Download as pdf or txt
Download as pdf or txt
You are on page 1of 25

Cell Biol Toxicol (2023) 39:827–851

https://doi.org/10.1007/s10565-022-09778-2

REVIEW

Ferroptosis: From regulation of lipid peroxidation


to the treatment of diseases
Yonghui Lv · Meiying Wu · Zhe Wang ·
Junqing Wang

Received: 28 June 2022 / Accepted: 11 November 2022 / Published online: 2 December 2022
© The Author(s), under exclusive licence to Springer Nature B.V. 2022

Abstract Ferroptosis is a regulated cell death Interestingly, researchers found that drug-resistant or
mainly manifested by iron-dependent lipid peroxide highly aggressive cancer cells are more prone to fer-
accumulation. The leading cause of ferroptosis is the roptosis. Therefore, ferroptosis may be a potential
imbalance of intracellular oxidative systems (e.g., strategy to eliminate cancer cells. In addition, links
LOXs, POR, ROS) and antioxidant systems (e.g., between ferroptosis and other diseases, such as neu-
GSH/GPx4, CoQ10/FSP1, ­BH4/GCH1), which is reg- rological disorders and ischemia–reperfusion injury,
ulated by a complex network. In the past decade, this have also been found. Understanding these diseases
metabolic network has been continuously refined, and from the perspective of ferroptosis may provide new
the links with various pathophysiological processes insights into clinical treatment. Herein, the metabolic
have been gradually established. Apoptosis has been processes in ferroptosis are reviewed, and the poten-
regarded as the only form of regulated cell death for tial mechanisms and targets of ferroptosis in different
a long time, and the application of chemotherapeutic diseases are summarized.
drugs to induce apoptosis of cancer cells is the main-
stream method. However, studies have reported that Keywords Ferroptosis · Iron metabolism · Lipid
cancer cells’ key features are resistance to apoptosis metabolism · Redox regulation · Diseases
and chemotherapeutics. For high proliferation, can-
cer cells often have very active lipid metabolism and Abbreviations
iron metabolism, which pave the way for ferroptosis. GPx4 Glutathione peroxidase 4
ACSL4 Acyl-CoA synthetase long chain
family member 4
Y. Lv · M. Wu (*) · J. Wang (*) LPCAT3 Lysophosphatidylcholine acyltrans-
School of Pharmaceutical Sciences, Shenzhen Campus ferase 3
of Sun Yat-Sen University, Shenzhen 518107, China
e-mail: wumy53@mail.sysu.edu.cn LOXs Lipoxygenases
GSH Glutathione
J. Wang
e-mail: wangjunqing@mail.sysu.edu.cn PUFA Polyunsaturated fatty acid
MUFA Monounsaturated fatty acid
Y. Lv
e-mail: lvyh9@mail2.sysu.edu.cn PUFA-PLs Polyunsaturated phospholipids
AA Arachidonic acid
Z. Wang (*) AdA Adrenoyl acid
Department of Pathology, The Eighth Affiliated Hospital, NADPH Nicotinamide adenine dinucleotide
Sun Yat-Sen University, Shenzhen 518033, China
e-mail: wangzh379@mail.sysu.edu.cn phosphate

Vol.: (0123456789)
13
828 Cell Biol Toxicol (2023) 39:827–851

HMGB1 High mobility group box 1 Nomenclature Committee on Cell Death, accidental
VDAC2/3 Voltage dependent anion channel-3/2 cell death (ACD) and regulated cell death (RCD) are
NOX Nitrogen oxides two modes of cell death (Kroemer et al. 2005). ACD
TFR1 Transferrin receptor 1 is considered a biologically uncontrolled death result-
LIP Labile iron pool ing from non-physiological stress. In contrast, RCD
CARS Cysteinyl tRNA synthetase is a gene-regulated cell suicide process that occurs in
SLC7A11 Solute carrier family 7 member 11 multicellular organisms to meet development needs
HSPB1 Heat shock protein family B (small) and maintain homeostasis. RCD was first observed
member 1 during metamorphic development in amphibians
Nrf2 Nuclear factor erythroid 2-related (Vogt and Vogt 1842). However, RCD was not widely
factor 2 studied until the term “apoptosis” was proposed in
ROS Reactive oxygen species 1972 (Kerr et al. 1972). Apoptosis has long been con-
NCOA4 Nuclear receptor coactivator 4 sidered the only form of RCD in mammalian cells.
DMT1 Divalent metal transporter 1 However, this view is continually challenged by the
FPN1 Ferroportin 1 discovery of the non-apoptotic form of RCD (e.g.,
OA Oleic acid pyroptosis, autophagy, necroptosis, ferroptosis). Fer-
PA Palmitic acid roptosis has attracted much attention since Brent
LA Linoleic acid R. Stockwell first proposed it in 2012 (Dixon et al.
ALOXs Arachidonic acid lipoxygenases 2012). As the name suggests, iron plays an important
G6PDH Glucose 6-phosphate dehydrogenase role in ferroptosis. Unlike caspase-mediated apop-
GDH Glutamate dehydrogenase tosis, ferroptosis is associated with lipid peroxides
PGDH 6-Phosphogluconate dehydrogenase which lead to lethal disruption of the plasma mem-
SCP2 Sterol carrier protein 2 brane. This process is highly dependent on intracellu-
DHA Docosahexaenoic acid lar iron levels (Cao and Dixon 2016). How to identify
POR Cytochrome P450 reductase ferroptosis? The following four criteria can generally
CoQ10 Coenzyme Q10 be used to determine whether cells undergo ferrop-
AMPK AMP-activated protein kinase tosis (Stockwell et al. 2017). (1) Cell death caused
ACC​ Acetyl-CoA carboxylase by cystine uptake inhibition should be eliminated by
LKB1 Serine/threonine-protein kinase 1 β-mercaptoethanol. (2) Cell death caused by reactive
GLS Glutaminase oxygen species should be prevented by iron chela-
α-KG α-Ketoglutaric acid tors (e.g., deferoxamine) and antioxidants (e.g., BHT,
OTUB1 Ubiquitin aldehyde binding 1 vitamin E). (3) The morphological changes of cells
GSSG Oxidized glutathione should be mainly mitochondria shrinkage and dam-
AIFM2/FSP1 Apoptosis inducing factor mitochon- age and no other characteristic changes. (4) The cell
dria associated 2 death process can be interrupted by ferroptosis inhibi-
ESCRT-III Endosomal sorting complex tors (e.g., liproxstatin-1, ferrostatin-1) but not by
BH4 Tetrahydrobiopterin other RCD inhibitors.
GCH1 GTP-cyclohydrolase 1 Apoptosis has long been considered the only form
EMT Epithelial-mesenchymal transition of RCD (Sauler et al. 2019). The application of chem-
DAMPs Damage-associated molecular pattern otherapeutic drugs to induce apoptosis is one of the
IRI Ischemia-reperfusion injury important methods for cancer treatment. However,
previous studies have reported that cancer cells have
intrinsic or acquired resistance to apoptosis, which
Introduction is one of the key features of cancer (Mohammad
et al. 2015). Interestingly, drug-resistant or highly
Cell death plays an essential role in various life pro- aggressive cancer cells were highly susceptible to
cesses, such as proliferation, growth, development, ferroptosis (Viswanathan et al. 2017). Furthermore,
and immunity (Vaux and Korsmeyer 1999). Accord- hyperproliferative cancer cells tend to be rich in iron
ing to the international standard proposed by the (Weinberg 1996), which causes cancer cells to be

Vol:. (1234567890)
13
Cell Biol Toxicol (2023) 39:827–851 829

sensitive to ferroptosis. Therefore, ferroptosis may have different morphological and biochemical charac-
become a new strategy to eliminate cancer cells (Xu teristics, as detailed in Table 1.
et al. 2021a). Recent evidence suggests that ferrop-
tosis is also closely related to neurological disorders,
ischemia–reperfusion injury, immunity, and infection Ferroptosis: redox homeostasis pushing
(Qiu et al. 2020). The growing research on ferroptosis boundaries
reveals more potential therapeutic targets for clinical
applications. Here, we review the metabolic regula- Ferroptosis is a complex oxidative and antioxidant
tion of ferroptosis and highlight the potential mecha- regulatory network (Fig. 1) (Jiang et al. 2021; Stock-
nisms and targets of ferroptosis in various diseases. well 2022). The introduction of PUFAs (mainly AA
and AdA) into the plasma membrane is mediated by
ACSL4/LPCAT3 (Doll et al. 2017). These polyun-
Ferroptosis: a unique form of regulated cell death saturated phospholipids (PUFA-PLs) are important
substrates for generating lipid peroxides. Normally,
Morphological and biochemical features of ferrop- lipid peroxides are maintained at physiological levels
tosis are unique in RCD. Morphologically, the cell by intracellular antioxidant systems (e.g., GSH/GPx4,
membrane does not rupture and blister. The mito- CoQ10/FSP1, ­BH4/GCH1). However, these enzymes
chondrial morphology changed significantly, mani- are inhibited or downregulated in ferroptosis (Tang
fested as shrinkage, loss or reduction of mitochondrial and Kroemer 2020). Ferroptosis is iron-dependent.
cristae, increased membrane density, and rupture of In cells, iron is mainly stored in the form of ferritin.
the outer mitochondrial membrane. In addition, there Ferritinophagy increases the level of the intracellular
was no change in nuclear size and no chromatin con- labile iron pool (LIP), which promotes the ROS gen-
densation. Biochemically, a large amount of iron eration through Fenton and Haber–Weiss reactions
and ROS are accumulated, and the MAPK pathway (Kruszewski 2003). In addition, the enzymes that cat-
is activated. Glutamate/cystine antiporter activity is alyze lipid peroxidation are mostly heme or non-heme
inhibited, resulting in the depletion of the intracel- iron enzymes.
lular reducing agent GSH. In addition, the release
of arachidonic acid mediators is increased. Immuno-
logically, DAMPs are released to promote inflamma- Lipid peroxidation
tory responses. Multiple ferroptosis regulators were
upregulated, including VDAC2/3, Ras, NOX, TFR1, Enzymatic oxidation
p53, and CARS. In contrast, anti-ferroptosis regula-
tors such as GPx4, SLC7A11, HSPB1, and Nrf2 were The ALOX family consists of six members, ALOX5,
downregulated (Xie et al. 2016). However, the mor- ALOX12, ALOX12B, ALOX5, ALOX15B, and
phology of apoptotic cells has markedly different fea- ALOXE3, which are a class of non-heme iron dioxy-
tures, including retraction of pseudopodia, membrane genases (Yang et al. 2016). ALOX5 oxidizes AA to
blebbing, chromatin condensation, cell, nuclear vol- 5-hydroxyperoxyeicosatetraenoic acid (5-HPETE).
ume reduction, and nuclear fragmentation. In addi- ALOX (12, 12B, 15, 15B) can catalyze AA to
tion, apoptotic cells have unique biochemical charac- 15-HPETE and 12-HPETE with incomplete regi-
teristics, such as activation of caspases (Cohen 1997), oselectivity (Fig. 2) (Gaschler and Stockwell 2017).
fragmentation of oligonucleosomal DNA, and phos- ALOX15 regulates RSL3-induced ferroptosis upon
phatidylserine eversion. Immunologically, apoptotic binding to phosphatidylethanolamine-binding pro-
cells often trigger anti-inflammatory responses and tein 1 (PEBP1). Zhou et al. found that inhibition of
immune silencing, while in some cases, they trigger ALOX12 alleviated GPx4 depletion-induced ferrop-
immune responses by releasing DAMPs. In apoptotic tosis in HT22 cells (Zhou et al. 2020). Probst and
cells, many key regulators are up-regulated, including colleagues showed that RLS3-induced ferroptosis
p53, Bax, Bak, etc. Simultaneously, it downregulated in acute lymphoblastic leukemia cells was inhibited
multiple anti-apoptotic family proteins (Hengartner by nordihydroguaiaretic acid, a selective inhibitor
2000). Pyroptosis, autophagy, and other RCD also of ALOX12/15 (Probst et al. 2017). Furthermore,

Vol.: (0123456789)
13
Table 1  The characteristics of common RCD (Chen et al. 2021a; Li et al. 2020b; Nguyen et al. 2020; Xia et al. 2019; Xie et al. 2016)
830

Type Ferroptosis Apoptosis Autophagy Pyroptosis Necroptosis

13
Vol:. (1234567890)
Identification Regulated cell death is Apoptosis is a regulated Autophagy is a process that Pyroptosis mediated by Traumatic cell death due to
caused by the accumula- cell death mediated by engulfs self-cytoplasmic the gasdermin family physicochemical factors
tion of iron-dependent various proteases. It is proteins or organelles of proteins is a form of
lipid peroxides, which is characterized by the to form vesicles that regulated cell death that
morphologically and bio- cleavage of specific fuse with lysosomes for releases pro-inflammatory
chemically distinct from aspartate residues of degradation factors and lyses cells
other RCD target proteins by these
proteases
Morphological character- Plasma membrane without cell membrane blistering; Autophagosome formation Loss of membrane integ- Rupture in the plasma mem-
istics rupture and blebbing; Nuclei and cell size reduc- such as macroautophagy rity; brane;
Mitochondrial shrinkage; tion; and microautophagy Chromatin condensation; Decreased cell volume;
Increased mitochondrial Chromatin condensation; Bubbling; swelling; Cytoplasm and organelle
membrane density; Nuclear fragmentation Release inflammatory swelling;
Loss or reduction of mito- factors Mitochondrial cristae rupture
chondria crista; until disappearance;
Loss of mitochondrial outer Chromatin condensation
membrane integrity;
Normal nuclear volume
and without chromatin
condensation
Biochemical characteristics Accumulation of iron and Activation of pro-apoptotic Conversion of MAP1 Activation of CASP (1, 4, Decreased ATP levels;
ROS; Bcl-2 family and caspases LC3B-I to MAP1 LC3B- 5) and GSDMD; Activation of MLKL and
Activated MAPKs; proteins; II; Inflammasome; RIP (1, 3);
Inhibition of system XC − ; Oligonucleosomal DNA Substrate (e.g., p62) degra- GSDMD cleavage; DAMPs such as HMGB1 are
Increased GSH and fragmentation; dation; GSDMD-N-mediated pore released;
NADPH oxidation; Phosphatidylserine ever- Separation of Beclin-1 from formation; Overactive PARP1;
Δψm dissipation; sion; Bcl-2/XL; Release of IL1β and IL18 Cytosolic necrosome forma-
Arachidonic acid mediators Δψm dissipation Increased autophagic flux tion
such as 11-HETE and and lysosomal activity
15-HETE release;
Cannot be modulated by
inhibitors of other RCD,
such as caspase, cathep-
sin, calpain proteases,
RIPK1, and cyclophilin
D;
Cell Biol Toxicol (2023) 39:827–851
Table 1  (continued)
Type Ferroptosis Apoptosis Autophagy Pyroptosis Necroptosis

