13-Hydroxyoctadecadienoic acid

Last updated
13-Hydroxyoctadecadienoic acid
13-HODE.svg
Names
Preferred IUPAC name
(9Z,11E,13S)-13-Hydroxyoctadeca-9,11-dienoic acid
Other names
13(S)-HODE, 13S-HODE
Identifiers
3D model (JSmol)
ChEBI
ChEMBL
ChemSpider
KEGG
PubChem CID
UNII
  • InChI=1S/C18H32O3/c1-2-3-11-14-17(19)15-12-9-7-5-4-6-8-10-13-16-18(20)21/h7,9,12,15,17,19H,2-6,8,10-11,13-14,16H2,1H3,(H,20,21)/b9-7-,15-12+/t17-/m0/s1
    Key: HNICUWMFWZBIFP-IRQZEAMPSA-N
  • 9Z,11E,13R:InChI=1S/C18H32O3/c1-2-3-11-14-17(19)15-12-9-7-5-4-6-8-10-13-16-18(20)21/h7,9,12,15,17,19H,2-6,8,10-11,13-14,16H2,1H3,(H,20,21)/b9-7-,15-12+/t17-/m1/s1
  • 9E,11E:CCCCCC(/C=C/C=C/CCCCCCCC(=O)O)O
  • 9Z,11E,13S:CCCCC[C@@H](/C=C/C=C\CCCCCCCC(=O)O)O
  • 9Z,11E,13R:CCCCC[C@H](/C=C/C=C\CCCCCCCC(=O)O)O
Properties
C18H32O3
Molar mass 296.451 g·mol−1
Except where otherwise noted, data are given for materials in their standard state (at 25 °C [77 °F], 100 kPa).

13-Hydroxyoctadecadienoic acid (13-HODE) is the commonly used term for 13(S)-hydroxy-9Z,11E-octadecadienoic acid (13(S)-HODE). The production of 13(S)-HODE is often accompanied by the production of its stereoisomer, 13(R)-hydroxy-9Z,11E-octadecadienoic acid (13(R)-HODE). The adjacent figure gives the structure for the (S) stereoisomer of 13-HODE. Two other naturally occurring 13-HODEs that may accompany the production of 13(S)-HODE are its cis-trans (i.e., 9E,11E) isomers viz., 13(S)-hydroxy-9E,11E-octadecadienoic acid (13(S)-EE-HODE) and 13(R)-hydroxy-9E,11E-octadecadienoic acid (13(R)-EE-HODE). Studies credit 13(S)-HODE with a range of clinically relevant bioactivities; recent studies have assigned activities to 13(R)-HODE that differ from those of 13(S)-HODE; and other studies have proposed that one or more of these HODEs mediate physiological and pathological responses, are markers of various human diseases, and/or contribute to the progression of certain diseases in humans. Since, however, many studies on the identification, quantification, and actions of 13(S)-HODE in cells and tissues have employed methods that did not distinguish between these isomers, 13-HODE is used here when the actual isomer studied is unclear.

Contents

A similar set of 9-Hydroxyoctadecadienoic acid (9-HODE) metabolites (i.e., 9(S)-HODE), 9(R)-HODE, 9(S)-EE-HODE), and 9(R)-EE-HODE) occurs naturally and particularly under conditions of oxidative stress forms concurrently with the 13-HODEs; the 9-HODEs have overlapping and complementary but not identical activities with the 13-HODEs. Some recent studies measuring HODE levels in tissue have lumped the four 9-HODEs with the four 13-HODEs to report only on total HODEs (tHODEs). tHODEs have been proposed to be markers for certain human diseases. Other studies have lumped together the 9-(S), 9(R), 13 (S)-, and 13(R)-HODEs along with the two ketone metabolites of these HODEs, 13-oxoODE (13-oxo-9Z,12E-octadecadienoic acid) and 9-oxoODE, reporting only on total OXLAMs (oxidized linoleic acid metabolites); the OXLAMs have been implicated in working together to signal for pain perception.

Pathways making 13-HODEs

15-Lipoxygenase 1

15-lipoxygenase 1 (ALOX15), while best known for converting the 20 carbon polyunsaturated fatty acid, arachidonic acid, into a series of 15-hydroxylated arachidonic acid metabolites (see 15-hydroxyicosatetraenoic acid), actually prefers as its substrate the 18 carbon polyunsaturated fatty acid, linoleic acid, over arachidonic acid, converting it to 13-hydroperoxy-9Z,11E-octadecadienoic acid (13-HpODE). [1] [2] The enzyme acts in a highly stereospecific manner, forming 13(S)-hydroperoxy-9Z,11E-octadecadienoic acid (13(S)-HpODE) but comparatively little or no 13(R)-hydroperoxy-9Z,11E-octadecadienoic acid (13(R)-HpODE) -. [3] [4] In cells, 13(S)-HpODE is rapidly reduced by peroxidases to 13(S)-HODE. [1] [5] ALOX15 is fully capable of metabolizing the linoleic acid that is bound to phospholipid [6] or cholesterol [7] to form 13(S)-HpODE-bound phospholipids and cholesterol that are rapidly converted to their corresponding 13(S)-HODE-bound products.

15-lipoxygenase 2

15-lipoxygenase type 2 (ALOX15B) strongly prefers arachidonic acid over linoleic acid and in consequence is relatively poor in metabolizing linoleic acid to 13(S)-HpODE (which is then converted to 13(S)-HODE) compared to 15-lipoxygenase 1; [8] nonetheless, it can contribute to the production of these metabolites. [2] [9]

Cyclooxygenases 1 and 2

Cyclooxygenase 1 (COX-1) and cyclooxygenase 2 (COX-2) metabolize linoleic acid to 13(S)-HODE with COX-2 exhibiting a higher preference for linoleic acid and therefore producing far more of this product than its COX-1 counterpart; [10] consequently, COX-2 appears to be the principle COX making 13(S)-HODE in cells expressing both enzymes. [11] Concurrently with their production of 13(S)-HODE, these enzymes also produce smaller amounts of 9(R)-HODE. [12] [11]

Cytochrome P450

Cytochrome P450 microsomal enzymes metabolize linoleic acid to a mixture of 13-HODEs and 9-HODEs; these reactions produce racemic mixtures in which the R stereoisomer predominates, for instance by a R/S ratio of 80%/20% for both 13-HODE and 9-HODE in human liver microsomes. [13] [14] [15]

Free radical and singlet oxygen oxidations

Oxidative stress in cells and tissues produces Free radical and singlet oxygen oxidations of linoleic acid to generate 13-HpODEs, 9-HpODEs, 13-HODEs, and 9-HODEs; these non-enzymatic reactions produce or are suspected but not proven to produce approximately equal amounts of their S and R stereoisomers. [16] [17] [18] Free radical oxidations of linoleic acid also produce 13-EE-HODE, 9-hydroxy-10E,12-E-octadecadienoic acid, 9-hydroxy-10E,12-Z-octadecadienoic acid, and 11-hydroxy-9Z,12Z-octadecadienoic acid while singlet oxygen attacks on linoleic acid produce (presumably) racemic mixtures of 9-hydroxy-10E,12-Z-octadecadienoic acid, 10-hydroxy-8E,12Z-octadecadienoic acid, and 12-hydroxy-9Z-13-E-octadecadienoic acid. [19] 4-Hydroxynonenal (i.e. 4-hydroxy-2E-nonenal or HNE) is also a peroxidation product of 13-HpODE. [20] Since oxidative stress commonly produces both free radicals and singlet oxygen, most or all of these products may form together in tissues undergoing oxidative stress. Free radical and singlet oxygen oxidations of linoleic acid produce a similar set of 13-HODE metabolites (see 9-Hydroxyoctadecadienoic acid). Studies attribute these oxidations to be major contributors to 13-HODE production in tissues undergoing oxidative stress including in humans sites of inflammation, steatohepatitis, cardiovascular disease-related atheroma plaques, neurodegenerative disease, etc. (see oxidative stress). [21] [19]

Metabolism of 13(S)-HODE

Like most polyunsaturated fatty acids and mono-hydroxyl polyunsaturated fatty acids, 13(S)-HODE is rapidly and quantitatively incorporated into phospholipids; [22] the levels of 13(S)-HODE esterified to the sn-2 position of phosphatidylcholine, phosphatidylinositol, and phosphatidylethanolamine in human psoriasis lesions are significantly lower than those in normal skin; this chain shortening pathway may be responsible for inactivating 13(S)-HODE. [23] 13(S)-HODE is also metabolized by peroxisome-dependent β-oxidations to chain-shortened 16-carbon, 14-carbon, and 12-carbon products which are released from the cell; [24] this chain-shortening pathway may serve to inactive and dispose of 13(S)-HODE.

