ArticlePDF AvailableLiterature Review

ACSL4-Mediated Ferroptosis and Its Potential Role in Central Nervous System Diseases and Injuries

Authors:

Abstract and Figures

As an iron-dependent regulated form of cell death, ferroptosis is characterized by iron-dependent lipid peroxidation and has been implicated in the occurrence and development of various diseases, including nervous system diseases and injuries. Ferroptosis has become a potential target for intervention in these diseases or injuries in relevant preclinical models. As a member of the Acyl-CoA synthetase long-chain family (ACSLs) that can convert saturated and unsaturated fatty acids, Acyl—CoA synthetase long-chain familymember4 (ACSL4) is involved in the regulation of arachidonic acid and eicosapentaenoic acid, thus leading to ferroptosis. The underlying molecular mechanisms of ACSL4-mediated ferroptosis will promote additional treatment strategies for these diseases or injury conditions. Our review article provides a current view of ACSL4-mediated ferroptosis, mainly including the structure and function of ACSL4, as well as the role of ACSL4 in ferroptosis. We also summarize the latest research progress of ACSL4-mediated ferroptosis in central nervous system injuries and diseases, further proving that ACSL4-medicated ferroptosis is an important target for intervention in these diseases or injuries.
Content may be subject to copyright.
Citation: Jia, B.; Li, J.; Song, Y.; Luo,
C. ACSL4-Mediated Ferroptosis and
Its Potential Role in Central Nervous
System Diseases and Injuries. Int. J.
Mol. Sci. 2023,24, 10021. https://
doi.org/10.3390/ijms241210021
Academic Editors: Hari
Shanker Sharma and Anna Atlante
Received: 12 April 2023
Revised: 1 June 2023
Accepted: 6 June 2023
Published: 12 June 2023
Copyright: © 2023 by the authors.
Licensee MDPI, Basel, Switzerland.
This article is an open access article
distributed under the terms and
conditions of the Creative Commons
Attribution (CC BY) license (https://
creativecommons.org/licenses/by/
4.0/).
International Journal of
Molecular Sciences
Review
ACSL4-Mediated Ferroptosis and Its Potential Role in Central
Nervous System Diseases and Injuries
Bowen Jia, Jing Li, Yiting Song and Chengliang Luo *
Department of Forensic Medicine, School of Basic Medicine and Biological Sciences, Soochow University,
Suzhou 215123, China
*Correspondence: clluo@suda.edu.cn
Abstract:
As an iron-dependent regulated form of cell death, ferroptosis is characterized by iron-
dependent lipid peroxidation and has been implicated in the occurrence and development of various
diseases, including nervous system diseases and injuries. Ferroptosis has become a potential target for
intervention in these diseases or injuries in relevant preclinical models. As a member of the Acyl-CoA
synthetase long-chain family (ACSLs) that can convert saturated and unsaturated fatty acids, Acyl—
CoA synthetase long-chain familymember4 (ACSL4) is involved in the regulation of arachidonic
acid and eicosapentaenoic acid, thus leading to ferroptosis. The underlying molecular mechanisms
of ACSL4-mediated ferroptosis will promote additional treatment strategies for these diseases or
injury conditions. Our review article provides a current view of ACSL4-mediated ferroptosis, mainly
including the structure and function of ACSL4, as well as the role of ACSL4 in ferroptosis. We also
summarize the latest research progress of ACSL4-mediated ferroptosis in central nervous system
injuries and diseases, further proving that ACSL4-medicated ferroptosis is an important target for
intervention in these diseases or injuries.
Keywords: ACSL4; ferroptosis; nervous system diseases; nervous system injuries
1. Introduction
Cell death is a common process in all living organisms, and diverse types of cell death
have been classified over time. According to the latest recommendations of the Cell Death
Nomenclature Committee in 2018, there are two types of cell death, that is, accidental cell
death (ACD) and regulated cell death (RCD) [
1
]. As a new form of regulated cell death
(RCD), ferroptosis is iron-dependent and characterized by the intracellular accumulation
of lipid peroxides to lethal levels [
2
]. However, ferroptosis is distinct from apoptosis,
various forms of necrosis, and autophagy in terms of morphology, biochemistry, and gene
expression. The typical symptoms of mitochondrial cristae reduction or disappearance and
outer membrane rupture differ from those of apoptotic cells, which are characterized by
membrane blistering and contraction [
3
]. ACSLs are a family of enzymes that can convert
saturated and unsaturated fatty acids with chain lengths of 8–22 to fatty acid acyl-CoA
esters [
4
]. ACSLs mediate fatty acid metabolism and are widely involved in endoplasmic
reticulum ER stress and ferroptosis. In particular, ACSL4 is a key enzyme in the production
of lipid peroxides, thus promoting ferroptosis in cells [
5
]. In this review, we will mainly
discuss the role of ACSL4 in the process of ferroptosis and investigate the effect of ACSL4
on nervous system diseases or injuries.
2. The Structure and Physiological Function of ACSL4
2.1. The Structure of ACSL4
ACSLs play a crucial role in activating long- and ultra-long-chain fatty acids to form
fatty acid acyl-CoA esters. It is made up of five members of the ACSLs family and can be
divided into two groups according to the composition of the homologous sequences. One is
Int. J. Mol. Sci. 2023,24, 10021. https://doi.org/10.3390/ijms241210021 https://www.mdpi.com/journal/ijms
Int. J. Mol. Sci. 2023,24, 10021 2 of 17
composed of ACSL1, ACSL5, and ACSL6, the other group consists of ACSL3 and ACSL4 [
6
].
The gene for ACSL4 is situated on the X chromosome of the human body, and the subcellular
localization of ACSL4 is mainly in the secretion pathway of endosome and peroxisome [
7
].
In addition, ACSL4 is transferred to the plasma membrane and the mitochondria-associated
membrane, which is responsible for fatty acid synthesis and beta-oxidation [
7
]. ACSL4 is
formed from five regions: an NH2 terminal, luciferase-like regions 1 and 2, the ligand that
connects two luciferase-like regions, and a C terminal [
8
]. ACSL4 is highly expressed in the
brain, adrenal glands, testes, and ovaries. In the ACSLs family, the luciferin-like region 2
and the C terminal amino acids sequence are identical, suggesting that the two regions are
the crux of the reaction catalyzed by ACSLs. The absence of 50 NH2-corresponding amino
acids may lead to differential responses of ACSLs to fatty acid preferences [
6
,
9
]. ACSL4
specifically exhibits preference for 20-carbon polyunsaturated fatty acid (PUFA) substrates,
including arachidonic acid (AA) and adrenic acid (AdA) [10].
2.2. The Physiological Function of ACSL4
Long-chain fatty acids (carbon chain length 14 to 24) are significant nutrients for the
formation and maintenance of cell membranes, energy supply with storage, membrane
anchoring of proteins in protein post-translational modification (PTM) pathways, transport
and localization pathways, and signal transduction, as well as protein interactions, etc. [
6
]
First phase fatty acid transporters (FATPs) bind and transport long-chain fatty acids to
target cells. ACSLs then catalyze the intracellular free long-chain fatty acids to acyl-CoA.
PUFA is an acronym for polyunsaturated fatty acids, which are a category of fatty acids
with multiple double bonds in their carbon chains. These fatty acids are divided into
two primary groups:
ω
-3 (n-3) and
ω
-6 (n-6) fatty acids, based on the location of the
first double bond from the methyl end of the fatty acid chain [
11
]. In the mammalian
ACSLs family [
12
], ACSL4 tends to catalyze several PUFAs to polyunsaturated fatty acid
coenzyme A (PUFAs-CoA), primarily including arachidonic acid 20:4 and adrenic acid
22:4. Following their formation, PUFAs-CoA are esterified into phospholipids by different
lysophosphatidylcholine acyltransferases (LPCATs). This process facilitates the incorpora-
tion of long-chain polyunsaturated fatty acids into cellular lipid membranes [
8
]. It enhances
membrane fluidity and facilitates the transportation of substances required for maintain-
ing normal cellular physiological functions. ACSL4 is involved in distinct biochemical
processes across different organelles. Within mitochondria, ACSL4 primarily contributes
to fatty acid synthesis and
β
-oxidation [
13
]. Among the peroxidases, ACSL4 is mainly
involved in
β
-oxidation and the synthesis of alkyl lipids. In the endoplasmic reticulum
(ER), it promotes glycerolipid synthesis and
ω
-oxidation, serving as the primary pathway
for catabolism of medium-chain fatty acids when
β
-oxidation is impaired [
14
,
15
]. The afore-
mentioned findings highlight the critical involvement of ACSL4 in fatty acid metabolism,
and it is the only subunit of the ACSLs family that plays an essential and direct role in
the process of ferroptosis. However, the influence of ACSL4 on the catalytic selectivity for
exogenous fatty acids can vary among different tissues and cell types, potentially owing
to variations in cell type and intracellular fatty acid composition [16,17]. For example, the
absence of ACSL4 in lipocytes reduces the incorporation of AA into phospholipids and
correspondingly reduces the level of 4-hydroxynonenal, the lipid peroxidation product
of AA [
6
]. ACSL4 deficiency plays a role in obesity-associated adipocyte dysfunction
because the ability of PUFA to synthesize phospholipids is abruptly diminished, resulting
in alterations in lipid composition and a high-fat diet and leading to fat accumulation
and adipocyte death [
18
]. The overexpression of ACSL4 in human arterial smooth muscle
cells stimulates the production of phosphatidylinositol (PI) and phosphatidylinositol (PE)
from exogenous AA, resulting in a decreased release of cytokine-dependent PGE2 [
19
,
20
].
Moreover, ACSL4 expression stimulates the generation of PE from exogenous AA and
oleic acid (OA) in fibroblast-like COS-7 cells. Dissociated AA is also implicated in the
synthesis of phosphatidylcholine (PC) in COS-7 cells, and steroidogenic cells regulate AA
release via the acyl-CoA thioesterase 2 (ACOT2) pathway [
21
]. In this pathway, ACSL4
Int. J. Mol. Sci. 2023,24, 10021 3 of 17
catalyzes the conversion of free intracellular AA into AA-CoA and provides it to ACOT2,
which subsequently transports AA to the mitochondria [
22
]. The released AA undergoes
progressive conversion through the lipoxygenase pathway, leading to the formation of the
steroidogenic acute regulatory (StAR) protein [
17
]. The StAR protein regulates the transport
of cholesterol into the inner mitochondrial membrane and serves as a critical rate-limiting
enzyme in steroid hormone biosynthesis [
23
]. AA and cyclic adenosine monophosphate
(cAMP) transduce signals from hormone receptors into the nucleus through two different
pathways and jointly regulate steroid production and STAR gene expression [6,24].
The overexpression of ACSL4 may result in false positive results by disrupting protein
distribution within cells. To investigate its physiological function, researchers commonly
use gene silencing or knockout experiments. For example, knockout of ACSL4 in rat fi-
broblast 3Y1 cells led to a reduction of AA-containing phospholipid levels following IL-1
β
stimulation, while AA-containing PC and PI levels were less affected [
6
,
25
]. In mouse em-
bryonic fibroblasts, ACSL4 knockout significantly decreased the level of polyunsaturated
fatty acid PE and inhibited ferroptosis [
26
]. Additionally, recombinant glutathione peroxidase
4 (GPX4)-ACSL4 double knockout cells showed significant resistance to ferroptosis. ACSL4
facilitates the esterification of CoA to free fatty acids in an ATP-dependent manner, thereby
activating fatty acid oxidation or lipid biosynthesis. This enzyme is also responsible for
the enrichment of long-chain unsaturated
ω
-6 fatty acids in the cell membrane [
27
,
28
].
However, ACSL4-KO cells exhibited increased sensitivity to ferroptosis upon supplemen-
tation with exogenous AA or AdA (along with other long-chain PUFAs), likely due to
the different kinetic properties of ACSL enzymes. ACSL4 prefers to use AA and AdA to
synthesize phospholipids at low concentrations in the presence of other fatty acids, while
other ACSL-like enzymes use other fatty acids. This concept is physiologically relevant
because the molar percentage of plasma AA levels is at least one to two orders of magnitude
lower than other fatty acids such as oleic acid, suggesting that ACSL4 is responsible for
activating AA at physiological concentrations while other ACSL-like enzymes may activate
AA when intracellular AA levels are elevated [26].
Thus, the regulation of PUFA by ACSL4 plays a crucial physiological role. Additionally,
ACSL4 serves other physiological functions apart from its role in lipid metabolism. A study
conducted by Cho revealed that ACSL4-deficient pure mice mostly perish during embryonic
development, while heterozygous female mice with ACSL4 deficiency experience decreased
fertility and compromised offspring quality [29,30].
2.3. The Regulation Mechanism of ACSL4
ACSL4 transcription is negatively regulated by miR-211-5p, miR-204A-5p, miR-34A-
5p, miR-424-5p, miR-205, and miR-34a [
5
], the expression of which is also inhibited by the
activation of integrin
α
6
β
4-mediated Src and signal transduction and transcription activator
3 (STAT3) or androgen receptors [
31
]. Furthermore, free AA may alter the levels of the
ACSL4 enzyme through the promotion of ubiquitination and proenzyme degradation [
32
].
Moreover, cyclic adenosine monophosphate (cAMP), special protein 1 (SP1), tyrosine
phosphatase SHP2 [
33
], and proto-oncogene transcriptional co-activator YAP were shown
to positively regulate ACSL4 expression [
5
]. ACSL4’s proximal promoter region contains
a cAMP response element binding site that initiates ACSL4 transcription by binding to
cAMP. The transcription of the ACSL4 gene can be triggered by the release of YAP activity
to enhance ferroptosis [
34
]. In addition, Zhang et al., found that the activation of PKC
β
II,
one of the isoforms of PKC (protein kinase C), amplified lipid peroxidation through the
phosphorylation and activation of ACSL4, which could directly phosphorylate ACSL4
Thr328. Furthermore, the lipid peroxidation PKC
β
II-ACSL4 positive feedback mechanism
could enhance the level of lipid peroxidation to induce ferroptosis [35].
