Skip to main content
The Journal of Biological Chemistry logoLink to The Journal of Biological Chemistry
. 2025 Aug 26;301(10):110629. doi: 10.1016/j.jbc.2025.110629

Recent updates in mammalian oxylipin biochemistry

Valerie B O’Donnell 1,
PMCID: PMC12481908  PMID: 40876591

Abstract

Oxylipins represent large families of bioactive lipids generated through the oxygenation of fatty acids. Although originally discovered almost 100 years ago, significant interest in these lipids continues today with many important discoveries on their structures and functions still being made, particularly in regards to mammalian biology. This review will highlight some aspects of the current status of the field, focusing on recent findings that continue to challenge our views about how oxylipins are made and act in human health and disease. Areas to be covered include: an update on the ever-expanding roles of prostaglandinE2 in cancer, octadecanoid structures and nomenclature, complex forms of oxylipins, and challenges and considerations for oxylipin analysis including potential clinical utility.

Keywords: lipid, oxylipin, eicosanoid, cyclooxygenase, prostaglandin, lipoxygenase


The term oxylipin generally refers to oxygenated metabolites of primarily monounsaturated fatty acid and polyunsaturated fatty acid (PUFA) that originate mainly from linoleic acid (LA), arachidonic acid, eicosapentaneoic acid, and docosahexanoic acid and they can be either enzymatic or nonenzymatic in origin. The first use of “oxylipin” was in 1991 when Hamberg described the formation of vicinal dihydroxy fatty acids (FAs) in the red alga Gracilariopsis lemaneiformis (1). Since then it has become used as an umbrella term to encompass well-known families of bioactive oxygenated FAs, such as octadecanoids (C18) (2), eicosanoids (C20) (3, 4), docosanoids (C22) (5, 6), as well as prostaglandins (PGs) and leukotrienes. In the LIPID MAPS Classification and Nomenclature, these lipids are all grouped under the category fatty acyls, within main classes that refer to their hydrocarbon chain lengths (https://lipidmaps.org/databases/lmsd/browse). Oxylipins have been known about for almost 100 years, with early work during the 1930s by Kurzrock and Lieb (7), and von Euler (8), on PGs. Later, in the second half of the 20th century, PGs and thromboxanes were structurally characterized by Hamberg, Bergström, and Samuelsson (9, 10), leading to the awarding of a Nobel Prize. Since that time, there has been an explosion of research into the biology and biochemistry of this large and expanding group of lipids, which continues to show no sign of waning in interest. The purpose of this review is to focus on recent findings in the field of mammalian oxylipin research and highlight gaps in knowledge that remain to be addressed, as well as discuss their analysis for both research and clinical applications.

The ever-expanding role of PGE2 in cancer

PGE2 was one of the first members of the PG family to be identified and characterized many years ago by Bergström, Sjövall, Samuelsson et al.(11, 12, 13). It is a product of microsomal prostaglandin E synthase (mPGES), acting on PGH2, itself a product of either cyclooxygenase-1 or cyclooxygenase-2 (COX). PGE2 is formed at varying levels depending on the specific biological context and signals potently at very low concentrations, binding and activating G protein–coupled receptors (GPCRs), termed EP1-4. This evokes a huge range of tissue and organ specific responses that are even still being delineated as our knowledge of how this important lipid participates in biology continue to be revealed.

PGE2 plays important and diverse roles in cancer, for example, through stimulating tumor growth and progression, promoting cell proliferation, preventing apoptosis, and increasing invasion and metastasis. These effects are mediated particularly through EP 1, 2 and 4 receptors, signaling through diverse mechanisms including PKC/NF-κB/Forkhead box protein C2, and EGFR/PI3K (14, 15, 16). A urinary metabolite of PGE2, PGE-M is elevated in human cancers including gastric and colon and correlates with poor outcomes (17, 18, 19), suggesting the lipid as a potential therapeutic target, as well as a biomarker (20). Since the early 2000s there has been huge interest in delineating how PGE2 promotes cancer and the involvement of PGE2-driven inflammation in this process. Major efforts were made to develop selective inhibitors of its signaling, as summarized in a recent review (21). Initially, based on compelling population level data, nonsteroidal anti-inflammatory drugs (NSAIDs) and aspirin were trialed, however due to their potential for side effects, including gastrointestinal bleeding, this has not made it into routine clinical use. A separate approach relates to inhibition of mPGES, to prevent generation of PGE2 directly, aiming to avoid the cardiovascular impacts of NSAIDs such as higher blood pressure due to prostacyclin inhibition. Although there is interest in development of mPGES inhibitors, only two phase I studies are listed on clinicaltrials.org, looking at evaluating safety, tolerability, and pharmacokinetics in healthy and elderly subjects of a drug, GRC27864 (NCT02361034, NCT02179645), and no studies in cancer are listed. These completed already in 2014/15, and showed good safety and efficacy in vivo (22), but follow on trials do not seem to have been initiated since.

As a more targeted approach, EP2 and EP4 antagonists are being actively tested both for their ability to slow gastric cancers and modulate immune responses. As yet, these have not made it into clinical use, although several trials are ongoing. An attractive hypothesis for this therapy relates to the combined use of EP receptor antagonists with immune checkpoint inhibitors. This takes into account how PGE2 acts in the immune system, for example, through contributing to immune evasion by recruiting regulatory T cells (Tregs) to the tumor microenvironment (23). In this context, Tregs can suppress the ability of cytotoxic T cells (CD8+ T cells) from targeting cancer cells. PGE2 also regulates macrophages, altering their polarization toward immunosuppression (24). As an example of a potential mechanism by which inflammation and PGE2 might influence colon cancer development, recent studies have shown how the lipid disrupts gut microbiota Treg communication via EP4 to increase inflammation, with this dysregulated in aging (25, 26).

