Download as pdf or txt
Download as pdf or txt
You are on page 1of 12

Molecular and Cellular Biochemistry

https://doi.org/10.1007/s11010-023-04783-3

Novel molecular mechanisms of doxorubicin cardiotoxicity: latest


leading‑edge advances and clinical implications
Y. Robert Li1 · Kassim Traore2 · Hong Zhu3

Received: 11 April 2023 / Accepted: 4 June 2023


© The Author(s), under exclusive licence to Springer Science+Business Media, LLC, part of Springer Nature 2023

Abstract
Doxorubicin (Dox) is among the most widely used cancer chemotherapeutic drugs. The clinical use of Dox is, however,
limited due to its cardiotoxicity. Studies over the past several decades have suggested various mechanisms of Dox-induced
cardiotoxicity (DIC). Among them are oxidative stress, topoisomerase inhibition, and mitochondrial damage. Several novel
molecular targets and signaling pathways underlying DIC have emerged over the past few years. The most notable advances
include discovery of ferroptosis as a major form of cell death in Dox cytotoxicity, and elucidation of the involvement of
cardiogenetics and regulatory RNAs as well as multiple other targets in DIC. In this review, we discuss these advances,
focusing on latest cutting-edge research discoveries from mechanistic studies reported in influential journals rather than
surveying all research studies available in the literature.

Keywords  Cardiogenetics · Cardiomyopathy · Cardiotoxicity · Doxorubicin · Ferroptosis · Heme · Hemopexin ·


Macrophage · Mitochondria · Nrf1 · Regulatory RNA

Abbreviations RCD Regulated cell death


circRNA Circular RNA SLC Solute carrier
DIC Doxorubicin-induced cardiotoxicity SNP Single nucleotide polymorphisms
Dox Doxorubicin TOP2B Topoisomerase 2β
ERK Extracellular regulated kinase
GPx4 Glutathione peroxidase-4
hiPSC-CMs Human induced pluripotent stem cell- Introduction
derived cardiomyocytes
HO-1 Heme oxygenase-1 Doxorubicin is a member of the anthracycline anticancer
lncRNA Long non-coding RNA drug class; other members of this drug class include dauno-
miRNA MicroRNA rubicin and epirubicin. Dox was initially isolated in 1967 in
Nrf1 Nuclear factor E2-related factor 1 the Farmitalia Research Laboratories in Italy from cultures
Nrf2 Nuclear factor E2-related factor 2 of a mutant Streptomyces peucetius (Streptomyces peucetius
OCT3 Cation transporter 3 caesius) [1]. The drug was first approved in 1974 by the
RARG​ Retinoic acid receptor gamma U.S. Food and Drug Administration (FDA) for clinical use.
Presently, Dox remains one of the most widely used cancer
chemotherapeutic agents. It is effective for a range of human
* Y. Robert Li cancers, including leukemias, Wilms tumor, neuroblastoma,
yli@campbell.edu
soft tissue and bone sarcomas, breast carcinoma, ovarian
1
Department of Pharmacology, Campbell University Jerry carcinoma, transitional cell bladder carcinoma, thyroid car-
Wallace School of Osteopathic Medicine, Buies Creek, cinoma, gastric carcinoma, Hodgkin’s disease, malignant
NC 27560, USA lymphoma, and lung cancer [2]. Despite its well-established
2
Department of Biochemistry, Duquesne University College anticancer efficacy, the clinical use of Dox is limited due to
of Osteopathic Medicine, Pittsburgh, PA 15282, USA its cardiotoxicity. In many cases, Dox-induced cardiotox-
3
Department of Physiology and Pathophysiology, Campbell icity (DIC) has been linked to the development of dilated
University Jerry Wallace School of Osteopathic Medicine, cardiomyopathy and heart failure. In fact, Dox-induced
Buies Creek, NC 27560, USA

13
Vol.:(0123456789)
Molecular and Cellular Biochemistry

cardiomyopathy is among the most common etiologies of Molecular pathways underlying


cancer chemotherapy-associated heart failure [3, 4]. doxorubicin‑induced myocardial ferroptosis
Although Dox has been used in the treatment of human
cancers for over five decades, the molecular mechanisms HO‑1 upregulation
underlying its cardiotoxicity remain partially understood.
Multiple theories have been proposed to explain DIC, which A study reported in 2019 by Fang et  al. [9], for the
include redox cycling in mitochondria, topoisomerase inhi- first time, demonstrated that ferroptosis mediates Dox-
bition, and oxidative stress [2, 5]. Studies over the past few induced cardiomyopathy in mice. Using canonical apop-
years have identified several novel molecular targets and tosis and/or necroptosis-defective ­Ripk3−/−, ­Mlkl−/−, or
signaling pathways underlying DIC. This mini-review sur- ­Fadd−/−Mlkl−/−mice, the authors found that Dox-induced
veys latest cutting-edge mechanistic findings on DIC and myocardial ferroptosis drives cardiomyopathy and mortal-
discusses emerging concepts that may lead to the develop- ity following Dox exposure. They further showed that inhi-
ment of more effective strategies for cardioprotection in bition of ferroptosis by ferrostatin-1 markedly ameliorates
Dox-treated cancer patients. To this end, the article focuses Dox cardiomyopathy. RNA-sequencing analysis revealed
on latest advances in areas related to ferroptosis, cardioge- upregulation of heme oxygenase-1 (HO-1) by Dox as a
netics, regulatory RNAs, and other emerging targets under- major mechanism of ferroptotic cardiomyopathy. HO-1
lying DIC. degrades heme and releases free iron in cardiomyocytes,
which in turn leads to the generation of oxidized lipids
in the mitochondrial membrane. Notably, iron chelation
therapy with dexrazoxane (the only FDA-approved drug
Ferroptosis in doxorubicin cardiotoxicity for protecting against DIC) alleviates Dox cardiomyopathy
in mice. Moreover, mitochondrially targeted antioxidant
Overview of regulated cell death MitoTEMPO rescues Dox cardiomyopathy, supporting
oxidative damage of mitochondria as a major mechanism
It is well known that Dox causes apoptosis in cardiomyo- underlying Dox-induced ferroptotic cardiac injury [9].
cytes, and cardiac apoptosis is the most commonly described Another important finding of the study is that upregula-
form of cell death induced by Dox in both in  vitro and tion of HO-1 by Dox occurs via an Nrf2 (nuclear factor
in vivo models [6]. Apoptosis is the prototypical form of E2-related factor 2)-dependent mechanism. Consequently,
regulated cell death (RCD); the newly discovered other Dox-induced accumulation of free iron (non-heme iron) in
forms of RCD include necroptosis, pyroptosis, ferroptosis, cardiomyocytes is abolished in Nrf2-null mice [9].
entotic cell death, netotic cell death, parthanatos, lysosome- The study by Fang et al., however, did not report the
dependent cell death, autophagy-dependent cell death, alka- effect of Nrf2 knockout on Dox-induced cardiomyopa-
liptosis, and oxeiptosis [7], as well as cuproptosis, a most thy. Nrf2 is the central regulator of a wide range of genes
recently identified form of RCD underlying copper-induced whose products are involved in diverse cellular processes,
cytotoxicity [8]. including antioxidative cytoprotection, antiinflammation,
and drug metabolism, as well as regulation of many other
metabolic pathways [15]. As such, the overall impact of
Ferroptosis as a major form of cell death Nrf2 signaling on Dox cardiomyopathy would be dictated
in doxorubicin cardiotoxicity by the balanced effects of both detrimental factors (in this
case, upregulation of HO-1 and the consequent accumula-
In addition to inducing apoptosis, Dox also causes ferrop- tion of free iron) and protective factors, such as antioxidant
tosis, which appears to be a major form of cell death under- enzymes that have been shown to be protective against
lying Dox-induced cardiomyopathy in mice [9]. Ferropto- DIC. In this context, a protective role for Nrf2 signaling
sis differs from other forms of cell death; it results from and HO-1 expression in doxorubicin-induced cardiotoxic-
iron-dependent lipid peroxide accumulation triggered by ity has been reported previously in other models of DIC
insufficiency of glutathione peroxidase-4 (GPx4) [10–12]. [16, 17]. The exact reasons for the discrepancy between
Dysregulated ferroptosis has been implicated in various dis- the findings by Fang et al. and those by others remain
ease processes, including oxidative tissue injury and tumo- unknown. Nevertheless, the study by Fang et al. conclu-
rigenesis [13]. The notion of ferroptosis as a major form of sively demonstrates a causal role of iron-dependent ferrop-
Dox-induced cell death is in line with the well-established tosis in Dox cardiomyopathy and the involvement of mito-
causal role of iron in DIC [14]. The sections below discuss chondrial oxidative stress in initiating the cardiac injury.
latest novel findings on the molecular events underlying
Dox-induced cardiac ferroptosis.