Immune characteristics DAMPs such as HMGB1 Immune silent and anti- Frequent inhibition of Pro-inflammatory Usually releases DAMPs to
are released inflammatory; inflammasome activation; promote inflammation;
Under certain circum- Non-classical secretion of Under certain circumstances,
stances, the release of cytokines pro-inflamma- anti-inflammatory
DAMPs to elicit an tory in some cases;
immune response
Key regulators Pro-ferroptosis: VDAC2/3, Proapoptotic: CARD8, Genes: ATG (1, 3–9) etc Regulators: CASP (1, 4, 5) Regulators: AURKA, RIP
ACSL4, Ras, NCOA4, GZMB, CASP (2, 3, Regulators: LC3, Beclin and GSDMD (1, 3), Ppm1b, MLKL,
Cell Biol Toxicol (2023) 39:827–851

NOX, CARS, TFR1, p53 6–10), p53, Bcl-2 family 1, ATG proteins family, LUBAC, ESCRT-III,
Anti-ferroptosis: GPx4, Anti-apoptotic: HSPA1B, mTOR, DRAM3, AMPK, cIAPs
SLC7A11, HSPB1, Nrf2, CARD6, Bcl-XL, NOX5 TFEB, ­Na+/K+-ATPase Genes: COL4A3BP, EDD1,
FSP1, RPL8, IREB2, CS, CD40, MPG, DCC1, CA9,
ATP5G3, TTC35, ACSL3 NPEPL1, SLC25A15,
SIRT5
Inducers GSH depletion: Erastin, Intrinsic apoptosis: DNA Rapamycin, Lithium, Metformin, DPP8/9 inhibi- TNF-α, Z-VAD-FMK
IKE, DPI2, BSO, BAY damage, Cytosolic C ­ a2+ Valproate, and Carbamaz- tor, Anthocyanin, Iron,
87–2243, SAS, Sorafenib, overload, etc epine DHA, α-NETA, DOX,
Glutamate, Cyst(e)inase Extrinsic apoptosis: FASL, Cisplatin, lobaplatin,
GPx4 inactivation: RSL3, UNC5B, DCC Paclitaxel, BI2536,
DPI (7/ML162, 10/ L61H10
ML210) DPI (12, 13,
17, 18, 19) Altretamine,
FIN56, Simvastatin, With-
aferin A, Lovastatin acid,
Fluvastatin;
Iron loading: F
­ INO2, Hemo-
globin, (NH4)2Fe(SO4)2,
Hemin, Salinomycin,
Lapatinib + Siramesine,
Amino acid deple-
tion + Cornell dots, BAY
11–7085;
Others: CIL (41, 56, 69,
70, 75, 79), Artemisinin
derivatives, Lanperisone

Vol.: (0123456789)
13
831
Table 1  (continued)
832

Type Ferroptosis Apoptosis Autophagy Pyroptosis Necroptosis


Inhibitors Antioxidants: Vitamin E, Apoptosis inhibitors: ILP- VPS34 inhibitors: PIK-III, CASP inhibitors: Wedelol- RIP1 inhibitors: Nec-1

13
Vol:. (1234567890)
Trolox, U0126 2, XIAP, c-IAP (1, 2), Vps34-In1, SAR 405 actone, Ac-YVAD-CMK, MLKL inhibitors: NSA
ROS inhibitors: SRS (8–72, Bruce/Apollon, ML-IAP/ PI3K inhibitors: Methy- VX765, Z-YVAD (OMe)- RIPK3 inhibitors: GSK872,
11–92, 12–45, 13–37, LIVIN, NAIP, Survivin ladenine, Wortmannin, FMK HS-1371
16–86), Ferrostatin-1 Caspase inhibitors: Z-VAD/ LY294002 NLRP3 inhibitors:
Iron chelators: DFO, Sola- IETD/DEVD-FMK, ULK1 inhibitors: MRT MCC950, CY-09, Isoli-
mine, Ciclopirox olamine, Emricasan, Q-DEVD/ (68,921, 67,307), SBI- quiritigenin, Oridonin,
2, 2- Bipyridyl LEHD/VD-OPh, Ivachtin, 0206965 Glybenclamide
Others: β-mercaptoethanol, Ac-DEVD-CHO Beclin1 inhibitors: Spau- GSDMD cleavage: Ac-
Cycloheximide, Ebselen, tin-1 FLTD-CMK
Aminooxyacetic acid H+-ATPase inhibitors:
Bafilomycin A1, Con-
canamycin A
Lysosome inhibitors: Chlo-
roquine, Hydrochloroquin
MAPKs mitogen-activated protein kinases, HETE hydroxyeicosatetraenoic acids, CASP caspase, GSDMD gasdermin D, RIP receptor-interacting protein, MLKL mixed lineage
kinase domain-like, PARP1 poly (ADP-ribose) polymerase 1, CARS cysteinyl-tRNA synthetase, FSP1 ferroptosis suppressor protein 1, RPL8 ribosomal protein L8, IREB2 iron
response element binding protein 2, CARD8 caspase recruitment domain-containing protein 8, GZMB granzyme B, UNC5B unc-5 netrin receptor B, ATG​autophagy, LC3 micro-
tubule-associated protein light chain 3, DRAM3 damage-regulated autophagy modulator 3, TFEB transcription factor EB, mTOR mammalian target of rapamycin, cIAPs inhibitor
of apoptosis proteins, LUBAC linear ubiquitin chain assembly complex, Ppm1b protein phosphatase 1B, AURKA aurora kinase A, MPG N-methylpurine-DNA glycosylase, CA9
carbonic anhydrase 9, SIRT5 sirtuin 5, NPEPL1 aminopeptidase like 1, DPI2 diphenylene iodonium 2, DPP8 dipeptidyl peptidase 8, NAIP neuronal apoptosis inhibitor protein
Cell Biol Toxicol (2023) 39:827–851
Cell Biol Toxicol (2023) 39:827–851 833

Fig. 1  The regulatory network of ferroptosis. The conver- complex is taken up into cells by TFR1-mediated endocytosis.
sion of PUFAs to PUFA-PLs sensitizes cells to ferroptosis. Iron overload promotes ferroptosis by accelerating Fenton and
In contrast, MUFA-PLs inhibit ferroptosis by reducing the Haber–Weiss reactions. GSH/GPx4, CoQ10/FSP1, and ­BH4/
abundance of PUFA-PLs. Under energy stress, PUFA produc- GCH1 are the three major antioxidant metabolic axes. GPx4
tion is reduced due to the activation of the LKB1-AMPK-ACC inhibits lipid peroxidation in the presence of GSH. Ubiquinol
pathway. Glutamine, on the one hand, promotes ferroptosis by and ­BH4 can neutralize hydroxyl and alkoxy radicals to inhibit
increasing ROS production; on the other hand, it inhibits fer- lipid peroxide accumulation
roptosis by promoting cystine uptake. The iron-transferrin

buthionine sulfoximine-induced intracellular GSH of ER-localized oxidoreductase. Tang et al. reported


depletion was lethal in wild-type cells but not in that in clear cell renal cell carcinoma and mela-
ALOX12/15-deficient cells (Hamilton and Wedner noma cells, POR could promote the peroxidation
1985). Cytochrome p450 reductase (POR) is a class of PUFA-PLs, resulting in cancer cell death in an

Vol.: (0123456789)
13
834 Cell Biol Toxicol (2023) 39:827–851

Fig. 2  Arachidonic acid


undergoes peroxidation
under the catalysis of differ-
ent lipoxygenases to gener-
ate various peroxides

ALOXs-independent manner (Tang and Kroemer leads to the destruction of RH on the plasma mem-
2020). Yan et al. reported that in Hela cells, POR brane. (iii) Termination: While the radical chain reac-
could promote the production of hydrogen peroxide tion spreads, some ROO· will react with other ROO·
by transferring electrons from NADPH to oxygen, to produce R = O, lipid alcohol, and oxygen. Alter-
which promotes ferroptosis (Yan et al. 2021). natively, in the presence of a free radical scavenger
(·Astabilized), ROO· is converted to ROOA.
Free radical oxidation

Non-enzymatic oxidation is mainly initiated by fer- Lipid antioxidation


rous iron. Ferrous iron in LIP catalyzes the produc-
tion of hydroxyl radicals from hydrogen peroxide GSH/GPx4
through Fenton and Haber–Weiss reactions (Fig. 3a)
(Kruszewski 2003; Ou et al. 2022). Subsequently, the As one of the major intracellular antioxidant enzymes,
radical chain reaction introduces oxygen molecules GPx4 is a GSH-dependent enzyme with the ability
into the polyunsaturated acyl chains of phospholipids to inhibit the accumulation of lipid hydroperoxide
to form lipid hydroperoxides. These lipid hydroper- (ROOH) (Forcina and Dixon 2019). The inactivation
oxides are further catalyzed by ferrous iron, resulting of GPx4 leads to rapid accumulation of intracellular
in the continuous expansion of lipid peroxidation in lipid peroxides, which induces ferroptosis (Fig. 1).
the plasma membrane, ultimately triggering ferropto- Hangauer et al. reported that drug resistance of cancer
sis. Specifically, the radical chain reaction is divided cells is related to GPx4 activity (Hangauer et al. 2017).
into three stages (Fig. 3b) (Doll and Conrad 2017). (i) Cancer cells overexpressing GPx4 are resistant to
Chain initiation: PUFA-PLs (labeled RH) is attacked treatment with buthionine sulfoximine and hypericin.
by hydroxyl radical, which leads to the loss of one Induction of GPx4 expression in breast cancer cells
hydrogen atom and becomes an alkyl radical (R·). An that do not express GPx4 results in increased cancer
oxygen molecule is introduced into R· to form lipid cell drug resistance (Zhang et al. 2020). Cao and col-
peroxy radicals (ROO·). (ii) Propagation: The gener- leagues showed that Iron chelators or antioxidants
ated ROO· reacts with other RH to produce new lipid could attenuate cell death caused by GPx4 inactiva-
hydroperoxide (ROOH) and R·, which indicates that tion (Cao and Dixon 2016). This further confirms that
free radicals propagate between different phospho- GPx4 activity is critical for ferroptosis. Furthermore,
lipid molecules. Then, ROOH is catalyzed by fer- in erastin-induced ferroptosis, HSP90 degrades GPx4
rous to form alkoxy radical (RO·). RO· attacks other through a chaperone-mediated autophagy process.
phospholipids (RH) to produce more R·. The gener- Application of the HSP90 inhibitor CDDO or knock-
ated R· repeats the above process continuously, which down of the HSP90 subunit can inhibit erastin-induced
Vol:. (1234567890)
13
Cell Biol Toxicol (2023) 39:827–851 835

Fig. 3  The oxidation of phospholipids. a Oxygen is con- ides (ROOH). The ROOH either participates in Fenton and
verted into superoxide radicals (·O2−) under the catalysis of Haber–Weiss reactions to generate alkoxy radicals (RO·) or
various enzymes in the cell, such as CYP, and NOX. Hydro- is reduced to lipid alcohol (ROH) by GPx4. b Under the ini-
gen peroxide produced under the catalysis of superoxide dis- tiation of hydroxyl radicals, RH undergoes radical chain reac-
mutase is converted into hydroxyl radicals through the Fen- tions, including chain initiation, chain propagation, and chain
ton and Haber–Weiss reactions. PUFA-PLs (RH) react with termination
oxygen under the catalysis of LOXs to generate lipid perox-

ferroptosis (Chen et al. 2021e). System XC- is a CoQ10/AIFM2


sodium-independent transporter with the ability to
uptake cystine and efflux glutamate. SLC7A11, a Coenzyme Q10 is an endogenous retinoid compound
multichannel transmembrane protein, is one of the that not only participates in the mitochondrial respira-
two subunits that make up system XC- (Fig. 1). Lew- tory chain but can also neutralize various free radicals
erenz et al. have reported that SLC7A11 promotes to exert a powerful antioxidant effect (Crane 2001).
GSH biosynthesis by increasing cystine uptake (Lew- Ubiquinol is the reduced form of coenzyme Q10,
erenz et al. 2013). Upregulated GSH can increase the which can prevent lipid peroxidation. Apoptosis-
activity of GPx4 to inhibit ferroptosis. The inactiva- inducing factor mitochondria-associated 2 (AIFM2,
tion of ACSL4 can partially abolish ferroptosis caused also known as FSP1) can mediate ubiquinol produc-
by the inactivation of SLC7A11 or GPx4 (Koppula tion (Fig. 1) (Dai et al. 2020). Different subcellular
et al. 2021). Liu et al. described that OTUB1 deubiq- localizations of AIFM2 have different cellular regu-
uitinates SLC7A11 to increase SLC7A11 stability, latory roles. Membrane AIFM2 can reduce coen-
resulting in the suppression of ferroptosis (Liu et al. zyme Q10 to ubiquinol under the action of NADPH,
2019). In addition, tumor suppressor genes (e.g., p53, thereby inhibiting ferroptosis (Bersuker et al. 2019).
BAP1) and proto-oncogene (KRAS) can also induce Notably, although iFSP1 can inhibit the activity of
SLC7A11 expression to inhibit ferroptosis. However, AIFM2, treatment with iFSP1 alone does not induce
it is worth noting that not all tumors had elevated lev- ferroptosis (Zheng et al. 2021). Nawarskas et al.
els of SLC7A11 expression. For example, ARID1A- described that statin could increase the sensitivity of
deficient tumors or tumors with gain-of-function cells to ferroptosis by inhibiting the production of
mutations in p53 show reduced levels of SLC7A11 coenzyme Q10 (Fig. 1) (Nawarskas 2005). In addi-
expression (Sasaki et al. 2020). tion, Kuang et al. reported that exogenous coenzyme

Vol.: (0123456789)
13
836 Cell Biol Toxicol (2023) 39:827–851

Q10 analogs idebenone also inhibit ferroptosis FSP1 (Doll et al. 2019). In addition, NADPH can also
(Kuang et al. 2020). The ESCRT-III-dependent mem- promote cystine uptake by SLC7A11 and elevate the
brane repair mechanism mediated by CHMP5 and production of GSH (Koppula et al. 2021). Overall,
CHMP6 may be activated with the accumulation of NADPH can enhance the antioxidant capacity of cells
AIFM2 in the cell membrane, which inhibits ferrop- through multiple pathways and reduce susceptibility
tosis (Liu et al. 2021). to ferroptosis.

