You are on page 1of 6

Prostaglandins and Other Lipid Mediators 144 (2019) 106363

Contents lists available at ScienceDirect

Prostaglandins and Other Lipid Mediators


journal homepage: www.elsevier.com/locate/prostaglandins

Role of acyl-CoA synthetase ACSL4 in arachidonic acid metabolism T



Hiroshi Kuwata, Shuntaro Hara
Division of Health Chemistry, Department of Healthcare and Regulatory Sciences, School of Pharmacy, Showa University, Tokyo, Japan

A R T I C LE I N FO A B S T R A C T

Keywords: The activation of long-chain free fatty acids is the first step reaction of their usage in the cells and tissues, which
acyl-CoA synthetase are catalyzed by a family of enzymes called acyl-coenzyme A synthetases long-chain isoform (ACSL). The five
ACSL4 ACSL enzymes identified in mammals are thought to have specific and differing functions. Among them, ACSL4
Arachidonic acid is a unique isozyme that preferentially catalyzes several polyunsaturated fatty acids (PUFAs) such as arachidonic
Eicosanoid
acid (AA), and ACSL4 is thought to be an important isozyme for PUFA metabolism. Recent studies revealed that
Polyunsaturated fatty acid
ACSL4 is involved in biological responses including inflammation, steroidogenesis, cell death, female fertility,
and cancer. ACSL4 and its substrate PUFAs are thus likely to contribute to these responses. However, the roles of
ACSL4 in PUFA metabolism are not fully understood. In this review, we describe the recent progress in ACSL4
research including the involvement of this enzyme in AA metabolism.

1. Introduction suggests a link between it and various pathological events (e.g., in-
flammation and cancer).
Long-chain fatty acids are important nutrients that are used as fuel Because the characteristics and functions of ACS family proteins are
for energy, the post-translational modification of proteins, components discussed in other reviews [1,8], in this review we summarize the re-
of membrane lipids, precursors of bioactive lipid mediators, and more. cent progress regarding the functions of ACSL4, with a focus on roles of
For most biological events (anabolic and catabolic cellular responses), ACSL4 in PUFA metabolism.
free long-chain fatty acids must be converted to their respective acyl-
coenzyme A (acyl-CoA) forms. The enzymes catalyzing the first step of 2. Roles of ACSL4 in eicosanoid biosynthesis following
the activation of fatty acids are acyl-CoA synthetases (ACS) which can proinflammatory stimulation
activate short-, medium-, long-, and very-long-chain fatty acids [1], and
are referred to as ACSS, ACSM, ACSL, and ACSVL, respectively. The The regulation of eicosanoid biosynthesis is quite complex, and
ACS family also includes ACS bubblegum (ACSBG) and ACS family several AA-metabolizing enzymes such as phospholipase A2 (PLA2),
(ACSF) enzymes [2,3]. Because the use of the fatty acids is essential for cyclooxygenase (COX), lipoxygenase (LOX), and cytochrome P450
cell homeostasis, the members of the ACS family are evolutionarily (CYP) enzymes are known to be involved in this regulation. In general,
conserved enzymes and are expressed in bacteria, yeasts, protozoans, AA released after cell activation is metabolized by COX or LOX, and
insects, fish, and mammals [3]. then the oxidized AA is converted into bioactive eicosanoids by each
Among the ACS family enzymes, ACSL — which catalyzes a broad terminal eicosanoid synthase (Fig. 1). Cytochrome P450 (CYP) mono-
range of fatty acid substrates from 10 to 22 carbons — consists of five oxygenase catalyzes the production of epoxyeicosatrienoic acids (EETs)
ACSL isozymes (ACSL1, ACSL3, ACSL4, ACSL5, and ACSL6) in mam- and hydroxyeicosatetraenoic acids (HETEs), and both EETs and HETEs
mals. The substrate specificities of the ACSL enzymes differ among the are activated by ACSL enzymes [9,10], resulting in the production of
five isozymes [4–6], and in particular, ACSL4 prefers polyunsaturated EET- and HETE-CoAs. These oxidized acyl-CoAs are esterified into
fatty acid (PUFAs) such as arachidonic acid (AA) and eicosapentaenoic phospholipids [11], which cause changes in properties of the mem-
acid (EPA) as its substrate [7]. Indeed, several studies have shown that brane lipid bilayer [12]. Studies of eicosanoid biosynthesis have fo-
ACSL4 plays a role in AA metabolism in a manner that is dependent on cused on the regulation of eicosanoid-producing enzymes, but it re-
the cell types (Table 1). The PUFAs are precursors of bioactive lipid mains unknown whether all of the released free AA uses these
mediators such as eicosanoids, and the unique feature of ACSL4 metabolic pathways.


Corresponding author at: Division of Health Chemistry, Department of Healthcare and Regulatory Sciences, School of Pharmacy, Showa University, 1-5-8
Hatanodai, Shinagawa-ku, Tokyo 142-8555, Japan.
E-mail address: haras@pharm.showa-u.ac.jp (S. Hara).

https://doi.org/10.1016/j.prostaglandins.2019.106363
Received 31 March 2019; Received in revised form 15 June 2019; Accepted 11 July 2019
Available online 12 July 2019
1098-8823/ © 2019 Elsevier Inc. All rights reserved.
H. Kuwata and S. Hara Prostaglandins and Other Lipid Mediators 144 (2019) 106363

Table 1
Relationships between ACSL and eicosanoid biosynthesis.
Cell-Types Stimulation Eicosanoids Experimental methods Eicosanoid release Ref.