13
Molecular and Cellular Biochemistry

GPx4 downregulation

In line with findings by Fang et al. [9], a subsequent study


by Tadokoro et al. [18] demonstrated that mitochondria-
dependent ferroptosis plays a pivotal role in Dox cardiomyo-
pathy in mice. They showed that Dox deceases expression
of GPx4 (a cellular suppressor of ferroptosis) and induces
excessive lipid peroxidation through accumulation of Dox-
Fe2+ complex in mitochondria, leading to mitochondria-
dependent ferroptosis. They also demonstrated that mito-
chondria-dependent ferroptosis is a major cause of DOX
cardiomyopathy. Notably, compared with wild-type mice,
Dox-induced cardiomyopathy and left ventricular dysfunc-
tion are ameliorated in GPx4-overexpressing mice and exac-
erbated in GPx4-heteroknockout ­(GPx4±) mice, indicating
a causal involvement of GPx4 downregulation in Dox car-
diomyopathy [18]. Moreover, the authors found that chela-
tion of intramitochondrial ­Fe2+ by Mito-FerroGreen, but not Fig. 1  Novel mechanisms by which doxorubicin induces iron over-
load, lipid peroxidation, and ferroptosis. As illustrated, both Nrf2/
­ e3+ by deferoxamine or dexrazoxane, prevents
chelation of F HO-1 and Alas1/heme synthesis pathways as well as GPx4 downreg-
Dox-induced lipid peroxidation and ferroptosis in cultured ulation may be involved in doxorubicin-induced iron overload, lipid
cardiomyocytes, suggesting that pharmacological chelation peroxidative, and mitochondrial ferroptosis, leading to cardiomyopa-
of mitochondrial F ­ e2+ may be a more effective approach thy. See text for detailed description. (+) denotes increase or activa-
tion; (−) denotes decrease or inhibition. This diagram is based pri-
to preventing DIC than F ­ e3+ chelation (such as by dexra- marily on novel findings reported by Fang et al. [9], Tadokoro et al.
zoxane) [18]. Indeed, the clinical efficacy of dexrazoxane [18], and Abe et al. [21]
in preventing DIC in cancer patients has been shown to be
rather limited [19, 20].
of free iron and consequent iron overload. On the other hand,
Suppression of heme biosynthesis Dox downregulates GPx4, an enzyme responsible for elimi-
nating lipid hydroperoxides and acting as an endogenous
More recently, Abe et al. further investigated the molecu- suppressor of ferroptosis [22]. Iron accumulation in mito-
lar events leading to mitochondria-dependent ferroptosis chondria promotes oxidative stress, lipid peroxidation, and
underlying DIC [21]. Using cultured cardiomyocytes, they ferroptosis, leading to cardiomyopathy and left ventricular
showed that Dox accumulates in mitochondria by intercalat- dysfunction. Hence, selective chelation of mitochondrial
ing into mitochondrial DNA (mtDNA), inducing ferroptosis iron may be a promising approach to suppressing ferropto-
in an mtDNA content-dependent manner. Moreover, Dox sis and thereby protecting against cardiomyopathy and heart
suppresses the synthesis of heme by decreasing the level failure in Dox-treated cancer patients. It, however, should
of 5′-aminolevulinate synthase 1 (Alas1), the rate-limiting be noted that cardioprotective modalities must not hinder
enzyme in heme biosynthesis, thereby impairing iron utili- the anticancer activity of Dox. In this context, iron chela-
zation, resulting in iron overload and mitochondria-depend- tion, including mitochondrial iron chelation has been shown
ent ferroptosis. As a result, either Alas1 overexpression or to suppress tumor growth and metastasis in animal models
supplementation with 5-aminolevulinic acid (5-ALA), the [23]. It is, thus, imperative to determine the effect of iron
product of Alas1, prevents Dox-induced iron overload and chelation on Dox’s anticancer activity.
ferroptosis in cardiomyocytes. Notably, administration of
5-ALA to mice also suppresses Dox-induced iron overload
and ferroptotic cardiomyopathy [21]. Hence, the study iden- Cardiogenetics of doxorubicin cardiotoxicity
tifies Alas1 as a novel molecular target of Dox and suggests
a therapeutic potential of 5-ALA in DIC. Overview of cardiogenetics
In summary, as illustrated in Fig. 1, induction of myo-
cardial iron-dependent ferroptosis is a major cellular event Cardiogenetics is the study of the genetic causes of heart
underlying DIC. Dox may cause mitochondrial iron accumu- diseases and how genetic variations affect cardiac patholo-
lation via two mechanisms: (1) upregulation of HO-1, lead- gies and their response to drug therapy. As an important
ing to degradation of heme to release free iron; and (2) inhi- aspect of cardiogenetics, studying the effects of genetic
bition of heme synthesis, resulting in decreased utilization variations on patients’ response to cardiovascular drugs has

13
Molecular and Cellular Biochemistry

Fig. 2  Role of SNPs in
doxorubicin cardiotoxicity. As
illustrated, multiple SNPs can
downregulate SLC28A3, lead-
ing to reduced transport of Dox
into cardiomyocytes and thereby
decreased Dox cardiotoxicity
(upper panel). On the other
hand, certain SNPs may affect
various cellular signaling path-
ways to augment Dox cardio-
toxicity (lower panel). See text
for detailed discussion of the
original findings on which this
diagram is primarily based

critical implications in precision medicine. Indeed, personal- transporters and retinoic acid receptor gamma (RARG), as
ized therapies based on pharmacogenetic data have greatly well as non-coding SNPs underlying DIC (Fig. 2).
improved the clinical management of various cardiovascular
diseases, especially atherosclerotic coronary artery disease Drug transporters
[24–26]. On the other hand, studies of the effects of genetic
variations on drug-induced cardiac injury, including Dox Multiple pharmacogenomic studies have identified the
cardiomyopathy have also advanced our understanding of synonymous genomic (exonic) variant rs7853758 (G > A,
the mechanisms of drug-induced cardiotoxicity and may L461L) and the intronic variant rs885004 in SLC28A3
contribute to the development of strategies to identify sus- (solute carrier family 28 member 3) to be associated with a
ceptible patients so as to minimize the risk of cardiotoxicity. lower incidence of DIC [30–32]. However, the true causal
The following sections discuss latest mechanistic studies on variant(s), the cardioprotective mechanism of this locus, the
major genetic variations affecting DIC. role of SLC28A3 in DIC, and the suitability of SLC28A3
as a target for cardioprotective drugs had remained unde-
termined until a very recent study by Magdy et al. [33].
Genetic variations in doxorubicin cardiotoxicity: SLC28A3, also known as concentrative nucleoside trans-
recent mechanistic studies porter 3 (CNT3), plays an important role in mediating the
cellular entry of a broad array of physiological nucleosides
While there have been many reports on the association of and synthetic anticancer nucleoside analog drugs [34].
diverse single nucleotide polymorphisms (SNPs) with Dox The association of the above genetic variations of
cardiotoxicity [27–29], conclusive, mechanistic studies to SLC28A3 with DIC suggests that this membrane trans-
establish the causality are relatively few. Described below porter may also mediate Dox uptake into cardiac cells.
are notable mechanistic studies on genetic variations in drug To define the role of SCL28A3 genetic variants in
DIC, Magdy et  al. [33] recruited 6 well-phenotyped,