13(S)-HODE is oxidized to 13-oxo-9Z,11E-octadecadienoic acid (13-oxo-HODE or 13-oxoODE) by a NAD+-dependent 13-HODE dehydrogenase, the protein for which has been partially purified from rat colon. [25] [26] [27] The formation of 13-oxo-ODE may represent the first step in 13(S)-HODEs peroxisome-dependent chain shortening but 13-oxo-ODE has its own areas of biological importance: it accumulates in tissues, [28] [29] is bioactive, [30] [31] and may have clinically relevance as a marker for [32] [33] and potential contributor to [33] human disease. 13-Oxo-ODE itself may react with glutathione in a non-enzymatic Michael reaction or a glutathione transferase-dependent reaction to form 13-oxo-ODE products containing an 11 trans double bound and glutathione attached to carbon 9 in a mixture of S and R diastereomers; these two diastereomers are major metabolites of 13(S)-HODE in cultured HT-29 human colon cancer cells. [34] Colonic mucosal explants from Sprague-Dawley rats and human colon cancer HT29 cells add glutathione to 13-oxo-ODE in a Michael reaction to form 13-oxo-9-glutatione-11(E)-octadecenoic acid; this conjugation reaction appears to be enzymatic and mediated by a glutathione transferase. [35] [36] Since this conjugate may be rapidly exported from the cell and has not yet been characterized for biological activity, it is not clear if this transferase reaction serves any function beyond removing 13-oxo-ODE from the cell to limit its activity. [34]

Activities

Stimulation of peroxisome proliferator-activated receptors

13-HODE, 13-oxoODE, and 13-EE-HODE (along with their 9-HODE counterparts) directly activate peroxisome proliferator-activated receptor gamma (PPARγ). [37] [38] [39] This activation appears responsible for the ability of 13-HODE (and 9-HODE) to induce the transcription of PPARγ-inducible genes in human monocytes as well as to stimulate the maturation of these cells to macrophages. [37] 13(S)-HODE (and 9(S)-HODE) also stimulate the activation of peroxisome proliferator-activated receptor beta (PPARβ) in a model cell system; 13-HODE (and 9-HODE) are also proposed to contribute to the ability of oxidized low-density lipoprotein (LDL) to activate PPARβl: LDL containing phospholipid-bound 13-HODE (and 9-HODE) is taken up by the cell and then acted on by phospholipases to release the HODEs which in turn directly activate PPARβl. [40]

Stimulation of TRPV1 receptor

13(S)-HODE, 13(R)-HODE and 13-oxoODE, along with their 9-HODE counterparts, also act on cells through TRPV1. TRPV1 is the transient receptor potential cation channel subfamily V member 1 receptor (also termed capsaicin receptor or vanilloid receptor 1). These 6 HODEs, dubbed, oxidized linoleic acid metabolites (OXLAMs), individually but also and possibly to a greater extent when acting together, stimulate TRPV1-dependent responses in rodent neurons, rodent and human bronchial epithelial cells, and in model cells made to express rodent or human TRPV1. This stimulation appears due to a direct interaction of these agents on TRPV1 although reports disagree on the potencies of the (OXLAMs) with, for example, the most potent OXLAM, 9(S)-HODE, requiring at least 10 micromoles/liter [41] or a more physiological concentration of 10 nanomoles/liter [30] to activate TRPV1 in rodent neurons. The OXLAM-TRPV1 interaction is credited with mediating pain sensation in rodents (see below).

Stimulation of GPR132 receptor

13(S)-HpODE, and 13(S)-HODE directly activate human (but not mouse) GPR132 (G protein coupled receptor 132, also termed G2A) in Chinese hamster ovary cells made to express these receptors; they are, however, far weaker GPR132 activators than 9(S)-HpODE or 9(S)-HODE. [42] [43] GPR132 was initially described as a pH sensing receptor; the role(s) of 13(S)-HpODE and 13(S)-HODE as well as 9(S)-HpODE, 9(S)HODE, and a series or GPR132-activating arachidonic acid hydroxy metabolites (i.e. HETEs) in activating G2A under the physiological and pathological conditions in which G2A is implicated (see GPR132 for a lists of these conditions) have not yet been determined. This determination, as it might apply to humans, is made difficult by the inability of these HODEs to activate rodent GPR132 and therefore to be analyzed in rodent models.

Involvement in mitochondria degradation

In the maturation of the red blood cell lineage (see erythropoiesis) from mitochondria-bearing reticulocytes to mature mitochondria-free erythrocytes in rabbits, the mitochondria accumulate phospholipid-bound 13(S)-HODE in their membranes due to the action of a lipoxygenase which (in rabbits, mice, and other sub-primate vertebrates) directly metabolizes linoleic acid-bound phospholipid to 13(S)-HpODE-bound phospholipid which is rapidly reduced to 13(S)-HODE-bound phospholipid. [44] It is suggested that the accumulation of phospholipid-bound 13(S)-HpODE and/or 13(S)-HODE is a critical step in rendering mitochondria more permeable thereby triggering their degradation and thence maturation to erythrocytes. [44] [45] However, functional inactivation of the phospholipid-attacking lipoxygenase gene in mice does not cause major defects in erythropoiesis. [46] It is suggested that mitochondrial degradation proceeds through at least two redundant pathways besides that triggered by lipoxygenase-dependent formation of 13(S)-HpODE- and 13(S)-HODE-bound phospholipids viz., mitochondrial digestion by autophagy and mitochondrial exocytosis. [47] In all events, formation of 13(S)-HODE bound to phospholipid in mitochondrial membranes is one pathway by which they become more permeable and thereby subject to degradation and, as consequence of their release of deleterious elements, to cause cell injury. [48]

Stimulation of blood leukocytes

13-HODE (and 9-HODE) are moderately strong stimulators of the directed migration (i.e. chemotaxis) of cow and human neutrophils in vitro [49] whereas 13(R)-HODE (and 9(R)-HODE, and 9(S)-HODE) are weak stimulators of the in vitro directed migration of the human cytotoxic and potentially tissue-injuring lymphocytes, i.e. natural killer cells. [50] These effects may contribute to the pro-inflammatory and tissue-injuring actions ascribed to 13-HODEs (and 9-HODEs).

Involvement in human diseases

Atherosclerosis

In atherosclerosis, an underlying cause of Coronary artery disease and strokes, atheromatous plaques accumulate in the vascular tunica intima thereby narrowing blood vessel size and decreasing blood flow. In an animal model and in humans 13-HODE (primarily esterified to cholesterol, phospholipids, and possibly other lipids) is a dominant component of these plaques. [51] [52] [53] [54] Since these studies found that early into the progression of the plaques, 13-HODE consisted primarily of the S stereoisomer while more mature plaques contained equal amounts of S and R stereoisomers, it was suggested that 15-LOX-1 contributes to early accumulation while cytochrome and/or free radical pathways contributes to the later accumulation of the plaques. Further studies suggest that 13(S)-HODE contributes to plaque formation by activating the transcription factor, PPARγ (13(R)-HODE lacks this ability [55] ), which in turn stimulates the production of two receptors on the surface of macrophages resident in the plaques, 1) CD36, a scavenger receptor for oxidized low density lipoproteins, native lipoproteins, oxidized phospholipids, and long-chain fatty acids, and 2) adipocyte protein 2 (aP2), a fatty acid binding protein; this may cause macrophages to increase their uptake of these lipids, transition to lipid-laden foam cells, and thereby increase plaque size. [56] The 13(S)-HODE/PPARγ axis also causes macrophages to self-destruct by activating apoptosis-inducing pathways;, this effect may also contribute to increases in plaque size. [57] These studies suggest that 13-HODE-producing metabolic pathways, [56] PPARγ, [56] [57] CD36, [58] and aP2 [59] may be therapeutic targets for treating atherosclerosis-related diseases. Indeed, Statins, which are known to suppress cholesterol synthesis by inhibiting an enzyme in the cholesterol synthesis pathway, 3-hydroxy-3-methyl-glutaryl-CoA reductase HMG-CoA reductase, are widely used to prevent atherosclerosis and atherosclerosis-related diseases. Statins also inhibit PPARγ in human macrophages, vascular endothelial cells, and smooth muscle cells; this action may contribute to their anti-atherogenic effect. [60]