2+
Rat neutrophiles Ca -ionophore (A23187) 5-HETE 6-keto-PGF1α PGE2 Triacsin C upregulated [14]
Human endothelial cells vasopressin 6-keto-PGF1α Triacsin C no effect [16]
angiotensin-II
bradykinin
Ca2+-ionophore (ionomycin)
Human breast cancer MDA-MB-231 no 5-HETE 12-HETE 15-HETE Knockdown of ACSL4 downregulated [31]
cells
Human breast cancer MCF-7 cells no 5-HETE 12-HETE 15-HETE Overexpression of ACSL4 upregulated
Human arterial smooth muscle cells IL-1β PGE2 Overexpression of ACSL4v1 downregulated [19]
no Triacsin C or Rosiglitazone upregulated
IL-1β Knockdown of ACSL4 downregulated
Rat fibroblastic 3Y1 cells IL-1β PGs 15-HETE 11-HETE (5,11-diHETE) Triacsin C or Knockdown of upregulated [15,21]
ACSL4
Mouse embryonic fibroblasts IL-1β PGE2 PGD2 PGF2α Triacsin C
Human fibroblastic WI-38 cells IL-1β
Human monocytic THP-1 cells lipopolysaccaride
Mouse gonadal white adipose tissue high fat diet COX-, LOX-, and CYP450-derived adipocyte-specific ACSL4 no effect [36]
oxylipins knockout

HETE, hydroxyeicosatetraenoic acid; 6-keto-PGF1α, stable metabolite of PGI2; ACSL4v1, ACSL4 transcript variant 1.

Fig. 1. ACSL4 in eicosanoid biosynthesis.


ACSL4: acyl-CoA synthetase 4, COX: cycloox-
ygenase, CYP: cytochrome P450, EETs: epox-
yeicosatrienoic acids, HETEs: hydro-
xyeicosatetraenoic acids, LOX: lipoxygenase,
LTs: leukotrienes, MAGL: monoacylglycerol li-
pase, PGs: prostaglandins, PLA2: phospholipase
A2, TX: thromboxane.

Oh-ishi et al. demonstrated that following calcium-ionophore with triacsin C enhanced the release of PGE2.
A23187 stimulation, the treatment of peritoneal neutrophils with We also observed that the knockdown of ACSL4 markedly enhances
triacsin C (which inhibits ACSL activity [13]) markedly enhanced the the eicosanoid biosynthesis from interleukin (IL)-1β-treated rat fibro-
release of eicosanoids as well as AA [14]. The enhancement of the re- blastic 3Y1 cells [15,21]. This effect is specific to the ACSL4 isoform,
lease of eicosanoids by triacsin C treatment was also observed in several because the knockdown of ACSL1 or ACSL3 failed to enhance the ei-
cell types, including human fibroblastic WI-38 cells (Table 1 [15],). cosanoid release. The knockdown of ACSL4 not only enhanced the le-
These results suggest that an inhibition of the activation of AA fol- vels of eicosanoids; it also accelerated the release of eicosanoids after
lowing stimulation may cause the increased levels of free AA and then stimulation. In light of these results, we speculated that most of the AA
the conversion of AA into eicosanoids. However, this hypothesis may released from phospholipids after IL-1β-stimulation is eliminated
not apply to all cell types, such as human endothelial cells [16]. The promptly via the ACSL4 pathway.
reason for this is not yet known, but there may be intricate mechanisms
underlying the biosynthesis of eicosanoids. 3. Involvement of ACSL4 in the release of AA into the
Because triacsin C is now known to inhibit the activities of ACSL1, intramitochondrial compartment
ACSL3, and ACSL4 [17,18], it is apparent that the above-mentioned
effects of triacsin C are mediated by the inhibition of these enzymes. In contrast to the biosynthesis of eicosanoids via the PLA2 pathway
Further, ACSL4 is likely to be involved in the incorporation of free AA mentioned above, the regulation of the release of AA from steroidogenic
following stimulation, since this isoform prefers PUFAs as its substrate. cells may use an alternative AA-releasing pathway mediated by the
Consistent with this notion, Golej et al. [19] have shown that the ACSL4-mitochondrial acyl-CoA thioesterase (ACOT2) pathway rather
overexpression of ACSL4 in human arterial smooth muscle cells reduced than the PLA2 pathway [22,23]. In this alternative pathway, ACSL4
the cytokine-dependent release of PGE2 compared to the release by catalyzes the conversion of intracellular free AA into AA-CoA and
mock-transfected cells. In addition, the treatment of these cells with provides it to ACOT2, which releases AA in mitochondria. The released
rosiglitazone (which inhibits ACSL4 activity [20]) as well as treatment AA in turn is metabolized by the lipoxygenase pathway to induce the

