15-Hydroxyeicosatetraenoic acid

Last updated
15-Hydroxyeicosatetraenoic acid
15(S)-HETE.svg
Names
Preferred IUPAC name
(5Z,8Z,11Z,13E,15S)-15-Hydroxyicosa-5,8,11,13-tetraenoic acid
Other names
15-HETE, 15(S)-HETE, 15(S)-HETE
Identifiers
3D model (JSmol)
ChemSpider
ECHA InfoCard 100.214.805 OOjs UI icon edit-ltr-progressive.svg
PubChem CID
UNII
  • InChI=1S/C20H32O3/c1-2-3-13-16-19(21)17-14-11-9-7-5-4-6-8-10-12-15-18-20(22)23/h4-5,8-11,14,17,19,21H,2-3,6-7,12-13,15-16,18H2,1H3,(H,22,23)/b5-4-,10-8-,11-9-,17-14+/t19-/m0/s1
    Key: JSFATNQSLKRBCI-VAEKSGALSA-N
  • CCCCC[C@@H](/C=C/C=C\C/C=C\C/C=C\CCCC(=O)O)O
Properties
C20H32O3
Molar mass 320.473 g·mol−1
Except where otherwise noted, data are given for materials in their standard state (at 25 °C [77 °F], 100 kPa).

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. [1] [2]

Contents

Some cell types (e.g. platelets) metabolize arachidonic acid to the stereoisomer of 15(S)-HpETE, 15(R)-HpETE. Both stereoisomers may also be formed as result of the metabolism of arachidonic acid by cellular microsomes or as a result of arachidonic acid auto-oxidation. Similar to 15(S)-HpETEs, 15(R)-HpETE may be rapidly reduced to 15(R)-HETE. These R,S stereoisomers differ only in having their hydroxy residue in opposite orientations. While the two R stereoisomers are sometimes referred to as 15-HpETE and 15-HETE, proper usage should identify them as R stereoisomers. 15(R)-HpETE and 15(R)-HETE lack some of the activity attributed to their S stereoisomers but can be further metabolized to bioactive products viz., the 15(R) class of lipoxins (also termed epi-lipoxins). [3]

15(S)-HETE, 15(S)-HpETE, and many of their derivative metabolites are thought to have physiologically important functions. They appear to act as hormone-like autocrine and paracrine signaling agents that are involved in regulating inflammatory and perhaps other responses. [1] [2] [4] Clinically, drugs that are stable analogs, and therefore mimic the anti-inflammatory actions of the lipoxins and drugs that block the production or actions of the pro-inflammatory eoxins may prove useful for treating acute and chronic inflammatory disorders. [5]

Nomenclature and stereoisomers

15(S)-HETE is unambiguously designated by a shortened version of its IUPAC name viz., 15(S)-hydroxy-5Z,8Z,11Z,13E-eicosatetraenoic acid. In this terminology S refers to the absolute configuration of the chirality of the hydroxy functional group at carbon position 15. Its 15(R) enantiomer is designated 15(R)-hydroxy-5Z,8Z,11Z,13E-eicosatetraenoic acid. Z and E give the cis–trans isomerism about each double bond at carbon positions 5, 8, 11, and 13 with Z indicating cis and E indicating trans isomerism. Both stereoisomers are produced from their corresponding S and R 15-HpETE stereoisomers, i.e. 15(S)-hydroperoxy-5Z,8Z,11Z,13E-eicosatetraenoic acid (15(S)-HpETE) and 15(R)-hydroperoxy-5Z,8Z,11Z,13E-eicosatetraenoic acid (15(R)-HpETE).

Production

Human cells release arachidonic acid (i.e. 5Z,8Z,11Z,14Z-eicosatetraenoic acid) from its storage site in phospholipids by reactions that involve phospholipase C and/or lipase enzymes. This release is stimulated or enhanced by cell stimulation. The freed arachidonic acid is then converted to 15-hydroperoxy/hydroxy products by one or more of the following five pathways.

15-Lipoxygenase-1: Cells metabolize arachidonic acid with 15-lipoxygenase-1 (i.e., 15-LO-1, ALOX15) to form 15(S)-HpETE as a major product and 12(S)-hydroperoxy-5Z,8Z,10E,15Z-eicosatetraenoic acid (12(S)-HpETE) and 14(S),15(S)-trans-oxido-5Z,8Z,11Z-14,15-leukotriene A4 as minor products; 15(S)-HpETE and 12(S)-HpETE are rapidly converted to 15(S)-HETE and 12(S)-hydroxy-5Z,8Z,10E,15Z-eicosatetraenoic acid (12(S)-hydroxyeicosatetraenoic acid), (i.e. 12(S)-HETE), respectively, or further metabolized through other enzyme pathways; 14(S),15(S)-trans-oxido-5Z,8Z,11Z-14,15-leukotriene A4 is metabolized by 15-LO-1 to various isomers of 8,15(S)-dihydroxy-5S,8S,11Z,13S-eicosatetraenoic acids, e.g. 8,15(S)-LTB4's. [6] [7] [8] [9] [10]

15-Lipoxygenase-2: Cells also used 15-lipoxygenase 2 (i.e. 15-LOX-2 or ALOX15B) to make 15(S)-HpETE and 15(S)-HETE. However this enzyme has a preference for metabolizing linoleic acid rather than arachidonic acid. It therefore forms linoleic acid metabolites (e.g. 13-hydoxyperoxy/hydroxy-octadecadienoic and 9-hydroperoxy/hydroxyl-octadecadienoic acids) in greater amounts than 15(S)-HpETE and 15(S)-HETE. 15-LOX-2 also differs from 15-LOX-1 in that it does not make 12(S)-HpETE or the leukotriene A4 isomer cited above. [10]

Cyclooxygenase: Cells can use prostaglandin-endoperoxide synthase 1 (i.e. cyclooxygenenase-1 or COX-1) and prostaglandin-endoperoxide synthase 2 (COX-2) to metabolize arachidonic acid primarily to prostaglandins but also to small amounts of 11(R)-HETE and a racemic mixture of 15-HETEs composed of ~22% 15(R)-HETE and ~78% 15(S)-HETE. [11] When pretreated with aspirin, however, COX-1 is inactive while COX-2 attacks arachidonic acid to produce almost exclusively 15(R)-HETE along with its presumed precursor 15(R)-HpETE. [11] [12] [13]

Microsome metabolism: Human and rat microsomal cytochrome P450s, e.g. CYP2C19, metabolize arachidonic acid to a racemic mixture of 15-HETEs, i.e., 15(R,S)-HETEs, >90% of which is the 15(R) stereoisomer. [14] [15]

Autoxidation: The spontaneous and non-enzymatically induced autoxidation of arachidonic acid yields 15(R,S)-hydroperoxy-5Z,8Z,11Z,13E-eicosatetraenoic acids. This non-enzymatic reaction is promoted in cells undergoing oxidative stress. Cells forming this racemic mixture of 15-hydroperoxy products may convert then to 15(R,S)-HETEs and other products. However, the uncontrolled overproduction of the 15-hydroperoxy products may react with other elements to produce cell injury. [16] [17]

Further metabolism

The newly formed products formed by the pathways cited in the previous section are bioactive but may also flow into down-stream pathways to form other metabolites with a different sets of bioactivity. The initially formed 15(S)-HpETE may be further metabolized by its parent cell or pass it to nearby cell by a process termed transcellular metabolism.