Asthma

In guinea pigs, 13(S)-HODE, when injected intravenously, causes a narrowing of lung airways and, when inhaled as an aerosol, mimics the asthmatic hypersensitivity to agents that cause bronchoconstriction by increasing airway narrowing responses to methacholine and histamine. [61] In a mouse model of allergen-induced asthma, 13-HODE levels are elevated, [62] in the latter mouse model, the injection of antibody directed against 13(S)-HODE reduced many of the pathological and physiological features of asthma. [48] mouse forced to overexpress in lung the mouse enzyme (12/15-lipoxygenase) that metabolizes linoleic acid to 13(S)-HODE exhibited elevated levels of this metabolite in lung as well as various pathological and physiological features of asthma, [62] and the instillation of 13(S)HODE replicated many of these features of asthma, [48] In the mouse model of asthma and in the human disease, epithelial cells of lung airways show various pathological changes including disruption of their mitochondria [48] [62] [63] 13(S)-HODE causes similar disruptive changes in the mitochondria of cultured Beas 2B human airway epithelial cells. [48] Furthermore, humans with asthma exhibit increased levels of 13-HODE in their blood, sputum, and washings form their lung alveola (i.e. bronchoalveolar lavage fluid of BAL) and human eosinophils, which are implicated in contributing to human asthma, metabolize linoleic acid to 13-HODE (and 9-HODE) to a far greater extent than any other type of leukocyte. [64] The mechanism responsible for 13-HODE's impact on airway epithelial cells may involve its activation of the TRPV1 receptor (see previous section on TRPV1): this receptor is highly expressed in mouse and human airway epithelial cells and in Beas 2B human airway epithelial cells and, furthermore, suppression of TRPV1 expression as well as a TPRV1 receptor inhibitor (capsazepan) block mouse airway responses to 13(S)-HODE. [48] While much further work is needed, these pre-clinical studies allow that 13(S)-HODE, made at least in part by eosinophils and operating through TRPV1, may be responsible for the airways damage which occurs in the more severe forms of asthma and that pharmacological inhibitors of TRPV1 may eventually proved to be useful additions to the treatment of asthma.

Cancer

Colon cancer

Familial adenomatous polyposis is a syndrome that includes the propensity to develop colorectal cancer (and other cancers) due to the inheritance of defective mutations in either the APC (adenomatous polyposis coli) or MUTYH gene. These mutations lead to several abnormalities in the regulation of the growth of colon epithelial cells that ultimately lead to the development of intestinal polyps which have a high risk of turning cancerous. [65] One of the abnormalities found in the APC disease is progressive reductions in 15-lipoxygenase 1 along with its product, 13-HODE (presumed but not unambiguously shown to be the S stereoisomer) as the colon disease advances from polyp to malignant stages; 15-HETE, 5-lipoxygenase, 12-lipoxygenase, and 15-lipoxygenase-2, and selected metabolites of the latter lipoxygenases show no such association. [66] [67] [68] Similarly selective reductions in 15-lipoxygenase 1 and 13-HODE occur in non-hereditary colon cancer. [69] [70] [66] 13(S)-HODE inhibits the proliferation and causes the death (apoptosis) of cultured human colon cancer cells. [55] [69] [71] [70] Animal model studies also find that the 15-lipoxygenase 1 / 13-HODE axis inhibits the development of drug-induced colon cancer as well as the growth of human colon cancer cell explants. [67] These results suggest that 15-lipoxygenase 1 and its 13(S)-HODE product are factors in promoting genetically-associated and -non-associated colon cancers; they function by contributing to the suppression of the development and/or growth of this cancer and when reduced or absent allow its unrestrained, malignant growth.

Breast cancer

13(S)-HODE stimulates the proliferation of human MCF-7 estrogen receptor positive and MBA-MD-231 estrogen receptor negative human breast cancer cell lines (see List of breast cancer cell lines) in culture); [72] its production appears necessary for epidermal growth factor and tumor growth factor α to stimulate cultured BT-20 human breast cancer cells to proliferate [73] and for human breast cancer xenografts to grow in mice.; [74] and among a series of 10 polyunsaturated fatty acid metabolites quantified in human breast cancer tissue, only 13-HODE (stereoisomer not defined) was significantly elevated in rapidly growing, compared to slower growing, cancers. [72] The results of these studies suggest that 13(S)-HODE may act to promote the growth of breast cancer in humans.

Prostate cancer

15-LOX 1 is overexpressed in prostate cancerous compared to non-cancerous prostate tissue and the levels of its expression in cultured various human prostate cancer cell lines correlates positively with their rates of proliferation and increases the proliferation response of prostate cancer cells to epidermal growth factor and insulin-like growth factor 1); its levels in human prostate cancer tissues also correlates positively with the cancers' severity as judged by the cancers' Gleason score; and overexpressed 15-LOX 1 appears to not only increase prostate cancer cell proliferation, but also promotes its cell survival by stimulating production and of insulin-like growth factor 1 and possibly altering the Bcl-2 pathway of cellular apoptosis as well as increases prostate tumor vascularization and thereby metastasis by stimulating production of vascular endothelial growth factor. These 15-LOX 1 effects appear due to the enzyme's production of 13(S)-HODE. [75] [76] [77] The 15-LOX 1/13(S)-HODE axis also promotes the growth of prostate cancer in various animal models. [78] [79] In one animal model the pro-growth effects of 15-LOX 1 were altered by dietary targeting: increases in dietary linoleic acid, an omega-6 fatty acid, promoted while increases in dietary stearidonic acid, an omega-3 fatty acid reduced the growth of human prostate cancer explants. [80] These effects could be due to the ability of the linoleic acid diet to increase the production of the 15-Lox 1 metabolite, 13-HODE, [80] and the ability of the stearidonic acid to increase the production of docosahexaenoic acid and the 15-LOX-1 metabolites of docosahexaenoic acid, 17S-hydroperoxy-docosa-hexa-4Z,7Z,10Z,13 Z,15E,19Z-enoate(17-HpDHA, 17S-hydroxy-docosahexa-4Z,7Z,10Z,13Z,15E,19Z-enoate(17-HDHA), 10S,17S-dihydroxy-docosahexa-4Z,7Z,11E,13Z,15E,19Z-enoate(10,17-diHDHA, protectin DX), and 7S,17S-dihydroxy-docsahexa-4Z,8E,10Z,13Z,15E,19Z-enoate(7,17-diHDHA, protectin D5), all of which are inhibitors of cultured human prostate cancer cell proliferation. [81] [82] [83]

Markers for disease

13-HODE levels are elevated, compared to appropriate controls, in the low density lipoproteins isolated from individuals with rheumatoid arthritis, [84] in the high-density lipoprotein fraction of patients with diabetes, [85] in the serum of individuals with polycystic kidney disease. [86] or chronic pancreatitis, [87] and in the plasma of individuals with alcoholic and non-alcoholic steatohepatitis. [88] [89] The level of total HODEs, which includes various 13-HODE and 9-HODE isomers, are elevated in the plasma and erythrocytes of patients with Alzheimer's disease and in the plasma but not erythrocytes of patients with vascular dementia compared to normal volunteers. [90] See 9-hydroxyoctadecadienoic acid section on 9-HODEs as markers of disease involving oxidative stress for further details. These studies suggest that high levels of the HODEs may be useful to indicate the presence and progression of the cited diseases. Since, however, the absolute values of HODEs found in different studies vary greatly, since HODE levels vary with dietary linoleic acid intake, since HODEs may form during the processing of tissues, and since abnormal HODE levels are not linked to a specific disease, the use of these metabolites as markers has not attained clinical usefulness. [21] [91] [92] [19] HODE markers may find usefulness as markers of specific disease, type of disease, and/or progression of disease when combined with other disease markers. [19] [93]

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 Neo-Latin word 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">Oxidative stress</span> Free radical toxicity

Oxidative stress reflects an imbalance between the systemic manifestation of reactive oxygen species and a biological system's ability to readily detoxify the reactive intermediates or to repair the resulting damage. Disturbances in the normal redox state of cells can cause toxic effects through the production of peroxides and free radicals that damage all components of the cell, including proteins, lipids, and DNA. Oxidative stress from oxidative metabolism causes base damage, as well as strand breaks in DNA. Base damage is mostly indirect and caused by the reactive oxygen species generated, e.g., O2 (superoxide radical), OH (hydroxyl radical) and H2O2 (hydrogen peroxide). Further, some reactive oxidative species act as cellular messengers in redox signaling. Thus, oxidative stress can cause disruptions in normal mechanisms of cellular signaling.

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

Lipoxygenases (LOX) are a family of (non-heme) iron-containing 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.

