Eicosanoid

Last updated
Pathways in biosynthesis of eicosanoids from arachidonic acid: there are parallel paths from EPA & DGLA. Eicosanoid synthesis.svg
Pathways in biosynthesis of eicosanoids from arachidonic acid: there are parallel paths from EPA & DGLA.

Eicosanoids are signaling molecules made by the enzymatic or non-enzymatic oxidation of arachidonic acid or other polyunsaturated fatty acids (PUFAs) that are, similar to arachidonic acid, around 20 carbon units in length. Eicosanoids are a sub-category of oxylipins, i.e. oxidized fatty acids of diverse carbon units in length, and are distinguished from other oxylipins by their overwhelming importance as cell signaling molecules. Eicosanoids function in diverse physiological systems and pathological processes such as: mounting or inhibiting inflammation, allergy, fever and other immune responses; regulating the abortion of pregnancy and normal childbirth; contributing to the perception of pain; regulating cell growth; controlling blood pressure; and modulating the regional flow of blood to tissues. In performing these roles, eicosanoids most often act as autocrine signaling agents to impact their cells of origin or as paracrine signaling agents to impact cells in the proximity of their cells of origin. Some eicosanoids, such as prostaglandins, may also have endocrine roles as hormones to influence the function of distant cells. [1] [2]

Contents

There are multiple subfamilies of eicosanoids, including most prominently the prostaglandins, thromboxanes, leukotrienes, lipoxins, resolvins, and eoxins. [1] For each subfamily, there is the potential to have at least 4 separate series of metabolites, two series derived from ω-6 PUFAs (arachidonic and dihomo-gamma-linolenic acids), one series derived from the ω-3 PUFA (eicosapentaenoic acid), and one series derived from the ω-9 PUFA (mead acid). This subfamily distinction is important. Mammals, including humans, are unable to convert ω-6 into ω-3 PUFA. In consequence, tissue levels of the ω-6 and ω-3 PUFAs and their corresponding eicosanoid metabolites link directly to the amount of dietary ω-6 versus ω-3 PUFAs consumed. [3] Since certain of the ω-6 and ω-3 PUFA series of metabolites have almost diametrically opposing physiological and pathological activities, it has often been suggested that the deleterious consequences associated with the consumption of ω-6 PUFA-rich diets reflects excessive production and activities of ω-6 PUFA-derived eicosanoids, while the beneficial effects associated with the consumption of ω-3 PUFA-rich diets reflect the excessive production and activities of ω-3 PUFA-derived eicosanoids. [4] [5] [6] [7] In this view, the opposing effects of ω-6 PUFA-derived and ω-3 PUFA-derived eicosanoids on key target cells underlie the detrimental and beneficial effects of ω-6 and ω-3 PUFA-rich diets on inflammation and allergy reactions, atherosclerosis, hypertension, cancer growth, and a host of other processes.

Nomenclature

Fatty acid sources

"Eicosanoid" (from Greek eicosa-  'twenty') is the collective term [8] for straight-chain PUFAs (polyunsaturated fatty acids) of 20 carbon units in length that have been metabolized or otherwise converted to oxygen-containing products. The PUFA precursors to the eicosanoids include:

Abbreviation

A particular eicosanoid is denoted by a four-character abbreviation, composed of:

The stereochemistry of the eicosanoid products formed may differ among the pathways. For prostaglandins, this is often indicated by Greek letters (e.g. PGF versus PGF). For hydroperoxy and hydroxy eicosanoids an S or R designates the chirality of their substituents (e.g. 5S-hydroxy-eicosateteraenoic acid [also termed 5(S)-, 5S-hydroxy-, and 5(S)-hydroxy-eicosatetraenoic acid] is given the trivial names of 5S-HETE, 5(S)-HETE, 5S-HETE, or 5(S)-HETE). Since eicosanoid-forming enzymes commonly make S isomer products either with marked preference or essentially exclusively, the use of S/R designations has often been dropped (e.g. 5S-HETE is 5-HETE). Nonetheless, certain eicosanoid-forming pathways do form R isomers and their S versus R isomeric products can exhibit dramatically different biological activities. [11] Failing to specify S/R isomers can be misleading. Here, all hydroperoxy and hydroxy substituents have the S configuration unless noted otherwise.

Classic eicosanoids

Current usage limits the term eicosanoid to:

Hydroxyeicosatetraenoic acids, leukotrienes, eoxins and prostanoids are sometimes termed "classic eicosanoids" [20] [21] [22]

Nonclassic eicosanoids

In contrast to the classic eicosanoids, several other classes of PUFA metabolites have been termed 'novel', 'eicosanoid-like' or 'nonclassic eicosanoids'. [23] [24] [25] [26] These included the following classes:

Metabolism of eicosapentaenoic acid to HEPEs, leukotrienes, prostanoids, and epoxyeicosatetraenoic acids as well as the metabolism of dihomo-gamma-linolenic acid to prostanoids and mead acid to 5(S)-hydroxy-6E,8Z,11Z-eicosatrienoic acid (5-HETrE), 5-oxo-6,8,11-eicosatrienoic acid (5-oxo-ETrE), LTA3, and LTC3 involve the same enzymatic pathways that make their arachidonic acid-derived analogs.

Biosynthesis

Eicosanoids typically are not stored within cells but rather synthesized as required. They derive from the fatty acids that make up the cell membrane and nuclear membrane. These fatty acids must be released from their membrane sites and then metabolized initially to products which most often are further metabolized through various pathways to make the large array of products we recognize as bioactive eicosanoids.

Fatty acid mobilization

Eicosanoid biosynthesis begins when a cell is activated by mechanical trauma, ischemia, other physical perturbations, attack by pathogens, or stimuli made by nearby cells, tissues, or pathogens such as chemotactic factors, cytokines, growth factors, and even certain eicosanoids. The activated cells then mobilize enzymes, termed phospholipase A2's (PLA2s), capable of releasing ω-6 and ω-3 fatty acids from membrane storage. These fatty acids are bound in ester linkage to the SN2 position of membrane phospholipids; PLA2s act as esterases to release the fatty acid. There are several classes of PLA2s with type IV cytosolic PLA2s (cPLA2s) appearing to be responsible for releasing the fatty acids under many conditions of cell activation. The cPLA2s act specifically on phospholipids that contain AA, EPA or GPLA at their SN2 position. cPLA2 may also release the lysophospholipid that becomes platelet-activating factor. [30]

Peroxidation and reactive oxygen species

Next, the free fatty acid is oxygenated along any of several pathways; see the Pathways table. The eicosanoid pathways (via lipoxygenase or COX) add molecular oxygen (O2). Although the fatty acid is symmetric, the resulting eicosanoids are chiral; the oxidations proceed with high stereoselectivity (enzymatic oxidations are considered practically stereospecific).

Four families of enzymes initiate or contribute to the initiation of the catalysis of fatty acids to eicosanoids:

Two different enzymes may act in series on a PUFA to form more complex metabolites. For example, ALOX5 acts with ALOX12 or aspirin-treated COX-2 to metabolize arachidonic acid to lipoxins and with cytochrome P450 monooxygenase(s), bacterial cytochrome P450 (in infected tissues), or aspirin-treated COX2 to metabolize eicosapentaenoic acid to the E series resolvins (RvEs) (see Specialized pro-resolving mediators). When this occurs with enzymes located in different cell types and involves the transfer of one enzyme's product to a cell which uses the second enzyme to make the final product it is referred to as transcellular metabolism or transcellular biosynthesis. [33]

The oxidation of lipids is hazardous to cells, particularly when close to the nucleus. There are elaborate mechanisms to prevent unwanted oxidation. COX, the lipoxygenases, and the phospholipases are tightly controlled—there are at least eight proteins activated to coordinate generation of leukotrienes. Several of these exist in multiple isoforms. [7]

Oxidation by either COX or lipoxygenase releases reactive oxygen species (ROS) and the initial products in eicosanoid generation are themselves highly reactive peroxides. LTA4 can form adducts with tissue DNA. Other reactions of lipoxygenases generate cellular damage; murine models implicate 15-lipoxygenase in the pathogenesis of atherosclerosis. [34] [35] The oxidation in eicosanoid generation is compartmentalized; this limits the peroxides' damage. The enzymes that are biosynthetic for eicosanoids (e.g., glutathione-S-transferases, epoxide hydrolases, and carrier proteins) belong to families whose functions are involved largely with cellular detoxification. This suggests that eicosanoid signaling might have evolved from the detoxification of ROS.

The cell must realize some benefit from generating lipid hydroperoxides close-by its nucleus. PGs and LTs may signal or regulate DNA-transcription there; LTB4 is ligand for PPARα. [5] (See diagram at PPAR).

Structures of selected eicosanoids
Prostaglandin E1.svg Thromboxane A2 acsv.svg Leukotriene B4.svg
Prostaglandin E1. The 5-member ring is characteristic of the class.Thromboxane A2. Oxygens
have moved into the ring.
Leukotriene B4. Note the 3 conjugated double bonds.
Prostaglandin I2.png Leukotriene E4.svg
Prostacyclin I2. The second ring distinguishes it from the prostaglandins.Leukotriene E4, an example of a cysteinyl leukotriene.

