Next Article in Journal
Is There a Role of Warburg Effect in Prostate Cancer Aggressiveness? Analysis of Expression of Enzymes of Lipidic Metabolism by Immunohistochemistry in Prostate Cancer Patients (DIAMOND Study)
Next Article in Special Issue
The Epigenetic Reader Methyl-CpG-Binding Protein 2 (MeCP2) Is an Emerging Oncogene in Cancer Biology
Previous Article in Journal
The Efficacy and Safety of Immune Checkpoint Inhibitor and Tyrosine Kinase Inhibitor Combination Therapy for Advanced or Metastatic Renal Cell Carcinoma: A Multicenter Retrospective Real-World Cohort Study
Previous Article in Special Issue
The Role of microRNAs in Multidrug Resistance of Glioblastoma
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Synthesis and Significance of Arachidonic Acid, a Substrate for Cyclooxygenases, Lipoxygenases, and Cytochrome P450 Pathways in the Tumorigenesis of Glioblastoma Multiforme, Including a Pan-Cancer Comparative Analysis

by
Jan Korbecki
1,
Ewa Rębacz-Maron
2,
Patrycja Kupnicka
1,
Dariusz Chlubek
1 and
Irena Baranowska-Bosiacka
1,*
1
Department of Biochemistry and Medical Chemistry, Pomeranian Medical University in Szczecin, Powstańców Wlkp. 72, 70-111 Szczecin, Poland
2
Department of Ecology and Anthropology, Institute of Biology, University of Szczecin, Wąska 13, 71-415 Szczecin, Poland
*
Author to whom correspondence should be addressed.
Cancers 2023, 15(3), 946; https://doi.org/10.3390/cancers15030946
Submission received: 5 December 2022 / Revised: 25 January 2023 / Accepted: 31 January 2023 / Published: 2 February 2023
(This article belongs to the Special Issue Oncogenes and Tumor Suppressor Genes in Brain Tumor)

Abstract

:

Simple Summary

Glioblastoma multiforme is a brain tumor with a very unfavorable prognosis, where the vast majority of patients do not survive a year after diagnosis. One line of research that may help in designing more successful therapeutic approaches is the synthesis and metabolism of arachidonic acid, which is then converted into a large number of different lipid mediators, including prostaglandins and leukotrienes (by cyclooxygenases and lipoxygenases, respectively). In this paper, we discuss the synthesis of arachidonic acid in glioblastoma multiforme tumors as well as the significance of lipid mediators synthesized from arachidonic acid, which can increase the proliferation of glioblastoma multiforme cancer cells, cause angiogenesis, inhibit the anti-tumor response of the immune system, and be responsible for resistance to treatment.

Abstract

Glioblastoma multiforme (GBM) is one of the most aggressive gliomas. New and more effective therapeutic approaches are being sought based on studies of the various mechanisms of GBM tumorigenesis, including the synthesis and metabolism of arachidonic acid (ARA), an omega-6 polyunsaturated fatty acid (PUFA). PubMed, GEPIA, and the transcriptomics analysis carried out by Seifert et al. were used in writing this paper. In this paper, we discuss in detail the biosynthesis of this acid in GBM tumors, with a special focus on certain enzymes: fatty acid desaturase (FADS)1, FADS2, and elongation of long-chain fatty acids family member 5 (ELOVL5). We also discuss ARA metabolism, particularly its release from cell membrane phospholipids by phospholipase A2 (cPLA2, iPLA2, and sPLA2) and its processing by cyclooxygenases (COX-1 and COX-2), lipoxygenases (5-LOX, 12-LOX, 15-LOX-1, and 15-LOX-2), and cytochrome P450. Next, we discuss the significance of lipid mediators synthesized from ARA in GBM cancer processes, including prostaglandins (PGE2, PGD2, and 15-deoxy-Δ12,14-PGJ2 (15d-PGJ2)), thromboxane A2 (TxA2), oxo-eicosatetraenoic acids, leukotrienes (LTB4, LTC4, LTD4, and LTE4), lipoxins, and many others. These lipid mediators can increase the proliferation of GBM cancer cells, cause angiogenesis, inhibit the anti-tumor response of the immune system, and be responsible for resistance to treatment.

1. Introduction

Glioblastoma multiforme (GBM) is one of the most aggressive brain tumors and has the worst prognosis, with an average survival of about one year [1,2,3]. In order to either improve existing therapies or develop new approaches, the mechanisms of GBM tumorigenesis are being intensively investigated, including those involving arachidonic acid (ARA) C20:4n-6 and the lipid mediators formed from this fatty acid.
PUFAs, in particular arachidonic acid ARA C20:4n-6, eicosapentaenoic acid (EPA) C20:5n-3, and docosahexaenoic acid (DHA) C22:6n-3, can be converted into lipid mediators, such as eicosanoids [4], and pro-resolving lipid mediators [5]. Eicosanoids are 20-carbon lipid mediators synthesized from ARA C20:4n-6, dihomo-γ-linolenic acid C20:3n-6, and EPA C20:5n-3 using cyclooxygenases (COX) and lipoxygenases (LOX), resulting in the formation of prostaglandins and leukotrienes, respectively [4]. Eicosanoids have pro-inflammatory properties, although there are also lipid mediators with anti-inflammatory properties, such as 15-deoxy-Δ12,14-PGJ2 (15d-PGJ2) [6]. EPA and DHA can be converted into pro-resolving lipid mediators with LOX, cytochrome P450, and acetylated cyclooxygenase-2 (COX-2) [5]. This conversion produces lipoxins and resolvins, although it should be mentioned here that free PUFAs, including ARA, are the activators of peroxisome proliferator-activated receptors (PPAR)α and PPARγ [7].
All of the aforementioned groups of ARA metabolites have either pro- or anti-cancer properties in GBM tumors, which indicates their significance in GBM tumor development. Despite their important role, some groups of these lipid mediators are little-known and rarely studied, and there is no paper in the literature that reviews the body of research in this area. The aim of this paper is to fill this gap and at the same time generate more interest in the role of ARA metabolites in GBM.

2. Methodology

This study’s major objective is to characterize the significance of all ARA C20:4n-6-derived lipid mediators, their receptors, and the enzymes responsible for their production in the tumorigenic pathways in GBM. The PubMed search engine (https://pubmed.ncbi.nlm.nih.gov accessed on 1 October 2022) was used for this purpose. Due to the fact that many of the lipid mediators produced from ARA C20:4n-6 have not yet been investigated in the context of GBM, two additional sources were used to conduct a bioinformatic analysis of every gene in GBM, namely, the transcriptomics analysis carried out by Seifert et al. [8] and the Gene Expression Profiling Interactive Analysis (GEPIA) web server (http://gepia.cancer-pku.cn accessed on 20 October 2022) [9].
The analyses posted on the GEPIA portal include the analysis of nearly 10,000 samples from 33 different cancers deposited in the Cancer Genome Atlas (TCGA) [10] along with the analysis of more than 8000 healthy tissue samples posted in Genotype-Tissue Expression (GTEx) [11,12]. The GEPIA served as a source of data on differences in the expression of given genes between GBM tumor and healthy brain tissue, and for linking the expression of a given gene to GBM patient prognosis.
A transcriptomics analysis was performed by Seifert et al. [8] on nearly 17,000 different genes in various grades of glioma, including GBM, from 45 patients. These results were normalized with a control: an analysis of gene expression in brain samples from 21 epilepsy patients from the REpository of Molecular BRAin Neoplasia DaTa (Rembrandt) [13], which served as a second source of data on differences in the expression of genes between GBM tumors and healthy brain tissue.

3. Arachidonic Acid Biosynthesis and Glioblastoma Multiforme

3.1. Arachidonic Acid Biosynthesis

ARA C20:4n-6 in humans is not synthesized de novo but from linoleic acid C18:2n-6 in the PUFA biosynthesis pathway (Figure 1) [14]. Linoleic acid C18:2n-6 in its activated form, linoleoyl-CoA C18:2n-6, undergoes desaturation with fatty acid desaturase 2 (FADS2)/Δ6-desaturase (D6D), which is accompanied by the formation of γ-linolenoyl-CoA C18:3n-6. Subsequently, the hydrocarbon chain in this fatty acyl-CoA is elongated with two carbons through the elongation of the long-chain fatty acid family members 5 (ELOVL5), accompanied by the formation of dihomo-γ-linolenoyl-CoA C20:3n-6. At the same time, an alternative pathway for the synthesis of dihomo-γ-linolenoyl-CoA C20:3n-6 from linoleoyl-CoA C18:2n-6 is also possible [15]. Linoleoyl-CoA C18:2n-6 is first elongated with ELOVL5 and then desaturated by FADS2. This means that these two enzymes can catalyze the formation of dihomo-γ-linolenoyl-CoA C20:3n-6 in reverse order. In this alternative pathway of PUFA biosynthesis, FADS2 shows activity not of Δ6-desaturase but of Δ8-desaturase. In the latter reaction, the hydrocarbon chain in dihomo-γ-linolenoyl-CoA C20:3n-6 is desaturated with fatty acid desaturase 1 (FADS1)/Δ5-desaturase (D5D), which is accompanied by the production of arachidonyl-CoA C20:4n-6. In the same way as arachidonyl-CoA C20:4n-6, EPA-CoA C20:5n-3 can also be synthesized from α-linolenoyl-CoA C18:3n-3 [14]. Arachidonyl-CoA C20:4n-6 is an activated form of ARA that participates in metabolic pathways, including lipid synthesis pathways. Once synthesized, arachidonyl-CoA C20:4n-6 is used to make lipids, particularly phospholipids. Incorporated into phospholipids, ARA C20:4n-6 is stored and then released by phospholipases A2 (PLA2) as a free fatty acid [16]. Arachidonyl-CoA C20:4n-6 can also be further elongated via elongation of the long-chain fatty acid family members 2 (ELOVL2) and ELOVL5 in a synthesis pathway similar to the synthesis of DHA C22:6n-3 from EPA C20:5n-3 [14,17,18,19].

3.2. Arachidonic Acid Biosynthesis Pathway in Glioblastoma Multiforme Tumors

Expression of FADS2, an enzyme important for the viability and self-renewal of GBM cancer stem cells [20], is higher in GBM tumors than in healthy brain tissue, according to GEPIA [9] and the transcriptomics analysis performed by Seifert et al. [8]. However, our study showed that FADS2 may have lower expression in tumors than in the peritumoral area in GBM patients [21]. Discrepancies between our results and the data from GEPIA and transcriptomics analysis performed by Seifert et al. may have resulted from studying different groups of patients. FADS2 expression in GBM tumors does not differ between men and women [21]. According to the GEPIA portal, higher FADS2 expression does not affect the prognosis for GBM patients [9]. Studies in GBM models show that FADS2 expression is higher in GBM cancer stem cells than in other GBM cancer cells [20].
The expression of FADS1, which is also important for the viability and self-renewal of GBM cancer stem cells [20], does not differ between GBM tumors and healthy brain tissue, according to GEPIA [9], Seifert et al. [8], and previous results from our research team [21]. According to the GEPIA portal, a higher FADS1 expression does not affect the prognosis for GBM patients [9]. FADS1 expression is higher in GBM cancer stem cells than in other GBM cancer cells [20].
ELOVL5 expression is higher in GBM tumors compared to healthy brain tissue, according to GEPIA [9] and Seifert et al. [8]. However, previous results from our research team did not show significant differences in the expression of ELOVL5 in GBM tumor tissue versus the peritumoral area [22]. Discrepancies between our results and the data from GEPIA and transcriptomics analysis performed by Seifert et al. may have resulted from studying different groups of patients. In addition, we observed that ELOVL5 expression was lower in GBM tumors in women relative to both the peritumoral area and GBM tumors in men [22]. Higher ELOVL5 expression does not affect the prognosis for GBM patients, according to GEPIA [9]. ELOVL5 expression can be higher in a GBM tumor as a result of hypoxia, as shown by our experiments with U87 MG line cells [22]. This is very important because hypoxia in a GBM tumor also increases the expression of COX-2 [23], an enzyme that converts ARA into prostanoids. This means that hypoxia increases the production of ARA and, at the same time, its conversion into prostanoids.

4. Phospholipase A2 Superfamily and the Release of Arachidonic Acid from Cell Membrane Phospholipids in Glioblastoma Multiforme

4.1. Phospholipase A2 Superfamily

The production of prostaglandins and leukotrienes requires a substrate for COX and LOX, namely, free ARA C20:4n-6, which is cleaved from cell membrane phospholipids by PLA2. Enzymes with PLA2 activity cleave either a fatty acid or a short acyl group from phospholipids at the sn-2 position [16]. All of these enzymes form the phospholipase A2 superfamily, which can be divided into six types. Three of these types are important in the release of ARA C20:4n-6 as well as other PUFA from cell membrane phospholipids [16]:
  • cytosolic phospholipase A2 (cPLA2),
  • calcium-independent phospholipase A2 (iPLA2), and
  • secretory phospholipase A2 (sPLA2).
The remaining PLA2 types include:
  • platelet-activating factor acetyl hydrolases (PAF-AH),
  • lysosomal phospholipase A2, and
  • adipose phospholipase A2.
In humans, seven representatives of cPLA2 are distinguished, namely, cPLA2α/PLA2G4A to cPLA2ζ/PLA2G4F. These enzymes, activated by Ca2+ [16], belong to the group IV (GIV) PLA2. Significantly, cPLA2γ/PLA2G4C lacks a Ca2+ binding domain and is not sensitive to this second messenger [24]. cPLA2α is additionally activated by phosphorylation and has the highest activity towards phosphatidylcholine (PC), phosphatidylethanolamine (PE), and, to a lesser extent, towards other glycerophospholipids [16]. cPLA2 have a specificity for cleaving PUFA from glycerophospholipids, particularly ARA C20:4n-6. cPLA2α shows the highest specificity for cleaving ARA C20:4n-6 [25,26], to a lesser extent, EPA C20:5n-3, and, to an even lesser extent, other PUFAs, e.g., linoleic acid C18:2n-3. cPLA2γ also has the highest specificity for cleaving ARA C20:4n-6 and a twice-lower specificity for cleaving both linoleic acid C18:2n-3 and oleic acid C16:1n-9 [26].
In humans, there are six representatives of iPLA2: iPLA2β to iPLA2η [16]. All of these enzymes belong to the GVI PLA2. They are activated by ATP [27], and their activity is independent of Ca2+ levels and reduced by calmodulin [28]. Enzymes in this group show different specificities for cleaving fatty acids from phospholipids at the sn-2 position. Depending on the enzymes, they show a higher ability to release a given fatty acid, e.g., oleic acid C16:1n-9 [27] or ARA C20:4n-6 [29].
Seventeen different groups of PLA2 have been classified to date, which includes sPLA2 [16]. Some sPLA2 groups consist of only the sPLA2 found in the venom of snakes, insects such as bees, and scorpions [16,30,31,32]. In humans, there are nine representatives of sPLA2 [16]. These enzymes cleave fatty acids from phospholipids at the sn-2 position without showing specificity to a particular fatty acid [16,33]. Once secreted into the intercellular space, sPLA2 not only cause the release of ARA C20:4n-6 but can also activate their receptor PLA2R1 [34].
After fatty acids are cleaved from phospholipids by PLA2, free fatty acids are formed, most commonly ARA C20:4n-6 and lysophosphatidylcholine (LPC) if PC was the reaction substrate (Figure 2). LPC can then be converted to lysophosphatidic acid (LPA) by the action of enzymes with lysophospholipase D (lysoPLD) activity [35,36]. An extracellular enzyme with lysoPLD activity is autotaxin (ATX)/ENPP2 [35,36]. Importantly, if the substrate for PLA2 is phosphatidic acid (PA), then LPA is formed directly [37]. LPA is a lipid mediator that acts through its six receptors (from lysophosphatidic acid receptor 1 (LPAR1) to LPAR6) [38].

4.2. Cytosolic Phospholipase A2 and Calcium-Independent Phospholipase A2 in Glioblastoma Multiforme

Expression of cPLA2α/PLA2G4A is upregulated in GBM tumors compared to healthy brain tissue [39]. This is also confirmed by bioinformatics analysis on the GEPIA portal [9] and the transcriptomics analysis by Seifert et al. [8]. At the same time, the expression of cPLA2β/PLA2G4B is lower, and the expressions of cPLA2γ/PLA2G4C, cPLA2δ/PLA2G4D, cPLA2ε/PLA2G4E, and cPLA2ζ/PLA2G4F are unchanged, according to GEPIA [9]. The expression of cPLA2γ/PLA2G4C is lower, and cPLA2ζ/PLA2G4F is not different in GBM tumors relative to healthy brain tissue, according to the transcriptomics analysis by Seifert et al. [8]. For six of the iPLA2, expression in GBM tumor does not differ compared to healthy brain tissue, according to GEPIA [9]. The expression of iPLA2β/PLA2G6 and iPLA2δ/PNPLA6 is lower in GBM tumor than in the healthy brain, according to the transcriptomics analysis by Seifert et al. [8]. Expressions of the remaining iPLA2 do not differ between GBM tumors and healthy brain tissue.
In the case of iPLA2η/PNPLA4, higher expression in GBM tumors is associated with a worse prognosis for the patient, according to GEPIA (Table 1) [9]. For iPLA2ζ/PNPLA2, there is a trend (p = 0.087) of worse prognosis and higher expression of this gene in the GBM tumor.
cPLA2 are activated in GBM cells, in particular, by sPLA2 enzymes [40,41]. This is associated with the induction of cPLA2 phosphorylation via MAPK kinase cascades as well as with an increase in cytoplasmic Ca2+ levels via phospholipase C-γ (PLC-γ) activation.
cPLA2α increases the proliferation of GBM cells, although the effect is not large. The most significant property of cPLA2α in GBM cells is causing chemoresistance to temozolomide (TMZ) and other chemotherapeutics, such as doxorubicin and 5-fluorouracil [39]. At the same time, the increased activity of cPLA2 may also decrease the viability of GBM cells, where TMZ induces the phosphorylation of cPLA2. This increases the activation of this enzyme [42] and thus leads to an increase in the level of free ARA 20:4n-6, whose excess reduces the viability of GBM cells. The reason for this may be in the activation of PPAR by this fatty acid [7,43,44] and the generation of reactive oxygen species (ROS) [45].
PLA2 may also be important in the interaction of GBM cells with endothelial cells. GBM cells cause an increase in the expression and activity of cPLA2 and iPLA2 in endothelial cells [46,47]. An increase in cPLA2 activity in endothelial cells can also be caused by radiation therapy [48]. A rise in the activity of cPLA2 and iPLA2 leads to the production of LPA [49]. GBM cancer cells may also increase COX-2 expression in endothelial cells, which increases the production of prostanoids including prostaglandin E2 (PGE2) [47]. LPA and PGE2 increase the proliferation and migration of endothelial cells [46,47,49]. This is also a mechanism of angiogenesis as a side effect of GBM radiotherapy [47,48]. At the same time, angiogenesis can be inhibited by pericytes [47].
Dying endothelial cells in a GBM tumor can secrete PGE2 that increases the proliferation of GBM cells [50]. This is associated with the processing of iPLA2β by caspase 3 [16,51], which increases the activity of this iPLA2 and, thus, leads to an increase in PGE2 production [50].

4.3. Secretory Phospholipase A2 in Glioblastoma Multiforme

Analyses on the GEPIA portal indicate that PLA2G5 expression is higher in GBM tumors [9]. There is also elevated expression of PLA2G2A, PLA2G12A, and PLA2G15 but no other sPLA2 in GBM tumors [9]. The transcriptomics analysis by Seifert et al. showed that the expressions of PLA2G2A and PLA2G5 are higher in GBM tumors than in healthy brain tissue [8]. This is the same as the data from the GEPIA web server. However, Seifert et al. showed that the expression of PLA2G12A and of the other sPLA2 enzymes is not different in GBM tumors relative to healthy brain tissue [8]. Wu et al. also showed that PLA2G5 expression is higher in gliomas than in healthy tissue and increases with tumor grade [52].
Higher expression of certain sPLA2 in GBM tumors is associated with a worse prognosis. According to GEPIA, these include PLA2G1B and PLA2G15 [9]. Wu et al. showed a higher number of sPLA2 affecting prognosis. In particular, worse prognoses in patients with GBM are associated with higher expression of PLA2G1B, PLA2G2E, PLA2G3, and PLA2G5 [52].
PLA2G5 is significant for tumorigenesis in low-grade gliomas and GBM. This suggests that a high expression of this sPLA2 is associated with a worse prognosis in patients with GBM and low-grade gliomas (Table 2) [52]. Analyses on the GEPIA portal show no significant association between the expression of the aforementioned sPLA2 and the GBM patient prognosis [9].
sPLA2 are secreted outside the cells where they perform their function. They have their own receptor, PLA2R1, from the C-type lectin superfamily and mannose receptor family [34], located in the cell membrane, through which it passes once. According to both GEPIA [9] and Seifert et al. [8], PLA2R1 expression does not differ between GBM tumors and healthy brain tissue. An above-average expression of this receptor in a GBM tumor is associated with a worse prognosis for the patient [9], indicating that sPLA2 may act on PLA2R1 and be pro-tumorigenic.
sPLA2 may act by participating in the production of LPA, a lipid mediator that has six different receptors [38]. According to GEPIA, LPAR3 expression is downregulated in GBM tumors relative to healthy brain tissue [9], whereas LPAR5 and LPAR6 expression is upregulated in GBM tumors. The expression of other LPA receptors is not altered in GBM tumors. The transcriptomics analysis by Seifert et al. shows that LPAR1 expression is lower, and LPAR6 expression is higher in GBM tumors relative to healthy brain tissue [8]. The expression of other LPA receptors does not differ between GBM tumors and healthy brain tissue.
sPLA2 also have the same catalytic properties as other PLA2. They cause the release of ARA 20:4n-6 from cell membrane phospholipids; this reaction produces free ARA 20:4n-6 and LPC. The latter is converted into LPA in the intercellular space by ATX [53], which is secreted by GBM cancer cells [54,55] and whose expression in GBM tumors is higher than in healthy brain tissue [53] and is elevated by interaction with microglial cells [55]. At the same time, GEPIA reports that ATX expression is not altered in GBM tumors [9], and Seifert et al. showed that it is lower [8] than in healthy brain tissue. The level of ATX expression in the tumor is not associated with prognosis severity for patients with GBM [9].
Another important source of ATX in the GBM tumor microenvironment is microglial cells [55], where ATX expression is upregulated by GBM cells, especially under hypoxia. Microglial cells also express the LPAR1 receptor and can respond to LPA [55].
Increased expression of various sPLA2 [52] and ATX [53] in GBM tumors also results in increased LPA production. GBM cancer cells show a loss of primary cilia, which leads to an increase in the distribution of LPAR1 in the plasma membrane of these cells and to an enhancement of signal transduction by this receptor as a result of a greater association of G proteins with this receptor [56].
LPA causes GBM cells to migrate [53,54,55,57,58] due to the activation of LPAR1, which results in the activation of protein kinase C (PKC)α. This is responsible for the phosphorylation of the progesterone receptor at the Ser400 residue [59,60]. GBM cancer cell migration is also facilitated by the LPA-induced decrease in oligodendrocyte adhesion [54]. It is also worth mentioning that in addition to LPAR1, the receptor for advanced glycation end products (RAGE) may be another important receptor causing GBM cancer cell migration [61].
LPA increases the proliferation of GBM cancer cells [55]. The effect of LPA on proliferation depends on LPAR1 receptors [55] and RAGE [61], and it occurs via the activation of two signaling pathways. The first is the Rho → sodium-hydrogen antiporter 1 (NHE-1) pathway, which leads to an increase in intracellular pH and, thus, the proliferation of GBM cancer cells [62]. The second pathway is the activation of extracellular signal-regulated kinase (ERK) mitogen-activated protein kinase (MAPK) by the phosphatidylinositol-4,5-bisphosphate 3-kinase (PI3K) → PKC pathway [62], which can also be initiated by epidermal growth factor receptor (EGFR) transactivation. Studies on PLA2G2A have shown that this sPLA2 increases GBM cancer cell proliferation via EGFR transactivation [63,64,65]. This is associated with the activation of PKC, which activates EGFR [64]. EGFR activation results in the activation of the Src → ERK MAPK → Akt/PKB → mammalian target of rapamycin (mTOR) → ribosomal protein 70 S6 kinase (p70S6K) pathway [63,65]. Its consequence is an increase in the proliferation of GBM cancer cells.
sPLA2 can also increase GBM cancer cell proliferation indirectly through the activation of cPLA2 inside a GBM cell [40]. This process is independent of LPA.
LPA inhibits FasL-induced apoptosis [66] due to the LPA-induced activation of thyroid hormone receptor-interacting protein 6 (TRIP6). TPIP6 binds directly to Fas receptor (FasR)/CD95, which inhibits the induction of apoptosis by this receptor [66].
LPA causes radioresistance of GBM cancer cells [48,58]. These effects are a result of the LPA-induced activation of LPAR1 [53,55] and LPAR3 [48].
Phosphorylation of the progesterone receptor by LPA increases vascular endothelial growth factor (VEGF) expression in GBM cancer cells [60], the most important growth factor in angiogenesis. LPA is also important in radiotherapy-induced angiogenesis in GBM tumors [58]. An increase in tumor vascularization during exposure to ionizing radiation can be inhibited by ATX inhibitors, which could have some clinical application in future therapies against GBM [58].
The aforementioned actions of LPA were carried out on various models of specific GBM cell lines. Significantly, the action of LPA may be more pronounced in GBM cancer stem cells than non-cancer stem cells, as the former show much higher expression of LPAR1 and LPAR3 [67].
LPAR1 is important in the development of GBM. Higher expression of this receptor in GBM tumors is associated with a worse prognosis [55]. At the same time, an analysis on the GEPIA portal did not link LPAR1 and LPAR3 expression to prognosis severity for GBM patients [9]. In addition, it did not show that the expression of the other LPA receptors had an effect on the prognosis for GBM patients.

4.4. Pan-Cancer Analysis of Phospholipase A2 Genes and Comparison of GBM Expression against Other Cancers

We also performed a pan-cancer analysis of the expression of the PLA2 genes with the GEPIA portal [9].
In GBM, but not in lower grade gliomas, there is higher expression of cPLA2α/PLA2G4A compared to healthy brain tissue [8,9]. Among the analyzed 31 tumor types, only four more had higher expression of this PLA2, and eight other types showed a decrease. For this reason, higher expression of this enzyme in GBM tumors can be considered characteristic for this cancer.
In GBM, the expression of cPLA2 β/PLA2G4B is decreased relative to healthy brain tissue [9], similar to lower grade glioma and 19 other types of cancer. This indicates that the decreased expression of this PLA2 in tumor is a hallmark of cancer.
Seifert et al. also indicates that cPLA2γ/PLA2G4C expression may be downregulated in GBM tumors relative to healthy brain tissue [8]. According to a pan-cancer analysis based on the GEPIA, cPLA2γ/PLA2G4C expression is downregulated in nine types of tumors but not in GBM or lower grade gliomas, whereas it is upregulated in seven types of tumors [9]. Changes in cPLA2γ/PLA2G4C expression in GBM tumors could be a hallmark of cancer.
Seifert et al. also showed a decrease in the expression of iPLA2β/PLA2G6 and iPLA2δ/PNPLA6 in GBM tumors relative to healthy brain tissue [8]. According to GEPIA, iPLA2β/PLA2G6 expression is downregulated in 15 tumor types (Table 3) [9], whereas iPLA2δ/PNPLA6 expression is only downregulated in three types. For this reason, it can be thought that decreased iPLA2β/PLA2G6 expression may be a hallmark of cancer. In contrast, reduced expression of iPLA2δ/PNPLA6 is characteristic of GBM.
Available sources [8,9] show that PLA2G2A, PLA2G5, PLA2G12A, and PLA2G15 undergo increased expression in GBM relative to healthy brain tissue. Changes in sPLA2’s expression in GBM are characteristic of this cancer. All listed sPLA2 undergo increased expression only in certain types of cancer (apart from GBM): PLA2G2A (in 2); PLA2G5 (in 1); PLA2G12A (in 4); PLA2G15 (in 3).
PLA2G2A expression is downregulated in 18 out of 31 types of cancer, indicating that it is generally downregulated in cancer (Table 4). In contrast, increased expression of PLA2G2A may occur in GBM [8,9], which may be characteristic of GBM. On the other hand, in 17 out of 31 cancers, there is a higher expression of PLA2G7 in the tumor than in healthy tissue. Its expression in a GBM tumor is not different from its expression in healthy brain tissue [8,9].

4.5. Lysophospholipid Acyltransferases in Glioblastoma Multiforme

When discussing the importance of PLA2 in tumorigenesis in GBM, it is also important to mention enzymes that catalyze the opposite reaction to the enzymes in question. An example of this is lysophosphatidylcholine acyltransferases (LPCAT), which catalyze the opposite reaction towards PC [68]. LPCAT causes the formation of PC from LPC and fatty acyl-CoA. For this reason, LPCAT decreases the level of LPA, a lipid mediator important in cancer processes in GBM. According to the GEPIA portal, GBM tumors have higher expressions of LPCAT1, LPCAT2, and LPCAT3, but lower expression of LPCAT4/LPEAT2 relative to healthy brain tissue [9]. In addition, according to Seifert et al., the expression of LPCAT1 and LPCAT3 is higher in GBM tumors than in healthy brain tissue [8]. In contrast, LPCAT4 expression is lower in GBM tumors. This confirms the results obtained from the GEPIA database. An increase in the expression of the aforementioned enzymes may contribute to a decrease in LPA level but also contribute to the intense remodeling of phospholipids in the cell membranes of GBM cells. At the same time, according to the GEPIA database, the expression of the mentioned enzymes does not affect the prognosis severity of GBM patients [9].

4.6. Acyl-CoA Thioesterases and Arachidonic Acid C20:4n-6 in Glioblastoma Multiforme

The most important pathway for the formation of free ARA C20:4n-6 is through PLA2 activity. However, free ARA C20:4n-6 can be formed from hydrolysis of arachidonyl-CoA by acyl-CoA thioesterases (ACOT) [69], a group of nine enzymes that cause hydrolysis of fatty acyl-CoA to free fatty acid and CoA [69,70]. An example of an enzyme from this group is ACOT7, which shows activity towards arachidonyl-CoA and saturated fatty acyl-CoA [69,70,71]. According to GEPIA and Seifert et al., there is a reduction in ACOT7 expression in GBM tumors relative to healthy brain tissue [8,9], where higher expression of this enzyme is associated with a worse prognosis for a GBM patient [9], suggesting the involvement of ACOT7 in tumorigenesis in GBM.
According to GEPIA and Seifert et al., there is also elevated expression of ACOT9 in GBM tumors [8,9], an enzyme showing the highest activity to myristoyl-CoA [69,70,72] and low activity to longer acyl-CoA. Importantly, the expression level of ACOT9 is not associated with the prognosis for a patient with GBM [9]. According to GEPIA, the expression of other ACOT does not differ between GBM tumors and healthy brain tissue [9]. In addition, Seifert et al. indicate that the expression of ACOT4 and ACOT8 in GBM tumors is lower than in healthy brain tissue [8].