3. ACSL4 in Ferroptosis
As a form of cell death driven by iron-dependent lipid peroxidation, ferroptosis was
proposed in 2012 by Drs. Brent R. Stockwell, Scott Dixon, and members of their labora-
Int. J. Mol. Sci. 2023,24, 10021 4 of 17
tory [
36
38
]. In general, ferroptosis has three essential features: (1) oxidation of PUFAs
(including membrane phospholipids); (2) redox activities related to iron utilization; (3) loss
of lipid hydrogen peroxide (LOOH) repair capacity [
8
]. Morphologically, ferroptosis cells
exhibit typical changes in mitochondrial membrane shrinkage, reduction or disappear-
ance of mitochondrial cristae, and rupture of the outer membrane, whereas mitochondria
usually show swelling in other forms of cell death [
39
41
]. Furthermore, treated with the
ferroptosis inducer erastin, the nuclei of cancer cells retained their structural integrity and
no nuclear pyknosis or chromatin edge clustering was observed [
42
]. These morphologic
features distinguish ferroptosis from apoptosis and necrosis [43].
Alterations in fatty acid metabolism serve as markers that indicate various patho-
logical conditions and metabolic disorders. Lipid metabolism disorders are observed in
ferroptosis and are heavily reliant on specific lipid metabolism proteins involved in the
metabolism of AA and AdA. To identify the major genes associated with lipid peroxida-
tion in ferroptosis, the research teams of Sebastian Doll and Bettina Proneth performed
two independent genetic experiments. By simultaneously analyzing a short palindromic
repeat-based genetic screen and another transcriptome microarray assay after comparing
ferroptosis-sensitive and resistant cells, ACSL4 gene expression was found to be indis-
pensable in the process of the oxidation of arachidonic acid-phosphatidylethanolamine
(AA-PE) and adrenal acid-phosphatidylethanolamine (AdA-PE) [
26
]. Among the ACSLs
family members, ACSL4 is the lipid metabolism enzyme which is most closely related to
ferroptosis. The overexpression of ACSL4 leads to the catalysis of diverse PUFAs, including
AA/AdA, thereby modifying the composition of cellular lipids and increasing cellular
susceptibility to ferroptosis [
44
]. In general, AdA and AA are first activated by ACSL4
during ferroptosis, followed by the formation of AdA-CoA and AA-CoA derivatives at
ER-associated oxidation centers. AdA-CoA and AA-CoA are then esterified by LPCAT3
to AdA-PE and AA-PE, which are then oxidized by 15-lipoxygenase (15-LOX) to produce
lipid hydroperoxide, giving rise to ferroptosis (Figure 1).
By cloning the mouse ACSL4 gene cDNA into a lentiviral vector, Yu Cui and Yan
Zhang demonstrated that cortical lentivirus administration injected into the left brain after
tMCAO surgery resulted in increased infarct size and decreased neurological function in
the ACSL4-overexpressing brain. Additionally, confocal microscopy revealed neuronal
death and heightened microglial activation in ACSL4-overexpressing mice, leading to
the release of substantial amounts of neurotoxic factors such as reactive oxygen species
(ROS) [
45
,
46
]. In the cerebral ischemia/reperfusion model, ACSL4 knockdown attenuates
ischemic brain injury while ACSL4 overexpression exacerbates ischemic brain injury [
26
].
Furthermore, ACSL4 contributes to neuronal death by promoting ferroptosis, and therefore,
inhibiting the esterification of AA/AdA to PE through pharmacological or genetic inhibi-
tion of ACSL4 has emerged as a specific approach to counteracting ferroptosis [
47
]. ACSL4
is also a potential target for tumor treatment, as studies have demonstrated its inhibitory
effect on glioma cell proliferation through the activation of the ferroptosis pathway [
48
].
Suppression of the thrombin-ACSL4 pathway may reduce neuronal ferroptosis following is-
chemic stroke [
49
]. Additionally, paeonol exhibits significant inhibition of ACSL4-mediated
neuronal ferroptosis induced by ferroptosis inducers [
50
]. In conclusion, ACSL4-mediated
fatty acid activation of AA/AdA is a key step in ferroptosis. The expression level or enzyme
activity of the ACSL4 protein is a vital biological factor for ferroptosis in cells and tissues,
which can be used as a biomarker for ferroptosis susceptibility and as a therapeutic target
for the treatment of ferroptosis-related diseases. Overall, the overexpression of ACSL4
promotes ferroptosis by regulating PUFAs, particularly when PUFAs reach hazardous
levels.
Int. J. Mol. Sci. 2023,24, 10021 5 of 17
Int.J.Mol.Sci.2023,24,xFORPEERREVIEW5of18
Figure1.Duringferroptosis,PKCβIIphosphorylatestheThr328siteofACSL4,directlyactivating
ACSL4andfacilitatingthebiosynthesisofPUFAlipids.PUFAs,primarilyAA20:4andAdA22:4,
areactivatedbyACSL4andthenformPUFA-CoAbybindingwithcoenzymeA(CoA)attheendo-
plasmicreticulumoxidationcenter,aprocessthatconsumesadenosinetriphosphate(ATP).PUFA-
CoAisesteriedtoPUFA-PEthroughtheassistanceofLPCAT3.Subsequently,itundergoesoxida-
tionby15-lipoxygenase(15-LOX),resultingintheproductionoflipidhydroperoxidesthatcontrib-
utetoirondepletion.Additionally,Fe2+canbereleasedfromthelabileironpool,leadingtothe
generationofreactiveoxygenspecies(ROS)suchasHO·throughtheFentonreaction.Consequently,
lipidperoxides,includingLOOH,canaccumulateviaasimilarreactionmediatedbyFe,resulting
inachainreactionthatproducesasignicantnumberoflipidradicals.SystemXCfacilitatesthe
exchangeofcysteineandglutamate,enablinghighlyspeciccysteineuptake.Oncecysteineenters
thecytoplasm,itundergoesreductiontocysteine,followedbycatalysisbyγ-glutamylcysteinesyn-
thase(γ-GCS)andglutathionesynthase(GSS)toproduceglutathionefromcysteine.Twomolecules
ofreducedglutathione(GSH)serveaselectrondonors,reducingPE-AA-OOHandPE-AdA-OOH
totheirrespectivealcohols,PE-AA-OHandPE-AdA-OH,andgeneratingoxidizedglutathione.Fur-
thermore,ACSL4directlyinhibitsGPX4,leadingtoferroptosis.However,steroid-producingcells
regulatethereleaseofAAthroughtheACOT2pathway.Inthispathway,ACSL4catalyzesthecon-
versionofintracellularfreeAAtoAA-coenzymeAandsuppliesittoACOT2,whichsubsequently
releasesAAintothemitochondria.ThereleasedAAisprogressivelymetabolizedthroughthelipox-
ygenasepathway,inducingStAR,althoughitsroleinferroptosisremainsunclear.PUFAs,polyun-
saturatedfayacids;AA,ArachidonicAcid;AdA,AdrenalAcid;PUFA-CoA,PolyunsaturatedFay
Acid-CoenzymeA;AA-CoA,ArachidonicAcid-CoenzymeA;AdA-CoA,AdrenalAcid-Coenzyme
A;ATP ,AdenosineTriphosphate;LPCAT3,Lysophosphatidylcholineacyltransferase3;PE-AA,
Phosphatidylethanolamine-ArachidonicAcid;PE-AA,Phosphatidylethanolamine-AdrenalAcid;
15-LOX,15-lipoxygenase;PE-AA-OOH,Phosphatidylethanolamine-ArachidonicAcidHydroper-
oxide;PE-AdA-OOH,Phosphatidylethanolamine-AdrenalAcidHydroperoxide;PE-AA-OO.,Phos-
phatidylethanolamine-ArachidonicAcidPeroxylRadical;PE-AdA-OO.,Phosphatidylethanola-
mine-AdrenalAcidPeroxylRadical;PE-AA-OH,Phosphatidylethanolamine-ArachidonicAcidAl-
cohol;PE-AdA-OH,Phosphatidylethanolamine-AdrenalAcidAlcohol;Glu,GlutamicAcid;Cys,
Cysteine;GSH,Glutathione;GSSG,GlutathioneDisulde;GPX4,GlutathionePeroxidase4;ACOT2,
acyl-CoAthioesterase2;StAR,steroidogenicacuteregulatory;PKCβII,proteinkinaseCβII.
BycloningthemouseACSL4genecDNAintoalentiviralvector,YuCuiandYa n
Zhangdemonstratedthatcorticallentivirusadministrationinjectedintotheleftbrainafter
tMCAOsurgeryresultedinincreasedinfarctsizeanddecreasedneurologicalfunctionin
theACSL4-overexpressingbrain.Additionally,confocalmicroscopyrevealedneuronal
deathandheightenedmicroglialactivationinACSL4-overexpressingmice,leadingtothe
Figure 1.
During ferroptosis, PKC
β
II phosphorylates the Thr328 site of ACSL4, directly activat-
ing ACSL4 and facilitating the biosynthesis of PUFA lipids. PUFAs, primarily AA 20:4 and AdA
22:4, are activated by ACSL4 and then form PUFA-CoA by binding with coenzyme A (CoA) at the
endoplasmic reticulum oxidation center, a process that consumes adenosine triphosphate (ATP).
PUFA-CoA is esterified to PUFA-PE through the assistance of LPCAT3. Subsequently, it undergoes
oxidation by 15-lipoxygenase (15-LOX), resulting in the production of lipid hydroperoxides that
contribute to iron depletion. Additionally, Fe
2+
can be released from the labile iron pool, lead-
ing to the generation of reactive oxygen species (ROS) such as HO
·
through the Fenton reaction.
Consequently, lipid peroxides, including LOOH, can accumulate via a similar reaction mediated
by Fe, resulting in a chain reaction that produces a significant number of lipid radicals. System
X
C
facilitates the exchange of cysteine and glutamate, enabling highly specific cysteine uptake.
Once cysteine enters the cytoplasm, it undergoes reduction to cysteine, followed by catalysis by
γ
-glutamylcysteine synthase (
γ
-GCS) and glutathione synthase (GSS) to produce glutathione from
cysteine. Two molecules of reduced glutathione (GSH) serve as electron donors, reducing PE-AA-
OOH and PE-AdA-OOH to their respective alcohols, PE-AA-OH and PE-AdA-OH, and generating
oxidized glutathione. Furthermore, ACSL4 directly inhibits GPX4, leading to ferroptosis. However,
steroid-producing cells regulate the release of AA through the ACOT2 pathway. In this pathway,
ACSL4 catalyzes the conversion of intracellular free AA to AA-coenzyme A and supplies it to ACOT2,
which subsequently releases AA into the mitochondria. The released AA is progressively metabolized
through the lipoxygenase pathway, inducing StAR, although its role in ferroptosis remains unclear.
PUFAs, polyunsaturated fatty acids; AA, Arachidonic Acid; AdA, Adrenal Acid; PUFA-CoA, Polyun-
saturated Fatty Acid-Coenzyme A; AA-CoA, Arachidonic Acid-Coenzyme A; AdA-CoA, Adrenal
Acid-Coenzyme A; ATP, Adenosine Triphosphate; LPCAT3, Lysophosphatidylcholine acyltrans-
ferase 3; PE-AA, Phosphatidylethanolamine-Arachidonic Acid; PE-AA, Phosphatidylethanolamine-
Adrenal Acid; 15-LOX, 15-lipoxygenase; PE-AA-OOH, Phosphatidylethanolamine-Arachidonic
Acid Hydroperoxide; PE-AdA-OOH, Phosphatidylethanolamine-Adrenal Acid Hydroperox-
ide; PE-AA-OO
.
, Phosphatidylethanolamine-Arachidonic Acid Peroxyl Radical; PE-AdA-OO
.
,
Phosphatidylethanolamine-Adrenal Acid Peroxyl Radical; PE-AA-OH, Phosphatidylethanolamine-
Arachidonic Acid Alcohol; PE-AdA-OH, Phosphatidylethanolamine-Adrenal Acid Alcohol; Glu,
Glutamic Acid; Cys, Cysteine; GSH, Glutathione; GSSG, Glutathione Disulfide; GPX4, Glutathione
Peroxidase 4; ACOT2, acyl-CoA thioesterase 2; StAR, steroidogenic acute regulatory; PKC
β
II, protein
kinase C βII.
Int. J. Mol. Sci. 2023,24, 10021 6 of 17
Recently, Leslie Magtanong et al., discovered that ACSL4 serves as a context-specific
regulator of ferroptosis. Through an overview of previous studies, Magtanong high-
lighted ACSL4’s role in inducing ferroptosis, primarily attributed to its inhibitory effect
on GPX4 [
51
]. The relationship between ACSL4 and GPX4 has been a prominent area of
investigation in ferroptosis research. For instance, the team led by Bo Chu demonstrated
that ACSL4 is necessary in ferroptosis induced by erastin or GPX4 inhibitors, whereas it
is dispensable in P53-mediated ferroptosis [
52
]. Shui et al., also reported that lipids can
be directly generated through photodynamic therapy (PDT) with exogenous oxygen radi-
cals, initiating lipid peroxidation independent of ACSL4 and lipoxygenases (ALOXs) [53].
Furthermore, Pang et al., identified that edaravone can alleviate spinal cord injury by
modulating the GPX4/ACSL4/5-LOX pathway [
54
]. Li et al., discovered that baicalein
improves cerebral ischemia-reperfusion injury through the GPX4/ACSL4/ACSL3 axis [
55
].
Wang et al., demonstrated that Seco Lupan Triterpen Derivatives induce ferroptosis via
the GPX4/ACSL4 axis [
56
]. These findings provide a theoretical foundation for further
elucidating the mechanisms of ferroptosis.
4. ACSL4 in Neurological Diseases and Injuries
4.1. ACSL4 in Brain Injury
Traumatic Brain Injury (TBI) is one of the world’s most serious health problems
with high morbidity and mortality [
57
,
58
]. TBI and its complications place an enormous
economic burden on families and society [
59
62
], and an increasing number of studies
have shown that ACSL4 plays an important role in the process of ferroptosis induced
after TBI [
63
]. ACSL4 turns membrane phospholipids into AA/AdA-CoA, which is the
initial step to lipid peroxides [
64
]. Hogan discovered the elevated level of PUFAs in
TBI, and that the occurrence of lipid peroxidation-mediated injury is associated with
brain injury [
65
]. Xiao found that, 6 h after controlled cortical injury (CCI), the mRNA
level of ACSL4 increased [
66
], and a significant increase in ACSL4 was observed after
injury according to Kenny [
67
] (Table 1). However, as biomarkers related to ferroptosis,
GPX4 and ACSL4 [
68
,
69
] were differentially expressed only in the early post-TBI period,
suggesting that the most active stage of ferroptosis may occur early after injury. Using
baicalein could abate PE oxidation and provided histological and cognitive protection
in postinjury [
67
,
70
,
71
] (Table 1). Furthermore, the application of ferroptosis inhibitors
ferristatin-1 and ferristatin II in the TBI mouse model can inhibit iron deposition, neuronal
degeneration, and reduce brain injury of TBI [
72
], which testifies the existence of ferroptosis
in TBI. As previously discussed, the accumulation of oxidized AA- or AdA-containing PE
leads to ferroptosis. Therefore, inhibition of ACSL4 and thus the formation of AA- and
AdA-esterified PE may also protect against TBI.