Prostanoid pathways

Both COX1 and COX2 (also termed prostaglandin-endoperoxide synthase-1 (PTGS1) and PTGS2, respectively) metabolize arachidonic acid by adding molecular O2 between carbons 9 and 11 to form an endoperoxide bridge between these two carbons, adding molecular O2 to carbon 15 to yield a 15-hydroperoxy product, creating a carbon-carbon bond between carbons 8 and 12 to create a cyclopentane ring in the middle of the fatty acid, and in the process making PGG2, a product that has two fewer double bonds than arachidonic acid. The 15-hydroperoxy residue of PGG2 is then reduced to a 15-hydroxyl residue thereby forming PGH2. PGH2 is the parent prostanoid to all other prostanoids. It is metabolized by (see diagram in Prostanoid): a) The prostaglandin E synthase pathway in which any one of three isozymes, PTGES, PTGES2, or PTGES3, convert PGH2 to PGE2 (subsequent products of this pathway include PGA2 and PGB2 (see Prostanoid § Biosynthesis of prostaglandins); b) PGF synthase which converts PGH2 to PGF2α; c) Prostaglandin D2 synthase which converts PGH2 to PGD2 (subsequent products in this pathway include 15-dPGJ2 (see Cyclopentenone prostaglandin); d) Thromboxane synthase which converts PGH2 to TXA2 (subsequent products in this pathway include TXB2); and e) Prostacyclin synthase which converts PGH2 to PGI2 (subsequent products in this pathway include 6-keto-PGFα. [36] [37] These pathways have been shown or in some cases presumed to metabolize eicosapentaenoic acid to eicosanoid analogs of the sited products that have three rather than two double bonds and therefore contain the number 3 in place of 2 attached to their names (e.g. PGE3 instead of PGE2). [38]

The PGE2, PGE1, and PGD2 products formed in the pathways just cited can undergo a spontaneous dehydration reaction to form PGA2, PGA1, and PGJ2, respectively; PGJ2 may then undergo a spontaneous isomerization followed by a dehydration reaction to form in series Δ12-PGJ2 and 15-deoxy-Δ12,14-PGJ2. [39]

PGH2 has a 5-carbon ring bridged by molecular oxygen. Its derived PGS have lost this oxygen bridge and contain a single, unsaturated 5-carbon ring with the exception of thromboxane A2 which possesses a 6-member ring consisting of one oxygen and 5 carbon atoms. The 5-carbon ring of prostacyclin is conjoined to a second ring consisting of 4 carbon and one oxygen atom. And, the 5 member ring of the cyclopentenone prostaglandins possesses an unsaturated bond in a conjugated system with a carbonyl group that causes these PGs to form bonds with a diverse range of bioactive proteins (for more see the diagrams at Prostanoid).

Hydroxyeicosatetraenoate (HETE) and leukotriene (LT) pathways

The enzyme 5-lipoxygenase (5-LO or ALOX5) converts arachidonic acid into 5-hydroperoxyeicosatetraenoic acid (5-HPETE), which may be released and rapidly reduced to 5-hydroxyeicosatetraenoic acid (5-HETE) by ubiquitous cellular glutathione-dependent peroxidases. [40] Alternately, ALOX5 uses its LTA synthase activity to act convert 5-HPETE to leukotriene A4 (LTA4). LTA4 is then metabolized either to LTB4 by leukotriene A4 hydrolase or leukotriene C4 (LTC4) by either LTC4 synthase or microsomal glutathione S-transferase 2 (MGST2). Either of the latter two enzymes act to attach the sulfur of cysteine's thio- (i.e. SH) group in the tripeptide glutamate-cysteine-glycine to carbon 6 of LTA4 thereby forming LTC4. After release from its parent cell, the glutamate and glycine residues of LTC4 are removed step-wise by gamma-glutamyltransferase and a dipeptidase to form sequentially LTD4 and LTE4. [41] [42] The decision to form LTB4 versus LTC4 depends on the relative content of LTA4 hydrolase versus LTC4 synthase (or glutathione S-transferase in cells; eosinophils, mast cells, and alveolar macrophages possess relatively high levels of LTC4 synthase and accordingly form LTC4 rather than or to a far greater extent than LTB4. 5-LOX may also work in series with cytochrome P450 oxygenases or aspirin-treated COX2 to form Resolvins RvE1, RvE2, and 18S-RvE1 (see Specialized pro-resolving mediators § EPA-derived resolvins).

The enzyme arachidonate 12-lipoxygenase (12-LO or ALOX12) metabolizes arachidonic acid to the S stereoisomer of 12-hydroperoxyeicosatetraenoic acid (12-HPETE) which is rapidly reduced by cellular peroxidases to the S stereoisomer of 12-hydroxyeicosatetraenoic acid (12-HETE) or further metabolized to hepoxilins (Hx) such as HxA3 and HxB. [43] [44]

The enzymes 15-lipoxygenase-1 (15-LO-1 or ALOX15) and 15-lipoxygenase-2 (15-LO-2, ALOX15B) metabolize arachidonic acid to the S stereoisomer of 15-hydroperoxyeicosatetraenoic acid (15(S)-HPETE) which is rapidly reduced by cellular peroxidases to the S stereoisomer of 15-hydroxyeicosatetraenoic acid (15(S)-HETE). [45] [46] The 15-lipoxygenases (particularly ALOX15) may also act in series with 5-lipoxygenase, 12-lipoxygenase, or aspirin-treated COX2 to form the lipoxins and epi-lipoxins or with P450 oxygenases or aspirin-treated COX2 to form Resolvin E3 (see Specialized pro-resolving mediators § EPA-derived resolvins).

A subset of cytochrome P450 (CYP450) microsome-bound ω-hydroxylases metabolize arachidonic acid to 20-hydroxyeicosatetraenoic acid (20-HETE) and 19-hydroxyeicosatetraenoic acid by an omega oxidation reaction. [47]

Epoxyeicosanoid pathway

The human cytochrome P450 (CYP) epoxygenases, CYP1A1, CYP1A2, CYP2C8, CYP2C9, CYP2C18, CYP2C19, CYP2E1, CYP2J2, and CYP2S1 metabolize arachidonic acid to the non-classic epoxyeicosatrienoic acids (EETs) by converting one of the fatty acid's double bonds to its epoxide to form one or more of the following EETs, 14,15-ETE, 11,12-EET, 8,9-ETE, and 4,5-ETE. [48] [49] 14,15-EET and 11,12-EET are the major EETs produced by mammalian, including human, tissues. [49] [50] [51] [52] [53] The same CYPs but also CYP4A1, CYP4F8, and CYP4F12 metabolize eicosapentaenoic acid to five epoxide epoxyeicosatetraenoic acids (EEQs) viz., 17,18-EEQ, 14,15-EEQ, 11,12-EEQ. 8,9-EEQ, and 5,6-EEQ. [54]

Function, pharmacology, and clinical significance

The following table lists a sampling of the major eicosanoids that possess clinically relevant biological activity, the cellular receptors (see Cell surface receptor) that they stimulate or, where noted, antagonize to attain this activity, some of the major functions which they regulate (either promote or inhibit) in humans and mouse models, and some of their relevancies to human diseases.

EicosanoidTargeted receptorsFunctions regulatedClinical relevancy
PGE2 PTGER1, PTGER2, PTGER3, PTGER4 inflammation; fever; pain perception; allodynia; parturition NSAIDs inhibit its production to reduce inflammation, fever, and pain; used to promote labor in childbirth; an abortifacient [37] [55] [56]
PGD2 Prostaglandin DP1 receptor 1, Prostaglandin DP2 receptor allergy reactions; allodynia; hair growthNSAIDs may target it to inhibit allodynia and male-pattern hair loss [37] [57] [58] [59] [60]
TXA2 Thromboxane receptor α and βblood platelet aggregation; blood clotting; allergic reactionsNSAIDs inhibit its production to reduce incidence of strokes and heart attacks [37] [61]
PGI2 Prostacyclin receptor platelet aggregation, vascular smooth muscle contractionPGI2 analogs used to treat vascular disorders like pulmonary hypertension, Raynaud's syndrome, and Buerger's disease [62] [63] [64]
15-d-Δ12,14-PGJ2 PPARγ, Prostaglandin DP2 receptor inhibits inflammation and cell growthinhibits diverse inflammatory responses in animal models; structural model for developing anti-inflammatory agents [12] [59] [60]
20-HETE ?vasoconstriction, inhibits plateletsinactivating mutations in the 20-HETE-forming enzyme, CYP2U1, associated with hereditary spastic paraplegia [65]
5-Oxo-ETE OXER1 chemotactic factor for and activator of eosinophilsstudies needed to determine if inhibiting its production or action inhibits allergic reactons [32]
LTB4 LTB4R, LTB4R2 chemotactic factor for and activator of leukocytes; inflammationstudies to date shown no clear benefits of LTB4 receptor antagonists for human inflammatory diseases [66] [67] [68]
LTC4 CYSLTR1, CYSLTR2, GPR17 vascular permeability; vascular smooth muscle contraction; allergyantagonists of CYSLTR1 used in asthma as well as other allergic and allergic-like reactions [69] [70]
LTD4 CYSLTR1, CYSLTR2, GPR17 vascular permeability; vascular smooth muscle contraction; allergyantagonists of CYSLTR1 used in asthma as well as other allergic and allergic-like reactions [66]
LTE4 GPR99 increases vascular permeability and airway mucin secretionthought to contribute to asthma as well as other allergic and allergic-like reactions [71]
LxA4 FPR2 inhibits functions of pro-inflammatory cellsSpecialized pro-resolving mediators class of inflammatory reaction suppressors [72] [73]
LxB4 FPR2, GPR32, AHR inhibits functions of pro-inflammatory cellsSpecialized pro-resolving mediators class of inflammatory reaction suppressors [72] [73]
RvE1 CMKLR1, inhibits BLT, TRPV1, TRPV3, NMDAR, TNFR inhibits functions of pro-inflammatory cellsSpecialized pro-resolving mediators class of inflammatory reaction suppressors; also suppresses pain perception [74] [75] [76]
RvE2 CMKLR1, receptor antagonist of BLT inhibits functions of pro-inflammatory cellsSpecialized pro-resolving mediators class of inflammatory reaction suppressors [72] [73] [76] [77]
14,15-EET ? vasodilation, inhibits platelets and pro-inflammatory cellsrole(s) in human disease not yet proven [78] [79]

Prostanoids

Many of the prostanoids are known to mediate local symptoms of inflammation: vasoconstriction or vasodilation, coagulation, pain, and fever. Inhibition of COX-1 and/or the inducible COX-2 isoforms is the hallmark of NSAIDs (non-steroidal anti-inflammatory drugs), such as aspirin. Prostanoids also activate the PPARγ members of the steroid/thyroid family of nuclear hormone receptors, and directly influence gene transcription. [80] Prostanoids have numerous other relevancies to clinical medicine as evidence by their use, the use of their more stable pharmacological analogs, of the use of their receptor antagonists as indicated in the following chart.