BH4/GCH1
The metabolic regulation of ferroptosis
Tetrahydrobiopterin ­(BH4) is an active cofactor
involved in redox processes. ­ BH4 has antioxidant Labile iron pool
properties both in vitro and in vivo. Kraft and co-
workers revealed that overexpression of GCH1 ­(BH4 The occurrence and development of ferroptosis
synthesis rate-limiting enzyme) abrogates lipid per- require the participation of iron. Although the exact
oxidation and shows strong protection against IKE- mechanism between ferroptosis and iron metabolism
induced ferroptosis independent of other ferropto- is unclear, the critical role of iron metabolism in fer-
sis-related antioxidant systems (Kraft et al. 2019). roptosis is beyond doubt (Doll and Conrad 2017).
­ H4/GCH1 path-
Soula et al. also proposed that the B For example, (1) ferroptosis can be terminated by the
way is a GPx4-independent antioxidant system. The iron chelator. (2) LIP mediates the production of ROS
accumulation of NADP and GSSG was found in by Fenton and Haber–Weiss reactions (Kruszewski
­BH4-deficient cells, while elevated levels of CoQ10 2003). (3) The enzymes related to lipid peroxidation
were found in GCH1-overexpressing cells (Soula (e.g., LOXs) are mainly heme and non-heme iron
et al. 2020). Furthermore, GCH1 has no protec- enzymes.
tive effect against apoptosis induced by apoptosis- The transport of extracellular iron into the LIP
inducing agents (Wei et al. 2020). This indicates that is mediated by various enzymes (TFR1, STEAP3,
GCH1 can act as a selective target of ferroptosis. DMT1) (Fig. 1) (Dunn et al. 2007). Ferrous iron in
LIP can convert peroxides into free radicals, the main
NADPH reactive oxygen species in cells, through the Fenton
and Haber–Weiss reactions (Dixon and Stockwell
NADPH is a major intracellular reducing agent which 2014; Kruszewski 2003). Gao et al. demonstrated that
can maintain intracellular redox homeostasis (Fig. 1). ferroptosis induced by erastin could be prevented by
NADPH can be produced through the pentose phos- gene knockout of TFR1 (Gao et al. 2015). Zhang et al.
phate pathway (PPP) (Stincone et al. 2015). The reported that miR-222 could inhibit TFRC-induced
silencing of the G6PDH and PGDH genes sensitizes ferroptosis in LX-2 cells (Zhang et al. 2022a). Intra-
HCC cells to ferroptosis-inducing agents. Yan et al. cellular iron is either used to synthesize iron-con-
documented that NADPH can regulate ferroptosis taining enzymes or stored as ferritin. Autophagic
in several ways. NADPH-cytochrome p450 reduc- degradation of ferritin (ferritinophagy) elevates the
tase (POR) and NADH-cytochrome b5 reductase intracellular LIP, sensitizing cells to ferroptosis.
(CYB5R1) can promote the production of lipid perox- NCOA4 is a key enzyme mediating the above process
ides. Occasionally, hydrogen peroxide is produced by (Mancias et al. 2014).
POR and CYB5R1, transferring electrons to ambient Solute carrier family 40 member 1 (SLC40A1/
oxygen. The hydrogen peroxide then promotes fer- FPN1) can mediate the export of non-heme iron
roptosis through Fenton and Haber–Weiss reactions across the membrane (Fig. 1) (Donovan et al. 2005).
(Yan et al. 2021). As a major reducing agent, NADPH FPN1 overexpression increases cellular resistance to
assists glutathione reductase in reducing GSSG to ferroptosis, which is a negative regulator. Hepcidin
glutathione GSH (Kaneko et al. 2002). The regen- antimicrobial peptide (HAMP), a peptide hormone
eration of ­BH4 requires the participation of NADPH secreted by the liver, can induce the internalization
(Thöny et al. 2000). NADPH also inhibits ferropto- and degradation of FPN1 (Liu et al. 2005). Ferritin
sis by reducing coenzyme Q10 to ubiquinol through is released extracellularly as exosomes mediated by

Vol:. (1234567890)
13
Cell Biol Toxicol (2023) 39:827–851 837

prominin-2, which inhibits ferroptosis (Brown et al. et al. 2019). Monounsaturated fatty acids (MUFAs)
2019). Brown et al. have shown that RSL3 can selec- are fatty acids with only one double bond. Unlike
tively induce the overexpression of prominin-2 in PUFAs, MUFAs (e.g., OA) have inhibitory effects
ferroptosis-insensitive cells but not in ferroptosis-sen- on ferroptosis (Magtanong et al. 2019). Why do dif-
sitive cells (Brown et al. 2019). This suggests that the ferent types of fatty acids have opposite effects on
knockdown of prominin-2 may promote ferroptosis. ferroptosis? Probably because PUFAs are impor-
tant substrates for unsaturated phospholipids syn-
Other iron‑related proteins thesis. The type of unsaturated phospholipids in the
cell membrane determines how sensitive the cell is
Other iron-related proteins that can regulate fer- to ferroptosis. Furthermore, vertebrates cannot syn-
roptosis include PCBP1, heat shock protein family thesize ω6 and ω3 PUFAs de novo due to the lack
B small member 1 (HSPB1), and heme oxygenase of Δ12 and Δ15 (Fig. 4). Therefore, 18:2n-6 and
(HMOX1). Philpott et al. have shown that loss of 18:3n-3 must be obtained from the diet (Henderson
PCBP1 in mouse hepatocytes increases the LIP, 1996; Tocher 2003). In addition to de novo synthe-
leading to ferroptosis (Philpott et al. 2020). A pre- sis, PUFAs or MUFAs can be taken up by fatty acid
vious study reported that HSPB1 expression was transporter (FATP), fatty acid translocase (CD36),
increased in erastin-induced ferroptosis cells (Sun and fatty acid-binding protein (FABP) (Fig. 1)
et al. 2015). Furthermore, HSPB1 can counter- (Bonen et al. 2007).
act ferroptosis by inhibiting TFR1-mediated iron
uptake (Fig. 1) (Chen et al. 2006). HMOX1 has a ACSL4/LPCAT3
dual role in ferroptosis. For one, HMOX1 can pro-
mote ferroptosis, as the HMOX1 inhibitor zinc ACSLs are responsible for introducing fatty acids
protoporphyrin IX prevents erastin-induced fer- into the plasma membrane, which have five members
roptosis. Conversely, the HMOX1 inducer heme in humans (ACSL1, ACSL3, ACSL4, ACSL5, and
promotes erastin-induced ferroptosis. In addition, ACSL6). They have different substrate preferences
CO generated by HMOX1 decomposing heme can and tissue specificities in fatty acid metabolism (Quan
also promote erastin-induced ferroptosis (Kwon et al. 2021). ACSL3 is biased to catalyze the produc-
et al. 2015). Notably, heme/hemoglobin-induced tion of OA-CoA, while ACSL4 is biased to catalyze
ferroptosis is ERK1/2-independent in hemorrhagic the formation of AA-CoA and AdA-CoA (Kuwata
stroke, which differs from erastin-induced ERK1/2- and Hara 2019). Particularly, ACSL4 is selectively
dependent ferroptosis (Zille et al. 2022). Second, expressed in different cancer cells. The expression
HMOX1 can inhibit ferroptosis under certain con- level of ACSL4 was significantly higher in ferropto-
ditions. For example, the knockdown of HMOX1 sis-sensitive cells than in ferroptosis-resistant cells
increased erastin-induced ferroptosis in renal proxi- (Doll et al. 2017). The overexpression of ACSL4 was
mal tubular cells (Adedoyin et al. 2018). observed in ferroptosis of hepatocellular carcinoma
induced by sorafenib (Feng et al. 2021). ACSL3 is
PUFAs/MUFAs often associated with ferroptosis inhibition. ACSL3
tends to catalyze the production of MUFA-CoA (e.g.,
The different types of fatty acids play different OA-CoA), which remodels the plasma membrane,
regulatory roles in ferroptosis (Das 2019). Polyun- reducing phospholipid peroxidation (Doll et al. 2017).
saturated fatty acids (PUFAs) refer to straight-chain Exogenous MUFAs can inhibit erastin or RSL3-
fatty acids with two or more double bonds. Treat- induced ferroptosis, which is ineffective in ACSL3-
ment with exogenous PUFAs increases the sensitiv- deficient cells (Magtanong et al. 2019). LPCAT is
ity of cells to ferroptosis (Magtanong et al. 2019). responsible for incorporating fatty acid acyl chains
The activation of lipophagy can increase endogenous into phosphatidylcholine. As a downstream protease
PUFAs and promote ferroptosis (Chen et al. 2021e). of ACSL4, LPCAT3 tends to incorporate AA-CoA
However, deuterated-PUFAs, in which the hydrogen and AdA-CoA into the plasma membrane to pro-
atom at the bisallyl position is replaced by deuterium mote ferroptosis (Hashidate-Yoshida et al. 2015). In
atom make cells resistant to ferroptosis (Magtanong addition, SCP2 promotes ferroptosis by transporting

Vol.: (0123456789)
13
838 Cell Biol Toxicol (2023) 39:827–851

Fig. 4  The biosynthesis


of long-chain PUFAs. Δ4,
Δ5, Δ6, Δ8, Δ9, Δ12, Δ15,
fatty acyl desaturases; FAS,
fatty acid synthase; ACC,
acetyl-CoA carboxylase;
ELOVL, very long-chain
fatty acid elongase; β, per-
oxisomal β-oxidation

phospholipids (including lipid peroxides) between (Zhao et al. 2020). It is worth mentioning that
different organelle membranes (Kriska et al. 2010). mitophagy can maintain the number of mitochon-
dria. SLC25A37 and SLC25A28 are degraded
Mitochondrial respiratory chain and activated through the PINK1-PRKN pathway,
resulting in mitochondrial iron accumulation. How-
Glucose is metabolized in different ways in tumor ever, the effect of mitophagy on ferroptosis remains
and normal cells. Unlike most normal cells, tumor mysterious (Chen et al. 2021e).
cells tend to ferment glucose to lactate, even in a
condition of oxygen sufficient to support oxida- AMPK
tive phosphorylation. This way of metabolism in
tumors is called aerobic glycolysis (WARBURG Under energy stress, tumor cells activate the AMPK
1956). Compared with oxidative phosphorylation, pathway to increase ATP production. However,
aerobic glycolysis is far less efficient in generat- AMPK can also phosphorylate and inhibit ACC,
ing ATP. To meet ATP demand, tumor cells absorb the rate-limiting enzyme for de novo fatty acid syn-
large amounts of glucose and undergo high rates thesis (Fig. 1) (Park et al. 2002). The inhibition of
of aerobic glycolysis. Simultaneously, a lot of ACC in ferroptosis-sensitive tumor cells results in
NADPH is produced. NADPH possesses the ability a significant reduction in the abundance of PUFAs,
to maintain cellular redox homeostasis and inhibit which reduces ferroptosis sensitivity (Lee et al.
ferroptosis (see above). The normal progress of 2020). In addition, AMPK can supplement NADPH
the mitochondrial respiratory chain produces a supply by enhancing fatty acid oxidative metabo-
lot of ROS. However, the abnormal accumulation lism, further reducing ferroptosis sensitivity (Jeon
of ROS can cause cellular damage (Poljsak et al. et al. 2012).
2013). In addition, ferroptosis inducer erastin can
inhibit the binding of tubulin and mitochondrial Glutamine
voltage-dependent anion channels (VDACs), keep-
ing VDACs open and enhancing mitochondrial res- The non-essential amino acid glutamine is mainly
piration, which in turn increases ROS production absorbed through SLC1A5 and SLC38A1 (Fig. 1).

Vol:. (1234567890)
13
Cell Biol Toxicol (2023) 39:827–851 839

Glutamine is converted to α-KG by GLS and GDH fibronectin, thereby increasing invasiveness and drug
(Xiao et al. 2016). α-KG accelerates the generation resistance. By targeting EMT-MET and CSCs, breast
of ROS through the tricarboxylic acid cycle and cancer cells have the potential to induce ferroptosis.
oxidative phosphorylation, which promotes ferrop- Ma et al. showed that cancer cell death induced by the
tosis. In addition, intracellular glutamate can also co-administration of siramesine and lapatinib could
promote the uptake of extracellular cystine through be inhibited by vitamin E (Ma et al. 2016). Ironomy-
system XC-, which reduces ferroptosis sensitivity cin (AM5) induces ferritinophagy in CSCs, leading to
(Lewerenz et al. 2006). iron overload and induction of ferroptosis (Mai et al.
2017). The diterpenoid natural product pleuromutilin
promotes ferroptosis in breast cancer cells by inhibit-
The ferroptosis in diseases ing thioredoxin (Llabani et al. 2019).

