality and function. In primary cultured neonatal rat cardiomyocytes, downregulation of MITOL expression by siRNA markedly reduced GPX4 localized in the mitochondria, promoting the accumulation of lipid peroxides in the mitochondria and induced cell death by ferroptosis. In MITOL-knockdown cells, the glutathione-degrading enzyme ChaC Glutathione Specific Gamma-Glutamylcyclotransferase 1 (CHAC1) expression was upregulated, resulting in a decrease in the glutathione/oxidized glutathione (GSH/GSSG) ratio. Improving the GSH/GSSG ratio through the administration of N-acetylcysteine or knockdown of CHAC1 suppressed ferroptosis by recovering the expression of GPX4 in MITOL knockdown NRVMs. In DOX-treated cultured cardiomyocytes, both MITOL and GPX4 decreased, but the forced expression of MITOL maintained the GPX4 content and prevented DOX-induced ferroptosis. Sensitivity to DOX cardiotoxicity increased in mice lacking MITOL. These results suggest that MITOL plays a role in protecting cardiomyocytes from DOX-induced toxicity by regulating mitochondrial glutathione levels and GPX4 expression. As ferroptosis caused by MITOL knockdown is a cardiomyocyte-specific phenomenon, the authors believe that it may explain at least part of the mechanism through which cardiomyocytes are more dysfunctional after DOX treatment than other cells. If it is possible for us to examine the expression of MITOL in the heart, we may be able to conduct risk stratification of cardiotoxicity caused by DOX. Moreover, compounds that upregulate MITOL may be a promising strategy to mitigate DOX-induced cardiotoxicity. Other studies have shown that the administration of GSH and glutamate, necessary for GSH synthesis, can reduce the cardiotoxicity of DOX [131,132,133]. GSH, a co-factor of GPX4, is a tripeptide of cysteine, glutamate, and glycine. Cysteine is often depleted in the cells. Cystine/glutamate antiporter solute carrier family 7 member 11 (SLC7A11, also known as System Xc−) promotes cystine uptake and glutathione biosynthesis. Erastin and sulfasalazine inhibit GSH synthesis by inhibiting SLC7A11, leading to ferroptosis [134].

Nrf2

Nrf2 and its repressor Kelch-like ECH-associated protein 1 (KEAP1) are key metabolism, oxidative stress, and inflammation regulators. In recent years, several reports have advocated the regulation of DOX cardiotoxicity by Nrf2 [135,136,137]. Bardoxolone methyl, an activator of the Nrf2-KEAP1 pathway and a potential treatment for chronic kidney disease, reduces DOX-induced cardiotoxicity in 3D cardiac spheroids, in which human cardiomyocytes are grown in combination with human cardiac fibroblasts and endothelial cells. Exposure of hepatocellular carcinoma cells (HCC) to ferroptosis-inducing compounds, such as elastin and sorafenib, promotes the nuclear accumulation of Nrf2 through the inactivation of KEAP1 by p62 expression. Knockdown of p62, quinone oxidoreductase-1, Hmox-1, and ferritin heavy chain-1 promotes ferroptosis in HCC cells in response to elastin and sorafenib [107]. The ubiquitin E3 ligase TRIM21 interacts with p62 and ubiquitinates it, negatively regulating the p62-KEAP1-Nrf2 antioxidant pathway [138]. Cardiac tissue lacking TRIM21 is protected from DOX-induced ferroptosis by homeostatic activation of Nrf2. As mentioned above, selenium inhibits ferroptosis by promoting the expression of the antioxidant GPX4; however, selenium also exerts a protective effect against DOX-induced cardiotoxicity by inhibiting the Nrf2-NLRP3 pathway [139]. It has been reported that Hemin stimulates Nrf-2/Hmox-1 and inhibits the TLR-5/NF-κB/TNF-α pathway, thereby exerting antioxidant and anti-inflammatory effects and reducing DOX-induced cardiotoxicity in a dose-dependent manner [140]. In contrast, there are reports that Nrf2 activation is involved in ferroptosis induction. Hassannia et al. reported that elevated Nrf2 activates Hmox1, increasing the amount of unstable Fe(II) ions, resulting in lipid peroxides production, ultimately inducing ferroptosis [141]. Fang et al. also concluded that DOX causes an increase in Nrf2, leading to cardiotoxicity via a similar mechanism [99].

Ferroptosis Suppressor Protein 1 (FSP1)

Another known defense mechanism against ferroptosis is FSP1, a GPX4- and GSH-independent ubiquinone reductase [142,143]. FSP1 was described as a pro-apoptotic gene apoptosis-inducing factor mitochondria-associated 2 (AIFM2) before it was renamed. The reduced ubiquinol scavenges peroxyl radicals (ROO•) causing lipid peroxidation. FSP1 uses NAD(P)H to catalyze the regeneration of ubiquinone (coenzyme Q10 (CoQ10)). To date, no evidence has been published regarding the direct relationship between FSP1 and DOX cardiotoxicity, but the fact that the administration of CoQ10 minimized DOX-induced toxicity, such as ECG changes, oxidative stress, lipid peroxidation, and changes in cardiac tissue, may suggest an association between DOX-induced cardiotoxicity and FSP1 [144,145].

2. Mitochondria Providing a Place for DOX-Induced Cardiotoxicity

Whether the mechanism of DOX cardiotoxicity is apoptosis or ferroptosis, the mitochondria are important places where toxicity is exerted [146,147,148,149,150,151,152,153,154,155]. It has been theorized that mitochondria-mediated apoptosis (endogenous pathway) is one of the mechanisms of DOX cardiotoxicity [147,150,156,157,158,159,160,161,162,163]. Mitochondria also play a central role in ferroptosis induction [99,100,101]. DOX disrupts protein complexes between major respiratory chain proteins, such as uncoupling protein 3 and cytochrome c oxidase, and induces abnormal mitochondrial respiration and various types of cell death through a Bnip3-dependent mechanism [80,98,162]. Mitochondria retain their homeostasis through repeated fission and fusion. DOX fragments mitochondria by inducing the activation of dynamin protein 1 (Drp1), which is important for mitochondrial fission [157,163,164]. Conversely, the induction of the mitochondrial fusion factor Mitofusin2 by DOX acts protectively against DOX-induced toxicity [164]. In addition, the involvement of mitochondrial fission protein 1 (Mtfp1), a new molecule regulating mitochondrial dynamics, has also been proposed [165]. Mechanistically, DOX upregulates the Mtfp1 expression, which induces mitochondrial fission and apoptosis. Moreover, in DOX-treated cardiomyocytes, the decreased expression of a long non-coding RNA called CMDL-1, which is involved in post-translational modification (phosphorylation) of Drp1, has also been reported to be associated with Drp1-induced promotion of mitochondrial fission and cell death [166]. Collectively, there is no shortage of evidence suggesting the importance of mitochondria in DOX cardiotoxicity.

3. Conclusions

This article summarizes the mechanisms involved in DOX cardiotoxicity, mainly related to two forms of cell death, apoptosis and ferroptosis, by focusing on the basic research in this century. Numerous previous studies suggest that various types of cell death are involved in DOX cardiotoxicity. The underlying mechanisms that induce multiple types of cell death and their common regulators may serve as therapeutic targets to alleviate DOX-induced cardiotoxicity. We await the results of basic research that will lead to a therapeutic strategy to mitigate cardiotoxicity, while maintaining the high anti-tumor efficacy of DOX or inhibiting the progression of delayed cardiac dysfunction after cancer eradication.