<span class="mw-page-title-main">TRPV1</span> Human protein for regulating body temperature

The transient receptor potential cation channel subfamily V member 1 (TRPV1), also known as the capsaicin receptor and the vanilloid receptor 1, is a protein that, in humans, is encoded by the TRPV1 gene. It was the first isolated member of the transient receptor potential vanilloid receptor proteins that in turn are a sub-family of the transient receptor potential protein group. This protein is a member of the TRPV group of transient receptor potential family of ion channels. Fatty acid metabolites with affinity for this receptor are produced by cyanobacteria, which diverged from eukaryotes at least 2000 million years ago (MYA). The function of TRPV1 is detection and regulation of body temperature. In addition, TRPV1 provides a sensation of scalding heat and pain (nociception). In primary afferent sensory neurons, it cooperates with TRPA1 to mediate the detection of noxious environmental stimuli.

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">GPR132</span> Protein-coding gene in the species Homo sapiens

G protein coupled receptor 132, also termed G2A, is classified as a member of the proton sensing G protein coupled receptor (GPR) subfamily. Like other members of this subfamily, i.e. GPR4, GPR68 (OGR1), and GPR65 (TDAG8), G2A is a G protein coupled receptor that resides in the cell surface membrane, senses changes in extracellular pH, and can alter cellular function as a consequence of these changes. Subsequently, G2A was suggested to be a receptor for lysophosphatidylcholine (LPC). However, the roles of G2A as a pH-sensor or LPC receptor are disputed. Rather, current studies suggest that it is a receptor for certain metabolites of the polyunsaturated fatty acid, linoleic acid.

<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).

<span class="mw-page-title-main">ALOX12B</span> Protein-coding gene in the species Homo sapiens

Arachidonate 12-lipoxygenase, 12R type, also known as ALOX12B, 12R-LOX, and arachidonate lipoxygenase 3, is a lipoxygenase-type enzyme composed of 701 amino acids and encoded by the ALOX12B gene. The gene is located on chromosome 17 at position 13.1 where it forms a cluster with two other lipoxygenases, ALOXE3 and ALOX15B. Among the human lipoxygenases, ALOX12B is most closely related in amino acid sequence to ALOXE3

<span class="mw-page-title-main">ALOXE3</span> Protein-coding gene in the species Homo sapiens

Epidermis-type lipoxygenase 3 is a member of the lipoxygenase family of enzymes; in humans, it is encoded by the ALOXE3 gene. This gene is located on chromosome 17 at position 13.1 where it forms a cluster with two other lipoxygenases, ALOX12B and ALOX15B. Among the human lipoxygenases, ALOXE3 is most closely related in amino acid sequence to ALOX12B. ALOXE3, ALOX12B, and ALOX15B are often classified as epidermal lipoxygenases, in distinction to the other three human lipoxygenases, because they were initially defined as being highly or even exclusively expressed and functioning in skin. The epidermis-type lipoxygenases are now regarded as a distinct subclass within the multigene family of mammalian lipoxygenases with mouse Aloxe3 being the ortholog to human ALOXE3, mouse Alox12b being the ortholog to human ALOX12B, and mouse Alox8 being the ortholog to human ALOX15B [supplied by OMIM]. ALOX12B and ALOXE3 in humans, Alox12b and Aloxe3 in mice, and comparable orthologs in other in other species are proposed to act sequentially in a multistep metabolic pathway that forms products that are structurally critical for creating and maintaining the skin's water barrier function.

<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.

<span class="mw-page-title-main">9-Hydroxyoctadecadienoic acid</span> Chemical compound

9-Hydroxyoctadecadienoic acid (or 9-HODE) has been used in the literature to designate either or both of two stereoisomer metabolites of the essential fatty acid, linoleic acid: 9(S)-hydroxy-10(E),12(Z)-octadecadienoic acid (9(S)-HODE) and 9(R)-hydroxy-10(E),12(Z)-octadecadienoic acid (9(R)-HODE); these two metabolites differ in having their hydroxy residues in the S or R configurations, respectively. The accompanying figure gives the structure for 9(S)-HETE. Two other 9-hydroxy linoleic acid derivatives occur in nature, the 10E,12E isomers of 9(S)-HODE and 9(R)-HODE viz., 9(S)-hydroxy-10E,12E-octadecadienoic acid (9(S)-EE-HODE) and 9(R)-hydroxy-10E,12E-octadecadienoic acid (13(R)-EE-HODE); these two derivatives have their double bond at carbon 12 in the E or trans configuration as opposed to the Z or cis configuration. The four 9-HODE isomers, particularly under conditions of oxidative stress, may form together in cells and tissues; they have overlapping but not identical biological activities and significances. Because many studies have not distinguished between the S and R stereoisomers and, particularly in identifying tissue levels, the two EE isomers, 9-HODE is used here when the isomer studied is unclear.

<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.

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.

Specialized pro-resolving mediators are a large and growing class of cell signaling molecules formed in cells by the metabolism of polyunsaturated fatty acids (PUFA) by one or a combination of lipoxygenase, cyclooxygenase, and cytochrome P450 monooxygenase enzymes. Pre-clinical studies, primarily in animal models and human tissues, implicate SPM in orchestrating the resolution of inflammation. Prominent members include the resolvins and protectins.