13
Molecular and Cellular Biochemistry

Dox-treated pediatric patients from the original associa- associated with cardiovascular risk [36, 37], it is important
tion study cohort, and generated human induced pluri- to determine if such genetic variations also affect patients’
potent stem cell-derived cardiomyocytes (hiPSC-CMs) susceptibility to DIC.
from them. With this approach, the authors showed that Although both SLC28A3 and SLC22A3 act as Dox
the patient-specific hiPSC-CMs recapitulate the cardio- uptake transporters in the heart, the relative importance of
protective effect of the SNP rs7853758. SLC28A3 gene these transporters in modulating DIC in a particular patient
knockdown and overexpression using CRISPR/Cas9 remains unclear. In addition, other transporter such as the
reduces and increases Dox uptake into the cardiomyocytes, organic cation transporters SLC22A4 and SLC22A17 are
respectively, altering their sensitivity to Dox cytotoxic- also implicated in Dox uptake into cardiomyocytes [33].
ity. These observations confirm the role of this locus in Hence, it is likely that the eventual impact would be deter-
DIC. Notably, upon fine mapping of the SLC28A3 locus, mined by the relative cardiac abundance and activity of these
the authors discovered that a linked SNP rs11140490, transporters in a particular patient. Indeed, genetic variations
rather than the originally identified rs7853758, is actu- in genes encoding membrane transporters are common in
ally the causal cardioprotective variant within that locus. human populations [38, 39].
Mechanistically, rs11140490 exerts its action by upregu-
lating a SLC28A3-overlapping antisense long non-cod- RARG​
ing RNA (lncRNA) SLC28A3-AS1. Overexpression of
SLC28A3-AS1 downregulates SCL28A3, reducing Dox In addition to modulating Dox transport, genetic variations
uptake into the cardiomyocytes. Moreover, via screening a may also affect the intracellular machinery underlying Dox
drug library using hiPSC-CMs followed by in vivo valida- cytotoxicity. For example, a missense variant rs2229774
tion in a mouse model of DIC, the authors demonstrated (S427L) in the RARG gene is associated with increased
that the SLC competitive inhibitor, desipramine protects susceptibility to DIC in pediatric cancer patients [40].
against DIC without compromising Dox anticancer effi- Recently, Burridge and associates showed that hiPSC-CMs
cacy. Hence, these novel findings establish SLC28A3 as from patients who had rs2229774 and suffered DIC are more
a transporter for Dox uptake into cardiomyocyte and pro- sensitive to Dox cytotoxicity [41]. Mechanistically, this
vide a basis for genetic testing of rs11140490 to identify RARG variant effect is mediated via increased expression of
patients who are protected from DIC. The findings also topoisomerase 2β (TOP2B; a critical mediator of DIC [42])
point to two potential therapeutic options: either using and decreased activation of the cardioprotective extracellu-
the lncRNA SLC28A3-AS1 or developing a desipramine lar regulated kinase (ERK) pathway. The authors also used
analog to protect against DIC [33]. The work by Magdy patient-specific hiPSC-CMs as a drug discovery platform
et al. represents an excellent example of a mechanistic and demonstrated that the RARG agonist CD1530 attenu-
cardiogenetic study to establish a causal role of a genetic ates DIC in mice. The findings hence provide a rationale for
variant in DIC. The study also demonstrates the power of clinical prechemotherapy genetic screening for rs2229774
patient-specific hiPSC-CMs in mechanistic investigation and a foundation for the clinical use of RARG agonist
of cardiogenetics of DIC as well as cardioprotective drug treatment to protect cancer patients from DIC [41]. More
discovery (see below). recently, using CRISPR-Cas9-genome-edited hiPSC-CMs,
Indeed, in a recent study, using hiPSC-CMs from patients Huang et al. reported that the RARG variant rs2229774 has
receiving Dox, Huang et al. probed the transcriptomic land- impaired ability to activate its target genes in response to
scape of solute carriers and identified another transporter Dox, including gene pathways involved in DNA repair and
SLC22A3 (also known as organic cation transporter 3 or consequently an inability to mediate DNA repair after Dox
OCT3) as a critical player regulating cardiac accumulation treatment. This results in enhanced susceptivity to Dox-
of Dox [35]. They showed that OCT3 deficiency markedly induced cardio-cytotoxicity [43]. The findings by Huang
reduces Dox accumulation in cardiac tissue and protects et al. thus suggest another novel cellular pathway mediated
mice from Dox-induced cardiac injury and left ventricular by RARG that could be targeted for protecting against DIC.
dysfunction. They demonstrated that pharmacological inhi-
bition of OCT3 (e.g., by nilotinib) preserves cardiac function Non‑coding SNPs
following treatment with Dox without affecting its plasma
levels or antitumor effects in multiple models of leukemia Genetic variations in the non-coding sequences may also
and breast cancer. Hence, the findings identify OCT3 as a affect DIC. In this regard, a genome-wide association study
previously unrecognized Dox uptake transporter in cardio- (GWAS) identified an association of a non-coding SNP
myocytes that can be targeted pharmacologically to aug- rs28714259 (G/A) with an increased risk of developing Dox-
ment the therapeutic index of Dox [35]. As SNPs in OCT3 induced heart failure [44]. Using hiPSC-CMs with either
gene have been identified in human populations and to be intrinsic polymorphism or CRISPR-Cas9-mediated deletion

13
Molecular and Cellular Biochemistry

of rs28714259 locus, Schneider and coworkers [45] recently [53]. CircITCH (circular RNA ITCH [E3 ubiquitin-protein
found that rs28714259 disrupts glucocorticoid receptor (GR) ligase]) is a broad-spectrum tumor-suppressive circRNA.
binding and thus blocks GR-activated expression of genes Early studies suggested that the host gene of CircITCH,
involved in adaptive survival responses to Dox-induced ITCH (E3 ubiquitin-protein ligase), is involved in DIC [54].
cardio-cytotoxicity. As a result, rs28714259 risk allele or Using hiPSC-CMs and specimens from cancer patients who
loss of rs28714259 locus decreases dexamethasone-induced suffered from Dox-induced cardiomyopathy, Han et al. found
protection of cardiomyocyte viability and contractility after that CircITCH is expressed in both human cardiac tissue
doxorubicin exposure [45]. Glucocorticoids remain a stand- and hiPSC-CMs, and its expression is significantly down-
ard therapy in the concurrent treatment with Dox to prevent regulated in DIC. Overexpression of CircITCH in hiPSC-
nausea and allergic reactions [46]. Hence, the above findings CMs protects against Dox cytotoxicity. Mechanistically,
highlight additional benefits of adjunctive glucocorticoid CircITCH functions as a competing endogenous RNA to
therapy in reducing DIC in cancer patients [45]. sequester miR-330-5p, resulting in activation of cytopro-
tective SIRT6, BIRC5, and ATP2A2 and the consequently
increased resistance of cardiomyocytes to Dox-induced
Regulatory RNAs in doxorubicin mitochondrial oxidative stress and DNA damage. In this
cardiotoxicity context, miR-330-5p interacts with the 3′-UTRs of SIRT6,
BIRC5, and ATP2A2 mRNAs to cause their downregulation;
Overview of regulatory RNAs accordingly, sequestering miR-330-5p by CircITCH pre-
vents this downregulation, thereby augmenting the expres-
Regulatory RNAs are composed mostly of non-coding sion of these cytoprotective factors. Notably, overexpression
RNAs (ncRNAs). Among the regulatory RNAs, microRNAs of CircITCH in mice represses the levels of miR-330-5p,
(miRNAs), long non-coding RNAs (lncRNAs), and circular upregulates SIRT6, BIRC5, and ATP2A2, and attenuates
RNAs (circRNAs) have received the most attention in recent Dox-induced cardiomyopathy and left ventricular dysfunc-
years for their ability to modulate diverse physiological and tion. Collectively, these findings reveal a novel role of the
pathophysiological processes [47–49], including cardiovas- “CircITCH/miR-330-5p/SIRT6-BIRC5-ATP2A2” axis as a
cular homeostasis and pathologies [50–52]. Over the past competing endogenous RNA network governing DIC [53].
few years, the potential involvement of a number of regula- The findings also point to the feasibility of using a circu-
tory RNAs, including miRNAs, lncRNAs, and circRNAs, in lar RNA (i.e., CircITCH)-based modality to protect against
DIC has been reported in the literature though high-quality DIC in cancer patients. In this regard, circRNAs exhibit high
mechanistic studies published in influential journals are rela- druggability due to their high stability arising from special
tively few. Hence, in the sections below, we focus only on closed loop structure [55].
discussing the novel findings from these few mechanistic
studies. Circular RNA CircINSR

Regulatory RNAs in doxorubicin cardiotoxicity: A circular RNA derived from the insulin receptor locus,
recent mechanistic studies known as CircINSR, has recently been shown to also
protect against DIC [56]. While insulin receptor signal-
lncRNA SLC28A3‑AS1 ing plays an important role in cardiac homeostasis [57,
58], the role of CircINSR, a highly species-conserved
As discussed earlier, the SLC28A3-overlapping antisense circRNA derived from the insulin receptor gene, in car-
lncRNA SLC28A3-AS1 is a critical mediator of the associa- diac physiology and disease remains unclear. Lu et al.
tion of SNP rs11140490 with decreased SLC28A3 expres- [56] recently showed that CircINSR is downregulated in
sion (and thus decreased Dox uptake into cardiomyocytes) heart failure in both animal models and humans, including
[33]. This illustrates a scenario where a lncRNA mediates heart failure induced by Dox. CircINSP downregulation
an SNP-induced downregulation of a membrane transporter leads to mitochondrial damage and cardiomyocyte death,
for Dox. and cardiac dysfunction. Conversely, overexpression of
CircINSR prevents Dox-induced cytotoxicity in cultured
Circular RNA CircITCH human cardiomyocytes and dramatically protects against
Dox-induced cardiomyopathy and ventricular dysfunction
In an exhaustive mechanistic study, Han et al. discovered in mice. Mechanistically, CircINSR physically interacts
that the tumor-suppressive human circular RNA CircITCH with the single-stranded DNA-binding protein 1 (SSBP1)
sequesters miR-330-5p to ameliorate DIC, revealing an inti- to maintain mitochondrial function by stabilizing mito-
mate interaction between a circRNA and an miRNA in DIC chondrial DNA during Dox exposure, thereby preventing