Moreover, knockdown of ACSL4 by specific shRNA inhibited erastin-induced ferrop-
tosis in HepG2 and HL60 cells (ferroptosis-sensitive cells) [
69
]. The inhibition of ACSL4
expression by shRNA only reduced MDA production, thus reducing the final production
of lipid peroxidation, while Fe
2+
did not accumulate in HepG2 and HL60 cells after erastin
treatment. These findings suggest that ACSL4 induces neuronal ferroptosis by regulating
lipid peroxidation rather than iron accumulation.
Int. J. Mol. Sci. 2023,24, 10021 7 of 17
Table 1. Models of neurodegenerative diseases and outcomes after intervention.
Diseases Biological Model Intervention Measure Consequence Reference
Traumatic Brain Injury
Controlled cortical impact
(CCI) /ACSL4 expression level
increased [66]
CCI Baicalein Decreased ferroptotic PE
oxidation [67]
Ischemic stroke
Middle cerebral artery
occlusion (MCAO) /ACSL4 increased after
decreasing 1–3 h of ischemia [46]
MCAO liproxstatin-1/Rosiglitazo-
ne/
Lipid peroxidation index
was significantly inhibited in
comparison with untreated
group
[73]
Hemorrhagic stroke
Oxygen and glucose
deprivation (OGD) /ACSL4 mRNA expression
was significantly increased [46]
OGD Paeonol Paeonol inhibited the
expression of ACSL4 [50]
Alzheimer’s disease APPswe transgenic mice /
Aβaccumulates in brain
tissue due to lipid
peroxidation
[74]
APPswe/PSEN1dE9
(APP/PS1) double
transgene mice
tetrahydroxy stilbene
glycoside (TSG)
TSG inhibited the expression
of ACSL4 [75]
Parkinson’s disease PD mice model
1-methyl-4-phenyl-1,2,3,6-
tetrahydropyridine
(MPTP)
The expression of ACSL4
significantly increased [76]
PD mice model β-hydroxybutyrate (BHB) BHB inhibits ferroptosis in
PD model [77]
Spinal cord injury Spinal cord Edaravone Reduces ACSL4 levels [54]
contusion injury model
Multiple sclerosis Experimental autoimmune
encephalitis (EAE) model ACSL4-KO
Knocking down the ACSL4
gene considerably reduced
the severity of EAE and the
clinical score of EAE mice
[78]
4.2. ACSL4 in Stroke
4.2.1. ACSL4 in Ischemic Stroke
Ischemic stroke is currently one of the leading causes of human mortality, accounting
for 80% of all strokes [
79
]. Currently, the only treatment options for patients with ischemic
stroke are surgery or thrombolysis with tissue plasminogen activator, but the prognosis
remains poor [
80
]. Recent studies have found that ferroptosis is closely related to the onset
and development of stroke, which may be a potential direction for stroke treatment [81].
The ferroptosis inhibitors liproxstatin-1 and ferrostatin-1 could prevent cerebral is-
chemia reperfusion injury induced by stroke in mice [
82
]. To observe the temporal pattern
of ACSL4 expression after focal ischemia, Cui et al., subjected mice to transient middle cere-
bral artery occlusion (tMCAO) for 1 h followed by reperfusion. The expression of ACSL4 in
the ipsilateral cortex decreased significantly after 1 to 3 h of ischemia and was higher than
that in the contralateral cortex after 6 h of reoxygenation. This suggests that, in the early
stages of focal ischemia, the expression of ACSL4 is down-regulated [
46
]. Hypoxia-inducing
factor 1-alpha (HIF-1
α
) mediated decreased ACSL4 expression after oxygen and glucose
deprivation (OGD). Knockdown of ACSL4 can alleviate ischemic brain damage, and the
overexpression of ACSL4 can exacerbate ischemic brain damage [
83
,
84
]. Chen et al., estab-
lished a transient ischemic model in mice with middle cerebral artery occlusion (MCAO)
after intravenous administration of rosiglitazone 1 h before MCAO. After inhibition of
ACSL4 with rosiglitazone (RSG), the decrease of GPX4 was greatly attenuated (Table 1).
Neurological function was significantly improved at 72 h after stroke, and cerebral infarct
volume was reduced. This study demonstrated that inhibition of ACSL4 could promote
recovery of neurological function after stroke by inhibiting ferroptosis [
73
]. Tuo et al., found
that, during the period of I/R, reduction of ACSL4 could be the result of modification after
translation. They also discovered that ACSL4 can mediate thrombin cytotoxicity which can
be blocked by the ACSL4 inhibitor pioglitazone (PIO). These results suggest that thrombin
Int. J. Mol. Sci. 2023,24, 10021 8 of 17
may contribute to neuronal cell death through the promotion of ACSL4-dependent ferrop-
tosis, and that reduction of ACSL4 may contribute to the inhibition of thrombin-induced
ferroptosis [49].
4.2.2. ACSL4 in Hemorrhagic Stroke
Intracerebral hemorrhage (ICH) is one of the most common and refractory diseases
in the world [
85
]. The hematoma after intracerebral hemorrhage causes progressive brain
tissue damage [
86
], and it is closely associated with ferroptosis during its development [
87
].
ACSL4 mRNA expression was significantly increased in brain microvascular endothelial
cells (BMVECs) treated with the hypothermic oxygen–glucose deprivation intracerebral
hemorrhage model cells (OGD/H). ACSL4 inhibits miR-106b-5p, promoting ferroptosis.
Target gene analysis identified ACSL4 as a target gene of miR-106b-5p in OGD/H ICH
model cells [
46
] (Table 1). The overexpression of ACSL4 countered the effects of miR-
106b-5p, suppressed the viability of ICH cells, and stimulated ferroptosis. These results
suggest that ACSL4 promotes ferroptosis, decreasing the cellular function of BMVECs,
which is consistent with the findings of Xie et al. [
88
]. Furthermore, H19 acts as a competing
endogenous RNA ceRNA and regulates the proliferation and ferroptosis of BMVECs
through the miR-106b-5p/ACSL4 axis [
81
,
82
]. H19 knockdown may prevent ICH by
regulating miR-106b-5p/ACSL4, making this axis a potential therapeutic target for ICH
treatment.
Paeonol (PAN, 2
0
-hydroxy-4
0
-methoxy acetophenone) is a natural product isolated
from Paeoniflora [
89
]. Zheng’s team used hemin to mimic ICH in HT22 cells and found that
hemin significantly up-regulated ACSL4 expression in neuronal cells, while PAN partially
reversed this phenomenon. Additionally, RNA pull-down experiments identified UPF1 and
ACSL4 as downstream targets of HOTAIR in ICH, and PAN could inhibit ICH progression
by mediating the HOTAIR/UPF1/ACSL4 axis, which may serve as a new medicine for
cerebral hemorrhage [50,89] (Table 1).
In recent years, the role of ferroptosis in early brain injury (EBI) of subarachnoid
hemorrhage (SAH) has been highlighted. Ferroptosis is involved in the pathogenesis
of EBI after SAH through various pathways, including the activation of ACSL4, iron
metabolism disorders [
90
], and the down-regulation of GPX4 and ferroptosis suppressor
protein 1 (FSP1) [
91
]. Western blot and immunofluorescence experiments have confirmed
the expression level of ACSL4 in brain tissue after SAH, which increases and then decreases.
The immunofluorescence assay also revealed the colocalization of ACSL4 with the neuronal
marker NEUN in the brain, which significantly increased 24 h after SAH [
92
]. Using
siRNA technology to silence ACSL4 expression, inflammation, blood–brain barrier (BBB)
damage, oxidative stress, cerebral edema, behavioral and cognitive deficits, and neuronal
death were reduced, while the number of surviving neurons increased. Similar results
were obtained with ferroptosis inhibitors [
27
]. Therefore, early intervention to reduce the
oxidative response and ferroptosis may be an effective treatment for SAH. For example,
puerarin can activate SIRT1 or the AMPK/PGC1
α
/NRF2 pathway to alleviate oxidative
stress and reduce ferroptosis in EBI after SAH [93].
4.3. ACSL4 in Alzheimer’s Disease
Alzheimer’s disease (AD) is a degenerative disease of the central nervous system
that primarily affects people over the age of 65 [
94
]. Its clinical manifestations are mainly
the decline of memory, language, and other cognitive abilities. The main pathological
features are amyloid beta peptide (A
β
) [
95
,
96
] as the core component of senile plaques and
neurofibrillary tangles caused by tau hyperphosphorylation [97,98]. Under the increasing
aging trend, due to the unclear pathogenesis of Alzheimer’s disease and the lack of cost-
effective clinical treatment, Alzheimer’s disease places a heavy burden on patients and
medical social security [97,99].
Lipids constitute a vital component of the brain, comprising approximately 40% to 75%
of its dry weight and accounting for up to 80% of the myelin sheath. They play crucial roles
Int. J. Mol. Sci. 2023,24, 10021 9 of 17
in energy metabolism, signal transduction, and various other processes [
100
]. In the context
of AD, the elevated presence of free radicals leads to lipid peroxidation, which is closely
associated with the initial pathological changes observed in AD [
101
]. Furthermore, there is
evidence linking ROS to brain damage in AD [102]. Previous studies have shown that the
level of total free fatty acids in the hippocampus of AD patients is significantly decreased
and the level of ACSL4 is significantly increased. Furthermore, high levels of free MDA
and 4-hydroxynonenal (4-HNE) are detected in several brain regions, and GPX4 expression
is down-regulated, proving the existence of ferroptosis and lipid peroxidation in AD
brain [
103
]. The abnormal folding and aggregation of A
β
in the brain is one of the hallmark
pathological changes of Alzheimer’s disease [
104
]. It has been reported that A
β
oligomer
can cause long-term enhancement impairment in the hippocampus of experimental rats,
and abnormally activate microglia proinflammatory phenotype and complement system,
inducing neuroinflammation and synaptic loss. A
β
is potentially associated with lipid
peroxidation in ferroptosis [
104
], it has the ability to integrate into the lipid bilayer of
neurons, leading to the production of hydrogen peroxide. However, in the presence of
oxidation-reducing metal ions, such as Fe
2+
, a Fenton reaction can occur, resulting in the
generation of a substantial amount of ROS that further target unsaturated lipids. This
exacerbates oxidative damage to lipids, proteins, and DNA [
105
]. Praticòet al., observed
the accumulation of Aβthrough lipid peroxidation and oxidative stress in an APP mouse
model [
74
]. Gao et al., conducted experiments using tetrahydroxy stilbene glycoside (TSG)
on mouse models of AD and found that TSG reduced the formation and accumulation of
A
β
(Table 1). Furthermore, compared to the non-intervention group, the TSG-treated group
exhibited a certain reduction in indices associated with lipid peroxidation in ferroptosis [
75
].
4.4. ACSL4 in Parkinson’s Disease
Parkinson’s disease (PD) is a widespread chronic degenerative disorder that commonly
affects motor skills, language, and other functions of the central nervous system [
106
,
107
].
PD is characterized by the damage to dopamine (DA) neurons in the pars compactus
nigra (SNpc), which may result in muscle rigidity, static tremors, sleep disturbances, motor
retardation, abnormal postural reflexes, sensory disturbances, autonomic nervous system
dysfunction, and other clinical symptoms [76].
Through the assessment of PD patients, it has been observed that the iron content
in glial and dopaminergic neurons is abnormally elevated compared to that in healthy
individuals, and this elevation is positively associated with the severity of PD [
108
110
].
Research has identified the formation of Lewy bodies, composed mainly of
α
-synuclein
nucleoprotein [
111
,
112
], as a distinctive hallmark of PD, occurring within the cytoplasm
of substantia nigra neurons [
113
,
114
]. Notably,
α
-synuclein exhibits a strong affinity for
lipid binding. In addition,
α
-synuclein not only mediates the formation of membrane
PUFAs [115], but also regulate the metabolism of AA [77]. ACSL4 demonstrates a specific
preference for AA, and PUFA has the ability to induce α-synuclein aggregation [116,117].
Recent research has indicated that the inhibition of SP1 can confer neuroprotective
effects in PD models [
118
]. Additionally, Ma et al., demonstrated that SP1 has the abil-
ity to reverse the impact of repressor element-1 silencing transcription factor (REST) on
erastin-induced LUHMES cell viability, ROS production, ferroptosis, and neuronal damage,
implying that REST may alleviate PD by reducing SP1 activity [
118
]. Additionally, the
study revealed that the overexpression of REST down-regulates ACSL4 in erastin-induced
LUHMES cells [
118
]. The interaction between miR-494-3p and ACSL4, REST, or SP1 was ex-
amined using luciferin chromatin immunoprecipitation or EMSA. The results revealed that
the repression of miR-494-3p could prevent ferroptosis and neuronal damage by regulating
the SP1/ACSL4 axis in PD by targeting REST.REST is a downstream gene of miR-494-3p,
which can inhibit ferroptosis neuronal damage induced by SP1, ROS, and mitochondrial
damage in LUHMES cells [
118
]. Furthermore, Song et al., discovered that 1-methyl-4-
phenyl-1,2,3,6-tetrahydropyridine (MPTP) treatment significantly up-regulates ACSL4
expression and down-regulates GPX4 expression in PD mice (Table 1). Apoferritin [
119
]
Int. J. Mol. Sci. 2023,24, 10021 10 of 17
treatment leads to reduced ASCL4 expression and increased expression of ferroptosis sup-
pressor protein 1 (FSP1) [
76
]. Moreover, Yu et al., demonstrated that
β
-hydroxybutyrate
(BHB) directly affects the stability of ACSL4 mRNA through zinc finger protein 36 (ZFP36),
exerting inhibitory effects on ferroptosis [
77
] (Table 1). The aforementioned studies offer
insights for potential future PD treatments by targeting ACSL4 to inhibit ferroptosis [
109
].