Author Contributions

Writing and editing, H.K., J.E., H.I., H.M., K.S., Y.K. and M.S.; conceptualization and supervision, M.S. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Rosen, M.R.; Myerburg, R.J.; Francis, D.P.; Cole, G.D.; Marbán, E. Translating Stem Cell Research to Cardiac Disease Therapies: Pitfalls and Prospects for Improvement. J. Am. Coll. Cardiol. 2014, 64, 922–937. [Google Scholar] [CrossRef] [Green Version]
  2. Ewer, M.S.; Ewer, S.M. Cardiotoxicity of Anticancer Treatments. Nat. Rev. Cardiol. 2015, 12, 620. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  3. Armstrong, G.T.; Oeffinger, K.C.; Chen, Y.; Kawashima, T.; Yasui, Y.; Leisenring, W.; Stovall, M.; Chow, E.J.; Sklar, C.A.; Mulrooney, D.A.; et al. Modifiable Risk Factors and Major Cardiac Events among Adult Survivors of Childhood Cancer. J. Clin. Oncol. 2013, 31, 3673–3680. [Google Scholar] [CrossRef]
  4. Patnaik, J.L.; Byers, T.; DiGuiseppi, C.; Dabelea, D.; Denberg, T.D. Cardiovascular Disease Competes with Breast Cancer as the Leading Cause of Death for Older Females Diagnosed with Breast Cancer: A Retrospective Cohort Study. Breast Cancer Res. 2011, 13, R64. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Abdel-Qadir, H.; Austin, P.C.; Lee, D.S.; Amir, E.; Tu, J.V.; Thavendiranathan, P.; Fung, K.; Anderson, G.M. A Population-Based Study of Cardiovascular Mortality Following Early-Stage Breast Cancer. JAMA Cardiol. 2017, 2, 88–93. [Google Scholar] [CrossRef] [PubMed]
  6. Hasin, T.; Gerber, Y.; McNallan, S.M.; Weston, S.A.; Kushwaha, S.S.; Nelson, T.J.; Cerhan, J.R.; Roger, V.L. Patients with Heart Failure Have an Increased Risk of Incident Cancer. J. Am. Coll. Cardiol. 2013, 62, 881–886. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Stoltzfus, K.C.; Zhang, Y.; Sturgeon, K.; Sinoway, L.I.; Trifiletti, D.M.; Chinchilli, V.M.; Zaorsky, N.G. Fatal Heart Disease among Cancer Patients. Nat. Commun. 2020, 11, 2011. [Google Scholar] [CrossRef] [PubMed]
  8. Parent, S.; Pituskin, E.; Paterson, D.I. The Cardio-Oncology Program: A Multidisciplinary Approach to the Care of Cancer Patients With Cardiovascular Disease. Can. J. Cardiol. 2016, 32, 847–851. [Google Scholar] [CrossRef] [PubMed]
  9. Ewer, M.S.; Lippman, S.M. Type II Chemotherapy-Related Cardiac Dysfunction: Time to Recognize a New Entity. J. Clin. Oncol. 2005, 23, 2900–2902. [Google Scholar] [CrossRef]
  10. Zhang, Z.; Yu, X.; Wang, Z.; Wu, P.; Huang, J. Anthracyclines Potentiate Anti-Tumor Immunity: A New Opportunity for Chemoimmunotherapy. Cancer Lett. 2015, 369, 331–335. [Google Scholar] [CrossRef]
  11. Hortobágyi, G.N. Anthracyclines in the Treatment of Cancer. An Overview. Drugs 1997, 54, 1–7. [Google Scholar] [CrossRef] [PubMed]
  12. Sawicki, K.T.; Sala, V.; Prever, L.; Hirsch, E.; Ardehali, H.; Ghigo, A. Preventing and Treating Anthracycline Cardiotoxicity: New Insights. Annu. Rev. Pharmacol. Toxicol. 2021, 61, 309–332. [Google Scholar] [CrossRef]
  13. Wallace, K.B.; Sardão, V.A.; Oliveira, P.J. Mitochondrial Determinants of Doxorubicin-Induced Cardiomyopathy. Circ. Res. 2020, 126, 926–941. [Google Scholar] [CrossRef]
  14. Cui, N.; Wu, F.; Lu, W.J.; Bai, R.; Ke, B.; Liu, T.; Li, L.; Lan, F.; Cui, M. Doxorubicin-Induced Cardiotoxicity Is Maturation Dependent due to the Shift from Topoisomerase IIα to IIβ in Human Stem Cell Derived Cardiomyocytes. J. Cell. Mol. Med. 2019, 23, 4627–4639. [Google Scholar] [CrossRef] [Green Version]
  15. Kalyanaraman, B. Teaching the Basics of the Mechanism of Doxorubicin-Induced Cardiotoxicity: Have We Been Barking up the Wrong Tree? Redox. Biol. 2020, 29, 101394. [Google Scholar] [CrossRef] [PubMed]
  16. Vejpongsa, P.; Yeh, E.T. Topoisomerase 2β: A Promising Molecular Target for Primary Prevention of Anthracycline-Induced Cardiotoxicity. Clin. Pharmacol. Ther. 2014, 95, 45–52. [Google Scholar] [CrossRef] [PubMed]
  17. Zhang, S.; Liu, X.; Bawa-Khalfe, T.; Lu, L.S.; Lyu, Y.L.; Liu, L.F.; Yeh, E.T. Identification of the Molecular Basis of Doxorubicin-induced Cardiotoxicity. Nat. Med. 2012, 18, 1639–1642. [Google Scholar] [CrossRef] [PubMed]
  18. Jiang, J.; Mohan, N.; Endo, Y.; Shen, Y.; Wu, W.J. Type IIB DNA Topoisomerase Is Downregulated by Trastuzumab and Doxorubicin to Synergize Cardiotoxicity. Oncotarget 2018, 9, 6095–6108. [Google Scholar] [CrossRef] [Green Version]
  19. Van Dalen, E.C.; Caron, H.N.; Dickinson, H.O.; Kremer, L.C. Cardioprotective Interventions for Cancer Patients Receiving Anthracyclines. Cochrane Database Syst. Rev. 2011, 2011, Cd003917. [Google Scholar] [CrossRef] [PubMed]
  20. Tebbi, C.K.; London, W.B.; Friedman, D.; Villaluna, D.; De Alarcon, P.A.; Constine, L.S.; Mendenhall, N.P.; Sposto, R.; Chauvenet, A.; Schwartz, C.L. Dexrazoxane-Associated Risk for Acute Myeloid Leukemia/Myelodysplastic Syndrome and Other Secondary Malignancies in Pediatric Hodgkin′s Disease. J. Clin. Oncol. 2007, 25, 493–500. [Google Scholar] [CrossRef]
  21. Rabelo, E.; De Angelis, K.; Bock, P.; Gatelli Fernandes, T.; Cervo, F.; Belló Klein, A.; Clausell, N.; Cláudia Irigoyen, M. Baroreflex Sensitivity and Oxidative Stress in Adriamycin-Induced Heart Failure. Hypertension 2001, 38, 576–580. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  22. Wattanapitayakul, S.K.; Chularojmontri, L.; Herunsalee, A.; Charuchongkolwongse, S.; Niumsakul, S.; Bauer, J.A. Screening of Antioxidants from Medicinal Plants for Cardioprotective Effect against Doxorubicin Toxicity. Basic Clin. Pharmacol. Toxicol. 2005, 96, 80–87. [Google Scholar] [CrossRef] [PubMed]
  23. Burridge, P.W.; Li, Y.F.; Matsa, E.; Wu, H.; Ong, S.G.; Sharma, A.; Holmström, A.; Chang, A.C.; Coronado, M.J.; Ebert, A.D.; et al. Human Induced Pluripotent Stem Cell-Derived Cardiomyocytes Recapitulate the Predilection of Breast Cancer Patients to Doxorubicin-Induced Cardiotoxicity. Nat. Med. 2016, 22, 547–556. [Google Scholar] [CrossRef] [Green Version]
  24. Arafa, M.H.; Mohammad, N.S.; Atteia, H.H.; Abd-Elaziz, H.R. Protective Effect of Resveratrol against Doxorubicin-Induced Cardiac Toxicity and Fibrosis in Male Experimental Rats. J. Physiol. Biochem. 2014, 70, 701–711. [Google Scholar] [CrossRef] [PubMed]
  25. Abe, J.; Yamada, Y.; Takeda, A.; Harashima, H. Cardiac Progenitor Cells Activated by Mitochondrial Delivery of Resveratrol Enhance the Survival of a Doxorubicin-Induced Cardiomyopathy Mouse Model via the Mitochondrial Activation of a Damaged Myocardium. J. Control. Release 2018, 269, 177–188. [Google Scholar] [CrossRef] [PubMed]
  26. Chen, Y.; Daosukho, C.; Opii, W.O.; Turner, D.M.; Pierce, W.M.; Klein, J.B.; Vore, M.; Butterfield, D.A.; St Clair, D.K. Redox Proteomic Identification of Oxidized Cardiac Proteins in Adriamycin-Treated Mice. Free Radic. Biol. Med. 2006, 41, 1470–1477. [Google Scholar] [CrossRef] [PubMed]
  27. Chaiswing, L.; Cole, M.P.; St Clair, D.K.; Ittarat, W.; Szweda, L.I.; Oberley, T.D. Oxidative Damage Precedes Nitrative Damage in Adriamycin-Induced Cardiac Mitochondrial Injury. Toxicol. Pathol. 2004, 32, 536–547. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  28. Gilleron, M.; Marechal, X.; Montaigne, D.; Franczak, J.; Neviere, R.; Lancel, S. NADPH Oxidases Participate to Doxorubicin-Induced Cardiac Myocyte Apoptosis. Biochem. Biophys. Res. Commun. 2009, 388, 727–731. [Google Scholar] [CrossRef] [PubMed]