5. Cyclooxygenase Pathway and Prostanoids in Glioblastoma Multiforme

5.1. Cyclooxygenase Pathway

Free PUFA, including ARA C20:4n-6, can be converted into prostanoids. This synthesis proceeds in two steps: the first reaction is catalyzed by COX: cyclooxygenase-1 (COX-1) and COX-2, whereas the second reaction is catalyzed by a prostanoid-specific synthase. The substrates for the production of prostanoids are dihomo-γ-linolenic acid C20:3n-6, ARA C20:4n-6, and EPA C20:5n-3, which are converted into 1-series [73], 2-series [73,74], and 3-series [75] prostaglandins or thromboxanes, respectively.
The most important prostanoids for tumorigenic processes in GBM are the 2-series prostanoids produced from ARA C20:4n-6. ARA C20:4n-6 is converted to prostaglandin G2 (PGG2) and then to prostaglandin H2 (PGH2) by COX [76,77,78], although during this reaction, the peroxygenated ARA C20:4n-6 can decompose with the generation of free radicals [79]. Cyclooxygenases also produce 9-hydroxyoctadecadienoic acid (9-HODE) from linoleic acid 18:2n-6 [80]. This compound is a ligand for PPARγ [81], transient receptor potential vanilloid 1 (TRPV1) [82], and G2A/GPR132 [83]; the latter is also a receptor for many lipid mediators produced in the LOX pathway.
COX-1 (another name is prostaglandin-endoperoxide synthase 1 (PTGS1)) is a constitutive enzyme with a constant level of expression [84]. A second enzyme with the same activity is COX-2 (another name is prostaglandin-endoperoxide synthase 2 (PTGS2)) [85], an inducible enzyme that is regulated at the transcriptional level and is characterized by rapid degradation of the COX-2 protein [86]. The half-life of the COX-2 protein is only 5 h.
Sometimes, cyclooxygenase-3 (COX-3), a variant of COX-1 that retains intron 1 in its mRNA, is also mentioned in the context of conversion to prostanoids [87]. Although there is expression of the COX-3 protein, which is longer than COX-1, this enzyme has the same activity as the other cyclooxygenases. In mice and dogs, COX-3 is more sensitive to the inhibitors acetaminophen and phenacetin. Humans also have a variant of COX-1, but it is as sensitive to these inhibitors as standard COX-1 [88].
PGH2 is unstable and undergoes spontaneous nonenzymatic conversion, mainly with PGE2 and, in smaller amounts, with prostaglandin D2 (PGD2) [78]. In the synthesis of PGE2, we can distinguish three synthases: membrane-bound prostaglandin E synthase-1 (mPGES-1)/PTGES [89,90,91], membrane-bound prostaglandin E synthase-2 (mPGES-2)/PTGES2 [92], and cytosolic prostaglandin E synthase (cPGES)/PTGES3 [93]. These synthases are dependent on glutathione, which serves to reduce the endoperoxide bridge in PGH2 with the formation of a single hydroxyl group. In addition, cPGES forms a complex with heat shock protein 90 (Hsp90), which is important in the activity of this PGE2 synthase [94]. mPGES-1 and mPGES-2 bind with either COX-1 or COX-2 [92,95,96], whereas cPGES binds only with COX-1 [93,97]. mPGES-1 is an inducible enzyme whose expression under the influence of inflammatory reactions increases following the expression of COX-2 [96]. mPGES-2 [96] and cPGES [93] are constitutive enzymes, meaning that their expression is not altered by inflammatory reactions.
In plasma, PGE2 undergoes enzymatic dehydration to PGA2 [98], which can isomerize to PGC2 via enzymes with PGA isomerase activity, and can then be isomerized to PGB2 via enzymes with PGC isomerase activity [98,99]. Importantly, detailed studies of the enzymes involved in these reactions are lacking.
PGH2 can also be enzymatically converted to other prostanoids by the appropriate synthase [97]. PGD2 is formed from this prostaglandin with the participation of hematopoietic-type prostaglandin D2 synthase (H-PGDS)/HPGDS and lipocalin-type prostaglandin D2 synthase (L-PGDS)/PTGDS [78]. It is also possible that pro-inflammatory prostaglandins are spontaneously converted into other prostaglandins with anti-inflammatory properties as a mechanism for regulating inflammatory responses [100].
PGD2 undergoes transformations to form the following prostaglandins: 15-deoxy-Δ12,14-PGD2 (15d-PGD2), PGJ2, Δ12-PGJ2, and 15d-PGJ2 [101,102]. PGD2 undergoes spontaneous non-enzymatic conversion to PGJ2 via dehydration or with Δ15-PGD2 [101,102]. PGJ2 can be spontaneously transformed directly into 15d-PGJ2 [102]. PGJ2 can be transformed with the participation of albumin into Δ12-PGJ2 [101,102,103,104].
As PGA2, PGJ2, 15d-PGJ2, and Δ12-PGJ2 have the same ring structure as cyclopentenone, they are classified as cyclopentenone prostaglandins [105]. Cyclopentenone prostaglandins have reactive electrophilic carbon atoms, which are responsible for the properties of this group of prostaglandins. These prostaglandins are inhibitors of nuclear factor κB (NF-κB) [6] and activators of PPARα and PPARγ [7,43]; thus, they have anti-inflammatory and anti-tumor properties.
It is possible that PGH2 can be converted to other prostanoids, such as TxA2 produced by thromboxane A synthase 1 (TBXAS1) [106,107]. TxA2 is unstable, as it undergoes non-enzymatic conversion to TxB2 with a TxA2 half-life of less than 40 s [108]; for this reason, TxA2 acts only locally at the site of synthesis.
TBXAS1 is responsible for the production of TxA2 and can also catalyze the conversion reaction of PGH2 into 12S-hydroxyheptadeca-5Z,8E,10E-trienoic acid (12-HHT) and malondialdehyde [106,109]. 12-HHT, produced by TBXAS1 in similar amounts to TxA2, is a ligand for leukotriene B4 receptor 2 (LTB4R2) [110,111,112].
PGH2 can be converted into PGI2 with PGIS (Figure 3) [113] or into PGF with aldoketoreductase (AKR)1B1 and AKR1C3 [114,115]. PGF can also be synthesized from PGE2 by AKR1C1 and AKR1C2 [114]. After synthesis, prostanoids are secreted outside the cell by multidrug resistance-associated protein 4 (MRP4)/ATP binding cassette subfamily C member 4 (ABCC4) [116].
Prostaglandins are first taken into the cell via prostaglandin transporter (PGT)/solute carrier organic anion transporter family member 2A1 (SLCO2A1), and they are inactivated and degraded [117,118]. Organic anions transporting polypeptide 3 (OATP3) and OATP4 are also involved in PGE2 uptake [119]. Then, prostaglandins are reduced by 15-hydroxyprostaglandin dehydrogenase (15-PGDH)/HPGD [118]. This reaction produces 15-keto-PGE2 from PGE2, a PPARγ ligand [120]. In a subsequent catabolic reaction, 15-keto-PGE2 is reduced by 12-hydroxyeicosanoid dehydrogenase (12-HEDH)/prostaglandin reductase 1 (PTGR1) [121] and prostaglandin reductase 2 (PTGR2) [120] through 15-oxoprostaglandin-Δ13-reductase (13-PGR) activity. This produces 13,14-dihydro-15-keto-PGE2 from 15-keto-PGE2.
Importantly, 13,14-dihydro-15-keto-PGE2 is unstable. It converts to 13,14-dihydro-15-keto-PGA2, and in this form, it combines with proteins, such as with albumin in plasma [122]. 13,14-dihydro-15-keto-PGA2 can also be converted to 11-deoxy-13,14-dihydro-15-keto-11,16-cyclo-PGE2 and occur in the blood in this form [122,123].
PGE2 can also be inactivated and degraded by β-oxidation [124,125]. It is first converted to PGE2-CoA [125], and then it is oxidized in peroxisomes and mitochondria, accompanied by the production of either dinor-PGE2 or tetranor-PGE1 [124].
PGE2 also undergoes ω-oxidation [126]. As a consequence of β-oxidation and ω-oxidation and also the action of 15-PGDH and PTGR1/2, 7α-hydroxy-5,11-diketotetranor-prosta-1,16-dioic acid is formed from PGE2, and then is excreted in the urine [127,128].
Acetylated COX-2 exhibits different catalytic properties than native COX-2. Although non-steroidal anti-inflammatory drugs (NSAID) prevent COX-2 catalytic activity, some NSAIDs cause acetylation of the COX-2 catalytic center. An example of such an NSAID is aspirin (acetylsalicylic acid), which causes changes in the catalytic properties of the enzyme. Acetylated COX-2 converts ARA C20:4n-6 into 15R-hydroxyeicosatetraenoic acid (15R-HETE) [129,130,131,132], whereas acetylated COX-1 has no catalytic activity [133].
Acetylated COX-2 also converts 5S-hydroxyeicosatetraenoic acid (5S-HETE) (the product of 5-lipoxygenase (5-LOX) activity) into 5S,15R-dihydroxyeicosatetraenoic acid (5S,15R-diHETE) [130,131]. Native COX-2 converts 5S-HETE into 5S,11R-diHETE, 5S,15R-diHETE, and 5S,15S-diHETE [130,131]. Then, 5-LOX converts 15R-HETE into 15-epi- lipoxin A4 (15-epi-LXA4) which has anti-inflammatory properties [134]. Another name for 15-epi-LXA4 is aspirin-triggered lipoxin (ATL). Acetylated COX-2 can also convert DHA C22:6n-3 and EPA C20:5n-3 into anti-inflammatory lipid mediators [5]. This means that aspirin has anti-inflammatory effects not only by inhibiting COX activity but also by causing the synthesis of lipid mediators with anti-inflammatory properties.
In addition to ARA C20:4n-6, dihomo-γ-linolenic acid C20:3n-6 and EPA C20:5n-3 are also converted with cyclooxygenases into 1-series prostaglandins [73] and 3-series prostaglandins [75], respectively. EPA C20:5n-3 reduces PGE2 production by COX-1 and, to a lesser extent, by COX-2 [135]. PGE3 binds to the same PGE2 receptors with less intracellular signal transduction efficiency [75]. PGE3 displaces PGE2 from the shared receptor, resulting in a decrease in the receptor’s activity. This means that PGE3 has anti-cancer properties.
PGE1 can also inhibit the proliferation of various cancer cells [136,137], although peroxidation of dihomo-γ-linolenic acid C20:3n-6 with COX-2 can result in the formation of PGH1 and the breakdown of the processed intermediate into free radicals [79]. COX-2 causes C-15 oxygenation of ARA C20:4n-6 and dihomo-γ-linolenic acid C20:3n-6. COX-2 can also catalyze C-8 oxygenation of dihomo-γ-linolenic acid C20:3n-6 [79,138], which often leads to the breakdown of the intermediate product and the formation of 8-hydroxyoctanoic acid (8-OH); this compound inhibits proliferation and is responsible for the antiproliferative properties of dihomo-γ-linolenic acid C20:3n-6 in cells with COX-2 expression [79,138], which is important for the inhibition of FADS1/D5D activity [139]. In the PUFA synthesis pathway, γ-linolenic acid C18:3n-6 in the acyl-CoA form is first elongated with Elovl5 to dihomo-γ-linolenic acid C20:3n-6 [14] and is then desaturated to ARA C20:4n-6 with FADS1/D5D. The reduction of FADS1/D5D activity results in the accumulation of dihomo-γ-linolenic acid C20:3n-6 in the cell. If such a cell has a high COX-2 expression, this fatty acid will either be converted into PGE1, or it will be broken during the reaction catalyzed by COX-2. This results in the formation of 8-OH-octanoic acid which inhibits tumor cell proliferation with a developed drug targeting FADS1/D5D activity [139].

5.2. Cyclooxygenase Pathway and Glioblastoma Multiforme

After ARA C20:4n-6 is released from cell membrane phospholipids, it is processed with COX and LOX. In the healthy brain, ARA C20:4n-6 is processed mainly with LOX, whereas in GBM tumors, it is processed mainly with COX, as shown by experiments on C6 cells [140].
COX-1 expression [141] and COX-2 expression [141,142] are elevated in GBM tumors compared to healthy brain tissue, whereas according to GEPIA and Seifert et al., just COX-1 expression is elevated [8,9]. The expression of all three PGE2 synthases, i.e., mPGES-1, mPGES-2, and cPGES, is also elevated in GBM [143], although according to GEPIA, only cPGES expression is higher compared to its expression in the healthy brain [9]. In contrast, Seifert et al. showed no change in PGE2 synthase expression in GBM tumors [8]. cPGES is enzymatically bound with just COX-1 [93,97]. Therefore, it is possible that COX-1-cPGES may play an important role in the production of PGE2 in GBM tumors. According to the GEPIA portal, there are also changes in the expressions of other prostaglandin synthases. In a GBM tumor, there is increased expression of H-PGDS but decreased expression of L-PGDS [9], both synthases involved in PGD2 synthesis. In contrast, Seifert et al. showed that the expression of H-PGDS and L-PGDS in a GBM tumor is lower than their expressions in a healthy brain [8]. According to GEPIA in a GBM tumor, there is also increased expression of AKR1B1, decreased expression of AKR1C1 and AKR1C2, and no change in AKR1C3 expression [9]. Similarly, Seifert et al. showed that in a GBM tumor, there is higher expression of AKR1B1 and decreased expression of AKR1C1, but there is no difference in the expressions of AKR1C2 or AKR1C3 between the GBM tumor and healthy brain tissue [8]. AKR1B1 is involved in the synthesis of PGF [115], whereas AKR1C1 and AKR1C2 are involved in the conversion of PGE2 into PGF [114]. Expression of the TxA2 synthesizing synthase TBXAS1 [8,9,144] is also elevated in GBM tumors, which may explain the increased expression and production of TxA2 and the higher TxA2/PGI2 ratio in GBM tumors than in healthy brain tissue [145,146].
As for receptors for prostaglandins, according to the GEPIA portal, there is an elevated expression of PTGER4/EP4 and TBXA2R/TP in the tumor relative to healthy brain tissue [9], these two being receptors for PGE2 and TxA2, respectively. In contrast, Seifert et al. showed that the expression of prostanoid receptors in GBM tumors did not differ relative to the healthy brain [8].
According to the GEPIA portal, the expression of MRP4/ABCC4 [9], a transporter responsible for the secretion of prostaglandins from the cell, is also increased in GBM tumors. The transcriptomics analysis by Seifert et al. did not confirm this [8]. GEPIA and Seifert et al. show no change in the expressions of PGT/SLCO2A1, 15-PGDH, 12-HEDH/PTGR1, and PTGR2 [8,9]—the first is a transporter that takes prostaglandins into the cell, and the second, third, and fourth are prostaglandin-degrading enzymes.
COX-2 is important in GBM tumor function. Its expression in GBM tumors is upregulated by hypoxia [23] and EGFR activation [147,148] as well as the action of epidermal growth factor receptor variant III (EGFRvIII) [147] and hepatocyte growth factor (HGF) [149]. COX-2 expression and biosynthesis of the most important product of this enzyme, PGE2, is present in GBM cancer cells. However, PGE2 in GBM tumors may not come mainly from GBM cancer cells but rather from tumor-associated macrophages (TAM) [150].
Under the influence of increased COX expression, there is increased production of PGE2, which is involved in tumorigenesis. PGE2 increases the expression of many factors relevant to tumorigenesis in GBM tumors—in particular, S100 calcium-binding protein A9 (S100A9) [151], interleukin 6 (IL-6) [152], and CXC motif chemokine ligand 8 (CXCL8)/interleukin 8 (IL-8) [153]. PGE2 also elevates proliferation [154,155,156] and causes migration of GBM cancer cells [156]. The effects on proliferation and migration are dependent on the receptors EP2 and EP4 [155,156], and perhaps also EP3. Activation of EP3 results in the activation of transient receptor potential melastatin 7 (TRPM7), which increases the proliferation and migration of GBM cells [157].
COX-2 is also important for GBM cancer stem cells. COX-2 expression, and with it, the production of PGE2, is higher in GBM cancer stem cells than in differentiated GBM cells [158,159]. This lipid mediator activates the Wnt pathway in GBM cancer stem cells, leading to the self-renewal and proliferation of these cells.
PGE2 induces angiogenesis in GBM tumors. Therefore, COX-2 expression is positively correlated with microvessel density in GBM tumors [160]. Notably, PGE2 causes vasculogenic mimicry of GBM cells, which promotes angiogenesis [161]. In GBM cells, PGE2 also increases the expression of CXCL8/IL-8 [153], which has pro-angiogenic properties [162].
PGE2 causes cancer immune evasion. Through EP4, PGE2 increases the expression of tryptophan-2,3-dioxygenase (TDO) [163], an enzyme that converts tryptophan into a signaling molecule that reduces immune cell activity.
PGE2 also affects tumor-associated cells which are important in cancer immune evasion. PGE2 increases the recruitment of myeloid-derived suppressor cells (MDSC) to the tumor niche in GBM [164] and interferes with the cytotoxic function of various immune cells, as shown by experiments in other cancer models. When acting chronically, PGE2 impairs the cytotoxic function of natural killer (NK) cells [165,166], dendritic cells [167], and T cells [168]. PGE2 also causes M2 polarization of macrophages [169], immunosuppressive cells that promote tumor growth.
PGE2 also causes radiation resistance [170,171] and TMZ resistance in GBM [172]. COX-2 expression and PGE2 production in GBM cancer cells are upregulated by TMZ [173] and ionizing radiation [170], which is related to caspase 3 activation in damaged cells and subsequent NF-κB activation [174]. Then, NF-κB increases COX-2 expression and, thus, the production of PGE2 that trans-activates EGFR and activates the β-catenin pathway, which has a pro-survival effect and leads to resistance to further therapy [170]. Through EP1 and EP3, PGE2 increases the intensity of β-oxidation and tricarboxylic acid cycle activity in mitochondria [172], leading to TMZ resistance. In response to ionizing radiation, healthy brain tissue also induces increased production of PGE2 and pro-inflammatory cytokines [175], which increases GBM cell migration as well as causes tumor recurrence [176].
PGD2 is also produced in GBM tumors [177]. At physiological concentrations, this prostaglandin increases the proliferation and migration of GBM cells, but, at concentrations of several micromoles, it decreases the viability and inhibits the proliferation of the GBM cells studied [177,178,179]. This effect may be due to 15d-PGJ2, which has anti-cancer properties [180]. PGD2 is non-enzymatically converted to 15d-PGJ2 [100]. High concentrations of PGD2 result in an accumulation of 15d-PGJ2 to a level that causes a measurable reduction in the viability of GBM cancer cells. Cyclopentenone prostaglandins, particularly PGJ2, Δ12-PGJ2, and 15d-PGJ2, have anti-tumor properties, as demonstrated in in vitro studies on GBM cells. These prostaglandins inhibit tumor cell proliferation through PPARγ activation [181,182].
TxA2 may also play an important role in tumorigenic processes in GBM. In GBM cells, TxA2 increases the expression of IL-6, which participates in tumorigenesis [183]. TBXAS1 inhibitors induce apoptosis and inhibit the migration and proliferation of GBM cancer cells [144,184,185], indicating an autocrine effect of TxA2. In addition, in an in vivo model, TBXAS1 inhibitors inhibited angiogenesis and GBM tumor growth [185]. The described inhibitors increased the sensitivity of GBM cells to alkylation chemotherapy [185] and radiotherapy [186].
Given the role of COX-2 in tumorigenesis in GBM, high COX-2 expression in GBM tumors is associated with poorer patient prognoses [160,187,188], although the GEPIA data showed no correlation between COX-1 and COX-2 expression and patient prognosis severity [9]. In addition, the expression of other prostanoid metabolism enzymes worsens the prognosis for GBM patients, in particular, high expression of mPGES-1, the synthase responsible for the production of PGE2 [121]. This is confirmed with the GEPIA data [9], although the expression levels of other PGE2 synthases are not associated with prognosis severity [9,121]. Of the other prostaglandin synthases, high expression of AKR1B1, a PGF-producing synthase [115], in GBM tumors is associated with poorer patient prognoses [9].
According to the GEPIA portal, expression of MRP4/ABCC4, a transporter that secretes prostaglandins from the cell, does not affect the prognosis for GBM patients [9]. Higher expression of certain prostaglandin receptors worsens the prognosis for patients with GBM. In particular, a worse prognosis is associated with higher expression of PTGER1/EP1 and PTGIR/IP [9], which are receptors for PGE2 and PGI2, respectively.
Higher expression of G2A/GPR132 is also associated with a worse prognosis (p = 0.052) in GBM patients [9]. G2A/GPR132 is a receptor for 9-HODE [83], a product of the activity of COX that process linoleic acid 18:2n-6 [80]. The role of this receptor in GBM has not been thoroughly investigated, although studies on fibroblasts have shown that G2A/GPR132 is an oncogene [189].
Prognosis severity is also affected by the expression level of enzymes involved in prostaglandin inactivation. High expression of 15-PGDH in GBM tumors is associated with a better prognosis [121]. The opposite is true for the expression of 12-HEDH/PTGR1, the enzyme that catalyzes the second prostaglandin inactivation reaction [121]. PGT/SLCO2A1 expression levels are not associated with prognosis severity. On the other hand, according to the GEPIA portal, the expressions of PGT/SLCO2A1, 15-PGDH, PTGR1, and PTGR2 do not affect the prognosis for patients with GBM [9].
Prostaglandin levels in GBM tumors may also be associated with a worse prognosis, particularly higher levels of PGE2 and PGF [121]. PGD2 levels in GBM tumors do not affect prognosis severity [121]. At the same time, these lipid mediators are often unstable, transforming into other lipid mediators with lesser or different properties within a short time after synthesis. For this reason, they may act locally in the immediate vicinity of the site of their synthesis.
Relating enzyme expression and levels of the discussed prostaglandins to prognosis makes it possible to estimate the significant impact of a particular pathway on cancer processes. In GBM tumors, higher expressions of production enzymes and levels of PGE2 (COX-2, mPGES-1) and PGF (COX-2, AKR1B1) are responsible for worse prognoses [9,121]. On the other hand, higher expression of the prostaglandin-inactivating enzyme, 15-PGDH, is associated with better prognoses (Table 5) [121]. For this reason, NSAIDs are being investigated as either potential drugs [190,191] or agents with chemopreventive properties against GBM. Various meta-analyses inconclusively discuss the chemopreventive properties of NSAIDs, such as aspirin. Depending on the meta-analyses cited, regular use of NSAIDs, including aspirin, may either reduce the risk [192,193] or have no effect [194] on the risk of developing glioma or GBM. Nevertheless, the COX pathway produces prostaglandins that exhibit pro-cancer and anti-cancer properties. A better option may be to develop drugs that specifically target only particular prostaglandins relevant to tumorigenic processes in GBM, namely PGE2 and PGF [9,121]. It may be possible to develop drugs that are specific inhibitors of mPGES-1.

5.3. Pan-Cancer Analysis of Genes Related to the COX Pathway and GBM

Changes in the expression of various genes in GBM tumors relative to healthy tissue may be the result of tumor-specific neoplastic processes or specific mechanisms found only in GBM. For this reason, we performed a pan-cancer analysis of the expression of the genes involved in the COX pathway based on the data available in the GEPIA web server [9]. It showed that increased or decreased expression of a given gene relative to healthy tissue does not occur in all types of cancer. At the same time, in some cases, a certain trend of changes in the expressions of the genes studied can be observed. An example of this is TBXAS1, whose expression is increased in nine types of cancer but decreased in another four types of cancer. Similarly, the expression of mPGES-1/PTGES is increased in eight types of cancers but decreased in three types of cancers. Some genes tend to undergo decreased expression in tumors. An example of this is 15-PGDH/HPGD, whose expression is reduced in 18 types of cancer but increased in two types of cancer relative to healthy tissue. Another example is the expression of PGIS/PTGIS, decreased in 17 types of cancer but elevated only in pancreatic adenocarcinoma.
According to GEPIA, there is an increase in COX-1/PTGS1 expression in GBM tumors, which is the same as in lower grade glioma. In seven types of tumors, this gene is overexpressed, but in seven more types, its expression is reduced. This indicates that the increased expression of COX-1/PTGS1 in gliomas (GBM and lower grade gliomas) is specific to these diseases. Some studies also show increased expression of PGE2 synthases (mPGES-1/PTGES, mPGES-2/PTGES2 and cPGES/PTGES3) [9,143], although GEPIA confirms it is only for cPGES/PTGES3 [9]. According to GEPIA, in lower grade glioma, there are no changes in the expression of PGE2 synthases relative to healthy brain tissue. According to GEPIA, expression of cPGES/PTGES3 is increased in 11 types of tumors but is decreased in one type. For this reason, the increase in cPGES/PTGES3 expression in GBM can be considered cancer-specific, just like mPGES-1/PTGES, which has increased expression in eight types of cancer and decreased in three. According to GEPIA, only four types of cancers have increased expression of mPGES-2/PTGES2, which shows that this enzyme may not be cancer-specific.
According to GEPIA in GBM, there is also increased expression of H-PGDS/HPGDS but decreased expression of L-PGDS/PTGDS [9]. At the same time, Seifert et al. showed that the expression of both PGD2 synthases is decreased in GBM tumors [8]. H-PGDS/HPGDS expression is also upregulated in lower grade glioma. H-PGDS/HPGDS expression is downregulated in five tumor types and upregulated in an equal number of tumor types. Changes in H-PGDS/HPGDS expression can be specific to gliomas. L-PGDS/PTGDS expression is lower in GBM compared to healthy brain tissue [8,9]. L-PGDS/PTGDS expression is decreased in almost all types of tumors and, thus, can be deemed specific to cancer.
In GBM, as in lower grade glioma, there is increased expression of TBXAS1 [8,9,144]. The expression of this enzyme is elevated in nine types of tumors, which means it may be cancer-specific.
In GBM tumors, there is also upregulation of AKR1B1 expression but downregulation of AKR1C1 and AKR1C2 expressions relative to healthy brain tissue [8,9]. Lower grade gliomas show no changes in the expressions of these enzymes. The expression of AKR1B1 increases in nine types of tumors. AKR1C1 and AKR1C2, on the other hand, have decreased expressions in 14 types of tumors, which indicates that these changes may be cancer-specific.
PGIS/PTGIS expression is downregulated in 17 types of tumors. At the same time, in GBM tumors, PGIS/PTGIS expression does not differ from healthy brain tissue [8,9].
In GBM and lower grade glioma, there is an increase in MRP4/ABCC4 expression [9]. This transporter also has increased expression in another four types of tumors but decreased expression in two types of tumors. Changes in MRP4/ABCC4 expression may be specific to gliomas.
Finally, 15-PGDH/HPGD expression is often downregulated in tumors (Table 6). This was shown by a pan-cancer analysis in which 18 out of 31 cancers had decreased expression of this enzyme. At the same time, in gliomas (GBM and lower grade glioma), there were no changes in 15-PGDH/HPGD expression relative to healthy brain tissue.

6. Lipoxygenases and Arachidonic Acid in Glioblastoma Multiforme

6.1. Lipoxygenases Pathway

In addition to the COX pathway, PUFA can be transformed with LOX. These enzymes exhibit dioxygenase activity, catalyzing the insertion of a hydroperoxyl group into a PUFA, most commonly ARA 20:4n-6. Hydroperoxyeicosatetraenoic acids (HpETE) are then formed from ARA 20:4n-6, which are further processed in the lipoxygenase pathway. The names of LOX enzymes are related to their sites of formation and the configuration of the hydroperoxyl group in ARA 20:4n-6. In humans, there are six LOX:
  • epidermal lipoxygenase 3/arachidonate lipoxygenase 3 (eLOX3/ALOXE3),
  • 5-lipoxygenase/arachidonate 5-lipoxygenase (5-LOX/ALOX5),
  • 12S-lipoxygenase/arachidonate 12-lipoxygenase, 12S type (12S-LOX/ALOX12),
  • 12R-lipoxygenase/arachidonate 12-lipoxygenase, 12R type (12R-LOX/ALOX12B),
  • 15-lipoxygenase-1/arachidonate 15-lipoxygenase (15-LOX-1/ALOX15), also known as 12/15-LOX, and
  • 15-lipoxygenase-2/arachidonate 15-lipoxygenase type B (15-LOX-2/ALOX15B).
The ALOX5 gene is found on chromosome 10. The other LOX form a gene cluster on 17p13.1 [195,196]. There is also a mouse 8-LOX [197], whose sequence is 78% identical to that of human 15-LOX-2/ALOX15B [197,198]. It is likely that mouse 8-LOX and human 15-LOX-2/ALOX15B are derived from a common ancestor, which was indirectly confirmed by mutagenesis experiments on these two enzymes. Changing only two amino acids in either mouse 8-LOX or human 15-LOX-2/ALOX15B alters the catalytic properties of these two enzymes in 15-LOX and 8-LOX, respectively [197].

6.1.1. Epidermal Lipoxygenase 3

The ALOXE3 gene forms a gene cluster on 17p13.1 together with other LOX [196]. The highest expression of the ALOXE3 gene is found in the skin [196,199]; very low expression of this gene is found in the brain, placenta, pancreas, ovary, and testis.
eLOX3/ALOXE3 shows no significant activity against ARA 20:4n-6 or linoleic acid C18:2n-6 [200], which is related to the low availability of molecular oxygen in the active center of this enzyme [201]. For this reason, the processing of ARA 20:4n-6 by eLOX3/ALOXE3 is very inefficient, but eLOX3/ALOXE3 can exhibit dioxygenase activity to ARA 20:4n-6.
eLOX3/ALOXE3 has hydroperoxide isomerase activity [200]. eLOX3/ALOXE3 converts HpETE into hydroxy-epoxyeicosatrienoic acid, which is the main product of eLOX3/ALOXE3 activity. eLOX3/ALOXE3 also converts HpETE into oxo-eicosatetraenoic acid (oxo-ETE)/ketoeicosatetraenoic acid (KETE) [200,202]. 15S-HpETE is converted by eLOX3/ALOXE3 into either 13R-hydroxy-14S,15S-epoxyeicosa-5Z,8Z,11Z-trienoic acid or 15-oxo-ETE [200].
eLOX3/ALOXE3 also converts 12S-HpETE into hepoxilin A3 (HxA3), HxB3 [200,203], or 12-oxo-ETE [204,205]. On the other hand, 12R-HpETE is converted by eLOX3/ALOXE3 into either 11,12-bis-epi-HxA3 or 12-oxo-ETE [200].
In addition, eLOX3/ALOXE3 shows activity to 5-HpETE and other HpETEs [202]. Because HETE and oxo-ETE [206] as well as hepoxilins [207] exhibit biological activity, eLOX3/ALOXE3 affects biological and pathological processes, particularly in the skin, where expression of this enzyme is highest. For this reason, mutations in the ALOXE3 gene lead to ichthyosis [208,209,210].

6.1.2. 5-Lipoxygenase

The best-studied LOX is 5-LOX/ALOX5. The highest expression of 5-LOX/ALOX5 is found in the bone marrow, appendix, lung, urinary bladder, spleen, and lymph node [199]. This enzyme converts ARA 20:4n-6 to 5S-hydroperoxyeicosatetraenoic acid (5-HpETE) and then to leukotriene A4 (LTA4) [211]. Importantly, 5-lipoxygenase-activating protein (FLAP)/ALOX5AP is required for the activity of 5-LOX/ALOX5. FLAP/ALOX5AP is a substrate carrier [212,213]. 5-HpETE is an activator of PPARα [214]; for this reason, if it is not converted to other lipid mediators, then it will activate this nuclear receptor. Subsequently, LTA4 is converted to other lipid mediators, in particular to other leukotrienes. LTA4 can also undergo spontaneous conversion to 5,6-diHETE, 5,12-diHETE, and 5-oxo-ETE [215]. In turn, 5-HpETE is converted to 5-hydroxyeicosatetraenoic acid (5-HETE) with glutathione peroxidase [216]. The identified receptor for 5-HETE is G2A/GPR132 [83]; this receptor is also activated by other lipid mediators, such as various HETE and 9-HODE.
5-oxo-ETE can also be formed from 5-HETE with the participation of an enzyme with 5-hydroxyeicosanoid dehydrogenase (5-HEDH) activity [217,218,219]. 5-oxo-ETE is an important lipid mediator with a receptor oxoeicosanoid receptor 1 (OXER1)/GPR99 [220,221,222].
LTA4 is a precursor for the production of other leukotrienes and lipoxins; it is converted to lipoxins in a reaction catalyzed by 12-LOX or 15-LOX [223]. LTA4 can also be converted to LTB4 by LTA4 hydrolase (LTA4H) [224,225]. LTA4H also has aminopeptidase activity unrelated to the production of leukotrienes [225]; this activity is important in moderating the immune response [226]. LTB4 has its own membrane receptors: LTB4R1/BLT1 [227] and LTB4R2/BLT2 [228]. Inside the cell, LTA4 and LTB4 activate PPARα, by which these leukotrienes can exert anti-inflammatory effects [7,214].
Glutathione can be attached to LTA4 by LTC4 synthase (LTC4S) (Figure 4) [229,230]. LTC4 is then formed. LTC4S combines with 5-LOX and FLAP to increase the efficiency of LTC4 production with ARA 20:4n-6 [231]. Subsequently, amino acids from the conjugated glutathione in LTC4 can be removed. As a consequence of this, LTC4 is converted into other leukotrienes, namely LTD4, LTE4, and LTF4. All of these leukotrienes, together with LTC4, form a group called cysteinyl leukotrienes. LTD4 is then formed from LTC4 with the involvement of γ-glutamyltransferase 1 (GGT1) and γ-glutamyltransferase 5 (GGT5) [232]. Subsequently, LTD4 can be converted to LTE4 with the participation of dipeptidase 1 (DPEP1) and dipeptidase 2 (DPEP2) [233,234]. LTC4 can also be converted to LTF4 with the participation of carboxypeptidase A [235]. Amino acids can be attached back to cysteine in cysteinyl leukotriene, as exemplified by the conversion of LTE4 to LTF4 with the participation of an enzyme with γ-glutamyltranspeptidase activity [236]. LTF4, however, has a much weaker effect than LTE4, and the latter reaction can be considered an inactivation of LTE4.
Once synthesized, leukotrienes are secreted from the cell. LTC4 is secreted from cells by multidrug resistance-associated proteins (MRP) [237]. In particular, MRP1/ABCC1 [238,239], MRP2/ABCC2 [240,241], MRP3/ABCC3 [242], MRP4/ABCC4 [243], MRP6/ABCC6 [244], MRP7/ABCC10 [245], and MRP8/ABCC11 [246] are responsible for this process. In contrast, OATP1/SLCO1C1 and OATP4 are responsible for the uptake of LTC4, particularly into liver cells where leukotrienes are degraded [119,247]. In contrast, LTB4 transport is still poorly studied; it is known that efflux of LTB4 occurs via MRP4/ABCC4 [243].
Once leukotrienes are secreted outside the cells, they can activate their membrane receptors. LTB4 has two receptors: LTB4R1/BLT1 [227] and LTB4R2/BLT2 [228], the former of which has a 20 times better dissociation constant (Kd) than LTB4R2 in binding LTB4 [228]. With that said, LTB4R2 can be activated by other ARA-derived lipid mediators. These include 12S-HETE, 12R-HETE, 15-HETE, 15-HpETE [248], and 12-HHT [110,111,112]. 12-HHT is formed together with malondialdehyde in a reaction catalyzed by TBXAS1, whose substrate is PGH2 [106,109]. In addition, 12-HHT can be formed independently of TBXAS1 but in smaller amounts [109].
The receptors for cysteinyl-leukotrienes are CysLTR1 [249] and CysLTR2 [250,251]. Both receptors show a 38% similarity in amino acid sequence [250]. CysLTR1 shows a high affinity for LTD4 and low affinity for LTC4 and LTE4, and it shows no affinity at all for LTB4 [249]. CysLTR2 has the best affinity for LTC4 and LTD4 and a very low affinity for LTE4, and it shows no affinity at all for LTB4 [250,251]. A receptor specific for LTE4 is 2-oxoglutarate receptor 1 (OXGR1)/GPR99 [252], which is also the receptor for 2-oxoglutarate. This receptor has a lower affinity for LTC4 and LTD4. Another identified receptor for cysteinyl-leukotrienes specifically for LTC4 and LTD4 is G protein-coupled receptor 17 (GPR17) [253], which is also activated by uridine diphosphate (UDP), UDP-glucose, and UDP-galactose [253]. Further studies have not confirmed that GPR17 is a receptor for UDP, LTC4, and LTD4 [254,255]. This receptor can, independently of its ligand, downregulate CysLTR1 [256], which means it can reduce the action of cysteinyl leukotrienes.
Leukotrienes can be inactivated and excreted. LTB4 is oxidized to 12-oxo-LTB4 with 12-hydroxyeicosanoid dehydrogenase (12-HEDH)/PTGR1 [257,258,259]. This enzyme is also involved in prostaglandin degradation [121]. Subsequently, 12-oxo-LTB4 is reduced with the formation of 12-oxo-10,11-dihydro-LTB4 with an enzyme with Δ10-reductase activity [260]. 12-oxo-10,11-dihydro-LTB4 can then be converted to 10,11-dihydro-LTB4 and 10,11-dihydro-12-epi-LTB4, which undergo ω-oxidation, β-oxidation, or elongation [257]; compounds formed after ω-oxidation and β-oxidation are excreted in the feces [261] and urine [262] as ω-carboxymetabolites of LTB4. HETE are similarly degraded, such as 12-HETE with the formation of 10,11-dihydro-12-HETE and 10,11-dihydro-12-oxo-ETE [263]. Cysteinyl-leukotrienes are first converted to LTE4 [264]; this leukotriene then undergoes ω-oxidation with the formation of ω-carboxy-tetranor-dihydro-LTE4, which is eliminated in the feces and urine.