4.5. ACSL4 in Spinal Cord Diseases
Spinal cord injury causes permanent or temporary changes in the function of the spinal
cord which can be divided into traumatic spinal cord injury (TSCI) and nontraumatic spinal
cord injury (NTSCI) [
120
]. The main symptoms include sensory–motor or autonomic nerve
dysfunction below the level of the spinal cord injury. TSCI is usually caused by external
physical shocks, such as car accidents, falls, sports, falling objects, or violent activities.
Globally, with the popularity of modern cars, the rise of various outdoor sports, and the
growth of the aging population, the incidence of TSCI presents an increasing and aging
trend.
Amyotrophic lateral sclerosis (ALS) is a neurodegenerative disorder characterized by
the progressive degeneration of motor neurons in the central nervous system, including
the brain and spinal cord. This degeneration leads to muscle paralysis, atrophy, and
functional impairment [
121
]. Although ALS and TSCI are distinct diseases, they share
certain similarities [
122
]. In rare cases, ALS can cause spinal cord injury, while TSCI can
result in motor neuron injury resembling ALS. Furthermore, both conditions are associated
with neurological damage and dysfunction, significantly impacting the affected individuals’
lives [
123
]. Edaravone shows promise as a potential therapeutic intervention by preventing
ferroptosis in ALS [124].
Edaravone, also known as 3-methyl-1-phenyl-2-pyrazolin-5-one, is a free radical
scavenger due to the lipophilicity of phenylmethyl, which allows edaravone to remain on
the membrane and scavenge lipid-reactive oxygen species [
125
,
126
]. By scavenging free
radicals, edaravone has the potential to mitigate oxidative stress and inhibit the activation of
ACSL4, and Yilin Pang and colleagues demonstrated that edaravone inhibits the ferroptosis
pathway following spinal cord injury in a contusion injury model [
54
]. 5-LOX and ACSL4
increased 2 days after injury, while edaravone significantly down-regulated their expression
and up-regulated GPX4/xCT in the acute phase of spinal cord injury [
54
]. There was no
significant change in the expression of 5-LOX and ACSL4 in each group 7 days after SCI,
suggesting that ferroptosis mainly occurred in the acute phase [
54
]. Edaravone regulates
GPX4/ ACSL4/5-LOX in the lower spinal segment of the lesion, and ACSL4 is expressed
in both the nucleus and cytoplasm of the injured spinal cord [54] (Table 1).
In addition, abnormalities in mitochondrial function and morphology have been ob-
served in ALS. In the context of neurodegenerative diseases, edaravone has demonstrated
a protective effect on mitochondria [
127
]. However, the precise mechanism underlying this
effect remains unclear. Considering that ACSL4 is localized to mitochondria-associated
membranes (MAMs), it is plausible that edaravone’s action on mitochondrial integrity
may prevent ACSL4 dysregulation or degradation [
128
]. Additionally, edaravone has been
shown to possess anti-inflammatory properties by inhibiting the production of inflam-
matory mediators. By attenuating the inflammatory response, edaravone may indirectly
modulate ACSL4 levels and potentially impact the pathogenesis of TSCI [129].
4.6. ACSL4 in Multiple Sclerosis
Multiple sclerosis (MS) is a disease characterized by inflammatory demyelination,
which involves the infiltration of immune cells into the central nervous system
(CNS)
[130,131]
, leading to recurrent demyelinating lesions with varying degrees of inflam-
mation, including inflammation throughout the entire lesion area, limited to the lesion
border or lack, and all of these were observed in MS patients [
132
]. Moreover, MS may cause
ongoing neurodegeneration (secondary progression), which leads to cumulative disability
over time [
133
]. To date, treatment of MS has reduced the frequency of relapses without
Int. J. Mol. Sci. 2023,24, 10021 11 of 17
affecting secondary progression. Iron acts as a co-factor for several enzymes that maintain
oligodendrocyte and myelin health, and may play a crucial role in remyelination [
134
].
Aberrant iron regulation in multiple sclerosis (MS) has been observed through magnetic
resonance imaging (MRI) and histological examinations, revealing iron deposition in gray
matter and a decrease in normal white matter [
132
]. These findings suggest a potential
association between MS and ferroptosis.
Given that MS is an immune-mediated disease with similarities to autoimmune dis-
orders, several drugs for MS have been developed using the experimental autoimmune
encephalitis (EAE) model. Genes implicated in ferroptosis were examined in the spinal cord
of EAE mice. The results indicated significant alterations in key ferroptosis-related genes,
including ACSL4 and GPX4. Furthermore, elevated levels of ACSL4 were detected prior to
the onset of clinical symptoms in EAE mice, and remained high in the chronic active areas
of MS patients. Notably, during the peak of EAE, the expression of ACSL4 was significantly
increased [
135
]. Knocking down the ACSL4 gene considerably reduced the severity of EAE
and the clinical score of EAE mice, indicating that ACSL4-mediated ferroptosis provoked
inflammation and promoted T-cell activation and CNS infiltration (Table 1). Therefore, the
inhibition of ACSL4 suppresses ferroptosis, which provides a potential therapeutic target
for the treatment of secondary neurodegeneration, but further clinical trials are needed to
test the efficacy of the drug [78,136].
5. Conclusions
Ferroptosis is an iron-dependent and novel form of regulated cell death in which lipid
peroxide levels accumulate to lethal levels. A variety of diseases, including nervous system
disorders and injuries, are associated with ferroptosis. According to reports on ferroptosis,
ACSL4 is one of the important enzymes in the ferroptosis pathway, which can be used as a
biomarker for ferroptosis and can promote ferroptosis. On the one hand, it can promote
tumor cell death; on the other hand, it also illustrates its role in disease-induced ferroptosis.
The pro-ferroptosis effect of ACSL4 was mainly due to its critical role in AA and AdA
metabolism and lipid peroxidation. Due to the different content and distribution of fatty
acids in different tumor cells and different tissue cells, as well as the different distribution
and content of ACSL4 and other related lipid metabolism enzymes, the sensitivity of
different tissues and different cells to ferroptosis varies greatly [
135
,
137
]. For example,
ACSL4 is highly expressed in patients with liver cancer and colon cancer, and the higher the
expression level is, the worse the prognosis is, while patients with gastric cancer [
138
] tend
to have a low expression of ACSL4. This is a very important clue for how to use ferroptosis
to inhibit tumor cells or inhibit ferroptosis to improve the prognosis of neurological diseases.
At present, it is urgent to accelerate the process of ferroptosis treatment of tumors without
toxic side effects on normal tissues through the pathological tissue analysis of tumors, or by
further exploring the time window of ACSL4 inhibitors or other ferroptosis inhibitors after
ischemia-reperfusion injury [
139
141
]. In addition, the possibility that ACSL4 plays a role
in genetic disorders cannot be ignored, as ACSL4 deletion mutations have been reported
in a family with Alport disease (also known as eye-ear-kidney syndrome). Moreover, the
specifics of ACSL4 deletion and complex disorders remain to be investigated. Finally, there
are still few studies on the lipid peroxidation pathway associated with ACSL4 in TBI. As a
type of injury with a high mortality rate, the role of ACSL4 in TBI needs to be further and
more widely explored.
Author Contributions:
B.J., J.L., Y.S. and C.L. carried out the studies of literature research and
performed the acquisition of data and writing in manuscript preparation. B.J., J.L. and Y.S. proofread
the manuscript and gave important comments from their area of expertise. C.L. supervised the
development of work, gave critical revisions, and edited the manuscript. All authors have read and
agreed to the published version of the manuscript.
Funding:
This work was supported by the National Natural Science Foundation of China (82271409,
81971163), the outstanding young backbone teacher of the “Blue Project” in Jiangsu Province
Int. J. Mol. Sci. 2023,24, 10021 12 of 17
(SR13450121), a Project Funded by the Priority Academic Program Development of Jiangsu Higher
Education Institutions (PAPD), the Key Program of Educational Commission of Anhui Province
(KJ2021A0826), and National Training Program of Innovation and Entrepreneurship for Undergradu-
ate (2021suda046).
Institutional Review Board Statement: Not applicable.
Informed Consent Statement: Not applicable.
Data Availability Statement:
Data sharing is not applicable to this article as no datasets were
generated or analyzed during the current study.
Conflicts of Interest: The authors declare that they have no competing interest.
References
1.
Galluzzi, L.; Vitale, I.; Aaronson, S.A.; Abrams, J.M.; Adam, D.; Agostinis, P.; Alnemri, E.S.; Altucci, L.; Amelio, I.; Andrews, D.W.;
et al. Molecular mechanisms of cell death: Recommendations of the Nomenclature Committee on Cell Death 2018. Cell Death
Differ. 2018,25, 486–541. [CrossRef]
2.
Dixon, S.J.; Lemberg, K.M.; Lamprecht, M.R.; Skouta, R.; Zaitsev, E.M.; Gleason, C.E.; Patel, D.N.; Bauer, A.J.; Cantley, A.M.; Yang,
W.S.; et al. Ferroptosis: An iron-dependent form of nonapoptotic cell death. Cell 2012,149, 1060–1072. [CrossRef]
3.
Jiang, X.; Stockwell, B.R.; Conrad, M. Ferroptosis: Mechanisms, biology and role in disease. Nat. Rev. Mol. Cell Biol.
2021
,22,
266–282. [CrossRef]
4.
Tang, Y.; Zhou, J.; Hooi, S.C.; Jiang, Y.; Lu, G. Fatty acid activation in carcinogenesis and cancer development: Essential roles of
long-chain acyl-CoA synthetases. Oncol. Lett. 2018,16, 1390–1396. [CrossRef] [PubMed]
5.
Quan, J.; Bode, A.M.; Luo, X. ACSL family: The regulatory mechanisms and therapeutic implications in cancer. Eur. J. Pharmacol.
2021,909, 174397. [CrossRef] [PubMed]
6.
Kuwata, H.; Hara, S. Role of acyl-CoA synthetase ACSL4 in arachidonic acid metabolism. Prostaglandins Other Lipid Mediat.
2019
,
144, 106363. [CrossRef] [PubMed]
7.
Radif, Y.; Ndiaye, H.; Kalantzi, V.; Jacobs, R.; Hall, A.; Minogue, S.; Waugh, M.G. The endogenous subcellular localisations of the
long chain fatty acid-activating enzymes ACSL3 and ACSL4 in sarcoma and breast cancer cells. Mol. Cell. Biochem.
2018
,448,
275–286. [CrossRef] [PubMed]
8.
Hou, J.; Jiang, C.; Wen, X.; Li, C.; Xiong, S.; Yue, T.; Long, P.; Shi, J.; Zhang, Z. ACSL4 as a Potential Target and Biomarker for
Anticancer: From Molecular Mechanisms to Clinical Therapeutics. Front. Pharmacol. 2022,13, 949863. [CrossRef]
9.
Fujino, T.; Kang, M.; Suzuki, H.; Iijima, H.; Yamamoto, T. Molecular Characterization and Expression of Rat Acyl-CoA Synthetase
3. J. Biol. Chem. 1996,271, 16748–16752. [CrossRef]
10.
Cao, Y.; Traer, E.; Zimmerman, G.A.; McIntyre, T.M.; Prescott, S.M. Cloning, Expression, and Chromosomal Localization of
Human Long-Chain Fatty Acid-CoA Ligase 4 (FACL4). Genomics 1998,49, 327–330. [CrossRef]
11.
Trautenberg, L.C.; Brankatschk, M.; Shevchenko, A.; Wigby, S.; Reinhardt, K. Ecological lipidology. Elife
2022
,11, e79288.
[CrossRef] [PubMed]
12.
Watkins, P.A.; Maiguel, D.; Jia, Z.; Pevsner, J. Evidence for 26 distinct acyl-coenzyme A synthetase genes in the human genome. J.
Lipid Res. 2007,48, 2736–2750. [CrossRef] [PubMed]
13.
Yang, Y.; Zhu, T.; Wang, X.; Xiong, F.; Hu, Z.; Qiao, X.; Yuan, X.; Wang, D. ACSL3 and ACSL4, Distinct Roles in Ferroptosis and
Cancers. Cancers 2022,14, 5896. [CrossRef] [PubMed]
14. Fujimoto, Y.; Itabe, H.; Kinoshita, T.; Homma, K.J.; Onoduka, J.; Mori, M.; Yamaguchi, S.; Makita, M.; Higashi, Y.; Yamashita, A.;
et al. Involvement of ACSL in local synthesis of neutral lipids in cytoplasmic lipid droplets in human hepatocyte HuH7. J. Lipid
Res. 2007,48, 1280–1292. [CrossRef] [PubMed]
15.
Zhao, J.; Zhang, H.; Fan, X.; Yu, X.; Huai, J. Lipid Dyshomeostasis and Inherited Cerebellar Ataxia. Mol. Neurobiol.
2022
,59,
3800–3828. [CrossRef]
16.
Ri, K.; Lee-Okada, H.; Yokomizo, T. Omega-6 highly unsaturated fatty acids in Leydig cells facilitate male sex hormone production.
Commun. Biol. 2022,5, 1001. [CrossRef]
17.
Castillo, A.F.; Orlando, U.D.; Maloberti, P.M.; Prada, J.G.; Dattilo, M.A.; Solano, A.R.; Bigi, M.M.; Ríos Medrano, M.A.; Torres,
M.T.; Indo, S.; et al. New inhibitor targeting Acyl-CoA synthetase 4 reduces breast and prostate tumor growth, therapeutic
resistance and steroidogenesis. Cell. Mol. Life Sci. 2021,78, 2893–2910. [CrossRef]
18.
Killion, E.A.; Reeves, A.R.; El Azzouny, M.A.; Yan, Q.; Surujon, D.; Griffin, J.D.; Bowman, T.A.; Wang, C.; Matthan, N.R.; Klett,
E.L.; et al. A role for long-chain acyl-CoA synthetase-4 (ACSL4) in diet-induced phospholipid remodeling and obesity-associated
adipocyte dysfunction. Mol. Metab. 2018,9, 43–56. [CrossRef]
19.
Golej, D.L.; Askari, B.; Kramer, F.; Barnhart, S.; Vivekanandan-Giri, A.; Pennathur, S.; Bornfeldt, K.E. Long-chain acyl-CoA
synthetase 4 modulates prostaglandin E2 release from human arterial smooth muscle cells. J. Lipid Res.