Six trials on the EP4 antagonist E7046/AN0025 are listed on clinicaltrials.gov. Two are terminated/suspended and two are active (NCT05191667, NCT04432857, NCT04975958, NCT02540291). In 2020, this drug was found to be safe and well tolerated in patients with various advanced tumors (27), and a trial in advanced rectal cancer in combination with chemoradiation completed in 2021 reporting encouraging preliminary efficacy results, and aiming to recommend phase 2 doses (NCT03152370) (28, 29). However, a trial that commenced in 2024 evaluating efficacy of this drug by progression free survival and other outcome measures appears to have been withdrawn (NCT05358691). A phase II trial of another EP4 antagonist (AAT-007) in advanced solid tumors was also withdrawn, this time due to the principal investigator moving institutions (NCT02538432), while a phase Ib study on the EP4 antagonist Grapiprant in combination with immune checkpoint inhibition completed in 2023 but has not yet reported (NCT03658772). Other studies on EP4 antagonists remain open and recruiting including a phase Ib/IIa study on CR6086 (NCT05205330), and a phase I/IIa study on HTL0039732, both in combination with immune checkpoint blockade (NCT05944237). Encouragingly, a dual EP2/EP4 antagonist was superior at reducing tumor burden in multiple mouse models to targeting EP4 alone (30). This approach is now being trialed in humans with a trial of a dual EP2/EP4 antagonist (TPST-1495) (NCT04344795) currently active with preliminary data reporting “pharmacodynamic activity, a manageable safety profile, and a potential signal of activity consistent with the preclinical data,” and an endometrial cancer therapy combination study is currently recruiting (NCT06129604) (31). Encouragingly, a phase II trial on this drug in familial adenomatous polyposis is about to open (NCT06557733), while another dual EP2/EP4 antagonist (MBF-362) recently completed a phase I evaluation for safety and preliminary efficacy in solid tumors (NCT05940571). Last, a study testing a triple antagonist of EP2, EP4, and the PGD2 receptor DP1 as monotherapy and in combination with immune checkpoint inhibition is recruiting (NCT06789172). This large suite of trials indicates significant pharma interest in the potential for PGE2 blockade in cancer, and over the next few years as phase II studies start to report, it will become clear whether there is a role for inhibition of EP antagonism in chemotherapy, particularly in combination with immune checkpoint blockade. Importantly, targeting EP receptors should avoid the side effects of NSAIDs. So far, urinary PGE-M has not become a routine biomarker for cancer diagnostics, but if EP antagonists prove clinically utility, then this may become a tool for patient stratification for therapies targeting PGE2. A summary of the strategies for blocking PGE2 formation and action in cancer is shown (Fig. 1).

Figure 1.

Figure 1

Strategies for preventing signaling actions of PGE2 in cancer. PGE2, prostaglandinE2.

Revisiting old lipids, in the form of octadecanoids

Octadecanoids are oxylipins derived from oxygenation of C18 PUFA, most commonly linoleate (LA, 18:2,ω6) and α-linolenate (18:3,ω3) (Fig. 2). Although much is known regarding their formation and function in plants (32), where they are essential for stress, growth, and development, less research has been undertaken into their roles in mammalian health and disease (33). Mono-hydroxy oxylipins derived from C18 PUFA are highly abundant in human tissues, with their plasma and serum levels being greater than those from other FA (34). Whether the lipids are formed during coagulation is not clear with one study showing that levels of oxylipins from LA increase significantly (35) in particular the 9- and 13-hydroxyoctadecanoids (HODE), from LA, while others showing little change (36, 37, 38). Currently, the sources of individual octadecanoids in blood are not clearly defined but could include circulating vascular cells, endothelium or organs such as liver, and enzymes that include both lipoxygenases (LOXs) (39) and COXs (40, 41). The best studied mammalian octadecanoids are CYP-derived epoxides and their corresponding diols which have been found to play roles in cardiac failure (42), adult respiratory distress syndrome (43), and regulation of vascular, cardiac, pulmonary, and renal health (reviewed in (44)). Since 2016, these lipids have been linked with skin physiology, pain, itch, FA uptake and transport, inflammation, and burn injury (45, 46, 47, 48, 49, 50, 51, 52). Increasing their levels in vivo reduces disease in various vascular disease models (53, 54, 55), suggesting they may play a role in dampening inflammation (52). In common with many other FA-derived oxylipins, monohydroxy, and epoxy octadecanoids are well known as low-affinity peroxisome proliferator–activated receptor γ ligands, and inducers, contributing to dampening inflammation including through suppression of NLR family pyrin domain–containing 3 inflammasome (56, 57, 58, 59, 60). This was first observed many years ago (60), and later shown to be how octadecanoids prevent lipopolysaccharide-driven inflammation (61). Thus, peroxisome proliferator–activated receptor γ activation/induction may largely explain how these lipids mediate vascular protective effects in many other models. Despite the large abundance of octadecanoids, in particular the HODEs, there remain many unanswered questions relating to their biological sources and signaling actions. Few specific GPCR for octadecanoids are yet known, with 9-HODE/GPR132 being the only International Union of Basic and Clinical Pharmacology recognized ligand binding pair, and with none recognized for epoxyeicosatrienoic acids (EETs), hydroxyoctadecanoic acids (HOMEs), or other octadecanoids (62). Low-affinity ligand binding has been claimed for several EETs and for 10-HOME with GPR40 (63, 64), although this is unlikely to be a truly specific GPCR-dependent signaling mechanism. In line with this, GPR40 is only recognized by International Union of Basic and Clinical Pharmacology as a free FA, but not octadecanoid receptor. 12,13-diHOME can increase calcium influx by TRPV1 in neurons, however as this is mediated via PKC activation at physiological lipid concentrations, it does not appear to due to direct receptor-ligand binding (65). Nitrated octadecanoids signal through their ability to participate in Michael additions with reactive thiol groups on proteins and this has been found to mediate diverse signaling actions that are overall anti-inflammatory in nature (66). Relating to skin physiology, an essential role for linoleate (including via retroconversion from arachidonic acid) in barrier formation has been known about since the 1980s (67, 68). Later, around 2010, several studies, including with KO mice, demonstrated a key role for LOXs in this process, as well as providing in vivo evidence of roles for 12R-LOX, eLOX3, and SDR9C7 (69, 70, 71, 72, 73). More recent studies provided the structures of the octadecanoid-containing ceramides in full (74, 75).

Figure 2.

Figure 2

Structures of various octadecanoids, showing positions of double bonds and geometric isomers.

The last few years has seen a resurgence in interest in octadecanoid mammalian biology, in part driven by development of new analytical approaches (76) and recent updates to their nomenclature and classification, both of which serve to highlight the diversity of these compounds and how little is known about them (77). Chiral methods enabled up to 103 standards to be separately analyzed, revealing approximately 20 of these to be detectable and quantifiable in human and murine plasma (78). A protocol for this was recently published here (79). Last, desorption electrospray ionization mass spectrometry–multiple reaction monitoring imaging was recently applied to lung tissue LA and α-linolenate metabolites (80). These new analytical tools for the study of octadecanoids will undoubtedly help to stimulate further research into their biological roles. Highlighting the major gaps in this field, a comprehensive review into octadecanoid biology and bioactivity published in 2025 (77) concluded that many gaps exist in the field including developing a consensus on tissue distribution and endogenous concentrations, what specific receptors might exist, their triggers for biosynthesis, and delineating the structures and functions of lesser known octadecanoids. With the new analytical tools available, these questions are ripe for further exploration in the years to come.