MedicineTypeMedical condition or useMedicineTypeMedical condition or use
Alprostadil PGE1 Erectile dysfunction, maintaining a patent ductus arteriosus in the fetus Beraprost PGI1 analog Pulmonary hypertension, avoiding reperfusion injury
Bimatoprost PGF2α analog Glaucoma, ocular hypertension Carboprost PGF2α analogLabor induction, abortifacient in early pregnancy
Dinoprostone PGE2Labor induction Iloprost PGI2 analog Pulmonary artery hypertension
Latanoprost PGF2α analog Glaucoma, ocular hypertension Misoprostol PGE1 analog Stomach ulcers labor induction, abortifacient
Travoprost PGF2α analog Glaucoma, ocular hypertension U46619 Longer lived TX analog Longer lived TX analogResearch only

Cyclopentenone prostaglandins

PGA1, PGA2, PGJ2, Δ12-PGJ2, and 15-deox-Δ12,14-PGJ2 exhibit a wide range of anti-inflammatory and inflammation-resolving actions in diverse animal models. [39] They therefore appear to function in a manner similar to specialized pro-resolving mediators although one of their mechanisms of action, forming covalent bonds with key signaling proteins, differs from those of the specialized pro-resolving mediators.

HETEs and oxo-ETEs

As indicated in their individual Wikipedia pages, 5-hydroxyeicosatetraenoic acid (which, like 5-oxo-eicosatetraenoic acid, acts through the OXER1 receptor), 5-oxo-eicosatetraenoic acid, 12-hydroxyeicosatetraenoic acid, 15-hydroxyeicosatetraenoic acid, and 20-hydroxyeicosatetraenoic acid show numerous activities in animal and human cells as well as in animal models that are related to, for example, inflammation, allergic reactions, cancer cell growth, blood flow to tissues, and/or blood pressure. However, their function and relevancy to human physiology and pathology have not as yet been shown.

Leukotrienes

The three cysteinyl leukotrienes, LTC4, LTD4, and LTE4, are potent bronchoconstrictors, increasers of vascular permeability in postcapillary venules, and stimulators of mucus secretion that are released from the lung tissue of asthmatic subjects exposed to specific allergens. They play a pathophysiological role in diverse types of immediate hypersensitivity reactions. [81] Drugs that block their activation of the CYSLTR1 receptor viz., montelukast, zafirlukast, and pranlukast, are used clinically as maintenance treatment for allergen-induced asthma and rhinitis; nonsteroidal anti-inflammatory drug-induced asthma and rhinitis (see aspirin-exacerbated respiratory disease); exercise- and cold-air induced asthma (see Exercise-induced bronchoconstriction); and childhood sleep apnea due to adenotonsillar hypertrophy (see Acquired non-inflammatory myopathy § Diet and Trauma Induced Myopathy). [82] [83] [84] [85] When combined with antihistamine drug therapy, they also appear useful for treating urticarial diseases such as hives. [86]

Lipoxins and epi-lipoxins

LxA4, LxB4, 15-epi-LxA4, and 15-epi-LXB4, like other members of the specialized pro-resolving mediators class of eicosanoids, possess anti-inflammatory and inflammation resolving activity. In a randomized controlled trial, AT-LXA4 and a comparatively stable analog of LXB4, 15R/S-methyl-LXB4, reduced the severity of eczema in a study of 60 infants [87] and, in another study, inhaled LXA4 decreased LTC4-initiated bronchoprovocation in patients with asthma. [88]

Eoxins

The eoxins (EXC4, EXD4, EXE5) are newly described. They stimulate vascular permeability in an ex vivo human vascular endothelial model system, [89] and in a small study of 32 volunteers EXC4 production by eosinophils isolated from severe and aspirin-intolerant asthmatics was greater than that from healthy volunteers and mild asthmatic patients; these findings have been suggested to indicate that the eoxins have pro-inflammatory actions and therefore potentially involved in various allergic reactions. [90] Production of eoxins by Reed-Sternburg cells has also led to suggestion that they are involve in Hodgkins disease. [91] However, the clinical significance of eoxins has not yet been demonstrated.

Resolvin metabolites of eicosapentaenoic acid

RvE1, 18S-RvE1, RvE2, and RvE3, like other members of the specialized pro-resolving mediators) class of eicosanoids, possess anti-inflammatory and inflammation resolving activity. A synthetic analog of RvE1 is in clinical phase III testing (see Phases of clinical research) for the treatment of the inflammation-based dry eye syndrome; along with this study, other clinical trials (NCT01639846, NCT01675570, NCT00799552 and NCT02329743) using an RvE1 analogue to treat various ocular conditions are underway. [88] RvE1 is also in clinical development studies for the treatment of neurodegenerative diseases and hearing loss. [92]

Other metabolites of eicosapentaenoic acid

The metabolites of eicosapentaenoic acid that are analogs of their arachidonic acid-derived prostanoid, HETE, and LT counterparts include: the 3-series prostanoids (e.g. PGE3, PGD3, PGF3α, PGI3, and TXA3), the hydroxyeicosapentaenoic acids (e.g. 5-HEPE, 12-HEPE, 15-HEPE, and 20-HEPE), and the 5-series LTs (e.g. LTB5, LTC5, LTD5, and LTE5). Many of the 3-series prostanoids, the hydroxyeicosapentaenoic acids, and the 5-series LT have been shown or thought to be weaker stimulators of their target cells and tissues than their arachidonic acid-derived analogs. They are proposed to reduce the actions of their arachidonate-derived analogs by replacing their production with weaker analogs. [93] [94] Eicosapentaenoic acid-derived counterparts of the Eoxins have not been described.

Epoxyeicosanoids

The epoxy eicosatrienoic acids (or EETs)—and, presumably, the epoxy eicosatetraenoic acids—have vasodilating actions on heart, kidney, and other blood vessels as well as on the kidney's reabsorption of sodium and water, and act to reduce blood pressure and ischemic and other injuries to the heart, brain, and other tissues; they may also act to reduce inflammation, promote the growth and metastasis of certain tumors, promote the growth of new blood vessels, in the central nervous system, regulate the release of neuropeptide hormones, and in the peripheral nervous system inhibit or reduce pain perception. [48] [49] [51]

The ω-3 and ω-6 series

The reduction in AA-derived eicosanoids and the diminished activity of the alternative products generated from ω-3 fatty acids serve as the foundation for explaining some of the beneficial effects of greater ω-3 intake.

Kevin Fritsche, Fatty Acids as Modulators of the Immune Response [95]

Arachidonic acid (AA; 20:4 ω-6) sits at the head of the "arachidonic acid cascade" – more than twenty eicosanoid-mediated signaling paths controlling a wide array of cellular functions, especially those regulating inflammation, immunity, and the central nervous system. [6]

In the inflammatory response, two other groups of dietary fatty acids form cascades that parallel and compete with the arachidonic acid cascade. EPA (20:5 ω-3) provides the most important competing cascade. DGLA (20:3 ω-6) provides a third, less prominent cascade. These two parallel cascades soften the inflammatory effects of AA and its products. Low dietary intake of these less-inflammatory fatty acids, especially the ω-3s, has been linked to several inflammation-related diseases, and perhaps some mental illnesses.

The U.S. National Institutes of Health and the National Library of Medicine state that there is 'A' level evidence that increased dietary ω-3 improves outcomes in hypertriglyceridemia, secondary cardiovascular disease prevention, and hypertension. There is 'B' level evidence ('good scientific evidence') for increased dietary ω-3 in primary prevention of cardiovascular disease, rheumatoid arthritis, and protection from ciclosporin toxicity in organ transplant patients. They also note more preliminary evidence showing that dietary ω-3 can ease symptoms in several psychiatric disorders. [96]

Besides the influence on eicosanoids, dietary polyunsaturated fats modulate immune response through three other molecular mechanisms. They (a) alter membrane composition and function, including the composition of lipid rafts; (b) change cytokine biosynthesis; and (c) directly activate gene transcription. [95] Of these, the action on eicosanoids is the best explored.

Mechanisms of ω-3 action

EFA sources: Essential fatty acid production and metabolism to form eicosanoids. At each step, the o-3 and o-6 cascades compete for the enzymes. EFA to Eicosanoids.svg
EFA sources: Essential fatty acid production and metabolism to form eicosanoids. At each step, the ω-3 and ω-6 cascades compete for the enzymes.

In general, the eicosanoids derived from AA promote inflammation, and those from EPA and from GLA (via DGLA) are less inflammatory, or inactive, or even anti-inflammatory and pro-resolving.

The figure shows the ω-3 and -6 synthesis chains, along with the major eicosanoids from AA, EPA, and DGLA.

Dietary ω-3 and GLA counter the inflammatory effects of AA's eicosanoids in three ways, along the eicosanoid pathways:

  • Displacement—Dietary ω-3 decreases tissue concentrations of AA, so there is less to form ω-6 eicosanoids.
  • Competitive inhibition—DGLA and EPA compete with AA for access to the cyclooxygenase and lipoxygenase enzymes. So the presence of DGLA and EPA in tissues lowers the output of AA's eicosanoids.
  • Counteraction—Some DGLA and EPA derived eicosanoids counteract their AA derived counterparts.