2011
,52, 782–793.
[CrossRef]
20.
Askari, B.; Kanter, J.E.; Sherrid, A.M.; Golej, D.L.; Bender, A.T.; Liu, J.; Hsueh, W.A.; Beavo, J.A.; Coleman, R.A.; Bornfeldt,
K.E. Rosiglitazone Inhibits Acyl-CoA Synthetase Activity and Fatty Acid Partitioning to Diacylglycerol and Triacylglycerol
Int. J. Mol. Sci. 2023,24, 10021 13 of 17
via a Peroxisome Proliferator–Activated Receptor-
γ
–Independent Mechanism in Human Arterial Smooth Muscle Cells and
Macrophages. Diabetes 2007,56, 1143–1152. [CrossRef]
21.
Küch, E.; Vellaramkalayil, R.; Zhang, I.; Lehnen, D.; Brügger, B.; Stremmel, W.; Ehehalt, R.; Poppelreuther, M.; Füllekrug, J. Differ-
entially localized acyl-CoA synthetase 4 isoenzymes mediate the metabolic channeling of fatty acids towards phosphatidylinositol.
Biochim. Et Biophys. Acta (BBA) Mol. Cell Biol. Lipids 2014,1841, 227–239. [CrossRef] [PubMed]
22.
Wang, W.; Hao, X.; Han, L.; Yan, Z.; Shen, W.; Dong, D.; Hasbargen, K.; Bittner, S.; Cortez, Y.; Greenberg, A.S.; et al. Tissue-Specific
Ablation of ACSL4 Results in Disturbed Steroidogenesis. Endocrinology 2019,160, 2517–2528. [CrossRef] [PubMed]
23.
Cornejo Maciel, F.; Maloberti, P.; Neuman, I.; Cano, F.; Castilla, R.; Castillo, F.; Paz, C.; Podestá, E.J. An arachidonic acid-preferring
acyl-CoA synthetase is a hormone-dependent and obligatory protein in the signal transduction pathway of steroidogenic
hormones. J. Mol. Endocrinol. 2005,34, 655–666. [CrossRef] [PubMed]
24.
Szczuko, M.; Kikut, J.; Komorniak, N.; Bilicki, J.; Celewicz, Z.; Zietek, M. The Role of Arachidonic and Linoleic Acid Derivatives
in Pathological Pregnancies and the Human Reproduction Process. Int. J. Mol. Sci. 2020,21, 9628. [CrossRef]
25.
Kuwata, H.; Hara, S. Inhibition of long-chain acyl-CoA synthetase 4 facilitates production of 5, 11-dihydroxyeicosatetraenoic acid
via the cyclooxygenase-2 pathway. Biochem. Biophys. Res. Commun. 2015,465, 528–533. [CrossRef]
26.
Doll, S.; Proneth, B.; Tyurina, Y.Y.; Panzilius, E.; Kobayashi, S.; Ingold, I.; Irmler, M.; Beckers, J.; Aichler, M.; Walch, A.; et al.
ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat. Chem. Biol. 2017,13, 91–98. [CrossRef]
27.
Qu, X.F.; Liang, T.Y.; Wu, D.G.; Lai, N.S.; Deng, R.M.; Ma, C.; Li, X.; Li, H.Y.; Liu, Y.Z.; Shen, H.T.; et al. Acyl-CoA synthetase long
chain family member 4 plays detrimental role in early brain injury after subarachnoid hemorrhage in rats by inducing ferroptosis.
CNS Neurosci. Ther. 2021,27, 449–463. [CrossRef]
28.
Kagan, V.E.; Mao, G.; Qu, F.; Angeli, J.P.F.; Doll, S.; Croix, C.S.; Dar, H.H.; Liu, B.; Tyurin, V.A.; Ritov, V.B.; et al. Oxidized
arachidonic and adrenic PEs navigate cells to ferroptosis. Nat. Chem. Biol. 2017,13, 81–90. [CrossRef]
29.
Cho, Y.; Kang, M.; Sone, H.; Suzuki, T.; Abe, M.; Igarashi, M.; Tokunaga, T.; Ogawa, S.; Takei, Y.A.; Miyazawa, T.; et al. Abnormal
Uterus with Polycysts, Accumulation of Uterine Prostaglandins, and Reduced Fertility in Mice Heterozygous for Acyl-CoA
Synthetase 4 Deficiency. Biochem. Biophys. Res. Commun. 2001,284, 993–997. [CrossRef]
30.
Chen, W.; Wang, C.; Hung, Y.; Weng, T.; Yen, M.; Lai, M. Systematic Analysis of Gene Expression Alterations and Clinical
Outcomes for Long-Chain Acyl-Coenzyme A Synthetase Family in Cancer. PLoS ONE 2016,11, e155660. [CrossRef]
31.
Brown, C.W.; Amante, J.J.; Goel, H.L.; Mercurio, A.M. The
α
6
β
4 integrin promotes resistance to ferroptosis. J. Cell Biol.
2017
,216,
4287–4297. [CrossRef] [PubMed]
32.
Nguyen, K.T.; Mun, S.; Yang, J.; Lee, J.; Seok, O.; Kim, E.; Kim, D.; An, S.Y.; Seo, D.; Suh, J.; et al. The MARCHF6 E3 ubiquitin
ligase acts as an NADPH sensor for the regulation of ferroptosis. Nat. Cell Biol. 2022,24, 1239–1251. [CrossRef] [PubMed]
33.
Cooke, M.; Orlando, U.; Maloberti, P.; Podestá, E.J.; Maciel, F.C. Tyrosine phosphatase SHP2 regulates the expression of acyl-CoA
synthetase ACSL4. J. Lipid Res. 2011,52, 1936–1948. [CrossRef] [PubMed]
34.
Wu, J.; Minikes, A.M.; Gao, M.; Bian, H.; Li, Y.; Stockwell, B.R.; Chen, Z.; Jiang, X. Intercellular interaction dictates cancer cell
ferroptosis via NF2–YAP signalling. Nature 2019,572, 402–406. [CrossRef] [PubMed]
35.
Zhang, H.; Hu, B.; Li, Z.; Du, T.; Shan, J.; Ye, Z.; Peng, X.; Li, X.; Huang, Y.; Zhu, X.; et al. PKC
β
II phosphorylates ACSL4 to
amplify lipid peroxidation to induce ferroptosis. Nat. Cell Biol. 2022,24, 88–98. [CrossRef]
36.
Stockwell, B.R. Ferroptosis turns 10: Emerging mechanisms, physiological functions, and therapeutic applications. Cell
2022
,185,
2401–2421. [CrossRef]
37. Yang, W.S.; Stockwell, B.R. Ferroptosis: Death by Lipid Peroxidation. Trends Cell Biol. 2016,26, 165–176. [CrossRef]
38.
Rui, T.; Wang, H.; Li, Q.; Cheng, Y.; Gao, Y.; Fang, X.; Ma, X.; Chen, G.; Gao, C.; Gu, Z.; et al. Deletion of ferritin H in neurons
counteracts the protective effect of melatonin against traumatic brain injury-induced ferroptosis. J. Pineal Res.
2021
,70, e12704.
[CrossRef]
39.
He, X.; Li, M.; Ye, Z.; You, X.; Wang, J.; Xiao, X.; Zhu, G.; Wei, J.; Zha, Y. Identification of Piperlongumine as Potent Inhibitor of
Necroptosis. Drug Des. Dev. Ther. 2023,17, 1387–1394. [CrossRef]
40.
Qiu, Y.; Shi, Y.; Zhu, N.; Zhang, S.; Zhang, C.; Gu, J.; He, P.; Dai, A.; Qin, L. A Lipid Perspective on Regulated Pyroptosis. Int. J.
Biol. Sci. 2023,19, 2333–2348. [CrossRef]
41.
Liu, J.; Kuang, F.; Kang, R.; Tang, D. Alkaliptosis: A new weapon for cancer therapy. Cancer Gene Ther.
2020
,27, 267–269.
[CrossRef] [PubMed]
42.
Cheng, Y.; Song, Y.; Chen, H.; Li, Q.; Gao, Y.; Lu, G.; Luo, C. Ferroptosis Mediated by Lipid Reactive Oxygen Species: A Possible
Causal Link of Neuroinflammation to Neurological Disorders. Oxidative Med. Cell. Longev.
2021
,2021, 5005136. [CrossRef]
[PubMed]
43.
Lei, G.; Zhuang, L.; Gan, B. Targeting ferroptosis as a vulnerability in cancer. Nat. Rev. Cancer
2022
,22, 381–396. [CrossRef]
[PubMed]
44.
Yi-di, Z.; Ya-xuan, R.; Jun, Z.; Chu-zhao, L.; Rui-hua, D. The Role and Research Progress of ACSL4 Gene. China Cattle Sci.
2020
,46,
52–58. [CrossRef]
45.
Zhou, L.; Li, F.; Xu, H.; Luo, C.; Wu, H.; Zhu, M.; Lu, W.; Ji, X.; Zhou, Q.; Zhu, D. Treatment of cerebral ischemia by disrupting
ischemia-induced interaction of nNOS with PSD-95. Nat. Med. 2010,16, 1439–1443. [CrossRef]
46.
Cui, Y.; Zhang, Y.; Zhao, X.; Shao, L.; Liu, G.; Sun, C.; Xu, R.; Zhang, Z. ACSL4 exacerbates ischemic stroke by promoting
ferroptosis-induced brain injury and neuroinflammation. Brain Behav. Immun. 2021,93, 312–321. [CrossRef]
Int. J. Mol. Sci. 2023,24, 10021 14 of 17
47.
Kuwata, H.; Yoshimura, M.; Sasaki, Y.; Yoda, E.; Nakatani, Y.; Kudo, I.; Hara, S. Role of long-chain acyl-coenzyme A synthetases
in the regulation of arachidonic acid metabolism in interleukin 1beta-stimulated rat fibroblasts. Biochim. Biophys. Acta
2014
,1841,
44–53. [CrossRef]
48.
Cheng, J.; Fan, Y.Q.; Liu, B.H.; Zhou, H.; Wang, J.M.; Chen, Q.X. ACSL4 suppresses glioma cells proliferation via activating
ferroptosis. Oncol. Rep. 2019,43, 147–158. [CrossRef]
49.
Tuo, Q.Z.; Liu, Y.; Xiang, Z.; Yan, H.F.; Zou, T.; Shu, Y.; Ding, X.L.; Zou, J.J.; Xu, S.; Tang, F.; et al. Thrombin induces ACSL4-
dependent ferroptosis during cerebral ischemia/reperfusion. Signal. Transduct. Target. Ther. 2022,7, 59. [CrossRef]
50.
Jin, Z.; Gao, W.; Liao, S.; Yu, T.; Shi, Q.; Yu, S.; Cai, Y. Paeonol inhibits the progression of intracerebral haemorrhage by mediating
the HOTAIR/UPF1/ACSL4 axis. ASN Neuro 2021,13, 166567834. [CrossRef]
51.
Magtanong, L.; Mueller, G.D.; Williams, K.J.; Billmann, M.; Chan, K.; Armenta, D.A.; Pope, L.E.; Moffat, J.; Boone, C.; Myers, C.L.;
et al. Context-dependent regulation of ferroptosis sensitivity. Cell Chem. Biol. 2022,29, 1409–1418. [CrossRef]
52.
Chu, B.; Kon, N.; Chen, D.; Li, T.; Liu, T.; Jiang, L.; Song, S.; Tavana, O.; Gu, W. ALOX12 is required for p53-mediated tumour
suppression through a distinct ferroptosis pathway. Nat. Cell Biol. 2019,21, 579–591. [CrossRef] [PubMed]
53. Shui, S.; Zhao, Z.; Wang, H.; Conrad, M.; Liu, G. Non-enzymatic lipid peroxidation initiated by photodynamic therapy drives a
distinct ferroptosis-like cell death pathway. Redox Biol. 2021,45, 102056. [CrossRef] [PubMed]
54.
Pang, Y.; Liu, X.; Wang, X.; Shi, X.; Ma, L.; Zhang, Y.; Zhou, T.; Zhao, C.; Zhang, X.; Fan, B.; et al. Edaravone Modulates Neuronal
GPX4/ACSL4/5-LOX to Promote Recovery after Spinal Cord Injury. Front. Cell Dev. Biol.
2022
,10, 849854. [CrossRef] [PubMed]
55.
Li, M.; Meng, Z.; Yu, S.; Li, J.; Wang, Y.; Yang, W.; Wu, H. Baicalein ameliorates cerebral ischemia-reperfusion injury by inhibiting
ferroptosis via regulating GPX4/ACSL4/ACSL3 axis. Chem. Biol. Interact. 2022,366, 110137. [CrossRef] [PubMed]
56.
Wang, H.; Wu, D.; Gao, C.; Teng, H.; Zhao, Y.; He, Z.; Chen, W.; Zong, Y.; Du, R. Seco-Lupane Triterpene Derivatives Induce
Ferroptosis through GPX4/ACSL4 Axis and Target Cyclin D1 to Block the Cell Cycle. J. Med. Chem.
2022
,65, 10014–10044.
[CrossRef]
57.
Feigin, V.L.; Nichols, E.; Alam, T.; Bannick, M.S.; Beghi, E.; Blake, N.; Culpepper, W.J.; Dorsey, E.R.; Elbaz, A.; Ellenbogen, R.G.;
et al. Global, regional, and national burden of neurological disorders, 1990–2016: A systematic analysis for the Global Burden of
Disease Study 2016. Lancet Neurol. 2019,18, 459–480. [CrossRef]
58.
Dewan, M.C.; Rattani, A.; Gupta, S.; Baticulon, R.E.; Hung, Y.C.; Punchak, M.; Agrawal, A.; Adeleye, A.O.; Shrime, M.G.; Rubiano,
A.M.; et al. Estimating the global incidence of traumatic brain injury. J. Neurosurg. 2018,2018, 1–18. [CrossRef]
59.
Maas, A.I.R.; Menon, D.K.; Adelson, P.D.; Andelic, N.; Bell, M.J.; Belli, A.; Bragge, P.; Brazinova, A.; Büki, A.; Chesnut, R.M.;
et al. Traumatic brain injury: Integrated approaches to improve prevention, clinical care, and research. Lancet Neurol.
2017
,16,
987–1048. [CrossRef]
60.