2
H. Kuwata and S. Hara Prostaglandins and Other Lipid Mediators 144 (2019) 106363

steroidogenic acute regulatory (StAR) protein [24,25]. StAR protein is a phospholipids compared to the adipocytes from control mice [36]. The
rate-limiting enzyme of steroid hormone biosynthesis, which controls lipidomic studies not only defined the role of ACSL4 in arachidonic acid
the transport of cholesterol into the inner mitochondrial membrane, incorporation within phospholipids; they also showed that the resulting
and the expression of StAR is regulated by AA or its metabolites in these linoleic acid incorporation increased with ACSL4 deficiency. Deficiency
cells [24,26,27]. of ACSL4 in adipocytes reduces the incorporation of AA into phos-
Several studies demonstrated that ACSL4 localizes on the mi- pholipids and AA pools, and also reduces the levels of the AA lipid
tochondria-associated membrane, endoplasmic reticulum, or peroxi- peroxidation product 4-hydroxynonenal [36]. These alternations of
somes of the liver and certain cancer cells [28–30]. Although further lipid composition by ACSL4 deficiency resulted in the protection of the
research is needed to clarify how ACSL4 can cross-talk with ACOT2, it high-fat diet-induced accumulation of adipose and liver fat, adipocyte
has been speculated that AA-CoA produced by ACSL4 at specific cellular death, gonadal white adipocyte tissue inflammation, and insulin re-
compartments may translocate to mitochondria via the acyl-CoA sistance, suggesting that ACSL4 is involved in the obesity-associated
binding protein DBI and the mitochondrial outer membrane protein adipocyte dysfunction. Thus, the control of the levels of PUFA-con-
TSPO [23]. taining phospholipids by ACSL4 enzyme may be involved in various
The perturbation of the regulation of AA concentration by the physiological and pathological processes.
knockdown of ACSL4 and ACOT2 markedly attenuated the adreno-
corticotropin (ACTH)- or cAMP-induced release of steroid hormone
such as progesterone, and supplementation with AA recovered the 5. ACSL4 in the brain
steroid hormone release from ACTH-treated steroidogenic cells [26,31].
Thus, the AA released by the ACSL4-ACOT2 pathway in mitochondria is ACSL4 transcripts are detected in a wide range of tissues, with
essential for steroidogenesis. Cho et al. demonstrated that female mice marked differences in their expression levels. Two splicing variants
heterozygous for ACSL4 deficiency had reduced fertility accompanied have been identified in ACSL4; ACSL4 variant 1 is a ubiquitous variant
by morphological changes of and prostaglandin accumulation in uterine of ACSL4, and ACSL4 variant 2 appears to be restricted to the brain
tissue [32]. Although the mechanism underlying the reduced fertility [6,7]. The ACSL4 variant 2 contains an N-terminal 41-amino acids ex-
observed in these mice is uncertain, it is possible that both perturbed tension sequence, which is predicted to form an integral membrane-
prostaglandin production and reduced AA supplementation via the associated domain. When these ACSL4 variants were transfected in
ACSL4-ACOT2 pathway (both of which attenuate steroid hormone COS-7 cells, the ACSL4 variant 1 localized to the inside of the plasma
production) may affect female fertility. membrane, whereas ACSL4 variant 2 located at the endoplasmic re-
Little is known about the cell types that use the ACSL4-ACOT2 axis ticulum and on lipid droplets [34]. Those results also showed that the
for AA release, but it has been established that the ACSL4-ACOT2 axis is overexpression of ACSL4 variants significantly increased the cellular
used in breast cancer cells and in steroidogenic cells. Maloberti et al. uptake of free AA to the same degree, suggesting that the ability of AA
showed that the expression levels of ACSL4 are correlated with the trapping is independent of the subcellular localization of ACSL4 var-
malignancy of breast cancer [33]. In that study, the knockdown of iants.
ACSL4 in the highly aggressive human breast cancer cell line MDA-MB- Although the functional differences between ACSL4 splicing var-
231 decreased the cells' proliferation, invasion, and migration, accom- iants are currently unknown, the differences in the subcellular locali-
panied by a reduction of several lipoxygenase products. The addition of zation of ACSL4 variants may contribute to their specific functions.
these LOX products restored the reduced cell proliferation induced by Meloni et al. showed that ACSL4 variant 2 is expressed in the human
knockdown of ACSL4. Thus, lipid mediators produced via the ACSL4- brain, and the cerebellum and corpus callosum in particular express
ACOT2 pathway may regulate the progression of certain cancers. only ACSL4 variant 2 [37]. ACSL4 variant 2 may exert its specific
functions in those brain regions, Other regions (e.g., the hippocampus)
4. The effects of ACSL4 on AA uptake and phospholipid express both ACSL4 variants. It is thus important to determine why
composition ACSL4 variants are distributed in specific regions of the brain and to
elucidate their functions.
Because ACSL4 prefers PUFAs such as AA as a substrate, this pre- Mutations of ACSL4 gene in human were found in two families with
ference seems to affect the fatty acid composition of several lipids. nonspecific mental retardation [38,39]. One of the mutations was
Indeed, there are several reports about the roles of ACSL4 in the cellular missense and the other was a splice-site change in ACSL4 gene. The
uptake of extracellular AA. The overexpression of ACSL4 in human level of the ACSL activity of lymphoblastoid cell lines from the affected
arterial smooth muscle cells markedly increased the incorporation of subjects of both families was low levels compared to normal cells. Thus,
exogenous AA into phosphatidylethanolamines and phosphatidylinosi- both mutations are null mutations. In the brain, the expression of
tols [19]. Similarly, the overexpression of ACSL4 in fibroblast-like COS- ACSL4 is localized in specific regions [39,40]. In the cerebellum, ACSL4
7 cells facilitated the incorporation of exogenous AA as well as oleic is expressed in the soma of Purkinje and granular cells and in the
acid (OA) in phosphatidylinositols [34]. In COS-7 cells, exogeneous AA, proximal dendritic region of Purkinje cells. In the hippocampus, ACSL4
but not OA, is also incorporated into phosphatidylcholines. Thus, the is expressed in the soma of pyramidal cells. These results suggest that
effects of ACSL4 on exogenous fatty acid incorporation may depend on ACSL4 is expressed in neurons and not in glial cells, and the decreased
the cell type. expression of ACSL4 appears to be related to development of the two
One of our investigations revealed that the knockdown of en- families’ mental retardation. It is unclear how the reduced ACSL4 ac-
dogenous ACSL4 expression in 3Y1 cells reduced the levels of several tivity could lead to mental retardation, and it is not known whether the
phosphatidylcholine and phosphatidylinositol species bearing AA fol- overproduction of eicosanoids or an alternation of the phospholipid
lowing IL-1β stimulation [15]. In this case, the levels of AA-containing composition in ACSL4 deficiency contributes to mental retardation.
phospholipids are minimally affected by the knockdown of ACSL4. However, because the inhibition of ACSL4 enhances AA-induced
More recently, it was observed that not only the levels of AA-containing apoptosis [41], it has been speculated that a decreased expression of
phospholipids but also those of other PUFA-bearing phospholipids are ACSL4 in brain may lead to precocious apoptosis in neurons and to a
regulated by ACSL4 [35,36]. The ablation of ACSL4 in mouse em- disruption of brain development [39]. Meloni et al. showed that ACSL4
bryonic fibroblasts markedly decreased the levels of PUFA-containing plays an important role in dendritic spine generation [37]. The me-
phosphatidylethanolamines [35], suppressing ferroptosis (see below). chanisms underlying the roles of ACSL4 in neural developments are
Further, adipocytes obtained from adipocyte-specific ACSL4-deficient thus of great interest.
mice fed a high-fat diet had reduced ability to incorporate PUFA into