13
Molecular and Cellular Biochemistry

Dox-induced mitochondrial damage and cardiomyocyte Other emerging targets of doxorubicin


death [56]. Notably, a human CircINSR mimics, synthe- cardiotoxicity
sized through in vitro transcription and subsequent circu-
larization via a DNA splint and enzymatic ligation, also In addition to the novel molecular targets and signaling
prevents Dox-induced cardiac cytotoxicity, highlighting pathways described in the preceding sections, several addi-
the feasibility of using CircINSR-based therapeutics for tional molecular and cellular targets have emerged over the
the intervention of DIC in cancer patients [56]. past few years to either underly DIC or serve as endogenous
In summary, multiple regulatory RNAs have been impli- defenses against DIC. The sections below highlight some
cated in DIC, either as mediators or protectors of DIC, notable studies.
depending on the mechanistic pathways involved (Fig. 3).
As regulatory RNAs play important roles in diverse physi-
ological and pathophysiological processes, targeting these Cardiac macrophages
molecules by Dox likely also causes diverse consequences.
Hence, future studies should determine not only the role of Macrophages are found in all tissues and exhibit great func-
regulatory RNAs in DIC, but also their impact on Dox’s tional diversities, including roles in physiological homeosta-
anticancer efficacy. Likewise, future investigation is war- sis and pathophysiological processes. Tissue macrophages
ranted to assess the potential adverse effects of regula- include: (1) those derived from blood monocytes, which
tory RNA-based therapeutics on body systems. This is in turn are derived from bone marrow progenitors and (2)
particularly relevant in view of the current high enthu- resident macrophages, which are derived from embryonic
siasm in developing regulatory RNA-based therapies for progenitors that populate all tissues, and persist through-
human diseases [55, 59]. On the other hand, mechanistic out life [60]. Cardiac macrophages play important roles
studies on regulatory RNAs in DIC not only deepen our in cardiac homeostasis and disease [61–64]. For example,
understanding of the molecular mechanisms by which Dox cardiac resident macrophages prevent fibrosis and stimulate
causes cardiomyopathy and heart failure, but also advance angiogenesis in response to cardiac pressure overload in
our knowledge of cardiac disease pathogenesis. mice [64]. This is in line with the notion that tissue resident
macrophages help resolve inflammation and tissue damage,
whereas recruited blood monocyte-derived macrophages
possess a proinflammatory phenotype, contributing to
inflammatory stress and tissue injury [60, 65, 66].
Cardiac macrophages are also involved in Dox-induced
cardiomyopathy [67]. In this regard, recruited blood-borne
macrophages with a proinflammatory phenotype constitute
the primary source of accumulated cardiac macrophages,
dominating the progression of Dox-induced cardiomyopathy
in mice. In contrast, proliferation of cardiac resident mac-
rophages driven by SR-A1-c-Myc signaling axis attenuates
Dox-induced cardiomyopathy by helping resolving inflam-
mation and retarding cardiomyopathy progression. Hence,
the SR-A1-c-Myc axis may represent a promising target to
treat Dox cardiomyopathy through augmentation of cardiac
resident reparative macrophage proliferation [67].

Circulating hemopexin

As aforementioned, iron plays a critical role in DIC, espe-


cially myocardial ferroptosis, and chelation of iron ame-
liorates Dox-induced cardiomyopathy and heart failure
Fig. 3  Role of circRNAs in doxorubicin cardiotoxicity. As illustrated, in cancer patients [68, 69]. A recent study demonstrated
circRNAs may modulate doxorubicin cardiotoxicity via either alter- that hemopexin (a circulating heme-binding protein [70])
ing endogenous cardioprotective defenses or regulating mitochon- modulates Dox-induced cardiomyopathy in patients and
drial DNA (mitoDNA) stability. See text for detailed description of
the original findings on which this diagram is primarily based. (+) mice [71]. Specifically, via proteomic analysis of plasma
denotes increase or activation; (−) denotes decrease or inhibition samples from Dox-treated patients, the authors showed that

13
Molecular and Cellular Biochemistry

increased hemopexin is associated with cardiac toxicity and Notably, using hiPSC-CMs, the authors also found that
dysfunction assessed by echocardiography. Administration overexpression of Nrf1 markedly attenuates Dox-induced
of hemopexin to wild-type mice treated with Dox improves cardio-cytotoxicity, as evidenced by improved cell viability
cardiac function. Conversely, hemopexin-knockout mice and preserved contractility [81]. This finding suggests that
show increased Dox cardiac toxicity compared to wild-type Nrf1 (a highly expressed CNC transcription factor in car-
mice. Mechanistically, hemopexin is likely transported to diomyocytes) may function as an important cytoprotector
the heart by circulating monocytes/macrophages and that against DIC. Further studies are warranted to determine the
hemopexin mitigates Dox-induced ferroptosis to confer cardioprotective function of Nrf1 in animal models of DIC.
cardioprotection. Together, these observations suggest that
hemopexin induction represents a compensatory response
during Dox treatment [71]. The findings may motivate addi- Conclusion and perspectives
tional research to determine whether the hemopexin path-
way could be leveraged for therapeutic benefit in patients While oxidative stress, mitochondrial damage, and topoi-
at risk of Dox cardiomyopathy. In this context, based on somerase inhibition remain the key mechanistic scheme of
its beneficial effects in reverting heme-induced damage in DIC, the past few years have witnessed a rapid growth in
animal models of sick cell disease (SCD) [72], hemopexin the number of high-quality mechanistic studies reported in
is currently under clinical trial to determine its efficacy in influential journals. These studies have uncovered a range
treating SCD patients (https://​sickl​ecell​socie​ty.​org/​csl889-​ of novel molecular and cellular targets/signaling pathways
clini​cal-​trial/). underlying DIC in animal models as well as patient-specific
hiPSC-CMs. These include mitochondrial iron-dependent
Nrf1 ferroptosis, genetic variations and regulatory RNAs, as well
as cardiac resident macrophages, circulating hemopexin,
The nuclear factor E2-related factor 1 (Nrf1) belongs to the and Nrf1 signaling. These novel findings not only broaden
Cap’N’Collar (CNC) family of leucine zipper transcription our understanding of the molecular mechanisms of DIC,
factors, which include also Nrf2 (nuclear factor E2-related but also provide opportunities to develop mechanistically
factor 2), Nrf3 (nuclear factor E2-related factor 3), and Nfe2 based modalities for DIC intervention in cancer patients. In
(nuclear factor E2) [73, 74]. While Nrf2 is the most exten- addition, these advances may also benefit the management
sively studied transcriptional activator of cytoprotective of cardiomyopathy caused by other drugs or pathological
genes, studies on Nrf1 are relatively fewer. Like Nrf2, Nrf1 conditions. It should be noted, however, that the primary
also binds to the antioxidant response element (ARE), but goal of cancer chemotherapy is to eradicate cancer cells, and
Nrf1 and Nrf2 play distinct roles in the activation of ARE- as such, cardioprotective approach in such patients should
dependent genes. For example, while both Nrf1 and Nrf2 not hinder the cancer-killing activity of the chemotherapy. It
bind with comparable affinity to the ARE in the metallothio- is thus imperative that experimental studies on cardioprotec-
nein-1 (MT1) gene promoter, only Nrf1 is able to upregulate tion of DIC should also simultaneously assess the impact on
reporter gene expression driven by the MT1 promoter [75]. Dox’s anticancer activity.
On the other hand, the inducible expression of many other As discussed in this review, multiple underlying mecha-
antioxidant and cytoprotective genes is largely dependent on nisms and numerous molecular targets are involved in DIC,
Nrf2. In contrast to Nrf2, Nrf1 is an essential gene during and each of them is concluded to be a significant cause of
development. Homozygous deletion of Nrf1 gene in mice DIC by the individual high-quality study. An immediate
results in late gestational embryonic lethality [76]. question is which one really plays a dominant role. While
Nrf1 is a critical regulator of multiple essential cellular there is no easy answer to this question, it is likely that the
processes, including cholesterol homeostasis [77], ferropto- significance of a particular finding made in a specific experi-
sis [78], and proteostasis [79, 80]. More recently, Cui et al. mental model might not be applicable to other experimental
showed that among the CNC family transcription factors, models or conditions. In this context, while studies using
Nrf1 is the most highly expressed member in the heart and animal models are critical for advancing knowledge, their
promotes heart regeneration and repair by regulating pro- validation and translatability into clinical medicine remain
teostasis and redox balance. [81]. Specifically, using a neo- a pressing challenge. It is hoped that continued systematic
natal mouse heart regeneration model, the authors demon- mechanistic studies would eventually lead to a clear pic-
strated that Nrf1 is activated in regenerating cardiomyocytes. ture of the predominant mechanism of DIC or elucidation
Genetic deletion of Nrf1 prevents regenerating cardiomyo- of a final common pathway at which distinct mechanisms
cytes from activating a transcriptional program required for converge to cause cardiomyopathy and heart failure in Dox-
heart regeneration. Conversely, Nrf1 overexpression protects treated patients.
the adult mouse heart from ischemia/reperfusion injury.