6.1.3. 12S-Lipoxygenase

ALOX12 gene expression is found in the esophagus and skin [199]. 12S-LOX/ALOX12 can participate in the conversion of LTA4 into lipoxins [223], but the best-described activity of 12S-LOX/ALOX12 is to catalyze the insertion of a hydroperoxyl group into ARA 20:4n-6 at position 12—12S-HpETE is then formed [265]—the compound which can also be formed with 15-LOX-1/ALOX15 [266].
12S-LOX can convert dihomo-γ-linolenic acid to 12S-hydroxy-8Z,10E,14Z-eicosatrienoic acid (12S-HETrE) [267,268]. In contrast, linoleic acid C18:2n-6 is not a substrate for 12S-LOX/ALOX12 [267]. 12S-HpETE can be converted to 12S-HETE, whose receptors are G protein-coupled receptor 31 (GPR31) [269] and G2A/GPR132 [83].
12S-HETE also activates PPARγ [270], as 12S-HpETE [200] and 12S-HETE can be converted to 12-oxo-ETE [260], a PPARγ ligand and activator [204]. 12-oxo-ETE can be converted back to 12S-HETE with an enzyme with 12-oxo-ETE reductase activity [271].
12S-HpETE can be converted to HxA3 (8-hydroxy-11,12-epoxyeicosatrienoic acid) or HxB3 (10-hydroxy-11,12-epoxyeicosatrienoic acid) with enzymes with hepoxilin synthase activity, for example, heme, as shown by experiments on hemoglobin and hemin [272,273]. Hepoxilin synthase activity is also demonstrated by eLOX3/ALOXE3, 12S-LOX/ALOX12, and 15-LOX-1/ALOX15, as shown by experiments on human, rat, and mouse models [200,203,274,275].
Then, HxA3 may bind glutathione via glutathione S-transferase at position 11 [276,277]. HxA3 then gives rise to 11-glutathionyl-HxA3, or otherwise HxA3-C. HxB3 is not subject to such modification [278]. HxA3-C can be produced in the brain and may be a neuromodulator [279]. Like cysteinyl-leukotrienes, HxA3-C can be converted to other cysteinyl-hepoxilins [279]. HxA3-C is converted to HxA3-D by γ-glutamyltranspeptidase. HxA3 and HxB3 can also be converted into trioxilin A3 (TrXA3) (8,11,12-trihydroxyepoxyeicosatrienoic acid) and TrXB3 (10,11,12-trihydroxyepoxyeicosatrienoic acid) with soluble epoxide hydrolase (sEH) (current name: epoxide hydrolase 2 (EPHX2)) [276,280]. HxA3 receptors are TRPV1 and transient receptor potential ankyrin 1 (TRPA1) [281,282]. HxA3 and TrXA3 are also antagonists of the TP receptor [283], the receptor for TxA2.

6.1.4. 12R-Lipoxygenase

In addition to 12S-LOX/ALOX12, there is a second enzyme with 12-LOX activity [195], namely 12R-LOX/ALOX12B [284]. This enzyme shows activity towards ARA C20:4n-6 but not linoleic acid C18:2n-6 [284]. 12R-LOX/ALOX12B transforms ARA C20:4n-6 into 12R-HpETE, a stereoisomer of the product of 12S-LOX/ALOX12’s enzyme activity. 12R-HpETE is converted to 11,12-bis-epi-HxA3 with eLOX3/ALOXE3 [200]. 12R-HpETE is a stereoisomer of 12S-HpETE. Similar to this compound, 12R-HpETE can also be converted to 12R-HETE [206], which is then converted to 12-oxo-ETE with an enzyme with 12-hydroxyeicosanoid dehydrogenase activity [206,260], including eLOX3/ALOXE3 [200].
The ALOX12B gene is only 38% similar to the ALOX12 gene. The highest expression of this enzyme is found in the skin, and it is much lower in the prostate and adrenal gland [196,199,284]. 12R-LOX is important in skin function; mutations in the ALOX12B gene lead to ichthyosis [208,210,285], as do mutations in the ALOXE3 gene. 12R-LOX/ALOX12B and eLOX3/ALOXE3 participate in a common pathway in lipid mediator production. 12R-LOX produces 12R-HpETE, which is converted to 11,12-bis-epi-HxA3 with eLOX3 (Figure 5) [200]. Under the influence of eLOX3/ALOXE3, 12-oxo-ETE is also formed from 12R-HpETE in small amounts [200].

6.1.5. 15-Lipoxygenases

Like the previously described LOX, 15-LOX catalyzes the formation of 15S-hydroperoxyeicosatetraenoic acids (15-HpETE) from ARA 20:4n-6 [286]. In humans, two 15-LOX isoforms are distinguished: 15-LOX-1/ALOX15 [287] and 15-LOX-2/ALOX15B [288]. The highest expression of 15-LOX-1/ALOX15 is found in the lung, and the lower expressions are in the skin, intestine, heart, lymph node, and testis [199]. The highest expression of 15-LOX-2/ALOX15B is found in the prostate and skin. Expression of this enzyme is also observed in the lung, esophagus, and cornea [196,199,288].
The enzymatic properties of the two isoforms differ. 15-LOX-1/ALOX15 catalyzes the formation of 15-HpETE, but it also converts part of the substrate, ARA 20:4n-6, into 12-HpETE [266]—for this reason, the enzyme owns its historical name: 12/15-LOX. 15-LOX-2/ALOX15B has no such activity [266,288].
15-LOX-1/ALOX15 shows much higher activity with linoleic acid C18:2n-6 than 15-LOX-2/ALOX15B (Figure 6) [266]. These enzymes convert linoleic acid C18:2n-6 into 13S-hydroperoxyoctadecadienoic acid (13-HpODE), which converts to 13S-hydroxyoctadecadienoic acid (13-HODE). The identified receptor for 13-HpODE is G2A/GPR132 [83]. 13-HODE also activates the TRPV1 receptor [82]. 13-HODE undergoes the same transformations as HETE and can be oxidized to 13-oxo-ODE. 13-oxo-ODE [289] and 13-HODE [290] are PPARγ ligands.
15-HpETE is transformed into many lipid mediators. It can be transformed into 15-HETE, which is an activator of PPARγ [270] and G2A/GPR132 [83]. 15-HpETE can be converted to 13R-hydroxy-14S,15S-epoxyeicosa-5Z,8Z,11Z-trienoic acid (14,15-HxB3 13R), 11S-hydroxy-14S,15S-epoxy-5Z,8Z,12E-eicosatrienoic acid (14,15-HxA3 11S), and 15-oxo-ETE [200,291]. 14,15-HxA3 11S, analogous to HxA3, can be conjugated with glutathione. This produces 14,15-HxA3-C 11S and cysteinyl-14,15-HxA3 11S, having conjugated glutathione without further amino acids, which is analogous to that of cysteinyl-leukotriene [291].
15-HpETE can also be converted to eoxins [292], which are isomers of leukotrienes.
15-HpETE can also be converted to lipoxins with 5-LOX [223], resulting in the formation of 5S,15S-dihydroperoxyeicosatetraenoic acid (5,15-diHpETE), and then converted to LXA4 or LXB4 [293]. 5-HpETE can also be converted with 15-LOX-1/ALOX15 into 5,15-diHpETE and, via the same pathway, be converted into LXA4 or LXB4 [293]. 15-HETE can be converted to LXA4 with 5-LOX/ALOX5 [294]. Lipoxins can also be formed from LTA4, which is processed by 15-LOX-1/ALOX15 or 12-LOX [293,295].
LXA4 is a lipid mediator with biological activity whose receptors are lipoxin A4 receptor (ALX)/formyl peptide receptor type 2 (FPR2) [296,297], aryl hydrocarbon receptor (AHR) [298], and estrogen receptors subtypes alpha (ERα) [299], the former of which is not a receptor for LXB4 [296]. The ALX/FPR2 receptor is responsible for the anti-inflammatory properties of lipoxins.
There are also cysteinyl lipoxins, which, just like cysteinyl leukotrienes, are lipoxins with conjugated glutathione at carbon 6 [294]. They are synthesized from 15-HETE, from which, with the participation of 5-LOX/ALOX5, 15-hydroxy-5,6-epoxy-eicosatetraenoic acid is formed, a compound similar in structure to LTA4. The epoxy group from these two compounds is converted to a hydroxyl group and conjugated glutathione [294]. However, it is not known whether cysteinyl lipoxins are essential lipid mediators or merely arise as a result of the nonspecificity of enzymes conjugating glutathione to various compounds.

6.2. Lipoxygenases in Glioblastoma Multiforme

In GBM tumors, ARA C20:4n-6 is mainly processed by COX, as shown by experiments on the C6 cell line [140]. In contrast, in the healthy brain, this PUFA is mainly processed by the LOX pathway. This shows that in GBM tumors, the LOX pathway may not be as important as the COX pathway, although it is still important in tumor mechanisms in GBM tumors.

6.2.1. 5-Lipoxygenase Pathway in Glioblastoma Multiforme

The expression of 5-LOX/ALOX5 in a GBM tumor is higher than in non-tumor brain tissue [300,301,302]. This is also confirmed by data obtained from the GEPIA portal [9] and from Seifert et al. transcriptomics analysis [8].
Expression of 5-LOX/ALOX5 in the GBM tumor is found in macrophage and microglial cells as well as in other cells, such as cancer cells [301,302]. It is higher in GBM cancer stem cells than in other GBM cancer cells [303]. According to GEPIA, higher expressions of FLAP/ALOX5AP, LTC4S, LTA4H, GGT5, and DPEP1 but not DPEP2 [9], the enzymes that synthesize LTB4 and LTE4 from the product of 5-LOX/ALOX5 activity, were also found in GBM tumors [224,225,229,230,232,234]. Seifert et al. showed that there are higher expressions of FLAP/ALOX5AP, LTA4H, and GGT5 in GBM tumors than in healthy brain tissue [8]. In contrast, LTC4S, DPEP1, and DPEP2 are not affected. The higher expression of enzymes responsible for leukotriene biosynthesis increases the production [304] and levels [305] of these lipid mediators further in GBM tumors than in healthy brain tissue, particularly cysteinyl-leukotrienes.
The expression level of 5-LOX/ALOX5 in GBM tumors does not affect prognosis [9,188], although simultaneous high expression of COX-2 and 5-LOX/ALOX5, two major ARA C20:4n-6 processing enzymes, is associated with a worse prognosis [188]. This shows that the two pathways in cooperation can impinge on prognosis severity.
The expression levels of most enzymes involved in leukotriene production and metabolism do not affect prognosis [9]. Only for GGT1, higher expression in GBM tumors is associated with a worse prognosis [9]. GGT5 expression showed a positive trend (p = 0.055) toward a worse prognosis. GGT1 and GGT5 are enzymes that catalyze the transformation of LTC4 into LTD4 [232], demonstrating that the transformation of cysteinyl leukotrienes may be important in tumorigenesis in GBM.
In addition, higher expression of 12-HEDH/PTGR1, an enzyme that degrades LTB4, as well as prostaglandins, may be associated with worse prognoses for GBM patients [121], although GEPIA did not confirm such a link [9]. In addition, GEPIA and Seifert et al. did not show that 12-HEDH/PTGR1 expression differs between GBM tumors and healthy brain tissue [8,9]. According to GEPIA [9] and Seifert et al. [8], expression levels of receptors for leukotrienes LTB4R1, LTB4R2, CysLTR1, CysLTR2, GPR17, and OXGR1/GPR99 do not differ between GBM tumors and healthy brain tissue. In addition, the expression levels of these receptors in GBM tumors do not affect prognosis [9].
Leukotrienes as well as the entire 5-LOX pathway are important in tumorigenesis in GBM. They may also be important in the onset of GBM and in the first stages of tumorigenesis. The GA genotype of rs2291427 in the ALOX5 gene is associated with a higher risk of GBM in men [306].
Expression of 5-LOX/ALOX5 is higher in GBM cancer stem cells than in other GBM cancer cells [303]. The products of 5-LOX/ALOX5 activity induce proliferation and self-renewal of GBM cancer stem cells. The effects of 5-LOX/ALOX5 on GBM cancer stem cells are autocrine in nature.
LTB4 also increases the proliferation of GBM cells [307]. This is associated with an increase in Ca2+ levels in the cytoplasm of GBM cells [307]. Studies of various cell lines show that 5-LOX/ALOX5 expression is present in only a portion of them [308,309]. Expression of 5-LOX/ALOX5 causes an autocrine increase in the proliferation of such a line and, thus, makes culture growth dependent on 5-LOX/ALOX5 activity. All GBM lines express LTA4H, LTB4R1/BLT1, LTB4R2/BLT2, and CysLTR2, but only some lines express LTC4S [309], indicating heterogeneity in the production of cysteinyl-leukotrienes and 5-HETE by GBM cancer cells.
The dependence of the proliferation of some GBM cancer cell lines on the 5-LOX pathway may be a potential therapeutic target for GBM treatment in personalized therapy. For this reason, the pan-LOX inhibitor Nordy [303,310], 5-LOX inhibitors such as caffeic acid [307], A861 [311], AA-863, and U-60,257 (pyriprost) [312], LTA4H inhibitors such as bestatin [311], and CysLTR1 and CysLTR2 receptor inhibitors such as montelukast and zafirlukast [313] have anti-tumor properties against GBM and inhibit proliferation. This is associated with decreased ERK MAPK activation and induction of apoptosis as a result of decreased expression of anti-apoptotic Bcl-2 and increased expression of pro-apoptotic Bax [308].
Cysteinyl leukotrienes may have anticancer properties by increasing the bioavailability of various chemotherapeutics. In the brain, as well as in GBM tumors, there is a blood-brain barrier (BBB) that is poorly permeable to many substances, including anticancer drugs [314]. However, cysteinyl leukotrienes have BBB permeability, as shown by experiments on rat RG-2 glioma tumors [315]. BBB permeability is highest for LTE4 [315], with cysteinyl leukotrienes not causing BBB permeability in healthy brain tissue [315,316]. For this reason, the administration of LTC4 prior to the administration of chemotherapeutics that pass poorly through the BBB increases the bioavailability of drugs such as cisplatin [317]. However, this method does not increase the bioavailability of all chemotherapeutics, as exemplified by paclitaxel [318].
The receptor for cysteinyl leukotrienes is GPR17 [253]. According to GEPIA [9] and Seifert et al. [8], the expression level of this receptor does not differ between GBM tumors and healthy brain tissue. Higher GPR17 expression is associated with better prognosis in patients with low-grade gliomas, according to the Chinese Glioma Genome Atlas (CGGA) [319] and GEPIA [9], but the expression of this receptor is not associated with prognosis in a GBM patient [9]. GPR17 expression is also higher in low-grade gliomas than in healthy brain tissue [319]. Activation of this receptor by the ligand inhibits proliferation in the G1 phase and induces apoptosis of GBM cell lines LN-229 and SNB-19 [319]. In addition, GPR17 ligands inhibit tumor growth, as shown by experiments using patient-derived xenograft mouse models. The action of GPR17 is associated with a decrease in the levels of cyclic adenosine monophosphate (cAMP) and Ca2+ in the cytoplasm, which reduces the activation of the PI3K → Akt/PKB pathway [319,320]. An increase in GPR17 expression can cause the proliferation and migration of GBM cells [321], particularly with an increase in the expression of this receptor by long non-coding RNA (lncRNA) colorectal neoplasia differentially expressed (CRNDE) in low-grade glioma cells [321].
The receptor for 5-HETE, and also other lipid mediators, is G2A/GPR132 [83]. Higher expression of this receptor, according to GEPIA, is associated with a worse prognosis for a GBM patient (p = 0.052) [9], yet there is no significant upregulation of this receptor expression in GBM tumors [8,9].
5-oxo-ETE may also play an important role in tumorigenic mechanisms in GBM. The receptor for this lipid mediator is OXER1/GPR99 [220,221,222]. The expression of this receptor does not differ between GBM tumor and healthy brain tissue [8,9]. According to GEPIA, higher expression of OXER1/GPR99, the receptor for 5-oxo-ETE, is associated with a worse prognosis for a GBM patient [9]. OXER1/GPR99 is also a receptor for 2-oxoglutarate, LTC4, and LTD4 [252]. There is a lack of thorough research on the importance of 5-oxo-ETE in tumorigenesis in GBM tumors.

6.2.2. 12-Lipoxygenase Pathway in Glioblastoma Multiforme

In GBM tumors, expression of 12S-LOX/ALOX12 and 12R-LOX/ALOX12B is not different from healthy brain tissue [8,9], nor is it associated with prognosis severity [9], nor is the expression of the receptor for 12S-HETE, i.e., GPR31, elevated and affecting prognosis [8,9]. In contrast, the expression of eLOX3/ALOXE3 in GBM tumors is lower than in other brain tissue [9,205]. On the other hand, the transcriptomics analysis by Seifert et al. showed no differences between eLOX3/ALOXE3 expression levels in GBM tumor and healthy brain tissue [8]. Downregulation of eLOX3/ALOXE3 expression in GBM tumor is associated with increased expression of miR-18a, which downregulates eLOX3/ALOXE3 expression [205]. At the same time, eLOX3/ALOXE3 expression is also not related to the prognoses of GBM patients [9].
12-LOX is involved in tumorigenesis in GBM. Studies on various cell lines have shown that 12-LOX expression is common in GBM cancer cells [309]. For this reason, 12-LOX inhibitors inhibit proliferation and reduce the viability of GBM cells [309,322]. 12-LOX inhibitors also inhibit the migration of GBM cells because they reduce the expression of matrix metalloproteinase 2 (MMP2) in these cells [309]. However, the exact mechanism of 12-LOX action on tumorigenic processes in GBM is poorly studied. The fact that eLOX3/ALOXE3 is anticancer in nature [205] suggests that a lipid mediator not formed by eLOX3/ALOXE3 is responsible for the pro-cancer properties of 12-LOX. Perhaps it is 12-HETE, a lipid mediator with proven pro-cancer properties in other cancers [323,324]. In addition, higher expression of G2A/GPR132, a receptor for 5-HETE, 12-HETE, 15-HETE, and 9-HODE, is associated with a worse prognosis for a GBM patient (p = 0.052) [9]. The oncogenic properties of G2A/GPR132 were also demonstrated in a study on fibroblasts [189], although there is no higher expression of G2A/GPR132 in GBM tumors than in healthy brain tissue [8,9].
12-LOX may also have anti-cancer properties. It converts ARA 20:4n-6 into 12-HpETE, a lipid from the hydroperoxyl group, and for this reason, it can cause lipid peroxidation, which, when free ARA 20:4n-6 is in excess and this PUFA is over-processed, has a destructive effect on the cell [325].
eLOX3/ALOXE3 has anti-tumor properties in GBM. eLOX3/ALOXE3 converts 12-HpETE into 12-oxo-ETE. In the absence of eLOX3/ALOXE3, 12-HpETE is converted to 12-HETE [205], meaning that eLOX3/ALOXE3 decreases 12-HETE production. This lipid mediator increases GBM cell migration. When 12-HETE production is decreased, GBM cell migration is reduced.
The lipid mediators produced by eLOX3/ALOXE3, including 12-oxo-ETE, have anti-tumor effects, particularly 12-oxo-ETE, which is a ligand for PPARγ [204,205]. Activation of this nuclear receptor inhibits proliferation and induces apoptosis of GBM cancer cells [326,327,328].
The products of eLOX3/ALOXE3 activity are hepoxilins and trioxilins [200,203], lipid mediators of physiological importance. However, there is a lack of studies on the importance of these lipid mediators in tumorigenesis in GBM.
Analysis on the GEPIA portal [9] and the transcriptomics analysis by Seifert et al. [8] showed no differences in the expression of EPHX2, the enzyme responsible for converting hepoxilins into trioxilins, between GBM tumors and healthy brain tissue [276,280]. At the same time, according to GEPIA, higher EPHX2 expression in GBM tumors is associated with a tendency toward a worse prognosis (p = 0.072), which may indicate that hepoxilins and trioxilins may have some role in neoplastic processes in GBM.

6.2.3. 15-Lipoxygenase Pathway in Glioblastoma Multiforme

GEPIA [9] and Seifert et al. [8] showed no differences in the expression of 15-LOX-1/ALOX15 and 15-LOX-2/ALOX15B between GBM tumors and healthy brain tissue. According to GEPIA, the expression level of these enzymes does not affect the prognosis for patients [9]. Studies on various GBM lines have shown differences in the expression of 15-LOX-1/ALOX15 and 15-LOX-2/ALOX15B in GBM cancer cells [309]. 15-LOX is important in the function of GBM cancer cells, and 15-LOX inhibitors reduce the viability and migration of GBM cancer cells [309]. On the other hand, increasing the expression and activity of 15-LOX-1/ALOX15 throughout the body may have an anti-tumor effect against GBM, as shown by gene therapy using an adenovirus transducing the ALOX15 gene [329]. This effect may depend on 13-HODE and 15-HETE.
All GBM lineages secrete 13-HODE, a product of the linoleic acid C18:2n-6 conversion with 15-LOX-1/ALOX15 and 15-LOX-2/ALOX15B [266]. 13-HODE increases MMP2 expression in GBM cells, which causes migration [309]. At the same time, 13-HODE also decreases the viability of GBM cells [309], which may depend on the activation of PPARγ via this lipid mediator [290]. This mechanism was confirmed in other cancers, including non-small cell lung cancer [330].
15-HETE can activate G2A/GPR132 [83]. Higher expression of this receptor. according to GEPIA. is associated with a worse prognosis for a GBM patient (p = 0.052) [9]. At the same time, the importance of this receptor in GBM has not been thoroughly investigated. Studies in other models have shown that G2A/GPR132 is an oncogene [189]; that is, 15-HETE through activation of G2A/GPR132 has a pro-cancer effect. At the same time, there is no significant upregulation of this receptor expression in GBM tumors [8,9].
The significance of lipoxins in GBM tumors has not been thoroughly investigated. The expression level of the LXA4 receptor ALX/FPR2 does not differ between GBM tumors and healthy brain tissue (Table 7) [8,9]. The expression level of this receptor in GBM tumors does not affect prognosis. However, it may be important in tumorigenesis in GBM tumors. Studies on U-87 MG cells have shown that silencing ALX/FPR2 reduces the proliferation and migration of the cells tested [331]. In addition, cells with silenced ALX/FPR2 showed lower expressions of VEGF, a major pro-angiogenic factor. However, this receptor is activated not only by LXA4 but also by other factors [332]—for this reason, the importance of LXA4 in tumorigenic processes in GBM cannot be determined.
The expression levels of various LOX are not associated with prognoses for GBM patients [9]. This indicates that the LOX pathway is not as relevant to cancer processes as other pathways. For this reason, drugs targeting LOX may show poor efficacy in GBM therapy. At the same time, the analyses performed in this study show that higher expression of OXER1 (the receptor for 5-oxo-ETE) and higher expression of G2A/GPR132 (the receptor for various HETE) are associated with poor prognosis [9]. This indicates a therapeutic target for future drugs developed for the treatment of GBM. In addition, higher expression of GGT1 in GBM tumors is associated with worse prognosis, and higher expression of GGT5 and EPHX2 is associated with a trend of worse prognosis for GBM patients. This indicates a future direction for research into tumor mechanisms in GBM.

6.3. Pan-Cancer Analysis of Genes Related to LOX Pathway and GBM

Similar to the COX pathway, we performed a pan-cancer analysis of the expression of the genes involved in the LOX pathway using the data from the GEPIA web server [9].
The expression of eLOX3/LOXE3 is reduced in GBM tumors. At the same time, there is no change in the expression of this enzyme relative to healthy brain tissue in lower grade gliomas. It is also reduced in two more types of tumors. For this reason, a decrease in eLOX3/LOXE3 expression may be considered specific to GBM.
In GBM tumors, there is elevated expression of 5-LOX/ALOX5 and FLAP/ALOX5AP relative to healthy brain tissue, which is similar to lower grade gliomas [9]. Expression of these proteins is elevated in 9 and 11 tumor types, respectively. In a similar number of tumor types, there is a reduction in the expressions of 5-LOX/ALOX5 and FLAP/ALOX5AP. This indicates that the elevated expressions of 5-LOX/ALOX5 and FLAP/ALOX5AP may be glioma-specific.
The expression of other LOX is not altered in GBM and lower grade gliomas, which is similar to most other types of cancer. In GBM tumors, there are elevated expressions of LTA4H/LTA4H and LTC4S/LTC4S relative to healthy tissue [9]. In lower grade gliomas, there is higher expression of only LTC4S/LTC4S [9]. According to Seifert et al., in II and III grade gliomas, there are higher expressions of LTA4H/LTA4H but not LTC4S/LTC4S relative to healthy brain tissue [8]. LTA4H/LTA4H expression is elevated in 4 out of 31 analyzed tumor types. LTC4S/LTC4S is upregulated in six tumor types but downregulated in eleven types [9]. Therefore, the elevated expression of LTA4H/LTA4H and LTC4S/LTC4S can be considered as specific to GBM and glioma, respectively.
GGT5 expression is upregulated in GBM and lower grade gliomas [8,9]. It is downregulated in eleven tumor types and upregulated in seven. Therefore, the elevation of GGT5 expression can be considered characteristic for gliomas.
DPEP1 expression is elevated in GBM tumors but not in lower grade gliomas (Table 8) [9]. It is decreased in six types of tumors but increased in four types, including GBM. For this reason, it can be thought that changes in DPEP1 expression are characteristic of GBM. EPHX2 expression is often decreased in tumors. In a pan-cancer analysis, 17 types of tumors had a reduced expression of this enzyme relative to healthy tissue. At the same time, in GBM tumors, EPHX2 expression does not differ relative to healthy brain tissue [8,9].

7. Cytochrome P450 Pathway in Glioblastoma Multiforme Tumors

7.1. Cytochrome P450 Pathway

In addition to the processing of ARA C20:4n-6 by COX and LOX, this fatty acid can also be converted into lipid mediators with cytochrome P450. It results in the formation of epoxyeicosatrienoic acids (EET) and HETE [333].
ARA C20:4n-6 can undergo either hydroxylation or epoxidation. The ω-hydroxylation reaction converts ARA C20:4n-6 into 20-hydroxyeicosatetraenoic acid (20-HETE). The enzymes responsible for this reaction are CYP1A2 [334], CYP1B1 [335], CYP2U1 [336], CYP4A11 [337,338], CYP4F2 [337,339], CYP4F3A, and CYP4F3B [339].
ARA C20:4n-6 can also be converted to 19-hydroxyeicosatetraenoic acid (19-HETE) in the (ω-1)-hydroxylation reaction. The cytochromes P450 responsible for this are CYP1B1 [335], CYP2C19 [340], CYP2E1 [334], and CYP2U1 [336].
ARA C20:4n-6 can also undergo hydroxylation at other positions with the formation of various HETE [335,340,341,342,343,344]. The cytochromes P450 carrying out this reaction include CYP1A2 [343], CYP1B1 [335], CYP2C9 [334,340], and CYP3A4 [343]. The HETE receptor, with an OH residue at positions 5 to 15, is G2A/GPR132 [83]. In contrast, receptors for 20-HETE include G-protein receptor 75 (GPR75) [345], transient receptor potential vanilloid 1 (TRPV1) channel [346], free fatty acid receptor 1 (FFAR1)/GPR40 [347], and PPARα [348]. HETE can then undergo ω-hydroxylation with CYP4F [333], resulting, for example, in the formation of 10,20-dihydroxyeicosatrienoic acid (10,20-DHET) from 10-HETE, which may be a mechanism for regulating the activity of these lipid mediators.
In the cytochrome P450 pathway, ARA C20:4n-6 can also undergo epoxidation with the formation of epoxyeicosatrienoic acids (EET). Because ARA C20:4n-6 has four double bonds, this reaction produces 5,6-EET, 8,9-EET, 11,12-EET, or 14,15-EET, albeit a given cytochrome P450 can produce mainly only some EET [340]. The enzymes responsible for this reaction are CYP1A2 [334], CYP1B1 [335], CYP2C8 [349,350], CYP2C9 [350], CYP2C19 [340], CYP2J2 [351], and CYP4X1 [352].
The receptor for EET is GPR40 [353]. 14,15-EET can activate receptors for prostaglandins, including PGE2 (PTGER2, PTGER3, and PTGER4), PGD2 (PTGDR), and PGF2α (PTGFR) [354,355]. EET can also activate PPARα (in particular, 11,12-EET [348] and PPARγ [356,357]).
Another important property of EET is that it enters the cell membrane and intracellular membranes. This is as a result of the incorporation of EET into glycerophospholipids at the sn-2 position [357,358,359]. EET can also be metabolized by EPHX1 and EPHX2 [357,360]. This is the same enzyme that catalyzes the conversion of hepoxilins (a hydroxy-epoxy derivative of ARA) to trioxilins [276,280]. EET are then converted to dihydroxyeicosatrienoic acid (DHET). In this form, particularly 14,15-DHET, they can activate PPARα [348,361].
EET can also undergo ω-hydroxylation with CYP4F [333]. For example, 8,9-EETs give rise to 20-hydroxy-8(9)-epoxyeicosatrienoic acid (20,8(9)-HEET) (Figure 7) [333,362]. EET can also be converted into either shorter or longer lipid mediators via β-oxidation and elongation, respectively [357]. Another possible reaction is the conversion of 5,6-EET, 8,9-EET, and 11,12-EET with COX [357,363,364], resulting in the formation of lipid mediators with proangiogenic properties. 5,6-epoxy-PGH2 is formed from 5,6-EET, [364]. In contrast, 11-hydroxy-8,9-EET (8,9,11-EHET) and 15-hydroxy-8,9-EET (8,9,15-EHET) are formed from 8,9-EET [364,365,366].
HETE and EET are the direct products of cytochrome P450 activity. However, cytochromes p450 are not only involved in the production of these ARA-derived lipid mediators. In addition, CYP4F and CYP4A cause ω-hydroxylation of the already discussed eicosanoids formed in COX and LOX pathways. CYP4A and CYP4F8 are responsible for the ω-hydroxylation and (ω-1)-hydroxylation of prostaglandins, respectively [126,333], and CYP4F is responsible for the transformation of LTB4 and lipoxins [333]. The aforementioned reactions often result in the inactivation of these lipid mediators.
It should be mentioned that the aforementioned cytochromes P450 are not only involved in the metabolism of ARA C20:4n-6. They can also metabolize other fatty acids [336], such as linoleic acid [367], and many drugs, including anticancer drugs [349,368].

7.2. Cytochrome P450 Pathway in Glioblastoma Multiforme Tumors

ARA C20:4n-6 is converted to 20-HETE [369], which increases the proliferation of GBM cells [370]. 20-HETE may also be an important pro-angiogenic factor in GBM tumors by acting on endothelial cells [369] and enhancing vascular mimicry of GBM cells [371]. Importantly, 20-HETE may not be produced by GBM cells [372] but by TAM and endothelial progenitor cells (EPCs) [373]. CYP2U1 [336,374], whose expression in GBM tumors is elevated relative to healthy brain tissue [8,9], may be responsible for 20-HETE production in GBM tumors. Nevertheless, there is very little research focused on 20-HETE production in GBM tumors.
In the rat glioma RG2 cell line, there is production of various lipid mediators, including 15-HETE, 12-HETE, 8-HETE, 5-HETE, 14,15-diHETE, 14,15-EET, 11,12-diHETE, and 11,12-EET [375]. In part, this may be due to the effect of elevated levels of glutamate in the intercellular space, which is characteristic for GBM tumors [376]. This amino acid increases the expression of CYP1B1 and CYP2U1 in GBM cells [374], leading to increased production of lipid mediators with these cytochrome P450 enzymes.
According to the GEPIA [9] and to Seifert et al. [8], the expression of most of the discussed cytochromes P450 do not differ between GBM tumors and healthy brain tissue. Both sources only show higher expression of CYP2U1 and lower expression of CYP4X1 in GBM tumors compared to healthy brain tissue. CYP2U1 is the cytochrome P450 producing 20-HETE and 19-HETE [336], which shows a possible source of these two lipid mediators in GBM tumors.
GEPIA, in contrast to Seifert et al. shows reduced expression of CYP2C8 in GBM tumors (Table 9). According to the GEPIA [9], the expression of this cytochrome P450 was not linked to the prognosis of GBM patients. Expression of the receptor for 20-HETE, i.e., GPR75, does not differ in GBM tumors compared to healthy brain tissue. The expression level of GPR75 is not associated with prognosis.
The expression of EPHX1 and EPHX2, enzymes involved in the conversion of EET to DHET, does not differ between GBM tumor and healthy brain tissue [8,9]. In addition, the expression levels of these enzymes are not associated with the prognosis of a GBM patient.