3
H. Kuwata and S. Hara Prostaglandins and Other Lipid Mediators 144 (2019) 106363

6. ACSL4 in cancer Ferroptosis, one of the regulated cell death processes that is distinct
from other types of cell-regulated cell death (e.g., apoptosis, necrosis,
In addition to breast cancer, ACSL4 has been found to be upregu- and autophagic cell death) is executed via reactive oxygen (ROS)- and
lated in several cancers including, colon, liver, and prostate cancer iron-dependent mechanisms [55–57]. Several ferroptosis-inducing
[33,42–45]. Cao et al. [42] reported that they detected ACSL4 expres- molecules have been discovered, and it has been revealed that the
sion predominantly in colon epithelium, and the signals were much disturbances of the appropriate controls of intracellular redox-related
higher in adenocarcinoma. In contrast, the expression level of ACSL4 in product levels, including glutathione as well as lipid hydroperoxide, are
colon adenoma was comparable to that in the adjacent normal tissues, important for the execution of this process of regulated cell death. For
suggesting that the upregulation of ACSL4 expression is correlated with example, the inhibition of glutamate/cysteine antiporter system Xc−, a
colon cancer development. A high expression level of ACSL4 is also key regulator of intracellular glutathione levels that causes a reduction
observed in human hepatocellular carcinoma and several hepatoma cell of cellular antioxidant glutathione, leads to ferroptosis. In line with this,
lines [46]. The forced expression of ACSL4 in ACSL4-negative hepato- the depletion of glutathione inactivates glutathione peroxidases (GPXs)
cellular carcinoma SNU398 cells promoted cancer cell growth and in- such as GPX4, which leads to the accumulation of oxidized phospho-
hibited ACSL inhibitor triacsin C-induced cell death; conversely, lipids and the induction of ferroptosis [58]. Treatments with lipophilic
triacsin C inhibited the growth of Hep 3B cells (which express high antioxidants suppressed the ferroptosis induced by the depletion of
levels of ACSL4), attenuating cell proliferation [43]. glutathione or by the inhibition of GPX4 [55]; thus, the accumulation of
Similar to hepatocellular carcinoma, the expression level of ACSL4 peroxidized phospholipid species is one of the critical events inducing
protein is upregulated in malignant prostatic tissues compared to be- ferroptosis.
nign tissues. It was revealed that the expression of ACSL4 in both breast Because GPX4 is a unique enzyme that can remove lipid peroxides
and prostate cancer cells is inversely correlated with sex hormone re- from membrane phospholipids [59] and since PUFA-containing phos-
ceptor expression [47]. In prostate cancer LNCaP cells, which express a pholipids are potential targets for lipid peroxidation, the biosynthetic
trace amount of ACSL4, the transfection of ACSL4 cDNA increased cell pathway for PUFA-containing phospholipids is important for the ex-
proliferation, migration, invasion, and anchorage-independent cell ecution of ferroptosis. Indeed, by performing a massive insertional
growth, whereas the knockdown of ACSL4 in prostate cancer cells ex- mutagenesis analysis, Dixon et al. identified ACSL4 and lysopho-
pressing endogenous ACSL4 attenuated cell proliferation, migration, sphatidylcholine acyl transferase 3, both of which are important for the
and invasion [45]. biosynthesis of PUFA-containing phospholipids as key enzymes for
In human gastric cancer, however, the expression of ACSL4 is fre- ferroptosis [60]. In addition, Yuan et al. showed that erastin dose-de-
quently down-regulated. Ye et al. [48] showed that approx. 50% of pendently induced ferroptotic cell death in ACSL4-expressing cells
gastric cancer specimens decreases in the expression levels of ACSL4 (such as HepG2 and HL60 cells) but had little effect on ACSL4-negative
mRNA by 67% of noncancerous gastric mucosa. The overexpression of cells (such as LNCaP and K562 cells); further, the knockdown of ACSL4
ACSL4 in gastric cancer cells inhibits cell growth and cell migration, in the former cell lines protected erastin-induced ferroptosis, and the
and, conversely, the knockdown of ACSL4 increases them. These results overexpression of ACSL4 in the latter cell lines facilitated ferroptosis
are inconsistent with the ACSL4 functions observed in the above-men- [61]. Thus, phospholipid peroxides produced via the ACSL4 pathway
tioned cancers, and further studies are needed to explain these dis- are considered the ‘executioners’ of ferroptosis, and researchers have
crepancies. tried to find the enigmatic lipid peroxidation products inducing fer-
More recently, Chen et al. analyzed the roles of ACSL4 as well as roptosis. AA- and adrenic acid (AdA)-containing phosphatidylethano-
other ACSL isozymes in various cancers by performing a systematic lamines, both of which are produced via the ACSL4 pathway, are oxi-
analysis of gene alterations and clinical outcomes [49]. The analysis dized during ferroptosis, and their peroxidized products act as the
revealed that ACSL4 is upregulated in colorectal, head and neck, executioners of ferroptosis [35,62].
kidney, myeloma, and liver cancer, but downregulated in bladder,
brain, breast, leukemia, and lung cancer. The data also showed that a
8. Conclusions
high expression of ACSL4 is associated with poor survival of patients
with colorectal cancer: in contrast, a low expression of ACSL4 is asso-
Long-chain fatty acids are important molecules that are active in
ciated with poor survival of patients with brain, breast, or lung cancer.
various metabolic and catabolic processes including energy storage and
These data clearly indicate a relationship between ACSL4 expression
membrane formation, and as a source of bioactive lipid mediators. The
pattern and cancer progression. However, the results obtained with
long-chain fatty acids in these processes must be activated by ACSL
breast cancer tissue are controversial because some findings are in-
enzymes in order to be used. The dietary intake of n-3 and n-6 PUFAs
consistent with these results [50], and more detailed investigations are
affects human health. Under these circumstances, ACSL4 activates
needed to better understand these relationships. In addition, the in-
PUFAs and participates in several biological responses such as in-
ducible expression of ACSL4 in these cancerous tissues seems to facil-
flammation, pregnancy, and cancer development and progression as
itate or inhibit cancer cell progression, and ACSL4 may thus become the
well as cell death, via the synthesis of PUFA-derived CoAs. ACSL4 is
molecular target for particular types of cancer drug development.
thus one of the principal enzymes that regulate the availability of
PUFAs, and ACSL4 might therefore become a potential target in the
7. ACSL4 in cell death
treatment of PUFA-mediated exacerbations of disease.
PUFAs have the potential to cause cytotoxicity that is either de-
pendent on or independent of oxidative stress against various types of Acknowledgments
cells, and the signaling pathways causing cytotoxic effects differ be-
tween cell types [41,51–53]. In some cases, ceramide- or unfolded We thank the members of our lab for their helpful discussions and
protein response (UPR)-mediated apoptosis is involved in the PUFA- comments. This work was supported in part by Grants-in-Aid for
induced cytotoxicity [52,54]. Because these cell death signals are in- Scientific Research (B) (nos. 25293033 and 16H05108) from the Japan
itiated by an excess amount of PUFA, the removal of unesterified PUFAs Society for the Promotion of Science, and by a Grant-in-Aid for
by their metabolic enzymes is a plausible mechanism of the suppression Scientific Research on Innovative Areas (no. 25116720) and a grant for
of PUFA-induced cytotoxic effects. Indeed, the overexpression of a a Private University High Technology Research Center Project from the
PUFA metabolic enzyme (such as COX-2 or ACSL4) reduces AA-induced Ministry of Education, Sports, Science, Culture and Technology of
apoptosis [41]. Japan.