13
Molecular and Cellular Biochemistry

Author contributions  All authors contributed to the review article topic form of nonapoptotic cell death. Cell 149:1060–1072. https://​doi.​
conception, literature searching and analysis, and the writing of the org/​10.​1016/j.​cell.​2012.​03.​042
manuscript. All authors read and approved the final manuscript. 11. Yang WS, Kim KJ, Gaschler MM, Patel M, Shchepinov MS,
Stockwell BR (2016) Peroxidation of polyunsaturated fatty acids
Funding  This work was supported in part by an IIG Grant (09A084) by lipoxygenases drives ferroptosis. Proc Natl Acad Sci U S A
from the American Institute for Cancer Research and a Grant 113:E4966–E4975. https://​doi.​org/​10.​1073/​pnas.​16032​44113
(CA192936) from the National Institutes of Health/National Cancer 12. Kagan VE, Mao G, Qu F, Angeli JP, Doll S, Croix CS, Dar HH,
Institute. Liu B, Tyurin VA, Ritov VB, Kapralov AA, Amoscato AA, Jiang
J, Anthonymuthu T, Mohammadyani D, Yang Q, Proneth B,
Data availability  Not applicable as this is a review article. Klein-Seetharaman J, Watkins S, Bahar I, Greenberger J, Mal-
lampalli RK, Stockwell BR, Tyurina YY, Conrad M, Bayir H
Declarations  (2017) Oxidized arachidonic and adrenic PEs navigate cells to
ferroptosis. Nat Chem Biol 13:81–90. https://​doi.​org/​10.​1038/​
Conflict of interest  The author has no relevant financial or non-finan- nchem​bio.​2238
cial interests to disclose. 13. Jiang X, Stockwell BR, Conrad M (2021) Ferroptosis: mecha-
nisms, biology and role in disease. Nat Rev Mol Cell Biol 22:266–
Ethical approval  Not applicable. This work does not involve animals 282. https://​doi.​org/​10.​1038/​s41580-​020-​00324-8
or human subjects. 14. Ichikawa Y, Ghanefar M, Bayeva M, Wu R, Khechaduri A, Naga
Prasad SV, Mutharasan RK, Naik TJ, Ardehali H (2014) Cardio-
toxicity of doxorubicin is mediated through mitochondrial iron
accumulation. J Clin Invest 124:617–630. https://d​ oi.o​ rg/1​ 0.1​ 172/​
JCI72​931
15. Li R, Jia Z, Zhu H (2019) Regulation of Nrf2 signaling. React
References Oxyg Species (Apex) 8:312–322. https://​doi.​org/​10.​20455/​ros.​
2019.​865
1. Bonadonna G, Monfardini S, De Lena M, Fossati-Bellani F (1969) 16. Li S, Wang W, Niu T, Wang H, Li B, Shao L, Lai Y, Li H, Janicki
Clinical evaluation of adriamycin, a new antitumour antibiotic. Br JS, Wang XL, Tang D, Cui T (2014) Nrf2 deficiency exaggerates
Med J 3:503–506 doxorubicin-induced cardiotoxicity and cardiac dysfunction. Oxid
2. Zhu H, Sarkar S, Scott L, Danelisen I, Trush MA, Jia Z, Li YR Med Cell Longev 2014:748524. https://​doi.​org/​10.​1155/​2014/​
(2016) Doxorubicin redox biology: redox cycling, topoisomer- 748524
ase inhibition, and oxidative stress. React Oxyg Species (Apex) 17. Hull TD, Boddu R, Guo L, Tisher CC, Traylor AM, Patel B,
1:189–198. https://​doi.​org/​10.​20455/​ros.​2016.​835 Joseph R, Prabhu SD, Suliman HB, Piantadosi CA, Agarwal A,
3. Higgins AY, O’Halloran TD, Chang JD (2015) Chemotherapy- George JF (2016) Heme oxygenase-1 regulates mitochondrial
induced cardiomyopathy. Heart Fail Rev 20:721–730. https://​doi.​ quality control in the heart. JCI Insight 1:e85817. https://​doi.​org/​
org/​10.​1007/​s10741-​015-​9502-y 10.​1172/​jci.​insig​ht.​85817
4. Page RL, O’Bryant CL, Cheng D, Dow TJ, Ky B, Stein CM, 18. Tadokoro T, Ikeda M, Ide T, Deguchi H, Ikeda S, Okabe K, Ishi-
Spencer AP, Trupp RJ, Lindenfeld J, American Heart Association kita A, Matsushima S, Koumura T, Yamada KI, Imai H, Tsutsui
Clinical P, Heart F, Transplantation Committees of the Council H (2020) Mitochondria-dependent ferroptosis plays a pivotal role
on Clinical C, Council on Cardiovascular S, Anesthesia, Coun- in doxorubicin cardiotoxicity. JCI Insight. https://d​ oi.o​ rg/1​ 0.1​ 172/​
cil on C, Stroke N, Council on Quality of C and Outcomes R jci.​insig​ht.​132747
(2016) Drugs That may cause or exacerbate heart failure: a scien- 19. de Baat EC, van Dalen EC, Mulder RL, Hudson MM, Ehrhardt
tific statement from the american heart association. Circulation MJ, Engels FK, Feijen EAM, Grotenhuis HB, Leerink JM,
134:e32-69. https://​doi.​org/​10.​1161/​CIR.​00000​00000​000426 Kapusta L, Kaspers GJL, Merkx R, Mertens L, Skinner R, Tiss-
5. Wallace KB, Sardao VA, Oliveira PJ (2020) Mitochondrial ing WJE, de Vathaire F, Nathan PC, Kremer LCM, Mavinkurve-
determinants of doxorubicin-induced cardiomyopathy. Circ Res Groothuis AMC, Armenian S (2022) Primary cardioprotection
126:926–941. https://d​ oi.o​ rg/1​ 0.1​ 161/C
​ IRCRE
​ SAHA.1​ 19.3​ 14681 with dexrazoxane in patients with childhood cancer who are
6. Christidi E, Brunham LR (2021) Regulated cell death pathways expected to receive anthracyclines: recommendations from the
in doxorubicin-induced cardiotoxicity. Cell Death Dis 12:339. international late effects of childhood cancer guideline harmoniza-
https://​doi.​org/​10.​1038/​s41419-​021-​03614-x tion group. Lancet Child Adolesc Health 6:885–894. https://​doi.​
7. Tang D, Kang R, Berghe TV, Vandenabeele P, Kroemer G (2019) org/​10.​1016/​S2352-​4642(22)​00239-5
The molecular machinery of regulated cell death. Cell Res 20. Macedo AVS, Hajjar LA, Lyon AR, Nascimento BR, Putzu A,
29:347–364. https://​doi.​org/​10.​1038/​s41422-​019-​0164-5 Rossi L, Costa RB, Landoni G, Nogueira-Rodrigues A, Ribeiro
8. Tsvetkov P, Coy S, Petrova B, Dreishpoon M, Verma A, Abdu- ALP (2019) Efficacy of dexrazoxane in preventing anthracycline
samad M, Rossen J, Joesch-Cohen L, Humeidi R, Spangler RD, cardiotoxicity in breast cancer. JACC CardioOncol 1:68–79.
Eaton JK, Frenkel E, Kocak M, Corsello SM, Lutsenko S, Kanarek https://​doi.​org/​10.​1016/j.​jaccao.​2019.​08.​003
N, Santagata S, Golub TR (2022) Copper induces cell death by 21. Abe K, Ikeda M, Ide T, Tadokoro T, Miyamoto HD, Furusawa S,
targeting lipoylated TCA cycle proteins. Science 375:1254–1261. Tsutsui Y, Miyake R, Ishimaru K, Watanabe M, Matsushima S,
https://​doi.​org/​10.​1126/​scien​ce.​abf05​29 Koumura T, Yamada KI, Imai H, Tsutsui H (2022) Doxorubicin
9. Fang X, Wang H, Han D, Xie E, Yang X, Wei J, Gu S, Gao F, causes ferroptosis and cardiotoxicity by intercalating into mito-
Zhu N, Yin X, Cheng Q, Zhang P, Dai W, Chen J, Yang F, Yang chondrial DNA and disrupting Alas1-dependent heme synthesis.
HT, Linkermann A, Gu W, Min J, Wang F (2019) Ferroptosis as a Sci Signal 15:eabn8017. https://​doi.​org/​10.​1126/​scisi​gnal.​abn80​
target for protection against cardiomyopathy. Proc Natl Acad Sci 17
U S A 116:2672–2680. https://​doi.​org/​10.​1073/​pnas.​18210​22116 22. Stockwell BR (2022) Ferroptosis turns 10: Emerging mecha-
10. Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zaitsev EM, nisms, physiological functions, and therapeutic applications. Cell
Gleason CE, Patel DN, Bauer AJ, Cantley AM, Yang WS, Mor- 185:2401–2421. https://​doi.​org/​10.​1016/j.​cell.​2022.​06.​003
rison B 3rd, Stockwell BR (2012) Ferroptosis: an iron-dependent