Generally, cancer cells have more active iron metab- Lung cancer
olism and higher levels of reactive oxygen species
than normal cells (Weinberg 1996). Many cancer- Lung cancer originating from the bronchial mucosa
related genes (e.g., p53, BAP1, and KRAS) are or lung glands is one of the malignant tumors that
also associated with ferroptosis. The activation of pose the greatest threat to human health (Barta et al.
the cancer canonical signaling pathway RAS-RAF- 2019). Currently, cisplatin combination therapy is
MEK-ERK is required for erastin-induced ferropto- commonly used to treat lung cancer. However, it has
sis (Dolma et al. 2003). Cancer cells that are highly been reported that lung cancers are resistant to cispl-
aggressive and resistant to conventional chemother- atin in some cases (Ashrafizadeh et al. 2021). There-
apeutics are more prone to ferroptosis (Viswanathan fore, new therapeutic strategies need to be explored
et al. 2017). Studies have reported that some neu- urgently. Zhang et al. pointed out that drug-resistant
rological disorders are associated with intracellular lung cancer cells tend to have higher expression lev-
iron accumulation, GPx4 inactivation, GSH deple- els of GPx4. The combination of GPx4 inhibitor and
tion, and abnormal lipoxygenase activity (Weiland cisplatin can not only induce non-small cell lung can-
et al. 2019). In addition, the link between ferroptosis cer (NSCLC) cell death but also effectively inhibit
and ischemia–reperfusion injury, infection, etc. has tumor growth in xenograft animal models. Mecha-
also been reported. This suggests that ferroptosis nistically, GPx4 inhibitors and cisplatin may overload
therapy may be a potential treatment for human dis- cancer cells with iron and trigger ferroptosis through
ease. The key mechanisms and targets of ferroptosis ferritinophagy (Zhang et al. 2020). In addition, cispl-
in human disease are summarized in Table 2. Below, atin-based chemotherapeutics can activate the Nrf2/
several common diseases linked to ferroptosis are xCT pathway in NSCLC cells, leading to drug resist-
reviewed. ance. When cisplatin was combined with erastin or
sorafenib to treat NSCLC, the upregulated Nrf2/xCT
pathway was inhibited, reducing cell resistance to
Cancer cisplatin-based chemotherapy (Li et al. 2020a). Over-
all, co-administration of ferroptosis inducers may be a
Breast cancer potential strategy for treating cisplatin-resistant lung
cancer.
Breast cancer is notorious for being difficult to treat
and metastasizes frequently. Breast cancer metas- Colorectal cancer
tasis is closely related to the EMT of cancer stem
cells (CSCs) (Takebe et al. 2011). The mesenchy- It has been confirmed that TP53 plays an important
mal phenotype epithelioid transformation (MET) role in colorectal cancer cells. TP53 can inhibit dipep-
is the inverse process of EMT and is associated tidyl peptidase 4 (DPP4). Silencing the TP53 gene
with tumor cell “homing” and proliferation (Brab- promotes DPP4-dependent lipid peroxidation, which
letz 2012). Breast cancer can be transformed into leads to ferroptosis in colorectal cancer cells (Xie
CSCs by transforming growth factor-β (TGF-β) or et al. 2017). Pagliara et al. showed that 5-fluorouracil

Vol.: (0123456789)
13
Table 2  The ferroptosis in human diseases
840

Category Diseases Key mechanisms Targets Regulators Ref

Cancer Breast cancer Iron overload; TFR1, SLC40A1, CISD1/2, HO-1, Lapatinib, MOF-Fe2+, Neratinib, Lin et al. (2021)

13
Vol:. (1234567890)
Ferritinophagy; ACSL1, ACSL4, SCL7A11, GPx4, Artesunate, α-eleostearic acid, Erastin,
Increased ACSL1 and ACSL4 expression; Nrf2, HMGCR​ Sulfasalazine, Metformin, RSL3,
Decrease SLC7A11, GPx4, and HMGCR protein levels; Fluvastatin
Pancreatic cancer Lysosomal iron-dependent ROS induced; ROS, L-cyst(e)ine, PDK, HSPA5, Artesunate, Cotylenin A and PEITC, Chen et al. (2021d)
Autophagy-dependent ferroptosis induced; SLC7A11, mTOR, GPx4, POLG, Cyst(e)inase, Dichloroacetate, EGCG,
L-cysteine and GSH depleted TFAM, NUPR1 IKE, Piperlongumine, Rapamycin,
Pyruvate oxidation and fatty acid synthesis are induced; RSL3, Ruscogenin, Zalcitabine,
Inhibition of GPx4 activity; ZZW-115
Induction of STING1-dependent autophagy;
NUPR1-LCN2-mediated iron depletion is inhibited;
Lung cancer Inhibition of K
­ RASG12C, GPx4, GCLC, PRIM2-SLC7A11, NFE2L2- KRAS, GPx4, GSH, ROS, SLC7A11, Adagrasib, BSO, Cisplatin, Curcumin, Wu et al. (2021b)
SLC7A11, USP14, FAF2, and NFE2L2-HMOX1 CaM, USP14 Erastin, Ginkgetin, Orlistat, Par-
Induction of lipid peroxidation, C
­ a2+/CaM signaling, and PINK1/ acetamol, PdPT, RSL3, Sorafenib,
Parkin-dependent mitosis; Sulforaphane, Zinc
Activation of autophagy;
Up-regulation of LC3-II/LC3-I, TP53, and BECN1;
Depletion of GSH
Hepatocellular carcinoma Inhibition of system XC-, Nrf2, GPx4, NUPR1, and S1R System XC-, Nrf2, GPx4, NUPR1, S1R Sorafenib, Quiescin Sulfhydryl Oxidase, Bekric et al. (2022), Su et al. (2020)
Artesunate, Erastin, RSL3, Solasonine,
ZZW-115, Haloperidol, Heteronemin
Gastric cancer Inhibition of CD01; c-Myb/CD01 Erastin, SASP Hao et al. (2017)
Upregulation of GPx4;
Colorectal cancer Inhibition of GPx4, DPP4 GPx4, DPP4, TMEM16F, ACSL4, RSL3, Cisplatin, β-elemene, Vitamin C, Xu et al. (2021b)
Activation of TMEM16F; SLC7A11, TP53, NCOA4, ACADSB Resibufogenin, IMCA, Bromelain, MiR-
Upregulation of ROS, DR5, and ACSL4; NAs, Erastin, Artesunate, Sorafenib,
Downregulating SLC7A11, TP53, and GSH; FeOOH nanospindles
Scavenging endogenous hydrogen sulfide (H2S);
Altered iron homeostasis
Clear cell renal cell carcinoma Inhibition of GSH, system XC − , NCOA4, EMP1-NOX4, and GPx4; VHL/HIF-2α, HILPDA, System XC-, Buthionine sulfoximine, Sorafenib, Artesu- Cosialls et al. (2021), Wang et al. (2021),
Upregulation of FTH, FTMT, and ROS levels; NCOA4, GPx4, NOX4, FTH, FTMT, nate, FINO2 Zhao et al. (2021a)
Enhancing DPP4 activity DPP4
Adrenocortical carcinoma Upregulation of GPx4 and ACSL4 Unknown Unknown Weigand et al. (2020)
Ovarian cancer Iron overload; TFR1, TAZ/NOX, GPx4 Erastin, Artesunate, Altretamine Bebber et al. (2020), Lin et al. (2020), Zuo
Reduce the depletion of NADPH; et al. (2020)
Inhibition of GPx4
Melanoma Inhibition of SLC7A11, GPx4, and NEDD4 SLC7A11, GPx4, GCL, NEDD4 Buthionine sulfoximine Chen et al. (2021b), Tsoi et al. (2018), Yang
et al. (2020)
Head and neck cancer Inhibition of CISD2, SLC7A11, LKB1, and antioxidant proteins; CISD2, SLC7A11, LKB1, Nrf2 Pioglitazone, Sulfasalazine, MiR-100-3p Figueroa-González et al. (2020), Kim et al.
(2018), Yu et al. (2019)
Brain cancer Downregulation of GSH; VDACs, system XC-, Iron, Cystine, Erastin, Sulfasalazine, Sorafenib, t-BHP, Qiu et al. (2020), Weiland et al. (2019)
Inhibition of GPx4; KEAP1, GPx4 Withaferin A, β-mercaptoethanol,
Iron overload; Zinc, CS
Upregulation of cystine uptake
Overactivation of heme oxygenase-1;
Increased ferritinophagy, depletion of GSSG, GSH, and inactivation
of system XC-
Blood cancer Acute lymphoblastic leukemia Promoting the production of ROS LOXs RSL3 Zhang et al. (2022b)
Acute myeloid leukemia Iron overload; Iron, System XC-, GPx4, GSH, ROS, APR-246, ATPR, TYP, DHA, CircK- Zhao et al. (2021b)
Inhibition of SLC7A11, GPx4, and ALDH3A2Upregulation of ROS TFR1, P53, HMGB1, ALDH3A2, DM4C
and P53; Hsa-let-7b-5p
Downregulation of TFR1 and HMGB1
Cell Biol Toxicol (2023) 39:827–851
Table 2  (continued)
Category Diseases Key mechanisms Targets Regulators Ref

Nervous system disease Parkinson’s Disease Iron overload; Iron, System XC-, GPx4, GSH, Lipid Sulfasalazine, Sorafenib, RSL3, FINO2, Weiland et al. (2019)
Ferritinophagy; peroxidation, Protein synthesis ART, t- BHP, DPI2, BSO, DFO, Fer-1,
Upregulation of DMTI expression and ROS; Vitamin E, Cp
Downregulation of SLC7A11, GSH, and CoQ10;
Inhibition of GPx4;

Alzheimer’s disease Depletes GSH System XC-, GPx4, Lipid peroxidation, Sulfasalazine, RSL3, FINO2, ART, BSO, Weiland et al. (2019)
Inhibition of GPx4; GSH, Protein synthesis Fer-1, Vitamin E, Cp, RPL13, CS
Upregulation of ROS;
Inhibition of ROS;

Huntington’s disease Depletes GSH; System XC-, GPx4, Lipid peroxidation, Sulfasalazine, RSL3, FINO2, ART, BSO, Weiland et al. (2019)
Directly inhibits GPx4, accumulation of lipid hydroperoxides GSH, LOXs Fer-1, Vitamin E, Zileuton
Cell Biol Toxicol (2023) 39:827–851

FINO2 inhibits GPx4 function and oxidizes iron;


ART interacts with lysosomal iron and generates ROS;
Inhibition of ROS;
Blocks LOX-induced lipid peroxidation;

Stroke Prevents cystine import; System XC-, GPx4, ROS, Iron, Seleno- Erastin, RSL3, Ferrostatins, Liproxstatins, Liu et al. (2022)
Inhibition of GPx4 and LOXs proteins Trolox, N-Acetylcysteine, Deferoxam-
Accumulation of ROS; ine, Ceruloplasmin, Selenium
Blocks lipid peroxidation;
Neutralizes toxic lipids;
Depletes iron;
Increases abundance of selenoproteins;

Traumatic brain injury Iron overload; Cyt C/CL, GSH, LOXs, GPx4, HIF-PHD Fer-1, Baicalein, N-acetylcysteine, JP4- Anthonymuthu et al. (2018), Karuppa-
Accumulation of ROS; 039, Adaptaquin gounder et al. (2016), Yao et al. (2021)
Upregulation of transferrin, LOXs, and ACSL4;
Inhibition of GPx4;
Ischemia–reperfusion Myocardial ischemia–reperfu- Accumulation of ROS; ER stress, SIRT1-p53/SLC7A11, p53/ Liproxstatin-1, Deferoxamine, USP22, Yang and Lin (2022)
injury sion injury Ferritinophagy; TfR1, AMPK, NOX2, ACSL4, GPx4, USP7, Lip-1, ELAVL1, Etomidate,
Downregulation of GPx4; VDAC1, GSH, Iron, MDA, Nrf2, Baicalin, Cyanidian-3-glucoside
Activation of the p53/TFR1 pathway, NOX2-related PCD; NCOA4, LC3II/LC3I, FTH1
Upregulation of VDAC1 levels;
Acute kidney injury Downregulation of GPx4 and GSH GPx4, SLC7A11, GSH, HO-1 Ferrostatin 1 or 16–86, PANX1 deletion, Li et al. (2021), Wu et al. (2021a)
Upregulation of HO-1, MDA, 4-HNE, and ROS MIF, Pachymic acid, XJB-5–131, Irisin
Immunity Unknown System XC-, GSH, HMGCR, Nrf2, ROS, Sulfasalazine, Glutamate, Sorafenib, Tang et al. (2020)
Ferrous Statins, Trigonelline, Artesunate, Iron,
Doxorubicin
Infection Unknown ALOXs Baicalein, Ferrostatin-1 Chen et al. (2021c)
Inflammation Inhibition of GPx4; ER stress, GPx4 ferrostatin-1, GSK2606414, Curculigoside, Chen et al. (2021c)
Upregulation of ROS; Liproxstatin-1, DFP, ROSI
Emancipation of HMGB1

HO-1 heme oxygenase-1, PDK pyruvate dehydrogenase kinases, POLG DNA polymerase gamma, TFAM mitochondrial transcription factor A, NUPR1 nuclear protein 1, CaM
calmodulin, S1R sigma-1 receptor, TMEM16F transmembrane protein 16F, HILPDA hypoxia-inducible lipid droplet-associated protein, FTMT mitochondrial ferritin, VDACs
voltage-dependent anion channels, KEAP1 kelch-like ECH-associated protein 1, ALDH3A2 aldehyde dehydrogenase 3 family member A2, MDA malondialdehyde, HMGCR​
hydroxymethylglutaryl-coenzyme A reductase, MOF-Fe2+ ­Fe2+-based metal–organic framework, RSL3 RAS-selective lethal, PEITC phenethyl isothiocyanate, EGCG​ epigallo-
catechin gallate, IKE imidazole ketone erastin, BSO buthioninesulfoximine, PdPT palladium pyrithione complex, SASP sulfasalazine, t-BHP tert-butyl hydroperoxide, APR-246
eprenetapopt, ATPR 4-amino-2-trifluoromethyl-phenyl retinate, TYP typhaneoside, ART​ artesunate, DPI diphenylene iodonium, DFO deferoxamine, Cp ceruloplasmin, CS citrate
synthase, Fer-1 ferrostatin-1, USP ubiquitin-specific protease, Lip-1 liproxstatin-1, ELAVL1 embryonic lethal-abnormal vision like protein 1, PANX1 pannexin1, MIF macrophage
migration inhibitory factor, DFP deferiprone, ROSI rosiglitazone, FTH1 ferritin heavy chain 1, GCL glutamate cysteine ligase, HIF-PHD hypoxia-inducible factor prolyl hydroxy-