4
H. Kuwata and S. Hara Prostaglandins and Other Lipid Mediators 144 (2019) 106363

References dependent and obligatory protein in the signal transduction pathway of steroido-
genic hormones, J. Mol. Endocrinol. 34 (2005) 655–666.
[27] X. Wang, C.L. Shen, M.T. Dyson, X. Yin, R.B. Schiffer, P. Grammas, D.M. Stocco, The
[1] P.A. Watkins, J.M. Ellis, Peroxisomal acyl-CoA synthetases, Biochim. Biophys. Acta involvement of epoxygenase metabolites of arachidonic acid in cAMP-stimulated
1822 (2012) 1411–1420. steroidogenesis and steroidogenic acute regulatory protein gene expression, J.
[2] S.J. Steinberg, J. Morgenthaler, A.K. Heinzer, K.D. Smith, P.A. Watkins, Very long- Endocrinol. 190 (2006) 871–878.
chain acyl-CoA synthetases. Human "bubblegum" represents a new family of pro- [28] T.M. Lewin, J.H. Kim, D.A. Granger, J.E. Vance, R.A. Coleman, Acyl-CoA synthetase
teins capable of activating very long-chain fatty acids, J. Biol. Chem. 275 (2000) isoforms 1, 4, and 5 are present in different subcellular membranes in rat liver and
35162–35169. can be inhibited independently, J. Biol. Chem. 276 (2001) 4674–4679.
[3] P.A. Watkins, D. Maiguel, Z. Jia, J. Pevsner, Evidence for 26 distinct acyl-coenzyme [29] Y. Radif, H. Ndiaye, V. Kalantzi, R. Jacobs, A. Hall, S. Minogue, M.G. Waugh, The
A synthetase genes in the human genome, J. Lipid Res. 48 (2007) 2736–2750. endogenous subcellular localisations of the long chain fatty acid-activating enzymes
[4] H. Iijima, T. Fujino, H. Minekura, H. Suzuki, M.J. Kang, T. Yamamoto, Biochemical ACSL3 and ACSL4 in sarcoma and breast cancer cells, Mol. Cell. Biochem. 448
studies of two rat acyl-CoA synthetases, ACS1 and ACS2, Eur. J. Biochem. 242 (2018) 275–286.
(1996) 186–190. [30] T.M. Lewin, C.G. Van Horn, S.K. Krisans, R.A. Coleman, Rat liver acyl-CoA syn-
[5] T. Fujino, M.J. Kang, H. Suzuki, H. Iijima, T. Yamamoto, Molecular characterization thetase 4 is a peripheral-membrane protein located in two distinct subcellular or-
and expression of rat acyl-CoA synthetase 3, J. Biol. Chem. 271 (1996) ganelles, peroxisomes, and mitochondrial-associated membrane, Arch. Biochem.
16748–16752. Biophys. 404 (2002) 263–270.
[6] M.J. Kang, T. Fujino, H. Sasano, H. Minekura, N. Yabuki, H. Nagura, H. Iijima, [31] P. Maloberti, R. Castilla, F. Castillo, F. Cornejo Maciel, C.F. Mendez, C. Paz,
T.T. Yamamoto, A novel arachidonate-preferring acyl-CoA synthetase is present in E.J. Podestá, Silencing the expression of mitochondrial acyl-CoA thioesterase I and
steroidogenic cells of the rat adrenal, ovary, and testis, Proc. Natl. Acad. Sci. U. S. A. acyl-CoA synthetase 4 inhibits hormone-induced steroidogenesis, FEBS J. 272
94 (1997) 2880–2884. (2005) 1804–1814.
[7] Y. Cao, E. Traer, G.A. Zimmerman, T.M. McIntyre, S.M. Prescott, Cloning, expres- [32] Y.Y. Cho, M.J. Kang, H. Sone, T. Suzuki, M. Abe, M. Igarashi, T. Tokunaga,
sion, and chromosomal localization of human long-chain fatty acid-CoA ligase 4 S. Ogawa, Y.A. Takei, T. Miyazawa, H. Sasano, T. Fujino, T.T. Yamamoto, Abnormal
(FACL4), Genomics 49 (1998) 327–330. uterus with polycysts, accumulation of uterine prostaglandins, and reduced fertility
[8] T.J. Grevengoed, E.L. Klett, R.A. Coleman, Acyl-CoA metabolism and partitioning, in mice heterozygous for acyl-CoA synthetase 4 deficiency, Biochem. Biophys. Res.
Annu. Rev. Nutr. 34 (2014) 1–30. Commun. 284 (2001) 993–997.
[9] E.L. Klett, S. Chen, M.L. Edin, L.O. Li, O. Ilkayeva, D.C. Zeldin, C.B. Newgard, [33] P.M. Maloberti, A.B. Duarte, U.D. Orlando, M.E. Pasqualini, A.R. Solano, C. Lopez-
R.A. Coleman, Diminished acyl-CoA synthetase isoform 4 activity in INS 832/13 Otin, E.J. Podesta, Functional interaction between acyl-CoA synthetase 4, lipoox-
cells reduces cellular epoxyeicosatrienoic acid levels and results in impaired glu- ygenases and cyclooxygenase-2 in the aggressive phenotype of breast cancer cells,
cose-stimulated insulin secretion, J. Biol. Chem. 288 (2013) 21618–21629. PLoS One 5 (2010) e15540.