Oxylipins as part of larger molecules, expanding bioactions

Although oxylipins are traditionally considered to be generated and to mediate their bioactivity as short-lived, locally acting free acid autocoids, it has become increasingly recognized that they also form through biochemically regulated processes attached to larger lipids such as phospholipids, endocannabinoids, glycerides, acylceramides, and sterol esters, and that these species serve distinct functions to their free acid counterparts. Enzymatic formation is very different from the free radical–dependent formation of oxidized phospholipids that was the subject of intense research in the 1980s to 1990s, instead involving either direct oxidation (phospholipids, endocannabinoids, sterol esters) or esterification of an endogenously formed oxylipins by the action of Lands cycle enzymes (phospholipids). In the case of phospholipids, acute activation of 5- or 12-LOXs and COX-1 or COX-2 in immune cells and platelets leads to endogenous generation of oxylipins that are rapidly esterified into phospholipids, in substantial amounts, termed enzymatically oxidized phospholipids (eoxPLs) (81, 82). In contrast, 15-LOX1 can directly oxygenate phospholipids in macrophages and eosinophils (83, 84). A more recently identified pathway in platelets involves formation of a 12-hydroxyeicosatetraenoic acid-lysophosphatidylcholine (12-HETE-lysophosphatidylethanolamine) by direct action of 12-LOX (85). This follows generation of the 2-arachidonyl-lysophosphatidylethanolamine substrate by calcium-independent phospholipase A2 (85). A very different mechanism for eoxPL formation involves incorporation of exogenously added oxylipins into membranes in cells in vitro. This was extensively researched in the 1990s, for both EETs and HETEs, and it was suggested that the lipids might represent a potential reservoir for oxylipins to be later released through hydrolysis. However, the oxylipin incorporation pattern is different for endogenous oxylipins (summarized here (81)) and a biological role for oxPL formed through such transcellular mechanisms still has not been yet proven. In contrast, eoxPL generated endogenously by platelets were shown to support coagulation and thrombosis in vitro, and in vivo in mouse models, through enhancing the bioactivity of phosphatidylserine (86, 87, 88, 89). Current gaps in our knowledge about eoxPL include exactly which Lands cycle enzymes are responsible for their generation, including the acyl-CoA synthetase long-chain family members that generate the CoA intermediates, and how this process is regulated during inflammation and innate immunity. In this regard, a role for lysophosphatidylcholine acyltransferase-3 in selectively forming 12-LOX–derived diacyl-12-HETE-phosphatidylethanolamine in platelets was recently reported (82). In contrast, lysophosphatidylcholine acyltransferase-3 played no role in esterification of COX-1–derived 11-HETE or 15-HETE into platelet phospholipid pools, and the enzymes responsible remain so far unknown (82). Recently increased blood cell eoxPL were found to be related to elevated thrombotic risk, in rheumatoid arthritis (RA) and atherosclerotic cardiovascular disease (82, 90). In RA, this was driven by interleukin-6–dependent platelet activity in mice and associated with higher levels of anti-eoxPL-IgG in human RA (90).

In the case of endocannabinoids, direct oxygenation by COX-2 was reported many years ago, forming PG, thromboxane, and prostacyclin forms of glycerol esters and ethanolamides (91), some of which can interact with EP receptors (92). While there have been some reports on the bioactivity of these lipids over the years, including an anti-inflammatory role for glyceryl-PGD2 (93), a lot remains to be discovered relating to their mechanisms of formation and possible roles in other conditions where endocannabinoids are important. Esterified oxylipins were reported to be formed in plasma, including in lipoproteins where they accounted for >90% of the total oxylipins detected (94). Whether they are from autoxidation or enzymatic activity is not known but considering that this pool includes isoprostanes, it is possible that they are largely generated nonenzymatically. As a cautionary note, a more recent study showed their artefactual formation can occur during sample processing (95) (see here for a comprehensive review (96)). In plasma, the main source of esterified oxylipins appears to be glycerides, and also phospholipids. Relevance to disease was recently shown with significant changes seen in plasma from obese type 2 diabetics, where a potential involvement of 12-LOX was proposed based on positional isomer composition (95).

Challenges in oxylipin analysis for basic research and clinical utility

The analysis of oxylipins is best performed using high sensitivity LC-MS/MS, with around 200 oxylipin primary standards now being available commercially, as well as several deuterated forms. State-of-the-art LC-MS/MS methods can robustly quantify down around 0.1 to 1 pg on column, depending on the platform, and many studies have been published on their application to oxylipin analysis in diverse tissues including plasma. Although the basic principles of oxylipin analysis is not any different from analysis of any other biomolecules, there are some additional points that researchers need to be aware of. These relate to isomeric separation, unusual behavior of particular oxylipins on column, and potential for artefactual generation during sample processing. These were recently summarized in a set of recommendations published by an International Lipidomics Society Interest Group, which was coauthored by almost 100 analysts working in the field (97). The recommendations described are for basic research, not clinical purposes, where a far higher degree of validation is required. Indeed, oxylipin analysis has not yet made it into routine clinical use. So far a clear demonstration that a specific oxylipin would represent a useful biomarker is lacking, considering that (i) increases in some, for example, PGs, may be a general feature of most forms of inflammation, and so may not provide further specificity than existing markers such as C-reactive protein, (ii) there are challenges with standardizing their analysis across clinical laboratories with significant potential for artefactual generation during sample processing (e.g. platelet-derived 12-HETE or thromboxaneB2 which can form during venipuncture). A recent study which avoided sampling artefacts reported plasma 12-HETE correlated with disease severity in COVID-19 (98). This may indicate a signature of platelet activation, but whether 12-HETE could indicate clinical need for anticoagulation therapy has not yet been tested. Consistent with this, plasma P-selectin is elevated during thromboembolism in COVID-19 (99). If true, this suggests that analysis of platelet activation markers, including oxylipins, may have future clinical utility in guiding the need for thrombolytic intervention.

Urinary oxylipins were the focus of much research during the 1980s and 1990s since they do not suffer from sampling artefacts, and the PGE2 metabolite, PGE-M was reported to be increased in various cancers, as outlined earlier in this review. Recently, Milne et al. turned their attention to the analysis of glucuronide metabolites that might represent endogenous markers of oxylipin biosynthesis, particularly where the primary oxylipin is low or has been rapidly metabolized. Glucuronide metabolites of isoprostanes were easily detected and characterized in human urine and could be altered through dietary modification (100). In contrast, although liver microsomes were able to metabolize resolvinD1 and D5 to glucuronides in vitro (101), these forms have not yet been detected in urine or other tissues. In summary, during the last several years, rapid advances in LC-MS/MS instrumentation and widening availability of primary and deuterated standards has enabled oxylipins to be studied in large numbers at low concentrations, in ways that were simply not possible up to now. An ongoing challenge in the field is to ensure consistency in their analysis, aiming for reproduction of key findings, to enable a consensus on their generation, mechanisms of formation and bioactions in health and disease.

Conclusion

This review is intended as a short update to the field of mammalian oxylipin research. Overall, there is significant current interest in the clinical analysis of oxylipins, although in what context this will prove useful for diagnostics or therapeutics still somewhat needs to be defined. Anticoagulation or cancer therapeutics represent promising avenues considering the major problem of thrombosis in almost all forms of disease and the significant current investment in therapies targeting PGE2 in cancer. Novel metabolites of oxylipins, measured in urine are now being revisited with new forms being characterized. This is a welcome development considering sampling artefacts in plasma which do not apply to urine, and so, revisiting previous work that defined the urinary metabolites of several oxylipins would be a worthwhile endeavor for the field.