Iaccarino, C.; Carretta, A.; Nicolosi, F.; Morselli, C. Epidemiology of severe traumatic brain injury. J. Neurosurg. Sci.
2018
,62,
535–541. [CrossRef]
61.
Majdan, M.; Plancikova, D.; Maas, A.; Polinder, S.; Feigin, V.; Theadom, A.; Rusnak, M.; Brazinova, A.; Haagsma, J. Years of life
lost due to traumatic brain injury in Europe: A cross-sectional analysis of 16 countries. PLoS Med.
2017
,14, e1002331. [CrossRef]
[PubMed]
62.
Cheng, P.; Yin, P.; Ning, P.; Wang, L.; Cheng, X.; Liu, Y.; Schwebel, D.C.; Liu, J.; Qi, J.; Hu, G.; et al. Trends in traumatic brain
injury mortality in China, 2006–2013: A population-based longitudinal study. PLoS Med.
2017
,14, e1002332. [CrossRef] [PubMed]
63.
Geng, Z.; Guo, Z.; Guo, R.; Ye, R.; Zhu, W.; Yan, B. Ferroptosis and traumatic brain injury. Brain Res. Bull.
2021
,172, 212–219.
[CrossRef] [PubMed]
64.
Gaschler, M.M.; Stockwell, B.R. Lipid peroxidation in cell death. Biochem. Biophys. Res. Commun.
2017
,482, 419–425. [CrossRef]
[PubMed]
65.
Hogan, S.R.; Phan, J.H.; Alvarado-Velez, M.; Wang, M.D.; Bellamkonda, R.V.; Fernández, F.M.; LaPlaca, M.C. Discovery of
Lipidome Alterations Following Traumatic Brain Injury via High-Resolution Metabolomics. J. Proteome Res.
2018
,17, 2131–2143.
[CrossRef]
66.
Xiao, X.; Jiang, Y.; Liang, W.; Wang, Y.; Cao, S.; Yan, H.; Gao, L.; Zhang, L. miR-212-5p attenuates ferroptotic neuronal death after
traumatic brain injury by targeting Ptgs2. Mol. Brain 2019,12, 78. [CrossRef]
67.
Kenny, E.M.; Fidan, E.; Yang, Q.; Anthonymuthu, T.S.; New, L.A.; Meyer, E.A.; Wang, H.; Kochanek, P.M.; Dixon, C.E.; Kagan,
V.E.; et al. Ferroptosis Contributes to Neuronal Death and Functional Outcome After Traumatic Brain Injury. Crit. Care Med.
2019
,
47, 410–418. [CrossRef]
68.
Rui, T.; Li, Q.; Song, S.; Gao, Y.; Luo, C. Ferroptosis-relevant mechanisms and biomarkers for therapeutic interventions in
traumatic brain injury. Histol. Histopathol. 2020,35, 1105–1113. [CrossRef]
69.
Yuan, H.; Li, X.; Zhang, X.; Kang, R.; Tang, D. Identification of ACSL4 as a biomarker and contributor of ferroptosis. Biochem.
Biophys. Res. Commun. 2016,478, 1338–1343. [CrossRef]
70.
Zheng, B.; Zhou, X.; Pang, L.; Che, Y.; Qi, X. Baicalin suppresses autophagy-dependent ferroptosis in early brain injury after
subarachnoid hemorrhage. Bioengineered 2021,12, 7794–7804. [CrossRef]
71.
Chen, S.; Hsu, C.; Huang, W.; Wang, J. Post-injury baicalein improves histological and functional outcomes and reduces
inflammatory cytokines after experimental traumatic brain injury. Br. J. Pharmacol. 2008,155, 1279–1296. [CrossRef] [PubMed]
72.
Li, J.; Jia, B.; Cheng, Y.; Song, Y.; Li, Q.; Luo, C. Targeting Molecular Mediators of Ferroptosis and Oxidative Stress for Neurological
Disorders. Oxidative Med. Cell. Longev. 2022,2022, 3999083. [CrossRef] [PubMed]
Int. J. Mol. Sci. 2023,24, 10021 15 of 17
73.
Chen, J.; Yang, L.; Geng, L.; He, J.; Chen, L.; Sun, Q.; Zhao, J.; Wang, X. Inhibition of Acyl-CoA Synthetase Long-Chain Family
Member 4 Facilitates Neurological Recovery after Stroke by Regulation Ferroptosis. Front. Cell. Neurosci.
2021
,15, 632354.
[CrossRef] [PubMed]
74.
Praticò, D. Increased Lipid Peroxidation Precedes Amyloid Plaque Formation in an Animal Model of Alzheimer Amyloidosis. J.
Neurosci. 2001,21, 4183–4187. [CrossRef] [PubMed]
75.
Gao, Y.; Li, J.; Wu, Q.; Wang, S.; Yang, S.; Li, X.; Chen, N.; Li, L.; Zhang, L. Tetrahydroxy stilbene glycoside ameliorates Alzheimer’s
disease in APP/PS1 mice via glutathione peroxidase related ferroptosis. Int. Immunopharmacol.
2021
,99, 108002. [CrossRef]
[PubMed]
76.
Song, L.M.; Xiao, Z.X.; Zhang, N.; Yu, X.Q.; Cui, W.; Xie, J.X.; Xu, H.M. Apoferritin improves motor deficits in MPTP-treated mice
by regulating brain iron metabolism and ferroptosis. IScience 2021,24, 102431. [CrossRef]
77.
Yu, X.; Yang, Y.; Zhang, B.; Han, G.; Yu, J.; Yu, Q.; Zhang, L. Ketone Body
β
-Hydroxybutyric Acid Ameliorates Dopaminergic
Neuron Injury through Modulating Zinc Finger Protein 36/Acyl-CoA Synthetase Long-Chain Family Member Four Signaling
Axis-Mediated Ferroptosis. Neuroscience 2023,509, 157–172. [CrossRef]
78.
Constantinescu, C.S.; Farooqi, N.; O’Brien, K.; Gran, B. Experimental autoimmune encephalomyelitis (EAE) as a model for
multiple sclerosis (MS). Br. J. Pharmacol. 2011,164, 1079–1106. [CrossRef]
79.
Zhang, H.; Sun, X.; Xie, Y.; Zan, J.; Tan, W. Isosteviol Sodium Protects Against Permanent Cerebral Ischemia Injury in Mice via
Inhibition of NF-
κ
B–Mediated Inflammatory and Apoptotic Responses. J. Stroke Cerebrovasc. Dis.
2017
,26, 2603–2614. [CrossRef]
80. Blakeley, J.O.; Llinas, R.H. Thrombolytic therapy for acute ischemic stroke. J. Neurol. Sci. 2007,261, 55–62. [CrossRef]
81.
Weiland, A.; Wang, Y.; Wu, W.; Lan, X.; Han, X.; Li, Q.; Wang, J. Ferroptosis and Its Role in Diverse Brain Diseases. Mol. Neurobiol.
2019,56, 4880–4893. [CrossRef] [PubMed]
82.
Ahmad, S.; Elsherbiny, N.M.; Haque, R.; Khan, M.B.; Ishrat, T.; Shah, Z.A.; Khan, M.M.; Ali, M.; Jamal, A.; Katare, D.P.; et al.
Sesamin attenuates neurotoxicity in mouse model of ischemic brain stroke. Neurotoxicology
2014
,45, 100–110. [CrossRef] [PubMed]
83.
Hailin, Z.; Yong, C.; Yongfeng, B.A.; Chao, L.U.; Wei, H.U.; Renpeng, Z. Research progress on ferroptosis regulators and their role
in stroke. Chin. J. Clin. Pharmacol. Ther. 2021,26, 1320–1327. [CrossRef]
84.
Xichen, Y.; Simei, Z.; Yongdan, C.; Li, Z.; Chengcai, Z.; Pengyue, Z.; Yaju, J. Research Progress on the Mechanism of Ferroptosis in
Ischemic Stroke. J. Yunnan Univ. Chin. Med. 2022,45, 97–102. [CrossRef]
85.
Morotti, A.; Boulouis, G.; Dowlatshahi, D.; Li, Q.; Shamy, M.; Al-Shahi, S.R.; Rosand, J.; Cordonnier, C.; Goldstein, J.N.;
Charidimou, A. Intracerebral haemorrhage expansion: Definitions, predictors, and prevention. Lancet Neurol.
2023
,22, 159–171.
[CrossRef]
86.
You, M.; Long, C.; Wan, Y.; Guo, H.; Shen, J.; Li, M.; He, Q.; Hu, B. Neuron derived fractalkine promotes microglia to absorb
hematoma via CD163/HO-1 after intracerebral hemorrhage. Cell. Mol. Life Sci. 2022,79, 224. [CrossRef]
87.
Xu, J.; Chen, Z.; Yu, F.; Liu, H.; Ma, C.; Xie, D.; Hu, X.; Leak, R.K.; Chou, S.; Stetler, R.A.; et al. IL-4/STAT6 signaling facilitates
innate hematoma resolution and neurological recovery after hemorrhagic stroke in mice. Proc. Natl. Acad. Sci. USA
2020
,117,
32679–32690. [CrossRef]
88.
Xie, L.; Wang, Y.; Chen, Z. LncRNA Blnc1 mediates the permeability and inflammatory response of cerebral hemorrhage by
regulating the PPAR-γ/SIRT6/FoxO3 pathway. Life Sci. 2021,267, 118942. [CrossRef]
89.
Zhang, L.; Li, D.; Liu, L. Paeonol: Pharmacological effects and mechanisms of action. Int. Immunopharmacol.
2019
,72, 413–421.
[CrossRef]
90.
Liu, H.; Schwarting, J.; Terpolilli, N.A.; Nehrkorn, K.; Plesnila, N. Scavenging Free Iron Reduces Arteriolar Microvasospasms
after Experimental Subarachnoid Hemorrhage. Stroke 2021,52, 4033–4042. [CrossRef]
91.
Kuang, H.; Wang, T.; Liu, L.; Tang, C.; Li, T.; Liu, M.; Wang, T.; Zhong, W.; Wang, Y. Treatment of early brain injury after
subarachnoid hemorrhage in the rat model by inhibiting p53-induced ferroptosis. Neurosci. Lett.
2021
,762, 136134. [CrossRef]
[PubMed]
92.
Yuan, B.; Zhao, X.; Shen, J.; Chen, S.; Huang, H.; Zhou, X.; Han, Y.; Zhou, L.; Lu, X.; Wu, Q. Activation of SIRT1 Alleviates
Ferroptosis in the Early Brain Injury after Subarachnoid Hemorrhage. Oxidative Med. Cell. Longev.
2022
,2022, 9069825. [CrossRef]
[PubMed]
93.
Huang, Y.; Wu, H.; Hu, Y.; Zhou, C.; Wu, J.; Wu, Y.; Wang, H.; Lenahan, C.; Huang, L.; Nie, S.; et al. Puerarin Attenuates Oxidative
Stress and Ferroptosis via AMPK/PGC1
α
/Nrf2 Pathway after Subarachnoid Hemorrhage in Rats. Antioxidants
2022
,11, 1259.
[CrossRef] [PubMed]
94. 2023 Alzheimer’s disease facts and figures. Alzheimer’s Dement. 2023,19, 1598–1695. [CrossRef]
95. Rostagno, A.A. Pathogenesis of Alzheimer’s Disease. Int. J. Mol. Sci. 2022,24, 107. [CrossRef] [PubMed]
96. Jorfi, M.; Maaser-Hecker, A.; Tanzi, R.E. The neuroimmune axis of Alzheimer ’s disease. Genome Med. 2023,15, 6. [CrossRef]
97.
Mullane, K.; Williams, M. Alzheimer’s disease (AD) therapeutics—1: Repeated clinical failures continue to question the amyloid
hypothesis of AD and the current understanding of AD causality. Biochem. Pharmacol. 2018,158, 359–375. [CrossRef]
98. Jellinger, K.A. Pathobiological Subtypes of Alzheimer Disease. Dement. Geriatr. Cogn. Disord. 2021,49, 321–333. [CrossRef]
99.
Wimo, A.; Guerchet, M.; Ali, G.C.; Wu, Y.T.; Prina, A.M.; Winblad, B.; Jönsson, L.; Liu, Z.; Prince, M. The worldwide costs of
dementia 2015 and comparisons with 2010. Alzheimer’s Dement. 2017,13, 1–7. [CrossRef]
100.
von Bartheld, C.S. Myths and truths about the cellular composition of the human brain: A review of influential concepts. J. Chem.
Neuroanat. 2018,93, 2–15. [CrossRef]
Int. J. Mol. Sci. 2023,24, 10021 16 of 17
101.
Pratico, D.; Sung, S. Lipid peroxidation and oxidative imbalance: Early functional events in Alzheimer ’s disease. J. Alzheimers Dis.
2004,6, 171–175. [CrossRef] [PubMed]
102.
Pratico, D. Oxidative stress hypothesis in Alzheimer’s disease: A reappraisal. Trends Pharmacol. Sci.
2008
,29, 609–615. [CrossRef]
[PubMed]
103.
Zhu, Z.; Liu, Y.; Gong, Y.; Jin, W.; Topchiy, E.; Turdi, S.; Gao, Y.; Culver, B.; Wang, S.; Ge, W.; et al. Mitochondrial aldehyde
dehydrogenase (ALDH2) rescues cardiac contractile dysfunction in an APP/PS1 murine model of Alzheimer’s disease via
inhibition of ACSL4-dependent ferroptosis. Acta Pharmacol. Sin. 2022,43, 39–49. [CrossRef] [PubMed]
104.
Khan, S.; Barve, K.H.; Kumar, M.S. Recent Advancements in Pathogenesis, Diagnostics and Treatment of Alzheimer’s Disease.
Curr. Neuropharmacol. 2020,18, 1106–1125. [CrossRef] [PubMed]
105.
Butterfield, D.A.; Drake, J.; Pocernich, C.; Castegna, A. Evidence of oxidative damage in Alzheimer’s disease brain: Central role
for amyloid beta-peptide. Trends Mol. Med. 2001,7, 548–554. [CrossRef]
106.
Liang, Z.; Yongqun, L.; Qinchuan, X.; Shitong, Z.; Xiaoli, L.I. Ferroptosis regulatory signaling pathway and its research progress
in re-lated diseases. Chin. J. Clin. Pharmacol. Ther. 2022,27, 227–234. [CrossRef]
107.
Tolosa, E.; Garrido, A.; Scholz, S.W.; Poewe, W. Challenges in the diagnosis of Parkinson’s disease. Lancet Neurol.