[10] E.L. Klett, S. Chen, A. Yechoor, F.B. Lih, R.A. Coleman, Long-chain acyl-CoA syn- [34] E.M. Kuch, R. Vellaramkalayil, I. Zhang, D. Lehnen, B. Brugger, W. Sreemmel,
thetase isoforms differ in preferences for eicosanoid species and long-chain fatty R. Ehehalt, M. Poppelreuther, J. Fullekrug, Differentially localized acyl-CoA syn-
acids, J. Lipid Res. 58 (2017) 884–894. thetase 4 isoenzymes mediate the metabolic channeling of fatty acids towards
[11] K. Bernstrom, K. Kayganich, R.C. Murphy, F.A. Fitzpatrick, Incorporation and dis- phosphatidylinositol, Biochim. Biophys. Acta 1841 (2014) 227–239.
tribution of epoxyeicosatrienoic acids into cellular phospholipids, J. Biol. Chem. [35] S. Doll, B. Proneth, Y.Y. Tyurina, E. Panzilius, S. Kobayashi, I. Ingold, M. Irmler,
267 (1992) 3686–3690. J. Beckers, M. Aichler, A. Walch, H. Prokisch, D. Trumbach, G. Mao, F. Qu, H. Bayir,
[12] J.H. Capdevila, J.R. Falck, R.C. Harris, Cytochrome P450 and arachidonic acid J. Fullekrug, C.H. Scheel, W. Wurst, J.A. Schick, V.E. Kagan, J.P. Angeli, M. Conrad,
bioactivation: molecular and functional properties of the arachidonate mono- ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition, Nat.
oxygenase, J. Lipid Res. 41 (2000) 163–181. Chem. Biol. 13 (2017) 91–98.
[13] H. Tomoda, K. Igarashi, S. Omura, Inhibition of acyl-CoA synthetase by triacsins, [36] E.A. Killion, A.R. Reeves, M.A. El Azzouny, Q.W. Yan, D. Surujon, J.D. Griffin,
Biochim. Biophys. Acta 921 (1987) 595–598. T.A. Bowman, C. Wang, N.R. Matthan, E.L. Klett, D. Kong, J.W. Newman, X. Han,
[14] S. Oh-ishi, K. Yamaki, M. Abe, H. Tomoda, S. Omura, The acyl-CoA synthetase M.J. Lee, R.A. Coleman, A.S. Greenberg, A role for long-chain acyl-CoA synthetase-4
inhibitor triacsin C enhanced eicosanoid release in leukocytes, Jpn. J. Pharmacol. (ACSL4) in diet-induced phospholipid remodeling and obesity-associated adipocyte
59 (1992) 417–418. dysfunction, Mol. Metab. 9 (2018) 43–56.
[15] H. Kuwata, M. Yoshimura, Y. Sasaki, E. Yoda, Y. Nakatani, I. Kudo, S. Hara, Role of [37] I. Meloni, V. Parri, R. De Filippis, F. Ariani, R. Artuso, M. Bruttini, E. Katzaki,
long-chain acyl-coenzyme A synthetases in the regulation of arachidonic acid me- I. Longo, F. Mari, C. Bellan, C.G. Dotti, A. Renieri, The XLMR gene ACSL4 plays a
tabolism in interleukin 1β-stimulated rat fibroblasts, Biochim. Biophys. Acta 1841 role in dendritic spine architecture, Neuroscience 159 (2009) 657–669.
(2014) 44–53. [38] M. Piccini, F. Vitelli, M. Bruttini, B.R. Pober, J.J. Jonsson, M. Villanova, M. Zollo,
[16] M.T. Weis, M. Brady, M. Moore, J. Crumley, J.N. Stallone, Inhibiting long-chain G. Borsani, A. Ballabio, Renieri A. FACL4, A new gene encoding long-chain acyl-
fatty acyl CoA synthetase does not increase agonist-induced release of arachidonate CoA synthetase 4, is deleted in a family with Alport syndrome, elliptocytosis, and
metabolites from human endothelial cells, J. Vasc. Res. 42 (2005) 275–283. mental retardation, Genomics 47 (1998) 350–358.
[17] J.H. Kim, T.M. Lewin, R.A. Coleman, Expression and characterization of re- [39] I. Meloni, M. Muscettola, M. Raynaud, I. Longo, M. Bruttini, M.P. Moizard,
combinant rat Acyl-CoA synthetases 1, 4, and 5. Selective inhibition by triacsin C M. Gomot, J. Chelly, V. des Portes, J.P. Fryns, H.H. Ropers, B. Magi, C. Bellan,
and thiazolidinediones, J. Biol. Chem. 276 (2001) 24667–24673. N. Volpi, H.G. Yntema, S.E. Lewis, J.E. Schaffer, A. Renieri, FACL4, encoding fatty
[18] C.G. Van Horn, J.M. Caviglia, L.O. Li, S. Wang, D.A. Granger, R.A. Coleman, acid-CoA ligase 4, is mutated in nonspecific X-linked mental retardation, Nat.
Characterization of recombinant long-chain rat acyl-CoA synthetase isoforms 3 and Genet. 30 (2002) 436–440.
6: identification of a novel variant of isoform 6, Biochemistry 44 (2005) [40] Y. Cao, K.J. Murphy, T.M. McIntyre, G.A. Zimmerman, S.M. Prescott, Expression of