Dedication

This review is written in honor of William (Bill) Smith, a true gentleman scientist who I knew to be one of the founding fathers of COX enzymology and biochemistry in the late 20th century. During the 1990s and early 2000s, I followed very closely his seminal papers on the mechanisms of COX peroxidase activity, which informed greatly my own studies into how nitric oxide is consumed by COX peroxidase, through acting as a direct reducing substrate. Following this his demonstration of COX homodimers was revolutionary in the field and went on to explain the specific interactions of various inhibitors and FAs with the different isomers. For about 4 years in the early 2010s, I would spend a month a year in Robert (Bob) Murphy’s lab in Denver, learning how to analyze oxylipins using LC-MS/MS. Due to their great friendship, Bill was often visiting at the same time, and so we overlapped somewhat, which I saw as a wonderful opportunity not to be missed, to expand my knowledge of COX enzymology. Those days are greatly missed.

Conflict of interest

The authors declare that they have no conflicts of interest with the contents of this article.

Acknowledgments

I would like to gratefully acknowledge Drs Larry Marnett and Craig Wheelock for proofreading the draft.

Author contributions

V. O. D. conceptualization; V. O. D. writing–original draft; V. O. D. writing–review and editing.

Reviewed by members of the JBC Editorial Board. Edited by George M. Carman

References

  • 1.Gerwick W.H., Moghaddam M., Hamberg M. Oxylipin metabolism in the red alga gracilariopsis lemaneiformis: mechanism of formation of vicinal dihydroxy fatty acids. Arch. Biochem. Biophys. 1991;290:436–444. doi: 10.1016/0003-9861(91)90563-x. [DOI] [PubMed] [Google Scholar]
  • 2.Schade U.F., Burmeister I., Engel R. Increased 13-hydroxyoctadecadienoic acid content in lipopolysaccharide stimulated macrophages. Biochem. Biophys. Res. Commun. 1987;147:695–700. doi: 10.1016/0006-291x(87)90986-7. [DOI] [PubMed] [Google Scholar]
  • 3.Astarita G., Kendall A.C., Dennis E.A., Nicolaou A. Targeted lipidomic strategies for oxygenated metabolites of polyunsaturated fatty acids. Biochim. Biophys. Acta. 2015;1851:456–468. doi: 10.1016/j.bbalip.2014.11.012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 4.Dennis E.A., Norris P.C. Eicosanoid storm in infection and inflammation. Nat. Rev. Immunol. 2015;15:511–523. doi: 10.1038/nri3859. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Aveldaño M.I., Sprecher H. Synthesis of hydroxy fatty acids from 4, 7, 10, 13, 16, 19-[1-14C] docosahexaenoic acid by human platelets. J. Biol. Chem. 1983;258:9339–9343. [PubMed] [Google Scholar]
  • 6.VanRollins M., Murphy R.C. Autooxidation of docosahexaenoic acid: analysis of ten isomers of hydroxydocosahexaenoate. J. Lipid Res. 1984;25:507–517. [PubMed] [Google Scholar]
  • 7.Kurzrock R., Lieb C.C. Biochemical studies of human semen. II. The action of semen on the human uterus. Proc. Soc. Exp. Biol. Med. 1930;28:268. [Google Scholar]
  • 8.von Euler U.S. On the specific vasodilating and plain muscle stimulating substances from accessory genital glands in man and certain animals (prostaglandin and vesigIandin) J. Physiol. 1936;88:213. doi: 10.1113/jphysiol.1936.sp003433. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Bergstroem S., Ryhage R., Samuelsson B., Sjoevall J. Prostaglandins and related factors. 15. The structures of prostaglandin E1, F1-Alpha, and F1-Beta. J. Biol. Chem. 1963;238:3555–3564. [PubMed] [Google Scholar]
  • 10.Hamberg M., Samuelsson B. On the mechanism of the biosynthesis of prostaglandins E-1 and F-1-alpha. J. Biol. Chem. 1967;242:5336–5343. [PubMed] [Google Scholar]
  • 11.Bergström S., Sjövall J., Pihl A., Almin K., Magnéli A., Pestmalis H., et al. The isolation of prostaglandin. Acta Chem. Scand. 1957;11:1086. [Google Scholar]
  • 12.Bergström S., Sjövall J. The isolation of prostaglandin E from sheep prostate glands. Acta Chem. Scand. 1960;14:1701–1705. [Google Scholar]
  • 13.Samuelsson B. On the incorporation of oxygen in the conversion of 8, 11, 14-eicosatrienoic acid to prostaglandin E1. J. Am. Chem. Soc. 1965;87:3011–3013. doi: 10.1021/ja01091a043. [DOI] [PubMed] [Google Scholar]
  • 14.Bai X., Wang J., Guo Y., Pan J., Yang Q., Zhang M., et al. Prostaglandin E2 stimulates β1-integrin expression in hepatocellular carcinoma through the EP1 receptor/PKC/NF-κB pathway. Sci. Rep. 2014;4:6538. doi: 10.1038/srep06538. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Ji R., Chou C.L., Xu W., Chen X.B., Woodward D.F., Regan J.W. EP1 prostanoid receptor coupling to G i/o up-regulates the expression of hypoxia-inducible factor-1 alpha through activation of a phosphoinositide-3 kinase signaling pathway. Mol. Pharmacol. 2010;77:1025–1036. doi: 10.1124/mol.110.063933. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Wang Q., Morris R.J., Bode A.M., Zhang T. Prostaglandin pathways: opportunities for cancer prevention and therapy. Cancer Res. 2022;82:949–965. doi: 10.1158/0008-5472.CAN-21-2297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Johnson J.C., Schmidt C.R., Shrubsole M.J., Billheimer D.D., Joshi P.R., Morrow J.D., et al. Urine PGE-M: a metabolite of prostaglandin E2 as a potential biomarker of advanced colorectal neoplasia. Clin. Gastroenterol. Hepatol. 2006;4:1358–1365. doi: 10.1016/j.cgh.2006.07.015. [DOI] [PubMed] [Google Scholar]