Role in inflammation

Since antiquity, the cardinal signs of inflammation have been known as: calor (warmth), dolor (pain), tumor (swelling), and rubor (redness). The eicosanoids are involved with each of these signs.

Redness —An insect's sting will trigger the classic inflammatory response. Short acting vasoconstrictors — TXA2 — are released quickly after the injury. The site may momentarily turn pale. Then TXA2 mediates the release of the vasodilators PGE2 and LTB4. The blood vessels engorge and the injury reddens.
Swelling —LTB4 makes the blood vessels more permeable. Plasma leaks out into the connective tissues, and they swell. The process also loses pro-inflammatory cytokines.
Pain —The cytokines increase COX-2 activity. This elevates levels of PGE2, sensitizing pain neurons.
Heat —PGE2 is also a potent pyretic agent. Aspirin and NSAIDS—drugs that block the COX pathways and stop prostanoid synthesis—limit fever or the heat of localized inflammation.

History

In 1930, gynecologist Raphael Kurzrok and pharmacologist Charles Leib characterized prostaglandin as a component of semen. Between 1929 and 1932, Burr and Burr showed that restricting fat from animal's diets led to a deficiency disease, and first described the essential fatty acids. [97] In 1935, von Euler identified prostaglandin. In 1964, Bergström and Samuelsson linked these observations when they showed that the "classical" eicosanoids were derived from arachidonic acid, which had earlier been considered to be one of the essential fatty acids. [98] In 1971, Vane showed that aspirin and similar drugs inhibit prostaglandin synthesis. [99] Von Euler received the Nobel Prize in medicine in 1970, which Samuelsson, Vane, and Bergström also received in 1982. E. J. Corey received it in chemistry in 1990 largely for his synthesis of prostaglandins.

See also

Related Research Articles

<span class="mw-page-title-main">Arachidonic acid</span> Fatty acid used metabolically in many organisms

Arachidonic acid is a polyunsaturated omega-6 fatty acid 20:4(ω-6), or 20:4(5,8,11,14). It is structurally related to the saturated arachidic acid found in cupuaçu butter. Its name derives from the ancient Greek neologism arachis (peanut), but peanut oil does not contain any arachidonic acid.

<span class="mw-page-title-main">Leukotriene</span> Class of inflammation mediator molecules

Leukotrienes are a family of eicosanoid inflammatory mediators produced in leukocytes by the oxidation of arachidonic acid (AA) and the essential fatty acid eicosapentaenoic acid (EPA) by the enzyme arachidonate 5-lipoxygenase.

<span class="mw-page-title-main">Lipoxygenase</span>

Lipoxygenases (LOX) are a family of (non-heme) iron-containing enzymes, more specifically oxidative enzymes, most of which catalyze the dioxygenation of polyunsaturated fatty acids in lipids containing a cis,cis-1,4-pentadiene into cell signaling agents that serve diverse roles as autocrine signals that regulate the function of their parent cells, paracrine signals that regulate the function of nearby cells, and endocrine signals that regulate the function of distant cells.

<span class="mw-page-title-main">Hepoxilin</span> Chemical compound

Hepoxilins (Hx) are a set of epoxyalcohol metabolites of polyunsaturated fatty acids (PUFA), i.e. they possess both an epoxide and an alcohol residue. HxA3, HxB3, and their non-enzymatically formed isomers are nonclassic eicosanoid derived from acid the (PUFA), arachidonic acid. A second group of less well studied hepoxilins, HxA4, HxB4, and their non-enzymatically formed isomers are nonclassical eicosanoids derived from the PUFA, eicosapentaenoic acid. Recently, 14,15-HxA3 and 14,15-HxB3 have been defined as arachidonic acid derivatives that are produced by a different metabolic pathway than HxA3, HxB3, HxA4, or HxB4 and differ from the aforementioned hepoxilins in the positions of their hydroxyl and epoxide residues. Finally, hepoxilin-like products of two other PUFAs, docosahexaenoic acid and linoleic acid, have been described. All of these epoxyalcohol metabolites are at least somewhat unstable and are readily enzymatically or non-enzymatically to their corresponding trihydroxy counterparts, the trioxilins (TrX). HxA3 and HxB3, in particular, are being rapidly metabolized to TrXA3, TrXB3, and TrXC3. Hepoxilins have various biological activities in animal models and/or cultured mammalian tissues and cells. The TrX metabolites of HxA3 and HxB3 have less or no activity in most of the systems studied but in some systems retain the activity of their precursor hepoxilins. Based on these studies, it has been proposed that the hepoxilins and trioxilins function in human physiology and pathology by, for example, promoting inflammation responses and dilating arteries to regulate regional blood flow and blood pressure.

Most of the eicosanoid receptors are integral membrane protein G protein-coupled receptors (GPCRs) that bind and respond to eicosanoid signaling molecules. Eicosanoids are rapidly metabolized to inactive products and therefore are short-lived. Accordingly, the eicosanoid-receptor interaction is typically limited to a local interaction: cells, upon stimulation, metabolize arachidonic acid to an eicosanoid which then binds cognate receptors on either its parent cell or on nearby cells to trigger functional responses within a restricted tissue area, e.g. an inflammatory response to an invading pathogen. In some cases, however, the synthesized eicosanoid travels through the blood to trigger systemic or coordinated tissue responses, e.g. prostaglandin (PG) E2 released locally travels to the hypothalamus to trigger a febrile reaction. An example of a non-GPCR receptor that binds many eicosanoids is the PPAR-γ nuclear receptor.

Arachidonate 5-lipoxygenase, also known as ALOX5, 5-lipoxygenase, 5-LOX, or 5-LO, is a non-heme iron-containing enzyme that in humans is encoded by the ALOX5 gene. Arachidonate 5-lipoxygenase is a member of the lipoxygenase family of enzymes. It transforms essential fatty acids (EFA) substrates into leukotrienes as well as a wide range of other biologically active products. ALOX5 is a current target for pharmaceutical intervention in a number of diseases.

<span class="mw-page-title-main">Mead acid</span> Chemical compound

Mead acid is an omega-9 fatty acid, first characterized by James F. Mead. As with some other omega-9 polyunsaturated fatty acids, animals can make Mead acid de novo. Its elevated presence in the blood is an indication of essential fatty acid deficiency. Mead acid is found in large quantities in cartilage.

<span class="mw-page-title-main">ALOX15</span> Lipoxygenase found in humans

ALOX15 is, like other lipoxygenases, a seminal enzyme in the metabolism of polyunsaturated fatty acids to a wide range of physiologically and pathologically important products. ▼ Gene Function

<span class="mw-page-title-main">5-Hydroxyeicosatetraenoic acid</span> Chemical compound

5-Hydroxyeicosatetraenoic acid (5-HETE, 5(S)-HETE, or 5S-HETE) is an eicosanoid, i.e. a metabolite of arachidonic acid. It is produced by diverse cell types in humans and other animal species. These cells may then metabolize the formed 5(S)-HETE to 5-oxo-eicosatetraenoic acid (5-oxo-ETE), 5(S),15(S)-dihydroxyeicosatetraenoic acid (5(S),15(S)-diHETE), or 5-oxo-15-hydroxyeicosatetraenoic acid (5-oxo-15(S)-HETE).

<span class="mw-page-title-main">CYP4A11</span> Protein-coding gene in the species Homo sapiens

Cytochrome P450 4A11 is a protein that in humans is codified by the CYP4A11 gene.

<span class="mw-page-title-main">CYP4F8</span> Protein-coding gene in the species Homo sapiens

Cytochrome P450 4F8 is a protein that in humans is encoded by the CYP4F8 gene.

<span class="mw-page-title-main">CYP4F12</span> Protein-coding gene in the species Homo sapiens

Cytochrome P450 4F12 is a protein that in humans is encoded by the CYP4F12 gene.

Epoxygenases are a set of membrane-bound, heme-containing cytochrome P450 enzymes that metabolize polyunsaturated fatty acids to epoxide products that have a range of biological activities. The most thoroughly studied substrate of the CYP epoxylgenases is arachidonic acid. This polyunsaturated fatty acid is metabolized by cyclooxygenases to various prostaglandin, thromboxane, and prostacyclin metabolites in what has been termed the first pathway of eicosanoid production; it is also metabolized by various lipoxygenases to hydroxyeicosatetraenoic acids and leukotrienes in what has been termed the second pathway of eicosanoid production. The metabolism of arachidonic acid to epoxyeicosatrienoic acids by the CYP epoxygenases has been termed the third pathway of eicosanoid metabolism. Like the first two pathways of eicosanoid production, this third pathway acts as a signaling pathway wherein a set of enzymes metabolize arachidonic acid to a set of products that act as secondary signals to work in activating their parent or nearby cells and thereby orchestrate functional responses. However, none of these three pathways is limited to metabolizing arachidonic acid to eicosanoids. Rather, they also metabolize other polyunsaturated fatty acids to products that are structurally analogous to the eicosanoids but often have different bioactivity profiles. This is particularly true for the CYP epoxygenases which in general act on a broader range of polyunsaturated fatty acids to form a broader range of metabolites than the first and second pathways of eicosanoid production. Furthermore, the latter pathways form metabolites many of which act on cells by binding with and thereby activating specific and well-characterized receptor proteins; no such receptors have been fully characterized for the epoxide metabolites. Finally, there are relatively few metabolite-forming lipoxygenases and cyclooxygenases in the first and second pathways and these oxygenase enzymes share similarity between humans and other mammalian animal models. The third pathway consists of a large number of metabolite-forming CYP epoxygenases and the human epoxygenases have important differences from those of animal models. Partly because of these differences, it has been difficult to define clear roles for the epoxygenase-epoxide pathways in human physiology and pathology.