References

  1. 1 2 Soberman, R. J; Harper, T. W; Betteridge, D; Lewis, R. A; Austen, K. F (1985). "Characterization and separation of the arachidonic acid 5-lipoxygenase and linoleic acid omega-6 lipoxygenase (arachidonic acid 15-lipoxygenase) of human polymorphonuclear leukocytes". The Journal of Biological Chemistry. 260 (7): 4508–15. doi: 10.1016/S0021-9258(18)89293-6 . PMID   3920219.
  2. 1 2 Wecksler, Aaron T; Kenyon, Victor; Deschamps, Joshua D; Holman, Theodore R (2008). "Substrate Specificity Changes for Human Reticulocyte and Epithelial 15-Lipoxygenases Reveal Allosteric Product Regulation". Biochemistry. 47 (28): 7364–75. doi:10.1021/bi800550n. PMC   2603187 . PMID   18570379.
  3. Reinaud, O; Delaforge, M; Boucher, J.L; Rocchiccioli, F; Mansuy, D (1989). "Oxidative metabolism of linoleic acid by human leukocytes". Biochemical and Biophysical Research Communications. 161 (2): 883–91. doi:10.1016/0006-291X(89)92682-X. PMID   2735926.
  4. Kühn, Hartmut; Barnett, Jim; Grunberger, Dorit; Baecker, Preston; Chow, Joan; Nguyen, Binh; Bursztyn-Pettegrew, Hela; Chan, Hardy; Sigal, Elliott (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.
  5. Kuhn, Hartmut; Walther, Matthias; Kuban, Ralf Jürgen (2002). "Mammalian arachidonate 15-lipoxygenases". Prostaglandins & Other Lipid Mediators. 68–69: 263–290. doi:10.1016/S0090-6980(02)00035-7. PMID   12432923.
  6. Burri, L.; Hoem, N.; Banni, S.; Berge, K. (2012). "Marine Omega-3 Phospholipids: Metabolism and Biological Activities". International Journal of Molecular Sciences. 13 (11): 15401–15419. doi: 10.3390/ijms131115401 . PMC   3509649 . PMID   23203133.
  7. Cholesterol at the U.S. National Library of Medicine Medical Subject Headings (MeSH)
  8. 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. 94 (12): 6148–52. Bibcode:1997PNAS...94.6148B. doi: 10.1073/pnas.94.12.6148 . PMC   21017 . PMID   9177185.
  9. Wecksler, Aaron T; Kenyon, Victor; Garcia, Natalie K; Deschamps, Joshua D; Van Der Donk, Wilfred A; Holman, Theodore R (2009). "Kinetic and Structural Investigations of the Allosteric Site in Human Epithelial 15-Lipoxygenase-2". Biochemistry. 48 (36): 8721–30. doi:10.1021/bi9009242. PMC   2746553 . PMID   19645454.
  10. Laneuville, O; Breuer, D. K; Xu, N; Huang, Z. H; Gage, D. A; Watson, J. T; Lagarde, M; Dewitt, D. L; Smith, W. L (1995). "Fatty acid substrate specificities of human prostaglandin-endoperoxide H synthase-1 and -2. Formation of 12-hydroxy-(9Z, 13E/Z, 15Z)- octadecatrienoic acids from alpha-linolenic acid". The Journal of Biological Chemistry. 270 (33): 19330–6. doi: 10.1074/jbc.270.33.19330 . PMID   7642610.
  11. 1 2 Godessart, Nuria; Camacho, Mercedes; López-Belmonte, Jesús; Antón, Rosa; García, Montserrat; De Moragas, Josep-María; Vila, Luis (1996). "Prostaglandin H-Synthase-2 is the Main Enzyme Involved in the Biosynthesis of Octadecanoids from Linoleic Acid in Human Dermal Fibroblasts Stimulated with Interleukin-1β". Journal of Investigative Dermatology. 107 (5): 726–32. doi: 10.1111/1523-1747.ep12365616 . PMID   8875957.
  12. Hamberg, Mats; Samuelsson, Bengt (1980). "Stereochemistry in the formation of 9-hydroxy-10,12-octadecadienoic acid and 13-hydroxy-9,11-octadecadienoic acid from linoleic acid by fatty acid cyclooxygenase". Biochimica et Biophysica Acta (BBA) - Lipids and Lipid Metabolism. 617 (3): 545–7. doi:10.1016/0005-2760(80)90022-3. PMID   6768399.
  13. Lindstrom, Terry D; Aust, Steven D (1984). "Studies on cytochrome P-450-dependent lipid hydroperoxide reduction". Archives of Biochemistry and Biophysics. 233 (1): 80–7. doi:10.1016/0003-9861(84)90603-9. PMID   6431911.
  14. Oliw, Ernst 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): 258–63. doi:10.1016/0005-2760(93)90106-J. PMID   8443245.
  15. Ruparel, Shivani; Green, Dustin; Chen, Paul; Hargreaves, Kenneth M (2012). "The Cytochrome P450 Inhibitor, Ketoconazole, Inhibits Oxidized Linoleic Acid Metabolite-Mediated Peripheral Inflammatory Pain". Molecular Pain. 8: 1744–8069–8–73. doi: 10.1186/1744-8069-8-73 . PMC   3488501 . PMID   23006841.
  16. Frankel, E.N (1984). "Chemistry of free radical and singlet oxidation of lipids". Progress in Lipid Research. 23 (4): 197–221. doi:10.1016/0163-7827(84)90011-0. PMID   6100997.
  17. Spiteller, Peter; Spiteller, Gerhard (1998). "Strong dependence of the lipid peroxidation product spectrum whether Fe2+/O2 or Fe3+/O2 is used as oxidant". Biochimica et Biophysica Acta (BBA) - Lipids and Lipid Metabolism. 1392 (1): 23–40. doi:10.1016/S0005-2760(97)00209-9. PMID   9593808.
  18. Punta, Carlo; Rector, Christopher L; Porter, Ned A (2005). "Peroxidation of Polyunsaturated Fatty Acid Methyl Esters Catalyzed byN-Methyl Benzohydroxamic Acid: A New and Convenient Method for Selective Synthesis of Hydroperoxides and Alcohols". Chemical Research in Toxicology. 18 (2): 349–56. doi:10.1021/tx049685x. PMID   15720142.
  19. 1 2 3 4 Yoshida, Yasukazu; Umeno, Aya; Akazawa, Yoko; Shichiri, Mototada; Murotomi, Kazutoshi; Horie, Masanori (2015). "Chemistry of Lipid Peroxidation Products and Their Use as Biomarkers in Early Detection of Diseases". Journal of Oleo Science. 64 (4): 347–56. doi: 10.5650/jos.ess14281 . PMID   25766928.
  20. Riahi, Yael; Cohen, Guy; Shamni, Ofer; Sasson, Shlomo (2010). "Signaling and cytotoxic functions of 4-hydroxyalkenals". American Journal of Physiology. Endocrinology and Metabolism. 299 (6): E879–86. doi:10.1152/ajpendo.00508.2010. PMID   20858748. S2CID   6062445.
  21. 1 2 Ramsden, Christopher E; Ringel, Amit; Feldstein, Ariel E; Taha, Ameer Y; MacIntosh, Beth A; Hibbeln, Joseph R; Majchrzak-Hong, Sharon F; Faurot, Keturah R; Rapoport, Stanley I; Cheon, Yewon; Chung, Yoon-Mi; Berk, Michael; Douglas Mann, J (2012). "Lowering dietary linoleic acid reduces bioactive oxidized linoleic acid metabolites in humans". Prostaglandins, Leukotrienes and Essential Fatty Acids. 87 (4–5): 135–41. doi:10.1016/j.plefa.2012.08.004. PMC   3467319 . PMID   22959954.
  22. Cho, Y; Ziboh, V. A (1994). "Incorporation of 13-hydroxyoctadecadienoic acid (13-HODE) into epidermal ceramides and phospholipids: Phospholipase C-catalyzed release of novel 13-HODE-containing diacylglycerol". Journal of Lipid Research. 35 (2): 255–62. doi: 10.1016/S0022-2275(20)41214-3 . PMID   8169529.
  23. Grøn, B; Iversen, L; Ziboh, V; Kragballe, K (1993). "Monohydroxy fatty acids esterified to phospholipids are decreased in lesional psoriatic skin". Archives of Dermatological Research. 285 (8): 449–54. doi:10.1007/BF00376816. PMID   8274032. S2CID   25517644.
  24. Fang, X; Kaduce, T. L; Spector, A. A (1999). "13-(S)-hydroxyoctadecadienoic acid (13-HODE) incorporation and conversion to novel products by endothelial cells". Journal of Lipid Research. 40 (4): 699–707. doi: 10.1016/S0022-2275(20)32149-0 . PMID   10191294.
  25. Bull, A. W; Earles, S. M; Bronstein, J. C (1991). "Metabolism of oxidized linoleic acid: Distribution of activity for the enzymatic oxidation of 13-hydroxyoctadecadienoic acid to 13-oxooctadecadienoic acid in rat tissues". Prostaglandins. 41 (1): 43–50. doi:10.1016/0090-6980(91)90103-M. PMID   2020745.