  • 18.Shrubsole M.J., Cai Q., Wen W., Milne G., Smalley W.E., Chen Z., et al. Urinary prostaglandin E2 metabolite and risk for colorectal adenoma. Cancer Prev. Res. (Phila) 2012;5:336–342. doi: 10.1158/1940-6207.CAPR-11-0426. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Dong L.M., Shu X.O., Gao Y.T., Milne G., Ji B.T., Yang G., et al. Urinary prostaglandin E2 metabolite and gastric cancer risk in the shanghai women's health study. Cancer Epidemiol. Biomarkers Prev. 2009;18:3075–3078. doi: 10.1158/1055-9965.EPI-09-0680. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Wang D., DuBois R.N. Urinary PGE-M: a promising cancer biomarker. Cancer Prev. Res. 2013;6:507–510. doi: 10.1158/1940-6207.CAPR-13-0153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Wilson D.J., DuBois R.N. Role of prostaglandin E2 in the progression of gastrointestinal cancer. Cancer Prev. Res. 2022;15:355–363. doi: 10.1158/1940-6207.CAPR-22-0038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Sant S., Tandon M., Menon V., Gudi G., Kattige V., Khairatkar Joshi N., et al. GRC 27864, novel, microsomal prostaglandin E synthase-1 enzyme inhibitor: phase 1 study to evaluate safety, PK and biomarkers in healthy, adult subjects. Osteoarthritis Cartilage. 2018;26:S351–S352. [Google Scholar]
  • 23.Thumkeo D., Punyawatthananukool S., Prasongtanakij S., Matsuura R., Arima K., Nie H., et al. PGE2-EP2/EP4 signaling elicits immunosuppression by driving the mregDC-Treg axis in inflammatory tumor microenvironment. Cell Rep. 2022;39 doi: 10.1016/j.celrep.2022.110914. [DOI] [PubMed] [Google Scholar]
  • 24.Sinha P., Clements V.K., Fulton A.M., Ostrand-Rosenberg S. Prostaglandin E2 promotes tumor progression by inducing myeloid-derived suppressor cells. Cancer Res. 2007;67:4507–4513. doi: 10.1158/0008-5472.CAN-06-4174. [DOI] [PubMed] [Google Scholar]
  • 25.Crittenden S., Goepp M., Pollock J., Robb C.T., Smyth D.J., Zhou Y., et al. Prostaglandin E(2) promotes intestinal inflammation via inhibiting microbiota-dependent regulatory T cells. Sci. Adv. 2021;7 doi: 10.1126/sciadv.abd7954. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Goepp M., Milburn J.V., Zhang B., Dong Y., Tyrrell V., Zheng X., et al. Age-related impairment of intestinal inflammation resolution through an eicosanoid-immune-microbiota axis. Cell Host Microbe. 2025;33:671–687.e676. doi: 10.1016/j.chom.2025.04.014. [DOI] [PubMed] [Google Scholar]
  • 27.Hong D.S., Parikh A., Shapiro G.I., Varga A., Naing A., Meric-Bernstam F., et al. First-in-human phase I study of immunomodulatory E7046, an antagonist of PGE(2)-receptor E-type 4 (EP4), in patients with advanced cancers. J. Immunother. Cancer. 2020;8 doi: 10.1136/jitc-2019-000222. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Wyrwicz L., Saunders M.P., Hall M., Ng J., Prasad V.B., Lautermilch N., et al. 540P - a phase Ib study of E7046 (AN0025) in combination with radiotherapy/chemoradiotherapy (RT/CRT) in preoperative treatment of rectal cancer. Ann. Oncol. 2019;30 [Google Scholar]
  • 29.Wyrwicz L., Saunders M., Hall M., Ng J., Hong T., Xu S., et al. AN0025, a novel antagonist of PGE2-receptor E-type 4 (EP4), in combination with total neoadjuvant treatment of advanced rectal cancer. Radiother. Oncol. 2023;185 doi: 10.1016/j.radonc.2023.109669. [DOI] [PubMed] [Google Scholar]
  • 30.Francica B.J., Holtz A., Lopez J., Freund D., Chen A., Wang D., et al. Dual blockade of EP2 and EP4 signaling is required for optimal immune activation and antitumor activity against prostaglandin-expressing tumors. Cancer Res. Commun. 2023;3:1486–1500. doi: 10.1158/2767-9764.CRC-23-0249. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Ulahannan S.V., Powderly J.D., Johnson M.L., Krauss J.C., Sharma M., Davar D., et al. A phase 1 study of TPST-1495 as a single agent and in combination with pembrolizumab in patients with advanced solid tumors. J. Clin. Oncol. 2023;41:3107. [Google Scholar]
  • 32.Wasternack C. Jasmonates: an update on biosynthesis, signal transduction and action in plant stress response, growth and development. Ann. Bot. 2007;100:681–697. doi: 10.1093/aob/mcm079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Quaranta A., Revol-Cavalier J., Wheelock C.E. The octadecanoids: an emerging class of lipid mediators. Biochem. Soc. Trans. 2022;50:1569–1582. doi: 10.1042/BST20210644. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Psychogios N., Hau D.D., Peng J., Guo A.C., Mandal R., Bouatra S., et al. The human serum metabolome. PLoS One. 2011;6 doi: 10.1371/journal.pone.0016957. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Hellhake S., Meckelmann S.W., Empl M.T., Rentmeister K., Wißdorf W., Steinberg P., et al. Non-targeted and targeted analysis of oxylipins in combination with charge-switch derivatization by ion mobility high-resolution mass spectrometry. Anal. Bioanal. Chem. 2020;412:5743–5757. doi: 10.1007/s00216-020-02795-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Kolmert J., Fauland A., Fuchs D., Säfholm J., Gómez C., Adner M., et al. Lipid mediator quantification in isolated human and Guinea pig airways: an expanded approach for respiratory research. Anal. Chem. 2018;90:10239–10248. doi: 10.1021/acs.analchem.8b01651. [DOI] [PubMed] [Google Scholar]
  • 37.Pedersen T.L., Gray I.J., Newman J.W. Plasma and serum oxylipin, endocannabinoid, bile acid, steroid, fatty acid and nonsteroidal anti-inflammatory drug quantification in a 96-well plate format. Analytica Chim. Acta. 2021;1143:189–200. doi: 10.1016/j.aca.2020.11.019. [DOI] [PubMed] [Google Scholar]
  • 38.Rajan M.R., Sotak M., Barrenäs F., Shen T., Borkowski K., Ashton N.J., et al. Comparative analysis of obesity-related cardiometabolic and renal biomarkers in human plasma and serum. Sci. Rep. 2019;9 doi: 10.1038/s41598-019-51673-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Soberman R.J., Harper T.W., Betteridge D., Lewis R.A., Austen K.F. Characterization and separation of the arachidonic acid 5-lipoxygenase and linoleic acid omega-6 lipoxygenase (arachidonic acid 15-lipoxygenase) of human polymorphonuclear leukocytes. J. Biol. Chem. 1985;260:4508–4515. [PubMed] [Google Scholar]
  • 40.Laneuville O., Breuer D.K., Xu N., Huang Z.H., Gage D.A., Watson J.T., et al. Fatty acid substrate specificities of human prostaglandin-endoperoxide H synthase-1 and -2. Formation of 12-hydroxy-(9Z, 13E/Z, 15Z)- octadecatrienoic acids from alpha-linolenic acid. J. Biol. Chem. 1995;270:19330–19336. doi: 10.1074/jbc.270.33.19330. [DOI] [PubMed] [Google Scholar]