1635–1642. fatty acid-CoA ligase 4 during development and in brain, FEBS Lett. 467 (2000)
[19] D.L. Golej, B. Askari, F. Kramer, S. Barnhart, A. Vivekanandan-Giri, S. Pennathur, 263–267.
K.E. Bornfeldt, Long-chain acyl-CoA synthetase 4 modulates prostaglandin E₂ re- [41] Y. Cao, A.T. Pearman, G.A. Zimmerman, T.M. McIntyre, S.M. Prescott, Intracellular
lease from human arterial smooth muscle cells, J. Lipid Res. 52 (2011) 782–793. unesterified arachidonic acid signals apoptosis, Proc. Natl. Acad. Sci. U. S. A. 97
[20] B. Askari, J.E. Kanter, A.M. Sherrid, D.L. Golej, A.T. Bender, J. Liu, W.A. Hsueh, (2000) 11280–11285.
J.A. Beavo, R.A. Coleman, K.E. Bornfeldt, Rosiglitazone inhibits acyl-CoA synthe- [42] Y. Cao, K.B. Dave, T.P. Doan, S.M. Prescott, Fatty acid CoA ligase 4 is up-regulated
tase activity and fatty acid partitioning to diacylglycerol and triacylglycerol via a in colon adenocarcinoma, Cancer Res. 61 (2001) 8429–8434.
peroxisome proliferator-activated receptor-gamma-independent mechanism in [43] Y.C. Liang, C.H. Wu, J.C. Chu, C.K. Wang, L.F. Hung, Y.J. Wang, Y.S. Ho,
human arterial smooth muscle cells and macrophages, Diabetes 56 (2007) J.G. Chang, S.Y. Lin, Involvement of fatty acid-CoA ligase 4 in hepatocellular car-
1143–1152. cinoma growth: roles of cyclic AMP and p38 mitogen-activated protein kinase,
[21] H. Kuwata, S. Hara, Inhibition of long-chain acyl-CoA synthetase 4 facilitates pro- World J. Gastroenterol. 11 (2005) 2557–2563.
duction of 5, 11-dihydroxyeicosatetraenoic acid via the cyclooxygenase-2 pathway, [44] Y.K. Sung, M.K. Park, S.H. Hong, S.Y. Hwang, M.H. Kwack, J.C. Kim, M.K. Kim,
Biochem. Biophys. Res. Commun. 465 (2015) 528–533. Regulation of cell growth by fatty acid-CoA ligase 4 in human hepatocellular car-
[22] A.F. Castillo, F. Cornejo Maciel, R. Castilla, A. Duarte, P. Maloberti, C. Paz, cinoma cells, Exp. Mol. Med. 39 (2007) 477–482.
E.J. Podesta, cAMP increases mitochondrial cholesterol transport through the in- [45] X. Wu, F. Deng, Y. Li, G. Daniels, X. Du, Q. Ren, J. Wang, L.H. Wang, Y. Yang,
duction of arachidonic acid release inside this organelle in Leydig cells, FEBS J. 273 V. Zhang, D. Zhang, F. Ye, J. Melamed, M.E. Monaco, P. Lee, ACSL4 promotes
(2006) 5011–5021. prostate cancer growth, invasion and hormonal resistance, Oncotarget 6 (2015)
[23] P. Maloberti, F. Cornejo Maciel, A.F. Castillo, R. Castilla, A. Duarte, M.F. Toledo, 44849–44863.
F. Meuli, P. Mele, C. Paz, E.J. Podestá, Enzymes involved in arachidonic acid release [46] Y.K. Sung, S.Y. Hwang, M.K. Park, H.I. Bae, W.H. Kim, J.C. Kim, M. Kim, Fatty acid-
in adrenal and Leydig cells, Mol. Cell. Endocrinol. 265–266 (2007) 113–120. CoA ligase 4 is overexpressed in human hepatocellular carcinoma, Cancer Si. 94
[24] X.J. Wang, M.T. Dyson, Y. Jo, D.W. Eubank, D.M. Stocco, Involvement of 5-lipox- (2003) 421–424.
ygenase metabolites of arachidonic acid in cyclic AMP-stimulated steroidogenesis [47] M.E. Monaco, C.J. Creighton, P. Lee, X. Zou, M.K. Topham, D.M. Stafforini,
and steroidogenic acute regulatory protein gene expression, J. Steroid Biochem. Expression of long-chain fatty acyl-CoA synthetase 4 in breast and prostate cancers
Mol. Biol. 85 (2003) 159–166. is associated with sex steroid hormone receptor negativity, Transl. Oncol. 3 (2010)
[25] X. Wang, L.P. Walsh, A.J. Reinhart, D.M. Stocco, The role of arachidonic acid in 91–98.
steroidogenesis and steroidogenic acute regulatory (StAR) gene and protein ex- [48] X. Ye, Y. Zhang, X. Wang, Y. Li, Y. Gao, Tumor-suppressive functions of long-chain
pression, J. Biol. Chem. 275 (2000) 20204–20209. acyl-CoA synthetase 4 in gastric cancer, IUBMB Life 68 (2016) 320–327.
[26] F. Cornejo Maciel, P. Maloberti, I. Neuman, F. Cano, R. Castilla, F. Castillo, C. Paz, [49] W.C. Chen, C.Y. Wang, Y.H. Hung, T.Y. Weng, M.C. Yen, M.D. Lai, Systematic
E.J. Podesta, An arachidonic acid-preferring acyl-CoA synthetase is a hormone- analysis of gene expression alterations and clinical outcomes for long-chain acyl-