  26. Bull, A. W; Branting, C; Bronstein, J. C; Blackburn, M. L; Rafter, J (1993). "Increases in 13-hydroxyoctadecadienoic acid dehydrogenase activity during differentiation of cultured cells". Carcinogenesis. 14 (11): 2239–43. doi:10.1093/carcin/14.11.2239. PMID   8242849.
  27. Earles, Sonja M; Bronstein, Joel C; Winner, David L; Bull, Arthur W (1991). "Metabolism of oxidized linoleic acid: Characterization of 13-hydroxyoctadecadienoic acid dehydrogenase activity from rat colonic tissue". Biochimica et Biophysica Acta (BBA) - Lipids and Lipid Metabolism. 1081 (2): 174–80. doi:10.1016/0005-2760(91)90023-B. PMID   1998735.
  28. Kühn, H; Belkner, J; Wiesner, R; Alder, L (1990). "Occurrence of 9- and 13-keto-octadecadienoic acid in biological membranes oxygenated by the reticulocyte lipoxygenase". Archives of Biochemistry and Biophysics. 279 (2): 218–24. doi:10.1016/0003-9861(90)90484-G. PMID   2112367.
  29. Waddington, Emma; Sienuarine, Kishore; Puddey, Ian; Croft, Kevin (2001). "Identification and Quantitation of Unique Fatty Acid Oxidation Products in Human Atherosclerotic Plaque Using High-Performance Liquid Chromatography". Analytical Biochemistry. 292 (2): 234–44. doi:10.1006/abio.2001.5075. PMID   11355856.
  30. 1 2 Patwardhan, A. M; Scotland, P. E; Akopian, A. N; Hargreaves, K. M (2009). "Activation of TRPV1 in the spinal cord by oxidized linoleic acid metabolites contributes to inflammatory hyperalgesia". Proceedings of the National Academy of Sciences. 106 (44): 18820–4. doi: 10.1073/pnas.0905415106 . PMC   2764734 . PMID   19843694.
  31. Altmann, Reinhold; Hausmann, Martin; Spöttl, Tanja; Gruber, Michael; Bull, Arthur W; Menzel, Katrin; Vogl, Daniela; Herfarth, Hans; Schölmerich, Jürgen; Falk, Werner; Rogler, Gerhard (2007). "13-Oxo-ODE is an endogenous ligand for PPARγ in human colonic epithelial cells". Biochemical Pharmacology. 74 (4): 612–22. doi:10.1016/j.bcp.2007.05.027. PMID   17604003.
  32. Gouveia-Figueira, Sandra; Späth, Jana; Zivkovic, Angela M; Nording, Malin L (2015). "Profiling the Oxylipin and Endocannabinoid Metabolome by UPLC-ESI-MS/MS in Human Plasma to Monitor Postprandial Inflammation". PLOS ONE. 10 (7): e0132042. Bibcode:2015PLoSO..1032042G. doi: 10.1371/journal.pone.0132042 . PMC   4506044 . PMID   26186333.
  33. 1 2 Zein, Claudia O; Lopez, Rocio; Fu, Xiaoming; Kirwan, John P; Yerian, Lisa M; McCullough, Arthur J; Hazen, Stanley L; Feldstein, Ariel E (2012). "Pentoxifylline decreases oxidized lipid products in nonalcoholic steatohepatitis: New evidence on the potential therapeutic mechanism". Hepatology. 56 (4): 1291–9. doi:10.1002/hep.25778. PMC   3430813 . PMID   22505276.
  34. 1 2 Murphy, Robert C; Zarini, Simona (2002). "Glutathione adducts of oxyeicosanoids". Prostaglandins & Other Lipid Mediators. 68–69: 471–82. doi:10.1016/S0090-6980(02)00049-7. PMID   12432937.
  35. Bull, Arthur W; Bronstein, Joel C; Earles, Sonja M; Blackburn, Mary L (1996). "Formation of adducts between 13-oxooctadecadienoic acid (13-OXO) and protein- derived thiols, in vivo and in vitro". Life Sciences. 58 (25): 2355–65. doi:10.1016/0024-3205(96)00236-6. PMID   8649225.
  36. Blackburn, Mary L; Podgorski, Izabela; Bull, Arthur W (1999). "Specific protein targets of 13-oxooctadecadienoic acid (13-OXO) and export of the 13-OXO–glutathione conjugate in HT-29 cells". Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids. 1440 (2–3): 225–34. doi:10.1016/S1388-1981(99)00123-7. PMID   10521706.
  37. 1 2 Nagy, Laszlo; Tontonoz, Peter; Alvarez, Jacqueline G.A; Chen, Hongwu; Evans, Ronald M (1998). "Oxidized LDL Regulates Macrophage Gene Expression through Ligand Activation of PPARγ". Cell. 93 (2): 229–40. doi: 10.1016/S0092-8674(00)81574-3 . PMID   9568715. S2CID   7573475.
  38. Itoh, T; Fairall, L; Amin, K; Inaba, Y; Szanto, A; Balint, B. L; Nagy, L; Yamamoto, K; Schwabe, J. W (2008). "Structural basis for the activation of PPARgamma by oxidized fatty acids". Nature Structural & Molecular Biology. 15 (9): 924–31. doi:10.1038/nsmb.1474. PMC   2939985 . PMID   19172745.
  39. Yokoi, Hiroshi; Mizukami, Hajime; Nagatsu, Akito; Ohno, Takamasa; Tanabe, Hiroki; Inoue, Makoto (2009). "Peroxisome Proliferator-Activated Receptor γ Ligands Isolated from Adlay Seed (Coix lacryma-jobi L. Var. Ma-yuen STAPF.)". Biological & Pharmaceutical Bulletin. 32 (4): 735–40. doi: 10.1248/bpb.32.735 . PMID   19336916.
  40. Delerive, Philippe; Furman, Christophe; Teissier, Elisabeth; Fruchart, Jean-Charles; Duriez, Patrick; Staels, Bart (2000). "Oxidized phospholipids activate PPARα in a phospholipase A2-dependent manner". FEBS Letters. 471 (1): 34–8. doi:10.1016/S0014-5793(00)01364-8. PMID   10760508. S2CID   85113031.
  41. De Petrocellis, Luciano; Schiano Moriello, Aniello; Imperatore, Roberta; Cristino, Luigia; Starowicz, Katarzyna; Di Marzo, Vincenzo (2012). "A re-evaluation of 9-HODE activity at TRPV1 channels in comparison with anandamide: Enantioselectivity and effects at other TRP channels and in sensory neurons". British Journal of Pharmacology. 167 (8): 1643–51. doi:10.1111/j.1476-5381.2012.02122.x. PMC   3525867 . PMID   22861649.
  42. Obinata, Hideru; Izumi, Takashi (2009). "G2A as a receptor for oxidized free fatty acids". Prostaglandins & Other Lipid Mediators. 89 (3–4): 66–72. doi:10.1016/j.prostaglandins.2008.11.002. PMID   19063986.
  43. Yin, Hong; Chu, Alan; Li, Wei; Wang, Bin; Shelton, Fabiola; Otero, Francella; Nguyen, Deborah G; Caldwell, Jeremy S; Chen, Yu Alice (2009). "Lipid G Protein-coupled Receptor Ligand Identification Using β-Arrestin Path Hunter Assay". Journal of Biological Chemistry. 284 (18): 12328–38. doi: 10.1074/jbc.M806516200 . PMC   2673301 . PMID   19286662.
  44. 1 2 Kühn, H; Brash, A. R (1990). "Occurrence of lipoxygenase products in membranes of rabbit reticulocytes. Evidence for a role of the reticulocyte lipoxygenase in the maturation of red cells". The Journal of Biological Chemistry. 265 (3): 1454–8. doi: 10.1016/S0021-9258(19)40037-9 . PMID   2104842.
  45. Van Leyen, Klaus; Duvoisin, Robert M; Engelhardt, Harald; Wiedmann, Martin (1998). "A function for lipoxygenase in programmed organelle degradation". Nature. 395 (6700): 392–5. Bibcode:1998Natur.395..392V. doi:10.1038/26500. PMID   9759730. S2CID   4366265.
  46. Sun, D; Funk, C. D (1996). "Disruption of 12/15-lipoxygenase expression in peritoneal macrophages. Enhanced utilization of the 5-lipoxygenase pathway and diminished oxidation of low density lipoprotein". The Journal of Biological Chemistry. 271 (39): 24055–62. doi: 10.1074/jbc.271.39.24055 . PMID   8798642.
  47. Ivanov, Igor; Kuhn, Hartmut; Heydeck, Dagmar (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.
  48. 1 2 3 4 5 6 Mabalirajan, Ulaganathan; Rehman, Rakhshinda; Ahmad, Tanveer; Kumar, Sarvesh; Singh, Suchita; Leishangthem, Geeta D; Aich, Jyotirmoi; Kumar, Manish; Khanna, Kritika; Singh, Vijay P; Dinda, Amit K; Biswal, Shyam; Agrawal, Anurag; Ghosh, Balaram (2013). "Linoleic acid metabolite drives severe asthma by causing airway epithelial injury". Scientific Reports. 3: 1349. Bibcode:2013NatSR...3E1349M. doi:10.1038/srep01349. PMC   3583002 . PMID   23443229.