  • 41.Walsh P., Behrens N., Carvallo Chaigneau F.R., McEligot H., Agrawal K., Newman J.W., et al. A randomized placebo controlled trial of ibuprofen for respiratory syncytial virus infection in a bovine model. PLOS One. 2016;11 doi: 10.1371/journal.pone.0152913. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Sugiyama S., Hayakawa M., Nagai S., Ajioka M., Ozawa T. Leukotoxin, 9, 10-epoxy-12-octadecenoate, causes cardiac failure in dogs. Life Sci. 1987;40:225–231. doi: 10.1016/0024-3205(87)90336-5. [DOI] [PubMed] [Google Scholar]
  • 43.Moghaddam M.F., Grant D.F., Cheek J.M., Greene J.F., Williamson K.C., Hammock B.D. Bioactivation of leukotoxins to their toxic diols by epoxide hydrolase. Nat. Med. 1997;3:562–566. doi: 10.1038/nm0597-562. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Newman J.W., Morisseau C., Hammock B.D. Epoxide hydrolases: their roles and interactions with lipid metabolism. Prog. Lipid Res. 2005;44:1–51. doi: 10.1016/j.plipres.2004.10.001. [DOI] [PubMed] [Google Scholar]
  • 45.Chiba T., Thomas C.P., Calcutt M.W., Boeglin W.E., O'Donnell V.B., Brash A.R. The precise structures and stereochemistry of trihydroxy-linoleates esterified in human and porcine epidermis and their significance in skin barrier function. J. Biol. Chem. 2016;291:14540–14554. doi: 10.1074/jbc.M115.711267. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Ramsden C.E., Ringel A., Majchrzak-Hong S.F., Yang J., Blanchard H., Zamora D., et al. Dietary linoleic acid-induced alterations in pro- and anti-nociceptive lipid autacoids:implications for idiopathic pain syndromes? Mol. Pain. 2016;12 doi: 10.1177/1744806916636386. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Ramsden C.E., Domenichiello A.F., Yuan Z.-X., Sapio M.R., Keyes G.S., Mishra S.K., et al. A systems approach for discovering linoleic acid derivatives that potentially mediate pain and itch. Sci. Signal. 2017;10 doi: 10.1126/scisignal.aal5241. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Domenichiello A.F., Sapio M.R., Loydpierson A.J., Maric D., Goto T., Horowitz M.S., et al. Molecular pathways linking oxylipins to nociception in rats. J. Pain. 2021;22:275–299. doi: 10.1016/j.jpain.2020.09.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Stanford K.I., Lynes M.D., Takahashi H., Baer L.A., Arts P.J., May F.J., et al. 12,13-diHOME: an exercise-induced lipokine that increases skeletal muscle fatty acid uptake. Cell Metab. 2018;27:1111–1120.e1113. doi: 10.1016/j.cmet.2018.03.020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 50.Lynes M.D., Leiria L.O., Lundh M., Bartelt A., Shamsi F., Huang T.L., et al. The cold-induced lipokine 12,13-diHOME promotes fatty acid transport into brown adipose tissue. Nat. Med. 2017;23:631–637. doi: 10.1038/nm.4297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Bergmann C.B., McReynolds C.B., Wan D., Singh N., Goetzman H., Caldwell C.C., et al. sEH-derived metabolites of linoleic acid drive pathologic inflammation while impairing key innate immune cell function in burn injury. Proc. Natl. Acad. Sci. U. S. A. 2022;119 doi: 10.1073/pnas.2120691119. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Gilroy D.W., Edin M.L., De Maeyer R.P.H., Bystrom J., Newson J., Lih F.B., et al. CYP450-derived oxylipins mediate inflammatory resolution. Proc. Natl. Acad. Sci. U. S. A. 2016;113:E3240–E3249. doi: 10.1073/pnas.1521453113. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Revermann M., Schloss M., Barbosa-Sicard E., Mieth A., Liebner S., Morisseau C., et al. Soluble epoxide hydrolase deficiency attenuates neointima formation in the femoral cuff model of hyperlipidemic mice. Arterioscler. Thromb. Vasc. Biol. 2010;30:909–914. doi: 10.1161/ATVBAHA.110.204099. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 54.Zhang L.N., Vincelette J., Cheng Y., Mehra U., Chen D., Anandan S.K., et al. Inhibition of soluble epoxide hydrolase attenuated atherosclerosis, abdominal aortic aneurysm formation, and dyslipidemia. Arterioscler. Thromb. Vasc. Biol. 2009;29:1265–1270. doi: 10.1161/ATVBAHA.109.186064. [DOI] [PubMed] [Google Scholar]
  • 55.Luo P., Chang H.H., Zhou Y., Zhang S., Hwang S.H., Morisseau C., et al. Inhibition or deletion of soluble epoxide hydrolase prevents hyperglycemia, promotes insulin secretion, and reduces islet apoptosis. J. Pharmacol. Exp. Ther. 2010;334:430–438. doi: 10.1124/jpet.110.167544. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 56.Kumar N., Gupta G., Anilkumar K., Fatima N., Karnati R., Reddy G.V., et al. 15-Lipoxygenase metabolites of α-linolenic acid, [13-(S)-HPOTrE and 13-(S)-HOTrE], mediate anti-inflammatory effects by inactivating NLRP3 inflammasome. Sci. Rep. 2016;6 doi: 10.1038/srep31649. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Umeno A., Sakashita M., Sugino S., Murotomi K., Okuzawa T., Morita N., et al. Comprehensive analysis of PPARγ agonist activities of stereo-, regio-, and enantio-isomers of hydroxyoctadecadienoic acids. Biosci. Rep. 2020;40 doi: 10.1042/bsr20193767. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Itoh T., Fairall L., Amin K., Inaba Y., Szanto A., Balint B.L., et al. Structural basis for the activation of PPARgamma by oxidized fatty acids. Nat. Struct. Mol. Biol. 2008;15:924–931. doi: 10.1038/nsmb.1474. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Li X., Kempf S., Günther S., Hu J., Fleming I. 11,12-EET regulates PPAR-γ expression to modulate TGF-β-Mediated macrophage polarization. Cells. 2023;12 doi: 10.3390/cells12050700. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 60.Liu Y., Zhang Y., Schmelzer K., Lee T.-S., Fang X., Zhu Y., et al. The antiinflammatory effect of laminar flow: the role of PPARγ, epoxyeicosatrienoic acids, and soluble epoxide hydrolase. Proc. Natl. Acad. Sci. U. S. A. 2005;102:16747–16752. doi: 10.1073/pnas.0508081102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Samokhvalov V., Vriend J., Jamieson K.L., Akhnokh M.K., Manne R., Falck J.R., et al. PPARγ signaling is required for mediating EETs protective effects in neonatal cardiomyocytes exposed to LPS. Front. Pharmacol. 2014;5:242. doi: 10.3389/fphar.2014.00242. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.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. doi: 10.1074/jbc.M507787200. [DOI] [PubMed] [Google Scholar]