15(S)-HpETE may be:

15(S)-HETE may be:

15(R)-HpETE may be:

15(R)-HETE may be:

Activities

15(S)-HpETE and 15(S)-HETE

Most studies have analyzed the action of 15(S)-HETE but not that of its less stable precursor 15(S)-HpETE. Since this precursor is rapidly converted to 15(S)-HETE in cells, it is likely that the two metabolites share similar activities. In many studies, however, is not clear that these activities reflect their intrinsic action or reflect their conversion to the metabolites sited above.

15(S)-HpETE and 15(S)-HETE bind to and activate the G protein-coupled receptor, leukotriene B4 receptor 2, i.e. BLT2. [47] This receptor activation may mediate, at least in part, certain cell-stimulating activities of the two metabolites. BLT2 may be responsible in part or whole for mediating the growth-promoting and anti-apoptosis (i.e. anti-cell death) activities of 15(S)-HETE in cultured human breast cancer cells; [48] human cancer colon cells, [49] human hepatocellular HepG2 and SMMC7721 cancer cells; [50] mouse 3T3 cells (a fibroblast cell line); [51] rat PA adventitia fibroblasts; [52] baby hamster kidney cells; [53] and diverse types of vascular endothelial cells. [54] [55] [56] [57] These growth-stimulating effects could contribute to the progression of the cited cancer types in animal models or even humans [48] [49] and the excess fibrosis that causes the narrowing of pulmonary arteries in hypoxia-induced pulmonary hypertension [51] or narrowing of portal arteries in the portal hypertension accompanying liver cirrhosis. [58] 15(S)-HETE may also act through BLT2 to stimulate an immediate contractile response in rat pulmonary arteries [59] and its angiogenic effect on human umbilical [55] and dermal [54] vascular endothelial cells.

15(S)-HpETE and 15(S)-HETE also directly bind with and activate peroxisome proliferator-activated receptor gamma. [60] This activation may contribute to the ability of 15(S)-HETE to inhibit the growth of cultured human prostate cancer PC-3, LNCaP, and DU145 cell lines and non-malignant human prostate cells; [61] [62] lung adenocarcinoma A549 cells; [63] human colorectal cancer cells; [64] corneal epithelial cells; [65] and Jurkat T-cell leukemia cells. [66] The decline in the level of 15(S)-HpETE-forming enzymes and consequential fall in cellular 15-HETE production that occurs in human prostate cancer cells may be one mechanism by which this and perhaps other human cancer cells (e.g. those of the colon, rectum, and lung) avoid the apoptosis-inducing actions of 15(S)-HpETE and/or 15(S)-HETE and thereby proliferate and spread. [67] [68] In this scenario, 15(S)-HETE and one of its forming enzymes, particularly 15-LOX-2, appear to act as tumor suppressors.

Some of the inhibitory effects of 15(S)-HpETE and 15(S)-HETE, particularly when induced by high concentrations (e.g. >1-10 micromolar), may be due to a less specific mechanism: 15(S)-HpETE and to a lesser extent 15(S)-HETE induce the generation of reactive oxygen species. These species trigger cells to activate their death programs, i.e. apoptosis, and/or are openly toxic to the cells. [69] [70] [66] [71] [72] 15(S)-HpETE and 15(S)-HETE inhibit angiogenesis and the growth of cultured human chronic myelogenous leukemia K-562 cells by a mechanism that is associated with the production of reactive oxygen species. [55] [73] [74]

Several bifunctional electrophilic breakdown products of 15(S)-HpETE, e.g. 4-hydroxy-2(E)-nonenal, 4-hydroperoxy-2(E)-nonenal, 4-oxo-2(E)-nonenal, and cis-4,5-epoxy-2(E)-decanal, are mutagens in mammalian cells and thereby may contripute to the development and/or progression of human cancers. [38]

15(R)-HETE

Similar to 15(S)-HpETE and 15(S)-HETE and with similar potency, 15(R)-HETE binds with and activates peroxisome proliferator-activated receptor gamma. [60] The precursor of 15(R)-HETE, 15(R)-HpETE may, similar to 15(S)-HpETE, break down to the mutagenic products 4-hydroxy-2(E)-nonenal, 4-hydroperoxy-2(E)-nonenal, 4-oxo-2(E)-nonenal, and cis-4,5-epoxy-2(E)-decanal and therefore be involved in cancer development and/or progression. [38]

15-Oxo-ETE

In cultured human monocytes of the THP1 cell line, 15-oxo-ETE inactivates IKKβ (also known as IKK2) thereby blocking this cell's NF-κB-mediated pro-inflammatory responses (e.g. lipopolysaccharide-induced production of TNFα, interleukin 6, and IL1B) while concurrently activating anti-oxidant responses upregulated through the anti-oxidant response element (ARE) by forcing cytosolic KEAP1 to release NFE2L2 which then moves to the nucleus, binds ARE, and induces production of, e.g. hemoxygenase-1, NADPH-quinone oxidoreductase, and possibly glutamate-cysteine ligase modifier. [75] By these actions, 15-oxo-ETE may dampen inflammatory and/or oxidative stress responses. In a cell-free system, 15-oxo-ETE is a moderately potent (IC50=1 μM) inhibitor of 12-lipoxygenase but not other human lipoxygenases. [76] This effect could also have anti-inflammatory and anti-oxidative effects by blocking the formation of 12-HETE and hepoxilins. 15-Oxo-ETE is an example of an α,β unsaturated ketone electrophile. These ketones are highly reactive with nucleophiles, adducting to, for example, the cysteines in transcription and transcription-related regulatory factors and enzymes to form their alkylated and thereby often inactivated products. [76] [77] It is presumed that the preceding activities of 15-oxo-ETE reflect its adduction to the indicated elements. [75] 15-Oxo-ETE, at 2-10 μM, also inhibits the proliferation of cultured human umbilical vein endothelial cells and LoVo human colorectal cancer cells [78] [79] and at the extremely high concentration of 100 μM inhibits the proliferation of cultured MBA-MD-231 and MCF7 breast cancer cells as well as SKOV3 ovarian cancer cells. [80] They may use a similar "protein-adduction" mechanism; if so the target protein(s) for these effects have not been defined or even suggested. This 15-oxo-ETE action may prove to inhibit the remodeling of blood vessels and reduce the growth of the cited cell types and cancers. At sub-micromolar concentrations, 15-oxo-ETE has weak chemotaxis activity for human monocytes and could serve to recruit this white blood cell into inflammatory responses. [81]