Vol.: (0123456789)
13
lase domain
841
842 Cell Biol Toxicol (2023) 39:827–851

could induce the activation of ribosomal protein L3 inhibitors can enhance the dependence on oxidative
(RPL3) in TP53-deficient colorectal cancer cells by phosphorylation. This metabolic switch promotes fer-
inhibiting the expression of cystathionine-β-synthase, roptosis in melanoma cells (Haq et al. 2013; Schöckel
which may affect the level of cysteine in cancer cells et al. 2015). The application of exogenous PUFAs
(Pagliara et al. 2016). In addition, it has been reported can increase the sensitivity of melanoma cell lines
that RPL3 reduces the expression of system XC- and to ferroptosis. The expression level of SLC7A11 was
induces ferroptosis in TP53 mutant lung cancer cells decreased in BRAFV600E melanoma cells during
(Russo et al. 2017). The co-administration of albizi- vemurafenib or trametinib treatment (Osrodek et al.
abioside A and cisplatin can promote ferroptosis by 2019), and SLC7A11 is considered a marker of fer-
inactivating GPx4 in colon cancer cells (Wei et al. roptosis-susceptible melanoma cells. In addition, the
2018). Bromelain can upregulate the expression of sensitivity of melanoma cells to ferroptosis is also
ACSL4 in KRAS-mutated colorectal cancer cells to related to GPx4. Knockout of the GPx4 gene was
promote ferroptosis (Park et al. 2018). Therefore, tar- lethal in drug-resistant A375 melanoma cells but did
geting TP53 or ACSL4 to induce ferroptosis in colo- not affect non-resistant A375 melanoma cells. More
rectal cancer has potential clinical applications. importantly, the lethality of GPx4 knockout in drug-
resistant A375 melanoma cells could be terminated
Clear cell renal cell carcinoma with the ferroptosis inhibitor ferrostatin-1 (Hangauer
et al. 2017). In addition, the Nrf2 gene also plays an
Clear cell renal cell carcinoma (CCRCC) is a com- important role in melanoma. Upregulation of Nrf2
mon renal cell carcinoma closely associated with induces the expression of CHAC1 and AKR in mela-
mutations in the von Hippel Lindau (VHL) gene. noma cells, thereby degrading lipid peroxides and
CCRCC is highly sensitive to GSH depletion-induced inhibiting ferroptosis (Gagliardi et al. 2019).
ferroptosis due to mutations in the VHL gene. In
addition, mutations in the VHL gene lead to persis-
tent stabilization of hypoxia-inducible factor (HIF). Nervous system diseases
Zou et al. pointed out that HIF2α specifically enriches
PUFAs in CCRCC, thereby increasing susceptibility Parkinson’s disease (PD) is characterized by neuronal
to ferroptosis (Zou et al. 2018). The use of PT2385 death in the substantia nigra pars compacta (SNpc),
(HIF inhibitor) can slightly improve the survival rate which causes motor dysfunction (Phani et al. 2012).
of CCRCC patients, but prolonged use can lead to Bharath et al. showed that iron accumulation, GSH
acquired resistance (Courtney et al. 2020). In addi- depletion, and elevated ROS levels are common in
tion to VHL genes, p53 and BAP1 are also frequently SNpc neurons (Bharath et al. 2002). Guiney et al.
deleted or mutated in CCRCC. The inactivation of pointed out that ferroptosis is the major cell death
BAP1 leads to the upregulation of SLC7A11, which pathway for dopaminergic neurons (Guiney et al.
inhibits ferroptosis (Han et al. 2021). Furthermore, 2017). Zeng et al. indicated that motor function was
CCRCC is highly sensitive to intracellular glutamate improved in the PD animal model with the treatment
and cystine levels. CCRCC neutralizes intracellular of iron chelators (Zeng et al. 2021). Astrocytes have
lipid peroxides mainly through the GSH/GPx4 path- a strong iron storage capacity and the ability to pre-
way. It has been found that ferroptosis in CCRCC can vent neuronal iron overload. Furthermore, astrocytes
be effectively induced by inhibiting the GSH biosyn- can provide neurons with antioxidants, such as the
thetic pathway (Miess et al. 2018). glutathione S-transferase Mu2 (Fernandez-Fernan-
dez et al. 2012). Therefore, the lack of interaction
Melanoma between neuronal cells and astrocytes may be respon-
sible for their ferroptosis.
Melanoma is one of the most serious skin cancers. A typical feature of Alzheimer’s disease (AD) is
Generally, the MAPK pathway is hyperactivated due neuronal degeneration related to learning and mem-
to alterations in the BRAF, RAS or NF1 genes, which ory (Yankner 1996). Xie and co-works have shown
promote melanoma cell survival. In melanoma cells, that lipid peroxidation and iron dysregulation are fre-
BRAF can inhibit oxidative metabolism, and BRAF quently present in neuronal cells of AD patients (Xie

Vol:. (1234567890)
13
Cell Biol Toxicol (2023) 39:827–851 843

et al. 2014). Intracellular iron levels in neurons are (Fan et al. 2021). Dabkowski et al. reported that
often inversely correlated with cognitive performance mitochondrial GPx4 overexpression in cardiomyo-
in AD patients. The main cause of ferroptosis is the cytes could alleviate cardiac ischemia–reperfu-
imbalance of iron metabolism and lipid redox homeo- sion injury (Dabkowski et al. 2008). Furthermore,
stasis. Therefore, ferroptosis provides a new perspec- DMT1 appears to have an important role in AMI.
tive for understanding the pathological mechanism of Song et al. reported that myocardial injury can
AD. Hambright et al. showed that knocking out GPx4 be alleviated in AMI mice by targeting DMT1
in hippocampal neurons caused significant cognitive expression with exosomes from human umbilical
impairment in mice, which could be ameliorated by cord blood-derived MSCs (Song et al. 2021). Fer-
ferroptosis inhibitor liproxstatin-1 (Hambright et al. rostatin-1 and iron chelators improve acute and
2017). Treatment of animal models with the anti- chronic IRI-induced heart failure (Li et al. 2021).
oxidants MitoQ and SOD2 also improved cognitive I/R-related cardiac injury can also be alleviated by
impairment. Furthermore, treatment with alpha-lipoic promoting GSH production (Ou et al. 2021). This
acid (ALA) ameliorated AD symptoms in the P301S suggests that ferroptosis may be a new strategy for
tau murine model (Zhang et al. 2018). preventing and treating myocardial disease.
Huntington’s disease (HD) is characterized by IRI is also one of the leading causes of acute renal
chorea movement, an autosomal dominant neurode- failure (Grynberg et al. 2021). Scindia et al. reported
generative disorder (Berardelli et al. 1999). Wu et al. that renal IRI is closely related to iron metabolism
showed that lipid peroxidation, iron accumulation, (Scindia et al. 2015). Erastin-induced proximal tubule
and reduced GPx4 activity were found in neurons of cell death can be prevented by ferrostatin 1 and defer-
HD patients and animal models (Wu et al. 2018). In oxamine (Linkermann 2016). GPx4 also has a vital
addition, the application of iron chelators can improve role in renal IRI. For example, knockout of GPx4
cognitive function in HD model animals (Ward et al. resulted in renal failure in model mice, which was
2015). ameliorated by ferroptosis inhibitors (Friedmann
Angeli et al. 2014). Furthermore, the inhibition of
ACSL4 activity with thiazolidinediones also partially
Ischemia–reperfusion injury prevented the death of GPx4 knockout mice (Doll
et al. 2017).
Iron overload is a major cause of myocardial I/R For neuronal I/R injury, iron overload is criti-
injury. In the early stage of I/R, iron from ferritin cal. For example, deferoxamine reduces infarct size
degradation promotes the Fenton reaction, which after cerebral reperfusion (Hanson et al. 2009). Guan
accelerates ROS production. Gao et al. reported et al. indicated that the learning and memory abili-
that the inhibition of glutaminolysis attenuated ties of I/R-injured gerbils could be enhanced by the
I/R-induced cardiac injury (Gao et al. 2015). Fang application of galangin, which inhibits ferroptosis
et al. reported that Ferristatin-1 and iron chelators by up-regulating SLC7A11 and GPx4 (Guan et al.
could attenuate I/R-induced heart failure (Fang 2021). In addition, Carvacrol can attenuate I/R dam-
et al. 2019). Chen et al. showed that the adminis- age in hippocampal neurons by increasing the expres-
tration of deferoxamine before reperfusion rapidly sion of GPx4 (Guan et al. 2019). Ferroptosis has also
ameliorated oxidative stress (Chan et al. 2012). In been considered a potential therapeutic target for
addition, intravenous infusion of deferoxamine can hepatic IRI. The ratio of aspartate aminotransferase
reduce ROS production during coronary artery to alanine aminotransferase and the percentage of the
bypass grafting and protect the myocardium from necrotic area was reduced in a liproxstatin-1-treated
I/R injury (Paraskevaidis et al. 2005). Shan et al. mouse model of liver I/R injury (Angeli et al. 2014).
pointed out that Cyanidin-3-glucoside can effec- Fer-1 and α-tocopherol reduced ROS production and
tively alleviate I/R-induced myocardial injury by down-regulated Ptgs2 in a mouse model of liver IRI
reducing oxidative stress and iron overload (Shan (Yamada et al. 2020). In other organs, liproxstatin-1
et al. 2021). A study in a rat I/R model showed that or rosiglitazone can ameliorate intestinal I/R injury
baicalin attenuated myocardial ischemia–reperfu- by regulating the expression of GPx4 and Ptgs2 (Li
sion injury by inhibiting the expression of ACSL4 et al. 2019).

Vol.: (0123456789)
13
844 Cell Biol Toxicol (2023) 39:827–851

Immunity axis and increased free iron and lipid peroxides. Iron
chelators and ferroptosis inhibitors can reduce the
It has been reported that IFN-γ produced by ­CD8+ T death of macrophages infected with M. tuberculosis
cells can inhibit the expression of system XC- (Zitvo- (Amaral et al. 2019). This suggests that ferroptosis
gel and Kroemer 2019). It is suggested that the induc- is closely related to the pathological process of some
tion of cell death by immune cells may be related infections.
to ferroptosis. Uderhardt et al. reported that IL-4
and IL-13 could promote the expression of ALOXs,
while reducing the expression level of GPx4, result- Conclusions and perspectives
ing in the production of a lot of arachidonic acid
pro-inflammatory factors (Uderhardt et al. 2012). Over the past decade, ferroptosis has received grow-
For macrophages, ferroptosis inducers can promote ing attention from researchers. As continuous efforts
the release of HMGB1, which is required for mac- are made, research progress in this field is constantly
rophage inflammation in response to ferroptotic cells updated. The understanding of ferroptosis has shifted
(Wen et al. 2019). Xu et al. pointed out that ferrop- from disruption of cellular function to a complex net-
tosis plays a dual role in immune cells. On the one work of redox regulation. However, many questions
hand, ferroptosis leads to the inactivation of ­CD4+ related to ferroptosis remain unanswered. First of all,
and ­CD8+ T cells. On the other hand, the differen- what is the point of ferroptosis? One possible hypoth-
tiation of peripheral blood mononuclear cells can be esis is that ferroptosis is a compromise between the
promoted by ferroptosis (Xu et al. 2021c). However, benefits and risks of introducing PUFAs into cell
its underlying mechanism remains unclear. membranes. In short, the introduction of PUFAs into
the plasma membrane can confer additional func-
tions on cells (e.g., altering membrane fluidity). How-
Infection ever, this simultaneously increases the sensitivity of
the plasma membrane to intracellular ROS. Under
In response to the invasion of pathogenic microor- the catalysis of oxidase and iron, PUFA-PLs in the
ganisms, host cells recognize pathogen-associated plasma membrane continuously react with ROS to
molecular patterns (PAMPs) through multiple pat- generate lipid peroxides, which trigger ferroptosis
tern recognition receptors (PRRs) (Amarante-Mendes after exceeding the cellular antioxidant capacity. If
et al. 2018). PRRs interact with PAMPs to generate this hypothesis holds, what is the threshold for lipid
a large amount of ROS, mainly from the mitochon- peroxide-induced ferroptosis? Second, why is iron
drial respiratory chain and NADPH oxidase complex irreplaceable in ferroptosis? Although copper and
(Boller and Felix 2009). For one, the generated ROS cobalt can catalyze the Fenton and Haber–Weiss reac-
are crucial for the clearance of pathogenic microor- tions, only iron-mediated reactions can cause cellular
ganisms. Second, a lot of ROS can cause oxidative ferroptosis. Why iron? The free radicals generated
stress in host cells. It has been shown that pathogenic by Fenton and Haber–Weiss reactions are not spe-
microorganisms capable of producing lipoxygenase cific for DNA, lipid, and protein damage, which is
can attack lipid components on the host cell mem- inconsistent with the essential characteristics of RCD.
brane and induce ferroptosis in host cells. For exam- Perhaps, the activation of iron-containing enzymes
ple, although Pseudomonas aeruginosa does not have such as LOXs may be a breakthrough in answering
arachidonic acid-phosphatidylethanolamine in its this question. In apoptosis, caspases and other pro-
cell membrane, it can convert arachidonic acid into apoptotic proteins are activated. In autophagy, MAP1
15-HETE on the host cell membrane (Vance et al. LC3B-I is converted to MAP1 LC3B-II, and Beclin-1
2004). Consistently, Dar et al. showed that bacteria is dissociated from Bcl-2/XL. Compared to this, what
lacking lipoxygenase were unable to induce ferrop- are the exact executioners and biomarkers of ferrop-
tosis in human bronchial epithelial cells (Dar et al. tosis? Iron-dependent lipid peroxide accumulation
2019). In addition, Mycobacterium tuberculosis- may be an intermediate event in ferroptosis rather
induced macrophage death also has ferroptosis fea- than effector molecules. Peroxidation of PUFA-PLs
tures such as inhibition of the GSH/GPx4 antioxidant is currently considered to be the terminal event in