  29. Ascensão, A.; Lumini-Oliveira, J.; Machado, N.G.; Ferreira, R.M.; Gonçalves, I.O.; Moreira, A.C.; Marques, F.; Sardão, V.A.; Oliveira, P.J.; Magalhães, J. Acute Exercise Protects against Calcium-Induced Cardiac Mitochondrial Permeability Transition Pore Opening in Doxorubicin-Treated Rats. Clin. Sci. 2011, 120, 37–49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  30. Gao, J.; Xiong, Y.; Ho, Y.S.; Liu, X.; Chua, C.C.; Xu, X.; Wang, H.; Hamdy, R.; Chua, B.H. Glutathione Peroxidase 1-Deficient Mice Are More Susceptible to Doxorubicin-Induced Cardiotoxicity. Biochim. Biophys. Acta 2008, 1783, 2020–2029. [Google Scholar] [CrossRef] [Green Version]
  31. Lam, C.K.; Wu, J.C. Clinical Trial in a Dish: Using Patient-Derived Induced Pluripotent Stem Cells to Identify Risks of Drug-Induced Cardiotoxicity. Arterioscler. Thromb. Vasc. Biol. 2021, 41, 1019–1031. [Google Scholar] [CrossRef] [PubMed]
  32. Keshet, Y.; Seger, R. The MAP Kinase Signaling Cascades: A System of Hundreds of Components Regulates a Diverse Array of Physiological Functions. Methods Mol. Biol. 2010, 661, 3–38. [Google Scholar] [CrossRef]
  33. Wagner, E.F.; Nebreda, A.R. Signal Integration by JNK and p38 MAPK Pathways in Cancer Development. Nat. Rev. Cancer 2009, 9, 537–549. [Google Scholar] [CrossRef] [PubMed]
  34. Spallarossa, P.; Altieri, P.; Garibaldi, S.; Ghigliotti, G.; Barisione, C.; Manca, V.; Fabbi, P.; Ballestrero, A.; Brunelli, C.; Barsotti, A. Matrix Metalloproteinase-2 and -9 Are Induced Differently by Doxorubicin in H9c2 Cells: The Role of MAP Kinases and NAD(P)H Oxidase. Cardiovasc. Res. 2006, 69, 736–745. [Google Scholar] [CrossRef] [PubMed]
  35. Thandavarayan, R.A.; Watanabe, K.; Sari, F.R.; Ma, M.; Lakshmanan, A.P.; Giridharan, V.V.; Gurusamy, N.; Nishida, H.; Konishi, T.; Zhang, S.; et al. Modulation of Doxorubicin-Induced Cardiac Dysfunction in Dominant-Negative p38α Mitogen-Activated Protein KINASE mice. Free Radic. Biol. Med. 2010, 49, 1422–1431. [Google Scholar] [CrossRef] [PubMed]
  36. Guo, R.; Lin, J.; Xu, W.; Shen, N.; Mo, L.; Zhang, C.; Feng, J. Hydrogen Sulfide Attenuates Doxorubicin-Induced Cardiotoxicity by Inhibition of the p38 MAPK Pathway in H9c2 Cells. Int. J. Mol. Med. 2013, 31, 644–650. [Google Scholar] [CrossRef]
  37. George, S.A.; Kiss, A.; Obaid, S.N.; Venegas, A.; Talapatra, T.; Wei, C.; Efimova, T.; Efimov, I.R. p38δ Genetic Ablation Protects Female Mice from Anthracycline Cardiotoxicity. Am. J. Physiol. Heart Circ. Physiol. 2020, 319, H775–H786. [Google Scholar] [CrossRef] [PubMed]
  38. Maruyama, S.; Shibata, R.; Ohashi, K.; Ohashi, T.; Daida, H.; Walsh, K.; Murohara, T.; Ouchi, N. Adiponectin Ameliorates Doxorubicin-Induced Cardiotoxicity through Akt Protein-Dependent Mechanism. J. Biol. Chem. 2011, 286, 32790–32800. [Google Scholar] [CrossRef] [Green Version]
  39. Lai, H.C.; Liu, T.J.; Ting, C.T.; Sharma, P.M.; Wang, P.H. Insulin-like Growth Factor-1 Prevents Loss of Electrochemical Gradient in Cardiac Muscle Mitochondria via Activation of PI 3 Kinase/Akt Pathway. Mol. Cell. Endocrinol. 2003, 205, 99–106. [Google Scholar] [CrossRef]
  40. Hang, P.; Zhao, J.; Sun, L.; Li, M.; Han, Y.; Du, Z.; Li, Y. Brain-Derived Neurotrophic Factor Attenuates Doxorubicin-Induced Cardiac Dysfunction through Activating Akt Signalling in Rats. J. Cell. Mol. Med. 2017, 21, 685–696. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  41. Li, L.L.; Wei, L.; Zhang, N.; Wei, W.Y.; Hu, C.; Deng, W.; Tang, Q.Z. Levosimendan Protects against Doxorubicin-Induced Cardiotoxicity by Regulating the PTEN/Akt Pathway. BioMed Res. Int. 2020, 2020, 8593617. [Google Scholar] [CrossRef] [PubMed]
  42. Cao, Y.; Ruan, Y.; Shen, T.; Huang, X.; Li, M.; Yu, W.; Zhu, Y.; Man, Y.; Wang, S.; Li, J. Astragalus Polysaccharide Suppresses Doxorubicin-Induced Cardiotoxicity by Regulating the PI3k/Akt and p38MAPK Pathways. Autoxid. Med. Cell. Longev. 2014, 2014, 674219. [Google Scholar] [CrossRef] [Green Version]
  43. Zhang, X.; Hu, C.; Kong, C.Y.; Song, P.; Wu, H.M.; Xu, S.C.; Yuan, Y.P.; Deng, W.; Ma, Z.G.; Tang, Q.Z. FNDC5 Alleviates Oxidative Stress and Cardiomyocyte Apoptosis in Doxorubicin-Induced Cardiotoxicity via Activating AKT. Cell. Death Differ. 2020, 27, 540–555. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Wen, S.Y.; Tsai, C.Y.; Pai, P.Y.; Chen, Y.W.; Yang, Y.C.; Aneja, R.; Huang, C.Y.; Kuo, W.W. Diallyl Trisulfide Suppresses Doxorubicin-Induced Cardiomyocyte Apoptosis by Inhibiting MAPK/NF-κB Signaling through Attenuation of ROS Generation. Environ. Toxicol. 2018, 33, 93–103. [Google Scholar] [CrossRef] [PubMed]
  45. Hu, Y.H.; Liu, J.; Lu, J.; Wang, P.X.; Chen, J.X.; Guo, Y.; Han, F.H.; Wang, J.J.; Li, W.; Liu, P.Q. sFRP1 Protects H9c2 Cardiac Myoblasts from Doxorubicin-Induced Apoptosis by Inhibiting the Wnt/PCP-JNK Pathway. Acta Pharmacol. Sin. 2020, 41, 1150–1157. [Google Scholar] [CrossRef] [PubMed]
  46. Boutelle, A.M.; Attardi, L.D. p53 and Tumor Suppression: It Takes a Network. Trends Cell. Biol. 2021, 31, 298–310. [Google Scholar] [CrossRef] [PubMed]
  47. Chua, C.C.; Liu, X.; Gao, J.; Hamdy, R.C.; Chua, B.H. Multiple Actions of Pifithrin-Alpha on Doxorubicin-Induced Apoptosis in Rat Myoblastic H9c2 Cells. Am. J. Physiol. Heart Circ. Physiol. 2006, 290, H2606–H2613. [Google Scholar] [CrossRef] [PubMed]
  48. Zhang, C.; Feng, Y.; Qu, S.; Wei, X.; Zhu, H.; Luo, Q.; Liu, M.; Chen, G.; Xiao, X. Resveratrol Attenuates Doxorubicin-Induced Cardiomyocyte Apoptosis in Mice through SIRT1-Mediated Deacetylation of p53. Cardiovasc. Res. 2011, 90, 538–545. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  49. McSweeney, K.M.; Bozza, W.P.; Alterovitz, W.L.; Zhang, B. Transcriptomic Profiling Reveals p53 as a Key Regulator of Doxorubicin-Induced Cardiotoxicity. Cell. Death Discov. 2019, 5, 102. [Google Scholar] [CrossRef] [Green Version]
  50. Saleme, B.; Das, S.K.; Zhang, Y.; Boukouris, A.E.; Lorenzana Carrillo, M.A.; Jovel, J.; Wagg, C.S.; Lopaschuk, G.D.; Michelakis, E.D.; Sutendra, G. p53-Mediated Repression of the PGC1A (PPARG Coactivator 1α) and APLNR (Apelin Receptor) Signaling Pathways Limits Fatty Acid Oxidation Energetics: Implications for Cardio-oncology. J. Am. Heart Assoc. 2020, 9, e017247. [Google Scholar] [CrossRef]
  51. Ma, J.; Wang, Y.; Zheng, D.; Wei, M.; Xu, H.; Peng, T. Rac1 Signalling Mediates Doxorubicin-Induced Cardiotoxicity through both Reactive Oxygen Species-Dependent and -Independent Pathways. Cardiovasc. Res. 2013, 97, 77–87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  52. Li, J.; Wang, P.Y.; Long, N.A.; Zhuang, J.; Springer, D.A.; Zou, J.; Lin, Y.; Bleck, C.K.E.; Park, J.H.; Kang, J.G.; et al. p53 Prevents Doxorubicin Cardiotoxicity Independently of Its Prototypical Tumor Suppressor Activities. Proc. Natl. Acad. Sci. USA 2019, 116, 19626–19634. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Shizukuda, Y.; Matoba, S.; Mian, O.Y.; Nguyen, T.; Hwang, P.M. Targeted Disruption of p53 Attenuates Doxorubicin-Induced Cardiac Toxicity in Mice. Mol. Cell. Biochem. 2005, 273, 25–32. [Google Scholar] [CrossRef] [PubMed]