5-Oxo-15(S)-hydroxy-ETE

5-Oxo-15(S)-hydroxy-ETE is properly a member of the 5-HETE family of agonists which binds to the oxoeicosanoid receptor 1, a G protein-coupled receptor, to activate its various target cells. As such, it is a potent stimulator of leukocytes, particularly eosinophils, as well as other OXE1-bearing cells including MDA-MB-231, MCF7, and SKOV3 cancer cells (see 5-Hydroxyicosatetraenoic acid and 5-Oxo-eicosatetraenoic acid). [82] It also binds with and activates PPARγ and thereby can stimulate or inhibit cells independently of OXE1. [80]

Lipoxins

LXA4, LXB4, AT-LXA4, and AT-LXB4 are specialized proresolving mediators, i.e. they potently inhibit the progression and contribute to the resolution of diverse inflammatory and allergic reactions.

Eoxins

Eoxin A4, eoxin C4, eoxin D4, and eoxin E4 are analogs of leukotriene A4, C4, leukotriene D4, and E4. Formation of the leukotrienes is initiated by 5-lipoxygenase metabolism of arachidonic acid to form a 5,6-epoxide viz, leukotriene A4; the latter metabolite is then converted to C4, D4, and E4 in succession. Formation of the eoxins is initiated by a 15-lipoxyenase-mediated metabolism of arachiconic acid to a 14,15-epoxide, eoxin A4 followed by its serial conversion to epoxins C4, D4, and E4 using the same pathways and enzymes that metabolize leukotriene A4 to its down-stream products. Preliminary studies have found that the eoxins have pro-inflammatory actions, suggest that they are involved in severe asthma, aspirin-induced asthma attacks, and perhaps other allergic reactions. The production of eoxins by Reed-Sternburg cells has also led to suggestion that they are involve in the lymphoma of Hodgkins disease. [27] Drugs blocking the 15-lipoxygenases may be useful for inhibiting inflammation by reducing the production of the eoxins. [83]

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">Eicosanoid</span> Class of compounds

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. Eicosanoids may also act as endocrine agents to control the function of distant cells.

<span class="mw-page-title-main">Lipoxin</span> Acronym for lipoxygenase interaction product

A lipoxin (LX or Lx), an acronym for lipoxygenase interaction product, is a bioactive autacoid metabolite of arachidonic acid made by various cell types. They are categorized as nonclassic eicosanoids and members of the specialized pro-resolving mediators (SPMs) family of polyunsaturated fatty acid (PUFA) metabolites. Like other SPMs, LXs form during, and then act to resolve, inflammatory responses. Initially, two lipoxins were identified, lipoxin A4 (LXA4) and LXB4, but more recent studies have identified epimers of these two LXs: the epi-lipoxins, 15-epi-LXA4 and 15-epi-LXB4 respectively.

<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.

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">ALOX12</span> Protein-coding gene in the species Homo sapiens

ALOX12, also known as arachidonate 12-lipoxygenase, 12-lipoxygenase, 12S-Lipoxygenase, 12-LOX, and 12S-LOX is a lipoxygenase-type enzyme that in humans is encoded by the ALOX12 gene which is located along with other lipoyxgenases on chromosome 17p13.3. ALOX12 is 75 kilodalton protein composed of 663 amino acids.

<span class="mw-page-title-main">GPR31</span> Protein in humans

G-protein coupled receptor 31 also known as 12-(S)-HETE receptor is a protein that in humans is encoded by the GPR31 gene. The human gene is located on chromosome 6q27 and encodes a G-protein coupled receptor protein composed of 319 amino acids.

<span class="mw-page-title-main">Oxoeicosanoid receptor 1</span> Protein-coding gene in the species Homo sapiens

Oxoeicosanoid receptor 1 (OXER1) also known as G-protein coupled receptor 170 (GPR170) is a protein that in humans is encoded by the OXER1 gene located on human chromosome 2p21; it is the principal receptor for the 5-Hydroxyicosatetraenoic acid family of carboxy fatty acid metabolites derived from arachidonic acid. The receptor has also been termed hGPCR48, HGPCR48, and R527 but OXER1 is now its preferred designation. OXER1 is a G protein-coupled receptor (GPCR) that is structurally related to the hydroxy-carboxylic acid (HCA) family of G protein-coupled receptors whose three members are HCA1 (GPR81), HCA2, and HCA3 ; OXER1 has 30.3%, 30.7%, and 30.7% amino acid sequence identity with these GPCRs, respectively. It is also related to the recently defined receptor, GPR31, for the hydroxyl-carboxy fatty acid 12-HETE.

<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).

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.

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">Eoxin E4</span> Chemical compound

Eoxin E4, also known as 14,15-leukotriene E4, is an eoxin. Cells make eoxins by metabolizing arachidonic acid with a 15-lipoxygenase enzyme to form 15(S)-hydroperoxyeicosapentaenoic acid (i.e. 15(S)-HpETE). This product is then converted serially to eoxin A4 (i.e. EXA4), EXC4, EXD4, and EXE4 by LTC4 synthase, an unidentified gamma-glutamyltransferase, and an unidentified dipeptidase, respectively, in a pathway which appears similar if not identical to the pathway which forms leukotreines, i.e. LTA4, LTC4, LTD4, and LTE4. This pathway is schematically shown as follows:

<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.

5-Hydroxyeicosanoid dehydrogenase (5-HEDH) or more formally, nicotinamide adenine dinucleotide phosphate (NADP+)-dependent dehydrogenase, is an enzyme that metabolizes an eicosanoid product of arachidonate 5-lipoxygenase (5-LOX), 5(S)-hydroxy-6S,8Z,11Z,14Z-eicosatetraenoic acid (i.e. 5-(S)-HETE; see 5-HETE) to its 5-keto analog, 5-oxo-eicosatetraenoic acid (i.e. 5-oxo-6S,8Z,11Z,14Z-eicosatetraenoic acid or 5-oxo-ETE). It also acts in the reverse direction, metabolizing 5-oxo-ETE to 5(S)-HETE. Since 5-oxo-ETE is 30–100-fold more potent than 5(S)-HETE in stimulating various cell types, 5-HEDH is regarded as a regulator and promoter of 5(S)HETE's and thereby 5-LOX's influences on cell function. Although 5-HEDH has been evaluated in a wide range of intact cells and in crude microsome preparations, it has not yet been evaluated for its structure, for its gene, of in pure form; furthermore, most studies on it have been conducted in human tissues.