7.3. Pan-Cancer Analysis of Cytochrome P450 Genes and Comparison of GBM Expression against Other Cancers

Changes in the expression of various genes in GBM tumors relative to healthy tissue could be the result of tumor-specific neoplastic processes or specific mechanisms found only in GBM. For this reason, a pan-cancer analysis of the expression of the cytochromes P450 genes described above was performed using the GEPIA portal [9].
CYP2C8 expression was lower in GBM tumors relative to healthy brain tissue [9], similar to lower grade gliomas (Table 10). Downregulation of CYP2C8 expression occurs in a variety of tumors. Out of 31 analyzed cancers, seven show decreased expression of this enzyme, which shows that reduced expression of CYP2C8 is common in cancers. In 11 types of cancers out of 31, there is an increase in CYP2J2 expression. However, in GBM and lower grade gliomas, there is no change in the expression of this cytochrome P450. GEPIA also shows that in 8 out of 31 cancers, including GBM tumors, there is higher expression of CYP2U1 compared to healthy tissue. This indicates that elevated CYP2U1 expression may be associated with cancerous processes. In GBM and lower grade gliomas, there is lower expression of CYP4X1 compared to healthy brain tissue [8,9]. In the other seven types of tumors, there is also a decrease in the expression of this cytochrome p450, which suggests that decreased CYP4X1 expression in tumor may be a common feature of cancer.

8. Conclusions

The importance of the most important ARA C20:4n-6-derived lipid mediators in cancer mechanisms in GBM is very well understood. These compounds, particularly PGE2 and leukotrienes, cause the proliferation and migration of GBM cancer cells, are important in the function of GBM cancer stem cells, cause angiogenesis, and by acting on cells of the immune system, inhibit the body’s anti-tumor response. However, the importance in GBM cancer processes of lesser-known ARA C20:4n-6-derived lipid mediators has not yet been investigated. We are talking, for example, about EET, lipoxins, hepoxilins, and some prostanoids, including PGF and TxA2. Investigating the function of these compounds will provide a better understanding of GBM tumor function. It may also contribute to the development of new therapeutic approaches.

Author Contributions

J.K. writing—original draft preparation, writing—review and editing; E.R.-M. investigation; P.K. investigation; D.C. funding acquisition, supervision; I.B.-B. original draft preparation, writing—review and editing, supervision. All authors have read and agreed to the published version of the manuscript.