<span class="mw-page-title-main">12-Hydroxyeicosatetraenoic acid</span> Chemical compound

12-Hydroxyeicosatetraenoic acid (12-HETE) is a derivative of the 20 carbon polyunsaturated fatty acid, arachidonic acid, containing a hydroxyl residue at carbon 12 and a 5Z,8Z,10E,14Z Cis–trans isomerism configuration (Z=cis, E=trans) in its four double bonds. It was first found as a product of arachidonic acid metabolism made by human and bovine platelets through their 12S-lipoxygenase (i.e. ALOX12) enzyme(s). However, the term 12-HETE is ambiguous in that it has been used to indicate not only the initially detected "S" stereoisomer, 12S-hydroxy-5Z,8Z,10E,14Z-eicosatetraenoic acid (12(S)-HETE or 12S-HETE), made by platelets, but also the later detected "R" stereoisomer, 12(R)-hydroxy-5Z,8Z,10E,14Z-eicosatetraenoic acid (also termed 12(R)-HETE or 12R-HETE) made by other tissues through their 12R-lipoxygenase enzyme, ALOX12B. The two isomers, either directly or after being further metabolized, have been suggested to be involved in a variety of human physiological and pathological reactions. Unlike hormones which are secreted by cells, travel in the circulation to alter the behavior of distant cells, and thereby act as Endocrine signalling agents, these arachidonic acid metabolites act locally as Autocrine signalling and/or Paracrine signaling agents to regulate the behavior of their cells of origin or of nearby cells, respectively. In these roles, they may amplify or dampen, expand or contract cellular and tissue responses to disturbances.

<span class="mw-page-title-main">15-Hydroxyeicosatetraenoic acid</span> Chemical compound

15-Hydroxyeicosatetraenoic acid (also termed 15-HETE, 15(S)-HETE, and 15S-HETE) is an eicosanoid, i.e. a metabolite of arachidonic acid. Various cell types metabolize arachidonic acid to 15(S)-hydroperoxyeicosatetraenoic acid (15(S)-HpETE). This initial hydroperoxide product is extremely short-lived in cells: if not otherwise metabolized, it is rapidly reduced to 15(S)-HETE. Both of these metabolites, depending on the cell type which forms them, can be further metabolized to 15-oxo-eicosatetraenoic acid (15-oxo-ETE), 5(S),15(S)-dihydroxy-eicosatetraenoic acid (5(S),15(S)-diHETE), 5-oxo-15(S)-hydroxyeicosatetraenoic acid (5-oxo-15(S)-HETE), a subset of specialized pro-resolving mediators viz., the lipoxins, a class of pro-inflammatory mediators, the eoxins, and other products that have less well-defined activities and functions. Thus, 15(S)-HETE and 15(S)-HpETE, in addition to having intrinsic biological activities, are key precursors to numerous biologically active derivatives.

Eoxins are proposed to be a family of proinflammatory eicosanoids. They are produced by human eosinophils, mast cells, the L1236 Reed–Sternberg cell line derived from Hodgkin's lymphoma, and certain other tissues. These cells produce the eoxins by initially metabolizing arachidonic acid, an omega-6 (ω-6) fatty acid, via any enzyme possessing 15-lipoxygenase activity. The product of this initial metabolic step, 15(S)-hydroperoxyeicosatetraenoic acid, is then converted to a series of eoxins by the same enzymes that metabolize the 5-lipoxygenase product of arachidonic acid metabolism, i.e. 5-Hydroperoxy-eicosatetraenoic acid to a series of leukotrienes. That is, the eoxins are 14,15-disubstituted analogs of the 5,6-disubstituted leukotrienes.

<span class="mw-page-title-main">12-Hydroxyheptadecatrienoic acid</span> Chemical compound

12-Hydroxyheptadecatrienoic acid (also termed 12-HHT, 12(S)-hydroxyheptadeca-5Z,8E,10E-trienoic acid, or 12(S)-HHTrE) is a 17 carbon metabolite of the 20 carbon polyunsaturated fatty acid, arachidonic acid. It was discovered and structurally defined in 1973 by P. Wlodawer, Bengt I. Samuelsson, and M. Hamberg, as a product of arachidonic acid metabolism made by microsomes (i.e. endoplasmic reticulum) isolated from sheep seminal vesicle glands and by intact human platelets. 12-HHT is less ambiguously termed 12-(S)-hydroxy-5Z,8E,10E-heptadecatrienoic acid to indicate the S stereoisomerism of its 12-hydroxyl residue and the Z, E, and E cis-trans isomerism of its three double bonds. The metabolite was for many years thought to be merely a biologically inactive byproduct of prostaglandin synthesis. More recent studies, however, have attached potentially important activity to it.

<span class="mw-page-title-main">5-Oxo-eicosatetraenoic acid</span> Chemical compound

5-Oxo-eicosatetraenoic acid is a nonclassic eicosanoid metabolite of arachidonic acid and the most potent naturally occurring member of the 5-HETE family of cell signaling agents. Like other cell signaling agents, 5-oxo-ETE is made by a cell and then feeds back to stimulate its parent cell and/or exits this cell to stimulate nearby cells. 5-Oxo-ETE can stimulate various cell types particularly human leukocytes but possesses its highest potency and power in stimulating the human eosinophil type of leukocyte. It is therefore suggested to be formed during and to be an important contributor to the formation and progression of eosinophil-based allergic reactions; it is also suggested that 5-oxo-ETE contributes to the development of inflammation, cancer cell growth, and other pathological and physiological events.

<span class="mw-page-title-main">Epoxyeicosatetraenoic acid</span> Chemical compound

Epoxyeicosatetraenoic acids are a set of biologically active epoxides that various cell types make by metabolizing the omega 3 fatty acid, eicosapentaenoic acid (EPA), with certain cytochrome P450 epoxygenases. These epoxygenases can metabolize EPA to as many as 10 epoxides that differ in the site and/or stereoisomer of the epoxide formed; however, the formed EEQs, while differing in potency, often have similar bioactivities and are commonly considered together.

Cytochrome P450 omega hydroxylases, also termed cytochrome P450 ω-hydroxylases, CYP450 omega hydroxylases, CYP450 ω-hydroxylases, CYP omega hydroxylase, CYP ω-hydroxylases, fatty acid omega hydroxylases, cytochrome P450 monooxygenases, and fatty acid monooxygenases, are a set of cytochrome P450-containing enzymes that catalyze the addition of a hydroxyl residue to a fatty acid substrate. The CYP omega hydroxylases are often referred to as monoxygenases; however, the monooxygenases are CYP450 enzymes that add a hydroxyl group to a wide range of xenobiotic and naturally occurring endobiotic substrates, most of which are not fatty acids. The CYP450 omega hydroxylases are accordingly better viewed as a subset of monooxygenases that have the ability to hydroxylate fatty acids. While once regarded as functioning mainly in the catabolism of dietary fatty acids, the omega oxygenases are now considered critical in the production or break-down of fatty acid-derived mediators which are made by cells and act within their cells of origin as autocrine signaling agents or on nearby cells as paracrine signaling agents to regulate various functions such as blood pressure control and inflammation.