  49. Henricks, P. A; Engels, F; Van Der Vliet, H; Nijkamp, F. P (1991). "9- and 13-hydroxy-linoleic acid possess chemotactic activity for bovine and human polymorphonuclear leukocytes". Prostaglandins. 41 (1): 21–7. doi:10.1016/0090-6980(91)90101-K. PMID   2020743.
  50. Rolin, Johannes; Al-Jaderi, Zaidoon; Maghazachi, Azzam A (2013). "Oxidized lipids and lysophosphatidylcholine induce the chemotaxis and intracellular calcium influx in natural killer cells". Immunobiology. 218 (6): 875–83. doi:10.1016/j.imbio.2012.10.009. PMID   23200035. S2CID   37940201.
  51. Kühn, H; Belkner, J; Zaiss, S; Fährenklemper, T; Wohlfeil, S (1994). "Involvement of 15-lipoxygenase in early stages of atherogenesis". The Journal of Experimental Medicine. 179 (6): 1903–11. doi:10.1084/jem.179.6.1903. PMC   2191515 . PMID   8195716.
  52. Folcik, V A; Nivar-Aristy, R A; Krajewski, L P; Cathcart, M K (1995). "Lipoxygenase contributes to the oxidation of lipids in human atherosclerotic plaques". Journal of Clinical Investigation. 96 (1): 504–10. doi:10.1172/JCI118062. PMC   185224 . PMID   7615823.
  53. Waddington, Emma I; Croft, Kevin D; Sienuarine, Kishore; Latham, Bruce; Puddey, Ian B (2003). "Fatty acid oxidation products in human atherosclerotic plaque: An analysis of clinical and histopathological correlates". Atherosclerosis. 167 (1): 111–20. doi:10.1016/S0021-9150(02)00391-X. PMID   12618275.
  54. Kühn, H; Heydeck, D; Hugou, I; Gniwotta, C (1997). "In vivo action of 15-lipoxygenase in early stages of human atherogenesis". Journal of Clinical Investigation. 99 (5): 888–93. doi:10.1172/JCI119253. PMC   507896 . PMID   9062346.
  55. 1 2 Cabral, Marisol; Martín-Venegas, Raquel; Moreno, Juan José (2014). "Differential cell growth/apoptosis behavior of 13-hydroxyoctadecadienoic acid enantiomers in a colorectal cancer cell line". American Journal of Physiology. Gastrointestinal and Liver Physiology. 307 (6): G664–71. doi:10.1152/ajpgi.00064.2014. PMID   25035111.
  56. 1 2 3 Lee Kennedy, R; Vangaveti, Venkat; Jarrod, Ghassan; Shashidhar, Venkatesh; Shashidhar, Venkatesh; Baune, Bernhard T (2010). "Review: Free fatty acid receptors: Emerging targets for treatment of diabetes and its complications". Therapeutic Advances in Endocrinology and Metabolism. 1 (4): 165–75. doi:10.1177/2042018810381066. PMC   3474614 . PMID   23148161.
  57. 1 2 Vangaveti, Venkat N; Shashidhar, Venkatesh M; Rush, Catherine; Malabu, Usman H; Rasalam, Roy R; Collier, Fiona; Baune, Bernhard T; Kennedy, Richard L (2014). "Hydroxyoctadecadienoic Acids Regulate Apoptosis in Human THP-1 Cells in a PPARγ-Dependent Manner". Lipids. 49 (12): 1181–92. doi:10.1007/s11745-014-3954-z. PMID   25330944. S2CID   4062623.
  58. Febbraio, Maria; Podrez, Eugene A; Smith, Jonathan D; Hajjar, David P; Hazen, Stanley L; Hoff, Henry F; Sharma, Kavita; Silverstein, Roy L (2000). "Targeted disruption of the class B scavenger receptor CD36 protects against atherosclerotic lesion development in mice". Journal of Clinical Investigation. 105 (8): 1049–56. doi:10.1172/JCI9259. PMC   300837 . PMID   10772649.
  59. Makowski, Liza; Boord, Jeffrey B; Maeda, Kazuhisa; Babaev, Vladimir R; Uysal, K. Teoman; Morgan, Maureen A; Parker, Rex A; Suttles, Jill; Fazio, Sergio; Hotamisligil, Gökhan S; Linton, Macrae F (2001). "Lack of macrophage fatty-acid–binding protein aP2 protects mice deficient in apolipoprotein E against atherosclerosis". Nature Medicine. 7 (6): 699–705. doi:10.1038/89076. PMC   4027052 . PMID   11385507.
  60. Fukuda, Kazuki; Matsumura, Takeshi; Senokuchi, Takafumi; Ishii, Norio; Kinoshita, Hiroyuki; Yamada, Sarie; Murakami, Saiko; Nakao, Saya; Motoshima, Hiroyuki; Kondo, Tatsuya; Kukidome, Daisuke; Kawasaki, Shuji; Kawada, Teruo; Nishikawa, Takeshi; Araki, Eiichi (2015). "Statins meditate anti-atherosclerotic action in smooth muscle cells by peroxisome proliferator-activated receptor-γ activation". Biochemical and Biophysical Research Communications. 457 (1): 23–30. doi:10.1016/j.bbrc.2014.12.063. PMID   25529449.
  61. Henricks, P; Engels, F; Vanderlinde, H; Garssen, J; Nijkamp, F (1995). "13-Hydroxy-linoleic acid induces airway hyperresponsiveness to histamine and methacholine in guinea pigs in vivo". Journal of Allergy and Clinical Immunology. 96 (1): 36–43. doi:10.1016/S0091-6749(95)70030-7. PMID   7622761.
  62. 1 2 3 Mabalirajan, U; Dinda, A. K; Kumar, S; Roshan, R; Gupta, P; Sharma, S. K; Ghosh, B (2008). "Mitochondrial structural changes and dysfunction are associated with experimental allergic asthma". Journal of Immunology. 181 (5): 3540–8. doi: 10.4049/jimmunol.181.5.3540 . PMID   18714027.
  63. Thomas, Biju; Rutman, Andrew; Hirst, Robert A; Haldar, Pranab; Wardlaw, Andrew J; Bankart, John; Brightling, Christopher E; O'Callaghan, Christopher (2010). "Ciliary dysfunction and ultrastructural abnormalities are features of severe asthma". Journal of Allergy and Clinical Immunology. 126 (4): 722–729.e2. doi:10.1016/j.jaci.2010.05.046. PMID   20673980.
  64. Engels, F; Kessels, G. C; Henricks, P. A; Nijkamp, F. P (1996). "Preferential formation of 13-hydroxylinoleic acid by human peripheral blood eosinophils". Prostaglandins. 52 (2): 117–24. doi:10.1016/0090-6980(96)00057-3. PMID   8880897.
  65. Lung, M. S; Trainer, A. H; Campbell, I; Lipton, L (2015). "Familial colorectal cancer". Internal Medicine Journal. 45 (5): 482–91. doi: 10.1111/imj.12736 . PMID   25955461.
  66. 1 2 Shureiqi, I; Chen, D; Day, R. S; Zuo, X; Hochman, F. L; Ross, W. A; Cole, R. A; Moy, O; Morris, J. S; Xiao, L; Newman, R. A; Yang, P; Lippman, S. M (2010). "Profiling Lipoxygenase Metabolism in Specific Steps of Colorectal Tumorigenesis". Cancer Prevention Research. 3 (7): 829–38. doi:10.1158/1940-6207.CAPR-09-0110. PMC   2900425 . PMID   20570882.
  67. 1 2 Zuo, Xiangsheng; Shureiqi, Imad (2013). "Eicosanoid profiling in colon cancer: Emergence of a pattern". Prostaglandins & Other Lipid Mediators. 104–105: 139–43. doi:10.1016/j.prostaglandins.2012.08.004. PMC   3532570 . PMID   22960430.
  68. Kuhn, Hartmut; Banthiya, Swathi; Van Leyen, Klaus (2015). "Mammalian lipoxygenases and their biological relevance". Biochimica et Biophysica Acta (BBA) - Molecular and Cell Biology of Lipids. 1851 (4): 308–30. doi:10.1016/j.bbalip.2014.10.002. PMC   4370320 . PMID   25316652.
  69. 1 2 Shureiqi, I; Wojno, K. J; Poore, J. A; Reddy, R. G; Moussalli, M. J; Spindler, S. A; Greenson, J. K; Normolle, D; Hasan, A. A; Lawrence, T. S; Brenner, D. E (1999). "Decreased 13-S-hydroxyoctadecadienoic acid levels and 15-lipoxygenase-1 expression in human colon cancers". Carcinogenesis. 20 (10): 1985–95. doi: 10.1093/carcin/20.10.1985 . PMID   10506115.
  70. 1 2 Nixon, Jennifer B; Kim, Kyung-Su; Lamb, Patricia W; Bottone, Frank G; Eling, Thomas E (2004). "15-Lipoxygenase-1 has anti-tumorigenic effects in colorectal cancer". Prostaglandins, Leukotrienes and Essential Fatty Acids. 70 (1): 7–15. doi:10.1016/j.plefa.2003.06.001. PMID   14643174.
  71. Shureiqi, I; Jiang, W; Zuo, X; Wu, Y; Stimmel, J. B; Leesnitzer, L. M; Morris, J. S; Fan, H.-Z; Fischer, S. M; Lippman, S. M (2003). "The 15-lipoxygenase-1 product 13-S-hydroxyoctadecadienoic acid down-regulates PPAR-δ to induce apoptosis in colorectal cancer cells". Proceedings of the National Academy of Sciences. 100 (17): 9968–73. Bibcode:2003PNAS..100.9968S. doi: 10.1073/pnas.1631086100 . PMC   187904 . PMID   12909723.