<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.

References

  1. 1 2 Moreno, J. J. (2009). "New aspects of the role of hydroxyeicosatetraenoic acids in cell growth and cancer development". Biochemical Pharmacology. 77 (1): 1–10. doi:10.1016/j.bcp.2008.07.033. PMID   18761324.
  2. 1 2 Schneider, C; Pozzi, A (2011). "Cyclooxygenases and lipoxygenases in cancer". Cancer and Metastasis Reviews. 30 (3–4): 277–294. doi:10.1007/s10555-011-9310-3. PMC   3798028 . PMID   22002716.
  3. 1 2 3 Buckley, C. D.; Gilroy, D. W.; Serhan, C. N. (2014). "Proresolving lipid mediators and mechanisms in the resolution of acute inflammation". Immunity. 40 (3): 315–327. doi:10.1016/j.immuni.2014.02.009. PMC   4004957 . PMID   24656045.
  4. Zhu, D; Ran, Y (2012). "Role of 15-lipoxygenase/15-hydroxyeicosatetraenoic acid in hypoxia-induced pulmonary hypertension". The Journal of Physiological Sciences. 62 (3): 163–172. doi: 10.1007/s12576-012-0196-9 . PMC   10717549 . PMID   22331435. S2CID   2723454.
  5. Leeper KV (1993). "Diagnosis and treatment of pulmonary infections in adult respiratory distress syndrome". New Horizons (Baltimore, Md.). 1 (4): 550–562. PMID   8087575.
  6. Schewe, T; Halangk, W; Hiebsch, C; Rapoport, S. M. (1975). "A lipoxygenase in rabbit reticulocytes which attacks phospholipids and intact mitochondria". FEBS Letters. 60 (1): 149–152. doi: 10.1016/0014-5793(75)80439-x . PMID   6318. S2CID   46488283.
  7. Bernström K, Hammarström S (1981). "Metabolism of leukotriene D by porcine kidney". J Biol Chem. 256 (18): 9579–9582. doi: 10.1016/S0021-9258(19)68801-0 . PMID   6895224.
  8. Hopkins, N. K.; Oglesby, T. D.; Bundy, G. L.; Gorman, R. R. (1984). "Biosynthesis and metabolism of 15-hydroperoxy-5,8,11,13-eicosatetraenoic acid by human umbilical vein endothelial cells". The Journal of Biological Chemistry. 259 (22): 14048–14053. doi: 10.1016/S0021-9258(18)89853-2 . PMID   6438089.
  9. Sigal, E; Dicharry, S; Highland, E; Finkbeiner, W. E. (1992). "Cloning of human airway 15-lipoxygenase: Identity to the reticulocyte enzyme and expression in epithelium". The American Journal of Physiology. 262 (4 Pt 1): L392–L398. doi:10.1152/ajplung.1992.262.4.L392. PMID   1566855.
  10. 1 2 Brash, A. R.; Boeglin, W. E.; Chang, M. S. (1997). "Discovery of a second 15S-lipoxygenase in humans". Proceedings of the National Academy of Sciences of the United States of America. 94 (12): 6148–6152. Bibcode:1997PNAS...94.6148B. doi: 10.1073/pnas.94.12.6148 . PMC   21017 . PMID   9177185.
  11. 1 2 Mulugeta, S; Suzuki, T; Hernandez, N. T.; Griesser, M; Boeglin, W. E.; Schneider, C (2010). "Identification and absolute configuration of dihydroxy-arachidonic acids formed by oxygenation of 5S-HETE by native and aspirin-acetylated COX-2". The Journal of Lipid Research. 51 (3): 575–585. doi: 10.1194/jlr.M001719 . PMC   2817587 . PMID   19752399.
  12. Serhan, C. N.; Takano, T; Maddox, J. F. (1999). "Aspirin-Triggered 15-Epi-Lipoxin A4 and Stable Analogs of Lipoxin A4 are Potent Inhibitors of Acute Inflammation: Receptors and Pathways". Lipoxygenases and their Metabolites. Advances in Experimental Medicine and Biology. Vol. 447. pp. 133–149. doi:10.1007/978-1-4615-4861-4_13. ISBN   978-0-306-46044-9. PMID   10086190.
  13. Rowlinson, S. W.; Crews, B. C.; Goodwin, D. C.; Schneider, C; Gierse, J. K.; Marnett, L. J. (2000). "Spatial requirements for 15-(R)-hydroxy-5Z,8Z,11Z,13E-eicosatetraenoic acid synthesis within the cyclooxygenase active site of murine COX-2. Why acetylated COX-1 does not synthesize 15-(R)-hete". The Journal of Biological Chemistry. 275 (9): 6586–6591. doi: 10.1074/jbc.275.9.6586 . PMID   10692466.
  14. Oliw, E. H. (1993). "Bis-Allylic hydroxylation of linoleic acid and arachidonic acid by human hepatic monooxygenases". Biochimica et Biophysica Acta (BBA) - Lipids and Lipid Metabolism. 1166 (2–3): 258–263. doi:10.1016/0005-2760(93)90106-j. PMID   8443245.
  15. Bylund, J; Kunz, T; Valmsen, K; Oliw, E. H. (1998). "Cytochromes P450 with bisallylic hydroxylation activity on arachidonic and linoleic acids studied with human recombinant enzymes and with human and rat liver microsomes". The Journal of Pharmacology and Experimental Therapeutics. 284 (1): 51–60. PMID   9435160.
  16. Boeynaems, J. M.; Oates, J. A.; Hubbard, W. C. (1980). "Preparation and characterization of hydroperoxy-eicosatetraenoic acids (HPETEs)". Prostaglandins. 19 (1): 87–97. doi:10.1016/0090-6980(80)90156-2. PMID   7384539.
  17. O'Flaherty JT, Thomas MJ, Lees CJ, McCall CE (1981). "Neutrophil-aggregating activity of monohydroxyeicosatetraenoic acids". Am. J. Pathol. 104 (1): 55–62. PMC   1903737 . PMID   7258296.
  18. Marshall, Paul J.; Kulmacz, Richard J. (1988). "Prostaglandin H synthase: Distinct binding sites for cyclooxygenase and peroxidase substrates". Archives of Biochemistry and Biophysics. 266 (1): 162–170. doi:10.1016/0003-9861(88)90246-9. PMID   3140729.