2021
,20,
385–397. [CrossRef]
108.
Wang, Y.; Tang, B.; Zhu, J.; Yu, J.; Hui, J.; Xia, S.; Ji, J. Emerging Mechanisms and Targeted Therapy of Ferroptosis in Neurological
Diseases and Neuro-oncology. Int. J. Biol. Sci. 2022,18, 4260–4274. [CrossRef]
109.
Tang, F.; Zhou, L.; Li, P.; Jiao, L.; Chen, K.; Guo, Y.; Ding, X.; He, S.; Dong, B.; Xu, R.; et al. Inhibition of ACSL4 Alleviates
Parkinsonism Phenotypes by Reduction of Lipid Reactive Oxygen Species. Neurotherapeutics 2023. [CrossRef]
110.
Jia, R.; Liu, Y.; Shuai, K.; Zhou, C.; Chen, L.; Zhu, L.; Wu, X. The Relationship between Iron and LRRK2 in a 6-OHDA-Induced
Parkinson’s Disease Model. Int. J. Mol. Sci. 2023,24, 3709. [CrossRef]
111.
Lee, H.; Choi, C.; Lee, S. Membrane-bound
α
-Synuclein Has a High Aggregation Propensity and the Ability to Seed the
Aggregation of the Cytosolic Form*. J. Biol. Chem. 2002,277, 671–678. [CrossRef] [PubMed]
112.
Peng, Y.; Wang, C.; Xu, H.H.; Liu, Y.; Zhou, F. Binding of
α
-synuclein with Fe(III) and with Fe(II) and biological implications of
the resultant complexes. J. Inorg. Biochem. 2010,104, 365–370. [CrossRef] [PubMed]
113.
Singleton, A.B.; Farrer, M.; Johnson, J.; Singleton, A.; Hague, S.; Kachergus, J.; Hulihan, M.; Peuralinna, T.; Dutra, A.; Nussbaum,
R.; et al. α-Synuclein Locus Triplication Causes Parkinson’s Disease. Science 2003,302, 841. [CrossRef] [PubMed]
114.
Rademacher, D.J. Potential for Therapeutic-Loaded Exosomes to Ameliorate the Pathogenic Effects of
α
-Synuclein in Parkinson’s
Disease. Biomedicines 2023,11, 1187. [CrossRef] [PubMed]
115.
Koeglsperger, T.; Rumpf, S.L.; Schliesser, P.; Struebing, F.L.; Brendel, M.; Levin, J.; Trenkwalder, C.; Hoglinger, G.U.; Herms, J.
Neuropathology of incidental Lewy body & prodromal Parkinson’s disease. Mol. Neurodegener. 2023,18, 32. [CrossRef]
116.
Fecchio, C.; Palazzi, L.; de Laureto, P.P.
α
-Synuclein and Polyunsaturated Fatty Acids: Molecular Basis of the Interaction and
Implication in Neurodegeneration. Molecules 2018,23, 1531. [CrossRef]
117.
Sharon, R.; Bar-Joseph, I.; Frosch, M.P.; Walsh, D.M.; Hamilton, J.A.; Selkoe, D.J. The formation of highly soluble oligomers of
alpha-synuclein is regulated by fatty acids and enhanced in Parkinson’s disease. Neuron 2003,37, 583–595. [CrossRef]
118.
Ma, J.J.; Li, X.H.; Fan, Y.Y.; Yang, D.W.; Gu, Q.; Li, D.S.; Chen, S.Y.; Wu, S.P.; Zheng, J.H.; Yang, H.Q.; et al. miR-494-3p Promotes
Erastin-Induced Ferroptosis by Targeting REST to Activate the Interplay between SP1 and ACSL4 in Parkinson’s Disease. Oxidative
Med. Cell. Longev. 2022,2022, 7671324. [CrossRef]
119.
Kuruppu, A.I.; Turyanska, L.; Bradshaw, T.D.; Manickam, S.; Galhena, B.P.; Paranagama, P.; De Silva, R. Apoferritin and Dps as
drug delivery vehicles: Some selected examples in oncology. Biochim. Biophys. Acta-Gen. Subj. 2022,1866, 130067. [CrossRef]
120.
Anjum, A.; Yazid, M.D.I.; Fauzi Daud, M.; Idris, J.; Ng, A.M.H.; Selvi Naicker, A.; Ismail, O.H.R.; Athi Kumar, R.K.; Lokanathan,
Y. Spinal Cord Injury: Pathophysiology, Multimolecular Interactions, and Underlying Recovery Mechanisms. Int. J. Mol. Sci.
2020,21, 7533. [CrossRef]
121.
Cozzolino, M.; Ferri, A.; Teresa Carrì, M. Amyotrophic Lateral Sclerosis: From Current Developments in the Laboratory to
Clinical Implications. Antioxid. Redox Signal. 2008,10, 405–444. [CrossRef]
122.
Matsuzono, K.; Suzuki, M.; Miura, K.; Ozawa, T.; Mashiko, T.; Koide, R.; Tanaka, R.; Fujimoto, S. Higher incidence of cervical
spinal cord compression in amyotrophic lateral sclerosis: A single-institute cohort study. Neurol. Sci.
2022
,43, 1079–1086.
[CrossRef]
123.
Sykova, E.; Cizkova, D.; Kubinova, S. Mesenchymal Stem Cells in Treatment of Spinal Cord Injury and Amyotrophic Lateral
Sclerosis. Front. Cell Dev. Biol. 2021,9, 695900. [CrossRef]
124.
Spasi´c, S.; Nikoli´c-Koki´c, A.; Mileti´c, S.; Orešˇcanin-Duši´c, Z.; Spasi´c, M.B.; Blagojevi´c, D.; Stevi´c, Z. Edaravone May Prevent
Ferroptosis in ALS. Curr. Drug Targets 2020,21, 776–780. [CrossRef] [PubMed]
125. Rothstein, J.D. Edaravone: A new drug approved for ALS. Cell 2017,171, 725. [CrossRef] [PubMed]
126.
Zhang, Y.; Sun, C.; Zhao, C.; Hao, J.; Zhang, Y.; Fan, B.; Li, B.; Duan, H.; Liu, C.; Kong, X.; et al. Ferroptosis inhibitor SRS 16-86
attenuates ferroptosis and promotes functional recovery in contusion spinal cord injury. Brain Res.
2019
,1706, 48–57. [CrossRef]
[PubMed]
127.
Angelova, P.R.; Esteras, N.; Abramov, A.Y. Mitochondria and lipid peroxidation in the mechanism of neurodegeneration: Finding
ways for prevention. Med. Res. Rev. 2021,41, 770–784. [CrossRef]
Int. J. Mol. Sci. 2023,24, 10021 17 of 17
128.
Cha, S.J.; Kim, K. Effects of the Edaravone, a Drug Approved for the Treatment of Amyotrophic Lateral Sclerosis, on Mitochondrial
Function and Neuroprotection. Antioxidants 2022,11, 195. [CrossRef]
129.
Zhou, X.; Zhao, R.; Lv, M.; Xu, X.; Liu, W.; Li, X.; Gao, Y.; Zhao, Z.; Zhang, Z.; Li, Y.; et al. ACSL4 promotes microglia-mediated
neuroinflammation by regulating lipid metabolism and VGLL4 expression. Brain Behav. Immun. 2023,109, 331–343. [CrossRef]
130. Compston, A.; Coles, A. Multiple sclerosis. Lancet 2008,372, 1502–1517. [CrossRef]
131. Doshi, A.; Chataway, J. Multiple sclerosis, a treatable disease. Clin. Med. 2016,16, s53–s59. [CrossRef] [PubMed]
132.
Oh, J.; Vidal-Jordana, A.; Montalban, X. Multiple sclerosis: Clinical aspects. Curr. Opin. Neurol.
2018
,31, 752–759. [CrossRef]
[PubMed]
133. Marcus, R. What Is Multiple Sclerosis? JAMA 2022,328, 2078. [CrossRef] [PubMed]
134.
Stadelmann, C.; Timmler, S.; Barrantes-Freer, A.; Simons, M. Myelin in the Central Nervous System: Structure, Function, and
Pathology. Physiol. Rev. 2019,99, 1381–1431. [CrossRef] [PubMed]
135.
Luoqian, J.; Yang, W.; Ding, X.; Tuo, Q.Z.; Xiang, Z.; Zheng, Z.; Guo, Y.J.; Li, L.; Guan, P.; Ayton, S.; et al. Ferroptosis promotes
T-cell activation-induced neurodegeneration in multiple sclerosis. Cell. Mol. Immunol. 2022,19, 913–924. [CrossRef] [PubMed]
136.
Kipp, M.; Nyamoya, S.; Hochstrasser, T.; Amor, S. Multiple sclerosis animal models: A clinical and histopathological perspective.
Brain Pathol. 2017,27, 123–137. [CrossRef]
137. Yue, T.; Guo-dong, L.; Yue-Ming, J. Studies on fatty acid activation mediated by ACSL4 and tumor. J. Toxicol. 2018,32, 160–164.
138.
Ye, X.; Zhang, Y.; Wang, X.; Li, Y.; Gao, Y. Tumor-suppressive functions of long-chain acyl-CoA synthetase 4 in gastric cancer.
IUBMB Life 2016,68, 320–327. [CrossRef]
139.
Jiang, X.; Guo, S.; Zhang, Y.; Zhao, Y.; Li, X.; Jia, Y.; Xu, Y.; Ma, B. LncRNA NEAT1 promotes docetaxel resistance in prostate
cancer by regulating ACSL4 via sponging miR-34a-5p and miR-204-5p. Cell. Signal. 2020,65, 109422. [CrossRef]
140.
Wu, X.; Zhi, F.; Lun, W.; Deng, Q.; Zhang, W. Baicalin inhibits PDGF-BB-induced hepatic stellate cell proliferation, apoptosis,
invasion, migration and activation via the miR-3595/ACSL4 axis. Int. J. Mol. Med. 2018,41, 1992–2002. [CrossRef]
141.
Park, S.; Oh, J.; Kim, Y.; Choe, S.; Chun, C.; Jin, E. Suppression of ABCD2 dysregulates lipid metabolism via dysregulation of
miR-141:ACSL4 in human osteoarthritis. Cell Biochem. Funct. 2018,36, 366–376. [CrossRef] [PubMed]
Disclaimer/Publisher’s Note:
The statements, opinions and data contained in all publications are solely those of the individual
author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to
people or property resulting from any ideas, methods, instructions or products referred to in the content.
... xCT inhibition also attenuates glutamine metabolism and lowers nutrient flexibility (12). Acyl-CoA synthetase long-chain family member 4 (ACSL4) increases levels of polyunsaturated fatty acids (PUFAs), raising sensitivity to ferroptosis by increasing lipid peroxidation (13). Moreover, high levels of lipid droplets could proportionally elevate susceptibility to ferroptosis because of elevated PUFAs and lipophagy activation (14). ...
Article
NAD(P)H-quinone oxidoreductase 1 (NQO1) is an essential redox enzyme responsible for redox balance and energy metabolism. Despite of its importance, the brain contains high capacity of polyunsaturated fatty acids and maintains low levels of NQO1 expression. In this study, we examined how levels of NQO1 expression affects cell survival in response to toxic insults causing mitochondrial dysfunction and ferroptosis, and whether NQO1 has a potential as a biomarker in different stressed conditions. Following treatment with rotenone, overexpressed NQO1 in SH-SY5Y cells improved cell survival by reducing mitochondrial reductive stress via increased NAD⁺ supply without mitochondrial biogenesis. However, NQO1 overexpression boosted lipid peroxidation following treatment with RSL3 and erastin. A lipid droplet staining assay showed increased lipid droplets in cells overexpressing NQO1. In contrast, NQO1 knockdown protected cells against ferroptosis by increasing GPX4, xCT, and the GSH/GSSG system. Also, NQO1 knockdown showed lower iron contents and lipid droplets than non-transfectants and cells overexpressing NQO1, even though it could not attenuate cell death when exposed to rotenone. In summary, our study suggests that different NQO1 levels may have advantages and disadvantages depending on the surrounding environments. Thus, regulating NQO1 expression could be a potential supplementary tool when treating neuronal diseases.
... And the correlation analysis showed a positive correlation between AFF3 and ACSL4 expression (Fig. 6C). The induction of ferroptosis is dependent on the peroxidation of PL-PUFA, and the acyl coenzyme a synthase long-chain family member 4 (ACSL4) enzyme is involved in lipid metabolism and catalyzes the activation and membrane binding of PUFAs, leading to an increased susceptibility of cells to ferroptosis [31][32][33]. The western blot assay showed significantly increased protein levels of ACSL4 in DU145 and 22RV1 cell lines after overexpression of AFF3 while knockdown of AFF3 decreased the protein level of ACSL4 (Fig. 6D&E). ...
Article
Full-text available
Prostate cancer (PCa) is one of the most common cancers affecting the health of men worldwide. Castration-resistant prostate cancer (CRPC), the advanced and refractory phase of prostate cancer, has multiple mechanisms of resistance to androgen deprivation therapy (ADT) such as AR mutations, aberrant androgen synthase, and abnormal expression of AR-related genes. Based on the research of the AR pathway, new drugs for the treatment of CRPC have been developed in clinical practice, such as Abiraterone and enzalutamide. However, many areas in this pathway are still worth exploring. In this study, single-cell sequencing analysis was utilized to scrutinize significant genes in the androgen receptor (AR) pathway related to CRPC. Our analysis of single-cell sequencing combined with bulk-cell sequencing revealed a substantial downregulation of AR-regulated AFF3 in CRPC. Overexpression of AFF3 restricted the proliferation and migration of prostate cancer cells whilst also increasing their sensitivity towards enzalutamide, while knockdown of AFF3 had the opposite effect. To elucidate the mechanism of tumor inhibition by AFF3, we applied GSVA and GSEA to investigate the metabolic pathways related to AFF3 and revealed that AFF3 had an impact on fatty acids metabolism and ferroptosis through the regulation of ACSL4 protein expression. Based on correlation analysis and flow cytometry, we can speculate that AFF3 can impact the sensitivity of the CRPC cell lines to the ferroptosis inducer (RSL3) by regulating ACSL4. Therefore, our findings may provide new insights into the mechanisms of drug resistance in CRPC, and AFF3 may serve as a novel prognostic biomarker in prostate cancer.