  54. Zhu, W.; Zhang, W.; Shou, W.; Field, L.J. P53 Inhibition Exacerbates Late-Stage Anthracycline Cardiotoxicity. Cardiovasc. Res. 2014, 103, 81–89. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Feridooni, T.; Hotchkiss, A.; Remley-Carr, S.; Saga, Y.; Pasumarthi, K.B. Cardiomyocyte Specific Ablation of p53 Is Not Sufficient to Block Doxorubicin Induced Cardiac Fibrosis and Associated Cytoskeletal Changes. PLoS ONE 2011, 6, e22801. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Heineke, J.; Auger-Messier, M.; Xu, J.; Oka, T.; Sargent, M.A.; York, A.; Klevitsky, R.; Vaikunth, S.; Duncan, S.A.; Aronow, B.J.; et al. Cardiomyocyte GATA4 Functions as a Stress-Responsive Regulator of Angiogenesis in the Murine Heart. J. Clin. Investig. 2007, 117, 3198–3210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Kim, Y.; Ma, A.G.; Kitta, K.; Fitch, S.N.; Ikeda, T.; Ihara, Y.; Simon, A.R.; Evans, T.; Suzuki, Y.J. Anthracycline-Induced Suppression of GATA-4 Transcription Factor: Implication in the Regulation of Cardiac Myocyte Apoptosis. Mol. Pharmacol. 2003, 63, 368–377. [Google Scholar] [CrossRef]
  58. Kobayashi, S.; Volden, P.; Timm, D.; Mao, K.; Xu, X.; Liang, Q. Transcription Factor GATA4 Inhibits Doxorubicin-Induced Autophagy and Cardiomyocyte Death. J. Biol. Chem. 2010, 285, 793–804. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Chen, B.; Zhong, L.; Roush, S.F.; Pentassuglia, L.; Peng, X.; Samaras, S.; Davidson, J.M.; Sawyer, D.B.; Lim, C.C. Disruption of a GATA4/Ankrd1 Signaling Axis in Cardiomyocytes Leads to Sarcomere Disarray: Implications for Anthracycline Cardiomyopathy. PLoS ONE 2012, 7, e35743. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  60. Kumari, H.; Huang, W.H.; Chan, M.W.Y. Review on the Role of Epigenetic Modifications in Doxorubicin-Induced Cardiotoxicity. Front. Cardiovasc. Med. 2020, 7, 56. [Google Scholar] [CrossRef] [PubMed]
  61. Ferreira, L.L.; Cunha-Oliveira, T.; Veloso, C.D.; Costa, C.F.; Wallace, K.B.; Oliveira, P.J. Single Nanomolar Doxorubicin Exposure Triggers Compensatory Mitochondrial Responses in H9c2 Cardiomyoblasts. Food Chem. Toxicol. 2019, 124, 450–461. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  62. Song, R.; Yang, Y.; Lei, H.; Wang, G.; Huang, Y.; Xue, W.; Wang, Y.; Yao, L.; Zhu, Y. HDAC6 Inhibition Protects Cardiomyocytes against Doxorubicin-Induced Acute Damage by Improving α-Tubulin Acetylation. J. Mol. Cell. Cardiol. 2018, 124, 58–69. [Google Scholar] [CrossRef] [PubMed]
  63. Piotrowska, I.; Isalan, M.; Mielcarek, M. Early Transcriptional Alteration of Histone Deacetylases in a Murine Model of Doxorubicin-Induced Cardiomyopathy. PLoS ONE 2017, 12, e0180571. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Hanf, A.; Oelze, M.; Manea, A.; Li, H.; Münzel, T.; Daiber, A. The Anti-Cancer Drug Doxorubicin Induces Substantial Epigenetic Changes in Cultured Cardiomyocytes. Chem. Biol. Interact. 2019, 313, 108834. [Google Scholar] [CrossRef] [PubMed]
  65. Dykxhoorn, D.M.; Novina, C.D.; Sharp, P.A. Killing the Messenger: Short RNAs that Silence Gene Expression. Nat. Rev. Mol. Cell. Biol. 2003, 4, 457–467. [Google Scholar] [CrossRef] [PubMed]
  66. Piegari, E.; Russo, R.; Cappetta, D.; Esposito, G.; Urbanek, K.; Dell′Aversana, C.; Altucci, L.; Berrino, L.; Rossi, F.; De Angelis, A. MicroRNA-34a Regulates Doxorubicin-Induced Cardiotoxicity in Rat. Oncotarget 2016, 7, 62312–62326. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  67. Zhu, J.N.; Fu, Y.H.; Hu, Z.Q.; Li, W.Y.; Tang, C.M.; Fei, H.W.; Yang, H.; Lin, Q.X.; Gou, D.M.; Wu, S.L.; et al. Activation of miR-34a-5p/Sirt1/p66shc Pathway Contributes to Doxorubicin-Induced Cardiotoxicity. Sci. Rep. 2017, 7, 11879. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  68. Du, J.; Hang, P.; Pan, Y.; Feng, B.; Zheng, Y.; Chen, T.; Zhao, L.; Du, Z. Inhibition of miR-23a Attenuates Doxorubicin-Induced Mitochondria-Dependent Cardiomyocyte Apoptosis by Targeting the PGC-1α/Drp1 Pathway. Toxicol. Appl. Pharmacol. 2019, 369, 73–81. [Google Scholar] [CrossRef] [PubMed]
  69. Gupta, S.K.; Garg, A.; Avramopoulos, P.; Engelhardt, S.; Streckfuss-Bömeke, K.; Batkai, S.; Thum, T. miR-212/132 Cluster Modulation Prevents Doxorubicin-Mediated Atrophy and Cardiotoxicity. Mol. Ther. 2019, 27, 17–28. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  70. Berezin, A. Epigenetics in Heart Failure Phenotypes. BBA Clin. 2016, 6, 31–37. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Bartlett, J.J.; Trivedi, P.C.; Pulinilkunnil, T. Autophagic Dysregulation in Doxorubicin Cardiomyopathy. J. Mol. Cell. Cardiol. 2017, 104, 1–8. [Google Scholar] [CrossRef] [PubMed]
  72. Lee, K.H.; Cho, H.; Lee, S.; Woo, J.S.; Cho, B.H.; Kang, J.H.; Jeong, Y.M.; Cheng, X.W.; Kim, W. Enhanced-Autophagy by Exenatide Mitigates Doxorubicin-Induced Cardiotoxicity. Int. J. Cardiol. 2017, 232, 40–47. [Google Scholar] [CrossRef]
  73. Lv, X.; Zhu, Y.; Deng, Y.; Zhang, S.; Zhang, Q.; Zhao, B.; Li, G. Glycyrrhizin Improved Autophagy Flux via HMGB1-Dependent Akt/mTOR Signaling Pathway to Prevent Doxorubicin-Induced Cardiotoxicity. Toxicology 2020, 441, 152508. [Google Scholar] [CrossRef] [PubMed]
  74. Yin, J.; Guo, J.; Zhang, Q.; Cui, L.; Zhang, L.; Zhang, T.; Zhao, J.; Li, J.; Middleton, A.; Carmichael, P.L.; et al. Doxorubicin-Induced Mitophagy and Mitochondrial Damage Is Associated with Dysregulation of the PINK1/Parkin Pathway. Toxicol. In Vitro 2018, 51, 1–10. [Google Scholar] [CrossRef] [PubMed]
  75. Li, M.; Sala, V.; De Santis, M.C.; Cimino, J.; Cappello, P.; Pianca, N.; Di Bona, A.; Margaria, J.P.; Martini, M.; Lazzarini, E.; et al. Phosphoinositide 3-Kinase Gamma Inhibition Protects From Anthracycline Cardiotoxicity and Reduces Tumor Growth. Circulation 2018, 138, 696–711. [Google Scholar] [CrossRef]
  76. Wang, X.; Wang, X.L.; Chen, H.L.; Wu, D.; Chen, J.X.; Wang, X.X.; Li, R.L.; He, J.H.; Mo, L.; Cen, X.; et al. Ghrelin Inhibits Doxorubicin Cardiotoxicity by Inhibiting Excessive Autophagy through AMPK and p38-MAPK. Biochem. Pharmacol. 2014, 88, 334–350. [Google Scholar] [CrossRef]
  77. Xu, X.; Chen, K.; Kobayashi, S.; Timm, D.; Liang, Q. Resveratrol Attenuates Doxorubicin-Induced Cardiomyocyte Death via Inhibition of p70 S6 Kinase 1-Mediated Autophagy. J. Pharmacol. Exp. Ther. 2012, 341, 183–195. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Chourasia, A.H.; Macleod, K.F. Tumor Suppressor Functions of BNIP3 and Mitophagy. Autophagy 2015, 11, 1937–1938. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Dhingra, A.; Jayas, R.; Afshar, P.; Guberman, M.; Maddaford, G.; Gerstein, J.; Lieberman, B.; Nepon, H.; Margulets, V.; Dhingra, R.; et al. Ellagic Acid Antagonizes Bnip3-Mediated Mitochondrial Injury and Necrotic Cell Death of Cardiac Myocytes. Free Radic. Biol. Med. 2017, 112, 411–422. [Google Scholar] [CrossRef] [PubMed]