Vol:. (1234567890)
13
Cell Biol Toxicol (2023) 39:827–851 845

ferroptosis. There are several possibilities for lipid will gradually reveal the original appearance of fer-
peroxides to induce ferroptosis: (1) Destruction of a roptosis, which will ultimately become the corner-
phospholipid bilayer (e.g., pore formation). (2) Dis- stone for targeting ferroptosis in human diseases.
ruption of plasma membrane function. (3) Destruc-
tion of important components in cells. Finally, as an Author contribution Y.L. and J.W. wrote the main manu-
script text, Z.W. prepared figures, and M.W. prepared tables.
emerging field, the current experimental methods All authors reviewed the manuscript.
for ferroptosis research are still flawed. Most of the
mechanistic studies of ferroptosis are based on fluo- Funding This work was supported by the National Natural
rescent reporter molecules that detect lipid peroxides. Science Foundation of China (82001887 to JW and 81900105
However, whether this method can truly and accu- to ZW), in part by a horizontal project of Sun Yat-sen Univer-
sity (K21-75110–007), Shenzhen Science and Technology Pro-
rately reflect the lipid peroxides produced by ferrop- gram (JCYJ20210324115003009), and the Guangdong Basic
tosis remains questioned. and Applied Basic Research Foundation (2019A1515110326).
As mentioned above, there is a close connection
between ferroptosis and the pathological process Data availability Not applicable.
of various diseases. Cancer cells have abnormally
Declarations
active iron metabolism and lipid metabolism, so it
has an innate advantage of inducing ferroptosis in
Competing interests The authors declare no competing inter-
drug-resistant and highly aggressive cancer cells. ests.
Ferroptosis in various neurodegenerative diseases is
also gradually recognized and provides a new per- Ethics approval Not applicable.
spective for further understanding of the underly-
ing pathological mechanisms. Thus, the application Consent to participate Written informed consents were col-
of ferroptosis in treating human disease has poten- lected from all participants before enrollment.
tial clinical advantages. However, current ferrop-
tosis inducers or inhibitors are still not suitable for Consent for publication All contributing authors agree to the
clinical application. Identification and validation of publication of this article.
ferroptosis biomarkers are critical for developing
applicable ferroptosis drugs. The discovery of fer- Human ethics Not applicable.
roptosis biomarkers will not only help to uncover
Conflict of interest The authors declare no conflict of inter-
the intrinsic link between ferroptosis and specific est.
diseases but also evaluate the efficacy of ferroptosis
drugs. Although the link between ferroptosis and the
pathological process of various diseases has been
established, the link to the physiological process is References
still lacking. As an ancient RCD, does ferroptosis
have a specific physiological significance? Such as Adedoyin O, Boddu R, Traylor A, Lever JM, Bolisetty S,
affecting aging and immunity. Iron accumulation has George JF, et al. Heme oxygenase-1 mitigates ferroptosis
been observed in Caenorhabditis elegans adulthood in renal proximal tubule cells. Am J Physiol Renal Phys-
iol. 2018;314(5):F702–14.
and later life. Inhibition of iron accumulation signif- Amaral EP, Costa DL, Namasivayam S, Riteau N, Kamenyeva
icantly reduces age-related cell death and prolongs O, Mittereder L, et al. A major role for ferroptosis in
the lifespan of C. elegans (Jenkins et al. 2020). For mycobacterium tuberculosis–induced cell death and tis-
immunity, γ-interferon produced by immune cells sue necrosis. J Exp Med. 2019;216(3):556–70.
Amarante-Mendes GP, Adjemian S, Branco LM, Zanetti LC,
can sensitize tumor cells to ferroptosis, indicating Weinlich R, Bortoluci KR. Pattern recognition recep-
a molecular basis for the body to use ferroptosis to tors and the host cell death molecular machinery. Front
prevent disease. Immunol. 2018:2379.
In conclusion, the research related to ferroptosis Angeli JPF, Proneth B, Hammond VJ, Tyurina YY, Tyurin
VA, ODonnell VB et al. Inactivation of the ferroptosis
is still in its early stage, and some of its mechanisms regulator gpx4 triggers acute renal failure in a thera-
remain unclear. We believe that through the continu- peutically relevant mechanism. Free Radical Bio Med.
ous exploration of the above problems, researchers 2014;(76):S77–78.

Vol.: (0123456789)
13
846 Cell Biol Toxicol (2023) 39:827–851

Anthonymuthu TS, Kenny EM, Lamade AM, Kagan VE, Bayır Chen X, Kang R, Kroemer G, Tang D. Targeting ferroptosis in
H. Oxidized phospholipid signaling in traumatic brain pancreatic cancer: a double-edged sword. Trends Cancer.
injury. Free Radical Bio Med. 2018;124:493–503. 2021d;7(10):891–901.
Ashrafizadeh M, Zarrabi A, Hushmandi K, Hashemi F, Mogh- Chen X, Yu C, Kang R, Kroemer G, Tang D. Cellular deg-
adam ER, Owrang M, et al. Lung cancer cells and their radation systems in ferroptosis. Cell Death Differ.
sensitivity/resistance to cisplatin chemotherapy: role 2021e;28(4):1135–48.
of micrornas and upstream mediators. Cell Signal. Cohen GM. Caspases: the executioners of apoptosis. Biochem
2021;78:109871. J. 1997;326(Pt 1)(Pt 1):1–16.
Barta JA, Powell CA, Wisnivesky JP. Global epidemiology of Cosialls E, El Hage R, Dos Santos L, Gong C, Mehrpour M,
lung cancer. Ann Glob Health. 2019;85(1). Hamaï A. Ferroptosis: cancer stem cells rely on iron until
Bebber CM, Müller F, Prieto Clemente L, Weber J, von “to die for” it. Cells-Basel. 2021;10(11):2981.
Karstedt S. Ferroptosis in cancer cell biology. Cancers. Courtney KD, Ma Y, de Leon AD, Christie A, Xie Z, Woolford
2020;12(1). L, et al. Hif-2 complex dissociation, target inhibition,
Bekric D, Ocker M, Mayr C, Stintzing S, Ritter M, Kiesslich and acquired resistance with pt2385, a first-in-class hif-2
T, et al. Ferroptosis in hepatocellular carcinoma: mecha- inhibitor, in patients with clear cell renal cell carcinoma.
nisms, drug targets and approaches to clinical translation. Clin Cancer Res. 2020;26(4):793–803.
Cancers. 2022;14(7):1826. Crane FL. Biochemical functions of coenzyme q10. J Am Coll
Berardelli A, Noth J, Thompson PD, Bollen EL, Currà Nutr. 2001;20(6):591–8.
A, Deuschl G, et al. Pathophysiology of chorea and Dabkowski ER, Williamson CL, Hollander JM. Mitochon-
bradykinesia in huntington’s disease. Mov Disord. dria-specific transgenic overexpression of phospholipid
1999;14(3):398–403. hydroperoxide glutathione peroxidase (gpx4) attenu-
Bersuker K, Hendricks JM, Li Z, Magtanong L, Ford ates ischemia/reperfusion-associated cardiac dysfunc-
B, Tang PH, et al. The coq oxidoreductase fsp1 tion. Free Radic Bio Med. 2008;45(6):855–65.
acts parallel to gpx4 to inhibit ferroptosis. Nature. Dai E, Zhang W, Cong D, Kang R, Wang J, Tang D. Aifm2
2019;575(7784):688–92. blocks ferroptosis independent of ubiquinol metabolism.
Bharath S, Hsu M, Kaur D, Rajagopalan S, Andersen JK. Glu- Biochem Biophys Res Commun. 2020;523(4):966–71.
tathione, iron and parkinson’s disease. Biochem Pharma- Dar HH, Tyurina YY, Mikulska-Ruminska K, Shrivastava I,
col. 2002;64(5–6):1037–48. Ting H, Tyurin VA, et al. Pseudomonas aeruginosa uti-
Boller T, Felix G. A renaissance of elicitors: perception of lizes host polyunsaturated phosphatidylethanolamines to
microbe-associated molecular patterns and danger sig- trigger theft-ferroptosis in bronchial epithelium. J Clin
nals by pattern-recognition receptors. Annu Rev Plant Investig. 2019;128(10):4639–53.
Biol. 2009;60:379–406. Das UN. Saturated fatty acids, mufas and pufas regulate ferrop-
Bonen A, Chabowski A, Luiken JJ, Glatz JF. Is membrane tosis. Cell Chem Biol. 2019;26(3):309–11.
transport of FFA mediated by lipid, protein, or both? Dixon SJ, Lemberg KM, Lamprecht MR, Skouta R, Zait-
Mechanisms and regulation of protein-mediated cellular sev EM, Gleason CE, et al. Ferroptosis: an iron-
fatty acid uptake: molecular, biochemical, and physiolog- dependent form of nonapoptotic cell death. Cell.
ical evidence. Physiology (Bethesda). 2007;22:15–29. 2012;149(5):1060–72.
Brabletz T. Emt and met in metastasis: where are the cancer Dixon SJ, Stockwell BR. The role of iron and reactive oxygen
stem cells? Cancer Cell. 2012;22(6):699–701. species in cell death. Nat Chem Biol. 2014;10(1):9–17.
Brown CW, Amante JJ, Chhoy P, Elaimy AL, Liu H, Zhu LJ, Doll S, Conrad M. Iron and ferroptosis: a still ill-defined liai-
et al. Prominin2 drives ferroptosis resistance by stimulat- son. IUBMB Life. 2017;69(6):423–34.
ing iron export. Dev Cell. 2019;51(5):575–86. Doll S, Proneth B, Tyurina YY, Panzilius E, Kobayashi S, Ingold
Cao JY, Dixon SJ. Mechanisms of ferroptosis. Cell Mol Life I, et al. Acsl4 dictates ferroptosis sensitivity by shaping cel-
Sci. 2016;73(11):2195–209. lular lipid composition. Nat Chem Biol. 2017;13(1):91–8.
Chan W, Taylor AJ, Ellims AH, Lefkovits L, Wong C, Kingwell Doll S, Freitas FP, Shah R, Aldrovandi M, Da Silva MC,
BA et al. Effect of iron chelation on myocardial infarct Ingold I, et al. Fsp1 is a glutathione-independent ferrop-
size and oxidative stress in st-elevation–myocardial tosis suppressor. Nature. 2019;575(7784):693–8.
infarction. Circulation. 2012;5(2):270–78. Dolma S, Lessnick SL, Hahn WC, Stockwell BR. Identification
Chen H, Zheng C, Zhang Y, Chang Y, Qian Z, Shen X. Heat of genotype-selective antitumor agents using synthetic
shock protein 27 downregulates the transferrin recep- lethal chemical screening in engineered human tumor
tor 1-mediated iron uptake. Int J Biochem Cell Biol. cells. Cancer Cell. 2003;3(3):285–96.
2006;38(8):1402–16. Donovan A, Lima CA, Pinkus JL, Pinkus GS, Zon LI, Robine
Chen K, Zhang S, Jiao J, Zhao S. Ferroptosis and its potential S, et al. The iron exporter ferroportin/slc40a1 is essential
role in lung cancer: updated evidence from pathogenesis for iron homeostasis. Cell Metab. 2005;1(3):191–200.
to therapy. J Inflamm Res. 2021a;14:7079. Dunn LL, Rahmanto YS, Richardson DR. Iron uptake and
Chen X, Kang R, Kroemer G, Tang D. Broadening horizons: metabolism in the new millennium. Trends Cell Biol.
the role of ferroptosis in cancer. Nat Rev Clin Oncol. 2007;17(2):93–100.
2021b;18(5):280–96. Fan Z, Cai L, Wang S, Wang J, Chen B. Baicalin prevents
Chen X, Kang R, Kroemer G, Tang D. Ferroptosis in infec- myocardial ischemia/reperfusion injury through inhib-
tion, inflammation, and immunity. J Exp Med. iting acsl4 mediated ferroptosis. Front Pharmacol.
2021c;218(6):e20210518. 2021;12:628988.