Funding

This study was supported by the statutory budget of the Department of Biochemistry and Medical Chemistry at Pomeranian Medical University in Szczecin, Poland and the Institute of Biology, University of Szczecin, Poland.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Johnson, D.R.; Omuro, A.M.P.; Ravelo, A.; Sommer, N.; Guerin, A.; Ionescu-Ittu, R.; Shi, S.; Macalalad, A.; Uhm, J.H. Overall survival in patients with glioblastoma before and after bevacizumab approval. Curr. Med. Res. Opin. 2018, 34, 813–820. [Google Scholar] [CrossRef] [PubMed]
  2. Grech, N.; Dalli, T.; Mizzi, S.; Meilak, L.; Calleja, N.; Zrinzo, A. Rising Incidence of Glioblastoma Multiforme in a Well-Defined Population. Cureus 2020, 12, e8195. [Google Scholar] [CrossRef] [PubMed]
  3. Bilgin, E.; Duman, B.B.; Altintas, S.; Cil, T.; Gezercan, Y.; Okten, A.I. Predictors of Survival in Turkish Patients with Primary Glioblastoma. Turk. Neurosurg. 2021, 31, 641–653. [Google Scholar] [CrossRef]
  4. Gomes, R.N.; Felipe da Costa, S.; Colquhoun, A. Eicosanoids and cancer. Clinics 2018, 73, e530s. [Google Scholar] [CrossRef]
  5. Serhan, C.N.; Levy, B.D. Resolvins in inflammation: Emergence of the pro-resolving superfamily of mediators. J. Clin. Investig. 2018, 128, 2657–2669. [Google Scholar] [CrossRef]
  6. Straus, D.S.; Pascual, G.; Li, M.; Welch, J.S.; Ricote, M.; Hsiang, C.H.; Sengchanthalangsy, L.L.; Ghosh, G.; Glass, C.K. 15-deoxy-delta 12,14-prostaglandin J2 inhibits multiple steps in the NF-kappa B signaling pathway. Proc. Natl. Acad. Sci. USA 2000, 97, 4844–4849. [Google Scholar] [CrossRef]
  7. Krey, G.; Braissant, O.; L’Horset, F.; Kalkhoven, E.; Perroud, M.; Parker, M.G.; Wahli, W. Fatty acids, eicosanoids, and hypolipidemic agents identified as ligands of peroxisome proliferator-activated receptors by coactivator-dependent receptor ligand assay. Mol. Endocrinol. 1997, 11, 779–791. [Google Scholar] [CrossRef] [PubMed]
  8. Seifert, M.; Garbe, M.; Friedrich, B.; Mittelbronn, M.; Klink, B. Comparative transcriptomics reveals similarities and differences between astrocytoma grades. BMC Cancer 2015, 15, 952. [Google Scholar] [CrossRef] [PubMed]
  9. Tang, Z.; Li, C.; Kang, B.; Gao, G.; Li, C.; Zhang, Z. GEPIA: A web server for cancer and normal gene expression profiling and interactive analyses. Nucleic Acids Res. 2017, 45, W98–W102. [Google Scholar] [CrossRef]
  10. Cancer Genome Atlas Research Network; Weinstein, J.N.; Collisson, E.A.; Mills, G.B.; Shaw, K.R.; Ozenberger, B.A.; Ellrott, K.; Shmulevich, I.; Sander, C.; Stuart, J.M. The Cancer Genome Atlas Pan-Cancer analysis project. Nat. Genet. 2013, 45, 1113–1120. [Google Scholar] [CrossRef]
  11. Lonsdale, J.; Thomas, J.; Salvatore, M.; Phillips, R.; Lo, E.; Shad, S.; Hasz, R.; Walters, G.; Garcia, F.; Young, N.; et al. The Genotype-Tissue Expression (GTEx) project. Nat. Genet. 2013, 45, 580–585. [Google Scholar] [CrossRef] [PubMed]
  12. The GTEx Consortium; Ardlie, K.G.; Deluca, D.S.; Segrè, A.V.; Sullivan, T.J.; Young, T.R.; Gelfand, E.T.; Trowbridge, C.A.; Maller, J.B.; Tukiainen, T.; et al. The Genotype-Tissue Expression (GTEx) pilot analysis: Multitissue gene regulation in humans. Science 2015, 348, 648–660. [Google Scholar]
  13. Madhavan, S.; Zenklusen, J.C.; Kotliarov, Y.; Sahni, H.; Fine, H.A.; Buetow, K. Rembrandt: Helping personalized medicine become a reality through integrative translational research. Mol. Cancer Res. 2009, 7, 157–167. [Google Scholar] [CrossRef] [PubMed]
  14. Guillou, H.; Zadravec, D.; Martin, P.G.; Jacobsson, A. The key roles of elongases and desaturases in mammalian fatty acid metabolism: Insights from transgenic mice. Prog. Lipid Res. 2010, 49, 186–199. [Google Scholar] [CrossRef]
  15. Park, W.J.; Kothapalli, K.S.; Lawrence, P.; Tyburczy, C.; Brenna, J.T. An alternate pathway to long-chain polyunsaturates: The FADS2 gene product Delta8-desaturates 20:2n-6 and 20:3n-3. J. Lipid Res. 2009, 50, 1195–1202. [Google Scholar] [CrossRef] [PubMed]
  16. Dennis, E.A.; Cao, J.; Hsu, Y.H.; Magrioti, V.; Kokotos, G. Phospholipase A2 enzymes: Physical structure, biological function, disease implication, chemical inhibition, and therapeutic intervention. Chem. Rev. 2011, 111, 6130–6185. [Google Scholar] [CrossRef]
  17. Leonard, A.E.; Bobik, E.G.; Dorado, J.; Kroeger, P.E.; Chuang, L.T.; Thurmond, J.M.; Parker-Barnes, J.M.; Das, T.; Huang, Y.S.; Mukerji, P. Cloning of a human cDNA encoding a novel enzyme involved in the elongation of long-chain polyunsaturated fatty acids. Biochem. J. 2000, 350 Pt 3, 765–770. [Google Scholar] [CrossRef]
  18. Leonard, A.E.; Kelder, B.; Bobik, E.G.; Chuang, L.T.; Lewis, C.J.; Kopchick, J.J.; Mukerji, P.; Huang, Y.S. Identification and expression of mammalian long-chain PUFA elongation enzymes. Lipids 2002, 37, 733–740. [Google Scholar] [CrossRef]
  19. Kitazawa, H.; Miyamoto, Y.; Shimamura, K.; Nagumo, A.; Tokita, S. Development of a high-density assay for long-chain fatty acyl-CoA elongases. Lipids 2009, 44, 765–773. [Google Scholar] [CrossRef]
  20. Shakya, S.; Gromovsky, A.D.; Hale, J.S.; Knudsen, A.M.; Prager, B.; Wallace, L.C.; Penalva, L.O.F.; Brown, H.A.; Kristensen, B.W.; Rich, J.N.; et al. Altered lipid metabolism marks glioblastoma stem and non-stem cells in separate tumor niches. Acta Neuropathol. Commun. 2021, 9, 101. [Google Scholar] [CrossRef]
  21. Korbecki, J.; Kojder, K.; Jeżewski, D.; Simińska, D.; Tarnowski, M.; Kopytko, P.; Safranow, K.; Gutowska, I.; Goschorska, M.; Kolasa-Wołosiuk, A.; et al. Expression of SCD and FADS2 Is Lower in the Necrotic Core and Growing Tumor Area than in the Peritumoral Area of Glioblastoma Multiforme. Biomolecules 2020, 10, 727. [Google Scholar] [CrossRef] [PubMed]
  22. Korbecki, J.; Simińska, D.; Jeżewski, D.; Kojder, K.; Tomasiak, P.; Tarnowski, M.; Chlubek, D.; Baranowska-Bosiacka, I. Glioblastoma Multiforme Tumors in Women Have a Lower Expression of Fatty Acid Elongases ELOVL2, ELOVL5, ELOVL6, and ELOVL7 than in Men. Brain Sci. 2022, 12, 1356. [Google Scholar] [CrossRef]
  23. Jalota, A.; Kumar, M.; Das, B.C.; Yadav, A.K.; Chosdol, K.; Sinha, S. A drug combination targeting hypoxia induced chemoresistance and stemness in glioma cells. Oncotarget 2018, 9, 18351–18366. [Google Scholar] [CrossRef] [PubMed]
  24. Pickard, R.T.; Strifler, B.A.; Kramer, R.M.; Sharp, J.D. Molecular cloning of two new human paralogs of 85-kDa cytosolic phospholipase A2. J. Biol. Chem. 1999, 274, 8823–8831. [Google Scholar] [CrossRef]
  25. Sundler, R.; Winstedt, D.; Wijkander, J. Acyl-chain selectivity of the 85 kDa phospholipase A2 and of the release process in intact macrophages. Biochem. J. 1994, 301, 455–458. [Google Scholar] [CrossRef]
  26. Underwood, K.W.; Song, C.; Kriz, R.W.; Chang, X.J.; Knopf, J.L.; Lin, L.L. A novel calcium-independent phospholipase A2, cPLA2-gamma, that is prenylated and contains homology to cPLA2. J. Biol. Chem. 1998, 273, 21926–21932. [Google Scholar] [CrossRef] [PubMed]
  27. Ackermann, E.J.; Kempner, E.S.; Dennis, E.A. Ca(2+)-independent cytosolic phospholipase A2 from macrophage-like P388D1 cells. Isolation and characterization. J. Biol. Chem. 1994, 269, 9227–9233. [Google Scholar] [CrossRef]
  28. Jenkins, C.M.; Wolf, M.J.; Mancuso, D.J.; Gross, R.W. Identification of the calmodulin-binding domain of recombinant calcium-independent phospholipase A2beta. implications for structure and function. J. Biol. Chem. 2001, 276, 7129–7135. [Google Scholar] [CrossRef]
  29. Portilla, D.; Dai, G. Purification of a novel calcium-independent phospholipase A2 from rabbit kidney. J. Biol. Chem. 1996, 271, 15451–15457. [Google Scholar] [CrossRef]
  30. Hariprasad, G.; Kumar, M.; Srinivasan, A.; Kaur, P.; Singh, T.P.; Jithesh, O. Structural analysis of a group III Glu62-phospholipase A2 from the scorpion, Mesobuthus tamulus: Targeting and reversible inhibition by native peptides. Int. J. Biol. Macromol. 2011, 48, 423–431. [Google Scholar] [CrossRef]
  31. Dhananjaya, B.L.; Sudarshan, S. Inhibition of secretary PLA₂—VRV-PL-VIIIa of Russell’s viper venom by standard aqueous stem bark extract of Mangifera indica L. Trop Biomed. 2015, 32, 24–35. [Google Scholar]
  32. Krayem, N.; Gargouri, Y. Scorpion venom phospholipases A2: A minireview. Toxicon 2020, 184, 48–54. [Google Scholar] [CrossRef] [PubMed]
  33. Stadel, J.M.; Jones, C.; Livi, G.P.; Hoyle, K.; Kurdyla, J.; Roshak, A.; McLaughlin, M.M.; Pfarr, D.A.; Comer, S.; Strickler, J.; et al. Recombinant human secretory phospholipase A2: Purification and characterization of the enzyme for active site studies. J. Mol. Recognit. 1992, 5, 145–153. [Google Scholar] [CrossRef]
  34. Sukocheva, O.; Menschikowski, M.; Hagelgans, A.; Yarla, N.S.; Siegert, G.; Reddanna, P.; Bishayee, A. Current insights into functions of phospholipase A2 receptor in normal and cancer cells: More questions than answers. Semin. Cancer Biol. 2019, 56, 116–127. [Google Scholar] [CrossRef] [PubMed]
  35. Tokumura, A.; Majima, E.; Kariya, Y.; Tominaga, K.; Kogure, K.; Yasuda, K.; Fukuzawa, K. Identification of human plasma lysophospholipase D, a lysophosphatidic acid-producing enzyme, as autotaxin, a multifunctional phosphodiesterase. J. Biol. Chem. 2002, 277, 39436–39442. [Google Scholar] [CrossRef] [PubMed]
  36. Umezu-Goto, M.; Kishi, Y.; Taira, A.; Hama, K.; Dohmae, N.; Takio, K.; Yamori, T.; Mills, G.B.; Inoue, K.; Aoki, J.; et al. Autotaxin has lysophospholipase D activity leading to tumor cell growth and motility by lysophosphatidic acid production. J. Cell Biol. 2002, 158, 227–233. [Google Scholar] [CrossRef]
  37. Snider, A.J.; Zhang, Z.; Xie, Y.; Meier, K.E. Epidermal growth factor increases lysophosphatidic acid production in human ovarian cancer cells: Roles for phospholipase D2 and receptor transactivation. Am. J. Physiol. Cell Physiol. 2010, 298, C163–C170. [Google Scholar] [CrossRef]
  38. Fukushima, N.; Ishii, S.; Tsujiuchi, T.; Kagawa, N.; Katoh, K. Comparative analyses of lysophosphatidic acid receptor-mediated signaling. Cell. Mol. Life Sci. 2015, 72, 2377–2394. [Google Scholar] [CrossRef]
  39. Yang, L.; Zhang, H. Expression of Cytosolic Phospholipase A2 Alpha in Glioblastoma Is Associated with Resistance to Chemotherapy. Am. J. Med. Sci. 2018, 356, 391–398. [Google Scholar] [CrossRef]
  40. Hernández, M.; Burillo, S.L.; Crespo, M.S.; Nieto, M.L. Secretory phospholipase A2 activates the cascade of mitogen-activated protein kinases and cytosolic phospholipase A2 in the human astrocytoma cell line 1321N1. J. Biol. Chem. 1998, 273, 606–612. [Google Scholar] [CrossRef]
  41. Hernández, M.; Barrero, M.J.; Alvarez, J.; Montero, M.; Sánchez Crespo, M.; Nieto, M.L. Secretory phospholipase A2 induces phospholipase Cgamma-1 activation and Ca2+ mobilization in the human astrocytoma cell line 1321N1 by a mechanism independent of its catalytic activity. Biochem. Biophys. Res. Commun. 1999, 260, 99–104. [Google Scholar] [CrossRef] [PubMed]
  42. Tsuji, S.; Ohno, Y.; Nakamura, S.; Yamada, T.; Noda, Y.; Saio, M.; Iwama, T.; Shimazawa, M.; Hara, H. Temozolomide has anti-tumor effects through the phosphorylation of cPLA2 on glioblastoma cells. Brain Res. 2019, 1723, 146396. [Google Scholar] [CrossRef] [PubMed]
  43. Forman, B.M.; Chen, J.; Evans, R.M. Hypolipidemic drugs, polyunsaturated fatty acids, and eicosanoids are ligands for peroxisome proliferator-activated receptors alpha and delta. Proc. Natl. Acad. Sci. USA 1997, 94, 4312–4317. [Google Scholar] [CrossRef]
  44. Gao, Y.; Han, D.; Sun, L.; Huang, Q.; Gai, G.; Wu, Z.; Meng, W.; Chen, X. PPARα Regulates the Proliferation of Human Glioma Cells through miR-214 and E2F2. Biomed. Res. Int. 2018, 2018, 3842753. [Google Scholar] [CrossRef]
  45. Leaver, H.A.; Williams, J.R.; Smith, C.; Whittle, I.R. Intracellular oxidation by human glioma cell populations: Effect of arachidonic acid. Prostaglandins Leukot. Essent. Fat. Acids 2004, 70, 449–453. [Google Scholar] [CrossRef] [PubMed]
  46. Giurdanella, G.; Motta, C.; Muriana, S.; Arena, V.; Anfuso, C.D.; Lupo, G.; Alberghina, M. Cytosolic and calcium-independent phospholipase A2 mediate glioma-enhanced proangiogenic activity of brain endothelial cells. Microvasc. Res. 2011, 81, 1–17. [Google Scholar] [CrossRef] [PubMed]
  47. Anfuso, C.D.; Motta, C.; Giurdanella, G.; Arena, V.; Alberghina, M.; Lupo, G. Endothelial PKCα-MAPK/ERK-phospholipase A2 pathway activation as a response of glioma in a triple culture model. A new role for pericytes? Biochimie 2014, 99, 77–87. [Google Scholar] [CrossRef]
  48. Schleicher, S.M.; Thotala, D.K.; Linkous, A.G.; Hu, R.; Leahy, K.M.; Yazlovitskaya, E.M.; Hallahan, D.E. Autotaxin and LPA receptors represent potential molecular targets for the radiosensitization of murine glioma through effects on tumor vasculature. PLoS ONE 2011, 6, e22182. [Google Scholar] [CrossRef]
  49. Linkous, A.G.; Yazlovitskaya, E.M.; Hallahan, D.E. Cytosolic phospholipase A2 and lysophospholipids in tumor angiogenesis. J. Natl. Cancer Inst. 2010, 102, 1398–1412. [Google Scholar] [CrossRef]
  50. Mao, P.; Smith, L.; Xie, W.; Wang, M. Dying endothelial cells stimulate proliferation of malignant glioma cells via a caspase 3-mediated pathway. Oncol. Lett. 2013, 5, 1615–1620. [Google Scholar] [CrossRef]
  51. Lauber, K.; Bohn, E.; Kröber, S.M.; Xiao, Y.J.; Blumenthal, S.G.; Lindemann, R.K.; Marini, P.; Wiedig, C.; Zobywalski, A.; Baksh, S.; et al. Apoptotic cells induce migration of phagocytes via caspase-3-mediated release of a lipid attraction signal. Cell 2003, 113, 717–730. [Google Scholar] [CrossRef]
  52. Wu, C.; Su, J.; Wang, X.; Wang, J.; Xiao, K.; Li, Y.; Xiao, Q.; Ling, M.; Xiao, Y.; Qin, C.; et al. Overexpression of the phospholipase A2 group V gene in glioma tumors is associated with poor patient prognosis. Cancer Manag. Res. 2019, 11, 3139–3152. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  53. Kishi, Y.; Okudaira, S.; Tanaka, M.; Hama, K.; Shida, D.; Kitayama, J.; Yamori, T.; Aoki, J.; Fujimaki, T.; Arai, H. Autotaxin is overexpressed in glioblastoma multiforme and contributes to cell motility of glioblastoma by converting lysophosphatidylcholine to lysophosphatidic acid. J. Biol. Chem. 2006, 281, 17492–17500. [Google Scholar] [CrossRef] [PubMed]
  54. Hoelzinger, D.B.; Nakada, M.; Demuth, T.; Rosensteel, T.; Reavie, L.B.; Berens, M.E. Autotaxin: A secreted autocrine/paracrine factor that promotes glioma invasion. J. Neurooncol. 2008, 86, 297–309. [Google Scholar] [CrossRef] [PubMed]
  55. Amaral, R.F.; Geraldo, L.H.M.; Einicker-Lamas, M.; Spohr, T.C.L.d.S.e.; Mendes, F.; Lima, F.R.S. Microglial lysophosphatidic acid promotes glioblastoma proliferation and migration via LPA1 receptor. J. Neurochem. 2021, 156, 499–512. [Google Scholar] [CrossRef] [PubMed]
  56. Loskutov, Y.V.; Griffin, C.L.; Marinak, K.M.; Bobko, A.; Margaryan, N.V.; Geldenhuys, W.J.; Sarkaria, J.N.; Pugacheva, E.N. LPA signaling is regulated through the primary cilium: A novel target in glioblastoma. Oncogene 2018, 37, 1457–1471. [Google Scholar] [CrossRef] [PubMed]
  57. Malchinkhuu, E.; Sato, K.; Maehama, T.; Ishiuchi, S.; Yoshimoto, Y.; Mogi, C.; Kimura, T.; Kurose, H.; Tomura, H.; Okajima, F. Role of Rap1B and tumor suppressor PTEN in the negative regulation of lysophosphatidic acid--induced migration by isoproterenol in glioma cells. Mol. Biol. Cell 2009, 20, 5156–5165. [Google Scholar] [CrossRef] [PubMed]
  58. Bhave, S.R.; Dadey, D.Y.; Karvas, R.M.; Ferraro, D.J.; Kotipatruni, R.P.; Jaboin, J.J.; Hallahan, A.N.; Dewees, T.A.; Linkous, A.G.; Hallahan, D.E.; et al. Autotaxin Inhibition with PF-8380 Enhances the Radiosensitivity of Human and Murine Glioblastoma Cell Lines. Front. Oncol. 2013, 3, 236. [Google Scholar] [CrossRef]
  59. Valdés-Rives, S.A.; de la Fuente-Granada, M.; Velasco-Velázquez, M.A.; González-Flores, O.; González-Arenas, A. LPA1 receptor activation induces PKCα nuclear translocation in glioblastoma cells. Int. J. Biochem. Cell Biol. 2019, 110, 91–102. [Google Scholar] [CrossRef]
  60. Valdés-Rives, S.A.; Arcos-Montoya, D.; de la Fuente-Granada, M.; Zamora-Sánchez, C.J.; Arias-Romero, L.E.; Villamar-Cruz, O.; Camacho-Arroyo, I.; Pérez-Tapia, S.M.; González-Arenas, A. LPA1 Receptor Promotes Progesterone Receptor Phosphorylation through PKCα in Human Glioblastoma Cells. Cells 2021, 10, 807. [Google Scholar] [CrossRef]
  61. Rai, V.; Touré, F.; Chitayat, S.; Pei, R.; Song, F.; Li, Q.; Zhang, J.; Rosario, R.; Ramasamy, R.; Chazin, W.J.; et al. Lysophosphatidic acid targets vascular and oncogenic pathways via RAGE signaling. J. Exp. Med. 2012, 209, 2339–2350. [Google Scholar] [CrossRef]
  62. Cechin, S.R.; Dunkley, P.R.; Rodnight, R. Signal transduction mechanisms involved in the proliferation of C6 glioma cells induced by lysophosphatidic acid. Neurochem. Res. 2005, 30, 603–611. [Google Scholar] [CrossRef] [PubMed]
  63. Martín, R.; Hernández, M.; Ibeas, E.; Fuentes, L.; Salicio, V.; Arnés, M.; Nieto, M.L. Secreted phospholipase A2-IIA modulates key regulators of proliferation on astrocytoma cells. J. Neurochem. 2009, 111, 988–999. [Google Scholar] [CrossRef]
  64. Hernández, M.; Martín, R.; García-Cubillas, M.D.; Maeso-Hernández, P.; Nieto, M.L. Secreted PLA2 induces proliferation in astrocytoma through the EGF receptor: Another inflammation-cancer link. Neuro Oncol. 2010, 12, 1014–1023. [Google Scholar] [CrossRef]
  65. Martín, R.; Cordova, C.; Gutiérrez, B.; Hernández, M.; Nieto, M.L. A dangerous liaison: Leptin and sPLA2-IIA join forces to induce proliferation and migration of astrocytoma cells. PLoS ONE 2017, 12, e0170675. [Google Scholar] [CrossRef] [PubMed]
  66. Lai, Y.J.; Lin, V.T.; Zheng, Y.; Benveniste, E.N.; Lin, F.T. The adaptor protein TRIP6 antagonizes Fas-induced apoptosis but promotes its effect on cell migration. Mol. Cell. Biol. 2010, 30, 5582–5596. [Google Scholar] [CrossRef] [PubMed]
  67. Annabi, B.; Lachambre, M.P.; Plouffe, K.; Sartelet, H.; Béliveau, R. Modulation of invasive properties of CD133+ glioblastoma stem cells: A role for MT1-MMP in bioactive lysophospholipid signaling. Mol. Carcinog. 2009, 48, 910–919. [Google Scholar] [CrossRef]
  68. Kita, Y.; Shindou, H.; Shimizu, T. Cytosolic phospholipase A2 and lysophospholipid acyltransferases. Biochim. Biophys. Acta BBA-Mol. Cell Biol. Lipids 2019, 1864, 838–845. [Google Scholar] [CrossRef] [PubMed]
  69. Hunt, M.C.; Alexson, S.E. The role Acyl-CoA thioesterases play in mediating intracellular lipid metabolism. Prog. Lipid Res. 2002, 41, 99–130. [Google Scholar] [CrossRef]
  70. Hunt, M.C.; Yamada, J.; Maltais, L.J.; Wright, M.W.; Podesta, E.J.; Alexson, S.E. A revised nomenclature for mammalian acyl-CoA thioesterases/hydrolases. J. Lipid Res. 2005, 46, 2029–2032. [Google Scholar] [CrossRef]
  71. Miyazawa, S.; Furuta, S.; Hashimoto, T. Induction of a novel long-chain acyl-CoA hydrolase in rat liver by administration of peroxisome proliferators. Eur. J. Biochem. 1981, 117, 425–430. [Google Scholar] [CrossRef] [PubMed]
  72. Poupon, V.; Bègue, B.; Gagnon, J.; Dautry-Varsat, A.; Cerf-Bensussan, N.; Benmerah, A. Molecular cloning and characterization of MT-ACT48, a novel mitochondrial acyl-CoA thioesterase. J. Biol. Chem. 1999, 274, 19188–19194. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  73. Levin, G.; Duffin, K.L.; Obukowicz, M.G.; Hummert, S.L.; Fujiwara, H.; Needleman, P.; Raz, A. Differential metabolism of dihomo-gamma-linolenic acid and arachidonic acid by cyclo-oxygenase-1 and cyclo-oxygenase-2: Implications for cellular synthesis of prostaglandin E1 and prostaglandin E2. Biochem. J. 2002, 365, 489–496. [Google Scholar] [CrossRef] [PubMed]
  74. Nakatsugi, S.; Sugimoto, N.; Furukawa, M. Effects of non-steroidal anti-inflammatory drugs on prostaglandin E2 production by cyclooxygenase-2 from endogenous and exogenous arachidonic acid in rat peritoneal macrophages stimulated with lipopolysaccharide. Prostaglandins Leukot. Essent. Fat. Acids 1996, 55, 451–457. [Google Scholar] [CrossRef] [PubMed]
  75. Hawcroft, G.; Loadman, P.M.; Belluzzi, A.; Hull, M.A. Effect of eicosapentaenoic acid on E-type prostaglandin synthesis and EP4 receptor signaling in human colorectal cancer cells. Neoplasia 2010, 12, 618–627. [Google Scholar] [CrossRef]
  76. Nugteren, D.H.; Hazelhof, E. Isolation and properties of intermediates in prostaglandin biosynthesis. Biochim. Biophys. Acta 1973, 326, 448–461. [Google Scholar] [CrossRef]
  77. Hamberg, M.; Svensson, J.; Samuelsson, B. Prostaglandin endoperoxides. A new concept concerning the mode of action and release of prostaglandins. Proc. Natl. Acad. Sci. USA 1974, 71, 3824–3828. [Google Scholar] [CrossRef]
  78. Yu, R.; Xiao, L.; Zhao, G.; Christman, J.W.; van Breemen, R.B. Competitive enzymatic interactions determine the relative amounts of prostaglandins E2 and D2. J. Pharmacol. Exp. Ther. 2011, 339, 716–725. [Google Scholar] [CrossRef]
  79. Gu, Y.; Xu, Y.; Law, B.; Qian, S.Y. The first characterization of free radicals formed from cellular COX-catalyzed peroxidation. Free Radic. Biol. Med. 2013, 57, 49–60. [Google Scholar] [CrossRef]
  80. Engels, F.; Willems, H.; Nijkamp, F.P. Cyclooxygenase-catalyzed formation of 9-hydroxylinoleic acid by guinea pig alveolar macrophages under non-stimulated conditions. FEBS Lett. 1986, 209, 249–253. [Google Scholar] [CrossRef]
  81. Vangaveti, V.N.; Shashidhar, V.M.; Rush, C.; Malabu, U.H.; Rasalam, R.R.; Collier, F.; Baune, B.T.; Kennedy, R.L. Hydroxyoctadecadienoic acids regulate apoptosis in human THP-1 cells in a PPARγ-dependent manner. Lipids 2014, 49, 1181–1192. [Google Scholar] [CrossRef] [PubMed]
  82. Alsalem, M.; Wong, A.; Millns, P.; Arya, P.H.; Chan, M.S.; Bennett, A.; Barrett, D.A.; Chapman, V.; Kendall, D.A. The contribution of the endogenous TRPV1 ligands 9-HODE and 13-HODE to nociceptive processing and their role in peripheral inflammatory pain mechanisms. Br. J. Pharmacol. 2013, 168, 1961–1974. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  83. Obinata, H.; Hattori, T.; Nakane, S.; Tatei, K.; Izumi, T. Identification of 9-hydroxyoctadecadienoic acid and other oxidized free fatty acids as ligands of the G protein-coupled receptor G2A. J. Biol. Chem. 2005, 280, 40676–40683. [Google Scholar] [CrossRef]
  84. Wang, L.H.; Hajibeigi, A.; Xu, X.M.; Loose-Mitchell, D.; Wu, K.K. Characterization of the promoter of human prostaglandin H synthase-1 gene. Biochem. Biophys. Res. Commun. 1993, 190, 406–411. [Google Scholar] [CrossRef] [PubMed]
  85. Kosaka, T.; Miyata, A.; Ihara, H.; Hara, S.; Sugimoto, T.; Takeda, O.; Takahashi, E.; Tanabe, T. Characterization of the human gene (PTGS2) encoding prostaglandin-endoperoxide synthase 2. Eur. J. Biochem. 1994, 221, 889–897. [Google Scholar] [CrossRef]
  86. Mbonye, U.R.; Wada, M.; Rieke, C.J.; Tang, H.Y.; Dewitt, D.L.; Smith, W.L. The 19-amino acid cassette of cyclooxygenase-2 mediates entry of the protein into the endoplasmic reticulum-associated degradation system. J. Biol. Chem. 2006, 281, 35770–35778. [Google Scholar] [CrossRef]
  87. Chandrasekharan, N.V.; Dai, H.; Roos, K.L.; Evanson, N.K.; Tomsik, J.; Elton, T.S.; Simmons, D.L. COX-3, a cyclooxygenase-1 variant inhibited by acetaminophen and other analgesic/antipyretic drugs: Cloning, structure, and expression. Proc. Natl. Acad. Sci. USA 2002, 99, 13926–13931. [Google Scholar] [CrossRef]
  88. Qin, N.; Zhang, S.P.; Reitz, T.L.; Mei, J.M.; Flores, C.M. Cloning, expression, and functional characterization of human cyclooxygenase-1 splicing variants: Evidence for intron 1 retention. J. Pharmacol. Exp. Ther. 2005, 315, 1298–1305. [Google Scholar] [CrossRef]
  89. Ouellet, M.; Falgueyret, J.P.; Ear, P.H.; Pen, A.; Mancini, J.A.; Riendeau, D.; Percival, M.D. Purification and characterization of recombinant microsomal prostaglandin E synthase-1. Protein Expr. Purif. 2002, 26, 489–495. [Google Scholar] [CrossRef]
  90. Thorén, S.; Weinander, R.; Saha, S.; Jegerschöld, C.; Pettersson, P.L.; Samuelsson, B.; Hebert, H.; Hamberg, M.; Morgenstern, R.; Jakobsson, P.J. Human microsomal prostaglandin E synthase-1: Purification, functional characterization, and projection structure determination. J. Biol. Chem. 2003, 278, 22199–22209. [Google Scholar] [CrossRef]
  91. Huang, X.; Yan, W.; Gao, D.; Tong, M.; Tai, H.H.; Zhan, C.G. Structural and functional characterization of human microsomal prostaglandin E synthase-1 by computational modeling and site-directed mutagenesis. Bioorg. Med. Chem. 2006, 14, 3553–3562. [Google Scholar] [CrossRef]
  92. Murakami, M.; Nakashima, K.; Kamei, D.; Masuda, S.; Ishikawa, Y.; Ishii, T.; Ohmiya, Y.; Watanabe, K.; Kudo, I. Cellular prostaglandin E2 production by membrane-bound prostaglandin E synthase-2 via both cyclooxygenases-1 and -2. J. Biol. Chem. 2003, 278, 37937–37947. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  93. Tanioka, T.; Nakatani, Y.; Semmyo, N.; Murakami, M.; Kudo, I. Molecular identification of cytosolic prostaglandin E2 synthase that is functionally coupled with cyclooxygenase-1 in immediate prostaglandin E2 biosynthesis. J. Biol. Chem. 2000, 275, 32775–32782. [Google Scholar] [CrossRef] [PubMed]
  94. Tanioka, T.; Nakatani, Y.; Kobayashi, T.; Tsujimoto, M.; Oh-ishi, S.; Murakami, M.; Kudo, I. Regulation of cytosolic prostaglandin E2 synthase by 90-kDa heat shock protein. Biochem. Biophys. Res. Commun. 2003, 303, 1018–1023. [Google Scholar] [CrossRef] [PubMed]
  95. Schneider, A.; Zhang, Y.; Zhang, M.; Lu, W.J.; Rao, R.; Fan, X.; Redha, R.; Davis, L.; Breyer, R.M.; Harris, R.; et al. Membrane-associated PGE synthase-1 (mPGES-1) is coexpressed with both COX-1 and COX-2 in the kidney. Kidney Int. 2004, 65, 1205–1213. [Google Scholar] [CrossRef]
  96. Giannico, G.; Mendez, M.; LaPointe, M.C. Regulation of the membrane-localized prostaglandin E synthases mPGES-1 and mPGES-2 in cardiac myocytes and fibroblasts. Am. J. Physiol. Heart Circ. Physiol. 2005, 288, H165–H174. [Google Scholar] [CrossRef]
  97. Ueno, N.; Murakami, M.; Tanioka, T.; Fujimori, K.; Tanabe, T.; Urade, Y.; Kudo, I. Coupling between cyclooxygenase, terminal prostanoid synthase, and phospholipase A2. J. Biol. Chem. 2001, 276, 34918–34927. [Google Scholar] [CrossRef]
  98. Polet, H.; Levine, L. Metabolism of prostaglandins E, A, and C in serum. J. Biol. Chem. 1975, 250, 351–357. [Google Scholar] [CrossRef] [PubMed]
  99. Jones, R.L. Preparation of prostaglandins C: Chemical fixation of prostaglandin A isomerase to a gel support and partition chromatography of prostaglandins A, B and C. Prostaglandins 1974, 5, 283–290. [Google Scholar] [CrossRef]
  100. Lee, B.R.; Paing, M.H.; Sharma-Walia, N. Cyclopentenone Prostaglandins: Biologically Active Lipid Mediators Targeting Inflammation. Front. Physiol. 2021, 12, 640374. [Google Scholar] [CrossRef] [PubMed]
  101. Fitzpatrick, F.A.; Wynalda, M.A. Albumin-catalyzed metabolism of prostaglandin D2. Identification of products formed in vitro. J. Biol. Chem. 1983, 258, 11713–11718. [Google Scholar] [CrossRef] [PubMed]
  102. Shibata, T.; Kondo, M.; Osawa, T.; Shibata, N.; Kobayashi, M.; Uchida, K. 15-deoxy-delta 12,14-prostaglandin J2. A prostaglandin D2 metabolite generated during inflammatory processes. J. Biol. Chem. 2002, 277, 10459–10466. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  103. Mahmud, I.; Smith, D.L.; Whyte, M.A.; Nelson, J.T.; Cho, D.; Tokes, L.G.; Alvarez, R.; Willis, A.L. On the identification and biological properties of prostaglandin J2. Prostaglandins Leukot. Med. 1984, 16, 131–146. [Google Scholar] [CrossRef] [PubMed]
  104. Yamaguchi, S.; Aldini, G.; Ito, S.; Morishita, N.; Shibata, T.; Vistoli, G.; Carini, M.; Uchida, K. Delta12-prostaglandin J2 as a product and ligand of human serum albumin: Formation of an unusual covalent adduct at His146. J. Am. Chem. Soc. 2010, 132, 824–832. [Google Scholar] [CrossRef] [PubMed]
  105. Straus, D.S.; Glass, C.K. Cyclopentenone prostaglandins: New insights on biological activities and cellular targets. Med. Res. Rev. 2001, 21, 185–210. [Google Scholar] [CrossRef]
  106. Hsu, P.Y.; Tsai, A.L.; Kulmacz, R.J.; Wang, L.H. Expression, purification, and spectroscopic characterization of human thromboxane synthase. J. Biol. Chem. 1999, 274, 762–769. [Google Scholar] [CrossRef] [PubMed]
  107. Watkins, G.; Douglas-Jones, A.; Mansel, R.E.; Jiang, W.G. Expression of thromboxane synthase, TBXAS1 and the thromboxane A2 receptor, TBXA2R, in human breast cancer. Int. Semin. Surg. Oncol. 2005, 2, 23. [Google Scholar] [CrossRef]
  108. Hamberg, M.; Svensson, J.; Samuelsson, B. Thromboxanes: A new group of biologically active compounds derived from prostaglandin endoperoxides. Proc. Natl. Acad. Sci. USA 1975, 72, 2994–2998. [Google Scholar] [CrossRef]
  109. Matsunobu, T.; Okuno, T.; Yokoyama, C.; Yokomizo, T. Thromboxane A synthase-independent production of 12-hydroxyheptadecatrienoic acid, a BLT2 ligand. J. Lipid Res. 2013, 54, 2979–2987. [Google Scholar] [CrossRef]
  110. Okuno, T.; Iizuka, Y.; Okazaki, H.; Yokomizo, T.; Taguchi, R.; Shimizu, T. 12(S)-Hydroxyheptadeca-5Z, 8E, 10E-trienoic acid is a natural ligand for leukotriene B4 receptor 2. J. Exp. Med. 2008, 205, 759–766. [Google Scholar] [CrossRef]
  111. Liu, M.; Saeki, K.; Matsunobu, T.; Okuno, T.; Koga, T.; Sugimoto, Y.; Yokoyama, C.; Nakamizo, S.; Kabashima, K.; Narumiya, S.; et al. 12-Hydroxyheptadecatrienoic acid promotes epidermal wound healing by accelerating keratinocyte migration via the BLT2 receptor. J. Exp. Med. 2014, 211, 1063–1078. [Google Scholar] [CrossRef] [PubMed]
  112. Giusti, F.; Casiraghi, M.; Point, E.; Damian, M.; Rieger, J.; Bon, C.L.; Pozza, A.; Moncoq, K.; Banères, J.L.; Catoire, L.J. Structure of the agonist 12-HHT in its BLT2 receptor-bound state. Sci. Rep. 2020, 10, 2630. [Google Scholar] [CrossRef] [Green Version]
  113. Cathcart, M.C.; Gray, S.G.; Baird, A.M.; Boyle, E.; Gately, K.; Kay, E.; Cummins, R.; Pidgeon, G.P.; O’Byrne, K.J. Prostacyclin synthase expression and epigenetic regulation in nonsmall cell lung cancer. Cancer 2011, 117, 5121–5132. [Google Scholar] [CrossRef] [PubMed]
  114. Dozier, B.L.; Watanabe, K.; Duffy, D.M. Two pathways for prostaglandin F2 alpha synthesis by the primate periovulatory follicle. Reproduction 2008, 136, 53–63. [Google Scholar] [CrossRef] [PubMed]
  115. Bresson, E.; Boucher-Kovalik, S.; Chapdelaine, P.; Madore, E.; Harvey, N.; Laberge, P.Y.; Leboeuf, M.; Fortier, M.A. The human aldose reductase AKR1B1 qualifies as the primary prostaglandin F synthase in the endometrium. J. Clin. Endocrinol. Metab. 2011, 96, 210–219. [Google Scholar] [CrossRef] [PubMed]
  116. Reid, G.; Wielinga, P.; Zelcer, N.; van der Heijden, I.; Kuil, A.; de Haas, M.; Wijnholds, J.; Borst, P. The human multidrug resistance protein MRP4 functions as a prostaglandin efflux transporter and is inhibited by nonsteroidal antiinflammatory drugs. Proc. Natl. Acad. Sci. USA 2003, 100, 9244–9249. [Google Scholar] [CrossRef]
  117. Lu, R.; Kanai, N.; Bao, Y.; Schuster, V.L. Cloning, in vitro expression, and tissue distribution of a human prostaglandin transporter cDNA(hPGT). J. Clin. Investig. 1996, 98, 1142–1149. [Google Scholar] [CrossRef]
  118. Nomura, T.; Lu, R.; Pucci, M.L.; Schuster, V.L. The two-step model of prostaglandin signal termination: In vitro reconstitution with the prostaglandin transporter and prostaglandin 15 dehydrogenase. Mol. Pharmacol. 2004, 65, 973–978. [Google Scholar] [CrossRef]
  119. Cattori, V.; van Montfoort, J.E.; Stieger, B.; Landmann, L.; Meijer, D.K.; Winterhalter, K.H.; Meier, P.J.; Hagenbuch, B. Localization of organic anion transporting polypeptide 4 (Oatp4) in rat liver and comparison of its substrate specificity with Oatp1, Oatp2 and Oatp3. Pflugers Arch. 2001, 443, 188–195. [Google Scholar] [CrossRef]
  120. Chou, W.L.; Chuang, L.M.; Chou, C.C.; Wang, A.H.; Lawson, J.A.; FitzGerald, G.A.; Chang, Z.F. Identification of a novel prostaglandin reductase reveals the involvement of prostaglandin E2 catabolism in regulation of peroxisome proliferator-activated receptor gamma activation. J. Biol. Chem. 2007, 282, 18162–18172. [Google Scholar] [CrossRef]
  121. Panagopoulos, A.T.; Gomes, R.N.; Almeida, F.G.; da Costa Souza, F.; Veiga, J.C.E.; Nicolaou, A.; Colquhoun, A. The prostanoid pathway contains potential prognostic markers for glioblastoma. Prostaglandins Other Lipid Mediat. 2018, 137, 52–62. [Google Scholar] [CrossRef] [PubMed]
  122. Granström, E.; Hamberg, M.; Hansson, G.; Kindahl, H. Chemical instability of 15-keto-13,14-dihydro-PGE2: The reason for low assay reliability. Prostaglandins 1980, 19, 933–957. [Google Scholar] [CrossRef] [PubMed]
  123. Bremme, K.; Eneroth, P.; Gottlieb, C.; Kindahl, H.; Svanborg, K.; Nilsson, B.; Olsson, M.; Bygdeman, M. 15-Keto-13,14-dihydroprostaglandin E2- and F2 alpha-metabolite levels in blood from men and women given prostaglandin E2 orally. Prostaglandins Leukot. Essent. Fat. Acids 1989, 37, 169–176. [Google Scholar] [CrossRef] [PubMed]
  124. Okumura, T.; Nakayama, R.; Sago, T.; Saito, K. Identification of prostaglandin E metabolites from primary cultures of rat hepatocytes. Biochim. Biophys. Acta 1985, 837, 197–207. [Google Scholar]
  125. Schepers, L.; Casteels, M.; Vamecq, J.; Parmentier, G.; Van Veldhoven, P.P.; Mannaerts, G.P. Beta-oxidation of the carboxyl side chain of prostaglandin E2 in rat liver peroxisomes and mitochondria. J. Biol. Chem. 1988, 263, 2724–2731. [Google Scholar] [CrossRef]
  126. Powell, W.S.; Solomon, S. Formation of 20-hydroxyprostaglandins by lungs of pregnant rabbits. J. Biol. Chem. 1978, 253, 4609–4616. [Google Scholar] [CrossRef]
  127. Hamberg, M.; Samuelsson, B. The structure of the major urinary metabolite of prostaglandin E2 in man. J. Am. Chem. Soc. 1969, 91, 2177–2178. [Google Scholar] [CrossRef]
  128. Hamberg, M.; Samuelsson, B. On the metabolism of prostaglandins E 1 and E 2 in man. J. Biol. Chem. 1971, 246, 6713–6721. [Google Scholar] [CrossRef]
  129. Mancini, J.A.; O’Neill, G.P.; Bayly, C.; Vickers, P.J. Mutation of serine-516 in human prostaglandin G/H synthase-2 to methionine or aspirin acetylation of this residue stimulates 15-R-HETE synthesis. FEBS Lett. 1994, 342, 33–37. [Google Scholar] [CrossRef]
  130. Mulugeta, S.; Suzuki, T.; Hernandez, N.T.; Griesser, M.; Boeglin, W.E.; Schneider, C. Identification and absolute configuration of dihydroxy-arachidonic acids formed by oxygenation of 5S-HETE by native and aspirin-acetylated COX-2. J. Lipid Res. 2010, 51, 575–585. [Google Scholar] [CrossRef]
  131. Tejera, N.; Boeglin, W.E.; Suzuki, T.; Schneider, C. COX-2-dependent and -independent biosynthesis of dihydroxy-arachidonic acids in activated human leukocytes. J. Lipid Res. 2012, 53, 87–94. [Google Scholar] [CrossRef] [PubMed]
  132. Lucido, M.J.; Orlando, B.J.; Vecchio, A.J.; Malkowski, M.G. Crystal Structure of Aspirin-Acetylated Human Cyclooxygenase-2: Insight into the Formation of Products with Reversed Stereochemistry. Biochemistry 2016, 55, 1226–1238. [Google Scholar] [CrossRef] [PubMed]
  133. Loll, P.J.; Picot, D.; Garavito, R.M. The structural basis of aspirin activity inferred from the crystal structure of inactivated prostaglandin H2 synthase. Nat. Struct. Biol. 1995, 2, 637–643. [Google Scholar] [CrossRef] [PubMed]
  134. Fiorucci, S.; Distrutti, E.; de Lima, O.M.; Romano, M.; Mencarelli, A.; Barbanti, M.; Palazzini, E.; Morelli, A.; Wallace, J.L. Relative contribution of acetylated cyclo-oxygenase (COX)-2 and 5-lipooxygenase (LOX) in regulating gastric mucosal integrity and adaptation to aspirin. FASEB J. 2003, 17, 1171–1173. [Google Scholar] [CrossRef]
  135. Wada, M.; DeLong, C.J.; Hong, Y.H.; Rieke, C.J.; Song, I.; Sidhu, R.S.; Yuan, C.; Warnock, M.; Schmaier, A.H.; Yokoyama, C.; et al. Enzymes and receptors of prostaglandin pathways with arachidonic acid-derived versus eicosapentaenoic acid-derived substrates and products. J. Biol. Chem. 2007, 282, 22254–22266. [Google Scholar] [CrossRef]
  136. Mengeaud, V.; Nano, J.L.; Fournel, S.; Rampal, P. Effects of eicosapentaenoic acid, gamma-linolenic acid and prostaglandin E1 on three human colon carcinoma cell lines. Prostaglandins Leukot. Essent. Fat. Acids 1992, 47, 313–319. [Google Scholar] [CrossRef]
  137. Tabolacci, C.; Lentini, A.; Provenzano, B.; Gismondi, A.; Rossi, S.; Beninati, S. Similar antineoplastic effects of nimesulide, a selective COX-2 inhibitor, and prostaglandin E1 on B16-F10 murine melanoma cells. Melanoma Res. 2010, 20, 273–279. [Google Scholar] [CrossRef]
  138. Xu, Y.; Qi, J.; Yang, X.; Wu, E.; Qian, S.Y. Free radical derivatives formed from cyclooxygenase-catalyzed dihomo-γ-linolenic acid peroxidation can attenuate colon cancer cell growth and enhance 5-fluorouracil’s cytotoxicity. Redox Biol. 2014, 2, 610–618. [Google Scholar] [CrossRef]
  139. Xu, Y.; Yang, X.; Zhao, P.; Yang, Z.; Yan, C.; Guo, B.; Qian, S.Y. Knockdown of delta-5-desaturase promotes the anti-cancer activity of dihomo-γ-linolenic acid and enhances the efficacy of chemotherapy in colon cancer cells expressing COX-2. Free Radic. Biol. Med. 2016, 96, 67–77. [Google Scholar] [CrossRef]
  140. Ishizaki, Y.; Morita, I.; Murota, S. Arachidonic acid metabolism in cultured astrocytes from rat embryo and in C6 glioma cells. Brain Res. 1989, 494, 138–142. [Google Scholar] [CrossRef]
  141. Deininger, M.H.; Weller, M.; Streffer, J.; Mittelbronn, M.; Meyermann, R. Patterns of cyclooxygenase-1 and -2 expression in human gliomas in vivo. Acta Neuropathol. 1999, 98, 240–244. [Google Scholar] [CrossRef] [PubMed]
  142. Joki, T.; Heese, O.; Nikas, D.C.; Bello, L.; Zhang, J.; Kraeft, S.K.; Seyfried, N.T.; Abe, T.; Chen, L.B.; Carroll, R.S.; et al. Expression of cyclooxygenase 2 (COX-2) in human glioma and in vitro inhibition by a specific COX-2 inhibitor, NS-398. Cancer Res. 2000, 60, 4926–4931. [Google Scholar] [PubMed]
  143. Mattila, S.; Tuominen, H.; Koivukangas, J.; Stenbäck, F. The terminal prostaglandin synthases mPGES-1, mPGES-2, and cPGES are all overexpressed in human gliomas. Neuropathology 2009, 29, 156–165. [Google Scholar] [CrossRef] [PubMed]
  144. Giese, A.; Hagel, C.; Kim, E.L.; Zapf, S.; Djawaheri, J.; Berens, M.E.; Westphal, M. Thromboxane synthase regulates the migratory phenotype of human glioma cells. Neuro Oncol. 1999, 1, 3–13. [Google Scholar] [CrossRef]
  145. Castelli, M.G.; Butti, G.; Chiabrando, C.; Cozzi, E.; Fanelli, R.; Gaetani, P.; Silvani, V.; Paoletti, P. Arachidonic acid metabolic profiles in human meningiomas and gliomas. J. Neurooncol. 1987, 5, 369–375. [Google Scholar] [CrossRef] [PubMed]
  146. Zhao, S.; Jiang, X.; Xue, D.; Chen, D. Glioma prostaglandin levels correlate with brain edema. J. Tongji Med. Univ. 1998, 18, 115–118. [Google Scholar] [PubMed]
  147. Lo, H.W.; Cao, X.; Zhu, H.; Ali-Osman, F. Cyclooxygenase-2 is a novel transcriptional target of the nuclear EGFR-STAT3 and EGFRvIII-STAT3 signaling axes. Mol. Cancer Res. 2010, 8, 232–245. [Google Scholar] [CrossRef]
  148. Xu, K.; Shu, H.K. Transcription factor interactions mediate EGF-dependent COX-2 expression. Mol. Cancer Res. 2013, 11, 875–886. [Google Scholar] [CrossRef]
  149. Zhao, Y.; Sun, Y.; Zhang, H.; Liu, X.; Du, W.; Li, Y.; Zhang, J.; Chen, L.; Jiang, C. HGF/MET signaling promotes glioma growth via up-regulation of Cox-2 expression and PGE2 production. Int. J. Clin. Exp. Pathol. 2015, 8, 3719–3726. [Google Scholar]
  150. Nakano, Y.; Kuroda, E.; Kito, T.; Yokota, A.; Yamashita, U. Induction of macrophagic prostaglandin E2 synthesis by glioma cells. J. Neurosurg. 2006, 104, 574–582. [Google Scholar] [CrossRef]
  151. Huang, N.; Chen, S.; Deng, J.; Huang, Q.; Liao, P.; Wang, F.; Cheng, Y. Overexpression of S100A9 in human glioma and in-vitro inhibition by aspirin. Eur. J. Cancer Prev. 2013, 22, 585–595. [Google Scholar] [CrossRef] [PubMed]
  152. Fiebich, B.L.; Hüll, M.; Lieb, K.; Gyufko, K.; Berger, M.; Bauer, J. Prostaglandin E2 induces interleukin-6 synthesis in human astrocytoma cells. J. Neurochem. 1997, 68, 704–709. [Google Scholar] [CrossRef] [PubMed]
  153. Venza, I.; Visalli, M.; Fortunato, C.; Ruggeri, M.; Ratone, S.; Caffo, M.; Caruso, G.; Alafaci, C.; Tomasello, F.; Teti, D.; et al. PGE2 induces interleukin-8 derepression in human astrocytoma through coordinated DNA demethylation and histone hyperacetylation. Epigenetics 2012, 7, 1315–1330. [Google Scholar] [CrossRef] [PubMed]
  154. Kardosh, A.; Blumenthal, M.; Wang, W.J.; Chen, T.C.; Schönthal, A.H. Differential effects of selective COX-2 inhibitors on cell cycle regulation and proliferation of glioblastoma cell lines. Cancer Biol. Ther. 2004, 3, 55–62. [Google Scholar] [CrossRef] [Green Version]
  155. Payner, T.; Leaver, H.A.; Knapp, B.; Whittle, I.R.; Trifan, O.C.; Miller, S.; Rizzo, M.T. Microsomal prostaglandin E synthase-1 regulates human glioma cell growth via prostaglandin E2–dependent activation of type II protein kinase A. Mol. Cancer Ther. 2006, 5, 1817–1826. [Google Scholar] [CrossRef] [PubMed]
  156. Ferreira, M.T.; Miyake, J.A.; Gomes, R.N.; Feitoza, F.; Stevannato, P.B.; da Cunha, A.S.; Serachi, F.O.; Panagopoulos, A.T.; Colquhoun, A. Cyclooxygenase Inhibition Alters Proliferative, Migratory, and Invasive Properties of Human Glioblastoma Cells In Vitro. Int. J. Mol. Sci. 2021, 22, 4297. [Google Scholar] [CrossRef]
  157. Tian, Y.; Yang, T.; Yu, S.; Liu, C.; He, M.; Hu, C. Prostaglandin E2 increases migration and proliferation of human glioblastoma cells by activating transient receptor potential melastatin 7 channels. J. Cell Mol. Med. 2018, 22, 6327–6337. [Google Scholar] [CrossRef]
  158. Annabi, B.; Laflamme, C.; Sina, A.; Lachambre, M.P.; Béliveau, R. A MT1-MMP/NF-kappaB signaling axis as a checkpoint controller of COX-2 expression in CD133(+) U87 glioblastoma cells. J. Neuroinflamm. 2009, 6, 8. [Google Scholar] [CrossRef]
  159. Wu, M.; Guan, J.; Li, C.; Gunter, S.; Nusrat, L.; Ng, S.; Dhand, K.; Morshead, C.; Kim, A.; Das, S. Aberrantly activated Cox-2 and Wnt signaling interact to maintain cancer stem cells in glioblastoma. Oncotarget 2017, 8, 82217–82230. [Google Scholar] [CrossRef]
  160. El-Sayed, M.; Taha, M.M. Immunohistochemical expression of cycloxygenase-2 in astrocytoma: Correlation with angiogenesis, tumor progression and survival. Turk. Neurosurg. 2011, 21, 27–35. [Google Scholar] [CrossRef]
  161. Rong, X.; Huang, B.; Qiu, S.; Li, X.; He, L.; Peng, Y. Tumor-associated macrophages induce vasculogenic mimicry of glioblastoma multiforme through cyclooxygenase-2 activation. Oncotarget 2016, 7, 83976–83986. [Google Scholar] [CrossRef] [PubMed]
  162. Addison, C.L.; Daniel, T.O.; Burdick, M.D.; Liu, H.; Ehlert, J.E.; Xue, Y.Y.; Buechi, L.; Walz, A.; Richmond, A.; Strieter, R.M. The CXC chemokine receptor 2, CXCR2, is the putative receptor for ELR+ CXC chemokine-induced angiogenic activity. J. Immunol. 2000, 165, 5269–5277. [Google Scholar] [CrossRef] [PubMed]
  163. Ochs, K.; Ott, M.; Rauschenbach, K.J.; Deumelandt, K.; Sahm, F.; Opitz, C.A.; von Deimling, A.; Wick, W.; Platten, M. Tryptophan-2,3-dioxygenase is regulated by prostaglandin E2 in malignant glioma via a positive signaling loop involving prostaglandin E receptor-4. J. Neurochem. 2016, 136, 1142–1154. [Google Scholar] [CrossRef] [PubMed]
  164. Fujita, M.; Kohanbash, G.; Fellows-Mayle, W.; Hamilton, R.L.; Komohara, Y.; Decker, S.A.; Ohlfest, J.R.; Okada, H. COX-2 blockade suppresses gliomagenesis by inhibiting myeloid-derived suppressor cells. Cancer Res. 2011, 71, 2664–2674. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  165. Ma, X.; Holt, D.; Kundu, N.; Reader, J.; Goloubeva, O.; Take, Y.; Fulton, A.M. A prostaglandin E (PGE) receptor EP4 antagonist protects natural killer cells from PGE2-mediated immunosuppression and inhibits breast cancer metastasis. Oncoimmunology 2013, 2, e22647. [Google Scholar] [CrossRef]
  166. Park, A.; Lee, Y.; Kim, M.S.; Kang, Y.J.; Park, Y.J.; Jung, H.; Kim, T.D.; Lee, H.G.; Choi, I.; Yoon, S.R. Prostaglandin E2 Secreted by Thyroid Cancer Cells Contributes to Immune Escape Through the Suppression of Natural Killer (NK) Cell Cytotoxicity and NK Cell Differentiation. Front. Immunol. 2018, 9, 1859. [Google Scholar] [CrossRef]