5
H. Kuwata and S. Hara Prostaglandins and Other Lipid Mediators 144 (2019) 106363

coenzyme A synthetase family in cancer, PLoS One 11 (2016) e0155660. Biol. 26 (2016) 165–176.
[50] X. Wu, Y. Li, J. Wang, X. Wen, M.T. Marcus, G. Daniels, D.Y. Zhang, F. Ye, [57] Y. Xie, W. Hou, X. Song, Y. Yu, J. Huang, X. Sun, R. Kang, D. Tang, Ferroptosis:
L.H. Wang, X. Du, S. Adams, B. Singh, J. Zavadil, P. Lee, M.E. Monaco, Long chain process and function, Cell Dearh Differ 23 (2016) 369–379.
fatty acyl-CoA synthetase 4 is a biomarker for and mediator of hormone resistance [58] W.S. Yang, R. SriRamaratnam, M.E. Welsch, K. Shimada, R. Skouta,
in human breast cancer, PLoS One 8 (2013) e77060. V.S. Viswanathan, J.H. Cheah, P.A. Clemons, A.F. Shamji, C.B. Clish, L.M. Brown,
[51] A. Zajdel, A. Wilczok, M. Tarkowski, Toxic effects of n-3 polyunsaturated fatty acids A.W. Girotti, V.W. Cornish, S.L. Schreiber, B.R. Stockwell, Regulation of ferroptotic
in human lung A549 cells, Toxicol. In Vitro 30 (2015) 486–491. cancer cell death by GPX4, Cell 156 (2014) 317–331.
[52] S. Akagi, N. Kono, H. Ariyama, H. Shindou, T. Shimizu, H. Arai, [59] H. Imai, Y. Nakagawa, Biological significance of phospholipid hydroperoxide glu-
Lysophosphatidylcholine acyltransferase 1 protects against cytotoxicity induced by tathione peroxidase (PHGPx, GPx4) in mammalian cells, Free Radic. Biol. Med. 34
polyunsaturated fatty acids, FASEB J. 30 (2016) 2027–2039. (2003) 145–169.
[53] S. Serini, S. Trombino, F. Oliva, E. Piccioni, G. Monego, F. Resci, A. Boninsegna, [60] S.J. Dixon, G.E. Winter, L.S. Musavi, E.D. Lee, B. Snijder, M. Rebsamen, G. Superti-
N. Picci, F.O. Ranelletti, G. Calviello, Docosahexaenoic acid induces apoptosis in Furga, B.R. Stockwell, Human haploid cell genetics reveals roles for lipid metabo-
lung cancer cells by increasing MKP-1 and down-regulating p-ERK1/2 and p-p38 lism genes in nonapoptotic cell death, ACS Chem. Biol. 10 (2015) 1604–1609.
expression, Apoptosis 13 (2008) 1172–1183. [61] H. Yuan, X. Li, X. Zhang, R. Kang, D. Tang, Identification of ACSL4 as a biomarker
[54] T.A. Chan, P.J. Morin, B. Vogelstein, K.W. Kinzler, Mechanisms underlying non- and contributor of ferroptosis, Biochem. Biophys. Res. Commun. 478 (2016)
steroidal antiinflammatory drug-mediated apoptosis, Proc. Natl. Acad. Sci. U. S. A. 1338–1343.
95 (1998) 681–686. [62] V.E. Kagan, G. Mao, F. Qu, J.P. Angeli, S. Doll, C.S. Croix, H.H. Dar, B. Liu,
[55] S.J. Dixon, K.M. Lemberg, M.R. Lamprecht, R. Skouta, E.M. Zaitsev, C.E. Gleason, V.A. Tyurin, V.B. Ritov, A.A. Kapralov, A.A. Amoscato, J. Jiang, T. Anthonymuthu,
D.N. Patel, A.J. Bauer, A.M. Cantley, W.S. Yang, Morrison 3rd B, Stockwell BR. D. Mohammadyani, Q. Yang, B. Proneth, J. Klein-Seetharaman, S. Watkins, I. Bahar,
Ferroptosis: an iron-dependent form of nonapoptotic cell death, Cell 149 (2012) J. Greenberger, R.K. Mallampalli, B.R. Stockwell, Y.Y. Tyurina, M. Conrad, H. Bayir,
1060–1072. Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis, Nat. Chem. Biol.
[56] W.S. Yang, B.R. Stockwell, Ferroptosis: death by lipid peroxidation, Trends Cell 13 (2017) 81–90.

You might also like