References

  1. 1 2 "Eicosanoid Synthesis and Metabolism: Prostaglandins, Thromboxanes, Leukotrienes, Lipoxins". The Medical Biochemistry Page. 2024. Retrieved 9 April 2024.
  2. "15.2C: Chemistry of Hormones". Medicine LibreTexts. 2018-07-21. Retrieved 2024-04-09.
  3. Edwards IJ, O'Flaherty JT (2008). "Omega-3 Fatty Acids and PPARgamma in Cancer". PPAR Research. 2008: 358052. doi: 10.1155/2008/358052 . PMC   2526161 . PMID   18769551.
  4. DeCaterina, R; Basta, G (June 2001). "n-3 Fatty acids and the inflammatory response – biological background". European Heart Journal Supplements. 3, Suppl D: D42–D49. doi: 10.1016/S1520-765X(01)90118-X . S2CID   22691568.
  5. 1 2 Funk, Colin D. (30 November 2001). "Prostaglandins and Leukotrienes: Advances in Eicosanoid Biology". Science. 294 (5548): 1871–1875. Bibcode:2001Sci...294.1871F. doi:10.1126/science.294.5548.1871. PMID   11729303.
  6. 1 2 Piomelli, Daniele (2000). "Arachidonic Acid". Neuropsychopharmacology: The Fifth Generation of Progress. Archived from the original on 2006-07-15. Retrieved 2006-03-03.
  7. 1 2 Soberman, Roy J.; Christmas, Peter (2003). "The organization and consequences of eicosanoid signaling". J. Clin. Invest. 111 (8): 1107–1113. doi:10.1172/JCI18338. PMC   152944 . PMID   12697726.
  8. Beare-Rogers (2001). "IUPAC Lexicon of Lipid Nutrition" (PDF). Retrieved June 1, 2006.
  9. Prostacyclin—PGI—was previously classified as prostaglandin and retains its old PGI2 identifier.
  10. Eicosanoids with different letters have placement of double-bonds and different functional groups attached to the molecular skeleton. Letters indicate roughly the order the eicosanoids were first described in the literature. For diagrams for PG [A–H] see Cyberlipid Center. "Prostanoids". Archived from the original on 2007-02-08. Retrieved 2007-02-05.
  11. Rossi AG, Thomas MJ, O'Flaherty JT (1988). "Stereospecific actions of 5-hydroxyeicosatetraenoate". FEBS Letters. 240 (1–2): 163–166. doi: 10.1016/0014-5793(88)80360-0 . PMID   3191990. S2CID   43027447.
  12. 1 2 Straus DS, Glass CK (2001). "Cyclopentenone prostaglandins: new insights on biological activities and cellular targets". Medicinal Research Reviews. 21 (3): 185–210. doi:10.1002/med.1006.abs. PMID   11301410.
  13. Prasad KN, Hovland AR, Cole WC, Prasad KC, Nahreini P, Edwards-Prasad J, Andreatta CP (2000). "Multiple antioxidants in the prevention and treatment of Alzheimer disease: analysis of biologic rationale". Clinical Neuropharmacology. 23 (1): 2–13. doi:10.1097/00002826-200001000-00002. PMID   10682224.
  14. Xu Y, Qian SY (2014). "Anti-cancer activities of ω-6 polyunsaturated fatty acids". Biomedical Journal. 37 (3): 112–119. doi: 10.4103/2319-4170.131378 . PMC   4166599 . PMID   24923568.
  15. Gomolka B, Siegert E, Blossey K, Schunck WH, Rothe M, Weylandt KH (2011). "Analysis of omega-3 and omega-6 fatty acid-derived lipid metabolite formation in human and mouse blood samples". Prostaglandins & Other Lipid Mediators. 94 (3–4): 81–87. doi:10.1016/j.prostaglandins.2010.12.006. PMID   21236358.
  16. Zulfakar MH, Edwards M, Heard CM (2007). "Is there a role for topically delivered eicosapentaenoic acid in the treatment of psoriasis?". European Journal of Dermatology. 17 (4): 284–291. doi:10.1684/ejd.2007.0201 (inactive 31 January 2024). PMID   17540633.{{cite journal}}: CS1 maint: DOI inactive as of January 2024 (link)
  17. Caramia G (2012). "[Essential fatty acids and lipid mediators. Endocannabinoids]". La Pediatria Medica e Chirurgica: Medical and Surgical Pediatrics (in Italian). 34 (2): 65–72. doi: 10.4081/pmc.2012.2 . PMID   22730630.
  18. 1 2 3 4 Wiktorowska-Owczarek A, Berezińska M, Nowak JZ (2015). "PUFAs: Structures, Metabolism and Functions". Advances in Clinical and Experimental Medicine. 24 (6): 931–941. doi: 10.17219/acem/31243 . PMID   26771963.
  19. Tanaka N, Yamaguchi H, Furugen A, Ogura J, Kobayashi M, Yamada T, Mano N, Iseki K (2014). "Quantification of intracellular and extracellular eicosapentaenoic acid-derived 3-series prostanoids by liquid chromatography/electrospray ionization tandem mass spectrometry". Prostaglandins, Leukotrienes, and Essential Fatty Acids. 91 (3): 61–71. doi:10.1016/j.plefa.2014.04.005. PMID   24996760.
  20. Van Dyke TE, Serhan CN (2003). "Resolution of inflammation: a new paradigm for the pathogenesis of periodontal diseases". J. Dent. Res. 82 (2): 82–90. doi:10.1177/154405910308200202. PMID   12562878. S2CID   40812937.
  21. Serhan CN, Gotlinger K, Hong S, Arita M (2004). "Resolvins, docosatrienes, and neuroprotectins, novel omega-3-derived mediators, and their aspirin-triggered endogenous epimers: an overview of their protective roles in catabasis". Prostaglandins Other Lipid Mediat. 73 (3–4): 155–172. doi:10.1016/j.prostaglandins.2004.03.005. PMID   15290791.
  22. Anderle P, Farmer P, Berger A, Roberts MA (2004). "Nutrigenomic approach to understanding the mechanisms by which dietary long-chain fatty acids induce gene signals and control mechanisms involved in carcinogenesis". Nutrition (Burbank, Los Angeles County, Calif.). 20 (1): 103–108. doi:10.1016/j.nut.2003.09.018. PMID   14698023.
  23. Evans AR, Junger H, Southall MD, et al. (2000). "Isoprostanes, novel eicosanoids that produce nociception and sensitize rat sensory neurons". J. Pharmacol. Exp. Ther. 293 (3): 912–920. PMID   10869392.
  24. O'Brien WF, Krammer J, O'Leary TD, Mastrogiannis DS (1993). "The effect of acetaminophen on prostacyclin production in pregnant women". Am. J. Obstet. Gynecol. 168 (4): 1164–1169. doi:10.1016/0002-9378(93)90362-m. PMID   8475962.
  25. Behrendt H, Kasche A, Ebner von Eschenbach C, Risse U, Huss-Marp J, Ring J (2001). "Secretion of proinflammatory eicosanoid-like substances precedes allergen release from pollen grains in the initiation of allergic sensitization" (PDF). Int. Arch. Allergy Immunol. 124 (1–3): 121–125. doi:10.1159/000053688. PMID   11306946. S2CID   53331.
  26. Sarau HM, Foley JJ, Schmidt DB, et al. (1999). "In vitro and in vivo pharmacological characterization of SB 201993, an eicosanoid-like LTB4 receptor antagonist with anti-inflammatory activity". Prostaglandins Leukot. Essent. Fatty Acids. 61 (1): 55–64. doi:10.1054/plef.1999.0074. PMID   10477044.
  27. Czerska M, Zieliński M, Gromadzińska J (2016). "Isoprostanes - A novel major group of oxidative stress markers". International Journal of Occupational Medicine and Environmental Health. 29 (2): 179–190. doi: 10.13075/ijomeh.1896.00596 . PMID   26670350.
  28. Friedli O, Freigang S (2016). "Cyclopentenone-containing oxidized phospholipids and their isoprostanes as pro-resolving mediators of inflammation". Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids. 1862 (4): 382–392. doi: 10.1016/j.bbalip.2016.07.006 . PMID   27422370.
  29. Cuyamendous C, de la Torre A, Lee YY, Leung KS, Guy A, Bultel-Poncé V, Galano JM, Lee JC, Oger C, Durand T (2016). "The novelty of phytofurans, isofurans, dihomo-isofurans and neurofurans: Discovery, synthesis and potential application" (PDF). Biochimie. 130: 49–62. doi:10.1016/j.biochi.2016.08.002. PMID   27519299. S2CID   1504539.
  30. University of Kansas Medical Center (2004). "Eicosanoids and Inflammation" (PDF). Archived from the original (PDF) on 2005-05-16. Retrieved 2007-01-05.
  31. 1 2 3 4 Gabbs M, Leng S, Devassy JG, Monirujjaman M, Aukema HM (2015). "Advances in Our Understanding of Oxylipins Derived from Dietary PUFAs". Advances in Nutrition. 6 (5): 513–540. doi:10.3945/an.114.007732. PMC   4561827 . PMID   26374175.
  32. 1 2 Powell WS, Rokach J (2015). "Biosynthesis, biological effects, and receptors of hydroxyeicosatetraenoic acids (HETEs) and oxoeicosatetraenoic acids (oxo-ETEs) derived from arachidonic acid". Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids. 1851 (4): 340–355. doi:10.1016/j.bbalip.2014.10.008. PMC   5710736 . PMID   25449650.
  33. Capra V, Rovati GE, Mangano P, Buccellati C, Murphy RC, Sala A (2015). "Transcellular biosynthesis of eicosanoid lipid mediators". Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids. 1851 (4): 377–382. doi:10.1016/j.bbalip.2014.09.002. PMID   25218301.
  34. Cyrus, Tillmann; Witztum, Joseph L.; Rader, Daniel J.; Tangirala, Rajendra; Fazio, Sergio; Linton, Macrae F.; Funk, Colin D. (June 1999). "Disruption of the 12/15-lipoxygenase gene diminishes atherosclerosis in apo E–deficient mice". J Clin Invest. 103 (11): 1597–1604n. doi:10.1172/JCI5897. PMC   408369 . PMID   10359569.
  35. Schewe T. (Mar–Apr 2002). "15-lipoxygenase-1: a prooxidant enzyme". Biol. Chem. 383 (3–4): 365–374. doi:10.1515/BC.2002.041. PMID   12033428. S2CID   7487557.