  19. Yeh, H. C.; Tsai, A. L.; Wang, L. H. (2007). "Reaction mechanisms of 15-hydroperoxyeicosatetraenoic acid catalyzed by human prostacyclin and thromboxane synthases". Archives of Biochemistry and Biophysics. 461 (2): 159–168. doi:10.1016/j.abb.2007.03.012. PMC   2041921 . PMID   17459323.
  20. Ochi, H; Morita, I; Murota, S (1992). "Roles of glutathione and glutathione peroxidase in the protection against endothelial cell injury induced by 15-hydroperoxyeicosatetraenoic acid". Archives of Biochemistry and Biophysics. 294 (2): 407–411. doi:10.1016/0003-9861(92)90704-z. PMID   1314541.
  21. 1 2 3 Brezinski, M. E.; Serhan, C. N. (1990). "Selective incorporation of (15S)-hydroxyeicosatetraenoic acid in phosphatidylinositol of human neutrophils: Agonist-induced deacylation and transformation of stored hydroxyeicosanoids". Proceedings of the National Academy of Sciences of the United States of America. 87 (16): 6248–6252. Bibcode:1990PNAS...87.6248B. doi: 10.1073/pnas.87.16.6248 . PMC   54510 . PMID   2117277.
  22. 1 2 Legrand, A. B.; Lawson, J. A.; Meyrick, B. O.; Blair, I. A.; Oates, J. A. (1991). "Substitution of 15-hydroxyeicosatetraenoic acid in the phosphoinositide signaling pathway". The Journal of Biological Chemistry. 266 (12): 7570–7577. doi: 10.1016/S0021-9258(20)89485-X . PMID   1850411.
  23. 1 2 3 Bergholte, J. M.; Soberman, R. J.; Hayes, R; Murphy, R. C.; Okita, R. T. (1987). "Oxidation of 15-hydroxyeicosatetraenoic acid and other hydroxy fatty acids by lung prostaglandin dehydrogenase". Archives of Biochemistry and Biophysics. 257 (2): 444–450. doi:10.1016/0003-9861(87)90589-3. PMID   3662534.
  24. 1 2 3 4 Hammond, V. J.; Morgan, A. H.; Lauder, S; Thomas, C. P.; Brown, S; Freeman, B. A.; Lloyd, C. M.; Davies, J; Bush, A; Levonen, A. L.; Kansanen, E; Villacorta, L; Chen, Y. E.; Porter, N; Garcia-Diaz, Y. M.; Schopfer, F. J.; O'Donnell, V. B. (2012). "Novel keto-phospholipids are generated by monocytes and macrophages, detected in cystic fibrosis, and activate peroxisome proliferator-activated receptor-γ". Journal of Biological Chemistry. 287 (50): 41651–41666. doi: 10.1074/jbc.M112.405407 . PMC   3516716 . PMID   23060450.
  25. Alpert, S. E.; Walenga, R. W.; Mandal, A; Bourbon, N; Kester, M (1999). "15-HETE-substituted diglycerides selectively regulate PKC isotypes in human tracheal epithelial cells". The American Journal of Physiology. 277 (3 Pt 1): L457–L464. doi:10.1152/ajplung.1999.277.3.L457. PMID   10484452.
  26. 1 2 3 Feltenmark, S; Gautam, N; Brunnström, A; Griffiths, W; Backman, L; Edenius, C; Lindbom, L; Björkholm, M; Claesson, H. E. (2008). "Eoxins are proinflammatory arachidonic acid metabolites produced via the 15-lipoxygenase-1 pathway in human eosinophils and mast cells". Proceedings of the National Academy of Sciences. 105 (2): 680–685. Bibcode:2008PNAS..105..680F. doi: 10.1073/pnas.0710127105 . PMC   2206596 . PMID   18184802.
  27. 1 2 3 Claesson, H. E. (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.
  28. 1 2 Sachs-Olsen, C; Sanak, M; Lang, A. M.; Gielicz, A; Mowinckel, P; Lødrup Carlsen, K. C.; Carlsen, K. H.; Szczeklik, A (2010). "Eoxins: A new inflammatory pathway in childhood asthma". Journal of Allergy and Clinical Immunology. 126 (4): 859–867.e9. doi:10.1016/j.jaci.2010.07.015. PMID   20920774. S2CID   1137911.
  29. Jubiz, W; Rådmark, O; Lindgren, J. A.; Malmsten, C; Samuelsson, B (1981). "Novel leukotrienes: Products formed by initial oxygenation of arachidonic acid at C-15". Biochemical and Biophysical Research Communications. 99 (3): 976–986. doi:10.1016/0006-291x(81)91258-4. PMID   7247953.
  30. Maas, R. L.; Brash, A. R.; Oates, J. A. (1981). "A second pathway of leukotriene biosynthesis in porcine leukocytes". Proceedings of the National Academy of Sciences of the United States of America. 78 (9): 5523–5527. Bibcode:1981PNAS...78.5523M. doi: 10.1073/pnas.78.9.5523 . PMC   348778 . PMID   6272308.
  31. Kühn, H; Barnett, J; Grunberger, D; Baecker, P; Chow, J; Nguyen, B; Bursztyn-Pettegrew, H; Chan, H; Sigal, E (1993). "Overexpression, purification and characterization of human recombinant 15-lipoxygenase". Biochimica et Biophysica Acta (BBA) - Lipids and Lipid Metabolism. 1169 (1): 80–89. doi:10.1016/0005-2760(93)90085-n. PMID   8334154.
  32. 1 2 Vogler, S; Zimmermann, N; Leopold, C; De Joncheere, K (2011). "Pharmaceutical policies in European countries in response to the global financial crisis". Southern Med Review. 4 (2): 69–79. doi:10.5655/smr.v4i2.1004 (inactive 2024-04-05). PMC   3471176 . PMID   23093885.{{cite journal}}: CS1 maint: DOI inactive as of April 2024 (link)
  33. Chawengsub, Y; Gauthier, K. M.; Campbell, W. B. (2009). "Role of arachidonic acid lipoxygenase metabolites in the regulation of vascular tone". AJP: Heart and Circulatory Physiology. 297 (2): H495–H507. doi:10.1152/ajpheart.00349.2009. PMC   2724209 . PMID   19525377.
  34. Chawengsub, Y; Gauthier, K. M.; Nithipatikom, K; Hammock, B. D.; Falck, J. R.; Narsimhaswamy, D; Campbell, W. B. (2009). "Identification of 13-hydroxy-14,15-epoxyeicosatrienoic acid as an acid-stable endothelium-derived hyperpolarizing factor in rabbit arteries". Journal of Biological Chemistry. 284 (45): 31280–31290. doi: 10.1074/jbc.M109.025627 . PMC   2781526 . PMID   19737933.