13
Molecular and Cellular Biochemistry

23. Sandoval-Acuna C, Torrealba N, Tomkova V, Jadhav SB, Blaz- selectivity by concentrative nucleoside transporters. Elife
kova K, Merta L, Lettlova S, Adamcova MK, Rosel D, Brabek J, 3:e03604. https://​doi.​org/​10.​7554/​eLife.​03604
Neuzil J, Stursa J, Werner L, Truksa J (2021) Targeting mitochon- 35. Huang KM, Zavorka Thomas M, Magdy T, Eisenmann ED, Uddin
drial iron metabolism suppresses tumor growth and metastasis by ME, DiGiacomo DF, Pan A, Keiser M, Otter M, Xia SH, Li Y, Jin
inducing mitochondrial dysfunction and mitophagy. Cancer Res Y, Fu Q, Gibson AA, Bonilla IM, Carnes CA, Corps KN, Cop-
81:2289–2303. https://​doi.o​ rg/1​ 0.1​ 158/​0008-​5472.​CAN-​20-​1628 pola V, Smith SA, Addison D, Nies AT, Bundschuh R, Chen T,
24. Roden DM, Johnson JA, Kimmel SE, Krauss RM, Medina MW, Lustberg MB, Wang J, Oswald S, Campbell MJ, Yan PS, Baker
Shuldiner A, Wilke RA (2011) Cardiovascular pharmacogenom- SD, Hu S, Burridge PW, Sparreboom A (2021) Targeting OCT3
ics. Circ Res 109:807–820. https://​d oi.​o rg/​1 0.​1 161/​C IRCR​ attenuates doxorubicin-induced cardiac injury. Proc Natl Acad Sci
ESAHA.​110.​230995 U S A. https://​doi.​org/​10.​1073/​pnas.​20201​68118
25. O’Donnell CJ, Nabel EG (2011) Genomics of cardiovascular 36. Qi Q, Workalemahu T, Zhang C, Hu FB, Qi L (2012) Genetic
disease. N Engl J Med 365:2098–2109. https://​doi.​org/​10.​1056/​ variants, plasma lipoprotein(a) levels, and risk of cardiovascular
NEJMr​a1105​239 morbidity and mortality among two prospective cohorts of type 2
26. Semsarian C, Ingles J, Ross SB, Dunwoodie SL, Bagnall RD, diabetes. Eur Heart J 33:325–334. https://​doi.​org/​10.​1093/​eurhe​
Kovacic JC (2021) Precision medicine in cardiovascular disease: artj/​ehr350
genetics and impact on phenotypes: JACC focus seminar 1/5. J 37. Tregouet DA, Konig IR, Erdmann J, Munteanu A, Braund PS,
Am Coll Cardiol 77:2517–2530. https://​doi.​org/​10.​1016/j.​jacc.​ Hall AS, Grosshennig A, Linsel-Nitschke P, Perret C, DeSuremain
2020.​12.​071 M, Meitinger T, Wright BJ, Preuss M, Balmforth AJ, Ball SG,
27. Wojnowski L, Kulle B, Schirmer M, Schluter G, Schmidt A, Meisinger C, Germain C, Evans A, Arveiler D, Luc G, Ruidav-
Rosenberger A, Vonhof S, Bickeboller H, Toliat MR, Suk EK, ets JB, Morrison C, van der Harst P, Schreiber S, Neureuther K,
Tzvetkov M, Kruger A, Seifert S, Kloess M, Hahn H, Loeffler M, Schafer A, Bugert P, El Mokhtari NE, Schrezenmeir J, Stark K,
Nurnberg P, Pfreundschuh M, Trumper L, Brockmoller J, Hasen- Rubin D, Wichmann HE, Hengstenberg C, Ouwehand W, Con-
fuss G (2005) NAD(P)H oxidase and multidrug resistance protein trol WTC, C, Cardiogenics C, Ziegler A, Tiret L, Thompson JR,
genetic polymorphisms are associated with doxorubicin-induced Cambien F, Schunkert H and Samani NJ, (2009) Genome-wide
cardiotoxicity. Circulation 112:3754–3762. https://​doi.​org/​10.​ haplotype association study identifies the SLC22A3-LPAL2-LPA
1161/​CIRCU​LATIO​NAHA.​105.​576850 gene cluster as a risk locus for coronary artery disease. Nat Genet
28. Todorova VK, Makhoul I, Dhakal I, Wei J, Stone A, Carter W, 41:283–285. https://​doi.​org/​10.​1038/​ng.​314
Owen A, Klimberg VS (2017) Polymorphic variations associated 38. Leabman MK, Huang CC, DeYoung J, Carlson EJ, Taylor TR, de
with doxorubicin-induced cardiotoxicity in breast cancer patients. la Cruz M, Johns SJ, Stryke D, Kawamoto M, Urban TJ, Kroetz
Oncol Res 25:1223–1229. https://​doi.​org/​10.​3727/​09650​4017X​ DL, Ferrin TE, Clark AG, Risch N, Herskowitz I, Giacomini KM,
14876​24509​6439 Transporters POM (2003) Natural variation in human membrane
29. Sapkota Y, Qin N, Ehrhardt MJ, Wang Z, Chen Y, Wilson CL, transporter genes reveals evolutionary and functional constraints.
Estepp J, Rai P, Hankins JS, Burridge PW, Jefferies JL, Zhang J, Proc Natl Acad Sci U S A 100:5896–5901. https://​doi.​org/​10.​
Hudson MM, Robison LL, Armstrong GT, Mulrooney DA, Yasui 1073/​pnas.​07308​57100
Y (2021) Genetic variants associated with therapy-related cardio- 39. Schaller L, Lauschke VM (2019) The genetic landscape of the
myopathy among childhood cancer survivors of african ancestry. human solute carrier (SLC) transporter superfamily. Hum Genet
Cancer Res 81:2556–2565. https://​doi.​org/​10.​1158/​0008-​5472.​ 138:1359–1377. https://​doi.​org/​10.​1007/​s00439-​019-​02081-x
CAN-​20-​2675 40. Aminkeng F, Bhavsar AP, Visscher H, Rassekh SR, Li Y, Lee JW,
30. Visscher H, Ross CJ, Rassekh SR, Barhdadi A, Dube MP, Al- Brunham LR, Caron HN, van Dalen EC, Kremer LC, van der Pal
Saloos H, Sandor GS, Caron HN, van Dalen EC, Kremer LC, HJ, Amstutz U, Rieder MJ, Bernstein D, Carleton BC, Hayden
van der Pal HJ, Brown AM, Rogers PC, Phillips MS, Rieder MJ, MR, Ross CJ, Canadian Pharmacogenomics Network for Drug
Carleton BC, Hayden MR, Network CP, for Drug Safety C, (2012) Safety C (2015) A coding variant in RARG confers susceptibility
Pharmacogenomic prediction of anthracycline-induced cardiotox- to anthracycline-induced cardiotoxicity in childhood cancer. Nat
icity in children. J Clin Oncol 30:1422–1428. https://​doi.​org/​10.​ Genet 47:1079–1084. https://​doi.​org/​10.​1038/​ng.​3374
1200/​JCO.​2010.​34.​3467 41. Magdy T, Jiang Z, Jouni M, Fonoudi H, Lyra-Leite D, Jung G,
31. Visscher H, Ross CJ, Rassekh SR, Sandor GS, Caron HN, van Romero-Tejeda M, Kuo HH, Fetterman KA, Gharib M, Bur-
Dalen EC, Kremer LC, van der Pal HJ, Rogers PC, Rieder MJ, meister BT, Zhao M, Sapkota Y, Ross CJ, Carleton BC, Bernstein
Carleton BC, Hayden MR and Consortium C (2013) Validation of D, Burridge PW (2021) RARG variant predictive of doxorubicin-
variants in SLC28A3 and UGT1A6 as genetic markers predictive induced cardiotoxicity identifies a cardioprotective therapy. Cell
of anthracycline-induced cardiotoxicity in children. Pediatr Blood Stem Cell 28:2076–2089. https://​doi.​org/​10.​1016/j.​stem.​2021.​08.​
Cancer 60:1375–1381. https://​doi.​org/​10.​1002/​pbc.​24505 006
32. Aminkeng F, Ross CJ, Rassekh SR, Hwang S, Rieder MJ, Bhavsar 42. Zhang S, Liu X, Bawa-Khalfe T, Lu LS, Lyu YL, Liu LF, Yeh
AP, Smith A, Sanatani S, Gelmon KA, Bernstein D, Hayden MR, ET (2012) Identification of the molecular basis of doxorubicin-
Amstutz U, Carleton BC and Group CCPR (2016) Recommenda- induced cardiotoxicity. Nat Med 18:1639–1642. https://​doi.​org/​
tions for genetic testing to reduce the incidence of anthracycline- 10.​1038/​nm.​2919
induced cardiotoxicity. Br J Clin Pharmacol 82:683–695. https://​ 43. Huang H, Christidi E, Shafaattalab S, Davis MK, Tibbits GF,
doi.​org/​10.​1111/​bcp.​13008 Brunham LR (2022) RARG S427L attenuates the DNA repair
33. Magdy T, Jouni M, Kuo HH, Weddle CJ, Lyra-Leite D, Fonoudi response to doxorubicin in induced pluripotent stem cell-derived
H, Romero-Tejeda M, Gharib M, Javed H, Fajardo G, Ross CJD, cardiomyocytes. Stem Cell Rep 17:756–765. https://​doi.​org/​10.​
Carleton BC, Bernstein D, Burridge PW (2022) Identification 1016/j.​stemcr.​2022.​03.​002
of Drug Transporter Genomic Variants and Inhibitors That Pro- 44. Schneider BP, Shen F, Gardner L, Radovich M, Li L, Miller KD,
tect Against Doxorubicin-Induced Cardiotoxicity. Circulation Jiang G, Lai D, O’Neill A, Sparano JA, Davidson NE, Cameron D,
145:279–294. https://​doi.​org/​10.​1161/​CIRCU​LATIO​NAHA.​121.​ Gradus-Pizlo I, Mastouri RA, Suter TM, Foroud T, Sledge GW Jr
055801 (2017) Genome-wide association study for anthracycline-induced
34. Johnson ZL, Lee JH, Lee K, Lee M, Kwon DY, Hong J, Lee congestive heart failure. Clin Cancer Res 23:43–51. https://​doi.​
SY (2014) Structural basis of nucleoside and nucleoside drug org/​10.​1158/​1078-​0432.​CCR-​16-​0908