Vol:. (1234567890)
13
Cell Biol Toxicol (2023) 39:827–851 847

Fang X, Wang H, Han D, Xie E, Yang X, Wei J, et al. Ferropto- cancer cells are vulnerable to gpx4 inhibition. Nature.
sis as a target for protection against cardiomyopathy. Proc 2017;551(7679):247–50.
Natl Acad Sci. 2019;116(7):2672–80. Hanson LR, Roeytenberg A, Martinez PM, Coppes VG,
Feng J, Lu P, Zhu G, Hooi SC, Wu Y, Huang X, et al. Sweet DC, Rao RJ, et al. Intranasal deferoxamine pro-
Acsl4 is a predictive biomarker of sorafenib sensitiv- vides increased brain exposure and significant pro-
ity in hepatocellular carcinoma. Acta Pharmacol Sin. tection in rat ischemic stroke. J Pharmacol Exp Ther.
2021;42(1):160–70. 2009;330(3):679–86.
Fernandez-Fernandez S, Almeida A, Bolaños JP. Antioxidant Hao S, Yu J, He W, Huang Q, Zhao Y, Liang B, et al.
and bioenergetic coupling between neurons and astro- Cysteine dioxygenase 1 mediates erastin-induced fer-
cytes. Biochem J. 2012;443(1):3–11. roptosis in human gastric cancer cells. Neoplasia.
Figueroa-González G, Carrillo-Hernández JF, Perez-Rodriguez 2017;19(12):1022–32.
I, Cantú De León D, Campos-Parra AD, Martínez-Gutié- Haq R, Shoag J, Andreu-Perez P, Yokoyama S, Edelman
rrez AD et al. Negative regulation of serine threonine H, Rowe GC, et al. Oncogenic braf regulates oxida-
kinase 11 (stk11) through mir-100 in head and neck can- tive metabolism via pgc1α and mitf. Cancer Cell.
cer. Genes-Basel. 2020;11(9):1058. 2013;23(3):302–15.
Forcina GC, Dixon SJ. Gpx4 at the crossroads of lipid homeo- Hashidate-Yoshida T, Harayama T, Hishikawa D, Morimoto R,
stasis and ferroptosis. Proteomics. 2019;19(18):1800311. Hamano F, Tokuoka SM et al. Fatty acid remodeling by
Friedmann Angeli JP, Schneider M, Proneth B, Tyurina YY, lpcat3 enriches arachidonate in phospholipid membranes
Tyurin VA, Hammond VJ, et al. Inactivation of the fer- and regulates triglyceride transport. Elife. 2015;4.
roptosis regulator gpx4 triggers acute renal failure in Henderson RJ. Fatty acid metabolism in freshwater fish with
mice. Nat Cell Biol. 2014;16(12):1180–91. particular reference to polyunsaturated fatty acids. Arch
Gagliardi M, Cotella D, Santoro C, Corà D, Barlev NA, Pia- Anim Nutr. 1996;49(1):5–22.
centini M, et al. Aldo-keto reductases protect metastatic Hengartner MO. The biochemistry of apoptosis. Nature.
melanoma from er stress-independent ferroptosis. Cell 2000;407(6805):770–6.
Death Dis. 2019;10(12):1–15. Jenkins NL, James SA, Salim A, Sumardy F, Speed TP, Conrad
Gao M, Monian P, Quadri N, Ramasamy R, Jiang X. Glutami- M, et al. Changes in ferrous iron and glutathione promote
nolysis and transferrin regulate ferroptosis. Mol Cell. ferroptosis and frailty in aging caenorhabditis elegans.
2015;59(2):298–308. Elife. 2020;9:e56580.
Gaschler MM, Stockwell BR. Lipid peroxidation in cell death. Jeon S, Chandel NS, Hay N. Ampk regulates nadph homeosta-
Biochem Biophys Res Commun. 2017;482(3):419–25. sis to promote tumour cell survival during energy stress.
Grynberg K, Ozols E, Mulley WR, Davis RJ, Flavell Nature. 2012;485(7400):661–5.
RA, Nikolic-Paterson DJ, et al. Jun amino-terminal Jiang X, Stockwell BR, Conrad M. Ferroptosis: mechanisms,
kinase 1 signaling in the proximal tubule causes cell biology and role in disease. Nat Rev Mol Cell Bio.
death and acute renal failure in rat and mouse mod- 2021;22(4):266–82.
els of renal ischemia/reperfusion injury. Am J Pathol. Kaneko T, Iuchi Y, Kobayashi T, Fujii T, Saito H, Kurachi H,
2021;191(5):817–28. et al. The expression of glutathione reductase in the male
Guan X, Li X, Yang X, Yan J, Shi P, Ba L, et al. The neuro- reproductive system of rats supports the enzymatic basis
protective effects of carvacrol on ischemia/reperfusion- of glutathione function in spermatogenesis. Eur J Bio-
induced hippocampal neuronal impairment by ferroptosis chem. 2002;269(5):1570–8.
mitigation. Life Sci. 2019;235:116795. Karuppagounder SS, Alim I, Khim SJ, Bourassa MW, Slei-
Guan X, Li Z, Zhu S, Cheng M, Ju Y, Ren L, et al. Galangin man SF, John R, et al. Therapeutic targeting of oxygen-
attenuated cerebral ischemia-reperfusion injury by inhi- sensing prolyl hydroxylases abrogates atf4-dependent
bition of ferroptosis through activating the slc7a11/gpx4 neuronal death and improves outcomes after brain
axis in gerbils. Life Sci. 2021;264:118660. hemorrhage in several rodent models. Sci Transl Med.
Guiney SJ, Adlard PA, Bush AI, Finkelstein DI, Ayton S. Fer- 2016;8(328):328r–9r.
roptosis and cell death mechanisms in parkinson’s dis- Kerr JF, Wyllie AH, Currie AR. Apoptosis: a basic biologi-
ease. Neurochem Int. 2017;104:34–48. cal phenomenon with wideranging implications in tissue
Hambright WS, Fonseca RS, Chen L, Na R, Ran Q. Ablation kinetics. Brit J Cancer. 1972;26(4):239–57.
of ferroptosis regulator glutathione peroxidase 4 in fore- Kim EH, Shin D, Lee J, Jung AR, Roh J. Cisd2 inhibition
brain neurons promotes cognitive impairment and neuro- overcomes resistance to sulfasalazine-induced ferrop-
degeneration. Redox Biol. 2017;12:8–17. totic cell death in head and neck cancer. Cancer Lett.
Hamilton DL, Wedner HJ. The role of glutathione in lym- 2018;432:180–90.
phocyte activation. I. Comparison of inhibitory Koppula P, Zhuang L, Gan B. Cystine transporter slc7a11/xct
effects of buthionine sulfoximine and 2-cyclohexene- in cancer: ferroptosis, nutrient dependency, and cancer
1-one by nuclear size transformation. J Immunol. therapy. Protein Cell. 2021;12(8):599–620.
1985;135(4):2740–47. Kraft VA, Bezjian CT, Pfeiffer S, Ringelstetter L, Müller C,
Han A, Purwin TJ, Aplin AE. Roles of the bap1 tumor Zandkarimi F, et al. Gtp cyclohydrolase 1/tetrahydrobi-
suppressor in cell metabolism. Cancer Res. opterin counteract ferroptosis through lipid remodeling.
2021;81(11):2807–14. Acs Central Sci. 2019;6(1):41–53.
Hangauer MJ, Viswanathan VS, Ryan MJ, Bole D, Kriska T, Pilat A, Schmitt JC, Girotti AW. Sterol carrier pro-
Eaton JK, Matov A, et al. Drug-tolerant persister tein-2 (scp-2) involvement in cholesterol hydroperoxide

Vol.: (0123456789)
13
848 Cell Biol Toxicol (2023) 39:827–851

cytotoxicity as revealed by scp-2 inhibitor effects. J Lipid Liu J, Kang R, Tang D. Escrt-iii-mediated membrane repair
Res. 2010;51(11):3174–84. in cell death and tumor resistance. Cancer Gene Ther.
Kroemer G, El-Deiry WS, Golstein P, Peter ME, Vaux D, Van- 2021;28(1–2):1–4.
denabeele P, et al. Classification of cell death: recom- Liu Y, Fang Y, Zhang Z, Luo Y, Zhang A, Lenahan C, et al.
mendations of the nomenclature committee on cell death. Ferroptosis: an emerging therapeutic target in stroke. J
Cell Death Differ. 2005;12(12):1463–7. Neurochem. 2022;160(1):64–73.
Kruszewski M. Labile iron pool: the main determinant of cellu- Llabani E, Hicklin RW, Lee HY, Motika SE, Crawford LA,
lar response to oxidative stress. Mutat Res Fundam Mol Weerapana E, et al. Diverse compounds from pleuromu-
Mech Mutagen. 2003;531(1–2):81–92. tilin lead to a thioredoxin inhibitor and inducer of ferrop-
Kuang F, Liu J, Tang D, Kang R. Oxidative damage and anti- tosis. Nat Chem. 2019;11(6):521–32.
oxidant defense in ferroptosis. Front Cell Dev Biol. Ma S, Henson ES, Chen Y, Gibson SB. Ferroptosis is induced
2020:969. following siramesine and lapatinib treatment of breast
Kuwata H, Hara S. Role of acyl-coa synthetase acsl4 in ara- cancer cells. Cell Death Dis. 2016;7(7):e2307.
chidonic acid metabolism. Prostag Oth Lipid m. Magtanong L, Ko P, To M, Cao JY, Forcina GC, Tarangelo
2019;144:106363. A, et al. Exogenous monounsaturated fatty acids pro-
Kwon M, Park E, Lee S, Chung SW. Heme oxygenase-1 accel- mote a ferroptosis-resistant cell state. Cell Chem Biol.
erates erastin-induced ferroptotic cell death. Oncotarget. 2019;26(3):420–32.
2015;6(27):24393–403. Mai TT, Hamaï A, Hienzsch A, Cañeque T, Müller S, Wicinski
Lee H, Zandkarimi F, Zhang Y, Meena JK, Kim J, Zhuang L, J, et al. Salinomycin kills cancer stem cells by sequester-
et al. Energy-stress-mediated ampk activation inhibits ing iron in lysosomes. Nat Chem. 2017;9(10):1025–33.
ferroptosis. Nat Cell Biol. 2020;22(2):225–34. Mancias JD, Wang X, Gygi SP, Harper JW, Kimmel-
Lewerenz J, Hewett SJ, Huang Y, Lambros M, Gout PW, Kali- man AC. Quantitative proteomics identifies ncoa4 as
vas PW, et al. The cystine/glutamate antiporter system the cargo receptor mediating ferritinophagy. Nature.
xc− in health and disease: from molecular mechanisms 2014;509(7498):105–9.
to novel therapeutic opportunities. Antioxid Redox Sign. Miess H, Dankworth B, Gouw AM, Rosenfeldt M, Schmitz
2013;18(5):522–55. W, Jiang M, et al. The glutathione redox system is
Lewerenz J, Klein M, Methner A. Cooperative action of glu- essential to prevent ferroptosis caused by impaired lipid
tamate transporters and cystine/glutamate antiporter metabolism in clear cell renal cell carcinoma. Oncogene.
system xc- protects from oxidative glutamate toxicity. J 2018;37(40):5435–50.
Neurochem. 2006;98(3):916–25. Mohammad RM, Muqbil I, Lowe L, Yedjou C, Hsu H, Lin L
Li Y, Yan H, Xu X, Liu H, Wu C, Zhao L. Erastin/sorafenib et al. Broad targeting of resistance to apoptosis in cancer.
induces cisplatin-resistant non-small cell lung cancer cell Semin Cancer Biol. 2015;35 Suppl(0):S78–S103.
ferroptosis through inhibition of the nrf2/xct pathway. Nawarskas JJ. Hmg-coa reductase inhibitors and coenzyme
Oncol Lett. 2020a;19(1):323–33. q10. Cardiol Rev. 2005;13(2):76–9.
Li Z, Chen L, Chen C, Zhou Y, Hu D, Yang J, et al. Targeting Nguyen THP, Mahalakshmi B, Velmurugan BK. Functional
ferroptosis in breast cancer. Biomark Res. 2020b;8(1):58. role of ferroptosis on cancers, activation and deactivation
Li Y, Feng D, Wang Z, Zhao Y, Sun R, Tian D, et al. Ischemia- by various therapeutic candidates-an update. Chem-Biol
induced acsl4 activation contributes to ferroptosis-medi- Interact. 2020;317:108930.
ated tissue injury in intestinal ischemia/reperfusion. Cell Osrodek M, Hartman ML, Czyz M. Physiologically relevant
Death Differ. 2019;26(11):2284–99. oxygen concentration (6% o2) as an important compo-
Li X, Ma N, Xu J, Zhang Y, Yang P, Su X et al. Targeting fer- nent of the microenvironment impacting melanoma phe-
roptosis: pathological mechanism and treatment of notype and melanoma response to targeted therapeutics
ischemia-reperfusion injury. Oxid Med Cell Longev. in vitro. Int J Mol Sci. 2019;20(17):4203.
2021;2021. Ou W, Liang Y, Qing Y, Wu W, Xie M, Zhang Y, et al.
Lin H, Ho H, Chang Y, Wei C, Chu P. The evolving role of fer- Hypoxic acclimation improves cardiac redox homeosta-
roptosis in breast cancer: translational implications pre- sis and protects heart against ischemia-reperfusion injury
sent and future. Cancers. 2021;13(18):4576. through upregulation of o-glcnacylation. Redox Biol.
Lin X, Ping J, Wen Y, Wu Y. The mechanism of ferroptosis 2021;43:101994.
and applications in tumor treatment. Front Pharmacol. Ou M, Lin C, Wang Y, Lu Y, Wang W, Li Z, et al. Heterojunc-
2020:1061. tion engineered bioactive chlorella for cascade promoted
Linkermann A. Nonapoptotic cell death in acute kidney injury cancer therapy. J Control Release. 2022;345:755–69.
and transplantation. Kidney Int. 2016;89(1):46–57. Pagliara V, Saide A, Mitidieri E, di Villa BRD, Sorrentino R,
Liu X, Nguyen NH, Marquess KD, Yang F, Haile DJ. Regula- Russo G, et al. 5-fu targets rpl3 to induce mitochondrial
tion of hepcidin and ferroportin expression by lipopoly- apoptosis via cystathionine-β-synthase in colon cancer
saccharide in splenic macrophages. Blood Cells Mol Dis. cells lacking p53. Oncotarget. 2016;7(31):50333.
2005;35(1):47–56. Paraskevaidis IA, Iliodromitis EK, Vlahakos D, Tsiapras DP,
Liu T, Jiang L, Tavana O, Gu W. The deubiquitylase otub1 Nikolaidis A, Marathias A, et al. Deferoxamine infu-
mediates ferroptosis via stabilization of slc7a11. Cancer sion during coronary artery bypass grafting ameliorates
Res. 2019;79(8):1913–24. lipid peroxidation and protects the myocardium against