... ACSL4-and LPCAT3-mediated reactions are jointly required for the initiation of ferroptosis by control-ling the availability of PUFA substrates for phospholipid synthesis ( Figure 4). Specifically, ACSL4 catalyzes the addition of CoA to long-chain polyunsaturated-CoAs (PUFA-CoAs), preferentially of AA and AdA, therefore activating the biosynthesis of AA-or AdA-containing phospholipids (reviewed in [152]). PUFA-CoAs then become substrates of the lysophosphatidylcholine acyltransferase (LPCAT) enzymes, particularly LPCAT3, which esterifies the PUFA-CoAs into PUFA-phospholipid (reviewed in [153]). ...
Article
Ferroptosis is a type of programmed cell death that differs from apoptosis, autophagy, and necrosis and is related to several physio-pathological processes, including tumorigenesis, neu-rodegeneration, senescence, blood diseases, kidney disorders, and ischemia-reperfusion injuries. Ferroptosis is linked to iron accumulation, eliciting dysfunction of antioxidant systems, which favor the production of lipid peroxides, cell membrane damage, and ultimately, cell death. Thus, signaling pathways evoking ferroptosis are strongly associated with those protecting cells against iron excess and/or lipid-derived ROS. Here, we discuss the interaction between the metabolic pathways of ferroptosis and antioxidant systems, with a particular focus on transcription factors implicated in the regulation of ferroptosis, either as triggers of lipid peroxidation or as ferroptosis antioxidant defense pathways.
... Furthermore, the levels of irP2, which is involved in the regulation of iron metabolism, significantly increased in the brain in the PHZ group compared with that of the control group ( Fig. 3D and E). ACSL4 is a specific biomarker and driver of ferroptosis (41), while FSP1 has been identified as key molecules in independent pathways associated with ferroptosis inhibition (42). ACSL4 levels significantly increased and FSP1 levels significantly decreased in the PHZ group compared with that of the control group (Fig. 3G-i). ...
Article
Full-text available
Ferroptosis is driven by iron‑dependent accumulation of lipid hydroperoxides, and hemolytic hyperbilirubinemia causes accumulation of unconjugated bilirubin and iron. The present study aimed to assess the role of ferroptosis in hemolytic hyperbilirubinemia‑induced brain damage (HHIBD). Rats were randomly divided into the control, phenylhydrazine (PHZ) and deferoxamine (DFO) + PHZ groups, with 12 rats in each group. Ferroptosis‑associated biochemical and protein indicators were measured in the brain tissue of rats. We also performed tandem mass tag‑labeled proteomic analysis. The levels of iron and malondialdehyde were significantly higher and levels of glutathione (GSH) and superoxide dismutase activity significantly lower in the brain tissues of the PHZ group compared with those in the control group. HHIBD also resulted in significant increases in the expression of the ferroptosis‑related proteins acyl‑CoA synthetase long‑chain family member 4, ferritin heavy chain 1 and transferrin receptor and divalent metal transporter 1, as well as a significant reduction in the expression of ferroptosis suppressor protein 1. Kyoto Encyclopedia of Genes and Genomes pathway enrichment analysis demonstrated that the differentially expressed proteins of rat brain tissues between the control and PHZ groups were significantly involved in ferroptosis, GSH metabolism and fatty acid biosynthesis pathways. Pretreatment with DFO induced antioxidant activity and alleviated lipid peroxidation‑mediated HHIBD. In addition, PC12 cells treated with ferric ammonium citrate showed shrinking mitochondria, high mitochondrial membrane density, and increased lipid reactive oxygen species and intracellular ferrous iron, which were antagonized by pretreatment with ferrostatin‑1 or DFO, which was reversed by pretreatment with ferrostatin‑1 or DFO. The present study demonstrated that ferroptosis is involved in HHIBD and provided novel insights into candidate proteins that are potentially involved in ferroptosis in the brain during hemolytic hyperbilirubinemia.
Article
Full-text available
Ischemic stroke is a common neurological disease. Currently, there are no Food and Drug Administration-approved drugs that can maximize the improvement in ischemic stroke-induced nerve damage. Hence, treating ischemic stroke remains a clinical challenge. Ferroptosis has been increasingly studied in recent years, and it is closely related to the pathophysiological process of ischemic stroke. Iron overload, reactive oxygen species accumulation, lipid peroxidation, and glutamate accumulation associated with ferroptosis are all present in ischemic stroke. This article focuses on describing the relationship between ferroptosis and ischemic stroke and summarizes the relevant substances that ameliorate ischemic stroke-induced neurological damage by inhibiting ferroptosis. Finally, the problems in the treatment of ischemic stroke targeting ferroptosis are discussed, hoping to provide a new direction for its treatment.
Article
Full-text available
Purpose Excessive necroptosis contributes to the pathogenesis of several inflammatory and neurodegenerative diseases. Here, using a high-throughput screening approach, we investigated the anti-necroptosis effects of piperlongumine, an alkaloid isolated from the long pepper plant, in vitro and in a mouse model of systemic inflammatory response syndrome (SIRS). Methods A natural compound library was screened for anti-necroptosis effects in cellular. The underlying mechanism of action of the top candidate piperlongumine was explored by quantifying the necroptosis marker phosphorylated receptor-interacting protein kinase 1 (p-RIPK1) by Western blotting. The anti-inflammatory effect of piperlongumine was assessed in a tumor necrosis factor α (TNFα)-induced SIRS model in mice. Results Among the compounds investigated, piperlongumine significantly rescued cell viability. The half maximal effective concentration (EC50) of piperlongumine for inhibiting necroptosis was 0.47 μM in HT-29 cells, 6.41 μM in FADD-deficient Jurkat cells, and 2.33 µM in CCRF-CEM cells, while the half maximal inhibitory concentration (IC50) was 95.4 µM in HT-29 cells, 93.02 µM in FADD-deficient Jurkat cells, and 161.1 µM in CCRF-CEM cells. Piperlongumine also significantly inhibited TNFα-induced intracellular RIPK1 Ser166 phosphorylation in cell lines and significantly prevented decreases in body temperature and improved survival in SIRS mice. Conclusion As a potent necroptosis inhibitor, piperlongumine prevents phosphorylation of RIPK1 at its activation residue Ser166. Piperlongumine thus potently inhibits necroptosis at concentrations safe enough for human cells in vitro and inhibits TNFα-induced SIRS in mice. Piperlongumine has potential clinical translational value for the treatment of the spectrum of diseases associated with necroptosis, including SIRS.
Article
Full-text available
Background Parkinson’s disease (PD) is a progressive neurodegenerative disorder associated with a loss of dopaminergic (DA) neurons. Despite symptomatic therapies, there is currently no disease-modifying treatment to halt neuronal loss in PD. A major hurdle for developing and testing such curative therapies results from the fact that most DA neurons are already lost at the time of the clinical diagnosis, rendering them inaccessible to therapy. Understanding the early pathological changes that precede Lewy body pathology (LBP) and cell loss in PD will likely support the identification of novel diagnostic and therapeutic strategies and help to differentiate LBP-dependent and -independent alterations. Several previous studies identified such specific molecular and cellular changes that occur prior to the appearance of Lewy bodies (LBs) in DA neurons, but a concise map of such early disease events is currently missing. Methods Here, we conducted a literature review to identify and discuss the results of previous studies that investigated cases with incidental Lewy body disease (iLBD), a presumed pathological precursor of PD. Results Collectively, our review demonstrates numerous cellular and molecular neuropathological changes occurring prior to the appearance of LBs in DA neurons. Conclusions Our review provides the reader with a summary of early pathological events in PD that may support the identification of novel therapeutic and diagnostic targets and aid to the development of disease-modifying strategies in PD.
Article
Full-text available
Pyroptosis is a novel pro-inflammatory cell programmed death dependent on Gasdermin (GSMD) family-mediated membrane pore formation and subsequent cell lysis, accompanied by the release of inflammatory factors and expanding inflammation in multiple tissues. All of these processes have impacts on a variety of metabolic disorders. Dysregulation of lipid metabolism is one of the most prominent metabolic alterations in many diseases, including the liver, cardiovascular system, and autoimmune diseases. Lipid metabolism produces many bioactive lipid molecules, which are important triggers and endogenous regulators of pyroptosis. Bioactive lipid molecules promote pyroptosis through intrinsic pathways involving reactive oxygen species (ROS) production, endoplasmic reticulum (ER) stress, mitochondrial dysfunction, lysosomal disruption, and the expression of related molecules. Pyroptosis can also be regulated during the processes of lipid metabolism, including lipid uptake and transport, de novo synthesis, lipid storage, and lipid peroxidation. Taken together, understanding the correlation between lipid molecules such as cholesterol and fatty acids and pyroptosis during metabolic processes can help to gain insight into the pathogenesis of many diseases and develop effective strategies from the perspective of pyroptosis.
Article
Full-text available
Pathogenic forms of α-synuclein (α-syn) are transferred to and from neurons, astrocytes, and microglia, which spread α-syn pathology in the olfactory bulb and the gut and then throughout the Parkinson's disease (PD) brain and exacerbate neurodegenerative processes. Here, we review attempts to minimize or ameliorate the pathogenic effects of α-syn or deliver therapeutic cargo into the brain. Exosomes (EXs) have several important advantages as carriers of therapeutic agents including an ability to readily cross the blood-brain barrier, the potential for targeted delivery of therapeutic agents, and immune resistance. Diverse cargo can be loaded via various methods, which are reviewed herein, into EXs and delivered into the brain. Genetic modification of EX-producing cells or EXs and chemical modification of EX have emerged as powerful approaches for the targeted delivery of therapeutic agents to treat PD. Thus, EXs hold great promise for the development of next-generation therapeutics for the treatment of PD.
Article
Full-text available
The pathogenesis of Parkinson’s disease (PD) is very complex and still needs further exploration. Leucine-rich repeat kinase 2 (LRRK2) is associated with familial PD in mutant forms and sporadic PD in the wild-type form. Abnormal iron accumulation is found in the substantia nigra of PD patients, but its exact effects are not very clear. Here, we show that iron dextran exacerbates the neurological deficit and loss of dopaminergic neurons in 6-OHDA lesioned rats. 6-OHDA and ferric ammonium citrate (FAC) significantly increase the activity of LRRK2 as reflected by the phosphorylation of LRRK2, at S935 and S1292 sites. 6-OHDA-induced LRRK2 phosphorylation is attenuated by the iron chelator deferoxamine, especially at the S1292 site. 6-OHDA and FAC markedly induce the expression of pro-apoptotic molecules and the production of ROS by activating LRRK2. Furthermore, G2019S-LRRK2 with high kinase activity showed the strongest absorptive capacity for ferrous iron and the highest intracellular iron content among WT-LRRK2, G2019S-LRRK2, and kinase-inactive D2017A-LRRK2 groups. Taken together, our results demonstrate that iron promotes the activation of LRRK2, and active LRRK2 accelerates ferrous iron uptake, suggesting that there exists an interplay between iron and LRRK2 in dopaminergic neurons, providing a new perspective to uncover the underlying mechanisms of PD occurrence.
Article
Full-text available
Alzheimer’s disease (AD) is a genetically complex and heterogeneous disorder with multifaceted neuropathological features, including β-amyloid plaques, neurofibrillary tangles, and neuroinflammation. Over the past decade, emerging evidence has implicated both beneficial and pathological roles for innate immune genes and immune cells, including peripheral immune cells such as T cells, which can infiltrate the brain and either ameliorate or exacerbate AD neuropathogenesis. These findings support a neuroimmune axis of AD, in which the interplay of adaptive and innate immune systems inside and outside the brain critically impacts the etiology and pathogenesis of AD. In this review, we discuss the complexities of AD neuropathology at the levels of genetics and cellular physiology, highlighting immune signaling pathways and genes associated with AD risk and interactions among both innate and adaptive immune cells in the AD brain. We emphasize the role of peripheral immune cells in AD and the mechanisms by which immune cells, such as T cells and monocytes, influence AD neuropathology, including microglial clearance of amyloid-β peptide, the key component of β-amyloid plaque cores, pro-inflammatory and cytotoxic activity of microglia, astrogliosis, and their interactions with the brain vasculature. Finally, we review the challenges and outlook for establishing immune-based therapies for treating and preventing AD.
Article
Full-text available
Alzheimer’s disease (AD) is the most common type of dementia, accounting for 60% to 80% of all cases [...]
Article
Ferroptosis is a programmed cell death pathway that is recently linked to Parkinson's disease (PD), where the key genes and molecules involved are still yet to be defined. Acyl-CoA synthetase long-chain family member 4 (ACSL4) esterifies polyunsaturated fatty acids (PUFAs) which is essential to trigger ferroptosis, and is suggested as a key gene in the pathogenesis of several neurological diseases including ischemic stroke and multiple sclerosis. Here, we report that ACSL4 expression in the substantia nigra (SN) was increased in a 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-treated model of PD and in dopaminergic neurons in PD patients. Knockdown of ACSL4 in the SN protected against dopaminergic neuronal death and motor deficits in the MPTP mice, while inhibition of ACSL4 activity with Triacsin C similarly ameliorated the parkinsonism phenotypes. Similar effects of ACSL4 reduction were observed in cells treated with 1-methyl-4-phenylpyridinium (MPP+) and it specifically prevented the lipid ROS elevation without affecting the mitochondrial ROS changes. These data support ACSL4 as a therapeutic target associated with lipid peroxidation in PD.
Article
Acyl-CoA synthetase long-chain family member 4 (ACSL4) is an important isozyme in polyunsaturated fatty acid (PUFA) metabolism. The role of ACSL4 in the lipopolysaccharide (LPS)-induced inflammation of microglia, and the effects of ACSL4-mediated inflammation on the progression of Parkinson's disease (PD) are unknown. In this study, we found that ACSL4 expression was increased after LPS stimulation. Knocking down ACSL4 in microglia decreased proinflammatory cytokine production. Mechanistically, ACSL4 reduced vestigial-like family member 4(VGLL4) expression to promote NF-κB signal transduction; and ACSL4 regulated lipid composition after LPS stimulation. In addition, knocking down ACSL4 alleviated neuroinflammation in a systemic LPS model and acute l-methyl-4-phenyl-l,2,3,6-tetrahydropyridine (MPTP) model. These data revealed ACSL4 to be a novel regulator that promotes microglia-mediated neuroinflammation by regulating VGLL4 expression and lipid metabolism.