  36. Korbecki J, Baranowska-Bosiacka I, Gutowska I, Chlubek D (2014). "Cyclooxygenase pathways". Acta Biochimica Polonica. 61 (4): 639–649. doi: 10.18388/abp.2014_1825 . PMID   25343148.
  37. 1 2 3 4 Claar D, Hartert TV, Peebles RS (2015). "The role of prostaglandins in allergic lung inflammation and asthma". Expert Review of Respiratory Medicine. 9 (1): 55–72. doi:10.1586/17476348.2015.992783. PMC   4380345 . PMID   25541289.
  38. Simopoulos AP (2010). "Genetic variants in the metabolism of omega-6 and omega-3 fatty acids: their role in the determination of nutritional requirements and chronic disease risk". Experimental Biology and Medicine. 235 (7): 785–795. doi:10.1258/ebm.2010.009298. PMID   20558833. S2CID   207195131.
  39. 1 2 Surh YJ, Na HK, Park JM, Lee HN, Kim W, Yoon IS, Kim DD (2011). "15-Deoxy-Δ¹²,¹⁴-prostaglandin J₂, an electrophilic lipid mediator of anti-inflammatory and pro-resolving signaling". Biochemical Pharmacology. 82 (10): 1335–1351. doi:10.1016/j.bcp.2011.07.100. PMID   21843512.
  40. Powell, W. S.; Rokach, J (2013). "The eosinophil chemoattractant 5-oxo-ETE and the OXE receptor". Progress in Lipid Research. 52 (4): 651–665. doi:10.1016/j.plipres.2013.09.001. PMC   5710732 . PMID   24056189.
  41. Rådmark O, Werz O, Steinhilber D, Samuelsson B (2015). "5-Lipoxygenase, a key enzyme for leukotriene biosynthesis in health and disease". Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids. 1851 (4): 331–339. doi:10.1016/j.bbalip.2014.08.012. PMID   25152163.
  42. Ahmad S, Thulasingam M, Palombo I, Daley DO, Johnson KA, Morgenstern R, Haeggström JZ, Rinaldo-Matthis A (2015). "Trimeric microsomal glutathione transferase 2 displays one third of the sites reactivity". Biochimica et Biophysica Acta (BBA) - Proteins and Proteomics. 1854 (10 Pt A): 1365–1371. doi:10.1016/j.bbapap.2015.06.003. PMID   26066610.
  43. Pace-Asciak, C. R. (2009). "The hepoxilins and some analogues: A review of their biology". British Journal of Pharmacology. 158 (4): 972–981. doi:10.1111/j.1476-5381.2009.00168.x. PMC   2785520 . PMID   19422397.
  44. Dobrian, A. D.; Lieb, D. C.; Cole, B. K.; Taylor-Fishwick, D. A.; Chakrabarti, S. K.; Nadler, J. L. (2011). "Functional and pathological roles of the 12- and 15-lipoxygenases". Progress in Lipid Research. 50 (1): 115–131. doi:10.1016/j.plipres.2010.10.005. PMC   3012140 . PMID   20970452.
  45. Ivanov, I; Kuhn, H; Heydeck, D (2015). "Structural and functional biology of arachidonic acid 15-lipoxygenase-1 (ALOX15)". Gene. 573 (1): 1–32. doi:10.1016/j.gene.2015.07.073. PMC   6728142 . PMID   26216303.
  46. Wittwer, J; Hersberger, M (2007). "The two faces of the 15-lipoxygenase in atherosclerosis". Prostaglandins, Leukotrienes and Essential Fatty Acids. 77 (2): 67–77. doi:10.1016/j.plefa.2007.08.001. PMID   17869078.
  47. Kroetz DL, Xu F (2005). "Regulation and inhibition of arachidonic acid omega-hydroxylases and 20-HETE formation". Annual Review of Pharmacology and Toxicology. 45: 413–438. doi:10.1146/annurev.pharmtox.45.120403.100045. PMID   15822183.
  48. 1 2 Yang, L; Mäki-Petäjä, K; Cheriyan, J; McEniery, C; Wilkinson, I. B. (2015). "The role of epoxyeicosatrienoic acids in the cardiovascular system". British Journal of Clinical Pharmacology. 80 (1): 28–44. doi:10.1111/bcp.12603. PMC   4500322 . PMID   25655310.
  49. 1 2 3 Spector, A. A.; Kim, H. Y. (2015). "Cytochrome P450 epoxygenase pathway of polyunsaturated fatty acid metabolism". Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids. 1851 (4): 356–365. doi:10.1016/j.bbalip.2014.07.020. PMC   4314516 . PMID   25093613.
  50. Fer, M; Dréano, Y; Lucas, D; Corcos, L; Salaün, J. P.; Berthou, F; Amet, Y (2008). "Metabolism of eicosapentaenoic and docosahexaenoic acids by recombinant human cytochromes P450". Archives of Biochemistry and Biophysics. 471 (2): 116–125. doi:10.1016/j.abb.2008.01.002. PMID   18206980.
  51. 1 2 Shahabi, P; Siest, G; Meyer, UA; Visvikis-Siest, S (2014). "Human cytochrome P450 epoxygenases: Variability in expression and role in inflammation-related disorders". Pharmacology & Therapeutics. 144 (2): 134–161. doi:10.1016/j.pharmthera.2014.05.011. PMID   24882266.
  52. Frömel, T; Kohlstedt, K; Popp, R; Yin, X; Awwad, K; Barbosa-Sicard, E; Thomas, AC; Lieberz, R; Mayr, M; Fleming, I (2013). "Cytochrome P4502S1: A novel monocyte/macrophage fatty acid epoxygenase in human atherosclerotic plaques". Basic Research in Cardiology. 108 (1): 319. doi:10.1007/s00395-012-0319-8. PMID   23224081. S2CID   9158244.
  53. Fleming, I (2014). "The pharmacology of the cytochrome P450 epoxygenase/soluble epoxide hydrolase axis in the vasculature and cardiovascular disease". Pharmacological Reviews. 66 (4): 1106–1140. doi:10.1124/pr.113.007781. PMID   25244930. S2CID   39465144.
  54. Westphal, C; Konkel, A; Schunck, WH (2011). "CYP-eicosanoids--a new link between omega-3 fatty acids and cardiac disease?". Prostaglandins & Other Lipid Mediators. 96 (1–4): 99–108. doi:10.1016/j.prostaglandins.2011.09.001. PMID   21945326.
  55. Matsuoka T, Narumiya S (2007). "Prostaglandin receptor signaling in disease". TheScientificWorldJournal. 7: 1329–1347. doi: 10.1100/tsw.2007.182 . PMC   5901339 . PMID   17767353.
  56. Thomas J, Fairclough A, Kavanagh J, Kelly AJ (2014). "Vaginal prostaglandin (PGE2 and PGF2a) for induction of labour at term". The Cochrane Database of Systematic Reviews. 2014 (6): CD003101. doi:10.1002/14651858.CD003101.pub3. PMC   7138281 . PMID   24941907.
  57. Rossi A, Anzalone A, Fortuna MC, Caro G, Garelli V, Pranteda G, Carlesimo M (2016). "Multi-therapies in androgenetic alopecia: review and clinical experiences". Dermatologic Therapy. 29 (6): 424–432. doi:10.1111/dth.12390. hdl: 11573/877469 . PMID   27424565. S2CID   45963890.
  58. Garza LA, Liu Y, Yang Z, Alagesan B, Lawson JA, Norberg SM, Loy DE, Zhao T, Blatt HB, Stanton DC, Carrasco L, Ahluwalia G, Fischer SM, FitzGerald GA, Cotsarelis G (2012). "Prostaglandin D2 inhibits hair growth and is elevated in bald scalp of men with androgenetic alopecia". Science Translational Medicine. 4 (126): 126ra34. doi:10.1126/scitranslmed.3003122. PMC   3319975 . PMID   22440736.
  59. 1 2 Hata AN, Breyer RM (2004). "Pharmacology and signaling of prostaglandin receptors: multiple roles in inflammation and immune modulation". Pharmacology & Therapeutics. 103 (2): 147–166. doi:10.1016/j.pharmthera.2004.06.003. PMID   15369681.
  60. 1 2 Figueiredo-Pereira ME, Corwin C, Babich J (2016). "Prostaglandin J2: a potential target for halting inflammation-induced neurodegeneration". Annals of the New York Academy of Sciences. 1363 (1): 125–137. Bibcode:2016NYASA1363..125F. doi:10.1111/nyas.12987. PMC   4801700 . PMID   26748744.
  61. Hoxha M, Buccellati C, Capra V, Garella D, Cena C, Rolando B, Fruttero R, Carnevali S, Sala A, Rovati GE, Bertinaria M (2016). "In vitro pharmacological evaluation of multitarget agents for thromboxane prostanoid receptor antagonism and COX-2 inhibition" (PDF). Pharmacological Research. 103: 132–143. doi:10.1016/j.phrs.2015.11.012. hdl:2318/1551575. PMID   26621246. S2CID   12881002.
  62. Cruz JE, Ward A, Anthony S, Chang S, Bae HB, Hermes-DeSantis ER (2016). "Evidence for the Use of Epoprostenol to Treat Raynaud's Phenomenon With or Without Digital Ulcers: A Review of the Literature". The Annals of Pharmacotherapy. 50 (12): 1060–1067. doi:10.1177/1060028016660324. PMID   27465880. S2CID   38333954.
  63. O'Connell C, Amar D, Boucly A, Savale L, Jaïs X, Chaumais MC, Montani D, Humbert M, Simonneau G, Sitbon O (2016). "Comparative Safety and Tolerability of Prostacyclins in Pulmonary Hypertension". Drug Safety. 39 (4): 287–294. doi:10.1007/s40264-015-0365-x. PMID   26748508. S2CID   24852012.
  64. Cacione, Daniel G.; Macedo, Cristiane R.; do Carmo Novaes, Frederico; Baptista-Silva, Jose Cc (4 May 2020). "Pharmacological treatment for Buerger's disease". The Cochrane Database of Systematic Reviews. 5 (5): CD011033. doi:10.1002/14651858.CD011033.pub4. ISSN   1469-493X. PMC   7197514 . PMID   32364620.
  65. Citterio A, Arnoldi A, Panzeri E, D'Angelo MG, Filosto M, Dilena R, Arrigoni F, Castelli M, Maghini C, Germiniasi C, Menni F, Martinuzzi A, Bresolin N, Bassi MT (2014). "Mutations in CYP2U1, DDHD2 and GBA2 genes are rare causes of complicated forms of hereditary spastic paraparesis" (PDF). Journal of Neurology. 261 (2): 373–381. doi:10.1007/s00415-013-7206-6. hdl:2434/421160. PMID   24337409. S2CID   19189811.
  66. 1 2 Liu M, Yokomizo T (2015). "The role of leukotrienes in allergic diseases". Allergology International. 64 (1): 17–26. doi: 10.1016/j.alit.2014.09.001 . PMID   25572555.