Vol:. (1234567890)
13
Cell Biol Toxicol (2023) 39:827–851 849

reperfusion injury: immediate and long-term signifi- Stincone A, Prigione A, Cramer T, Wamelink MM, Campbell
cance. Eur Heart J. 2005;26(3):263–70. K, Cheung E, et al. The return of metabolism: biochem-
Park S, Oh J, Kim M, Jin E. Bromelain effectively suppresses istry and physiology of the pentose phosphate pathway.
kras-mutant colorectal cancer by stimulating ferroptosis. Biol Rev. 2015;90(3):927–63.
Anim Cells Syst. 2018;22(5):334–40. Stockwell BR, Friedmann Angeli JP, Bayir H, Bush AI, Con-
Park SH, Gammon SR, Knippers JD, Paulsen SR, Rubink rad M, Dixon SJ, et al. Ferroptosis: a regulated cell death
DS, Winder WW. Phosphorylation-activity relation- nexus linking metabolism, redox biology, and disease.
ships of ampk and acetyl-coa carboxylase in muscle. Cell. 2017;171(2):273–85.
Journal of applied physiology (Bethesda, Md. : 1985). Stockwell BR. Ferroptosis turns 10: emerging mechanisms,
2002;92(6):2475–82. physiological functions, and therapeutic applications.
Phani S, Loike JD, Przedborski S. Neurodegeneration and Cell. 2022;185(14):2401–21.
inflammation in parkinson’s disease. Parkinsonism Relat Su Y, Zhao B, Zhou L, Zhang Z, Shen Y, Lv H, et al. Ferrop-
D. 2012;18:S207–9. tosis, a novel pharmacological mechanism of anti-cancer
Philpott CC, Patel SJ, Protchenko O. Management versus mis- drugs. Cancer Lett. 2020;483:127–36.
cues in the cytosolic labile iron pool: the varied functions Sun X, Ou Z, Xie M, Kang R, Fan Y, Niu X, et al. Hspb1 as
of iron chaperones. Biochim Biophys Acta Mol Cell Res a novel regulator of ferroptotic cancer cell death. Onco-
2020;1867(11):118830. gene. 2015;34(45):5617–25.
Poljsak B, Šuput D, Milisav I. Achieving the balance between Takebe N, Warren RQ, Ivy SP. Breast cancer growth and
ROS and antioxidants: when to use the synthetic antioxi- metastasis: interplay between cancer stem cells, embry-
dants. Oxid Med Cell Longev. 2013;2013:956792. onic signaling pathways and epithelial-to-mesenchymal
Probst L, Dächert J, Schenk B, Fulda S. Lipoxygenase transition. Breast Cancer Res. 2011;13(3):1–11.
inhibitors protect acute lymphoblastic leukemia cells Tang D, Kroemer G. Ferroptosis. Curr Biol.
from ferroptotic cell death. Biochem Pharmacol. 2020;30(21):R1292–7.
2017;140:41–52. Tang R, Xu J, Zhang B, Liu J, Liang C, Hua J, et al. Ferropto-
Qiu Y, Cao Y, Cao W, Jia Y, Lu N. The application of ferropto- sis, necroptosis, and pyroptosis in anticancer immunity. J
sis in diseases. Pharmacol Res. 2020;159:104919. Hematol Oncol. 2020;13(1):110.
Quan J, Bode AM, Luo X. Acsl family: the regulatory mecha- Thöny B, Auerbach G, Blau N. Tetrahydrobiopterin bio-
nisms and therapeutic implications in cancer. Eur J Phar- synthesis, regeneration and functions. Biochem J.
macol. 2021;909:174397. 2000;347(1):1–16.
Russo A, Saide A, Smaldone S, Faraonio R, Russo G. Role of Tocher DR. Metabolism and functions of lipids and fatty acids
ul3 in multidrug resistance in p53-mutated lung cancer in teleost fish. Rev Fish Sci. 2003;11(2):107–84.
cells. Int J Mol Sci. 2017;18(3):547. Tsoi J, Robert L, Paraiso K, Galvan C, Sheu KM, Lay J, et al.
Sasaki M, Chiwaki F, Kuroda T, Komatsu M, Matsusaki K, Multi-stage differentiation defines melanoma subtypes with
Kohno T, et al. Efficacy of glutathione inhibitors for the differential vulnerability to drug-induced iron-dependent
treatment of arid1a-deficient diffuse-type gastric cancers. oxidative stress. Cancer Cell. 2018;33(5):890–904.
Biochem Biophys Res Commun. 2020;522(2):342–7. Uderhardt S, Herrmann M, Oskolkova OV, Aschermann S,
Sauler M, Bazan IS, Lee PJ. Cell death in the lung: the Bicker W, Ipseiz N, et al. 12/15-lipoxygenase orches-
apoptosis–necroptosis axis. Annu Rev Physiol. trates the clearance of apoptotic cells and maintains
2019;81:375–402. immunologic tolerance. Immunity. 2012;36(5):834–46.
Schöckel L, Glasauer A, Basit F, Bitschar K, Truong H, Erd- Vance RE, Hong S, Gronert K, Serhan CN, Mekalanos JJ. The
mann G, et al. Targeting mitochondrial complex i using opportunistic pathogen pseudomonas aeruginosa carries
bay 87–2243 reduces melanoma tumor growth. Cancer a secretable arachidonate 15-lipoxygenase. Proc Natl
Metab. 2015;3(1):1–16. Acad Sci. 2004;101(7):2135–9.
Scindia Y, Dey P, Thirunagari A, Liping H, Rosin DL, Floris Vaux DL, Korsmeyer SJ. Cell death in development. Cell.
M, et al. Hepcidin mitigates renal ischemia-reperfusion 1999;96(2):245–54.
injury by modulating systemic iron homeostasis. J Am Viswanathan VS, Ryan MJ, Dhruv HD, Gill S, Eichhoff OM,
Soc Nephrol. 2015;26(11):2800–14. Seashore-Ludlow B, et al. Dependency of a therapy-
Shan X, Lv Z, Yin M, Chen J, Wang J, Wu Q. The protective resistant state of cancer cells on a lipid peroxidase path-
effect of cyanidin-3-glucoside on myocardial ischemia- way. Nature. 2017;547(7664):453–7.
reperfusion injury through ferroptosis. Oxid Med Cell Vogt C, Vogt KC. Untersuchungen über die Entwicklungsge-
Longev. 2021;2021. schichte der Geburtshelferkröte (Alytes obstetricans).
Song Y, Wang B, Zhu X, Hu J, Sun J, Xuan J, et al. Human Jent & Gassmann, 1842.
umbilical cord blood–derived mscs exosome attenu- Wang H, Cheng Y, Mao C, Liu S, Xiao D, Huang J, et al.
ate myocardial injury by inhibiting ferroptosis in Emerging mechanisms and targeted therapy of ferropto-
acute myocardial infarction mice. Cell Biol Toxicol. sis in cancer. Mol Ther. 2021;29(7):2185–208.
2021;37(1):51–64. WARBURG O. On the origin of cancer cells. Science (New
Soula M, Weber RA, Zilka O, Alwaseem H, La K, Yen F, York, N.Y.). 1956;123(3191):309–14.
et al. Metabolic determinants of cancer cell sensitiv- Ward RJ, Dexter DT, Crichton RR. Neurodegenerative diseases
ity to canonical ferroptosis inducers. Nat Chem Biol. and therapeutic strategies using iron chelators. J Trace
2020;16(12):1351–60. Elem Med Bio. 2015;31:267–73.

Vol.: (0123456789)
13
850 Cell Biol Toxicol (2023) 39:827–851

Wei G, Sun J, Hou Z, Luan W, Wang S, Cui S, et al. Novel Yang Y, Lin X. Potential relationship between autophagy and
antitumor compound optimized from natural saponin ferroptosis in myocardial ischemia/reperfusion injury.
albiziabioside a induced caspase-dependent apoptosis Genes Dis. 2022.
and ferroptosis as a p53 activator through the mitochon- Yang WS, Kim KJ, Gaschler MM, Patel M, Shchepinov MS,
drial pathway. Eur J Med Chem. 2018;157:759–72. Stockwell BR. Peroxidation of polyunsaturated fatty
Wei X, Yi X, Zhu X, Jiang D. Posttranslational modifications acids by lipoxygenases drives ferroptosis. Proc Natl Acad
in ferroptosis. Oxid Med Cell Longev. 2020;2020. Sci. 2016;113(34):E4966–75.
Weigand I, Schreiner J, Röhrig F, Sun N, Landwehr L, Urlaub Yang Y, Luo M, Zhang K, Zhang J, Gao T, Connell DO,
H, et al. Active steroid hormone synthesis renders adren- et al. Nedd4 ubiquitylates vdac2/3 to suppress eras-
ocortical cells highly susceptible to type ii ferroptosis tin-induced ferroptosis in melanoma. Nat Commun.
induction. Cell Death Dis. 2020;11(3):1–12. 2020;11(1):1–14.
Weiland A, Wang Y, Wu W, Lan X, Han X, Li Q, et al. Fer- Yankner BA. Mechanisms of neuronal degeneration in alzhei-
roptosis and its role in diverse brain diseases. Mol Neu- mer’s disease. Neuron. 1996;16(5):921–32.
robiol. 2019;56(7):4880–93. Yao M, Liu T, Zhang L, Wang M, Yang Y, Gao J. Role of
Weinberg ED. The role of iron in cancer. Eur J Cancer Prev. ferroptosis in neurological diseases. Neurosci Lett.
1996:19–36. 2021;747:135614.
Wen Q, Liu J, Kang R, Zhou B, Tang D. The release and activ- Yu H, Yang C, Jian L, Guo S, Chen R, Li K, et al. Sul-
ity of hmgb1 in ferroptosis. Biochem Biophys Res Com- fasalazine-induced ferroptosis in breast cancer
mun. 2019;510(2):278–83. cells is reduced by the inhibitory effect of estro-
Wu J, Tuo Q, Lei P. Ferroptosis, a recent defined form of criti- gen receptor on the transferrin receptor. Oncol Rep.
cal cell death in neurological disorders. J Mol Neurosci. 2019;42(2):826–38.
2018;66(2):197–206. Zeng X, An H, Yu F, Wang K, Zheng L, Zhou W, et al. Ben-
Wu H, Wang F, Ta N, Zhang T, Gao W. The multifac- efits of iron chelators in the treatment of parkinson’s dis-
eted regulation of mitochondria in ferroptosis. Life. ease. Neurochem Res. 2021;46(5):1239–51.
2021a;11(3):222. Zhang Y, Wang D, Xu S, Zhang S, Fan Y, Yang Y, et al.
Wu S, Zhu C, Tang D, Dou QP, Shen J, Chen X. The role of fer- Α-lipoic acid improves abnormal behavior by mitiga-
roptosis in lung cancer. Biomark Res. 2021b;9(1):1–14. tion of oxidative stress, inflammation, ferroptosis, and
Xia X, Fan X, Zhao M, Zhu P. The relationship between fer- tauopathy in p301s tau transgenic mice. Redox Biol.
roptosis and tumors: a novel landscape for therapeutic 2018;14:535–48.
approach. Curr Gene Ther. 2019;19(2):117. Zhang X, Sui S, Wang L, Li H, Zhang L, Xu S, et al. Inhibition
Xiao D, Zeng L, Yao K, Kong X, Wu G, Yin Y. The glutamine- of tumor propellant glutathione peroxidase 4 induces fer-
alpha-ketoglutarate (akg) metabolism and its nutritional roptosis in cancer cells and enhances anticancer effect of
implications. Amino Acids. 2016;48(9):2067–80. cisplatin. J Cell Physiol. 2020;235(4):3425–37.
Xie A, Gao J, Xu L, Meng D. Shared mechanisms of neuro- Zhang J, Liu Y, Li Q, Xu A, Hu Y, Sun C. Ferroptosis in hema-
degeneration in alzheimer’s disease and parkinson’s dis- tological malignancies and its potential network with
ease. Biomed Res Int. 2014;2014. abnormal tumor metabolism. Biomed Pharmacother.
Xie Y, Hou W, Song X, Yu Y, Huang J, Sun X, et al. Fer- 2022a;148:112747.
roptosis: process and function. Cell Death Differ. Zhang Q, Qu Y, Zhang Q, Li F, Li B, Li Z et al. Exosomes
2016;23(3):369–79. derived from hepatitis b virus-infected hepatocytes pro-
Xie Y, Zhu S, Song X, Sun X, Fan Y, Liu J, et al. The tumor mote liver fibrosis via mir-222/tfrc axis. Cell Biol Toxi-
suppressor p53 limits ferroptosis by blocking dpp4 activ- col. 2022b:1–15.
ity. Cell Rep. 2017;20(7):1692–704. Zhao Y, Li Y, Zhang R, Wang F, Wang T, Jiao Y. The role of
Xu D, Lü Y, Li Y, Li S, Wang Z, Wang J. Ferroptosis resistance erastin in ferroptosis and its prospects in cancer therapy.
in cancer: an emerging crisis of new hope. BIO Integra- Oncotargets Ther. 2020;13:5429–41.
tion. 2021a;2(1):22–8. Zhao S, Li P, Wu W, Wang Q, Qian B, Li X, et al. Roles of
Xu S, He Y, Lin L, Chen P, Chen M, Zhang S. The emerging ferroptosis in urologic malignancies. Cancer Cell Int.
role of ferroptosis in intestinal disease. Cell Death Dis. 2021a;21(1):676.
2021b;12(4):289. Zhao Y, Huang Z, Peng H. Molecular mechanisms of ferrop-
Xu S, Min J, Wang F. Ferroptosis: an emerging player in tosis and its roles in hematologic malignancies. Front
immune cells. Sci Bull. 2021c;22:2257–60. Oncol. 2021b;11:743006.
Yamada N, Karasawa T, Wakiya T, Sadatomo A, Ito H, Zheng J, Sato M, Mishima E, Sato H, Proneth B, Con-
Kamata R, et al. Iron overload as a risk factor for rad M. Sorafenib fails to trigger ferroptosis across
hepatic ischemia-reperfusion injury in liver transplan- a wide range of cancer cell lines. Cell Death Dis.
tation: potential role of ferroptosis. Am J Transplant. 2021;12(7):1–10.
2020;20(6):1606–18. Zhou B, Liu J, Kang R, Klionsky DJ, Kroemer G, Tang D.
Yan B, Ai Y, Sun Q, Ma Y, Cao Y, Wang J, et al. Membrane Ferroptosis is a type of autophagy-dependent cell death.
damage during ferroptosis is caused by oxidation of Semin Cancer Biol. 2020;66:89–100.
phospholipids catalyzed by the oxidoreductases por and Zille M, Oses-Prieto JA, Savage SR, Karuppagounder SS,
cyb5r1. Mol Cell. 2021;81(2):355–69. Chen Y, Kumar A, et al. Hemin-induced death models

Vol:. (1234567890)
13
Cell Biol Toxicol (2023) 39:827–851 851

hemorrhagic stroke and is a variant of classical neuronal Publisher’s Note Springer Nature remains neutral with regard
ferroptosis. J Neurosci. 2022;42(10):2065–79. to jurisdictional claims in published maps and institutional
Zitvogel L, Kroemer G. Interferon-γ induces cancer cell ferrop- affiliations.
tosis. Cell Res. 2019;29(9):692–3.
Zou Y, Palte MJ, Deik AA, Li H, Eaton JK, Wang W Springer Nature or its licensor (e.g. a society or other partner)
et al. Hif-2α drives an intrinsic vulnerability to fer- holds exclusive rights to this article under a publishing
roptosis in clear cell renal cell carcinoma. BioRxiv. agreement with the author(s) or other rightsholder(s); author
2018:388041. self-archiving of the accepted manuscript version of this article
Zuo S, Yu J, Pan H, Lu L. Novel insights on targeting ferropto- is solely governed by the terms of such publishing agreement
sis in cancer therapy. Biomark Res. 2020;8(1):1–11. and applicable law.

Vol.: (0123456789)
13

You might also like