  167. Sharma, S.; Stolina, M.; Yang, S.C.; Baratelli, F.; Lin, J.F.; Atianzar, K.; Luo, J.; Zhu, L.; Lin, Y.; Huang, M.; et al. Tumor cyclooxygenase 2-dependent suppression of dendritic cell function. Clin. Cancer Res. 2003, 9, 961–968. [Google Scholar]
  168. Yuan, X.L.; Chen, L.; Li, M.X.; Dong, P.; Xue, J.; Wang, J.; Zhang, T.T.; Wang, X.A.; Zhang, F.M.; Ge, H.L.; et al. Elevated expression of Foxp3 in tumor-infiltrating Treg cells suppresses T-cell proliferation and contributes to gastric cancer progression in a COX-2-dependent manner. Clin. Immunol. 2010, 134, 277–288. [Google Scholar] [CrossRef]
  169. Liu, L.; Ge, D.; Ma, L.; Mei, J.; Liu, S.; Zhang, Q.; Ren, F.; Liao, H.; Pu, Q.; Wang, T.; et al. Interleukin-17 and prostaglandin E2 are involved in formation of an M2 macrophage-dominant microenvironment in lung cancer. J. Thorac. Oncol. 2012, 7, 1091–1100. [Google Scholar] [CrossRef]
  170. Brocard, E.; Oizel, K.; Lalier, L.; Pecqueur, C.; Paris, F.; Vallette, F.M.; Oliver, L. Radiation-induced PGE2 sustains human glioma cells growth and survival through EGF signaling. Oncotarget 2015, 6, 6840–6849. [Google Scholar] [CrossRef]
  171. Cook, P.J.; Thomas, R.; Kingsley, P.J.; Shimizu, F.; Montrose, D.C.; Marnett, L.J.; Tabar, V.S.; Dannenberg, A.J.; Benezra, R. Cox-2-derived PGE2 induces Id1-dependent radiation resistance and self-renewal in experimental glioblastoma. Neuro Oncol. 2016, 18, 1379–1389. [Google Scholar] [CrossRef] [PubMed]
  172. Tsai, Y.T.; Lo, W.L.; Chen, P.Y.; Ko, C.Y.; Chuang, J.Y.; Kao, T.J.; Yang, W.B.; Chang, K.Y.; Hung, C.Y.; Kikkawa, U.; et al. Reprogramming of arachidonate metabolism confers temozolomide resistance to glioblastoma through enhancing mitochondrial activity in fatty acid oxidation. J. Biomed. Sci. 2022, 29, 21. [Google Scholar] [CrossRef]
  173. Lombardi, F.; Augello, F.R.; Artone, S.; Ayroldi, E.; Giusti, I.; Dolo, V.; Cifone, M.G.; Cinque, B.; Palumbo, P. Cyclooxygenase-2 Upregulated by Temozolomide in Glioblastoma Cells Is Shuttled In Extracellular Vesicles Modifying Recipient Cell Phenotype. Front. Oncol. 2022, 12, 933746. [Google Scholar] [CrossRef] [PubMed]
  174. Feng, X.; Yu, Y.; He, S.; Cheng, J.; Gong, Y.; Zhang, Z.; Yang, X.; Xu, B.; Liu, X.; Li, C.Y.; et al. Dying glioma cells establish a proangiogenic microenvironment through a caspase 3 dependent mechanism. Cancer Lett. 2017, 385, 12–20. [Google Scholar] [CrossRef] [PubMed]
  175. Desmarais, G.; Charest, G.; Therriault, H.; Shi, M.; Fortin, D.; Bujold, R.; Mathieu, D.; Paquette, B. Infiltration of F98 glioma cells in Fischer rat brain is temporary stimulated by radiation. Int. J. Radiat. Biol. 2016, 92, 444–450. [Google Scholar] [CrossRef]
  176. Desmarais, G.; Charest, G.; Fortin, D.; Bujold, R.; Mathieu, D.; Paquette, B. Cyclooxygenase-2 inhibitor prevents radiation-enhanced infiltration of F98 glioma cells in brain of Fischer rat. Int. J. Radiat. Biol. 2015, 91, 624–633. [Google Scholar] [CrossRef]
  177. Ferreira, M.T.; Gomes, R.N.; Panagopoulos, A.T.; de Almeida, F.G.; Veiga, J.C.E.; Colquhoun, A. Opposing roles of PGD2 in GBM. Prostaglandins Other Lipid Mediat. 2018, 134, 66–76. [Google Scholar] [CrossRef]
  178. Keyaki, A.; Handa, H.; Yamashita, J.; Tokuriki, Y.; Otsuka, S.; Yamasaki, T.; Gi, H. Growth-inhibitory effect of prostaglandin D2 on mouse glioma cells. J. Neurosurg. 1984, 61, 912–917. [Google Scholar] [CrossRef]
  179. Westphal, M.; Neuss, M.; Herrmann, H.D. Prostaglandins: Antiproliferative effect of PGD 2 on cultured human glioma cells. Acta Neurochir. 1986, 83, 56–61. [Google Scholar] [CrossRef]
  180. Cho, W.H.; Choi, C.H.; Park, J.Y.; Kang, S.K.; Kim, Y.K. 15-deoxy-(Delta12,14)-prostaglandin J2 (15d-PGJ2) induces cell death through caspase-independent mechanism in A172 human glioma cells. Neurochem. Res. 2006, 31, 1247–1254. [Google Scholar] [CrossRef]
  181. Nakahata, N.; Abe, M.T.; Nakanishi, H. PGJ2 and delta 12PGJ2 inhibit cell growth accompanied with inhibition of phosphoinositide turnover in human astrocytoma cells. Prostaglandins 1990, 40, 405–416. [Google Scholar] [CrossRef] [PubMed]
  182. Morosetti, R.; Servidei, T.; Mirabella, M.; Rutella, S.; Mangiola, A.; Maira, G.; Mastrangelo, R.; Koeffler, H.P. The PPARgamma ligands PGJ2 and rosiglitazone show a differential ability to inhibit proliferation and to induce apoptosis and differentiation of human glioblastoma cell lines. Int. J. Oncol. 2004, 25, 493–502. [Google Scholar]
  183. Obara, Y.; Kurose, H.; Nakahata, N. Thromboxane A2 promotes interleukin-6 biosynthesis mediated by an activation of cyclic AMP-response element-binding protein in 1321N1 human astrocytoma cells. Mol. Pharmacol. 2005, 68, 670–679. [Google Scholar] [CrossRef] [PubMed]
  184. Yoshizato, K.; Zapf, S.; Westphal, M.; Berens, M.E.; Giese, A. Thromboxane synthase inhibitors induce apoptosis in migration-arrested glioma cells. Neurosurgery 2002, 50, 343–354. [Google Scholar] [PubMed]
  185. Schmidt, N.O.; Ziu, M.; Cargioli, T.; Westphal, M.; Giese, A.; Black, P.M.; Carroll, R.S. Inhibition of thromboxane synthase activity improves glioblastoma response to alkylation chemotherapy. Transl. Oncol. 2010, 3, 43–49. [Google Scholar] [CrossRef]
  186. Schauff, A.K.; Kim, E.L.; Leppert, J.; Nadrowitz, R.; Wuestenberg, R.; Brockmann, M.A.; Giese, A. Inhibition of invasion-associated thromboxane synthase sensitizes experimental gliomas to gamma-radiation. J. Neurooncol. 2009, 91, 241–249. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  187. Shono, T.; Tofilon, P.J.; Bruner, J.M.; Owolabi, O.; Lang, F.F. Cyclooxygenase-2 expression in human gliomas: Prognostic significance and molecular correlations. Cancer Res. 2001, 61, 4375–4381. [Google Scholar] [PubMed]
  188. Wang, X.; Chen, Y.; Zhang, S.; Zhang, L.; Liu, X.; Zhang, L.; Li, X.; Chen, D. Co-expression of COX-2 and 5-LO in primary glioblastoma is associated with poor prognosis. J. Neurooncol. 2015, 125, 277–285. [Google Scholar] [CrossRef]
  189. Zohn, I.E.; Klinger, M.; Karp, X.; Kirk, H.; Symons, M.; Chrzanowska-Wodnicka, M.; Der, C.J.; Kay, R.J. G2A is an oncogenic G protein-coupled receptor. Oncogene 2000, 19, 3866–3877. [Google Scholar] [CrossRef]
  190. Nam, D.H.; Park, K.; Park, C.; Im, Y.H.; Kim, M.H.; Lee, S.; Hong, S.C.; Shin, H.J.; Kim, J.H.; Eoh, W.; et al. Intracranial inhibition of glioma cell growth by cyclooxygenase-2 inhibitor celecoxib. Oncol. Rep. 2004, 11, 263–268. [Google Scholar] [CrossRef]
  191. Tuettenberg, J.; Grobholz, R.; Korn, T.; Wenz, F.; Erber, R.; Vajkoczy, P. Continuous low-dose chemotherapy plus inhibition of cyclooxygenase-2 as an antiangiogenic therapy of glioblastoma multiforme. J. Cancer Res. Clin. Oncol. 2005, 131, 31–40. [Google Scholar] [CrossRef] [PubMed]
  192. Zhang, T.; Yang, X.; Liu, P.; Zhou, J.; Luo, J.; Wang, H.; Li, A.; Zhou, Y. Association between nonsteroidal anti-inflammatory drugs use and risk of central nervous system tumors: A dose-response meta analysis. Oncotarget 2017, 8, 102486–102498. [Google Scholar] [CrossRef] [PubMed]
  193. Amirian, E.S.; Ostrom, Q.T.; Armstrong, G.N.; Lai, R.K.; Gu, X.; Jacobs, D.I.; Jalali, A.; Claus, E.B.; Barnholtz-Sloan, J.S.; Il’yasova, D.; et al. Aspirin, NSAIDs, and Glioma Risk: Original Data from the Glioma International Case-Control Study and a Meta-analysis. Cancer Epidemiol. Biomark. Prev. 2019, 28, 555–562. [Google Scholar] [CrossRef]
  194. Liu, Y.; Lu, Y.; Wang, J.; Xie, L.; Li, T.; He, Y.; Peng, Q.; Qin, X.; Li, S. Association between nonsteroidal anti-inflammatory drug use and brain tumour risk: A meta-analysis. Br. J. Clin. Pharmacol. 2014, 78, 58–68. [Google Scholar] [CrossRef]
  195. Funk, C.D.; Funk, L.B.; FitzGerald, G.A.; Samuelsson, B. Characterization of human 12-lipoxygenase genes. Proc. Natl. Acad. Sci. USA 1992, 89, 3962–3966. [Google Scholar] [CrossRef]
  196. Krieg, P.; Marks, F.; Fürstenberger, G. A gene cluster encoding human epidermis-type lipoxygenases at chromosome 17p13.1: Cloning, physical mapping, and expression. Genomics 2001, 73, 323–330. [Google Scholar] [CrossRef]
  197. Jisaka, M.; Kim, R.B.; Boeglin, W.E.; Brash, A.R. Identification of amino acid determinants of the positional specificity of mouse 8S-lipoxygenase and human 15S-lipoxygenase-2. J. Biol. Chem. 2000, 275, 1287–1293. [Google Scholar] [CrossRef] [PubMed]
  198. Fürstenberger, G.; Marks, F.; Krieg, P. Arachidonate 8(S)-lipoxygenase. Prostaglandins Other Lipid Mediat. 2002, 68–69, 235–243. [Google Scholar] [CrossRef]
  199. Fagerberg, L.; Hallström, B.M.; Oksvold, P.; Kampf, C.; Djureinovic, D.; Odeberg, J.; Habuka, M.; Tahmasebpoor, S.; Danielsson, A.; Edlund, K.; et al. Analysis of the human tissue-specific expression by genome-wide integration of transcriptomics and antibody-based proteomics. Mol. Cell. Proteom. 2014, 13, 397–406. [Google Scholar] [CrossRef]
  200. Yu, Z.; Schneider, C.; Boeglin, W.E.; Marnett, L.J.; Brash, A.R. The lipoxygenase gene ALOXE3 implicated in skin differentiation encodes a hydroperoxide isomerase. Proc. Natl. Acad. Sci. USA 2003, 100, 9162–9167. [Google Scholar] [CrossRef]
  201. Zheng, Y.; Brash, A.R. Dioxygenase activity of epidermal lipoxygenase-3 unveiled: Typical and atypical features of its catalytic activity with natural and synthetic polyunsaturated fatty acids. J. Biol. Chem. 2010, 285, 39866–39875. [Google Scholar] [CrossRef] [PubMed]
  202. Yu, Z.; Schneider, C.; Boeglin, W.E.; Brash, A.R. Human and mouse eLOX3 have distinct substrate specificities: Implications for their linkage with lipoxygenases in skin. Arch. Biochem. Biophys. 2006, 455, 188–196. [Google Scholar] [CrossRef] [PubMed]
  203. Gregus, A.M.; Dumlao, D.S.; Wei, S.C.; Norris, P.C.; Catella, L.C.; Meyerstein, F.G.; Buczynski, M.W.; Steinauer, J.J.; Fitzsimmons, B.L.; Yaksh, T.L.; et al. Systematic analysis of rat 12/15-lipoxygenase enzymes reveals critical role for spinal eLOX3 hepoxilin synthase activity in inflammatory hyperalgesia. FASEB J. 2013, 27, 1939–1949. [Google Scholar] [CrossRef] [PubMed]
  204. Higgins, C.B.; Zhang, Y.; Mayer, A.L.; Fujiwara, H.; Stothard, A.I.; Graham, M.J.; Swarts, B.M.; DeBosch, B.J. Hepatocyte ALOXE3 is induced during adaptive fasting and enhances insulin sensitivity by activating hepatic PPARγ. JCI Insight 2018, 3, e120794. [Google Scholar] [CrossRef] [PubMed]
  205. Yang, X.; Liu, J.; Wang, C.; Cheng, K.K.; Xu, H.; Li, Q.; Hua, T.; Jiang, X.; Sheng, L.; Mao, J.; et al. miR-18a promotes glioblastoma development by down-regulating ALOXE3-mediated ferroptotic and anti-migration activities. Oncogenesis 2021, 10, 15. [Google Scholar] [CrossRef] [PubMed]
  206. Powell, W.S.; Rokach, J. Biosynthesis, biological effects, and receptors of hydroxyeicosatetraenoic acids (HETEs) and oxoeicosatetraenoic acids (oxo-ETEs) derived from arachidonic acid. Biochim. Biophys. Acta 2015, 1851, 340–355. [Google Scholar] [CrossRef] [Green Version]
  207. Pace-Asciak, C.R. Pathophysiology of the hepoxilins. Biochim. Biophys. Acta 2015, 1851, 383–396. [Google Scholar] [CrossRef]
  208. Yu, Z.; Schneider, C.; Boeglin, W.E.; Brash, A.R. Mutations associated with a congenital form of ichthyosis (NCIE) inactivate the epidermal lipoxygenases 12R-LOX and eLOX3. Biochim. Biophys. Acta 2005, 1686, 238–247. [Google Scholar] [CrossRef]
  209. Wang, T.; Xu, C.; Zhou, X.; Li, C.; Zhang, H.; Lian, B.Q.; Lee, J.J.; Shen, J.; Liu, Y.; Lian, C.G. Homozygous ALOXE3 Nonsense Variant Identified in a Patient with Non-Bullous Congenital Ichthyosiform Erythroderma Complicated by Superimposed Bullous Majocchi’s Granuloma: The Consequences of Skin Barrier Dysfunction. Int. J. Mol. Sci. 2015, 16, 21791–21801. [Google Scholar] [CrossRef]
  210. Hotz, A.; Kopp, J.; Bourrat, E.; Oji, V.; Komlosi, K.; Giehl, K.; Bouadjar, B.; Bygum, A.; Tantcheva-Poor, I.; Hellström Pigg, M.; et al. Meta-Analysis of Mutations in ALOX12B or ALOXE3 Identified in a Large Cohort of 224 Patients. Genes 2021, 12, 80. [Google Scholar] [CrossRef]
  211. Ueda, N.; Kaneko, S.; Yoshimoto, T.; Yamamoto, S. Purification of arachidonate 5-lipoxygenase from porcine leukocytes and its reactivity with hydroperoxyeicosatetraenoic acids. J. Biol. Chem. 1986, 261, 7982–7988. [Google Scholar] [CrossRef] [PubMed]
  212. Mancini, J.A.; Abramovitz, M.; Cox, M.E.; Wong, E.; Charleson, S.; Perrier, H.; Wang, Z.; Prasit, P.; Vickers, P.J. 5-lipoxygenase-activating protein is an arachidonate binding protein. FEBS Lett. 1993, 318, 277–281. [Google Scholar] [CrossRef] [PubMed]
  213. Häfner, A.K.; Gerstmeier, J.; Hörnig, M.; George, S.; Ball, A.K.; Schröder, M.; Garscha, U.; Werz, O.; Steinhilber, D. Characterization of the interaction of human 5-lipoxygenase with its activating protein FLAP. Biochim. Biophys. Acta 2015, 1851, 1465–1472. [Google Scholar] [CrossRef] [PubMed]
  214. Narala, V.R.; Adapala, R.K.; Suresh, M.V.; Brock, T.G.; Peters-Golden, M.; Reddy, R.C. Leukotriene B4 is a physiologically relevant endogenous peroxisome proliferator-activated receptor-alpha agonist. J. Biol. Chem. 2010, 285, 22067–22074. [Google Scholar] [CrossRef]
  215. Falgueyret, J.; Riendeau, D. LTA4-derived 5-oxo-eicosatetraenoic acid: pH-dependent formation and interaction with the LTB4 receptor of human polymorphonuclear leukocytes. Biochim. Biophys. Acta 2000, 1484, 51–58. [Google Scholar] [CrossRef]
  216. Chiba, N.; Imai, H.; Narashima, K.; Arai, M.; Oshima, G.; Kunimoto, M.; Nakagawa, Y. Cellular glutathione peroxidase as a predominant scavenger of hydroperoxyeicosatetraenoic acids in rabbit alveolar macrophages. Biol. Pharm. Bull. 1999, 22, 1047–1051. [Google Scholar] [CrossRef] [Green Version]
  217. Erlemann, K.R.; Cossette, C.; Grant, G.E.; Lee, G.J.; Patel, P.; Rokach, J.; Powell, W.S. Regulation of 5-hydroxyeicosanoid dehydrogenase activity in monocytic cells. Biochem. J. 2007, 403, 157–165. [Google Scholar] [CrossRef]
  218. Patel, P.; Cossette, C.; Anumolu, J.R.; Erlemann, K.R.; Grant, G.E.; Rokach, J.; Powell, W.S. Substrate selectivity of 5-hydroxyeicosanoid dehydrogenase and its inhibition by 5-hydroxy-Delta6-long-chain fatty acids. J. Pharmacol. Exp. Ther. 2009, 329, 335–341. [Google Scholar] [CrossRef]
  219. Nagendra Reddy, C.; Ye, Q.; Patel, P.; Sivendran, S.; Chourey, S.; Wang, R.; Anumolu, J.R.; Grant, G.E.; Powell, W.S.; Rokach, J. Design and synthesis of affinity chromatography ligands for the purification of 5-hydroxyeicosanoid dehydrogenase. Bioorg. Med. Chem. 2017, 25, 116–125. [Google Scholar] [CrossRef]
  220. Hosoi, T.; Koguchi, Y.; Sugikawa, E.; Chikada, A.; Ogawa, K.; Tsuda, N.; Suto, N.; Tsunoda, S.; Taniguchi, T.; Ohnuki, T. Identification of a novel human eicosanoid receptor coupled to Gi/o. J. Biol. Chem. 2002, 277, 31459–31465. [Google Scholar] [CrossRef]
  221. Jones, C.E.; Holden, S.; Tenaillon, L.; Bhatia, U.; Seuwen, K.; Tranter, P.; Turner, J.; Kettle, R.; Bouhelal, R.; Charlton, S.; et al. Expression and characterization of a 5-oxo-6E,8Z,11Z,14Z-eicosatetraenoic acid receptor highly expressed on human eosinophils and neutrophils. Mol. Pharmacol. 2003, 63, 471–477. [Google Scholar] [CrossRef] [PubMed]
  222. Kalyvianaki, K.; Drosou, I.; Notas, G.; Castanas, E.; Kampa, M. Enhanced OXER1 expression is indispensable for human cancer cell migration. Biochem. Biophys. Res. Commun. 2021, 584, 95–100. [Google Scholar] [CrossRef] [PubMed]
  223. Serhan, C.N. Lipoxin biosynthesis and its impact in inflammatory and vascular events. Biochim. Biophys. Acta 1994, 1212, 1–25. [Google Scholar] [CrossRef] [PubMed]
  224. Rådmark, O.; Shimizu, T.; Jörnvall, H.; Samuelsson, B. Leukotriene A4 hydrolase in human leukocytes. Purification and properties. J. Biol. Chem. 1984, 259, 12339–12345. [Google Scholar] [CrossRef]
  225. Rudberg, P.C.; Tholander, F.; Andberg, M.; Thunnissen, M.M.; Haeggström, J.Z. Leukotriene A4 hydrolase: Identification of a common carboxylate recognition site for the epoxide hydrolase and aminopeptidase substrates. J. Biol. Chem. 2004, 279, 27376–27382. [Google Scholar] [CrossRef] [PubMed]
  226. Paige, M.; Wang, K.; Burdick, M.; Park, S.; Cha, J.; Jeffery, E.; Sherman, N.; Shim, Y.M. Role of leukotriene A4 hydrolase aminopeptidase in the pathogenesis of emphysema. J. Immunol. 2014, 192, 5059–5068. [Google Scholar] [CrossRef] [Green Version]
  227. Yokomizo, T.; Izumi, T.; Chang, K.; Takuwa, Y.; Shimizu, T. A G-protein-coupled receptor for leukotriene B4 that mediates chemotaxis. Nature 1997, 387, 620–624. [Google Scholar] [CrossRef] [PubMed]
  228. Yokomizo, T.; Kato, K.; Terawaki, K.; Izumi, T.; Shimizu, T. A second leukotriene B4 receptor, BLT2. A new therapeutic target in inflammation and immunological disorders. J. Exp. Med. 2000, 192, 421–432. [Google Scholar] [CrossRef]
  229. Lam, B.K.; Penrose, J.F.; Freeman, G.J.; Austen, K.F. Expression cloning of a cDNA for human leukotriene C4 synthase, an integral membrane protein conjugating reduced glutathione to leukotriene A4. Proc. Natl. Acad. Sci. USA 1994, 91, 7663–7667. [Google Scholar] [CrossRef]
  230. Ago, H.; Kanaoka, Y.; Irikura, D.; Lam, B.K.; Shimamura, T.; Austen, K.F.; Miyano, M. Crystal structure of a human membrane protein involved in cysteinyl leukotriene biosynthesis. Nature 2007, 448, 609–612. [Google Scholar] [CrossRef]
  231. Strid, T.; Svartz, J.; Franck, N.; Hallin, E.; Ingelsson, B.; Söderström, M.; Hammarström, S. Distinct parts of leukotriene C4 synthase interact with 5-lipoxygenase and 5-lipoxygenase activating protein. Biochem. Biophys. Res. Commun. 2009, 381, 518–522. [Google Scholar] [CrossRef] [PubMed]
  232. Wickham, S.; West, M.B.; Cook, P.F.; Hanigan, M.H. Gamma-glutamyl compounds: Substrate specificity of gamma-glutamyl transpeptidase enzymes. Anal. Biochem. 2011, 414, 208–214. [Google Scholar] [CrossRef] [PubMed]
  233. Adachi, H.; Kubota, I.; Okamura, N.; Iwata, H.; Tsujimoto, M.; Nakazato, H.; Nishihara, T.; Noguchi, T. Purification and characterization of human microsomal dipeptidase. J. Biochem. 1989, 105, 957–961. [Google Scholar] [CrossRef] [PubMed]
  234. Habib, G.M.; Shi, Z.Z.; Cuevas, A.A.; Lieberman, M.W. Identification of two additional members of the membrane-bound dipeptidase family. FASEB J. 2003, 17, 1313–1315. [Google Scholar] [CrossRef]
  235. Reddanna, P.; Prabhu, K.S.; Whelan, J.; Reddy, C.C. Carboxypeptidase A-catalyzed direct conversion of leukotriene C4 to leukotriene F4. Arch. Biochem. Biophys. 2003, 413, 158–163. [Google Scholar] [CrossRef]
  236. Bernström, K.; Hammarström, S. A novel leukotriene formed by transpeptidation of leukotriene E. Biochem. Biophys. Res. Commun. 1982, 109, 800–804. [Google Scholar] [CrossRef]
  237. Jedlitschky, G.; Keppler, D. Transport of leukotriene C4 and structurally related conjugates. Vitam. Horm. 2002, 64, 153–184. [Google Scholar]
  238. Zhang, D.W.; Nunoya, K.; Vasa, M.; Gu, H.M.; Cole, S.P.; Deeley, R.G. Mutational analysis of polar amino acid residues within predicted transmembrane helices 10 and 16 of multidrug resistance protein 1 (ABCC1): Effect on substrate specificity. Drug Metab. Dispos. 2006, 34, 539–546. [Google Scholar] [CrossRef]
  239. Johnson, Z.L.; Chen, J. Structural Basis of Substrate Recognition by the Multidrug Resistance Protein MRP1. Cell 2017, 168, 1075–1085. [Google Scholar] [CrossRef]
  240. Cui, Y.; König, J.; Buchholz, J.K.; Spring, H.; Leier, I.; Keppler, D. Drug resistance and ATP-dependent conjugate transport mediated by the apical multidrug resistance protein, MRP2, permanently expressed in human and canine cells. Mol. Pharmacol. 1999, 55, 929–937. [Google Scholar]
  241. Kamisako, T.; Leier, I.; Cui, Y.; König, J.; Buchholz, U.; Hummel-Eisenbeiss, J.; Keppler, D. Transport of monoglucuronosyl and bisglucuronosyl bilirubin by recombinant human and rat multidrug resistance protein 2. Hepatology 1999, 30, 485–490. [Google Scholar] [CrossRef] [PubMed]
  242. Lee, Y.M.; Cui, Y.; König, J.; Risch, A.; Jäger, B.; Drings, P.; Bartsch, H.; Keppler, D.; Nies, A.T. Identification and functional characterization of the natural variant MRP3-Arg1297His of human multidrug resistance protein 3 (MRP3/ABCC3). Pharmacogenetics 2004, 14, 213–223. [Google Scholar] [CrossRef] [PubMed]
  243. Rius, M.; Hummel-Eisenbeiss, J.; Keppler, D. ATP-dependent transport of leukotrienes B4 and C4 by the multidrug resistance protein ABCC4 (MRP4). J. Pharmacol. Exp. Ther. 2008, 324, 86–94. [Google Scholar] [CrossRef]
  244. Belinsky, M.G.; Chen, Z.S.; Shchaveleva, I.; Zeng, H.; Kruh, G.D. Characterization of the drug resistance and transport properties of multidrug resistance protein 6 (MRP6, ABCC6). Cancer Res. 2002, 62, 6172–6177. [Google Scholar] [PubMed]
  245. Chen, Z.S.; Hopper-Borge, E.; Belinsky, M.G.; Shchaveleva, I.; Kotova, E.; Kruh, G.D. Characterization of the transport properties of human multidrug resistance protein 7 (MRP7, ABCC10). Mol. Pharmacol. 2003, 63, 351–358. [Google Scholar] [CrossRef] [PubMed]
  246. Chen, Z.S.; Guo, Y.; Belinsky, M.G.; Kotova, E.; Kruh, G.D. Transport of bile acids, sulfated steroids, estradiol 17-beta-D-glucuronide, and leukotriene C4 by human multidrug resistance protein 8 (ABCC11). Mol. Pharmacol. 2005, 67, 545–557. [Google Scholar] [CrossRef] [PubMed]
  247. Li, L.; Lee, T.K.; Meier, P.J.; Ballatori, N. Identification of glutathione as a driving force and leukotriene C4 as a substrate for oatp1, the hepatic sinusoidal organic solute transporter. J. Biol. Chem. 1998, 273, 16184–16191. [Google Scholar] [CrossRef]
  248. Yokomizo, T.; Kato, K.; Hagiya, H.; Izumi, T.; Shimizu, T. Hydroxyeicosanoids bind to and activate the low affinity leukotriene B4 receptor, BLT2. J. Biol. Chem. 2001, 276, 12454–12459. [Google Scholar] [CrossRef] [Green Version]
  249. Lynch, K.R.; O’Neill, G.P.; Liu, Q.; Im, D.S.; Sawyer, N.; Metters, K.M.; Coulombe, N.; Abramovitz, M.; Figueroa, D.J.; Zeng, Z.; et al. Characterization of the human cysteinyl leukotriene CysLT1 receptor. Nature 1999, 399, 789–793. [Google Scholar] [CrossRef]
  250. Heise, C.E.; O’Dowd, B.F.; Figueroa, D.J.; Sawyer, N.; Nguyen, T.; Im, D.S.; Stocco, R.; Bellefeuille, J.N.; Abramovitz, M.; Cheng, R.; et al. Characterization of the human cysteinyl leukotriene 2 receptor. J. Biol. Chem. 2000, 275, 30531–30536. [Google Scholar] [CrossRef]
  251. Nothacker, H.P.; Wang, Z.; Zhu, Y.; Reinscheid, R.K.; Lin, S.H.; Civelli, O. Molecular cloning and characterization of a second human cysteinyl leukotriene receptor: Discovery of a subtype selective agonist. Mol. Pharmacol. 2000, 58, 1601–1608. [Google Scholar] [CrossRef] [PubMed]
  252. Kanaoka, Y.; Maekawa, A.; Austen, K.F. Identification of GPR99 protein as a potential third cysteinyl leukotriene receptor with a preference for leukotriene E4 ligand. J. Biol. Chem. 2013, 288, 10967–10972. [Google Scholar] [CrossRef] [PubMed]
  253. Ciana, P.; Fumagalli, M.; Trincavelli, M.L.; Verderio, C.; Rosa, P.; Lecca, D.; Ferrario, S.; Parravicini, C.; Capra, V.; Gelosa, P.; et al. The orphan receptor GPR17 identified as a new dual uracil nucleotides/cysteinyl-leukotrienes receptor. EMBO J. 2006, 25, 4615–4627. [Google Scholar] [CrossRef] [PubMed]
  254. Qi, A.D.; Harden, T.K.; Nicholas, R.A. Is GPR17 a P2Y/leukotriene receptor? examination of uracil nucleotides, nucleotide sugars, and cysteinyl leukotrienes as agonists of GPR17. J. Pharmacol. Exp. Ther. 2013, 347, 38–46. [Google Scholar] [CrossRef] [PubMed]
  255. Simon, K.; Merten, N.; Schröder, R.; Hennen, S.; Preis, P.; Schmitt, N.K.; Peters, L.; Schrage, R.; Vermeiren, C.; Gillard, M.; et al. The Orphan Receptor GPR17 Is Unresponsive to Uracil Nucleotides and Cysteinyl Leukotrienes. Mol. Pharmacol. 2017, 91, 518–532. [Google Scholar] [CrossRef] [PubMed]
  256. Maekawa, A.; Balestrieri, B.; Austen, K.F.; Kanaoka, Y. GPR17 is a negative regulator of the cysteinyl leukotriene 1 receptor response to leukotriene D4. Proc. Natl. Acad. Sci. USA 2009, 106, 11685–11690. [Google Scholar] [CrossRef]
  257. Wheelan, P.; Zirrolli, J.A.; Morelli, J.G.; Murphy, R.C. Metabolism of leukotriene B4 by cultured human keratinocytes. Formation of glutathione conjugates and dihydro metabolites. J. Biol. Chem. 1993, 268, 25439–25448. [Google Scholar] [CrossRef]
  258. Yokomizo, T.; Ogawa, Y.; Uozumi, N.; Kume, K.; Izumi, T.; Shimizu, T. cDNA cloning, expression, and mutagenesis study of leukotriene B4 12-hydroxydehydrogenase. J. Biol. Chem. 1996, 271, 2844–2850. [Google Scholar] [CrossRef] [Green Version]
  259. Tobin, D.M.; Roca, F.J.; Ray, J.P.; Ko, D.C.; Ramakrishnan, L. An enzyme that inactivates the inflammatory mediator leukotriene B4 restricts mycobacterial infection. PLoS ONE 2013, 8, e67828. [Google Scholar] [CrossRef]
  260. Wainwright, S.L.; Powell, W.S. Mechanism for the formation of dihydro metabolites of 12-hydroxyeicosanoids. Conversion of leukotriene B4 and 12-hydroxy-5,8,10,14-eicosatetraenoic acid to 12-oxo intermediates. J. Biol. Chem. 1991, 266, 20899–20906. [Google Scholar] [CrossRef]
  261. Hagmann, W.; Korte, M. Hepatic uptake and metabolic disposition of leukotriene B4 in rats. Biochem. J. 1990, 267, 467–470. [Google Scholar] [CrossRef] [PubMed]
  262. Berry, K.A.; Borgeat, P.; Gosselin, J.; Flamand, L.; Murphy, R.C. Urinary metabolites of leukotriene B4 in the human subject. J. Biol. Chem. 2003, 278, 24449–24460. [Google Scholar] [CrossRef] [PubMed]
  263. Wainwright, S.; Falck, J.R.; Yadagiri, P.; Powell, W.S. Metabolism of 12(S)-hydroxy-5,8,10,14-eicosatetraenoic acid and other hydroxylated fatty acids by the reductase pathway in porcine polymorphonuclear leukocytes. Biochemistry 1990, 29, 10126–10135. [Google Scholar] [CrossRef] [PubMed]
  264. Huber, M.; Müller, J.; Leier, I.; Jedlitschky, G.; Ball, H.A.; Moore, K.P.; Taylor, G.W.; Williams, R.; Keppler, D. Metabolism of cysteinyl leukotrienes in monkey and man. Eur. J. Biochem. 1990, 194, 309–315. [Google Scholar] [CrossRef]
  265. Chang, W.C.; Ning, C.C.; Lin, M.T.; Huang, J.D. Epidermal growth factor enhances a microsomal 12-lipoxygenase activity in A431 cells. J. Biol. Chem. 1992, 267, 3657–3666. [Google Scholar] [CrossRef]
  266. Wecksler, A.T.; Kenyon, V.; Deschamps, J.D.; Holman, T.R. Substrate specificity changes for human reticulocyte and epithelial 15-lipoxygenases reveal allosteric product regulation. Biochemistry 2008, 47, 7364–7375. [Google Scholar] [CrossRef]
  267. Ikei, K.N.; Yeung, J.; Apopa, P.L.; Ceja, J.; Vesci, J.; Holman, T.R.; Holinstat, M. Investigations of human platelet-type 12-lipoxygenase: Role of lipoxygenase products in platelet activation. J. Lipid Res. 2012, 53, 2546–2559. [Google Scholar] [CrossRef]
  268. Yeung, J.; Tourdot, B.E.; Adili, R.; Green, A.R.; Freedman, C.J.; Fernandez-Perez, P.; Yu, J.; Holman, T.R.; Holinstat, M. 12(S)-HETrE, a 12-Lipoxygenase Oxylipin of Dihomo-γ-Linolenic Acid, Inhibits Thrombosis via Gαs Signaling in Platelets. Arterioscler. Thromb. Vasc. Biol. 2016, 36, 2068–2077. [Google Scholar] [CrossRef] [Green Version]
  269. Guo, Y.; Zhang, W.; Giroux, C.; Cai, Y.; Ekambaram, P.; Dilly, A.K.; Hsu, A.; Zhou, S.; Maddipati, K.R.; Liu, J.; et al. Identification of the orphan G protein-coupled receptor GPR31 as a receptor for 12-(S)-hydroxyeicosatetraenoic acid. J. Biol. Chem. 2011, 286, 33832–33840. [Google Scholar] [CrossRef]
  270. Sun, L.; Xu, Y.W.; Han, J.; Liang, H.; Wang, N.; Cheng, Y. 12/15-Lipoxygenase metabolites of arachidonic acid activate PPARγ: A possible neuroprotective effect in ischemic brain. J. Lipid Res. 2015, 56, 502–514. [Google Scholar] [CrossRef]
  271. Falgueyret, J.P.; Leblanc, Y.; Riendeau, D. Stereoselective carbonyl reductases from rat skin and leukocyte microsomes converting 12-ketoeicosatetraenoic acid to 12(S)-HETE. FEBS Lett. 1990, 262, 197–200. [Google Scholar] [CrossRef] [PubMed]
  272. Pace-Asciak, C.R. Hemoglobin- and hemin-catalyzed transformation of 12L-hydroperoxy-5,8,10,14-eicosatetraenoic acid. Biochim. Biophys. Acta 1984, 793, 485–488. [Google Scholar] [CrossRef] [PubMed]
  273. Reynaud, D.; Demin, P.; Pace-Asciak, C.R. Hepoxilin A3 formation in the rat pineal gland selectively utilizes (12S)-hydroperoxyeicosatetraenoic acid (HPETE), but not (12R)-HPETE. J. Biol. Chem. 1994, 269, 23976–23980. [Google Scholar] [CrossRef] [PubMed]
  274. Nigam, S.; Patabhiraman, S.; Ciccoli, R.; Ishdorj, G.; Schwarz, K.; Petrucev, B.; Kühn, H.; Haeggström, J.Z. The rat leukocyte-type 12-lipoxygenase exhibits an intrinsic hepoxilin A3 synthase activity. J. Biol. Chem. 2004, 279, 29023–29030. [Google Scholar] [CrossRef] [PubMed]
  275. Krieg, P.; Rosenberger, S.; de Juanes, S.; Latzko, S.; Hou, J.; Dick, A.; Kloz, U.; van der Hoeven, F.; Hausser, I.; Esposito, I.; et al. Aloxe3 knockout mice reveal a function of epidermal lipoxygenase-3 as hepoxilin synthase and its pivotal role in barrier formation. J. Investig. Dermatol. 2013, 133, 172–180. [Google Scholar] [CrossRef]
  276. Pace-Asciak, C.R.; Lee, W.S. Purification of hepoxilin epoxide hydrolase from rat liver. J. Biol. Chem. 1989, 264, 9310–9313. [Google Scholar] [CrossRef]
  277. Laneuville, O.; Chang, M.; Reddy, C.C.; Corey, E.J.; Pace-Asciak, C.R. Isozyme specificity in the conversion of hepoxilin A3 (HxA3) into a glutathionyl hepoxilin (HxA3-C) by the Yb2 subunit of rat liver glutathione S-transferase. J. Biol. Chem. 1990, 265, 21415–21418. [Google Scholar] [CrossRef]
  278. Pace-Asciak, C.R.; Laneuville, O.; Chang, M.; Reddy, C.C.; Su, W.G.; Corey, E.J. New products in the hepoxilin pathway: Isolation of 11-glutathionyl hepoxilin A3 through reaction of hepoxilin A3 with glutathione S-transferase. Biochem. Biophys. Res. Commun. 1989, 163, 1230–1234. [Google Scholar] [CrossRef]
  279. Pace-Asciak, C.R.; Laneuville, O.; Su, W.G.; Corey, E.J.; Gurevich, N.; Wu, P.; Carlen, P.L. A glutathione conjugate of hepoxilin A3: Formation and action in the rat central nervous system. Proc. Natl. Acad. Sci. USA 1990, 87, 3037–3041. [Google Scholar] [CrossRef]
  280. Cronin, A.; Decker, M.; Arand, M. Mammalian soluble epoxide hydrolase is identical to liver hepoxilin hydrolase. J. Lipid Res. 2011, 52, 712–719. [Google Scholar] [CrossRef]
  281. Gregus, A.M.; Doolen, S.; Dumlao, D.S.; Buczynski, M.W.; Takasusuki, T.; Fitzsimmons, B.L.; Hua, X.Y.; Taylor, B.K.; Dennis, E.A.; Yaksh, T.L. Spinal 12-lipoxygenase-derived hepoxilin A3 contributes to inflammatory hyperalgesia via activation of TRPV1 and TRPA1 receptors. Proc. Natl. Acad. Sci. USA 2012, 109, 6721–6726. [Google Scholar] [CrossRef] [PubMed]
  282. Singh, N.K.; Rao, G.N. Emerging role of 12/15-Lipoxygenase (ALOX15) in human pathologies. Prog. Lipid Res. 2019, 73, 28–45. [Google Scholar] [CrossRef] [PubMed]
  283. Siangjong, L.; Goldman, D.H.; Kriska, T.; Gauthier, K.M.; Smyth, E.M.; Puli, N.; Kumar, G.; Falck, J.R.; Campbell, W.B. Vascular hepoxilin and trioxilins mediate vasorelaxation through TP receptor inhibition in mouse arteries. Acta Physiol. 2017, 219, 188–201. [Google Scholar] [CrossRef] [PubMed]
  284. Boeglin, W.E.; Kim, R.B.; Brash, A.R. A 12R-lipoxygenase in human skin: Mechanistic evidence, molecular cloning, and expression. Proc. Natl. Acad. Sci. USA 1998, 95, 6744–6749. [Google Scholar] [CrossRef]
  285. Zhang, L.; Hu, Y.; Lu, J.; Zhao, P.; Zhang, X.; Tan, L.; Li, J.; Xiao, C.; Zeng, L.; He, X. Identification of the first congenital ichthyosis case caused by a homozygous deletion in the ALOX12B gene due to chromosome 17 mixed uniparental disomy. Front. Genet. 2022, 13, 931833. [Google Scholar] [CrossRef]
  286. Bryant, R.W.; Bailey, J.M.; Schewe, T.; Rapoport, S.M. Positional specificity of a reticulocyte lipoxygenase. Conversion of arachidonic acid to 15-S-hydroperoxy-eicosatetraenoic acid. J. Biol. Chem. 1982, 257, 6050–6055. [Google Scholar] [CrossRef]
  287. Sigal, E.; Dicharry, S.; Highland, E.; Finkbeiner, W.E. Cloning of human airway 15-lipoxygenase: Identity to the reticulocyte enzyme and expression in epithelium. Am. J. Physiol. 1992, 262, L392–L398. [Google Scholar] [CrossRef]
  288. Brash, A.R.; Boeglin, W.E.; Chang, M.S. Discovery of a second 15S-lipoxygenase in humans. Proc. Natl. Acad. Sci. USA 1997, 94, 6148–6152. [Google Scholar] [CrossRef] [Green Version]
  289. Altmann, R.; Hausmann, M.; Spöttl, T.; Gruber, M.; Bull, A.W.; Menzel, K.; Vogl, D.; Herfarth, H.; Schölmerich, J.; Falk, W.; et al. 13-Oxo-ODE is an endogenous ligand for PPARgamma in human colonic epithelial cells. Biochem. Pharmacol. 2007, 74, 612–622. [Google Scholar] [CrossRef]
  290. Umeno, A.; Sakashita, M.; Sugino, S.; Murotomi, K.; Okuzawa, T.; Morita, N.; Tomii, K.; Tsuchiya, Y.; Yamasaki, K.; Horie, M.; et al. Comprehensive analysis of PPARγ agonist activities of stereo-, regio-, and enantio-isomers of hydroxyoctadecadienoic acids. Biosci. Rep. 2020, 40, BSR20193767. [Google Scholar] [CrossRef]
  291. Brunnström, A.; Hamberg, M.; Griffiths, W.J.; Mannervik, B.; Claesson, H.E. Biosynthesis of 14,15-hepoxilins in human l1236 Hodgkin lymphoma cells and eosinophils. Lipids 2011, 46, 69–79. [Google Scholar] [CrossRef] [PubMed]
  292. Feltenmark, S.; Gautam, N.; Brunnström, A.; Griffiths, W.; Backman, L.; Edenius, C.; Lindbom, L.; Björkholm, M.; Claesson, H.E. Eoxins are proinflammatory arachidonic acid metabolites produced via the 15-lipoxygenase-1 pathway in human eosinophils and mast cells. Proc. Natl. Acad. Sci. USA 2008, 105, 680–685. [Google Scholar] [CrossRef] [PubMed]
  293. Green, A.R.; Freedman, C.; Tena, J.; Tourdot, B.E.; Liu, B.; Holinstat, M.; Holman, T.R. 5S,15S-Dihydroperoxyeicosatetraenoic Acid (5,15-diHpETE) as a Lipoxin Intermediate: Reactivity and Kinetics with Human Leukocyte 5-Lipoxygenase, Platelet 12-Lipoxygenase, and Reticulocyte 15-Lipoxygenase-1. Biochemistry 2018, 57, 6726–6734. [Google Scholar] [CrossRef] [PubMed]
  294. Steinhilber, D.; Roth, H.J. New series of lipoxins isolated from human eosinophils. FEBS Lett. 1989, 255, 143–148. [Google Scholar] [CrossRef]
  295. Tornhamre, S.; Elmqvist, A.; Lindgren, J.A. 15-Lipoxygenation of leukotriene A4. Studies Of 12- and 15-lipoxygenase efficiency to catalyze lipoxin formation. Biochim. Biophys. Acta 2000, 1484, 298–306. [Google Scholar] [CrossRef]
  296. Fiore, S.; Maddox, J.F.; Perez, H.D.; Serhan, C.N. Identification of a human cDNA encoding a functional high affinity lipoxin A4 receptor. J. Exp. Med. 1994, 180, 253–260. [Google Scholar] [CrossRef]
  297. Romano, M.; Recchia, I.; Recchiuti, A. Lipoxin receptors. Sci. World J. 2007, 7, 1393–1412. [Google Scholar] [CrossRef]
  298. Schaldach, C.M.; Riby, J.; Bjeldanes, L.F. Lipoxin A4: A new class of ligand for the Ah receptor. Biochemistry 1999, 38, 7594–7600. [Google Scholar] [CrossRef]
  299. Russell, R.; Gori, I.; Pellegrini, C.; Kumar, R.; Achtari, C.; Canny, G.O. Lipoxin A4 is a novel estrogen receptor modulator. FASEB J. 2011, 25, 4326–4337. [Google Scholar] [CrossRef]
  300. Boado, R.J.; Pardridge, W.M.; Vinters, H.V.; Black, K.L. Differential expression of arachidonate 5-lipoxygenase transcripts in human brain tumors: Evidence for the expression of a multitranscript family. Proc. Natl. Acad. Sci. USA 1992, 89, 9044–9048. [Google Scholar] [CrossRef]
  301. Golubic, M.; Prayson, R.A.; Vargo, L.; Bondar, J.; Barnett, G.H. Increased expression of 5-lipoxygenase in glioblastoma multiforme. Adv. Exp. Med. Biol. 2003, 525, 205–208. [Google Scholar] [PubMed]
  302. Nathoo, N.; Prayson, R.A.; Bondar, J.; Vargo, L.; Arrigain, S.; Mascha, E.J.; Suh, J.H.; Barnett, G.H.; Golubic, M. Increased expression of 5-lipoxygenase in high-grade astrocytomas. Neurosurgery 2006, 58, 347–354. [Google Scholar] [CrossRef] [PubMed]
  303. Wang, B.; Yu, S.C.; Jiang, J.Y.; Porter, G.W.; Zhao, L.T.; Wang, Z.; Tan, H.; Cui, Y.H.; Qian, C.; Ping, Y.F.; et al. An inhibitor of arachidonate 5-lipoxygenase, Nordy, induces differentiation and inhibits self-renewal of glioma stem-like cells. Stem. Cell Rev. Rep. 2011, 7, 458–470. [Google Scholar] [CrossRef] [PubMed]
  304. Simmet, T.; Luck, W.; Winking, M.; Delank, W.K.; Peskar, B.A. Identification and characterization of cysteinyl-leukotriene formation in tissue slices from human intracranial tumors: Evidence for their biosynthesis under in vivo conditions. J. Neurochem. 1990, 54, 2091–2099. [Google Scholar] [CrossRef] [PubMed]
  305. Black, K.L.; Hoff, J.T.; McGillicuddy, J.E.; Gebarski, S.S. Increased leukotriene C4 and vasogenic edema surrounding brain tumors in humans. Ann. Neurol. 1986, 19, 592–595. [Google Scholar] [CrossRef]
  306. Jin, T.B.; Li, X.L.; Yang, H.; Jiri, M.; Shi, X.G.; Yuan, D.Y.; Kang, L.L.; Li, S.Q. Association of polymorphisms in FLT3, EGFR, ALOX5, and NEIL3 with glioblastoma in the Han Chinese population. Med. Oncol. 2013, 30, 718. [Google Scholar] [CrossRef]
  307. Kim, J.A.; Chung, Y.J.; Lee, Y.S. Intracellular Ca2+ mediates lipoxygenase-induced proliferation of U-373 MG human astrocytoma cells. Arch. Pharm. Res. 1998, 21, 664–670. [Google Scholar] [CrossRef]
  308. Lim, J.Y.; Oh, J.H.; Jung, J.R.; Kim, S.M.; Ryu, C.H.; Kim, H.T.; Jeun, S.S. MK886-induced apoptosis depends on the 5-LO expression level in human malignant glioma cells. J. Neurooncol. 2010, 97, 339–346. [Google Scholar] [CrossRef]
  309. Souza, F.D.C.; Ferreira, M.T.; Colquhoun, A. Influence of Lipoxygenase Inhibition on Glioblastoma Cell Biology. Int. J. Mol. Sci. 2020, 21, 8395. [Google Scholar] [CrossRef]
  310. Xu, J.P.; Liu, H.; Bian, X.W.; Chen, J.H.; Zhou, X.D.; Wu, Y.Z. Effect of nordy on biological behaviors of malignant glioma cell line U87MG and the analysis of differential expression proteome. Zhonghua Bing Li Xue Za Zhi 2007, 36, 609–613. [Google Scholar]
  311. Ishii, K.; Zaitsu, M.; Yonemitsu, N.; Kan, Y.; Hamasaki, Y.; Matsuo, M. 5-lipoxygenase pathway promotes cell proliferation in human glioma cell lines. Clin. Neuropathol. 2009, 28, 445–452. [Google Scholar] [CrossRef] [PubMed]
  312. Gáti, I.; Bergström, M.; Csóka, K.; Muhr, C.; Carlsson, J. Effects of the 5-lipoxygenase inhibitors AA-863 and U-60,257 on human glioma cell lines. Prostaglandins Leukot. Essent. Fat. Acids 1990, 40, 117–124. [Google Scholar] [CrossRef] [PubMed]
  313. Piromkraipak, P.; Parakaw, T.; Phuagkhaopong, S.; Srihirun, S.; Chongthammakun, S.; Chaithirayanon, K.; Vivithanaporn, P. Cysteinyl leukotriene receptor antagonists induce apoptosis and inhibit proliferation of human glioblastoma cells by downregulating B-cell lymphoma 2 and inducing cell cycle arrest. Can. J. Physiol. Pharmacol. 2018, 96, 798–806. [Google Scholar] [CrossRef] [PubMed]
  314. Abbott, N.J. Blood-brain barrier structure and function and the challenges for CNS drug delivery. J. Inherit. Metab. Dis. 2013, 36, 437–449. [Google Scholar] [CrossRef] [PubMed]
  315. Chio, C.C.; Baba, T.; Black, K.L. Selective blood-tumor barrier disruption by leukotrienes. J. Neurosurg. 1992, 77, 407–410. [Google Scholar] [CrossRef]
  316. Black, K.L.; King, W.A.; Ikezaki, K. Selective opening of the blood-tumor barrier by intracarotid infusion of leukotriene C4. J. Neurosurg. 1990, 72, 912–916. [Google Scholar] [CrossRef]
  317. Black, P.; Hand, C.M.; Vender, J.R.; Finkelstein, S.D. Chemotherapy in experimental brain tumor, part 2: Pretreatment with leukotriene C4 prolongs survival. J. Neurooncol. 1998, 36, 7–19. [Google Scholar] [CrossRef]
  318. Fellner, S.; Bauer, B.; Miller, D.S.; Schaffrik, M.; Fankhänel, M.; Spruss, T.; Bernhardt, G.; Graeff, C.; Färber, L.; Gschaidmeier, H.; et al. Transport of paclitaxel (Taxol) across the blood-brain barrier in vitro and in vivo. J. Clin. Investig. 2002, 110, 1309–1318. [Google Scholar] [CrossRef]
  319. Doan, P.; Nguyen, P.; Murugesan, A.; Subramanian, K.; Konda Mani, S.; Kalimuthu, V.; Abraham, B.G.; Stringer, B.W.; Balamuthu, K.; Yli-Harja, O.; et al. Targeting Orphan G Protein-Coupled Receptor 17 with T0 Ligand Impairs Glioblastoma Growth. Cancers 2021, 13, 3773. [Google Scholar] [CrossRef]
  320. Nguyen, P.; Doan, P.; Rimpilainen, T.; Konda Mani, S.; Murugesan, A.; Yli-Harja, O.; Candeias, N.R.; Kandhavelu, M. Synthesis and Preclinical Validation of Novel Indole Derivatives as a GPR17 Agonist for Glioblastoma Treatment. J. Med. Chem. 2021, 64, 10908–10918. [Google Scholar] [CrossRef]
  321. Hu, Y.; Luo, H.; Zhu, X.; Guo, H. CRNDE/ETS1/GPR17 Facilitates the Proliferation, Migration, and Invasion of Glioma. Comput. Math. Methods Med. 2021, 2021, 7566365. [Google Scholar] [CrossRef] [PubMed]
  322. Terpinskaya, T.I.; Osipov, A.V.; Kryukova, E.V.; Kudryavtsev, D.S.; Kopylova, N.V.; Yanchanka, T.L.; Palukoshka, A.F.; Gondarenko, E.A.; Zhmak, M.N.; Tsetlin, V.I.; et al. α-Conotoxins and α-Cobratoxin Promote, while Lipoxygenase and Cyclooxygenase Inhibitors Suppress the Proliferation of Glioma C6 Cells. Mar. Drugs 2021, 19, 118. [Google Scholar] [CrossRef]
  323. Ding, X.Z.; Tong, W.G.; Adrian, T.E. 12-lipoxygenase metabolite 12(S)-HETE stimulates human pancreatic cancer cell proliferation via protein tyrosine phosphorylation and ERK activation. Int. J. Cancer 2001, 94, 630–636. [Google Scholar] [CrossRef]
  324. Guo, A.M.; Liu, X.; Al-Wahab, Z.; Maddippati, K.R.; Ali-Fehmi, R.; Scicli, A.G.; Munkarah, A.R. Role of 12-lipoxygenase in regulation of ovarian cancer cell proliferation and survival. Cancer Chemother. Pharmacol. 2011, 68, 1273–1283. [Google Scholar] [CrossRef] [PubMed]
  325. Higuchi, Y.; Yoshimoto, T. Arachidonic acid converts the glutathione depletion-induced apoptosis to necrosis by promoting lipid peroxidation and reducing caspase-3 activity in rat glioma cells. Arch. Biochem. Biophys. 2002, 400, 133–140. [Google Scholar] [CrossRef] [PubMed]
  326. Strakova, N.; Ehrmann, J.; Dzubak, P.; Bouchal, J.; Kolar, Z. The synthetic ligand of peroxisome proliferator-activated receptor-gamma ciglitazone affects human glioblastoma cell lines. J. Pharmacol. Exp. Ther. 2004, 309, 1239–1247. [Google Scholar] [CrossRef]
  327. Grommes, C.; Landreth, G.E.; Sastre, M.; Beck, M.; Feinstein, D.L.; Jacobs, A.H.; Schlegel, U.; Heneka, M.T. Inhibition of in vivo glioma growth and invasion by peroxisome proliferator-activated receptor gamma agonist treatment. Mol. Pharmacol. 2006, 70, 1524–1533. [Google Scholar] [CrossRef] [PubMed]
  328. Liu, Y.; Shi, L.; Liu, Y.; Li, P.; Jiang, G.; Gao, X.; Zhang, Y.; Jiang, C.; Zhu, W.; Han, H.; et al. Activation of PPARγ mediates icaritin-induced cell cycle arrest and apoptosis in glioblastoma multiforme. Biomed. Pharmacother. 2018, 100, 358–366. [Google Scholar] [CrossRef] [PubMed]
  329. Viita, H.; Pacholska, A.; Ahmad, F.; Tietäväinen, J.; Naarala, J.; Hyvärinen, A.; Wirth, T.; Ylä-Herttuala, S. 15-Lipoxygenase-1 induces lipid peroxidation and apoptosis, and improves survival in rat malignant glioma. In Vivo 2012, 26, 1–8. [Google Scholar]
  330. Yuan, H.; Li, M.Y.; Ma, L.T.; Hsin, M.K.; Mok, T.S.; Underwood, M.J.; Chen, G.G. 15-Lipoxygenases and its metabolites 15(S)-HETE and 13(S)-HODE in the development of non-small cell lung cancer. Thorax 2010, 65, 321–326. [Google Scholar] [CrossRef] [Green Version]
  331. Liu, L.; Li, X.; Shi, J.; Li, L.; Wang, J.; Luo, Z.Z. Effects of FPR2 gene silencing on the proliferation, migration and invasion of human glioma U87 cells. Zhonghua Zhong Liu Za Zhi 2018, 40, 659–666. [Google Scholar] [PubMed]
  332. He, H.Q.; Ye, R.D. The Formyl Peptide Receptors: Diversity of Ligands and Mechanism for Recognition. Molecules 2017, 22, 455. [Google Scholar] [CrossRef] [PubMed]
  333. Johnson, A.L.; Edson, K.Z.; Totah, R.A.; Rettie, A.E. Cytochrome P450 ω-Hydroxylases in Inflammation and Cancer. Adv. Pharmacol. 2015, 74, 223–262. [Google Scholar]
  334. Rifkind, A.B.; Lee, C.; Chang, T.K.; Waxman, D.J. Arachidonic acid metabolism by human cytochrome P450s 2C8, 2C9, 2E1, and 1A2: Regioselective oxygenation and evidence for a role for CYP2C enzymes in arachidonic acid epoxygenation in human liver microsomes. Arch. Biochem. Biophys. 1995, 320, 380–389. [Google Scholar] [CrossRef] [PubMed]
  335. Choudhary, D.; Jansson, I.; Stoilov, I.; Sarfarazi, M.; Schenkman, J.B. Metabolism of retinoids and arachidonic acid by human and mouse cytochrome P450 1b1. Drug Metab. Dispos. 2004, 32, 840–847. [Google Scholar] [CrossRef] [PubMed]
  336. Chuang, S.S.; Helvig, C.; Taimi, M.; Ramshaw, H.A.; Collop, A.H.; Amad, M.; White, J.A.; Petkovich, M.; Jones, G.; Korczak, B. CYP2U1, a novel human thymus- and brain-specific cytochrome P450, catalyzes omega- and (omega-1)-hydroxylation of fatty acids. J. Biol. Chem. 2004, 279, 6305–6314. [Google Scholar] [CrossRef]