13
Molecular and Cellular Biochemistry

45. Wu X, Shen F, Jiang G, Xue G, Philips S, Gardner L, Cunning- 61. McNally EM (2020) Cardiac macrophages - keeping the engine
ham G, Bales C, Cantor E, Schneider BP (2022) A non-coding running clean. N Engl J Med 383:2474–2476. https://​doi.​org/​10.​
GWAS variant impacts anthracycline-induced cardiotoxic phe- 1056/​NEJMc​ibr20​30271
notypes in human iPSC-derived cardiomyocytes. Nat Commun 62. Hulsmans M, Clauss S, Xiao L, Aguirre AD, King KR, Hanley
13:7171. https://​doi.​org/​10.​1038/​s41467-​022-​34917-y A, Hucker WJ, Wulfers EM, Seemann G, Courties G, Iwamoto
46. Grunberg SM (2007) Antiemetic activity of corticosteroids in Y, Sun Y, Savol AJ, Sager HB, Lavine KJ, Fishbein GA, Capen
patients receiving cancer chemotherapy: dosing, efficacy, and DE, Da Silva N, Miquerol L, Wakimoto H, Seidman CE, Seidman
tolerability analysis. Ann Oncol 18:233–240. https://​doi.​org/​10.​ JG, Sadreyev RI, Naxerova K, Mitchell RN, Brown D, Libby P,
1093/​annonc/​mdl347 Weissleder R, Swirski FK, Kohl P, Vinegoni C, Milan DJ, Ellinor
47. (2019) A focus on regulatory RNAs. Nat Cell Biol 21: 535. PT, Nahrendorf M (2017) Macrophages facilitate electrical con-
https://​doi.​org/​10.​1038/​s41556-​019-​0325-2 duction in the heart. Cell 169:510–522. https://​doi.​org/​10.​1016/j.​
48. Chen LL (2020) The expanding regulatory mechanisms and cell.​2017.​03.​050
cellular functions of circular RNAs. Nat Rev Mol Cell Biol 63. Sugita J, Fujiu K, Nakayama Y, Matsubara T, Matsuda J, Oshima
21:475–490. https://​doi.​org/​10.​1038/​s41580-​020-​0243-y T, Liu Y, Maru Y, Hasumi E, Kojima T, Seno H, Asano K, Ishi-
49. Statello L, Guo CJ, Chen LL, Huarte M (2021) Gene regu- jima A, Tomii N, Yamazaki M, Kudo F, Sakuma I, Nagai R,
lation by long non-coding RNAs and its biological functions. Manabe I, Komuro I (2021) Cardiac macrophages prevent sudden
Nat Rev Mol Cell Biol 22:96–118. https://​d oi.​o rg/​1 0.​1 038/​ death during heart stress. Nat Commun 12:1910. https://​doi.​org/​
s41580-​020-​00315-9 10.​1038/​s41467-​021-​22178-0
50. Uchida S, Dimmeler S (2015) Long noncoding RNAs in cardio- 64. Revelo XS, Parthiban P, Chen C, Barrow F, Fredrickson G, Wang
vascular diseases. Circ Res 116:737–750. https://d​ oi.o​ rg/1​ 0.1​ 161/​ H, Yucel D, Herman A, van Berlo JH (2021) Cardiac resident
CIRCR​ESAHA.​116.​302521 macrophages prevent fibrosis and stimulate angiogenesis. Circ
51. Zhang Z, Salisbury D, Sallam T (2018) Long noncoding RNAs in Res 129:1086–1101. https://d​ oi.o​ rg/1​ 0.1​ 161/C
​ IRCRE
​ SAHA.1​ 21.​
atherosclerosis: JACC review topic of the week. J Am Coll Car- 319737
diol 72:2380–2390. https://​doi.​org/​10.​1016/j.​jacc.​2018.​08.​2161 65. Bleriot C, Chakarov S, Ginhoux F (2020) Determinants of resident
52. Gomes CPC, Schroen B, Kuster GM, Robinson EL, Ford K, tissue macrophage identity and function. Immunity 52:957–970.
Squire IB, Heymans S, Martelli F, Emanueli C, Devaux Y, Action https://​doi.​org/​10.​1016/j.​immuni.​2020.​05.​014
EU-CC (2020) Regulatory RNAs in heart failure. Circulation 66. Huang QQ, Doyle R, Chen SY, Sheng Q, Misharin AV, Mao Q,
141:313–328. https://d​ oi.​org/1​ 0.​1161/​CIRCU​LATIO​NAHA.​119.​ Winter DR, Pope RM (2021) Critical role of synovial tissue-
042474 resident macrophage niche in joint homeostasis and suppression
53. Han D, Wang Y, Wang Y, Dai X, Zhou T, Chen J, Tao B, Zhang of chronic inflammation. Sci Adv. https://​doi.​org/​10.​1126/​sciadv.​
J, Cao F (2020) The Tumor-suppressive human circular RNA Cir- abd05​15
cITCH sponges miR-330-5p to Ameliorate doxorubicin-induced 67. Zhang H, Xu A, Sun X, Yang Y, Zhang L, Bai H, Ben J, Zhu X,
cardiotoxicity through upregulating SIRT6, survivin, and SER- Li X, Yang Q, Wang Z, Wu W, Yang D, Zhang Y, Xu Y, Chen
CA2a. Circ Res 127:e108–e125. https://​doi.​org/​10.​1161/​CIRCR​ Q (2020) Self-maintenance of cardiac resident reparative mac-
ESAHA.​119.​316061 rophages attenuates doxorubicin-induced cardiomyopathy through
54. Otaki Y, Takahashi H, Watanabe T, Funayama A, Netsu S, Honda the SR-A1-c-Myc axis. Circ Res 127:610–627. https://​doi.​org/​10.​
Y, Narumi T, Kadowaki S, Hasegawa H, Honda S, Arimoto T, 1161/​CIRCR​ESAHA.​119.​316428
Shishido T, Miyamoto T, Kamata H, Nakajima O, Kubota I (2016) 68. Swain SM, Whaley FS, Gerber MC, Weisberg S, York M, Spicer
HECT-type ubiquitin E3 Ligase ITCH interacts with thioredoxin- D, Jones SE, Wadler S, Desai A, Vogel C, Speyer J, Mittelman A,
interacting protein and ameliorates reactive oxygen species- Reddy S, Pendergrass K, Velez-Garcia E, Ewer MS, Bianchine JR,
induced cardiotoxicity. J Am Heart Assoc 5:e002485. https://​doi.​ Gams RA (1997) Cardioprotection with dexrazoxane for doxoru-
org/​10.​1161/​JAHA.​115.​002485 bicin-containing therapy in advanced breast cancer. J Clin Oncol
55. Winkle M, El-Daly SM, Fabbri M, Calin GA (2021) Noncod- 15:1318–1332. https://​doi.​org/​10.​1200/​JCO.​1997.​15.4.​1318
ing RNA therapeutics - challenges and potential solutions. 69. Lipshultz SE, Rifai N, Dalton VM, Levy DE, Silverman LB, Lip-