  • 63.Park S.K., Herrnreiter A., Pfister S.L., Gauthier K.M., Falck B.A., Falck J.R., et al. GPR40 is a low-affinity epoxyeicosatrienoic acid receptor in vascular cells. J. Biol. Chem. 2018;293:10675–10691. doi: 10.1074/jbc.RA117.001297. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Miyamoto J., Mizukure T., Park S.-B., Kishino S., Kimura I., Hirano K., et al. A gut microbial metabolite of linoleic acid, 10-hydroxy-cis-12-octadecenoic acid, ameliorates intestinal epithelial barrier impairment partially via GPR40-MEK-ERK pathway. J. Biol. Chem. 2015;290:2902–2918. doi: 10.1074/jbc.M114.610733. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Zimmer B., Angioni C., Osthues T., Toewe A., Thomas D., Pierre S.C., et al. The oxidized linoleic acid metabolite 12,13-DiHOME mediates thermal hyperalgesia during inflammatory pain. Biochim. Biophys. Acta. 2018;1863:669–678. doi: 10.1016/j.bbalip.2018.03.012. [DOI] [PubMed] [Google Scholar]
  • 66.Schopfer F.J., Khoo N.K.H. Nitro-Fatty acid logistics: formation, biodistribution, signaling, and pharmacology. Trends Endocrinol. Metab. 2019;30:505–519. doi: 10.1016/j.tem.2019.04.009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Hansen H.S., Jensen B., von Wettstein-Knowles P. Apparent in vivo retroconversion of dietary arachidonic to linoleic acid in essential fatty acid-deficient rats. Biochim. Biophys. Acta. 1986;878:284–287. doi: 10.1016/0005-2760(86)90158-x. [DOI] [PubMed] [Google Scholar]
  • 68.Hansen H.S. The essential nature of linoleic acid in mammals. Trends Biochem. Sci. 1986;11:263–265. [Google Scholar]
  • 69.Zheng Y., Yin H., Boeglin W.E., Elias P.M., Crumrine D., Beier D.R., et al. Lipoxygenases mediate the effect of essential fatty acid in skin barrier formation: a proposed role in releasing omega-hydroxyceramide for construction of the corneocyte lipid envelope. J. Biol. Chem. 2011;286:24046–24056. doi: 10.1074/jbc.M111.251496. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 70.Moran J.L., Qiu H., Turbe-Doan A., Yun Y., Boeglin W.E., Brash A.R., et al. A mouse mutation in the 12R-lipoxygenase, Alox12b, disrupts formation of the epidermal permeability barrier. J. Invest. Dermatol. 2007;127:1893–1897. doi: 10.1038/sj.jid.5700825. [DOI] [PubMed] [Google Scholar]
  • 71.Epp N., Fürstenberger G., Müller K., de Juanes S., Leitges M., Hausser I., et al. 12R-lipoxygenase deficiency disrupts epidermal barrier function. J. Cell Biol. 2007;177:173–182. doi: 10.1083/jcb.200612116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Krieg P., Rosenberger S., de Juanes S., Latzko S., Hou J., Dick A., et al. Aloxe3 knockout mice reveal a function of epidermal lipoxygenase-3 as hepoxilin synthase and its pivotal role in barrier formation. J. Invest. Dermatol. 2013;133:172–180. doi: 10.1038/jid.2012.250. [DOI] [PubMed] [Google Scholar]
  • 73.Takeichi T., Hirabayashi T., Miyasaka Y., Kawamoto A., Okuno Y., Taguchi S., et al. SDR9C7 catalyzes critical dehydrogenation of acylceramides for skin barrier formation. J. Clin. Invest. 2020;130:890–903. doi: 10.1172/JCI130675. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Ohno Y., Nakamura T., Iwasaki T., Katsuyama A., Ichikawa S., Kihara A. Determining the structure of protein-bound ceramides, essential lipids for skin barrier function. iScience. 2023;26 doi: 10.1016/j.isci.2023.108248. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 75.Tyrrell V.J., Ali F., Boeglin W.E., Andrews R., Burston J., Birchall J.C., et al. Lipidomic and transcriptional analysis of the linoleoyl-omega-hydroxyceramide biosynthetic pathway in human psoriatic lesions. J. Lipid Res. 2021;62 doi: 10.1016/j.jlr.2021.100094. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 76.Quaranta A., Zöhrer B., Revol-Cavalier J., Benkestock K., Balas L., Oger C., et al. Development of a chiral supercritical fluid chromatography-tandem mass spectrometry and reversed-phase liquid chromatography-tandem mass spectrometry platform for the quantitative metabolic profiling of octadecanoid oxylipins. Anal. Chem. 2022;94:14618–14626. doi: 10.1021/acs.analchem.2c02601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Revol-Cavalier J., Quaranta A., Newman J.W., Brash A.R., Hamberg M., Wheelock C.E. The octadecanoids: synthesis and bioactivity of 18-Carbon oxygenated fatty acids in mammals, bacteria, and fungi. Chem. Rev. 2025;125:1–90. doi: 10.1021/acs.chemrev.3c00520. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78.Quaranta A., Zöhrer B., Revol-Cavalier J., Benkestock K., Balas L., Oger C., et al. Development of a chiral supercritical fluid chromatography–tandem mass spectrometry and reversed-phase liquid chromatography–tandem mass spectrometry platform for the quantitative metabolic profiling of octadecanoid oxylipins. Anal. Chem. 2022;94:14618–14626. doi: 10.1021/acs.analchem.2c02601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79.Salamin O., Wheelock C.E. In: Clinical Metabolomics: Methods and Protocols. Giera M., Sánchez-López E., editors. Springer US; New York, NY: 2025. Quantification of octadecanoids in human plasma using chiral supercritical fluid chromatography-tandem mass spectrometry; pp. 315–339. [DOI] [PubMed] [Google Scholar]
  • 80.Smith M.J., Nie M., Adner M., Säfholm J., Wheelock C.E. Development of a desorption electrospray Ionization–Multiple-Reaction-Monitoring mass spectrometry (DESI-MRM) workflow for spatially mapping oxylipins in pulmonary tissue. Anal. Chem. 2024;96:17950–17959. doi: 10.1021/acs.analchem.4c02350. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.O'Donnell V.B., Aldrovandi M., Murphy R.C., Krönke G. Enzymatically oxidized phospholipids assume center stage as essential regulators of innate immunity and cell death. Sci. Signal. 2019;12:eaau2293. doi: 10.1126/scisignal.aau2293. [DOI] [PubMed] [Google Scholar]