  80. Zheng, X.; Zhong, T.; Ma, Y.; Wan, X.; Qin, A.; Yao, B.; Zou, H.; Song, Y.; Yin, D. Bnip3 Mediates Doxorubicin-Induced Cardiomyocyte Pyroptosis via Caspase-3/GSDME. Life Sci. 2020, 242, 117186. [Google Scholar] [CrossRef] [PubMed]
  81. Ong, A.L.C.; Ramasamy, T.S. Role of Sirtuin1-p53 Regulatory Axis in Aging, Cancer and Cellular Reprogramming. Ageing Res. Rev. 2018, 43, 64–80. [Google Scholar] [CrossRef] [PubMed]
  82. Ruan, Y.; Dong, C.; Patel, J.; Duan, C.; Wang, X.; Wu, X.; Cao, Y.; Pu, L.; Lu, D.; Shen, T.; et al. SIRT1 Suppresses Doxorubicin-Induced Cardiotoxicity by Regulating the Oxidative Stress and p38MAPK Pathways. Cell. Physiol. Biochem. 2015, 35, 1116–1124. [Google Scholar] [CrossRef] [PubMed]
  83. Lou, Y.; Wang, Z.; Xu, Y.; Zhou, P.; Cao, J.; Li, Y.; Chen, Y.; Sun, J.; Fu, L. Resveratrol Prevents Doxorubicin-Induced Cardiotoxicity in H9c2 Cells through the Inhibition of Endoplasmic Reticulum Stress and the Activation of the Sirt1 Pathway. Int. J. Mol. Med. 2015, 36, 873–880. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  84. Cappetta, D.; Esposito, G.; Piegari, E.; Russo, R.; Ciuffreda, L.P.; Rivellino, A.; Berrino, L.; Rossi, F.; De Angelis, A.; Urbanek, K. SIRT1 Activation Attenuates Diastolic Dysfunction by Reducing Cardiac Fibrosis in a Model of Anthracycline Cardiomyopathy. Int. J. Cardiol. 2016, 205, 99–110. [Google Scholar] [CrossRef]
  85. Hu, C.; Zhang, X.; Song, P.; Yuan, Y.P.; Kong, C.Y.; Wu, H.M.; Xu, S.C.; Ma, Z.G.; Tang, Q.Z. Meteorin-like Protein Attenuates Doxorubicin-Induced Cardiotoxicity via Activating cAMP/PKA/SIRT1 Pathway. Redox. Biol. 2020, 37, 101747. [Google Scholar] [CrossRef] [PubMed]
  86. Wu, W.Y.; Cui, Y.K.; Hong, Y.X.; Li, Y.D.; Wu, Y.; Li, G.; Li, G.R.; Wang, Y. Doxorubicin Cardiomyopathy Is Ameliorated by Acacetin via Sirt1-Mediated Activation of AMPK/Nrf2 Signal Molecules. J. Cell. Mol. Med. 2020, 24, 12141–12153. [Google Scholar] [CrossRef]
  87. Liu, D.; Ma, Z.; Xu, L.; Zhang, X.; Qiao, S.; Yuan, J. PGC1α Activation by Pterostilbene Ameliorates Acute Doxorubicin Cardiotoxicity by Reducing Oxidative Stress via Enhancing AMPK and SIRT1 Cascades. Aging 2019, 11, 10061–10073. [Google Scholar] [CrossRef] [PubMed]
  88. Dutta, D.; Xu, J.; Dirain, M.L.; Leeuwenburgh, C. Calorie Restriction Combined with Resveratrol Induces Autophagy and Protects 26-Month-Old Rat Hearts from Doxorubicin-Induced Toxicity. Free Radic. Biol. Med. 2014, 74, 252–262. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  89. Yuan, Y.P.; Ma, Z.G.; Zhang, X.; Xu, S.C.; Zeng, X.F.; Yang, Z.; Deng, W.; Tang, Q.Z. CTRP3 Protected against Doxorubicin-Induced Cardiac Dysfunction, Inflammation and Cell Death via Activation of Sirt1. J. Mol. Cell. Cardiol. 2018, 114, 38–47. [Google Scholar] [CrossRef] [PubMed]
  90. Cui, L.; Guo, J.; Zhang, Q.; Yin, J.; Li, J.; Zhou, W.; Zhang, T.; Yuan, H.; Zhao, J.; Zhang, L.; et al. Erythropoietin Activates SIRT1 to Protect Human Cardiomyocytes against Doxorubicin-Induced Mitochondrial Dysfunction and Toxicity. Toxicol. Lett. 2017, 275, 28–38. [Google Scholar] [CrossRef]
  91. Albensi, B.C. What Is Nuclear Factor Kappa B (NF-κB) Doing in and to the Mitochondrion? Front. Cell. Dev. Biol. 2019, 7, 154. [Google Scholar] [CrossRef] [PubMed]
  92. Kim, D.S.; Woo, E.R.; Chae, S.W.; Ha, K.C.; Lee, G.H.; Hong, S.T.; Kwon, D.Y.; Kim, M.S.; Jung, Y.K.; Kim, H.M.; et al. Plantainoside D Protects Adriamycin-Induced Apoptosis in H9c2 Cardiac Muscle Cells via the Inhibition of ROS Generation and NF-kappaB Activation. Life Sci. 2007, 80, 314–323. [Google Scholar] [CrossRef] [PubMed]
  93. Wang, S.; Kotamraju, S.; Konorev, E.; Kalivendi, S.; Joseph, J.; Kalyanaraman, B. Activation of Nuclear Factor-kappaB during Doxorubicin-Induced Apoptosis in Endothelial Cells and Myocytes Is Pro-Apoptotic: The Role of Hydrogen Peroxide. Biochem. J. 2002, 367, 729–740. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  94. Li, D.; Li, J.; An, Y.; Yang, Y.; Zhang, S.Q. Doxorubicin-Induced Apoptosis in H9c2 Cardiomyocytes by NF-κB Dependent PUMA Upregulation. Eur. Rev. Med. Pharmacol. Sci. 2013, 17, 2323–2329. [Google Scholar] [PubMed]
  95. Wang, Z.Q.; Chen, M.T.; Zhang, R.; Zhang, Y.; Li, W.; Li, Y.G. Docosahexaenoic Acid Attenuates Doxorubicin-Induced Cytotoxicity and Inflammation by Suppressing NF-κB/iNOS/NO Signaling Pathway Activation in H9C2 Cardiac Cells. J. Cardiovasc. Pharmacol. 2016, 67, 283–289. [Google Scholar] [CrossRef] [PubMed]
  96. Guo, R.; Wu, K.; Chen, J.; Mo, L.; Hua, X.; Zheng, D.; Chen, P.; Chen, G.; Xu, W.; Feng, J. Exogenous Hydrogen Sulfide Protects against Doxorubicin-Induced Inflammation and Cytotoxicity by Inhibiting p38MAPK/NFκB Pathway in H9c2 Cardiac cells. Cell. Physiol. Biochem. 2013, 32, 1668–1680. [Google Scholar] [CrossRef]
  97. Zhao, Y.; Tian, X.; Liu, G.; Wang, K.; Xie, Y.; Qiu, Y. Berberine Protects Myocardial Cells against Anoxia-Reoxygenation Injury via p38 MAPK-Mediated NF-κB Signaling Pathways. Exp. Ther. Med. 2019, 17, 230–236. [Google Scholar] [CrossRef] [Green Version]
  98. Dhingra, R.; Guberman, M.; Rabinovich-Nikitin, I.; Gerstein, J.; Margulets, V.; Gang, H.; Madden, N.; Thliveris, J.; Kirshenbaum, L.A. Impaired NF-κB Signalling Underlies Cyclophilin D-Mediated Mitochondrial Permeability Transition Pore Opening in Doxorubicin Cardiomyopathy. Cardiovasc. Res. 2020, 116, 1161–1174. [Google Scholar] [CrossRef] [PubMed]
  99. Fang, X.; Wang, H.; Han, D.; Xie, E.; Yang, X.; Wei, J.; Gu, S.; Gao, F.; Zhu, N.; Yin, X.; et al. Ferroptosis as a Target for Protection against Cardiomyopathy. Proc. Natl Acad. Sci. USA 2019, 116, 2672–2680. [Google Scholar] [CrossRef] [Green Version]
  100. Kitakata, H.; Endo, J.; Matsushima, H.; Yamamoto, S.; Ikura, H.; Hirai, A.; Koh, S.; Ichihara, G.; Hiraide, T.; Moriyama, H.; et al. MITOL/MARCH5 Determines the Susceptibility of Cardiomyocytes to Doxorubicin-Induced Ferroptosis by Regulating GSH Homeostasis. J. Mol. Cell. Cardiol. 2021, 161, 116–129. [Google Scholar] [CrossRef] [PubMed]
  101. Tadokoro, T.; Ikeda, M.; Ide, T.; Deguchi, H.; Ikeda, S.; Okabe, K.; Ishikita, A.; Matsushima, S.; Koumura, T.; Yamada, K.I.; et al. Mitochondria-Dependent Ferroptosis Plays a Pivotal Role in Doxorubicin Cardiotoxicity. JCI Insight 2020, 5, e132747. [Google Scholar] [CrossRef]
  102. Christidi, E.; Brunham, L.R. Regulated Cell Death Pathways in Doxorubicin-Induced Cardiotoxicity. Cell. Death Dis. 2021, 12, 339. [Google Scholar] [CrossRef]
  103. Li, N.; Jiang, W.; Wang, W.; Xiong, R.; Wu, X.; Geng, Q. Ferroptosis and Its Emerging Roles in Cardiovascular Diseases. Pharmacol. Res. 2021, 166, 105466. [Google Scholar] [CrossRef] [PubMed]
  104. Koleini, N.; Nickel, B.E.; Edel, A.L.; Fandrich, R.R.; Ravandi, A.; Kardami, E. Oxidized Phospholipids in Doxorubicin-Induced Cardiotoxicity. Chem. Biol. Interact. 2019, 303, 35–39. [Google Scholar] [CrossRef]
  105. Mancardi, D.; Mezzanotte, M.; Arrigo, E.; Barinotti, A.; Roetto, A. Iron Overload, Oxidative Stress, and Ferroptosis in the Failing Heart and Liver. Antioxidants 2021, 10, 1864. [Google Scholar] [CrossRef] [PubMed]