  35. Bui, P; Imaizumi, S; Beedanagari, S. R.; Reddy, S. T.; Hankinson, O (2011). "Human CYP2S1 metabolizes cyclooxygenase- and lipoxygenase-derived eicosanoids". Drug Metabolism and Disposition. 39 (2): 180–190. doi:10.1124/dmd.110.035121. PMC   3033693 . PMID   21068195.
  36. Brash, A. R.; Yu, Z; Boeglin, W. E.; Schneider, C (2007). "The hepoxilin connection in the epidermis". FEBS Journal. 274 (14): 3494–3502. doi: 10.1111/j.1742-4658.2007.05909.x . PMID   17608720. S2CID   9799021.
  37. 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.
  38. 1 2 3 4 5 Lee, S. H.; Williams, M. V.; Dubois, R. N.; Blair, I. A. (2005). "Cyclooxygenase-2-mediated DNA damage". Journal of Biological Chemistry. 280 (31): 28337–28346. doi: 10.1074/jbc.M504178200 . PMID   15964853.
  39. 1 2 Lee, S. H.; Rangiah, K; Williams, M. V.; Wehr, A. Y.; Dubois, R. N.; Blair, I. A. (2007). "Cyclooxygenase-2-mediated metabolism of arachidonic acid to 15-oxo-eicosatetraenoic acid by rat intestinal epithelial cells". Chemical Research in Toxicology. 20 (11): 1665–1675. doi:10.1021/tx700130p. PMID   17910482.
  40. Brinckmann, R; Schnurr, K; Heydeck, D; Rosenbach, T; Kolde, G; Kühn, H (1998). "Membrane translocation of 15-lipoxygenase in hematopoietic cells is calcium-dependent and activates the oxygenase activity of the enzyme". Blood. 91 (1): 64–74. doi: 10.1182/blood.V91.1.64 . PMID   9414270.
  41. Maskrey, B. H.; Bermúdez-Fajardo, A; Morgan, A. H.; Stewart-Jones, E; Dioszeghy, V; Taylor, G. W.; Baker, P. R.; Coles, B; Coffey, M. J.; Kühn, H; O'Donnell, V. B. (2007). "Activated platelets and monocytes generate four hydroxyphosphatidylethanolamines via lipoxygenase". Journal of Biological Chemistry. 282 (28): 20151–20163. doi: 10.1074/jbc.M611776200 . PMID   17519227.
  42. Thomas, C. P.; Morgan, L. T.; Maskrey, B. H.; Murphy, R. C.; Kühn, H; Hazen, S. L.; Goodall, A. H.; Hamali, H. A.; Collins, P. W.; O'Donnell, V. B. (2010). "Phospholipid-esterified eicosanoids are generated in agonist-activated human platelets and enhance tissue factor-dependent thrombin generation". Journal of Biological Chemistry. 285 (10): 6891–6903. doi: 10.1074/jbc.M109.078428 . PMC   2844139 . PMID   20061396.
  43. 1 2 Serhan, C. N. (2005). "Lipoxins and aspirin-triggered 15-epi-lipoxins are the first lipid mediators of endogenous anti-inflammation and resolution". Prostaglandins, Leukotrienes and Essential Fatty Acids. 73 (3–4): 141–162. doi:10.1016/j.plefa.2005.05.002. PMID   16005201.
  44. Maas, R. L.; Turk, J; Oates, J. A.; Brash, A. R. (1982). "Formation of a novel dihydroxy acid from arachidonic acid by lipoxygenase-catalyzed double oxygenation in rat mononuclear cells and human leukocytes". The Journal of Biological Chemistry. 257 (12): 7056–67. doi: 10.1016/S0021-9258(18)34537-X . PMID   6806263.
  45. Serhan, C. N. (1989). "On the relationship between leukotriene and lipoxin production by human neutrophils: Evidence for differential metabolism of 15-HETE and 5-HETE". Biochimica et Biophysica Acta (BBA) - Lipids and Lipid Metabolism. 1004 (2): 158–168. doi:10.1016/0005-2760(89)90264-6. PMID   2546590.
  46. 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.
  47. Yokomizo, T; Kato, K; Hagiya, H; Izumi, T; Shimizu, T (2001). "Hydroxyeicosanoids bind to and activate the low affinity leukotriene B4 receptor, BLT2". Journal of Biological Chemistry. 276 (15): 12454–12459. doi: 10.1074/jbc.M011361200 . PMID   11278893.
  48. 1 2 O'Flaherty, J. T.; Wooten, R. E.; Samuel, M. P.; Thomas, M. J.; Levine, E. A.; Case, L. D.; Akman, S. A.; Edwards, I. J. (2013). "Fatty acid metabolites in rapidly proliferating breast cancer". PLOS ONE. 8 (5): e63076. Bibcode:2013PLoSO...863076O. doi: 10.1371/journal.pone.0063076 . PMC   3642080 . PMID   23658799.
  49. 1 2 Cabral, M; Martín-Venegas, R; Moreno, J. J. (2013). "Role of arachidonic acid metabolites on the control of non-differentiated intestinal epithelial cell growth". The International Journal of Biochemistry & Cell Biology. 45 (8): 1620–1628. doi:10.1016/j.biocel.2013.05.009. PMID   23685077.
  50. Ma, J; Zhang, L; Zhang, J; Liu, M; Wei, L; Shen, T; Ma, C; Wang, Y; Chen, Y; Zhu, D (2013). "15-lipoxygenase-1/15-hydroxyeicosatetraenoic acid promotes hepatocellular cancer cells growth through protein kinase B and heat shock protein 90 complex activation". The International Journal of Biochemistry & Cell Biology. 45 (6): 1031–1041. doi:10.1016/j.biocel.2013.02.018. PMID   23474367.
  51. 1 2 Nieves, D; Moreno, J. J. (2006). "Hydroxyeicosatetraenoic acids released through the cytochrome P-450 pathway regulate 3T6 fibroblast growth". The Journal of Lipid Research. 47 (12): 2681–9. doi: 10.1194/jlr.M600212-JLR200 . PMID   16980726.