  72. 1 2 O'Flaherty, Joseph T; Wooten, Rhonda E; Samuel, Michael P; Thomas, Michael J; Levine, Edward A; Case, L. Douglas; Akman, Steven A; Edwards, Iris 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.
  73. Reddy, Nagi; Everhart, Angela; Eling, Thomas; Glasgow, Wayne (1997). "Characterization of a 15-Lipoxygenase in Human Breast Carcinoma BT-20 Cells: Stimulation of 13-HODE Formation by TGFα/EGF". Biochemical and Biophysical Research Communications. 231 (1): 111–6. doi:10.1006/bbrc.1997.6048. PMID   9070230.
  74. Hill, Steven M; Blask, David E; Xiang, Shulin; Yuan, Lin; Mao, Lulu; Dauchy, Robert T; Dauchy, Erin M; Frasch, Tripp; Duplesis, Tamika (2011). "Melatonin and Associated Signaling Pathways that Control Normal Breast Epithelium and Breast Cancer". Journal of Mammary Gland Biology and Neoplasia. 16 (3): 235–45. doi:10.1007/s10911-011-9222-4. PMID   21773809. S2CID   22711432.
  75. Kelavkar, U; Glasgow, W; Eling, T. E (2002). "The effect of 15-lipoxygenase-1 expression on cancer cells". Current Urology Reports. 3 (3): 207–14. doi:10.1007/s11934-002-0066-8. PMID   12084190. S2CID   21497252.
  76. Hsi, Linda C; Wilson, Leigh C; Eling, Thomas E (2002). "Opposing Effects of 15-Lipoxygenase-1 and -2 Metabolites on MAPK Signaling in Prostate". Journal of Biological Chemistry. 277 (43): 40549–56. doi: 10.1074/jbc.M203522200 . PMID   12189136.
  77. Kelavkar, U. P; Cohen, C (2004). "15-lipoxygenase-1 expression upregulates and activates insulin-like growth factor-1 receptor in prostate cancer cells". Neoplasia. 6 (1): 41–52. doi:10.1016/S1476-5586(04)80052-6. PMC   1508629 . PMID   15068670.
  78. Kelavkar, Uddhav P; Parwani, Anil V; Shappell, Scott B; Martin, W. David (2006). "Conditional Expression of Human 15-Lipoxygenase-1 in Mouse Prostate Induces Prostatic Intraepithelial Neoplasia: The FLiMP Mouse Model". Neoplasia. 8 (6): 510–22. doi:10.1593/neo.06202. PMC   1601466 . PMID   16820097.
  79. Sen, Malabika; McHugh, Kevin; Hutzley, Justin; Philips, Brian J; Dhir, Rajiv; Parwani, Anil V; Kelavkar, Uddhav P (2006). "Orthotopic expression of human 15-lipoxygenase (LO)-1 in the dorsolateral prostate of normal wild-type C57BL/6 mouse causes PIN-like lesions". Prostaglandins & Other Lipid Mediators. 81 (1–2): 1–13. doi:10.1016/j.prostaglandins.2006.05.024. PMID   16997127.
  80. 1 2 Kelavkar, U. P; Hutzley, J; McHugh, K; Allen, K. G; Parwani, A (2009). "Prostate tumor growth can be modulated by dietarily targeting the 15-lipoxygenase-1 and cyclooxygenase-2 enzymes". Neoplasia. 11 (7): 692–9. doi:10.1593/neo.09334. PMC   2697355 . PMID   19568414.
  81. Edwards, I. J; Berquin, I. M; Sun, H; O'Flaherty, J. T; Daniel, L. W; Thomas, M. J; Rudel, L. L; Wykle, R. L; Chen, Y. Q (2004). "Differential Effects of Delivery of Omega-3 Fatty Acids to Human Cancer Cells by Low-Density Lipoproteins versus Albumin". Clinical Cancer Research. 10 (24): 8275–83. doi: 10.1158/1078-0432.CCR-04-1357 . PMID   15623603.
  82. o'Flaherty, Joseph T; Hu, Yungping; Wooten, Rhonda E; Horita, David A; Samuel, Michael P; Thomas, Michael J; Sun, Haiguo; Edwards, Iris J (2012). "15-Lipoxygenase Metabolites of Docosahexaenoic Acid Inhibit Prostate Cancer Cell Proliferation and Survival". PLOS ONE. 7 (9): e45480. Bibcode:2012PLoSO...745480O. doi: 10.1371/journal.pone.0045480 . PMC   3447860 . PMID   23029040.
  83. Hu, Yunping; Sun, Haiguo; o'Flaherty, Joseph T; Edwards, Iris J (2013). "15-Lipoxygenase-1-mediated metabolism of docosahexaenoic acid is required for syndecan-1 signaling and apoptosis in prostate cancer cells". Carcinogenesis. 34 (1): 176–82. doi:10.1093/carcin/bgs324. PMC   3584949 . PMID   23066085.
  84. Jira, Wolfgang; Spiteller, Gerhard; Richter, Andreas (1997). "Increased levels of lipid oxidation products in low density lipoproteins of patients suffering from rheumatoid arthritis". Chemistry and Physics of Lipids. 87 (1): 81–9. doi:10.1016/S0009-3084(97)00030-3. PMID   9219348.
  85. Lê, Quang Huy; El Alaoui, Meddy; Véricel, Evelyne; Ségrestin, Bérénice; Soulère, Laurent; Guichardant, Michel; Lagarde, Michel; Moulin, Philippe; Calzada, Catherine (2015). "Glycoxidized HDL, HDL Enriched with Oxidized Phospholipids and HDL from Diabetic Patients Inhibit Platelet Function". The Journal of Clinical Endocrinology & Metabolism. 100 (5): 2006–14. doi:10.1210/jc.2014-4214. PMC   4803888 . PMID   25794249.
  86. Klawitter, Jelena; Klawitter, Jost; McFann, Kim; Pennington, Alexander T; Abebe, Kaleab Z; Brosnahan, Godela; Cadnapaphornchai, Melissa A; Chonchol, Michel; Gitomer, Berenice; Christians, Uwe; Schrier, Robert W (2014). "Bioactive lipid mediators in polycystic kidney disease". Journal of Lipid Research. 55 (6): 1139–49. doi:10.1194/jlr.P042176. PMC   4031945 . PMID   24343898.
  87. Stevens, Tyler; Berk, Michael P; Lopez, Rocio; Chung, Yoon-Mi; Zhang, Renliang; Parsi, Mansour A; Bronner, Mary P; Feldstein, Ariel E (2012). "Lipidomic Profiling of Serum and Pancreatic Fluid in Chronic Pancreatitis". Pancreas. 41 (4): 518–22. doi:10.1097/MPA.0b013e31823ca306. PMID   22504378. S2CID   42972163.
  88. Yang, Lili; Latchoumycandane, Calivarathan; McMullen, Megan R; Pratt, Brian T; Zhang, Renliang; Papouchado, Bettina G; Nagy, Laura E; Feldstein, Ariel E; McIntyre, Thomas M (2010). "Chronic Alcohol Exposure Increases Circulating Bioactive Oxidized Phospholipids". Journal of Biological Chemistry. 285 (29): 22211–20. doi: 10.1074/jbc.M110.119982 . PMC   2903350 . PMID   20460374.
  89. Feldstein, Ariel E; Lopez, Rocio; Tamimi, Tarek Abu-Rajab; Yerian, Lisa; Chung, Yoon-Mi; Berk, Michael; Zhang, Renliang; McIntyre, Thomas M; Hazen, Stanley L (2010). "Mass spectrometric profiling of oxidized lipid products in human nonalcoholic fatty liver disease and nonalcoholic steatohepatitis". Journal of Lipid Research. 51 (10): 3046–54. doi:10.1194/jlr.M007096. PMC   2936759 . PMID   20631297.
  90. Yoshida, Yasukazu; Yoshikawa, Atsushi; Kinumi, Tomoya; Ogawa, Yoko; Saito, Yoshiro; Ohara, Kazuyuki; Yamamoto, Hirokazu; Imai, Yasuharu; Niki, Etsuo (2009). "Hydroxyoctadecadienoic acid and oxidatively modified peroxiredoxins in the blood of Alzheimer's disease patients and their potential as biomarkers". Neurobiology of Aging. 30 (2): 174–85. doi:10.1016/j.neurobiolaging.2007.06.012. PMID   17688973. S2CID   23418993.
  91. Yoshida, Yasukazu; Umeno, Aya; Shichiri, Mototada (2013). "Lipid peroxidation biomarkers for evaluating oxidative stress and assessing antioxidant capacity in vivo". Journal of Clinical Biochemistry and Nutrition. 52 (1): 9–16. doi:10.3164/jcbn.12-112. PMC   3541426 . PMID   23341691.
  92. Niki, Etsuo (2014). "Biomarkers of lipid peroxidation in clinical material". Biochimica et Biophysica Acta (BBA) - General Subjects. 1840 (2): 809–17. doi:10.1016/j.bbagen.2013.03.020. PMID   23541987.
  93. Liu, Yan; Wang, Duan; Li, Di; Sun, Ruifang; Xia, Min (2014). "Associations of retinol-binding protein 4 with oxidative stress, inflammatory markers, and metabolic syndrome in a middle-aged and elderly Chinese population". Diabetology & Metabolic Syndrome. 6 (1): 25. doi: 10.1186/1758-5996-6-25 . PMC   3938900 . PMID   24559154.