  106. Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang, W.S.; et al. Ferroptosis: An Iron-Dependent form of Nonapoptotic Cell Death. Cell 2012, 149, 1060–1072. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Sun, X.; Ou, Z.; Chen, R.; Niu, X.; Chen, D.; Kang, R.; Tang, D. Activation of the p62-Keap1-NRF2 Pathway Protects against Ferroptosis in Hepatocellular Carcinoma Cells. Hepatology 2016, 63, 173–184. [Google Scholar] [CrossRef]
  108. Alim, I.; Caulfield, J.T.; Chen, Y.; Swarup, V.; Geschwind, D.H.; Ivanova, E.; Seravalli, J.; Ai, Y.; Sansing, L.H.; Ste Marie, E.J.; et al. Selenium Drives a Transcriptional Adaptive Program to Block Ferroptosis and Treat Stroke. Cell 2019, 177, 1262–1279. [Google Scholar] [CrossRef] [Green Version]
  109. Xu, M.; Tao, J.; Yang, Y.; Tan, S.; Liu, H.; Jiang, J.; Zheng, F.; Wu, B. Ferroptosis Involves in Intestinal Epithelial Cell Death in Ulcerative Colitis. Cell. Death Dis. 2020, 11, 86. [Google Scholar] [CrossRef]
  110. Matsushita, M.; Freigang, S.; Schneider, C.; Conrad, M.; Bornkamm, G.W.; Kopf, M. T Cell Lipid Peroxidation Induces Ferroptosis and Prevents Immunity to Infection. J. Exp. Med. 2015, 212, 555–568. [Google Scholar] [CrossRef] [Green Version]
  111. Anbharasi, V.; Cao, N.; Feng, S.S. Doxorubicin Conjugated to D-Alpha-Tocopheryl Polyethylene Glycol Succinate and Folic Acid as a Prodrug for Targeted Chemotherapy. J. Biomed. Mater. Res. A 2010, 94, 730–743. [Google Scholar] [CrossRef] [PubMed]
  112. Oliveira, P.J.; Santos, M.S.; Wallace, K.B. Doxorubicin-Induced Thiol-Dependent Alteration of Cardiac Mitochondrial Permeability Transition and Respiration. Biochemistry 2006, 71, 194–199. [Google Scholar] [CrossRef] [Green Version]
  113. Berthiaume, J.M.; Oliveira, P.J.; Fariss, M.W.; Wallace, K.B. Dietary Vitamin E Decreases Doxorubicin-Induced Oxidative Stress without Preventing Mitochondrial Dysfunction. Cardiovasc. Toxicol. 2005, 5, 257–267. [Google Scholar] [CrossRef] [Green Version]
  114. Swain, S.M.; Whaley, F.S.; Gerber, M.C.; Weisberg, S.; York, M.; Spicer, D.; Jones, S.E.; Wadler, S.; Desai, A.; Vogel, C.; et al. Cardioprotection with Dexrazoxane for Doxorubicin-Containing Therapy in Advanced Breast Cancer. J. Clin. Oncol. 1997, 15, 1318–1332. [Google Scholar] [CrossRef] [PubMed]
  115. Hasinoff, B.B.; Herman, E.H. Dexrazoxane: How It Works in Cardiac and Tumor Cells. Is it a Prodrug or Is It a Drug? Cardiovasc. Toxicol. 2007, 7, 140–144. [Google Scholar] [CrossRef]
  116. Swain, S.M.; Vici, P. The Current and Future Role of Dexrazoxane as a Cardioprotectant in Anthracycline Treatment: Expert Panel Review. J. Cancer Res. Clin. Oncol. 2004, 130, 1–7. [Google Scholar] [CrossRef] [Green Version]
  117. Kotamraju, S.; Chitambar, C.R.; Kalivendi, S.V.; Joseph, J.; Kalyanaraman, B. Transferrin Receptor-Dependent Iron Uptake Is Responsible for Doxorubicin-Mediated Apoptosis in Endothelial Cells: Role of Oxidant-Induced Iron Signaling in Apoptosis. J. Biol. Chem. 2002, 277, 17179–17187. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Trofenciuc, N.M.; Bordejevic, A.D.; Tomescu, M.C.; Petrescu, L.; Crisan, S.; Geavlete, O.; Mischie, A.; Onel, A.F.M.; Sasu, A.; Pop-Moldovan, A.L. Toll-like Receptor 4 (TLR4) Expression Is Correlated with T2* Iron Deposition in Response to Doxorubicin Treatment: Cardiotoxicity Risk Assessment. Sci. Rep. 2020, 10, 17013. [Google Scholar] [CrossRef]
  119. Maccarinelli, F.; Gammella, E.; Asperti, M.; Regoni, M.; Biasiotto, G.; Turco, E.; Altruda, F.; Lonardi, S.; Cornaghi, L.; Donetti, E.; et al. Mice Lacking Mitochondrial Ferritin Are More Sensitive to Doxorubicin-Mediated Cardiotoxicity. J. Mol. Med. 2014, 92, 859–869. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  120. Ichikawa, Y.; Ghanefar, M.; Bayeva, M.; Wu, R.; Khechaduri, A.; Naga Prasad, S.V.; Mutharasan, R.K.; Naik, T.J.; Ardehali, H. Cardiotoxicity of Doxorubicin Is Mediated through Mitochondrial Iron Accumulation. J. Clin. Investig. 2014, 124, 617–630. [Google Scholar] [CrossRef] [Green Version]
  121. Ichikawa, Y.; Bayeva, M.; Ghanefar, M.; Potini, V.; Sun, L.; Mutharasan, R.K.; Wu, R.; Khechaduri, A.; Jairaj Naik, T.; Ardehali, H. Disruption of ATP-Binding Cassette B8 in Mice Leads to Cardiomyopathy through a Decrease in Mitochondrial Iron Export. Proc. Natl. Acad. Sci. USA 2012, 109, 4152–4157. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Bock, F.J.; Tait, S.W.G. Mitochondria as Multifaceted Regulators of Cell Death. Nat. Rev. Mol. Cell. Biol. 2020, 21, 85–100. [Google Scholar] [CrossRef] [PubMed]
  123. Liu, J.; Kang, R.; Tang, D. Signaling Pathways and Defense Mechanisms of Ferroptosis. FEBS J. 2021. [Google Scholar] [CrossRef]
  124. He, H.; Wang, L.; Qiao, Y.; Yang, B.; Yin, D.; He, M. Epigallocatechin-3-Gallate Pretreatment Alleviates Doxorubicin-Induced Ferroptosis and Cardiotoxicity by Upregulating AMPKα2 and Activating Adaptive Autophagy. Redox. Biol. 2021, 48, 102185. [Google Scholar] [CrossRef] [PubMed]
  125. Callus, B.A.; Vaux, D.L. Caspase Inhibitors: Viral, Cellular and Chemical. Cell. Death Differ. 2007, 14, 73–78. [Google Scholar] [CrossRef] [PubMed]
  126. Ma, W.; Wei, S.; Zhang, B.; Li, W. Molecular Mechanisms of Cardiomyocyte Death in Drug-Induced Cardiotoxicity. Front. Cell. Dev. Biol. 2020, 8, 434. [Google Scholar] [CrossRef]
  127. Ingold, I.; Berndt, C.; Schmitt, S.; Doll, S.; Poschmann, G.; Buday, K.; Roveri, A.; Peng, X.; Porto Freitas, F.; Seibt, T.; et al. Selenium Utilization by GPX4 Is Required to Prevent Hydroperoxide-Induced Ferroptosis. Cell 2018, 172, 409–422. [Google Scholar] [CrossRef] [Green Version]
  128. Yang, W.S.; SriRamaratnam, R.; Welsch, M.E.; Shimada, K.; Skouta, R.; Viswanathan, V.S.; Cheah, J.H.; Clemons, P.A.; Shamji, A.F.; Clish, C.B.; et al. Regulation of Ferroptotic Cancer Cell Death by GPX4. Cell 2014, 156, 317–331. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  129. Mohamed, H.E.; El-Swefy, S.E.; Hagar, H.H. The Protective Effect of Glutathione Administration on Adriamycin-Induced Acute Cardiac Toxicity in Rats. Pharmacol. Res. 2000, 42, 115–121. [Google Scholar] [CrossRef] [PubMed]
  130. Todorova, V.K.; Kaufmann, Y.; Hennings, L.J.; Klimberg, V.S. Glutamine Regulation of Doxorubicin Accumulation in Hearts Versus Tumors in Experimental Rats. Cancer Chemother. Pharmacol. 2010, 66, 315–323. [Google Scholar] [CrossRef]
  131. Todorova, V.K.; Kaufmann, Y.; Hennings, L.; Klimberg, V.S. Oral Glutamine Protects against Acute Doxorubicin-Induced Cardiotoxicity of Tumor-Bearing Rats. J. Nutr. 2010, 140, 44–48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Tang, D.; Chen, X.; Kang, R.; Kroemer, G. Ferroptosis: Molecular Mechanisms and Health Implications. Cell. Res. 2021, 31, 107–125. [Google Scholar] [CrossRef] [PubMed]
  133. Chen, M.; Samuel, V.P.; Wu, Y.; Dang, M.; Lin, Y.; Sriramaneni, R.; Sah, S.K.; Chinnaboina, G.K.; Zhang, G. Nrf2/HO-1 Mediated Protective Activity of Genistein Against Doxorubicin-Induced Cardiac Toxicity. J. Environ. Pathol. Toxicol. Oncol. 2019, 38, 143–152. [Google Scholar] [CrossRef] [PubMed]