  52. Zhang, L; Li, Y; Chen, M; Su, X; Yi, D; Lu, P; Zhu, D (2014). "15-LO/15-HETE mediated vascular adventitia fibrosis via p38 MAPK-dependent TGF-β". Journal of Cellular Physiology. 229 (2): 245–257. doi: 10.1002/jcp.24443 . PMID   23982954. S2CID   311866.
  53. Kiran Kumar, Y. V.; Raghunathan, A; Sailesh, S; Prasad, M; Vemuri, M. C.; Reddanna, P (1993). "Differential effects of 15-HPETE and 15-HETE on BHK-21 cell proliferation and macromolecular composition". Biochimica et Biophysica Acta (BBA) - Lipids and Lipid Metabolism. 1167 (1): 102–108. doi:10.1016/0005-2760(93)90223-v. PMID   8384883.
  54. 1 2 Zhang, B; Cao, H; Rao, G. N. (2005). "15(S)-hydroxyeicosatetraenoic acid induces angiogenesis via activation of PI3K-Akt-mTOR-S6K1 signaling". Cancer Research. 65 (16): 7283–7291. doi: 10.1158/0008-5472.CAN-05-0633 . PMID   16103079.
  55. 1 2 3 Soumya, S. J.; Binu, S; Helen, A; Anil Kumar, K; Reddanna, P; Sudhakaran, P. R. (2012). "Effect of 15-lipoxygenase metabolites on angiogenesis: 15(S)-HPETE is angiostatic and 15(S)-HETE is angiogenic". Inflammation Research. 61 (7): 707–718. doi:10.1007/s00011-012-0463-5. PMID   22450700. S2CID   2297892.
  56. Soumya, S. J.; Binu, S; Helen, A; Reddanna, P; Sudhakaran, P. R. (2013). "15(S)-HETE-induced angiogenesis in adipose tissue is mediated through activation of PI3K/Akt/mTOR signaling pathway". Biochemistry and Cell Biology. 91 (6): 498–505. doi:10.1139/bcb-2013-0037. PMID   24219292.
  57. Li, J; Zhang, Y; Liu, Y; Shen, T; Zhang, H; Xing, Y; Zhu, D (2015). "PGC-1α plays a major role in the anti-apoptotic effect of 15-HETE in pulmonary artery endothelial cells". Respiratory Physiology & Neurobiology. 205: 84–91. doi:10.1016/j.resp.2014.10.015. PMID   25447678. S2CID   27118439.
  58. Pandey, V; Sultan, M; Kashofer, K; Ralser, M; Amstislavskiy, V; Starmann, J; Osprian, I; Grimm, C; Hache, H; Yaspo, M. L.; Sültmann, H; Trauner, M; Denk, H; Zatloukal, K; Lehrach, H; Wierling, C (2014). "Comparative analysis and modeling of the severity of steatohepatitis in DDC-treated mouse strains". PLOS ONE. 9 (10): e111006. Bibcode:2014PLoSO...9k1006P. doi: 10.1371/journal.pone.0111006 . PMC   4210132 . PMID   25347188.
  59. Wang, Y; Liang, D; Wang, S; Qiu, Z; Chu, X; Chen, S; Li, L; Nie, X; Zhang, R; Wang, Z; Zhu, D (2010). "Role of the G-protein and tyrosine kinase--Rho/ROK pathways in 15-hydroxyeicosatetraenoic acid induced pulmonary vasoconstriction in hypoxic rats". Journal of Biochemistry. 147 (5): 751–764. doi:10.1093/jb/mvq010. PMID   20139061.
  60. 1 2 Naruhn, S; Meissner, W; Adhikary, T; Kaddatz, K; Klein, T; Watzer, B; Müller-Brüsselbach, S; Müller, R (2010). "15-hydroxyeicosatetraenoic acid is a preferential peroxisome proliferator-activated receptor beta/delta agonist". Molecular Pharmacology. 77 (2): 171–184. doi:10.1124/mol.109.060541. PMID   19903832. S2CID   30996954.
  61. Shappell, S. B.; Gupta, R. A.; Manning, S; Whitehead, R; Boeglin, W. E.; Schneider, C; Case, T; Price, J; Jack, G. S.; Wheeler, T. M.; Matusik, R. J.; Brash, A. R.; Dubois, R. N. (2001). "15S-Hydroxyeicosatetraenoic acid activates peroxisome proliferator-activated receptor gamma and inhibits proliferation in PC3 prostate carcinoma cells". Cancer Research. 61 (2): 497–503. PMID   11212240.
  62. Tang, S; Bhatia, B; Maldonado, C. J.; Yang, P; Newman, R. A.; Liu, J; Chandra, D; Traag, J; Klein, R. D.; Fischer, S. M.; Chopra, D; Shen, J; Zhau, H. E.; Chung, L. W.; Tang, D. G. (2002). "Evidence that arachidonate 15-lipoxygenase 2 is a negative cell cycle regulator in normal prostate epithelial cells". Journal of Biological Chemistry. 277 (18): 16189–16201. doi: 10.1074/jbc.M111936200 . PMID   11839751.
  63. Kudryavtsev, I. A.; Golenko, O. D.; Gudkova, M. V.; Myasishcheva, N. V. (2002). "Arachidonic acid metabolism in growth control of A549 human lung adenocarcinoma cells". Biochemistry. Biokhimiia. 67 (9): 1021–1026. doi:10.1023/A:1020526119866. PMID   12387716. S2CID   27912248.
  64. Chen, G. G.; Xu, H; Lee, J. F.; Subramaniam, M; Leung, K. L.; Wang, S. H.; Chan, U. P.; Spelsberg, T. C. (2003). "15-hydroxy-eicosatetraenoic acid arrests growth of colorectal cancer cells via a peroxisome proliferator-activated receptor gamma-dependent pathway". International Journal of Cancer. 107 (5): 837–843. doi:10.1002/ijc.11447. PMID   14566836. S2CID   36953974.
  65. Chang, M. S.; Schneider, C; Roberts, R. L.; Shappell, S. B.; Haselton, F. R.; Boeglin, W. E.; Brash, A. R. (2005). "Detection and subcellular localization of two 15S-lipoxygenases in human cornea". Investigative Ophthalmology & Visual Science. 46 (3): 849–856. doi: 10.1167/iovs.04-1166 . PMID   15728540.
  66. 1 2 Kumar, K. A.; Arunasree, K. M.; Roy, K. R.; Reddy, N. P.; Aparna, A; Reddy, G. V.; Reddanna, P (2009). "Effects of (15S)-hydroperoxyeicosatetraenoic acid and (15S)-hydroxyeicosatetraenoic acid on the acute- lymphoblastic-leukaemia cell line Jurkat: Activation of the Fas-mediated death pathway". Biotechnology and Applied Biochemistry. 52 (Pt 2): 121–133. doi:10.1042/BA20070264. PMID   18494609. S2CID   19055952.