  337. Lasker, J.M.; Chen, W.B.; Wolf, I.; Bloswick, B.P.; Wilson, P.D.; Powell, P.K. Formation of 20-hydroxyeicosatetraenoic acid, a vasoactive and natriuretic eicosanoid, in human kidney. Role of Cyp4F2 and Cyp4A11. J. Biol. Chem. 2000, 275, 4118–4126. [Google Scholar] [CrossRef]
  338. Gainer, J.V.; Bellamine, A.; Dawson, E.P.; Womble, K.E.; Grant, S.W.; Wang, Y.; Cupples, L.A.; Guo, C.Y.; Demissie, S.; O’Donnell, C.J.; et al. Functional variant of CYP4A11 20-hydroxyeicosatetraenoic acid synthase is associated with essential hypertension. Circulation 2005, 111, 63–69. [Google Scholar] [CrossRef]
  339. Fer, M.; Corcos, L.; Dréano, Y.; Plée-Gautier, E.; Salaün, J.P.; Berthou, F.; Amet, Y. Cytochromes P450 from family 4 are the main omega hydroxylating enzymes in humans: CYP4F3B is the prominent player in PUFA metabolism. J. Lipid Res. 2008, 49, 2379–2389. [Google Scholar] [CrossRef]
  340. Bylund, J.; Ericsson, J.; Oliw, E.H. Analysis of cytochrome P450 metabolites of arachidonic and linoleic acids by liquid chromatography-mass spectrometry with ion trap MS. Anal. Biochem. 1998, 265, 55–68. [Google Scholar] [CrossRef]
  341. Brash, A.R.; Boeglin, W.E.; Capdevila, J.H.; Yeola, S.; Blair, I.A. 7-HETE, 10-HETE, and 13-HETE are major products of NADPH-dependent arachidonic acid metabolism in rat liver microsomes: Analysis of their stereochemistry, and the stereochemistry of their acid-catalyzed rearrangement. Arch. Biochem. Biophys. 1995, 321, 485–492. [Google Scholar] [CrossRef] [PubMed]
  342. Carroll, M.A.; Balazy, M.; Margiotta, P.; Huang, D.D.; Falck, J.R.; McGiff, J.C. Cytochrome P-450-dependent HETEs: Profile of biological activity and stimulation by vasoactive peptides. Am. J. Physiol. 1996, 271, R863–R869. [Google Scholar] [CrossRef] [PubMed]
  343. Bylund, J.; Kunz, T.; Valmsen, K.; Oliw, E.H. Cytochromes P450 with bisallylic hydroxylation activity on arachidonic and linoleic acids studied with human recombinant enzymes and with human and rat liver microsomes. J. Pharmacol. Exp. Ther. 1998, 284, 51–60. [Google Scholar]
  344. Bednar, M.M.; Gross, C.E.; Russell, S.R.; Fuller, S.P.; Ahern, T.P.; Howard, D.B.; Falck, J.R.; Reddy, K.M.; Balazy, M. 16(R)-hydroxyeicosatetraenoic acid, a novel cytochrome P450 product of arachidonic acid, suppresses activation of human polymorphonuclear leukocyte and reduces intracranial pressure in a rabbit model of thromboembolic stroke. Neurosurgery 2000, 47, 1410–1419. [Google Scholar] [CrossRef]
  345. Garcia, V.; Gilani, A.; Shkolnik, B.; Pandey, V.; Zhang, F.F.; Dakarapu, R.; Gandham, S.K.; Reddy, N.R.; Graves, J.P.; Gruzdev, A.; et al. 20-HETE Signals Through G-Protein-Coupled Receptor GPR75 (Gq) to Affect Vascular Function and Trigger Hypertension. Circ. Res. 2017, 120, 1776–1788. [Google Scholar] [CrossRef]
  346. Wen, H.; Östman, J.; Bubb, K.J.; Panayiotou, C.; Priestley, J.V.; Baker, M.D.; Ahluwalia, A. 20-Hydroxyeicosatetraenoic acid (20-HETE) is a novel activator of transient receptor potential vanilloid 1 (TRPV1) channel. J. Biol. Chem. 2012, 287, 13868–13876. [Google Scholar] [CrossRef]
  347. Tunaru, S.; Bonnavion, R.; Brandenburger, I.; Preussner, J.; Thomas, D.; Scholich, K.; Offermanns, S. 20-HETE promotes glucose-stimulated insulin secretion in an autocrine manner through FFAR1. Nat. Commun. 2018, 9, 177. [Google Scholar] [CrossRef]
  348. Ng, V.Y.; Huang, Y.; Reddy, L.M.; Falck, J.R.; Lin, E.T.; Kroetz, D.L. Cytochrome P450 eicosanoids are activators of peroxisome proliferator-activated receptor alpha. Drug Metab. Dispos. 2007, 35, 1126–1134. [Google Scholar] [CrossRef]
  349. Dai, D.; Zeldin, D.C.; Blaisdell, J.A.; Chanas, B.; Coulter, S.J.; Ghanayem, B.I.; Goldstein, J.A. Polymorphisms in human CYP2C8 decrease metabolism of the anticancer drug paclitaxel and arachidonic acid. Pharmacogenetics 2001, 11, 597–607. [Google Scholar] [CrossRef]
  350. Kato, Y.; Mukai, Y.; Rane, A.; Inotsume, N.; Toda, T. The Inhibitory Effect of Telmisartan on the Metabolism of Arachidonic Acid by CYP2C9 and CYP2C8: An in Vitro Study. Biol. Pharm. Bull. 2017, 40, 1409–1415. [Google Scholar] [CrossRef]
  351. Wu, S.; Moomaw, C.R.; Tomer, K.B.; Falck, J.R.; Zeldin, D.C. Molecular cloning and expression of CYP2J2, a human cytochrome P450 arachidonic acid epoxygenase highly expressed in heart. J. Biol. Chem. 1996, 271, 3460–3468. [Google Scholar] [CrossRef] [PubMed]
  352. Stark, K.; Dostalek, M.; Guengerich, F.P. Expression and purification of orphan cytochrome P450 4X1 and oxidation of anandamide. FEBS J. 2008, 275, 3706–3717. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  353. Park, S.K.; Herrnreiter, A.; Pfister, S.L.; Gauthier, K.M.; Falck, B.A.; Falck, J.R.; Campbell, W.B. GPR40 is a low-affinity epoxyeicosatrienoic acid receptor in vascular cells. J. Biol. Chem. 2018, 293, 10675–10691. [Google Scholar] [CrossRef]
  354. Yang, C.; Kwan, Y.W.; Au, A.L.; Poon, C.C.; Zhang, Q.; Chan, S.W.; Lee, S.M.; Leung, G.P. 14,15-Epoxyeicosatrienoic acid induces vasorelaxation through the prostaglandin EP2 receptors in rat mesenteric artery. Prostaglandins Other Lipid Mediat. 2010, 93, 44–51. [Google Scholar] [CrossRef]
  355. Liu, X.; Qian, Z.Y.; Xie, F.; Fan, W.; Nelson, J.W.; Xiao, X.; Kaul, S.; Barnes, A.P.; Alkayed, N.J. Functional screening for G protein-coupled receptor targets of 14,15-epoxyeicosatrienoic acid. Prostaglandins Other Lipid Mediat. 2017, 132, 31–40. [Google Scholar] [CrossRef]
  356. Liu, Y.; Zhang, Y.; Schmelzer, K.; Lee, T.S.; Fang, X.; Zhu, Y.; Spector, A.A.; Gill, S.; Morisseau, C.; Hammock, B.D.; et al. The antiinflammatory effect of laminar flow: The role of PPARgamma, epoxyeicosatrienoic acids, and soluble epoxide hydrolase. Proc. Natl. Acad. Sci. USA 2005, 102, 16747–16752. [Google Scholar] [CrossRef]
  357. Spector, A.A.; Norris, A.W. Action of epoxyeicosatrienoic acids on cellular function. Am. J. Physiol. Cell Physiol. 2007, 292, C996–C1012. [Google Scholar] [CrossRef] [PubMed]
  358. Karara, A.; Dishman, E.; Falck, J.R.; Capdevila, J.H. Endogenous epoxyeicosatrienoyl-phospholipids. A novel class of cellular glycerolipids containing epoxidized arachidonate moieties. J. Biol. Chem. 1991, 266, 7561–7569. [Google Scholar] [CrossRef]
  359. Somani, S.T.; Zeigler, M.; Fay, E.E.; Leahy, M.; Bermudez, B.; Totah, R.A.; Hebert, M.F. Changes in erythrocyte membrane epoxyeicosatrienoic, dihydroxyeicosatrienoic, and hydroxyeicosatetraenoic acids during pregnancy. Life Sci. 2021, 264, 118590. [Google Scholar] [CrossRef]
  360. Edin, M.L.; Hamedani, B.G.; Gruzdev, A.; Graves, J.P.; Lih, F.B.; Arbes, S.J., 3rd; Singh, R.; Orjuela Leon, A.C.; Bradbury, J.A.; DeGraff, L.M.; et al. Epoxide hydrolase 1 (EPHX1) hydrolyzes epoxyeicosanoids and impairs cardiac recovery after ischemia. J. Biol. Chem. 2018, 293, 3281–3292. [Google Scholar] [CrossRef]
  361. Fang, X.; Hu, S.; Xu, B.; Snyder, G.D.; Harmon, S.; Yao, J.; Liu, Y.; Sangras, B.; Falck, J.R.; Weintraub, N.L.; et al. 14,15-Dihydroxyeicosatrienoic acid activates peroxisome proliferator-activated receptor-alpha. Am. J. Physiol. Heart Circ. Physiol. 2006, 290, H55–H63. [Google Scholar] [CrossRef]
  362. Le Quéré, V.; Plée-Gautier, E.; Potin, P.; Madec, S.; Salaün, J.P. Human CYP4F3s are the main catalysts in the oxidation of fatty acid epoxides. J. Lipid Res. 2004, 45, 1446–1458. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  363. Moreland, K.T.; Procknow, J.D.; Sprague, R.S.; Iverson, J.L.; Lonigro, A.J.; Stephenson, A.H. Cyclooxygenase (COX)-1 and COX-2 participate in 5,6-epoxyeicosatrienoic acid-induced contraction of rabbit intralobar pulmonary arteries. J. Pharmacol. Exp. Ther. 2007, 321, 446–454. [Google Scholar] [CrossRef] [PubMed]
  364. Rand, A.A.; Barnych, B.; Morisseau, C.; Cajka, T.; Lee, K.S.S.; Panigrahy, D.; Hammock, B.D. Cyclooxygenase-derived proangiogenic metabolites of epoxyeicosatrienoic acids. Proc. Natl. Acad. Sci. USA 2017, 114, 4370–4375. [Google Scholar] [CrossRef] [PubMed]
  365. Barnych, B.; Rand, A.A.; Cajka, T.; Lee, K.S.S.; Hammock, B.D. Synthesis of cyclooxygenase metabolites of 8,9-epoxyeicosatrienoic acid (EET): 11- and 15-hydroxy 8,9-EETs. Org. Biomol. Chem. 2017, 15, 4308–4313. [Google Scholar] [CrossRef] [PubMed]
  366. Rand, A.A.; Rajamani, A.; Kodani, S.D.; Harris, T.R.; Schlatt, L.; Barnych, B.; Passerini, A.G.; Hammock, B.D. Epoxyeicosatrienoic acid (EET)-stimulated angiogenesis is mediated by epoxy hydroxyeicosatrienoic acids (EHETs) formed from COX-2. J. Lipid Res. 2019, 60, 1996–2005. [Google Scholar] [CrossRef]
  367. Hildreth, K.; Kodani, S.D.; Hammock, B.D.; Zhao, L. Cytochrome P450-derived linoleic acid metabolites EpOMEs and DiHOMEs: A review of recent studies. J. Nutr. Biochem. 2020, 86, 108484. [Google Scholar] [CrossRef]
  368. Zanger, U.M.; Schwab, M. Cytochrome P450 enzymes in drug metabolism: Regulation of gene expression, enzyme activities, and impact of genetic variation. Pharmacol. Ther. 2013, 138, 103–141. [Google Scholar] [CrossRef]
  369. Chen, P.; Guo, M.; Wygle, D.; Edwards, P.A.; Falck, J.R.; Roman, R.J.; Scicli, A.G. Inhibitors of cytochrome P450 4A suppress angiogenic responses. Am. J. Pathol. 2005, 166, 615–624. [Google Scholar] [CrossRef]
  370. Guo, A.M.; Sheng, J.; Scicli, G.M.; Arbab, A.S.; Lehman, N.L.; Edwards, P.A.; Falck, J.R.; Roman, R.J.; Scicli, A.G. Expression of CYP4A1 in U251 human glioma cell induces hyperproliferative phenotype in vitro and rapidly growing tumors in vivo. J. Pharmacol. Exp. Ther. 2008, 327, 10–19. [Google Scholar] [CrossRef]
  371. Angara, K.; Rashid, M.H.; Shankar, A.; Ara, R.; Iskander, A.; Borin, T.F.; Jain, M.; Achyut, B.R.; Arbab, A.S. Vascular mimicry in glioblastoma following anti-angiogenic and anti-20-HETE therapies. Histol. Histopathol. 2017, 32, 917–928. [Google Scholar] [PubMed]
  372. Guo, M.; Roman, R.J.; Falck, J.R.; Edwards, P.A.; Scicli, A.G. Human U251 glioma cell proliferation is suppressed by HET0016 [N-hydroxy-N′-(4-butyl-2-methylphenyl)formamidine], a selective inhibitor of CYP4A. J. Pharmacol. Exp. Ther. 2005, 315, 526–533. [Google Scholar] [CrossRef]
  373. Wang, C.; Li, Y.; Chen, H.; Zhang, J.; Zhang, J.; Qin, T.; Duan, C.; Chen, X.; Liu, Y.; Zhou, X.; et al. Inhibition of CYP4A by a novel flavonoid FLA-16 prolongs survival and normalizes tumor vasculature in glioma. Cancer Lett. 2017, 402, 131–141. [Google Scholar] [CrossRef] [PubMed]
  374. Yu, X.; Wu, J.; Hu, M.; Wu, J.; Zhu, Q.; Yang, Z.; Xie, X.; Feng, Y.Q.; Yue, J. Glutamate affects the CYP1B1- and CYP2U1-mediated hydroxylation of arachidonic acid metabolism via astrocytic mGlu5 receptor. Int. J. Biochem. Cell Biol. 2019, 110, 111–121. [Google Scholar] [CrossRef] [PubMed]
  375. Zagorac, D.; Jakovcevic, D.; Gebremedhin, D.; Harder, D.R. Antiangiogenic effect of inhibitors of cytochrome P450 on rats with glioblastoma multiforme. J. Cereb. Blood Flow Metab. 2008, 28, 1431–1439. [Google Scholar] [CrossRef] [PubMed]
  376. Takano, T.; Lin, J.H.; Arcuino, G.; Gao, Q.; Yang, J.; Nedergaard, M. Glutamate release promotes growth of malignant gliomas. Nat. Med. 2001, 7, 1010–1015. [Google Scholar] [CrossRef]
Figure 1. ARA biosynthesis. ARA C20:4n-6 in humans is not synthesized de novo but from linoleic acid C18:2n-6. As linoleoyl-CoA C18:2n-6, this PUFA undergoes desaturation to γ-linolenoyl-CoA C18:3n-6 with FADS2/D6D. This fatty acyl-CoA is then converted to dihomo-γ-linolenoyl-CoA C20:3n-6 with ELOVL5 and, finally, to arachidonyl-CoA C20:4n-6 with FADS1/D5D. Dihomo-γ-linolenoyl-CoA C20:3n-6 can also be formed from linoleoyl-CoA via an alternative pathway. Linoleoyl-CoA C18:2n-6 first undergoes elongation with ELOVL5 and then desaturation with FADS2. The latter enzyme in this pathway exhibits Δ8-desaturase activity. —higher expression of given enzymes in GBM tumor relative to healthy tissue.
Figure 1. ARA biosynthesis. ARA C20:4n-6 in humans is not synthesized de novo but from linoleic acid C18:2n-6. As linoleoyl-CoA C18:2n-6, this PUFA undergoes desaturation to γ-linolenoyl-CoA C18:3n-6 with FADS2/D6D. This fatty acyl-CoA is then converted to dihomo-γ-linolenoyl-CoA C20:3n-6 with ELOVL5 and, finally, to arachidonyl-CoA C20:4n-6 with FADS1/D5D. Dihomo-γ-linolenoyl-CoA C20:3n-6 can also be formed from linoleoyl-CoA via an alternative pathway. Linoleoyl-CoA C18:2n-6 first undergoes elongation with ELOVL5 and then desaturation with FADS2. The latter enzyme in this pathway exhibits Δ8-desaturase activity. —higher expression of given enzymes in GBM tumor relative to healthy tissue.
Cancers 15 00946 g001
Figure 2. Importance of PLA2 in metabolism of ARA and production of lipids mediators from ARA. ARA C20:4n-6 is cleaved from PC by PLA2. This reaction also produces LPC, which can be converted in the intercellular space to LPA by ATX. LPA can be considered a lipid mediator because its biological activity is related to the activation of its specific receptors: LPAR1-LPAR6. Free ARA C20:4n-6, on the other hand, can be used for eicosanoid production in either the COX pathway or the LOX pathway. ↑—higher expression of given enzymes in GBM tumor relative to healthy tissue; ↓—lower expression of given enzymes in GBM tumor relative to healthy tissue.
Figure 2. Importance of PLA2 in metabolism of ARA and production of lipids mediators from ARA. ARA C20:4n-6 is cleaved from PC by PLA2. This reaction also produces LPC, which can be converted in the intercellular space to LPA by ATX. LPA can be considered a lipid mediator because its biological activity is related to the activation of its specific receptors: LPAR1-LPAR6. Free ARA C20:4n-6, on the other hand, can be used for eicosanoid production in either the COX pathway or the LOX pathway. ↑—higher expression of given enzymes in GBM tumor relative to healthy tissue; ↓—lower expression of given enzymes in GBM tumor relative to healthy tissue.
Cancers 15 00946 g002
Figure 3. COX pathway. After release by PLA2, ARA C20:4n-6 is converted into prostanoids with COX. It is transformed into PGH2 with either COX-1 or COX-2. Then, this prostaglandin is transformed into other prostaglandins (PGE2, PGD2, PGI2, and PGF) or TxA2 by the respective synthases. These lipid mediators undergo further transformations. TxA2 is unstable and undergoes a spontaneous transformation into TxB2. Similarly, PGD2 undergoes spontaneous transformation to PGJ2—this prostaglandin can then be transformed into 15-deoxy-Δ12,14-PGJ2 (15d-PGJ2) or Δ12-PGJ2. PGE2 can be transformed into PGA2, and then into PGC2 and PGB2. Prostanoids also undergo degradation. The figure shows an example of PGE2, which undergoes inactivation by oxidation with 15-PGDH and reduction with PTGR1/2. PGE2 can also undergo degradation by β-oxidation and ω-oxidation, followed by the action of 15-PGDH and PTGR1/2. The resulting degradation product is PGE2, which is removed from the body. ↑—higher expression of given enzymes in GBM tumor relative to healthy tissue; ↓—lower expression of given enzymes in GBM tumor relative to healthy tissue.
Figure 3. COX pathway. After release by PLA2, ARA C20:4n-6 is converted into prostanoids with COX. It is transformed into PGH2 with either COX-1 or COX-2. Then, this prostaglandin is transformed into other prostaglandins (PGE2, PGD2, PGI2, and PGF) or TxA2 by the respective synthases. These lipid mediators undergo further transformations. TxA2 is unstable and undergoes a spontaneous transformation into TxB2. Similarly, PGD2 undergoes spontaneous transformation to PGJ2—this prostaglandin can then be transformed into 15-deoxy-Δ12,14-PGJ2 (15d-PGJ2) or Δ12-PGJ2. PGE2 can be transformed into PGA2, and then into PGC2 and PGB2. Prostanoids also undergo degradation. The figure shows an example of PGE2, which undergoes inactivation by oxidation with 15-PGDH and reduction with PTGR1/2. PGE2 can also undergo degradation by β-oxidation and ω-oxidation, followed by the action of 15-PGDH and PTGR1/2. The resulting degradation product is PGE2, which is removed from the body. ↑—higher expression of given enzymes in GBM tumor relative to healthy tissue; ↓—lower expression of given enzymes in GBM tumor relative to healthy tissue.
Cancers 15 00946 g003
Figure 4. 5-LOX pathway. ARA C20:4n-6 is converted to 5-HpETE with 5-LOX. This enzyme also catalyzes the next step in leukotriene biosynthesis. It converts 5-HpETE into LTA4, which can then be converted into LTB4 with LTA4H, into LTC4 with LTC4S, or into 5-oxo-ETE. 5-HpETE can also be converted to 5-oxo-ETE. LTC4 can be converted to other cysteinyl leukotrienes. LTC4 can be converted to LTF4 with the involvement of carboxypeptidase A or to LTD4 with the involvement of GGT1 and GGT5. Subsequently, LTD4 can be converted into LTE4 with the participation of DPEP1 and DPEP2, and then converted into LTF4 with γ-glutamyltranspeptidase. ↑—higher expression of given enzymes in GBM tumor relative to healthy tissue.
Figure 4. 5-LOX pathway. ARA C20:4n-6 is converted to 5-HpETE with 5-LOX. This enzyme also catalyzes the next step in leukotriene biosynthesis. It converts 5-HpETE into LTA4, which can then be converted into LTB4 with LTA4H, into LTC4 with LTC4S, or into 5-oxo-ETE. 5-HpETE can also be converted to 5-oxo-ETE. LTC4 can be converted to other cysteinyl leukotrienes. LTC4 can be converted to LTF4 with the involvement of carboxypeptidase A or to LTD4 with the involvement of GGT1 and GGT5. Subsequently, LTD4 can be converted into LTE4 with the participation of DPEP1 and DPEP2, and then converted into LTF4 with γ-glutamyltranspeptidase. ↑—higher expression of given enzymes in GBM tumor relative to healthy tissue.
Cancers 15 00946 g004
Figure 5. 12-LOX pathway. ARA C20:4n-6 is converted to 12S-HpETE and 12R-HpETE with 12S-LOX and 12R-LOX, respectively. Either 12-oxo-ETE or the corresponding 12-HETE can be formed from these compounds. 12S-HpETE can also be converted to HxA3 or HxB3 with hemin and lipoxygenases: eLOX3, 12S-LOX, or 15-LOX-1. 12R-HpETE can undergo a similar conversion to 11,12-bis-epi-HxA3. HxA3 may undergo further transformations. HxA3 can be conjugated to glutathione. HxA3-C is then formed, from which amino acids can be detached—HxA3-D is then formed in a reaction similar to the transformation of cysteinyl-leukotrienes. HxA3 can also be converted to TrXA3. Arrows next to enzymes: higher or lower expression of given enzymes in GBM tumor relative to healthy tissue. ↓—lower expression of given enzymes in GBM tumor relative to healthy tissue.
Figure 5. 12-LOX pathway. ARA C20:4n-6 is converted to 12S-HpETE and 12R-HpETE with 12S-LOX and 12R-LOX, respectively. Either 12-oxo-ETE or the corresponding 12-HETE can be formed from these compounds. 12S-HpETE can also be converted to HxA3 or HxB3 with hemin and lipoxygenases: eLOX3, 12S-LOX, or 15-LOX-1. 12R-HpETE can undergo a similar conversion to 11,12-bis-epi-HxA3. HxA3 may undergo further transformations. HxA3 can be conjugated to glutathione. HxA3-C is then formed, from which amino acids can be detached—HxA3-D is then formed in a reaction similar to the transformation of cysteinyl-leukotrienes. HxA3 can also be converted to TrXA3. Arrows next to enzymes: higher or lower expression of given enzymes in GBM tumor relative to healthy tissue. ↓—lower expression of given enzymes in GBM tumor relative to healthy tissue.
Cancers 15 00946 g005
Figure 6. 15-LOX pathway. (A). Linoleic acid C18:2n-6 can be converted by 15-LOX-1 and 15-LOX-2 into 13-HpODE. This compound can then be converted into 13-HODE and 13-oxo-ODE. (B) 15-LOX-1 and 15-LOX-2 can convert ARA C20:4n-6 into 15-HpETE. 15-LOX-1 can also convert this fatty acid into 12-HpETE. 15-HpETE can then be converted into EXA4 and into cysteinyl-eoxins EXC4, EXD4, and EXE4. 15-HpETE can also be converted into hepoxilins 14,15-HxA3 11S, and 14,15-HxB3 13R. 14,15-HxA3 11S can be converted to cysteinyl hepoxilins, such as 14,15-HxA3-C 11S.
Figure 6. 15-LOX pathway. (A). Linoleic acid C18:2n-6 can be converted by 15-LOX-1 and 15-LOX-2 into 13-HpODE. This compound can then be converted into 13-HODE and 13-oxo-ODE. (B) 15-LOX-1 and 15-LOX-2 can convert ARA C20:4n-6 into 15-HpETE. 15-LOX-1 can also convert this fatty acid into 12-HpETE. 15-HpETE can then be converted into EXA4 and into cysteinyl-eoxins EXC4, EXD4, and EXE4. 15-HpETE can also be converted into hepoxilins 14,15-HxA3 11S, and 14,15-HxB3 13R. 14,15-HxA3 11S can be converted to cysteinyl hepoxilins, such as 14,15-HxA3-C 11S.
Cancers 15 00946 g006
Figure 7. Cytochrome P450 pathway. ARA 20:4n-6 can be converted in the cytochrome P450 pathway, resulting in the formation of various ETT and HETE. ETT can undergo further transformations where they are incorporated into glycerophospholipids in the sn-2 position; in this form, they build the cell membrane and intracellular membranes. In addition, the epoxide bond in ETT can be transformed by EPHX1 and EPHX2 into two hydroxyl groups, resulting in the formation of various DHET. ETT can also undergo ω-hydroxylation, which results in the formation of various HEET. ETT can be converted with COX. 5,6-EET then produces 5,6-epoxy-PGH2, whereas 8,9-EET produces either 8,9,11-EHET or 8,9,15-EHET. ↑—higher expression of given enzymes in GBM tumor relative to healthy tissue; ↓—lower expression of given enzymes in GBM tumor relative to healthy tissue.
Figure 7. Cytochrome P450 pathway. ARA 20:4n-6 can be converted in the cytochrome P450 pathway, resulting in the formation of various ETT and HETE. ETT can undergo further transformations where they are incorporated into glycerophospholipids in the sn-2 position; in this form, they build the cell membrane and intracellular membranes. In addition, the epoxide bond in ETT can be transformed by EPHX1 and EPHX2 into two hydroxyl groups, resulting in the formation of various DHET. ETT can also undergo ω-hydroxylation, which results in the formation of various HEET. ETT can be converted with COX. 5,6-EET then produces 5,6-epoxy-PGH2, whereas 8,9-EET produces either 8,9,11-EHET or 8,9,15-EHET. ↑—higher expression of given enzymes in GBM tumor relative to healthy tissue; ↓—lower expression of given enzymes in GBM tumor relative to healthy tissue.
Cancers 15 00946 g007
Table 1. Description of cPLA2 and iPLA2.
Table 1. Description of cPLA2 and iPLA2.
NameExpression Level in GBM Tumor Relative to Healthy TissueImpact on Prognosis with Higher Expression in GBM Tumors
SourceGEPIA [9]Seifert et al. [8]GEPIA [9]
cPLA2
cPLA2α/PLA2G4AHigher expression in the tumorHigher expression in the tumorNo significant impact on prognosis
cPLA2β/PLA2G4BLower expression in the tumorExpression does not changeNo significant impact on prognosis
cPLA2γ/PLA2G4CExpression does not changeLower expression in the tumorNo significant impact on prognosis
cPLA2δ/PLA2G4DExpression does not changeExpression does not changeNo significant impact on prognosis
cPLA2ε/PLA2G4EExpression does not changeExpression does not changeNo significant impact on prognosis
cPLA2ζ/PLA2G4FExpression does not changeExpression does not changeNo significant impact on prognosis
iPLA2
iPLA2β/PLA2G6Expression does not changeLower expression in the tumorNo significant impact on prognosis
iPLA2γ/PNPLA8Expression does not changeExpression does not changeNo significant impact on prognosis
iPLA2δ/PNPLA6Expression does not changeLower expression in the tumorNo significant impact on prognosis
iPLA2ε/PNPLA3Expression does not changeExpression does not changeNo significant impact on prognosis
iPLA2ζ/PNPLA2Expression does not changeExpression does not changeWorse prognosis p = 0.087
iPLA2η/PNPLA4Expression does not changeExpression does not changeWorse prognosis
Red background—higher expression in the tumor; blue background—lower expression in the tumor; red background—worse prognosis with higher expression of a given PLA2.
Table 2. Description of sPLA2 and sPLA2 receptors in GBM.
Table 2. Description of sPLA2 and sPLA2 receptors in GBM.
NameExpression Level in GBM Tumor Relative to Healthy TissueImpact on Prognosis with Higher Expression in GBM Tumors
SourceGEPIA [9]Seifert et al. [8]GEPIA [9]Wu et al. [52]
PLA2G1BExpression does not changeExpression does not changeWorse prognosis p = 0.078Worse prognosis
PLA2G2AHigher expression in the tumorHigher expression in the tumorNo significant impact on prognosisNo significant impact on prognosis
PLA2G2DExpression does not changeExpression does not changeNo significant impact on prognosisNo significant impact on prognosis
PLA2G2EExpression does not changeExpression does not changeN/AWorse prognosis
PLA2G2FExpression does not changeExpression does not changeN/ANo significant impact on prognosis
PLA2G3Expression does not changeExpression does not changeNo significant impact on prognosisWorse prognosis
PLA2G5Higher expression in the tumorHigher expression in the tumorNo significant impact on prognosisWorse prognosis
PLA2G7Expression does not changeExpression does not changeNo significant impact on prognosisNo significant impact on prognosis
PLA2G10Expression does not changeExpression does not changeN/ANo significant impact on prognosis
PLA2G12AHigher expression in the tumorExpression does not changeNo significant impact on prognosisNo significant impact on prognosis
PLA2G12BExpression does not changeExpression does not changeN/ANo significant impact on prognosis
PLA2G15Higher expression in the tumorExpression does not changeWorse prognosisNo significant impact on prognosis
PLA2G16Expression does not changeExpression does not changeNo significant impact on prognosisNo significant impact on prognosis
PLA2R1/PLA2R1Expression does not changeExpression does not changeWorse prognosis
Red background—higher expression in the tumor; red background—worse prognosis with higher expression of a given PLA2.
Table 3. Pan-cancer analysis of gene expression of cPLA2 and iPLA2.
Table 3. Pan-cancer analysis of gene expression of cPLA2 and iPLA2.
Name of CancercPLA2α/PLA2G4AcPLA2β/PLA2G4BcPLA2γ/PLA2G4CcPLA2δ/PLA2G4DcPLA2ε/PLA2G4EcPLA2ζ/PLA2G4FiPLA2β/PLA2G6iPLA2γ/PNPLA8iPLA2δ/PNPLA6iPLA2ε/PNPLA3iPLA2ζ/PNPLA2iPLA2η/PNPLA4
Adrenocortical carcinoma (ACC)=========
Bladder urothelial carcinoma (BLCA)==========
Breast invasive carcinoma (BRCA)========
Cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC)========
Cholangiocarcinoma (CHOL)======
Colon adenocarcinoma (COAD)======
Lymphoid neoplasm diffuse large B-cell lymphoma (DLBC)=========
Esophageal carcinoma (ESCA)==========
Glioblastoma multiforme (GBM)==========
Head and neck squamous cell carcinoma (HNSC)===========
Kidney chromophobe (KICH)==========
Kidney renal clear cell carcinoma (KIRC)==========
Kidney renal papillary cell carcinoma (KIRP)===========
Acute myeloid leukemia (LAML)=====
Brain lower grade glioma (LGG)==========
Liver hepatocellular carcinoma (LIHC)==========
Lung adenocarcinoma (LUAD)=====
Lung squamous cell carcinoma (LUSC)=====
Ovarian serous cystadenocarcinoma (OV)==========
Pancreatic adenocarcinoma (PAAD)========
Pheochromocytoma and paraganglioma (PCPG)==========
Prostate adenocarcinoma (PRAD)==========
Rectum adenocarcinoma (READ)======
Sarcoma (SARC)===========
Skin cutaneous melanoma (SKCM)====
Stomach adenocarcinoma (STAD)=========
Testicular germ cell tumors (TGCT)=====
Thyroid carcinoma (THCA)========
Thymoma (THYM)=======
Uterine corpus endometrial carcinoma (UCEC)=========
Uterine carcinosarcoma (UCS)=======
Red background, ↑—expression higher in tumors than in healthy tissue; blue background, ↓—expression lower in tumors than in healthy tissue; gray background, =—expression does not differ between tumors and healthy tissue.
Table 4. Pan-cancer analysis of gene expression of sPLA2 and sPLA2 receptors.
Table 4. Pan-cancer analysis of gene expression of sPLA2 and sPLA2 receptors.
Name of CancerPLA2G1BPLA2G2APLA2G2DPLA2G2EPLA2G2FPLA2G3PLA2G5PLA2G7PLA2G10PLA2G12APLA2G12BPLA2G15PLA2G16PLA2R1
Adrenocortical carcinoma (ACC)============
Bladder urothelial carcinoma (BLCA)==========
Breast invasive carcinoma (BRCA)==========
Cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC)==========
Cholangiocarcinoma (CHOL)============
Colon adenocarcinoma (COAD)==========
Lymphoid neoplasm diffuse large B-cell lymphoma (DLBC)=========
Esophageal carcinoma (ESCA)=======
Glioblastoma multiforme (GBM)==========
Head and neck squamous cell carcinoma (HNSC)===========
Kidney chromophobe (KICH)=============
Kidney renal clear cell carcinoma (KIRC)===========
Kidney renal papillary cell carcinoma (KIRP)=========
Acute myeloid leukemia (LAML)============
Brain lower grade glioma (LGG)==============
Liver hepatocellular carcinoma (LIHC)=============
Lung adenocarcinoma (LUAD)=========
Lung squamous cell carcinoma (LUSC)========
Ovarian serous cystadenocarcinoma (OV)=========
Pancreatic adenocarcinoma (PAAD)======
Pheochromocytoma and paraganglioma (PCPG)============
Prostate adenocarcinoma (PRAD)===========
Rectum adenocarcinoma (READ)==========
Sarcoma (SARC)============
Skin cutaneous melanoma (SKCM)=======
Stomach adenocarcinoma (STAD)==========
Testicular germ cell tumors (TGCT)=======
Thyroid carcinoma (THCA)==========
Thymoma (THYM)=========
Uterine corpus endometrial carcinoma (UCEC)=======
Uterine carcinosarcoma (UCS)=========
Red background, ↑—expression higher in tumor than in healthy tissue; blue background, ↓—expression lower in tumor than in healthy tissue; gray background, =—expression does not differ between tumor and healthy tissue.
Table 5. Description of individual enzymes involved in the synthesis, transport, and degradation of prostaglandins.
Table 5. Description of individual enzymes involved in the synthesis, transport, and degradation of prostaglandins.
NameBiochemical SignificanceExpression Level in GBM Tumor Relative to Healthy TissueImpact on Prognosis with Higher Expression in GBM Tumors
Source GEPIA [9]Seifert et al. [8]Other Data SourceGEPIA [9]Other Data Source
COX-1PGH2 synthesis from ARAHigher expression in the tumorHigher expression in the tumorHigher expression in the tumor [141]No significant impact on prognosisNo significant impact on prognosis [121]
COX-2PGH2 synthesis from ARAExpression does not changeExpression does not changeHigher expression in the tumor [141,142]No significant impact on prognosisWorse prognosis [160,187,188]
mPGES-1PGE2 synthesis from PGH2Expression does not changeExpression does not changeHigher expression in the tumor [143]Worse prognosisWorse prognosis [121]
mPGES-2PGE2 synthesis from PGH2Expression does not changeExpression does not changeHigher expression in the tumor [143]No significant impact on prognosisNo significant impact on prognosis [121]
cPGESPGE2 synthesis from PGH2Higher expression in the tumorExpression does not changeHigher expression in the tumor [143]No significant impact on prognosis
H-PGDSSynthesis of PGD2 from PGH2Higher expression in the tumorLower expression in the tumor No significant impact on prognosis
L-PGDSSynthesis of PGD2 from PGH2Lower expression in the tumorLower expression in the tumor No significant impact on prognosis
TBXAS1TxA2 synthesis from PGH2Higher expression in the tumorHigher expression in the tumorHigher expression in the tumor [144]No significant impact on prognosis
AKR1B1PGF synthesis from PGH2Higher expression in the tumorHigher expression in the tumor Worse prognosis
AKR1C1PGF synthesis from PGE2Lower expression in the tumorLower expression in the tumor No significant impact on prognosis
AKR1C2PGF synthesis from PGE2Lower expression in the tumorExpression does not change No significant impact on prognosis
AKR1C3PGF synthesis from PGH2Expression does not changeExpression does not change No significant impact on prognosis
PGISPGIF2 synthesis from PGH2Expression does not changeExpression does not change No significant impact on prognosis
MRP4Secretion of prostaglandins from the cellHigher expression in the tumorExpression does not change No significant impact on prognosis
PGT/SLCO2A1Uptake of prostaglandins into the cellExpression does not changeExpression does not change No significant impact on prognosis
15-PGDHFirst degradation reaction/formation of PPARγ ligand from PGE2Expression does not changeExpression does not change No significant impact on prognosisBetter prognosis [121]
PTGR1Second degradation/inactivation reaction of PPARγ ligand made from PGE2Expression does not changeExpression does not change No significant impact on prognosisWorse prognosis [121]
PTGR2Second degradation/inactivation reaction of PPARγ ligand made from PGE2Expression does not changeExpression does not change No significant impact on prognosisNo significant impact on prognosis
Red background—higher expression in the tumor; blue background—lower expression in the tumor; red background—worse prognosis with higher expression; blue background—better prognosis with higher expression.
Table 6. Pan-cancer analysis of expression of genes involved in COX pathway.
Table 6. Pan-cancer analysis of expression of genes involved in COX pathway.
Name of CancerCOX-1/PTGS1COX-2/PTGS2mPGES-1/PTGESmPGES-2/PTGES2cPGES/PTGES3H-PGDS/HPGDSL-PGDS/PTGDSTBXAS1AKR1B1AKR1C1AKR1C2AKR1C3PGIS/PTGISMRP4/ABCC4PGT/SLCO2A115-PGDH/HPGDPTGR1PTGR2
Adrenocortical carcinoma (ACC)=========
Bladder urothelial carcinoma (BLCA)=========
Breast invasive carcinoma (BRCA)===========
Cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC)==============
Cholangiocarcinoma (CHOL)========
Colon adenocarcinoma (COAD)=======
Lymphoid neoplasm diffuse large B-cell lymphoma (DLBC)=======
Esophageal carcinoma (ESCA)========
Glioblastoma multiforme (GBM)=========
Head and neck squamous cell carcinoma (HNSC)===============
Kidney chromophobe (KICH)============
Kidney renal clear cell carcinoma (KIRC)============
Kidney renal papillary cell carcinoma (KIRP)==========
Acute myeloid leukemia (LAML)=====
Brain lower grade glioma (LGG)============
Liver hepatocellular carcinoma (LIHC)============
Lung adenocarcinoma (LUAD)=============
Lung squamous cell carcinoma (LUSC)========
Ovarian serous cystadenocarcinoma (OV)===========
Pancreatic adenocarcinoma (PAAD)=
Pheochromocytoma and paraganglioma (PCPG)==============
Prostate adenocarcinoma (PRAD)==========
Rectum adenocarcinoma (READ)=====
Sarcoma (SARC)================
Skin cutaneous melanoma (SKCM)========
Stomach adenocarcinoma (STAD)===============
Testicular germ cell tumors (TGCT)=========
Thyroid carcinoma (THCA)============
Thymoma (THYM)=====
Uterine corpus endometrial carcinoma (UCEC)===========
Uterine carcinosarcoma (UCS)=============
Red background, ↑—expression higher in tumor than in healthy tissue; blue background, ↓—expression lower in tumor than in healthy tissue; gray background, =—expression does not differ between tumor and healthy tissue.
Table 7. Description of the various enzymes involved in the synthesis, action, and degradation of lipoxygenases along with their involvement in tumorigenesis in GBM.
Table 7. Description of the various enzymes involved in the synthesis, action, and degradation of lipoxygenases along with their involvement in tumorigenesis in GBM.
NameBiochemical SignificanceExpression Level In GBM Tumors Relative To Healthy Tissue Impact on Prognosis with Higher Expression in GBM Tumors
Source GEPIA [9]Seifert et al. [8]GEPIA [9]
eLOX3/ALOXE3Production of hepoxilins/hydroxy-epoxyeicosatrienoic acid and oxo-ETE from HpETELower expression in the tumorExpression does not changeNo significant impact on prognosis
5-LOX/ALOX55-HpETE production from ARA; the first enzyme in leukotrienes and the 5-oxo-ETE synthesis pathway; synthesis of lipoxins from 15-HpETE and 15-HETEHigher expression in the tumorHigher expression in the tumorNo significant impact on prognosis
FLAP/ALOX5APSubstrate carrier for 5-LOXHigher expression in the tumorHigher expression in the tumorNo significant impact on prognosis
12S-LOX/ALOX1212S-HpETE production from ARA; the first enzyme in the hepoxilin production pathway; production of lipoxins from LTA4Expression does not changeExpression does not changeNo significant impact on prognosis
12R-LOX/ALOX12B12R-HpETE production from ARAExpression does not changeExpression does not changeNo significant impact on prognosis
15-LOX-1/ALOX1515-HpETE production from ARA; 12-HpETE production from ARA; production of lipoxins, eoxins, 15-oxo-ETE and 15-HETE; production of 13-HpODE from linoleic acid C18:2n-6Expression does not changeExpression does not changeNo significant impact on prognosis
15-LOX-2/ALOX15B15-HpETE production from ARA; production of 15-HpETE, lipoxins, eoxins, 15-oxo-ETE and 15-HETEExpression does not changeExpression does not changeNo significant impact on prognosis
LTA4HLTB4 production from LTA4Higher expression in the tumorHigher expression in the tumorNo significant impact on prognosis
LTC4SLTC4 production from LTA4Higher expression in the tumorExpression does not changeNo significant impact on prognosis
GGT1LTD4 production from LTC4Expression does not changeExpression does not changeWorse prognosis
GGT5LTD4 production from LTC4Higher expression in the tumorHigher expression in the tumorWorse prognosis (p = 0.055)
DPEP1LTE4 production from LTD4Higher expression in the tumorExpression does not changeNo significant impact on prognosis
DPEP2LTE4 production from LTD4Expression does not changeExpression does not changeNo significant impact on prognosis
EPHX2Conversion of hepoxilins into trioxilinExpression does not changeExpression does not changeWorse prognosis (p = 0.072)
Receptors
LTB4R1LTB4 receptorExpression does not changeExpression does not changeNo significant impact on prognosis
LTB4R2LTB4 receptorExpression does not changeExpression does not changeNo significant impact on prognosis
CYSLTR1Cysteinyl-leukotrienes receptorExpression does not changeExpression does not changeNo significant impact on prognosis
CYSLTR2Cysteinyl-leukotrienes receptorExpression does not changeExpression does not changeNo significant impact on prognosis
OXER15-oxo-ETE receptorExpression does not changeExpression does not changeWorse prognosis
ALX/FPR2LXA4 receptorExpression does not changeExpression does not changeNo significant impact on prognosis
GPR17Cysteinyl-leukotrienes receptorExpression does not changeExpression does not changeNo significant impact on prognosis
GPR3112S-HETE receptorExpression does not changeExpression does not changeNo significant impact on prognosis
OXGR1/GPR99LTE4 receptorExpression does not changeExpression does not changeNo significant impact on prognosis
G2A/GPR1325-HETE, 12-HETE, 15-HETE, 9-HODE receptorExpression does not changeExpression does not changeWorse prognosis (p = 0.052)
Red background—higher expression in the tumor; blue background—lower expression in the tumor; red background—worse prognosis with higher expression.
Table 8. Pan-cancer analysis of expression of genes involved in the LOX pathway.
Table 8. Pan-cancer analysis of expression of genes involved in the LOX pathway.
Name of CancereLOX3/ALOXE35-LOX/ALOX5FLAP/ALOX5AP12S-LOX/ALOX1212R-LOX/ALOX12B15-LOX-1/ALOX1515-LOX-2/ALOX15BLTA4H/LTA4HLTC4S/LTC4SGGT1GGT5DPEP1DPEP2EPHX2
Adrenocortical carcinoma (ACC)=========
Bladder urothelial carcinoma (BLCA)==========
Breast invasive carcinoma (BRCA)=============
Cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC)===========
Cholangiocarcinoma (CHOL)=========
Colon adenocarcinoma (COAD)==========
Lymphoid neoplasm diffuse large B-cell lymphoma (DLBC)========
Esophageal carcinoma (ESCA)===========
Glioblastoma multiforme (GBM)=======
Head and neck squamous cell carcinoma (HNSC)==========
Kidney chromophobe (KICH)==========
Kidney renal clear cell carcinoma (KIRC)========
Kidney renal papillary cell carcinoma (KIRP)=======
Acute myeloid leukemia (LAML)=======
Brain lower grade glioma (LGG)==========
Liver hepatocellular carcinoma (LIHC)============
Lung adenocarcinoma (LUAD)=========
Lung squamous cell carcinoma (LUSC)=======
Ovarian serous cystadenocarcinoma (OV)==========
Pancreatic adenocarcinoma (PAAD)====
Pheochromocytoma and paraganglioma (PCPG)===========
Prostate adenocarcinoma (PRAD)=============
Rectum adenocarcinoma (READ)===========
Sarcoma (SARC)===========
Skin cutaneous melanoma (SKCM)======
Stomach adenocarcinoma (STAD)=============
Testicular germ cell tumors (TGCT)========
Thyroid carcinoma (THCA)===========
Thymoma (THYM)=======
Uterine corpus endometrial carcinoma (UCEC)=========
Uterine carcinosarcoma (UCS)=========
Red background, ↑—expression higher in tumor than in healthy tissue; blue background, ↓—expression lower in tumor than in healthy tissue; gray background, =—expression does not differ between tumor and healthy tissue.
Table 9. Significance of cytochromes P450 and GPR75 receptors in ARA metabolism and tumorigenic processes in GBM.
Table 9. Significance of cytochromes P450 and GPR75 receptors in ARA metabolism and tumorigenic processes in GBM.
NameExpression Level in GBM Tumor Relative to Healthy TissueImpact on Prognosis with Higher Expression in GBM Tumors
SourceGEPIA [9]Seifert et al. [8]GEPIA [9]
CYP1A2Expression does not changeExpression does not changeN/A
CYP1B1Expression does not changeExpression does not changeNo significant impact on prognosis
CYP2C8Lower expression in the tumorExpression does not changeNo significant impact on prognosis
CYP2C9Expression does not changeExpression does not changeN/A
CYP2C19Expression does not changeExpression does not changeN/A
CYP2J2Expression does not changeExpression does not changeNo significant impact on prognosis
CYP2U1Higher expression in the tumorHigher expression in the tumorNo significant impact on prognosis
CYP3A4Expression does not changeExpression does not changeWorse prognosis
p = 0.07
CYP4A11Expression does not changeExpression does not changeNo significant impact on prognosis
CYP4F2Expression does not changeExpression does not changeNo significant impact on prognosis
CYP4F3Expression does not changeN/ANo significant impact on prognosis
CYP4X1Lower expression in the tumorLower expression in the tumorNo significant impact on prognosis
GPR75Expression does not changeExpression does not changeNo significant impact on prognosis
Red background—higher expression in the tumor; blue background—lower expression in the tumor; red background—worse prognosis with higher expression.
Table 10. Pan-cancer analysis of the expression of the cytochromes P450 and GPR75 receptor genes in question.
Table 10. Pan-cancer analysis of the expression of the cytochromes P450 and GPR75 receptor genes in question.
Name of CancerCYP1A2CYP1B1CYP2C8CYP2C9CYP2C19CYP2J2CYP2U1CYP3A4CYP4A11CYP4F2CYP4F3CYP4X1GPR75
Adrenocortical carcinoma (ACC)===========
Bladder urothelial carcinoma (BLCA)===========
Breast invasive carcinoma (BRCA)=============
Cervical squamous cell carcinoma and endocervical adenocarcinoma (CESC)=========
Cholangiocarcinoma (CHOL)===
Colon adenocarcinoma (COAD)=========
Lymphoid neoplasm diffuse large B-cell lymphoma (DLBC)===========
Esophageal carcinoma (ESCA)=========
Glioblastoma multiforme (GBM)==========
Head and neck squamous cell carcinoma (HNSC)===========
Kidney chromophobe (KICH)========
Kidney renal clear cell carcinoma (KIRC)========
Kidney renal papillary cell carcinoma (KIRP)========
Acute myeloid leukemia (LAML)==========
Brain lower grade glioma (LGG)=========
Liver hepatocellular carcinoma (LIHC)=======
Lung adenocarcinoma (LUAD)============
Lung squamous cell carcinoma (LUSC)=============
Ovarian serous cystadenocarcinoma (OV)===========
Pancreatic adenocarcinoma (PAAD)=======
Pheochromocytoma and paraganglioma (PCPG)===========
Prostate adenocarcinoma (PRAD)============
Rectum adenocarcinoma (READ)=========
Sarcoma (SARC)==========
Skin cutaneous melanoma (SKCM)========
Stomach adenocarcinoma (STAD)========
Testicular germ cell tumors (TGCT)==========
Thyroid carcinoma (THCA)===========
Thymoma (THYM)==========
Uterine corpus endometrial carcinoma (UCEC)==========
Uterine carcinosarcoma (UCS)==========
Red background, ↑—expression higher in tumor than in healthy tissue; blue background, ↓—expression lower in tumor than in healthy tissue; gray background, =—expression does not differ between tumor and healthy tissue.
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.