  67. Bäck M, Dahlén SE, Drazen JM, Evans JF, Serhan CN, Shimizu T, Yokomizo T, Rovati GE (2011). "International Union of Basic and Clinical Pharmacology. LXXXIV: leukotriene receptor nomenclature, distribution, and pathophysiological functions". Pharmacological Reviews. 63 (3): 539–584. doi: 10.1124/pr.110.004184 . PMID   21771892. S2CID   5563700.
  68. Bäck M, Powell WS, Dahlén SE, Drazen JM, Evans JF, Serhan CN, Shimizu T, Yokomizo T, Rovati GE (2014). "Update on leukotriene, lipoxin and oxoeicosanoid receptors: IUPHAR Review 7". British Journal of Pharmacology. 171 (15): 3551–3574. doi:10.1111/bph.12665. PMC   4128057 . PMID   24588652.
  69. Cingi C, Muluk NB, Ipci K, Şahin E (2015). "Antileukotrienes in upper airway inflammatory diseases". Current Allergy and Asthma Reports. 15 (11): 64. doi:10.1007/s11882-015-0564-7. PMID   26385352. S2CID   38854822.
  70. Nettis E, D'Erasmo M, Di Leo E, Calogiuri G, Montinaro V, Ferrannini A, Vacca A (2010). "The employment of leukotriene antagonists in cutaneous diseases belonging to allergological field". Mediators of Inflammation. 2010: 1–6. doi: 10.1155/2010/628171 . PMC   2945673 . PMID   20886028.
  71. Kanaoka Y, Maekawa A, Austen KF (2013). "Identification of GPR99 protein as a potential third cysteinyl leukotriene receptor with a preference for leukotriene E4 ligand". The Journal of Biological Chemistry. 288 (16): 10967–10972. doi: 10.1074/jbc.C113.453704 . PMC   3630866 . PMID   23504326.
  72. 1 2 3 Romano M, Cianci E, Simiele F, Recchiuti A (2015). "Lipoxins and aspirin-triggered lipoxins in resolution of inflammation". European Journal of Pharmacology. 760: 49–63. doi:10.1016/j.ejphar.2015.03.083. PMID   25895638.
  73. 1 2 3 Chiang N, Serhan CN, Dahlén SE, Drazen JM, Hay DW, Rovati GE, Shimizu T, Yokomizo T, Brink C (2006). "The lipoxin receptor ALX: potent ligand-specific and stereoselective actions in vivo". Pharmacological Reviews. 58 (3): 463–487. doi:10.1124/pr.58.3.4. PMID   16968948. S2CID   6496181.
  74. Qu Q, Xuan W, Fan GH (2015). "Roles of resolvins in the resolution of acute inflammation". Cell Biology International. 39 (1): 3–22. doi:10.1002/cbin.10345. PMID   25052386. S2CID   10160642.
  75. Lim JY, Park CK, Hwang SW (2015). "Biological Roles of Resolvins and Related Substances in the Resolution of Pain". BioMed Research International. 2015: 830930. doi: 10.1155/2015/830930 . PMC   4538417 . PMID   26339646.
  76. 1 2 Serhan CN, Chiang N, Dalli J, Levy BD (2015). "Lipid mediators in the resolution of inflammation". Cold Spring Harbor Perspectives in Biology. 7 (2): a016311. doi:10.1101/cshperspect.a016311. PMC   4315926 . PMID   25359497.
  77. Serhan CN, Chiang N (2013). "Resolution phase lipid mediators of inflammation: agonists of resolution". Current Opinion in Pharmacology. 13 (4): 632–640. doi:10.1016/j.coph.2013.05.012. PMC   3732499 . PMID   23747022.
  78. Yang L, Mäki-Petäjä K, Cheriyan J, McEniery C, Wilkinson IB (2015). "The role of epoxyeicosatrienoic acids in the cardiovascular system". British Journal of Clinical Pharmacology. 80 (1): 28–44. doi:10.1111/bcp.12603. PMC   4500322 . PMID   25655310.
  79. Clinical trial number NCT00847899 for "Evaluation of Soluble Epoxide Hydrolase (s-EH) Inhibitor in Patients With Mild to Moderate Hypertension and Impaired Glucose Tolerance" at ClinicalTrials.gov
  80. Bos C, Richel D, Ritsema T, Peppelenbosch M, Versteeg H (2004). "Prostanoids and prostanoid receptors in signal transduction". Int J Biochem Cell Biol. 36 (7): 1187–1205. doi:10.1016/j.biocel.2003.08.006. PMID   15109566.
  81. Samuelsson B (May 1983). "Leukotrienes: mediators of immediate hypersensitivity reactions and inflammation". Science. 220 (4597): 568–575. Bibcode:1983Sci...220..568S. doi:10.1126/science.6301011. PMID   6301011.
  82. Haeggström JZ, Funk CD (2011). "Lipoxygenase and leukotriene pathways: biochemistry, biology, and roles in disease". Chemical Reviews. 111 (10): 5866–5898. doi:10.1021/cr200246d. PMID   21936577.[ permanent dead link ]
  83. Anwar Y, Sabir JS, Qureshi MI, Saini KS (2014). "5-lipoxygenase: a promising drug target against inflammatory diseases-biochemical and pharmacological regulation". Current Drug Targets. 15 (4): 410–422. doi:10.2174/1389450114666131209110745. PMID   24313690.
  84. Kar M, Altıntoprak N, Muluk NB, Ulusoy S, Bafaqeeh SA, Cingi C (March 2016). "Antileukotrienes in adenotonsillar hypertrophy: a review of the literature". European Archives of Oto-Rhino-Laryngology. 273 (12): 4111–4117. doi:10.1007/s00405-016-3983-8. PMID   26980339. S2CID   31311115.
  85. Oussalah A, Mayorga C, Blanca M, Barbaud A, Nakonechna A, Cernadas J, Gotua M, Brockow K, Caubet JC, Bircher A, Atanaskovic M, Demoly P, K Tanno L, Terreehorst I, Laguna JJ, Romano A, Guéant JL (April 2016). "Genetic variants associated with drugs-induced immediate hypersensitivity reactions: a PRISMA-compliant systematic review". Allergy. 71 (4): 443–462. doi: 10.1111/all.12821 . PMID   26678823. S2CID   13352894.
  86. Mitchell S, Balp MM, Samuel M, McBride D, Maurer M (2015). "Systematic review of treatments for chronic spontaneous urticaria with inadequate response to licensed first-line treatments". International Journal of Dermatology. 54 (9): 1088–1104. doi:10.1111/ijd.12727. PMID   25515967. S2CID   23250789.
  87. Wu SH, Chen XQ, Liu B, Wu HJ, Dong L (2013). "Efficacy and safety of 15(R/S)-methyl-lipoxin A(4) in topical treatment of infantile eczema". The British Journal of Dermatology. 168 (1): 172–178. doi:10.1111/j.1365-2133.2012.11177.x. PMID   22834636. S2CID   31721094.
  88. 1 2 Basil MC, Levy BD (2016). "Specialized pro-resolving mediators: endogenous regulators of infection and inflammation". Nature Reviews. Immunology. 16 (1): 51–67. doi:10.1038/nri.2015.4. PMC   5242505 . PMID   26688348.
  89. Feltenmark S, Gautam N, Brunnström A, Griffiths W, Backman L, Edenius C, Lindbom L, Björkholm M, Claesson HE (January 2008). "Eoxins are proinflammatory arachidonic acid metabolites produced via the 15-lipoxygenase-1 pathway in human eosinophils and mast cells". Proc. Natl. Acad. Sci. U.S.A. 105 (2): 680–685. Bibcode:2008PNAS..105..680F. doi: 10.1073/pnas.0710127105 . PMC   2206596 . PMID   18184802.
  90. James A, Daham K, Backman L, Brunnström A, Tingvall T, Kumlin M, Edenius C, Dahlén SE, Dahlén B, Claesson HE (2013). "The influence of aspirin on release of eoxin C4, leukotriene C4 and 15-HETE, in eosinophilic granulocytes isolated from patients with asthma". Int. Arch. Allergy Immunol. 162 (2): 135–142. doi:10.1159/000351422. PMID   23921438. S2CID   29180895.
  91. Claesson HE (2009). "On the biosynthesis and biological role of eoxins and 15-lipoxygenase-1 in airway inflammation and Hodgkin lymphoma". Prostaglandins & Other Lipid Mediators. 89 (3–4): 120–125. doi:10.1016/j.prostaglandins.2008.12.003. PMID   19130894.
  92. Serhan CN, Chiang N, Dalli J (2015). "The resolution code of acute inflammation: Novel pro-resolving lipid mediators in resolution". Seminars in Immunology. 27 (3): 200–215. doi:10.1016/j.smim.2015.03.004. PMC   4515371 . PMID   25857211.
  93. Guichardant M, Calzada C, Bernoud-Hubac N, Lagarde M, Véricel E (2015). "Omega-3 polyunsaturated fatty acids and oxygenated metabolism in atherothrombosis". Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids. 1851 (4): 485–495. doi:10.1016/j.bbalip.2014.09.013. PMID   25263947.
  94. Calder PC (2014). "Biomarkers of immunity and inflammation for use in nutrition interventions: International Life Sciences Institute European Branch work on selection criteria and interpretation". Endocrine, Metabolic & Immune Disorders Drug Targets. 14 (4): 236–244. doi:10.2174/1871530314666140709091650. PMID   25008763.
  95. 1 2 Fritsche, Kevin (August 2006). "Fatty Acids as Modulators of the Immune Response". Annual Review of Nutrition. 26: 45–73. doi:10.1146/annurev.nutr.25.050304.092610. PMID   16848700.
  96. National Institute of Health (2005-08-01). "Omega-3 fatty acids, fish oil, alpha-linolenic acid". Archived from the original on May 3, 2006. Retrieved March 26, 2006.
  97. Burr, G.O.; Burr, M.M. (1930). "On the nature and role of the fatty acids essential in nutrition". J. Biol. Chem. 86 (587): 587–621. doi: 10.1016/S0021-9258(20)78929-5 .
  98. Bergström, S.; Danielsson, H.; Samuelsson, B. (1964). "The enzymatic formation of prostaglandin E2 from arachidonic acid". Biochim. Biophys. Acta. 90 (207): 207–210. doi:10.1016/0304-4165(64)90145-x. PMID   14201168.
  99. Vane, J. R. (June 23, 1971). "Inhibition of prostaglandin synthesis as a mechanism of action for aspirin-like drugs". Nature New Biology. 231 (25): 232–235. doi:10.1038/newbio231232a0. PMID   5284360.