  67. Shappell, S. B.; Boeglin, W. E.; Olson, S. J.; Kasper, S; Brash, A. R. (1999). "15-lipoxygenase-2 (15-LOX-2) is expressed in benign prostatic epithelium and reduced in prostate adenocarcinoma". The American Journal of Pathology. 155 (1): 235–245. doi:10.1016/S0002-9440(10)65117-6. PMC   1866677 . PMID   10393855.
  68. Tang, D. G.; Bhatia, B; Tang, S; Schneider-Broussard, R (2007). "15-lipoxygenase 2 (15-LOX2) is a functional tumor suppressor that regulates human prostate epithelial cell differentiation, senescence, and growth (size)". Prostaglandins & Other Lipid Mediators. 82 (1–4): 135–146. doi:10.1016/j.prostaglandins.2006.05.022. PMID   17164141.
  69. Ochi, H; Morita, I; Murota, S (1992). "Mechanism for endothelial cell injury induced by 15-hydroperoxyeicosatetraenoic acid, an arachidonate lipoxygenase product". Biochimica et Biophysica Acta (BBA) - Molecular Cell Research. 1136 (3): 247–252. doi:10.1016/0167-4889(92)90113-p. PMID   1520701.
  70. MacCarrone, M; Ranalli, M; Bellincampi, L; Salucci, M. L.; Sabatini, S; Melino, G; Finazzi-Agrò, A (2000). "Activation of different lipoxygenase isozymes induces apoptosis in human erythroleukemia and neuroblastoma cells". Biochemical and Biophysical Research Communications. 272 (2): 345–350. doi:10.1006/bbrc.2000.2597. PMID   10833416.
  71. Dymkowska, D; Wojtczak, L (2009). "Arachidonic acid-induced apoptosis in rat hepatoma AS-30D cells is mediated by reactive oxygen species". Acta Biochimica Polonica. 56 (4): 711–715. doi: 10.18388/abp.2009_2506 . PMID   19949744.
  72. Cells. 32:1021-1027, 2011
  73. Soumya, S. J.; Binu, S; Helen, A; Reddanna, P; Sudhakaran, P. R. (2014). "15-LOX metabolites and angiogenesis: Angiostatic effect of 15(S)-HPETE involves induction of apoptosis in adipose endothelial cells". PeerJ. 2: e635. doi: 10.7717/peerj.635 . PMC   4207198 . PMID   25346880.
  74. Mahipal, S. V.; Subhashini, J; Reddy, M. C.; Reddy, M. M.; Anilkumar, K; Roy, K. R.; Reddy, G. V.; Reddanna, P (2007). "Effect of 15-lipoxygenase metabolites, 15-(S)-HPETE and 15-(S)-HETE on chronic myelogenous leukemia cell line K-562: Reactive oxygen species (ROS) mediate caspase-dependent apoptosis". Biochemical Pharmacology. 74 (2): 202–214. doi:10.1016/j.bcp.2007.04.005. PMID   17517376.
  75. 1 2 Snyder, N. W.; Golin-Bisello, F; Gao, Y; Blair, I. A.; Freeman, B. A.; Wendell, S. G. (2015). "15-Oxoeicosatetraenoic acid is a 15-hydroxyprostaglandin dehydrogenase-derived electrophilic mediator of inflammatory signaling pathways". Chemico-Biological Interactions. 234: 144–153. Bibcode:2015CBI...234..144S. doi:10.1016/j.cbi.2014.10.029. PMC   4414684 . PMID   25450232.
  76. 1 2 Armstrong, M. M.; Diaz, G; Kenyon, V; Holman, T. R. (2014). "Inhibitory and mechanistic investigations of oxo-lipids with human lipoxygenase isozymes". Bioorganic & Medicinal Chemistry. 22 (15): 4293–4297. doi:10.1016/j.bmc.2014.05.025. PMC   4112157 . PMID   24924423.
  77. Delmastro-Greenwood, M; Freeman, B. A.; Wendell, S. G. (2014). "Redox-dependent anti-inflammatory signaling actions of unsaturated fatty acids". Annual Review of Physiology. 76: 79–105. doi:10.1146/annurev-physiol-021113-170341. PMC   4030715 . PMID   24161076.
  78. Wei, C; Zhu, P; Shah, S. J.; Blair, I. A. (2009). "15-oxo-Eicosatetraenoic acid, a metabolite of macrophage 15-hydroxyprostaglandin dehydrogenase that inhibits endothelial cell proliferation". Molecular Pharmacology. 76 (3): 516–525. doi:10.1124/mol.109.057489. PMC   2730384 . PMID   19535459.
  79. Snyder, N. W.; Revello, S. D.; Liu, X; Zhang, S; Blair, I. A. (2013). "Cellular uptake and antiproliferative effects of 11-oxo-eicosatetraenoic acid". The Journal of Lipid Research. 54 (11): 3070–3077. doi: 10.1194/jlr.M040741 . PMC   3793611 . PMID   23945567.
  80. 1 2 O'Flaherty, J. T.; Rogers, L. C.; Paumi, C. M.; Hantgan, R. R.; Thomas, L. R.; Clay, C. E.; High, K; Chen, Y. Q.; Willingham, M. C.; Smitherman, P. K.; Kute, T. E.; Rao, A; Cramer, S. D.; Morrow, C. S. (2005). "5-Oxo-ETE analogs and the proliferation of cancer cells". Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids. 1736 (3): 228–236. doi:10.1016/j.bbalip.2005.08.009. PMID   16154383.
  81. Sozzani, S; Zhou, D; Locati, M; Bernasconi, S; Luini, W; Mantovani, A; O'Flaherty, J. T. (1996). "Stimulating properties of 5-oxo-eicosanoids for human monocytes: Synergism with monocyte chemotactic protein-1 and -3". Journal of Immunology. 157 (10): 4664–4671. doi: 10.4049/jimmunol.157.10.4664 . PMID   8906847. S2CID   23499393.
  82. O'Flaherty, J. T.; Kuroki, M; Nixon, A. B.; Wijkander, J; Yee, E; Lee, S. L.; Smitherman, P. K.; Wykle, R. L.; Daniel, L. W. (1996). "5-Oxo-eicosatetraenoate is a broadly active, eosinophil-selective stimulus for human granulocytes". Journal of Immunology. 157 (1): 336–342. doi: 10.4049/jimmunol.157.1.336 . PMID   8683135. S2CID   35264541.
  83. Sadeghian H, Jabbari A (2016). "15-Lipoxygenase inhibitors: a patent review". Expert Opinion on Therapeutic Patents. 26 (1): 65–88. doi:10.1517/13543776.2016.1113259. PMID   26560362. S2CID   20192361.