Nat Rev Drug Discov 20:629–651. https://​d oi.​o rg/​1 0.​1 038/​ sitz SR, Colan SD, Asselin BL, Barr RD, Clavell LA, Hurwitz
s41573-​021-​00219-z CA, Moghrabi A, Samson Y, Schorin MA, Gelber RD, Sallan SE
56. Lu D, Chatterjee S, Xiao K, Riedel I, Huang CK, Costa A, Cush- (2004) The effect of dexrazoxane on myocardial injury in doxoru-
man S, Neufeldt D, Rode L, Schmidt A, Juchem M, Leonardy J, bicin-treated children with acute lymphoblastic leukemia. N Engl
Buchler G, Blume J, Gern OL, Kalinke U, Wen Tan WL, Foo R, J Med 351:145–153. https://​doi.​org/​10.​1056/​NEJMo​a0351​53
Vink A, van Laake LW, van der Meer P, Bar C, Thum T (2022) 70. Takahashi N, Takahashi Y, Putnam FW (1985) Complete amino
A circular RNA derived from the insulin receptor locus protects acid sequence of human hemopexin, the heme-binding protein of
against doxorubicin-induced cardiotoxicity. Eur Heart J 43:4496– serum. Proc Natl Acad Sci U S A 82:73–77. https://​doi.​org/​10.​
4511. https://​doi.​org/​10.​1093/​eurhe​artj/​ehac3​37 1073/​pnas.​82.1.​73
57. Bertrand L, Horman S, Beauloye C, Vanoverschelde JL (2008) 71. Liu J, Lane S, Lall R, Russo M, Farrell L, Debreli Coskun M,
Insulin signalling in the heart. Cardiovasc Res 79:238–248. Curtin C, Araujo-Gutierrez R, Scherrer-Crosbie M, Trachtenberg
https://​doi.​org/​10.​1093/​cvr/​cvn093 BH, Kim J, Tolosano E, Ghigo A, Gerszten RE, Asnani A (2022)
58. Riehle C, Abel ED (2016) Insulin signaling and heart failure. Circ Circulating hemopexin modulates anthracycline cardiac toxicity
Res 118:1151–1169. https://d​ oi.o​ rg/1​ 0.1​ 161/C
​ IRCRE
​ SAHA.1​ 16.​ in patients and in mice. Sci Adv 8:edac9245. https://​doi.​org/​10.​
306206 1126/​sciadv.​adc92​45
59. Wu N, Qadir J, Yang BB (2022) CircRNA perspective: new strate- 72. Vinchi F, Costa da Silva M, Ingoglia G, Petrillo S, Brinkman
gies for RNA therapy. Trends Mol Med 28:530. https://d​ oi.o​ rg/1​ 0.​ N, Zuercher A, Cerwenka A, Tolosano E, Muckenthaler MU
1016/j.​molmed.​2022.​04.​001 (2016) Hemopexin therapy reverts heme-induced proinflamma-
60. Wynn TA, Chawla A, Pollard JW (2013) Macrophage biology tory phenotypic switching of macrophages in a mouse model of
in development, homeostasis and disease. Nature 496:445–455. sickle cell disease. Blood 127:473–486. https://​doi.​org/​10.​1182/​
https://​doi.​org/​10.​1038/​natur​e12034 blood-​2015-​08-​663245

13
Molecular and Cellular Biochemistry

73. Chan JY, Han XL, Kan YW (1993) Cloning of Nrf1, an NF-E2-re- 79. Radhakrishnan SK, Lee CS, Young P, Beskow A, Chan JY,
lated transcription factor, by genetic selection in yeast. Proc Natl Deshaies RJ (2010) Transcription factor Nrf1 mediates the protea-
Acad Sci U S A 90:11371–11375. https://​doi.​org/​10.​1073/​pnas.​ some recovery pathway after proteasome inhibition in mammalian
90.​23.​11371 cells. Mol Cell 38:17–28. https://​doi.​org/​10.​1016/j.​molcel.​2010.​
74. Sykiotis GP, Bohmann D (2010) Stress-activated cap’n’collar 02.​029
transcription factors in aging and human disease. Sci Signal. 80. Zhang Y, Nicholatos J, Dreier JR, Ricoult SJ, Widenmaier SB,
https://​doi.​org/​10.​1126/​scisi​gnal.​3112r​e3 Hotamisligil GS, Kwiatkowski DJ, Manning BD (2014) Coor-
75. Ohtsuji M, Katsuoka F, Kobayashi A, Aburatani H, Hayes JD, dinated regulation of protein synthesis and degradation by
Yamamoto M (2008) Nrf1 and Nrf2 play distinct roles in acti- mTORC1. Nature 513:440–443. https://​doi.​org/​10.​1038/​natur​
vation of antioxidant response element-dependent genes. J Biol e13492
Chem 283:33554–33562. https://​doi.​org/​10.​1074/​jbc.​M8045​ 81. Cui M, Atmanli A, Morales MG, Tan W, Chen K, Xiao X, Xu
97200 L, Liu N, Bassel-Duby R, Olson EN (2021) Nrf1 promotes
76. Chan JY, Kwong M, Lu R, Chang J, Wang B, Yen TS, Kan YW heart regeneration and repair by regulating proteostasis and
(1998) Targeted disruption of the ubiquitous CNC-bZIP transcrip- redox balance. Nat Commun 12:5270. https://​doi.​org/​10.​1038/​
tion factor, Nrf-1, results in anemia and embryonic lethality in s41467-​021-​25653-w
mice. EMBO J 17:1779–1787. https://​doi.​org/​10.​1093/​emboj/​
17.6.​1779 Publisher's Note Springer Nature remains neutral with regard to
77. Widenmaier SB, Snyder NA, Nguyen TB, Arduini A, Lee GY, jurisdictional claims in published maps and institutional affiliations.
Arruda AP, Saksi J, Bartelt A, Hotamisligil GS (2017) NRF1 Is
an ER Membrane sensor that is central to cholesterol homeostasis. Springer Nature or its licensor (e.g. a society or other partner) holds
Cell 171:1094–1109. https://​doi.​org/​10.​1016/j.​cell.​2017.​10.​003 exclusive rights to this article under a publishing agreement with the
78. Forcina GC, Pope L, Murray M, Dong W, Abu-Remaileh M, Ber- author(s) or other rightsholder(s); author self-archiving of the accepted
tozzi CR, Dixon SJ (2022) Ferroptosis regulation by the NGLY1/ manuscript version of this article is solely governed by the terms of
NFE2L1 pathway. Proc Natl Acad Sci U S A 119:e2118646119. such publishing agreement and applicable law.
https://​doi.​org/​10.​1073/​pnas.​21186​46119

13

You might also like