  134. Nordgren, K.K.; Wallace, K.B. Keap1 Redox-Dependent Regulation of Doxorubicin-Induced Oxidative Stress Response in Cardiac Myoblasts. Toxicol. Appl. Pharmacol. 2014, 274, 107–116. [Google Scholar] [CrossRef] [PubMed]
  135. Li, S.; Wang, W.; Niu, T.; Wang, H.; Li, B.; Shao, L.; Lai, Y.; Li, H.; Janicki, J.S.; Wang, X.L.; et al. Nrf2 Deficiency Exaggerates Doxorubicin-Induced Cardiotoxicity and Cardiac Dysfunction. Oxid. Med. Cell. Longev. 2014, 2014, 748524. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  136. Hou, K.; Shen, J.; Yan, J.; Zhai, C.; Zhang, J.; Pan, J.A.; Zhang, Y.; Jiang, Y.; Wang, Y.; Lin, R.Z.; et al. Loss of TRIM21 Alleviates Cardiotoxicity by Suppressing Ferroptosis Induced by the Chemotherapeutic Agent Doxorubicin. EBioMedicine 2021, 69, 103456. [Google Scholar] [CrossRef] [PubMed]
  137. Yang, H.B.; Lu, Z.Y.; Yuan, W.; Li, W.D.; Mao, S. Selenium Attenuates Doxorubicin-Induced Cardiotoxicity Through Nrf2-NLRP3 Pathway. Biol. Trace Elem. Res. 2021. [Google Scholar] [CrossRef] [PubMed]
  138. Refaie, M.M.M.; Shehata, S.; Ibrahim, R.A.; Bayoumi, A.M.A.; Abdel-Gaber, S.A. Dose-Dependent Cardioprotective Effect of Hemin in Doxorubicin-Induced Cardiotoxicity Via Nrf-2/HO-1 and TLR-5/NF-κB/TNF-α Signaling Pathways. Cardiovasc. Toxicol. 2021, 21, 1033–1044. [Google Scholar] [CrossRef] [PubMed]
  139. Hassannia, B.; Wiernicki, B.; Ingold, I.; Qu, F.; Van Herck, S.; Tyurina, Y.Y.; Bayır, H.; Abhari, B.A.; Angeli, J.P.F.; Choi, S.M.; et al. Nano-Targeted Induction of Dual Ferroptotic Mechanisms Eradicates High-Risk Neuroblastoma. J. Clin. Investig. 2018, 128, 3341–3355. [Google Scholar] [CrossRef]
  140. Doll, S.; Freitas, F.P.; Shah, R.; Aldrovandi, M.; da Silva, M.C.; Ingold, I.; Goya Grocin, A.; Xavier da Silva, T.N.; Panzilius, E.; Scheel, C.H.; et al. FSP1 Is a Glutathione-Independent Ferroptosis Suppressor. Nature 2019, 575, 693–698. [Google Scholar] [CrossRef] [PubMed]
  141. Bersuker, K.; Hendricks, J.M.; Li, Z.; Magtanong, L.; Ford, B.; Tang, P.H.; Roberts, M.A.; Tong, B.; Maimone, T.J.; Zoncu, R.; et al. The CoQ Oxidoreductase FSP1 Acts Parallel to GPX4 to Inhibit Ferroptosis. Nature 2019, 575, 688–692. [Google Scholar] [CrossRef] [PubMed]
  142. Botelho, A.F.M.; Lempek, M.R.; Branco, S.; Nogueira, M.M.; de Almeida, M.E.; Costa, A.G.; Freitas, T.G.; Rocha, M.; Moreira, M.V.L.; Barreto, T.O.; et al. Coenzyme Q10 Cardioprotective Effects Against Doxorubicin-Induced Cardiotoxicity in Wistar Rat. Cardiovasc. Toxicol. 2020, 20, 222–234. [Google Scholar] [CrossRef] [PubMed]
  143. Rahmanifard, M.; Vessal, M.; Noorafshan, A.; Karbalay-Doust, S.; Naseh, M. The Protective Effects of Coenzyme Q10 and Lisinopril Against Doxorubicin-Induced Cardiotoxicity in Rats: A Stereological and Electrocardiogram Study. Cardiovasc. Toxicol. 2021, 21, 936–946. [Google Scholar] [CrossRef] [PubMed]
  144. Green, P.S.; Leeuwenburgh, C. Mitochondrial Dysfunction Is an Early Indicator of Doxorubicin-Induced Apoptosis. Biochim. Biophys. Acta 2002, 1588, 94–101. [Google Scholar] [CrossRef] [Green Version]
  145. Childs, A.C.; Phaneuf, S.L.; Dirks, A.J.; Phillips, T.; Leeuwenburgh, C. Doxorubicin Treatment In Vivo Causes Cytochrome C Release and Cardiomyocyte Apoptosis, as Well as Increased Mitochondrial Efficiency, Superoxide Dismutase Activity, and Bcl-2:Bax Ratio. Cancer Res. 2002, 62, 4592–4598. [Google Scholar]
  146. Tokarska-Schlattner, M.; Wallimann, T.; Schlattner, U. Multiple Interference of Anthracyclines with Mitochondrial Creatine Kinases: Preferential Damage of the Cardiac Isoenzyme and Its Implications for Drug Cardiotoxicity. Mol. Pharmacol. 2002, 61, 516–523. [Google Scholar] [CrossRef] [Green Version]
  147. Forrest, G.L.; Gonzalez, B.; Tseng, W.; Li, X.; Mann, J. Human Carbonyl Reductase Overexpression in the Heart Advances the Development of Doxorubicin-Induced Cardiotoxicity in Transgenic Mice. Cancer Res. 2000, 60, 5158–5164. [Google Scholar]
  148. Clementi, M.E.; Giardina, B.; Di Stasio, E.; Mordente, A.; Misiti, F. Doxorubicin-Derived Metabolites induce Release of Cytochrome C and Inhibition of Respiration on Cardiac Isolated Mitochondria. Anticancer Res. 2003, 23, 2445–2450. [Google Scholar]
  149. Kluza, J.; Marchetti, P.; Gallego, M.A.; Lancel, S.; Fournier, C.; Loyens, A.; Beauvillain, J.C.; Bailly, C. Mitochondrial Proliferation during Apoptosis Induced by Anticancer Agents: Effects of Doxorubicin and Mitoxantrone on Cancer and Cardiac Cells. Oncogene 2004, 23, 7018–7030. [Google Scholar] [CrossRef] [Green Version]
  150. Kavazis, A.N.; Smuder, A.J.; Min, K.; Tümer, N.; Powers, S.K. Short-Term Exercise Training Protects against Doxorubicin-Induced Cardiac Mitochondrial Damage Independent of HSP72. Am. J. Physiol. Heart Circ. Physiol. 2010, 299, H1515–H1524. [Google Scholar] [CrossRef] [Green Version]
  151. Bugger, H.; Guzman, C.; Zechner, C.; Palmeri, M.; Russell, K.S.; Russell, R.R., III. Uncoupling Protein Downregulation in Doxorubicin-Induced Heart Failure Improves Mitochondrial Coupling but Increases Reactive Oxygen Species Generation. Cancer Chemother. Pharmacol. 2011, 67, 1381–1388. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Fajardo, G.; Zhao, M.; Berry, G.; Wong, L.J.; Mochly-Rosen, D.; Bernstein, D. β2-Adrenergic Receptors Mediate Cardioprotection through Crosstalk with Mitochondrial Cell Death Pathways. J. Mol. Cell. Cardiol. 2011, 51, 781–789. [Google Scholar] [CrossRef] [Green Version]
  153. Pereira, G.C.; Pereira, S.P.; Tavares, L.C.; Carvalho, F.S.; Magalhães-Novais, S.; Barbosa, I.A.; Santos, M.S.; Bjork, J.; Moreno, A.J.; Wallace, K.B.; et al. Cardiac Cytochrome c and Cardiolipin Depletion during Anthracycline-Induced Chronic Depression of Mitochondrial Function. Mitochondrion 2016, 30, 95–104. [Google Scholar] [CrossRef] [PubMed]
  154. Ahmed, L.A.; El-Maraghy, S.A. Nicorandil Ameliorates Mitochondrial Dysfunction in Doxorubicin-Induced Heart Failure in Rats: Possible Mechanism of Cardioprotection. Biochem. Pharmacol. 2013, 86, 1301–1310. [Google Scholar] [CrossRef] [PubMed]
  155. Narumi, T.; Shishido, T.; Otaki, Y.; Kadowaki, S.; Honda, Y.; Funayama, A.; Honda, S.; Hasegawa, H.; Kinoshita, D.; Yokoyama, M.; et al. High-Mobility Group Box 1-Mediated Heat Shock Protein Beta 1 Expression Attenuates Mitochondrial Dysfunction and Apoptosis. J. Mol. Cell. Cardiol. 2015, 82, 1–12. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  156. Moreira, A.C.; Branco, A.F.; Sampaio, S.F.; Cunha-Oliveira, T.; Martins, T.R.; Holy, J.; Oliveira, P.J.; Sardão, V.A. Mitochondrial Apoptosis-Inducing Factor Is Involved in Doxorubicin-Induced Toxicity on H9c2 Cardiomyoblasts. Biochim. Biophys. Acta 2014, 1842, 2468–2478. [Google Scholar] [CrossRef] [Green Version]
  157. Liang, X.; Wang, S.; Wang, L.; Ceylan, A.F.; Ren, J.; Zhang, Y. Mitophagy Inhibitor Liensinine Suppresses Doxorubicin-Induced Cardiotoxicity through Inhibition of Drp1-Mediated Maladaptive Mitochondrial Fission. Pharmacol. Res. 2020, 157, 104846. [Google Scholar] [CrossRef]