Share and Cite

MDPI and ACS Style

Korbecki, J.; Rębacz-Maron, E.; Kupnicka, P.; Chlubek, D.; Baranowska-Bosiacka, I. Synthesis and Significance of Arachidonic Acid, a Substrate for Cyclooxygenases, Lipoxygenases, and Cytochrome P450 Pathways in the Tumorigenesis of Glioblastoma Multiforme, Including a Pan-Cancer Comparative Analysis. Cancers 2023, 15, 946. https://doi.org/10.3390/cancers15030946

AMA Style

Korbecki J, Rębacz-Maron E, Kupnicka P, Chlubek D, Baranowska-Bosiacka I. Synthesis and Significance of Arachidonic Acid, a Substrate for Cyclooxygenases, Lipoxygenases, and Cytochrome P450 Pathways in the Tumorigenesis of Glioblastoma Multiforme, Including a Pan-Cancer Comparative Analysis. Cancers. 2023; 15(3):946. https://doi.org/10.3390/cancers15030946

Chicago/Turabian Style

Korbecki, Jan, Ewa Rębacz-Maron, Patrycja Kupnicka, Dariusz Chlubek, and Irena Baranowska-Bosiacka. 2023. "Synthesis and Significance of Arachidonic Acid, a Substrate for Cyclooxygenases, Lipoxygenases, and Cytochrome P450 Pathways in the Tumorigenesis of Glioblastoma Multiforme, Including a Pan-Cancer Comparative Analysis" Cancers 15, no. 3: 946. https://doi.org/10.3390/cancers15030946

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

Article Metrics

Back to TopTop