  • 82.Protty M.B., Tyrrell V.J., Hajeyah A.A., Morgan B., Costa D., Li Y., et al. Aspirin modulates generation of procoagulant phospholipids in cardiovascular disease, by regulating LPCAT3. J. Lipid Res. 2024;66 doi: 10.1016/j.jlr.2024.100727. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83.Maskrey B.H., Bermúdez-Fajardo A., Morgan A.H., Stewart-Jones E., Dioszeghy V., Taylor G.W., et al. Activated platelets and monocytes generate four hydroxyphosphatidylethanolamines via lipoxygenase. J. Biol. Chem. 2007;282:20151–20163. doi: 10.1074/jbc.M611776200. [DOI] [PubMed] [Google Scholar]
  • 84.Morgan A.H., Dioszeghy V., Maskrey B.H., Thomas C.P., Clark S.R., Mathie S.A., et al. Phosphatidylethanolamine-esterified eicosanoids in the mouse: tissue localization and inflammation-dependent formation in Th-2 disease. J. Biol. Chem. 2009;284:21185–21191. doi: 10.1074/jbc.M109.021634. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Liu X., Sims H.F., Jenkins C.M., Guan S., Dilthey B.G., Gross R.W. 12-LOX catalyzes the oxidation of 2-arachidonoyl-lysolipids in platelets generating eicosanoid-lysolipids that are attenuated by iPLA(2)γ knockout. J. Biol. Chem. 2020;295:5307–5320. doi: 10.1074/jbc.RA119.012296. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 86.Lauder S.N., Allen-Redpath K., Slatter D.A., Aldrovandi M., O'Connor A., Farewell D., et al. Networks of enzymatically oxidized membrane lipids support calcium-dependent coagulation factor binding to maintain hemostasis. Sci. Signal. 2017;10 doi: 10.1126/scisignal.aan2787. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Uderhardt S., Ackermann J.A., Fillep T., Hammond V.J., Willeit J., Santer P., et al. Enzymatic lipid oxidation by eosinophils propagates coagulation, hemostasis, and thrombotic disease. J. Exp. Med. 2017;214:2121–2138. doi: 10.1084/jem.20161070. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Allen-Redpath K., Aldrovandi M., Lauder S.N., Gketsopoulou A., Tyrrell V.J., Slatter D.A., et al. Phospholipid membranes drive abdominal aortic aneurysm development through stimulating coagulation factor activity. Proc. Natl. Acad. Sci. U. S. A. 2019;116:8038–8047. doi: 10.1073/pnas.1814409116. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Slatter D.A., Percy C.L., Allen-Redpath K., Gajsiewicz J.M., Brooks N.J., Clayton A., et al. Enzymatically oxidized phospholipids restore thrombin generation in coagulation factor deficiencies. JCI Insight. 2019;3 doi: 10.1172/jci.insight.98459. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90.Costa D., Hughes S., Jenkins R., Cardus Figueras A., Protty M., Tyrrell V., et al. Interleukin-6 elevates thrombosis via pro-coagulant phospholipids from platelet 12-lipoxygenase in rheumatoid arthritis. bioRxiv. 2025 doi: 10.1101/2025.03.26.645440. [preprint] [DOI] [Google Scholar]
  • 91.Kozak K.R., Crews B.C., Morrow J.D., Wang L.H., Ma Y.H., Weinander R., et al. Metabolism of the endocannabinoids, 2-arachidonylglycerol and anandamide, into prostaglandin, thromboxane, and prostacyclin glycerol esters and ethanolamides. J. Biol. Chem. 2002;277:44877–44885. doi: 10.1074/jbc.M206788200. [DOI] [PubMed] [Google Scholar]
  • 92.Nirodi C.S., Crews B.C., Kozak K.R., Morrow J.D., Marnett L.J. The glyceryl ester of prostaglandin E2 mobilizes calcium and activates signal transduction in RAW264. 7 cells. Proc. Natl. Acad. Sci. U. S. A. 2004;101:1840–1845. doi: 10.1073/pnas.0303950101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Alhouayek M., Masquelier J., Cani P.D., Lambert D.M., Muccioli G.G. Implication of the anti-inflammatory bioactive lipid prostaglandin D2-glycerol ester in the control of macrophage activation and inflammation by ABHD6. Proc. Natl. Acad. Sci. U. S. A. 2013;110:17558–17563. doi: 10.1073/pnas.1314017110. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 94.Shearer G.C., Newman J.W. Lipoprotein lipase releases esterified oxylipins from very low-density lipoproteins. Prostaglandins Leukot. Essent. Fatty Acids. 2008;79:215–222. doi: 10.1016/j.plefa.2008.09.023. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95.Ostermann A.I., Koch E., Rund K.M., Kutzner L., Mainka M., Schebb N.H. Targeting esterified oxylipins by LC–MS - effect of sample preparation on oxylipin pattern. Prostaglandins Other Lipid Mediat. 2020;146 doi: 10.1016/j.prostaglandins.2019.106384. [DOI] [PubMed] [Google Scholar]
  • 96.Shearer G.C., Newman J.W. Impact of circulating esterified eicosanoids and other oxylipins on endothelial function. Curr. Atheroscler. Rep. 2009;11:403–410. doi: 10.1007/s11883-009-0061-3. [DOI] [PubMed] [Google Scholar]
  • 97.Schebb N.H., Kampschulte N., Hagn G., Plitzko K., Meckelmann S.W., Ghosh S., et al. Technical recommendations for analyzing oxylipins by liquid chromatography–mass spectrometry. Sci. Signal. 2025;18:eadw1245. doi: 10.1126/scisignal.adw1245. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Meng H., Sengupta A., Ricciotti E., Mrčela A., Mathew D., Mazaleuskaya L.L., et al. Deep phenotyping of the lipidomic response in COVID-19 and non-COVID-19 sepsis. Clin. Transl. Med. 2023;13 doi: 10.1002/ctm2.1440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Fenyves B.G., Mehta A., Kays K.R., Beakes C., Margolin J., Goldberg M.B., et al. Plasma P-selectin is an early marker of thromboembolism in COVID-19. Am. J. Hematol. 2021;96:E468–e471. doi: 10.1002/ajh.26372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Milne G.L., Nogueira M.S., Gao B., Sanchez S.C., Amin W., Thomas S., et al. Identification of novel F(2)-isoprostane metabolites by specific UDP-glucuronosyltransferases. Redox Biol. 2024;70 doi: 10.1016/j.redox.2023.103020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101.Nogueira M.S., Sanchez S.C., Milne C.E., Amin W., Thomas S.J., Milne G.L. Resolvins D5 and D1 undergo phase II metabolism by uridine 5'-diphospho-glucuronosyltransferases. Prostaglandins Other Lipid Mediat. 2024;174 doi: 10.1016/j.prostaglandins.2024.106870. [DOI] [PubMed] [Google Scholar]

Articles from The Journal of Biological Chemistry are provided here courtesy of American Society for Biochemistry and Molecular Biology

RESOURCES