  158. Li, B.; Kim, D.S.; Yadav, R.K.; Kim, H.R.; Chae, H.J. Sulforaphane Prevents Doxorubicin-Induced Oxidative Stress and Cell Death in Rat H9c2 Cells. Int. J. Mol. Med. 2015, 36, 53–64. [Google Scholar] [CrossRef] [Green Version]
  159. Kotamraju, S.; Konorev, E.A.; Joseph, J.; Kalyanaraman, B. Doxorubicin-Induced Apoptosis in Endothelial Cells and Cardiomyocytes Is Ameliorated by Nitrone Spin Traps and Ebselen. Role of Reactive Oxygen and Nitrogen Species. J. Biol. Chem. 2000, 275, 33585–33592. [Google Scholar] [CrossRef] [Green Version]
  160. Xia, P.; Liu, Y.; Chen, J.; Coates, S.; Liu, D.X.; Cheng, Z. Inhibition of Cyclin-Dependent Kinase 2 Protects against Doxorubicin-Induced Cardiomyocyte Apoptosis and Cardiomyopathy. J. Biol. Chem. 2018, 293, 19672–19685. [Google Scholar] [CrossRef] [Green Version]
  161. Xia, P.; Chen, J.; Liu, Y.; Fletcher, M.; Jensen, B.C.; Cheng, Z. Doxorubicin Induces Cardiomyocyte Apoptosis and Atrophy through Cyclin-Dependent Kinase 2-Mediated Activation of Forkhead Box O1. J. Biol. Chem. 2020, 295, 4265–4276. [Google Scholar] [CrossRef]
  162. Dhingra, R.; Margulets, V.; Chowdhury, S.R.; Thliveris, J.; Jassal, D.; Fernyhough, P.; Dorn, G.W., II; Kirshenbaum, L.A. Bnip3 Mediates Doxorubicin-Induced Cardiac Myocyte Necrosis and Mortality through Changes in Mitochondrial Signaling. Proc. Natl. Acad. Sci. USA 2014, 111, E5537–E5544. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Catanzaro, M.P.; Weiner, A.; Kaminaris, A.; Li, C.; Cai, F.; Zhao, F.; Kobayashi, S.; Kobayashi, T.; Huang, Y.; Sesaki, H.; et al. Doxorubicin-Induced Cardiomyocyte Death Is Mediated by Unchecked Mitochondrial Fission and Mitophagy. FASEB J. 2019, 33, 11096–11108. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  164. Gharanei, M.; Hussain, A.; Janneh, O.; Maddock, H. Attenuation of Doxorubicin-Induced Cardiotoxicity by Mdivi-1: A Mitochondrial Division/Mitophagy Inhibitor. PLoS ONE 2013, 8, e77713. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. McLaughlin, D.; Zhao, Y.; O’Neill, K.M.; Edgar, K.S.; Dunne, P.D.; Kearney, A.M.; Grieve, D.J.; McDermott, B.J. Signalling Mechanisms Underlying Doxorubicin and Nox2 NADPH Oxidase-Induced Cardiomyopathy: Involvement of Mitofusin-2. Br. J. Pharmacol. 2017, 174, 3677–3695. [Google Scholar] [CrossRef] [Green Version]
  166. Aung, L.H.H.; Li, R.; Prabhakar, B.S.; Li, P. Knockdown of Mtfp1 Can Minimize Doxorubicin Cardiotoxicity by Inhibiting Dnm1l-Mediated Mitochondrial Fission. J. Cell. Mol. Med. 2017, 21, 3394–3404. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Schematic representation of Doxorubicin (DOX)-induced apoptosis pathway. DOX, doxorubicin; ROS, reactive oxygen species; MAPK, mitogen-activated protein kinase; JNK, Jun amino terminal kinase; ERK, extracellular signal-regulated kinase; NAC, N-acetylcysteine; TopII, opoisomerase II; PGC-1α, PPARG coactivator 1α; GPX1, glutathione peroxidase 1; HDAC, histone deacetylase; GATA4, GATA binding protein 4.
Figure 1. Schematic representation of Doxorubicin (DOX)-induced apoptosis pathway. DOX, doxorubicin; ROS, reactive oxygen species; MAPK, mitogen-activated protein kinase; JNK, Jun amino terminal kinase; ERK, extracellular signal-regulated kinase; NAC, N-acetylcysteine; TopII, opoisomerase II; PGC-1α, PPARG coactivator 1α; GPX1, glutathione peroxidase 1; HDAC, histone deacetylase; GATA4, GATA binding protein 4.
Ijms 23 01414 g001
Figure 2. Schematic representation of Doxorubicin (DOX)-induced ferroptosis pathway. DOX, doxorubicin; Tf, transferrin; GSH, glutathione; GSSG, oxidized glutathione; GPX4, glutathione and glutathione peroxidase 4; FSP1, ferroptosis suppressor protein 1; CHAC1, ChaC Glutathione Specific Gamma-Glutamylcyclotransferase 1; Nrf2, Nuclear factor erythroid 2-related factor 2; KEAP1, Kelch-like ECH-associated protein 1; ABCB8, ATP-binding cassette transporter 8; CoQ10, coenzyme Q10.
Figure 2. Schematic representation of Doxorubicin (DOX)-induced ferroptosis pathway. DOX, doxorubicin; Tf, transferrin; GSH, glutathione; GSSG, oxidized glutathione; GPX4, glutathione and glutathione peroxidase 4; FSP1, ferroptosis suppressor protein 1; CHAC1, ChaC Glutathione Specific Gamma-Glutamylcyclotransferase 1; Nrf2, Nuclear factor erythroid 2-related factor 2; KEAP1, Kelch-like ECH-associated protein 1; ABCB8, ATP-binding cassette transporter 8; CoQ10, coenzyme Q10.
Ijms 23 01414 g002
Table 1. Characteristics of type I and II chemotherapy-induced cardiotoxicity.
Table 1. Characteristics of type I and II chemotherapy-induced cardiotoxicity.
Type I (Myocardial Damage)Type II (Myocardial Dysfunction)
Representative agentsAnthracyclines (DOX)Trastuzumab
Bevacizumab
Sunitinib
Sorafenib
Dose-dependenceYesNo
MechanismFree radical formationInhibition of Erb signaling
DNA damage
Oxidative damage
Clinical manifestationUnderlying damage appears to be permanent and irreversibleHigh likelihood of recovery (to or near baseline cardiac status) in 2–4 months (reversible)
Biopsy presentationMyofibril disarrayMinimal changes have been reported
Vacuole formation
Effect of rechallengeHigh probability of recurrent dysfunction that is progressive, leading to intractable heart failure and deathIncreasing evidence for the relative safety of rechallenge; additional data needed
Modified from reference [2,9].
Table 2. Differences between features of apoptosis and ferroptosis.
Table 2. Differences between features of apoptosis and ferroptosis.
Apoptosis (Intrinsic Apoptosis)Ferroptosis
Biochemical characteristicsInvolvement of Bcl-2 family proteins
Release of cytochrome c from mitochondria
Activation of caspases
Peroxidation of cell membrane phospholipids catalyzed by iron ions (Fe2+)
Accumulation of lipid hydroxyl radicals
Key moleculesCaspase-3, Bcl-2, BAX, p53, NF-κBGPX4, GSH, Xc−, CHAC1, CoQ10, Nrf2
InhibitorsZVAD-FMK, Emricasan, Q-VD-Oph, IDN-6556, DEVD-CHOFer-1, Vitamin E, Liproxstatin-1, CoQ10
Morphological featuresChromatin condensation and fragmentation
DNA laddering
Cell shrinkage and bleb formation at the plasma membrane
Exposure of phosphatidylserine to the outer membrane
Decrease in mitochondrial cristae
Mitochondrial aggregation
Modified from reference [125,126,127].
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Kitakata, H.; Endo, J.; Ikura, H.; Moriyama, H.; Shirakawa, K.; Katsumata, Y.; Sano, M. Therapeutic Targets for DOX-Induced Cardiomyopathy: Role of Apoptosis vs. Ferroptosis. Int. J. Mol. Sci. 2022, 23, 1414. https://doi.org/10.3390/ijms23031414

AMA Style

Kitakata H, Endo J, Ikura H, Moriyama H, Shirakawa K, Katsumata Y, Sano M. Therapeutic Targets for DOX-Induced Cardiomyopathy: Role of Apoptosis vs. Ferroptosis. International Journal of Molecular Sciences. 2022; 23(3):1414. https://doi.org/10.3390/ijms23031414

Chicago/Turabian Style

Kitakata, Hiroki, Jin Endo, Hidehiko Ikura, Hidenori Moriyama, Kohsuke Shirakawa, Yoshinori Katsumata, and Motoaki Sano. 2022. "Therapeutic Targets for DOX-Induced Cardiomyopathy: Role of Apoptosis vs. Ferroptosis" International Journal of Molecular Sciences 23, no. 3: 1414. https://doi.org/10.3390/ijms23031414

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop