Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2011 Sep 13.
Published in final edited form as: Cardiovasc Hematol Agents Med Chem. 2011 Jul 1;9(3):154–164. doi: 10.2174/187152511797037619

12-Lipoxygenase: A Potential Target for Novel Anti-Platelet Therapeutics

Jennifer Yeung 1, Michael Holinstat 1,*
PMCID: PMC3171607  NIHMSID: NIHMS318781  PMID: 21838667

Abstract

Platelets play an essential role in the regulation of hemostasis and thrombosis and controlling their level of activation is central to prevention of occlusive clot formation and stroke. Although a number of anti-platelet targets have been identified to address this issue including COX-1, the P2Y12 receptor, the integrin αIIbβ3, and more recently the protease-activated receptor-1, these targets often result in a significant increased risk of bleeding which may lead to pathologies as serious as the thrombosis they were meant to treat including intracranial hemorrhage and gastrointestinal bleeding. Therefore, alternative approaches to treat uncontrolled platelet activation are warranted. Platelet-type 12-lipoxygenase is an enzyme which oxidizes the free fatty acid in the platelet resulting in the production of the stable metabolite 12-hydroxyeicosatetraenoic acid (12-HETE). The role of 12-HETE in the platelet has been controversial with reports associating its function as being both anti- and pro-thrombotic. In this review, the role of 12-lipoxygenase and its bioactive metabolites in regulation of platelet reactivity, clot formation, and hemostasis is described. Understanding the mechanisms by which 12-lipoxygenase and its metabolites modulate platelet function may lead to the development of a novel class of anti-platelet therapies targeting the enzyme in order to attenuate injury-induced clot formation, vessel occlusion and pathophysiological shifts in hemostasis.

Keywords: 12-LOX, anti-platelet therapeutics, eicosanoids, fatty acid oxidation, hemostasis, lipoxygenase, platelets, thrombosis

PLATELET THERAPY IN CARDIOVASCULAR DISEASE

Incidence of CVD

Recently, the American Heart Association reported that cardiovascular diseases (CVD) accounted for 33.6% of all deaths in the United States [1], and is the single leading cause of death in the nation and worldwide [2] occurring before age 65. Furthermore, more than 67.3% of adults over the age of 20 are reported to be obese, which is one of the major contributing factors to developing CVD along with smoking. Consequently, surgical procedures for cardiovascular complications have increased by 27% from 1997 to 2007 [1]. Despite these alarming statistics, the mortality rate resulting from CVD complications has slightly decreased, although the burden of the disease still remains high. This is due, in part, to the successful development and widespread use in the clinic of a number of anti-platelet drugs.

Current Anti-Platelet Drugs

One of the earliest targets for anti-platelet therapy was cyclooxygenase-1 (COX-1), responsible for the formation of prostanoids, which are involved in inflammation [3]. A number of pharmacological approaches have been developed over the years to inhibit COX activation in platelets including aspirin, indomethacin, and nonsteroidal anti-inflammatory drugs (NSAIDS) [4]. The efficacy in treating and preventing vascular-occulsive events with COX-1 inhibitors may be limiting in a segment of the population [5, 6]. One study for example, reported that greater than 33% of the patients with CVD exhibited aspirin resistance [3, 7]. These observations however are controversial since no generally acceptable definition of aspirin resistance exists and intra-individual differences in COX-1 inhibition can be highly variable [8]. Another target on the platelet which has successfully been inhibited in order to limit platelet activation is the ADP receptor, P2Y12. Targeting P2Y12 with clopidogrel, a pro-drug that binds to and inhibits receptor activation, has been shown to be effective in further reducing platelet activation when given as dual anti-platelet therapy with aspirin [9]. However, due to the nature of this pro-drug and the genetic variability of the cytochrome P450 enzyme required to convert clopidogrel to its active form, its utility is limited as is evident from the development of next generation P2Y12 inhibitors ticagrelor and prasugrel [10, 11]. Integrin IIb/IIIa inhibitors (abciximab, eptifibatide, tirofiban), although a breakthrough in antibody-based anti-platelet therapy [11-13], have resulted in an increase in bleeding complications following PCI. Furthermore, some patients developed frequent myocardial infarction and refractory ischemia post-tibrofiban administration [14].

Adverse side effects such as bleeding are of primary concern for current treatment against platelet activation in CVD therapy, especially prior to and during surgical procedures [15]. Thus, alternative strategies which would inhibit platelet activation while minimizing the bleeding side effect are warranted. A novel target for anti-platelet therapy in the treatment of patients with cardiovascular related diseases may be human 12-lipoxygenase (12-LOX). In order to develop potential 12-LOX inhibitors however, we must have a comprehensive understanding of its pathophysiological and biochemical implications and the role 12-LOX metabolites play in the vascular system. Our current knowledge of this enzyme and its oxidized products in platelets and other tissues is still limited, but the preliminary evidence shows the potential advantages of inhibiting 12-LOX on the platelet to treating human diseases.

Historical Overview of Lipoxygenases

Lipoxygenases (LOs) are a family of nonheme iron dioxygenases, originally known as lipoxidases that catalyze the oxidation of polyunsaturated fatty-acids such as linoleic acid or arachidonic acid (AA) containing the cis-methylene interrupted diene structure and esters yielding conjugated hydroperoxides [16]. The earliest lipoxygenase studies showed 15-lipoxygenase purified from soybean oxidized unsaturated moieties of fatty acids [17] at a specific carbon site of the acyl chain; that eventually allowed lipoxygenase isoforms to be identified according to their site of carbon oxidation and stereochemistry. The transformation of AA to 12(S)-hydroxy-5,8,10,14-eicosatetraenoic acid (12(S)-HETE) was first demonstrated in human and bovine platelets in the mid-1970s [18]. These metabolites, produced by 5-, 8 -, 12-, and 15- LO, have been reported to potentially form signaling lipid mediators that exert their effects through the G protein–coupled receptor (GPCR) [19]. Lipoxygenases and their metabolites have been demonstrated experimentally and clinically to be restricted to specific cells or tissues and show significant species specificity. To date, three isoforms of 12(S)-lipoxygenases have been identified in human accordingly to their cell type: epidermis, leukocyte, and platelet [20]. 12-LOX activation has been reported to play a role in a number of diseases including psoriasis [21] and ulcerative colitis [22, 23]. Despite intensive research in this field, the majority of the discoveries of 12-lipoxygenase functions and their implications have been established using animal models. Thus, our current understanding of the biological significance of these enzymes and products in humans is still limited. 12-LOX isoforms and their products will be the focus of this review showing their involvement in platelet reactivity contributing to hemostasis and thrombosis and the overall scheme in developing alternative targets to prevent vascular occlusion, myocardial infarction, and stroke.

OVERVIEW of 12-LOX

Activation of Platelets through a number of receptors is known to result in activation of cytosolic phospholipase A2 (cPLA2), a lipid lipase that generates free fatty acid such as AA from the phospholipid membrane by cleaving the sn-2 position [18]. Once AA is formed, it is available for oxidation by either COX-1 or 12-LOX and can produce a number of bioactive lipid metabolites [18]. In leukocytes, oxidized AA will result in the production of prostaglandin E2 (PGE2) and leukotriene B4 (LTB4) [24, 25] as pro-inflammatory effectors or thromboxane (TxA2) and 12-HETE in platelets. Recently, we have shown that COX-1 and 12-LOX-mediated signaling may rely on different pools of AA based on the kinetics of TxA2 and 12-HETE formation and their differential reliance on cPLA2 at the surface of the platelet [26]. 12-LOX activation is also thought to be important for dense granule secretion in platelets as well as normal platelet aggregation and adhesion [27]. This is not surprising considering blocking 12-LOX attenuates aggregation and integrin activation in the presence of thromboxane, collagen, thrombin, and protease-activated receptors (PARs) [28-30]. Additionally, 12-LOX has also been implicated to play a role in regulating calcium mobilization. A role for 12-LOX in the platelet using one of the classical 12-LOX inhibitors, baicalein, was first described in the mid-1990s, where stimulation with AA in the absence of 12-LOX resulted in a significant attenuation of thrombin-induced calcium [Ca2+]i transients and aggregation [27]. In addition to oxidation, 12-LOX in platelets has also been reported to have lipoxin synthase activity. Lipoxins, such as LXA4 and LXB4, are tetraene-containing eicosanoids generated from exogenous LTA4 that induces vasoconstriction of the smooth muscles and regulates neutrophil function via binding at specific recognition sites [31-34]. The molecular observations above confirm an important role for 12-LOX in human platelet reactivity and a renewed interest in this field attests to the therapeutic potential inherent with regulation of 12-LOX. Finally, although human studies to date are limited to ex vivo platelet reactivity and thrombosis, a number of animal models have added crucial information as to the potential role of 12-LOX in hemostasis including studies with 12/15-LO knockout mice, canines, porcine, and rabbits, show varying and sometimes unrelated physiological effects compared with humans [35] (Table 1). Although 12-LO targets and functions appear to be species related, 12-LO activation in a number of platelet models has been correlated to modulation of platelet reactivity in vivo (see Table 1). We must be careful not to interpret these studies to mean that 12-LOX is essential for normal platelet activation, but rather that elimination of 12-LOX protein or activity may be related to normal regulation of hemostasis and thrombosis. This is an area that will need further investigation in order to determine how the animal models translate to platelet function in the human. Recent work, however, does indicate that altered 12-LOX function may be related to defects in hemostasis in vivo [36].

Table 1.

12-LOX Expression and Function in Different Species

Species Cell Type Function
Human Adrenal glomerulosa cells LO activation participates in aldosterone’s stimulatory effects of angiotensin II (ANG) [37]
Epithelial lens cells (HLECs) Growth in response to EGF and insulin [38]
platelet 12-HETE and other metabolites formation [26, 39]; platelet activation [2, 40]
Mouse Pancreatic islets and adipose tissue Cytokine production in adipocytes and macrophages shown to impair insulin signaling such as tumor
necrosis factor-alpha (TNF-α), interleukin 6(IL-6), IL-12p40, and monocyte chemoattractant protein
(MCP-1) [35]
Melanoma cells Cell proliferation [39, 41]
Platelets 12-HETE and other metabolites formation shown to modulate platelet reactivity [42]
Porcine Anterior pituitary Possible gonadotroph regulation [43]
Rabbit Corneal epithelial cells Regulation of epithelial cell proliferation and the rate of corneal re-epithelialization following an
injury [44]
Rat Mesangial cells in renal Indirectly involved in the expression of P-cadherins [45-47]
Endothelium Vasconstriction [48]
Canine Gingival tissue, hepatocytes [49],
Kupffer cells [50] and platelets
[51]
Involved in neutrophil activity [50] and vasoconstriction and thrombosis [51]

12-LOX SUBSTRATES

Polyunsaturated Fatty Acids (PUFA) as Regulators of 12-LOX-Mediated Activity

A possible alternative approach to anti-platelet therapy may lie in the dietary intake of certain essential fatty acids (EFA). Large prospective studies have shown that there is a positive relationship between increased dietary intake of ω-3 fatty acids and reduced CVD. For instance, Eskimos from the West Coast of Greenland whose diet included large quantities of whale, fish, and seal, had a positive correlation with lower incidence of myocardial infarction, stroke, and mild tendency to bruise compared to the overall population [52]. Further, rats receiving Thomas-Hartroft thrombogenic diet were subsequently fed with unsaturated fatty acid supplements which comparatively showed reduced thromboplastin generation and mortality compared to control rats [53]. Interestingly, the incidence of mortality due to ischemic heart and cerebrovascular disease between 1950 and 1982 was found to be lower in fishing communities relative to farming villages in Japan [54-56]. This epidemiological study correlated the low level of CVD-related events with high levels of eicosapentaenoic acid (EPA) from fish. Thus, larger amount of specific PUFAs in the diet may result in an increased pool of fatty acids made available to 12-LOX in the platelet. Depending on the specific fatty acid substrates available, oxidation by 12-LOX may generate several pools of eicosanoids which may in turn act as key regulators of platelet reactivity. These nutritional studies, although supportive of a role for 12-LOX in mediating changes based on altered free fatty acid oxidation, lack the scientific rigor required in order to connect the observed benefit of PUFA supplementation with a substantive link to altered 12-LOX oxidized metabolites as being causative to these reported cardiovascular benefits. Furthermore, while the addition of PUFAs as a dietary supplement may be beneficial, the amount and type of fatty acids added to the diet needs to be carefully monitored in patients with existing cardiovascular risk. Previous studies for example, have suggested that docosahexaenoic acid (DHA) has an adverse effect on diabetics and elderly who are already suffering from a lower antioxidant potential. In one study, platelets from an elderly subject with low DHA were given low DHA supplements and exhibited lower lipid peroxidation activity, whereas incubated with high DHA concentration induced higher 12-HETE formation [57, 58]. As for EPA, fatty acid supplements given to patients resulted in attenuation of platelet aggregation. TxB2 production has also been shown to be inhibited in platelets pre-treated with EPA [57].

12-LOX Substrate Availability

Dietary intake of certain EFAs ( ω-3 or ω-6) have been shown to impact the type of metabolic products generated by 12-LOX. For instance AA, an ω-6 series fatty acid derived from cis-linoleic acid, can be found mostly in peanut oil. As for ω-3, fish oil, flaxeed, and algal oil are the common sources for α-linolenic acid (ALA), DHA, and EPA [59]. Fatty acids in platelet membranes containing DHA have been reported to be oxidized by 12-LOX to various hydroxyl docosahexaenoic acid (HDoHE) isoforms, but largely forming 14-HDoHE in an agonist and calcium-dependent manner [18]. A number of metabolic products are formed following 12-LOX oxidation of another fatty acid, EPA, which include AA, TxB3, 12(S)-hydroxyeicosapentanoic acid (12-HEPE), and hydroxy-5,8,10-14-heptadecatetraenoic acid (HhTE) [60, 61]. One study showed that EPA conversion to TxB3 depended on the presence of hydroxy-5,8,10, 14-eicosatetraenoic acid (12-HpETE [61], whereas, pre-incubation with 12-HETE did not induce EPA metabolism. In another case, pre-incubation with 12(S)-HpETE has been shown to increase the amount of nonesterified AA in collagen stimulated platelets, significantly enhancing platelet aggregation and the formation of TXB2 [62]. The eicosanoid metabolites, once formed, can have a number of regulatory functions in the platelet probably through both autocrine as well as paracrine signaling schemes. The physiological and cellular effects due to oxidized products are further discussed in detail below.

Direct 12-LOX Regulation of Eicosanoid Metabolites

Eicosanoid metabolites are able to be further oxidized by 12-LOX. 12-LOX can catalyze 5-HETE to generate 5(S), 12-dihydroxyeicosatetraenoic acid (12(S)-DHETE) and 15-HETE to 14,15-DHETE in platelets under conditions of prolonged exposure to the enzyme [63]. Similarly, exogenous AA and 5-HETE show reduced production of 5(S), 12(S)-DHETE. 11,12-DHETE has also been reported to be generated in a 12-LOX dependent manner [64].

BIOLOGICAL ROLE OF METABOLITES

Metabolite Profiling in the Blood

Profiling of lipid metabolites and mediators in whole blood in the presence of the calcium ionophore A23187, demonstrated that 12-LOX contributes a large proportion of the total products formed [65] compared with the other lipoxygenases. The predominant products were 12-HETE, 12-HEPE, and 12-hydroxydocosahexaenoic acid (12-HDHA). A significant number of 5-LOX products, including LTB4, LTB5, and PGE2, were also found in circulation under these conditions. Shifting ω-3 or ω-6 content in the lipid bilayer of the cells has been shown to contribute to differences in their respective lipid mediators and metabolites and thus regulate biochemical and physiological events in the cells of the individuals [60, 66].

12-LOX Metabolite Regulation of Various Tissues

12-LOX oxidation of various fatty acids in the platelet can result in the formation of a number of unique eicosanoid metabolites. Many, if not all, of these eicosanoids can play a regulatory role both within the platelet itself as well as at distal tissues and organ systems. Previous studies support a pro-inflammatory role for eicosanoid formed through 12-LOX oxidation of ω-3 fatty acids and AA in a number of animal models including the mouse and rabbit [67] Further, work in cell lines confirms a role for these eicosanoids in a number of distant tissue beds as well. For example, mouse pre-adipocytes 3T3-L1 cells pre-treated with 12(S)-HETE and 12(S)-HpETE induce upregulation of proinflammatory cytokine genes, such as tumor necrosis factor-alpha (TNF-α), interleukin 6(IL-6), IL-12p40, and monocyte chemoattractant protein (MCP-1) [35]. In the human epidermis, 12(R)-HETE, which is sterochemically different from platelet-derived 12(S)-HETE, have been reported to increase DNA synthesis and plays a significant role in psoriasis [68, 69].

12-LOX Metabolite Regulation of Platelets

The role of the AA oxidized metabolite in platelets, 12-HETE, is not well understood. Furthermore, published work in this area of research is controversial as 12-HETE has been reported to be pro-thrombotic, anti-thrombotic, a well as inert towards platelet activity. One study indicated that 12-HETE had no effect on either basal or thrombin-induced [Ca2+]i levels or aggregation [70]. Conversely, other reports showed a clear potentiation of thrombin-induced aggregation in platelets in the presence of 12-HETE [71]. 12-HpETE has also been shown to stimulate 12-LOX but inhibit COX-1 in lysed platelets[72] and one report indicated that 12(S)-HETE acts as an inhibitor of platelet and neutrophil PLA2 activity [73]. Additionally, the eicosanoid, 15-HETE, which is primarily produced in leukocytes, has been reported to act as an inhibitor of 12-LOX [74]. Other eicosanoid products, such as 12-HPEPE and 12-HEPE originating from 12-LOX oxidation of EPA, are thought to elicit an inhibitory effect on platelet aggregation [54, 55]. In addition to their effects on aggregation, 12-HPEPE and 12-HEPE have been shown to attenuate serotonin (5-HT) release mediated by AA and collagen in a dose dependent manner [75]. The level of fatty acid substrate available to 12-LOX may also play a role in its physiological regulation of platelet function. On the one hand, incorporation of DHA into one’s diet lowers lipid peroxidation, which leads to attenuation of platelet reactivity at 200 μM [76] by inhibiting TXA2 induced aggregation [77], whereas higher DHA concentrations of 400 μM resulted in increased prostaglandins [76]and potentiation of platelet reactivity. In a clinical trial, an elderly group receiving 30 mg of DHA per day resulted in lower lipid peroxidation levels and increased levels of vitamin E [78]. Additionally, elderly patients receiving 150 mg DHA and 30 mg EPA resulted in lower oxidative stress in platelets [57]. Contrary to these observations, one group has reported an increase in plasma lipid peroxidases in healthy individuals receiving 2 g EPA and 1.3 g DHA daily [79]. This paradoxical phenomenon has also been observed for the 12-LOX metabolite, 12-HEPE.

12-LOX Metabolite Regulation of CVD

Patients with essential hypertension, a significant risk factor for vascular occlusion [80], have shown an increase in the basal level of 12(S)-HETE in the platelets and in urinary excretion of 12(S)-HETE compared to healthy subjects [81]. Further, the protein levels of 12-LOX in the cytosolic fraction from hypertensive patients was much higher than in normotensive subjects. Thrombin-mediated 12(S)-HETE generation however, did not differ between the groups implying a potential role for genetic variation in the levels of 12-HETE formed [81]. SNP analysis has uncovered an association in the coding polymorphism of the 12-LOX gene with essential hypertension and urinary production of 12(S)-HETE [82].

Age is known to be directly correlated to an increased risk for cardiovascular disease [83, 84]. Similarly, platelet reactivity is also increased in older patients due, in part, to increasing levels of AA and TxA formation [85-87] Although 12-LOX product formation has not been studied in young versus older subjects, it is likely that 12-HETE is also increased in this segment of the population. Additionally, diabetics have been shown to exhibit an increase in PLA2 activity linked to an increase in TxA2 formation [88]. 12-LOX has been reported to have a link to platelet function in diabetes as well. 12-HETE levels in platelets from a group of Japanese patients with type 2 diabetes were observed to be decreased compared with healthy subjects [89], whereas noninsulin dependent patients in US showed an increase in 12-HETE formation in the urine samples compared to healthy subjects [90]. These controversial observations, although perplexing, support a link between the levels of 12-LOX activity in the platelet and diabetes.

12-LOX INHIBITORS

The studies described above show that regulating the amount of essential fatty acids and their metabolites via 12-LOX is essential in understanding both the pathophysiological processes of the platelets and CVD. Various groups have screened for potential natural and small molecule drugs targeting 12-LOX, however, many of these screens have failed due to problems with efficacy, off-target effects, and adverse events, both in animals and human platelets (Table 2). One of the earliest drugs tested on arachidonate 12-LOX was an acetylenic acid, 4,6-10-13-eicosatetrayonic acid (4,7,10,13-ETYA) [113]. This approach however, also targeted human peripheral neutrophil 5-LOX with an ID50 of 2-3 uM and other lipoxygenases from different sources and was therefore not developed further. Esculetin, also known as curcumin, was shown to inhibit 12-HETE production in both human and rat platelets [119], but did not inhibit formations of TxB2 and HHT [146]. Besides curcumin, baicalein (5,6,7-tihydroxyflavone), a compound extracted from Radix Scutellariae roots [96], was first reported to selectively inhibit 12-LOX in human platelets in the 1980s [147] without affecting cyclooxygenase activity [148]. In addition, platelet activation and ATP secretion stimulated by Chlyamydia pnemoniae was markedly reduced by this inhibitor [92]. More recent data suggests that baicalein inhibits cPLA2 in human platelets and that some of its effects may be due to a lower level of AA formation following initial platelet activation. Baicalein has also been reported to be an inhibitor of CYP2C9, an enzyme involved in drug metabolism [149] as well as other human LOs and COXs [95]. In addition to its’ off target effects, baicalein in rats showed that the amount of 12-HETE produced in the presence of the inhibitor and thrombin stimulation did not correlate with the potentiation of contractile responses in the artery [150].

Table 2.

12-Lipoxygenase Inhibitors and Targets

Inhibitor Origin Target(s)
CDC 12-LOX in human platelets; 5-LO in rats [91-93], 5-LO in human polymorphonuclear
leukocytes and monocytes [93, 94]
Baicalein Scutellariae roots human reticulocyte 15-LOX and 12-LOX in platelets [27, 95, 96] , prolyl-4 hydroxylases
(PHDs) in human hepatoma cells and mouse fibroblasts 3T3-L1 [97]; Raf-1 mediated
phosphorylation of MEK-1 in rat glioma cells [98]; phosphatidylinositol 3-kinase (PI3K)
inhibitor in human cancer cell lines e.g. colorectal, lung, gastric, ovarian, renal, melanoma,
breast, glioma, prostate [99]
(NDGA) Creosote bush 5-LOX, 12-LOX prostaglandin [100], inhibits HER2 and IGF-1 receptor tyrosine kinases
in human breast cancer cells, [101, 102]; neutrophil phospholipase A2 [103]
BW755C Phenidone analogue dual inhibitor 12-LOX and COX-1 in platelets [104]; 5-LOX in leukocytes [105]
AA-861 5-LO in human and guinea pig peritoneal leukocytes [106]; 12-LO in mouse epidermis
[107, 108]; tyrosine phosphatase of the receptor tyrosine kinase signaling pathway in
mouse embryonic fibroblasts [109]
Timegadine COX and LO in rabbit, rat, bovine, horse platelets [110-112] [110]
ETYA 12-LOX [113]; COX-1 [114-116], cytochrome P-450 ensymes in rat thyroid FRTLA-5
cells [117]; neutrophil phospholipase A2 [103]; Δ-6 desaturase in mouse hepatic cells
[118]
Esculetin 12-LOX in human platelets [61, 119]; 5-LO [120],
BHPP Hydroxamic acids 12/15-LO in rats and mice [121]
Panaxynol a polyacetylene compound from
Ginseng radix, Fang-Feng and
Panx ginseng
COX, leukocyte 12-LO and platelet 12-LO in humans and porcine. 15-LO in rabbit [122];
cholesterol acyltransferase in rat liver [123]; 15-hydroxyprostaglandin dehydro-
genase (PGDH) in rabbit gastric mucosa [124]
Falcarindiol Panaxynol analog leukocyte 12-LO and platelet 12-LO in human, and 15-LO [122]in rabbit reticulocyte;
GABA degradative enzymes GABA transaminase (GABA-T) [125]
BW A4C acetohydroaxmic acid 12- and 15-LO in bovine leukocyte, and 15-LO human leukocyte [126, 127]; secretory
type II phospholipase A2 (sPLA2-II) in guinea-pig alveolar macrophages [128]
Hinokitiol Derived from tropolone, a
constituent of the wood
Chymacyparis taiwanesis
12-LOX in human platelet [129, 130]; HIF-specific prolyl-4-hydroxylases (PHDs) [131]
in human HepG2 hepatoma and HeLa cervical epithelial cells; metalloprotease in
injects [132]
KY11449 Derived from Streptoverticillium
hadanonense
Bovine 12-LO, rat basophilic leukemia cells (RBL-1) 12-LO [133]
Gossypol Polyphenolic derived compound
from cotton plant
5-LO and 12-LO [134] in human neutrophils and platelets, and rat basophilic leukemia
cells (RBL-1) [135]; 11β-hydroxysteroid dehydrogenase (11β-HSD) activity in human and
rat kidney [136]; Bcl-2 in human ovarian cancer cell lines OV433 and TOV112D [137]
Catechins Green tea leaves soybean lipoxygenases and 12-LO in rabbit and human [130]; tumor-associated NOX
(tNOX) in human mammary adenocarcinoma cells (BT-20) and HeLa [138]
Quercetin LOs in rat mast cells, 12-LOX, and 5-LO in RBL-1[139]; neutrophil phospholipase
A2 [103]; P-glycoprotein-mediated efflux transport and CYP3A4 enzyme [140],
human glycolate oxidase [141], sPLA2 in human platelets [142]
MK 866 12-LOX in human platelets [127] , 5-LO in rats [143, 144]
12-LOX small molecule
inhibitors
Small molecule screen selectively targets 12-LOX in human platelets [145]

Other potential inhibitors which have shown little efficacy toward 12-LOX include 1) Dicranin (acetylenic fatty acid: 9,12,15-octadecatrien-6-ynoic aic), extracted from Dicranum Scoparium, which weakly inhibited COX-1, but resulted in an increase in 12-HETE [151], 2) Knipholone, which is isolated from the roots of Kniphofa foliosa and was shown to inhibit leukotriene synthesis, but only weakly inhibit 12-HETE production [152], and 3) OPC-29030 which inhibits thrombin-mediated 12(S)-HETE production [153]. Additionally, Hinokitiol, extracted from Japanese wood, was shown to be a selective 12-LOX inhibitor. Unfortunately, Hinokitiol has also been reported to be cytotoxic and terato-geneic on living tissues [129, 154]. Recently, there has been an increased interest in developing a highly selective small molecule inhibitor targeting 12-LOX. These compounds structurally exhibit greater selectivity than the previous natural inhibitors described above due to their selectivity in distinguishing and LO paralogs in species specific tissues/cells [145, 155]. These small molecule inhibitors may possibly reduce off-target effects in the system due to their greater selectivity and aid in clarifying the role of 12-LOX in the pathophysiology of thrombosis in the human.

CONCLUSIONS

Cardiovascular disease remains the leading cause of death in the world and is a growing problem both globally as well as within the United States. Research spanning over three decades has convincingly established a central role for platelet activation in the pathophysiology of cardiovascular disorders and acute coronary syndrome. Although current pharmacological therapy for treatment of diseases caused by blood clots, such as heart disease and stroke, has greatly improved the morbidity profile of patients with CVD, new approaches are warranted which alone, or in conjunction with currently approved pharmacological interventions such as aspirin or clopidogrel, will further decrease morbidity and mortality due to unwanted clot formation. Although targeting of enzymes such as COX-1 or surface receptors including P2Y12, PAR1, and integrin receptor αIIbβ3, has been extremely useful in decreasing morbidity due to MI, these therapies have failed to significantly shift the incidence of mortality in these patients [156, 157]. This may be due, in part, to the fact that these anti-platelet drugs do not fully attenuate platelet activation, can have delayed onset and long durations of action, and may result in significant morbidity them selves due to bleeding complications [156, 157]. New therapeutic approaches targeting the level of platelet activation necessary to inhibit vessel occlusion and stroke without significantly increasing bleeding are needed. To address this problem, inhibiting platelet activation of a secondary pathway may allow for further inhibition of clot growth and stability without significantly altering the bleeding profile following vascular insult as is observed with inhibition of secondary pathways including COX-1 and P2Y12. COX-1 inhibition for example, although only blocking formation of the weak agonist, TxA2, has proven to be one of the most prescribed pharmacological approaches in anti-platelet therapy. 12-LOX may be a viable future target for anti-platelet therapy. Studies have shown that a link exists between the levels of 12-LOX and cardiovascular risks such as type 2 diabetes and hypertension. Furthermore, 12-LOX metabolites such as 12-HETE, have been shown to potentiate platelet activation, thrombin generation, and calcium mobilization. Recent work using small molecule inhibitors now supports a pro-thrombotic role for 12-LOX in the human platelet. Thus, targeting this enzyme in concert with inhibition of targets such as COX-1 or P2Y12 may allow for attenuation of the platelet clotting cycle without a significant increased risk of bleeding. The next phase of 12-LOX investigations should focus on whether inhibiting 12-LOX in both in vitro as well as in vivo animal model systems can either inhibit platelet activation to a greater extent compared to aspirin or clopidogrel, or determine if inhibition of 12-LOX works synergistically with the already established pharmacological approaches in order to potentially shift the threshold for platelet activation further to the right on an agonist dose-response curve.

Although 12-LOX was identified in the early 1970s by Hamberg and Samuelsson [158], identifying the regulatory role of 12-LOX and its metabolites in platelet function has been difficult, in no small part due to the poor selectivity of naturally occurring lipoxygenase inhibitors (see Table 2). Recently however, several research groups have revisited this enzyme and are developing a number of natural and synthetic molecule approaches in order to identify highly selective inhibitors against platelet-type 12-lipoxygenase in the human. The first generation of these inhibitors is now being tested in human platelets and early results support the potential targeting of this enzyme for future use as an anti-platelet therapy. Prior to establishment of 12-LOX as a viable target, suitable animal models will need to be identified in order to determine not only the effectiveness and safety of 12-LOX inhibitors, but to also identify the role of 12-LOX in hemostasis in vivo. Platelet-type 12-LOX is not the only potential target in development, however, its relatively selective expression in megakaryocytes and platelets, and its pro-thrombotic activity in human platelets supports further development of this target for anti-platelet therapeutics.

ACKNOWLEDGEMENTS

This work was funded in part by the National Institutes of Health grant HL089457 to MH.

ABBREVIATIONS

CVD

Cardiovascular diseases

12-LOX

Human 12-lipoxygenase

5-HT

Serotonin

AA

Arachidonic acid

ALA

α-linolenic acid

COX

Cyclooxygenase

DHA

Docosahexaenoic acid

DHETE

Dihydroxyeicosatetraenoic acid

EFA

Essential fatty acids

EPA

Eicosapentaenoic acid

ETYA

Eicosatetrayonic acid

GPCR

G protein-coupled receptor

HDHA

Hydroxydocosahexaenoic acid

HDOHE

Hydroxyl docosahexaenoic acid

HEPE

Hydroxyeicosapentanoic acid

HETE

Hydroxy-5,8,10,14-eicosatetraenoic acid

HHTE

Hydroxy-5,8,10-14-heptadecatetraenoic acid

HPETE

Hydroxy-5,8,10,14-eicosatetraenoic acid

IL

Interleukin

LA

Cis-linoleic acid

LO

Lipoxygenase

LT

Leukotriene

LX

Lipoxin

MCP-1

Monocyte chemoattractant protein

NSAIDS

Nonsteroidal anti-inflammatory drugs

PAR

Protease-activated receptor

PG

Prostaglandin

PLA

Phospholipase

PUFA

Polyunsaturated fatty acid

SNP

Single nucleotide polymorphism

TNF-A

Tumor necrosis factor-alpha

TX

Thromboxane

Footnotes

CONFLICT OF INTEREST None of the authors has a conflict of interest for this manuscript.

REFERENCES

  • [1].Roger VL, Go AS, Lloyd-Jones DM, Adams RJ, Berry JD, Brown TM, Carnethon MR, Dai S, de Simone G, Ford ES, Fox CS, Fullerton HJ, Gillespie C, Greenlund KJ, Hailpern SM, Heit JA, Ho PM, Howard VJ, Kissela BM, Kittner SJ, Lackland DT, Lichtman JH, Lisabeth LD, Makuc DM, Marcus GM, Marelli A, Matchar DB, McDermott MM, Meigs JB, Moy CS, Mozaffarian D, Mussolino ME, Nichol G, Paynter NP, Rosamond WD, Sorlie PD, Stafford RS, Turan TN, Turner MB, Wong ND, Wylie-Rosett J. Heart disease and stroke statistics--2011 update: a report from the American Heart Association. Circulation. 2011;123(4):e18–e209. doi: 10.1161/CIR.0b013e3182009701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [2].Mendis S, Lindholm LH, Anderson SG, Alwan A, Koju R, Onwubere BJ, Kayani AM, Abeysinghe N, Duneas A, Tabagari S, Fan W, Sarraf-Zadegan N, Nordet P, Whitworth J, Heagerty A. Total cardiovascular risk approach to improve efficiency of cardiovascular prevention in resource constrain settings. J. Clin. Epidemiol. 2011 doi: 10.1016/j.jclinepi.2011.02.001. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • [3].Capone ML, Tacconelli S, Rodriguez LG, Patrignani P. NSAIDs and cardiovascular disease: transducing human pharmacology results into clinical read-outs in the general population. Pharmacol. Rep. 2010;62(3):530–535. doi: 10.1016/s1734-1140(10)70310-8. [DOI] [PubMed] [Google Scholar]
  • [4].Nawarskas JJ, Clark SM. Ticagrelor: a novel reversible oral antiplatelet agent. Cardiol. Rev. 2011;19(2):95–100. doi: 10.1097/CRD.0b013e3182099d86. [DOI] [PubMed] [Google Scholar]
  • [5].Trelle S, Reichenbach S, Wandel S, Hildebrand P, Tschannen B, Villiger PM, Egger M, Juni P. Cardiovascular safety of non-steroidal anti-inflammatory drugs: network meta-analysis. BMJ. 2011;342:c7086. doi: 10.1136/bmj.c7086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [6].Rafferty M, Walters MR, Dawson J. Anti-platelet therapy and aspirin resistance - clinically and chemically relevant? Curr. Med. Chem. 2010;17(36):4578–4586. doi: 10.2174/092986710794182962. [DOI] [PubMed] [Google Scholar]
  • [7].Patrono C, Garcia, Rodriguez LA, Landolfi R, Baigent C. Low-dose aspirin for the prevention of atherothrombosis. N. Engl. J. Med. 2005;353(22):2373–2383. doi: 10.1056/NEJMra052717. [DOI] [PubMed] [Google Scholar]
  • [8].Michelson AD, Cattaneo M, Eikelboom JW, Gurbel P, Kottke-Marchant K, Kunicki TJ, Pulcinelli FM, Cerletti C, Rao AK. Aspirin resistance: position paper of the Working Group on Aspirin Resistance. J. Thromb. Haemost. 2005;3(6):1309–1311. doi: 10.1111/j.1538-7836.2005.01351.x. [DOI] [PubMed] [Google Scholar]
  • [9].De Schryver EL, Algra A. Secondary stroke prevention with antithrombotic drugs. Curr. Vasc. Pharmacol. 2010;8(1):129–133. doi: 10.2174/157016110790226543. [DOI] [PubMed] [Google Scholar]
  • [10].Iyu D, Glenn JR, White AE, Fox SC, Dovlatova N, Heptinstall S. P2Y(12) and EP3 antagonists promote the inhibitory effects of natural modulators of platelet aggregation that act via cAMP. Platelets. 2011 doi: 10.3109/09537104.2011.576284. [Epub ahead of print] [DOI] [PubMed] [Google Scholar]
  • [11].Riva L, Di Pasquale G, Casella G, Calabrese D, Zagnoni S, Pallotti MG. Antiplatelet therapy in acute coronary syndromes: state of the art and new perspectives. G. Ital. Cardiol. (Rome) 2010;11(12 Suppl. 3):27S–33S. [PubMed] [Google Scholar]
  • [12].Leclerc JR. Platelet glycoprotein IIb/IIIa antagonists: lessons learned from clinical trials and future directions. Crit. Care Med. 2002;30(5 Suppl.):S332–340. doi: 10.1097/00003246-200205001-00025. [DOI] [PubMed] [Google Scholar]
  • [13].Kimmelstiel C, Phang R, Rehman A, Rand W, Miele R, Rhofiry J, MacIsaac DA, Gouveia W, Denier D, Becker RC. Short-term comparative outcomes associated with the use of GP IIb/IIIa antagonists in patients undergoing coronary intervention. J. Thromb. Thrombolysis. 2001;11(3):203–209. doi: 10.1023/a:1011904718960. [DOI] [PubMed] [Google Scholar]
  • [14].Zhao XQ, Theroux P, Snapinn SM, Sax FL. Intracoronary thrombus and platelet glycoprotein IIb/IIIa receptor blockade with tirofiban in unstable angina or non-Q-wave myocardial infarction. Angiographic results from the PRISM-PLUS trial (Platelet receptor inhibition for ischemic syndrome management in patients limited by unstable signs and symptoms). PRISM-PLUS Investigators. Circulation. 1999;100(15):1609–1615. doi: 10.1161/01.cir.100.15.1609. [DOI] [PubMed] [Google Scholar]
  • [15].Hennekens CH, Schneider WR, Hebert PR, Tantry US, Gurbel PA. Hypothesis formulation from subgroup analyses: nonadherence or nonsteroidal anti-inflammatory drug use explains the lack of clinical benefit of aspirin on first myocardial infarction attributed to “aspirin resistance”. Am. Heart J. 2010;159(5):744–748. doi: 10.1016/j.ahj.2009.11.033. [DOI] [PubMed] [Google Scholar]
  • [16].Bergstrom S, Holman RT. Total conjugation of linoleic acid in oxidation with lipoxidase. Nature. 1948;161(4080):55. doi: 10.1038/161055a0. [DOI] [PubMed] [Google Scholar]
  • [17].Theorell H, Holman RT, Akeson A. Crystalline lipoxidase. Acta Chem. Scand. 1947;1(6):571–576. doi: 10.3891/acta.chem.scand.01-0571. [DOI] [PubMed] [Google Scholar]
  • [18].Morgan LT, Thomas CP, Kuhn H, O’Donnell VB. Thrombin-activated human platelets acutely generate oxidized docosahexaenoic-acid-containing phospholipids via 12-lipoxygenase. Biochem. J. 2010;431(1):141–148. doi: 10.1042/BJ20100415. [DOI] [PubMed] [Google Scholar]
  • [19].Brash AR. Lipoxygenases: occurrence.; functions.; catalysis.; and acquisition of substrate. J. Biol. Chem. 1999;274(34):23679–23682. doi: 10.1074/jbc.274.34.23679. [DOI] [PubMed] [Google Scholar]
  • [20].Yoshimoto T, Takahashi Y. Arachidonate 12-lipoxygenases. Prostaglandins Other Lipid Mediat. 2002;68-69:245–262. doi: 10.1016/s0090-6980(02)00034-5. [DOI] [PubMed] [Google Scholar]
  • [21].Kragballe K, Fallon JD. Increased aggregation and arachidonic acid transformation by psoriatic platelets: evidence that platelet-derived 12-hydroxy-eicosatetraenoic acid increases keratinocyte DNA synthesis in vitro. Arch Dermatol. Res. 1986;278(6):449–453. doi: 10.1007/BF00455162. [DOI] [PubMed] [Google Scholar]
  • [22].McDonald CJ, Calabresi P. Psoriasis and occlusive vascular disease. Br. J. Dermatol. 1978;99(5):469–475. doi: 10.1111/j.1365-2133.1978.tb02012.x. [DOI] [PubMed] [Google Scholar]
  • [23].Yoshimura R, Matsuyama M, Tsuchida K, Kawahito Y, Sano H, Nakatani T. Expression of lipoxygenase in human bladder carcinoma and growth inhibition by its inhibitors. J. Urol. 2003;170(5):1994–1999. doi: 10.1097/01.ju.0000080296.54262.c8. [DOI] [PubMed] [Google Scholar]
  • [24].James MJ, Gibson RA, Cleland LG. Dietary polyunsaturated fatty acids and inflammatory mediator production. Am. J. Clin. Nutr. 2000;71(1 Suppl.):343S–348S. doi: 10.1093/ajcn/71.1.343s. [DOI] [PubMed] [Google Scholar]
  • [25].Willis AL. Prostaglandins and the future of medicine (an overview of currently evolving data and ideas). 1. Introductory comments: outline and scope of the series. Prostaglandins. 1974;5(6):506–511. doi: 10.1016/s0090-6980(74)80024-9. [DOI] [PubMed] [Google Scholar]
  • [26].Holinstat M, Boutaud O, Apopa PL, Vesci J, Bala M, Oates JA, Hamm HE. Protease-activated receptor signaling in platelets activates cytosolic phospholipase A2alpha differently for cyclooxygenase-1 and 12-lipoxygenase catalysis. Arterioscler Thromb. Vasc. Biol. 2011;31(2):435–442. doi: 10.1161/ATVBAHA.110.219527. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [27].Nyby MD, Sasaki M, Ideguchi Y, Wynne HE, Hori MT, Berger ME, Golub MS, Brickman AS, Tuck ML. Platelet lipoxygenase inhibitors attenuate thrombin- and thromboxane mimetic-induced intracellular calcium mobilization and platelet aggregation. J. Pharmacol. Exp. Ther. 1996;278(2):503–509. [PubMed] [Google Scholar]
  • [28].Prescott SM, Zimmerman GA, Stafforini DM, McIntyre TM. Platelet-activating factor and related lipid mediators. Annu. Rev. Biochem. 2000;69:419–445. doi: 10.1146/annurev.biochem.69.1.419. [DOI] [PubMed] [Google Scholar]
  • [29].Kroll MH, Schafer AI. Biochemical mechanisms of platelet activation. Blood. 1989;74(4):1181–1195. [PubMed] [Google Scholar]
  • [30].Jackson SP, Nesbitt WS, Kulkarni S. Signaling events underlying thrombus formation. J. Thromb. Haemost. 2003;1(7):1602–1612. doi: 10.1046/j.1538-7836.2003.00267.x. [DOI] [PubMed] [Google Scholar]
  • [31].Fiore S, Ryeom SW, Weller PF, Serhan CN. Lipoxin recognition sites. Specific binding of labeled lipoxin A4 with human neutrophils. J. Biol. Chem. 1992;267(23):16168–16176. [PubMed] [Google Scholar]
  • [32].Romano M, Chen XS, Takahashi Y, Yamamoto S, Funk CD, Serhan CN. Lipoxin synthase activity of human platelet 12-lipoxygenase. Biochem. J. 1993;296(Pt 1):127–133. doi: 10.1042/bj2960127. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [33].Serhan CN, Sheppard KA. Lipoxin formation during human neutrophil-platelet interactions. Evidence for the transformation of leukotriene A4 by platelet 12-lipoxygenase in vitro. J. Clin. Invest. 1990;85(3):772–780. doi: 10.1172/JCI114503. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [34].Weber PC. Fischer, S. Arachidonic acid and eicosapentaenoic acid metabolism in platelets and vessel walls. Med. Biol. 1984;62(2):129. [PubMed] [Google Scholar]
  • [35].Chakrabarti SK, Cole BK, Wen Y, Keller SR, Nadler JL. 12/15-lipoxygenase products induce inflammation and impair insulin signaling in 3T3-L1 adipocytes. Obesity (Silver Spring) 2009;17(9):1657–1663. doi: 10.1038/oby.2009.192. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [36].Kaur G, Jalagadugula G, Mao G, Rao AK. RUNX1/core binding factor A2 regulates platelet 12-lipoxygenase gene (ALOX12): studies in human RUNX1 haplodeficiency. Blood. 2010;115(15):3128–3135. doi: 10.1182/blood-2009-04-214601. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [37].Natarajan R, Yang DC, Lanting L, Nadler JL. Key role of P38 mitogen-activated protein kinase and the lipoxygenase pathway in angiotensin II actions in H295R adrenocortical cells. Endocrine. 2002;18(3):295–301. doi: 10.1385/ENDO:18:3:295. [DOI] [PubMed] [Google Scholar]
  • [38].Haque MS, Arora JK, Dikdan G, Lysz TW, Zelenka PS. The rabbit lens epithelial cell line N/N1003A requires 12-lipoxygenase activity for DNA synthesis in response to EGF. Mol. Vis. 1999;5:8. [PubMed] [Google Scholar]
  • [39].Onoda JM, Kantak SS, Piechocki MP, Awad W, Chea R, Liu B, Honn KV. Inhibition of radiation-enhanced expression of integrin and metastatic potential in B16 melanoma cells by a lipoxygenase inhibitor. Radiat. Res. 1994;140(3):410–418. [PubMed] [Google Scholar]
  • [40].Coffey MJ, Jarvis GE, Gibbins JM, Coles B, Barrett NE, Wylie OR, O’Donnell VB. Platelet 12-lipoxygenase activation via glycoprotein VI: involvement of multiple signaling pathways in agonist control of H(P)ETE synthesis. Circ. Res. 2004;94(12):1598–1605. doi: 10.1161/01.RES.0000132281.78948.65. [DOI] [PubMed] [Google Scholar]
  • [41].Raso E, Dome B, Somlai B, Zacharek A, Hagmann W, Honn KV, Timar J. Molecular identification.; localization and function of platelet-type 12-lipoxygenase in human melanoma progression.; under experimental and clinical conditions. Melanoma Res. 2004;14(4):245–250. doi: 10.1097/01.cmr.0000138479.00897.d8. [DOI] [PubMed] [Google Scholar]
  • [42].Johnson EN, Brass LF, Funk CD. Increased platelet sensitivity to ADP in mice lacking platelet-type 12-lipoxygenase. Proc. Natl. Acad. Sci. USA. 1998;95(6):3100–3105. doi: 10.1073/pnas.95.6.3100. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [43].Ikawa H, Yamamoto K, Takahashi Y, Ueda N, Hayashi Y, Yamamoto S, Ishimura K, Irahara M, Aono T. Arachidonate 12-lipoxygenase in porcine anterior pituitary cells: its localization and possible function in gonadotrophs. J. Endocrinol. 1996;148(1):33–41. doi: 10.1677/joe.0.1480033. [DOI] [PubMed] [Google Scholar]
  • [44].Ottino P, Taheri F, Bazan HE. Growth factor-induced proliferation in corneal epithelial cells is mediated by 12(S)-HETE. Exp. Eye. Res. 2003;76(5):613–622. doi: 10.1016/s0014-4835(03)00003-4. [DOI] [PubMed] [Google Scholar]
  • [45].Kang SW, Adler SG, Nast CC, LaPage J, Gu JL, Nadler JL, Natarajan R. 12-lipoxygenase is increased in glucose-stimulated mesangial cells and in experimental diabetic nephropathy. Kidney Int. 2001;59(4):1354–1362. doi: 10.1046/j.1523-1755.2001.0590041354.x. [DOI] [PubMed] [Google Scholar]
  • [46].Reddy MA, Adler SG, Kim YS, Lanting L, Rossi J, Kang SW, Nadler JL, Shahed A, Natarajan R. Interaction of MAPK and 12-lipoxygenase pathways in growth and matrix protein expression in mesangial cells. Am. J. Physiol. Renal Physiol. 2002;283(5):F985–994. doi: 10.1152/ajprenal.00181.2002. [DOI] [PubMed] [Google Scholar]
  • [47].Xu ZG, Miao LN, Cui YC, Jia Y, Yuan H, Wu M. Angiotensin II type 1 receptor expression is increased via 12-lipoxygenase in high glucose-stimulated glomerular cells and type 2 diabetic glomeruli. Nephrol. Dial. Transplant. 2009;24(6):1744–1752. doi: 10.1093/ndt/gfn703. [DOI] [PubMed] [Google Scholar]
  • [48].Zhou W, Wang XL, Kaduce TL, Spector AA, Lee HC. Impaired arachidonic acid-mediated dilation of small mesenteric arteries in Zucker diabetic fatty rats. Am. J. Physiol. Heart Circ. Physiol. 2005;288(5):H2210–2218. doi: 10.1152/ajpheart.00704.2004. [DOI] [PubMed] [Google Scholar]
  • [49].Tamai K, Dohi T, Ogawa T, Okamoto H, Tsujimoto A. Some properties of gingival 12-lipoxygenase activity in human and dog. Arch. Oral Biol. 1990;35(8):575–581. doi: 10.1016/0003-9969(90)90022-3. [DOI] [PubMed] [Google Scholar]
  • [50].Reddy GR, Ueda N, Suzuki T, Yamamoto S, Ishimura K, Kawada N, Mizoguchi Y. Characterization of arachidonate 12-lipoxygenase found in the liver of mongrel dog and its immunohistochemical localization in neutrophils. Tokushima J. Exp. Med. 1995;42(1-2):27–35. [PubMed] [Google Scholar]
  • [51].Hiraku S, Taniguchi K, Wakitani K, Omawari N, Kira H, Miyamoto T, Okegawa T, Kawasaki A, Ujiie A. Pharmacological studies on the TXA2 synthetase inhibitor (E)-3-[p-(1H-imidazol-1-ylmethyl)phenyl]-2-propenoic acid (OKY-046) Jpn. J. Pharmacol. 1986;41(3):393–401. doi: 10.1254/jjp.41.393. [DOI] [PubMed] [Google Scholar]
  • [52].Dyerberg J, Bang HO. Haemostatic function and platelet polyunsaturated fatty acids in Eskimos. Lancet. 1979;2(8140):433–435. doi: 10.1016/s0140-6736(79)91490-9. [DOI] [PubMed] [Google Scholar]
  • [53].Knauss HJ, Sheffner AL. Effect of unsaturated fatty acid supplements upon mortality and clotting parameters in rats fed thrombogenic diets. J. Nutr. 1967;93(3):393–400. doi: 10.1093/jn/93.3.393. [DOI] [PubMed] [Google Scholar]
  • [54].Tamura Y, Hirai A, Terano T, Takenaga M, Saitoh H, Tahara K, Yoshida S. Clinical and epidemiological studies of eicosapentaenoic acid (EPA) in Japan. Prog. Lipid Res. 1986;25(1-4):461–466. doi: 10.1016/0163-7827(86)90092-5. [DOI] [PubMed] [Google Scholar]
  • [55].Takenaga M, Hirai A, Terano T, Tamura Y, Kitagawa H, Yoshida S. Comparison of the in vitro effect of eicosapentaenoic acid (EPA)-derived lipoxygenase metabolites on human platelet function with those of arachidonic acid. Thromb. Res. 1986;41(3):373–384. doi: 10.1016/0049-3848(86)90248-3. [DOI] [PubMed] [Google Scholar]
  • [56].Lorenz R, Spengler U, Fischer S, Duhm J, Weber PC. Platelet function.; thromboxane formation and blood pressure control during supplementation of the Western diet with cod liver oil. Circulation. 1983;67(3):504–511. doi: 10.1161/01.cir.67.3.504. [DOI] [PubMed] [Google Scholar]
  • [57].Vericel E, Calzada C, Chapuy P, Lagarde M. The influence of low intake of n-3 fatty acids on platelets in elderly people. Atherosclerosis. 1999;147(1):187–192. doi: 10.1016/s0021-9150(99)00171-9. [DOI] [PubMed] [Google Scholar]
  • [58].Vericel E, Polette A, Bacot S, Calzada C, Lagarde M. Pro- and antioxidant activities of docosahexaenoic acid on human blood platelets. J. Thromb. Haemost. 2003;1(3):566–572. doi: 10.1046/j.1538-7836.2003.00076.x. [DOI] [PubMed] [Google Scholar]
  • [59].Siess W, Roth P, Scherer B, Kurzmann I, Bohlig B, Weber PC. Platelet-membrane fatty acids.; platelet aggregation.; and thromboxane formation during a mackerel diet. Lancet. 1980;1(8166):441–444. doi: 10.1016/s0140-6736(80)90995-2. [DOI] [PubMed] [Google Scholar]
  • [60].Larson MK, Shearer GC, Ashmore JH, Anderson-Daniels JM, Graslie EL, Tholen JT, Vogelaar JL, Korth AJ, Nareddy V, Sprehe M, Harris WS. Omega-3 fatty acids modulate collagen signaling in human platelets. Prostaglandins Leukot. Essent. Fatty Acids. 2011;84(3-4):93–98. doi: 10.1016/j.plefa.2010.11.004. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [61].Morita I, Takahashi R, Saito Y, Murota S. Stimulation of eicosapentaenoic acid metabolism in washed human platelets by 12-hydroperoxyeicosatetraenoic acid. J. Biol. Chem. 1983;258(17):10197–10199. [PubMed] [Google Scholar]
  • [62].Calzada C, Vericel E, Mitel B, Coulon L, Lagarde M. 12(S)-Hydroperoxy-eicosatetraenoic acid increases arachidonic acid availability in collagen-primed platelets. J. Lipid Res. 2001;42(9):1467–1473. [PubMed] [Google Scholar]
  • [63].Dadaian M, Westlund P. Eicosanoid metabolism in human platelets is modified by albumin. Adv. Exp. Med. Biol. 1999;469:23–27. doi: 10.1007/978-1-4615-4793-8_4. [DOI] [PubMed] [Google Scholar]
  • [64].Westlund P, Palmblad J, Falck JR, Lumin S. Synthesis, structural identification and biological activity of 11,12-dihydroxyeicosatetraenoic acids formed in human platelets. Biochim. Biophys. Acta. 1991;1081(3):301–307. doi: 10.1016/0005-2760(91)90286-q. [DOI] [PubMed] [Google Scholar]
  • [65].Gomolka B, Siegert E, Blossey K, Schunck WH, Rothe M, Weylandt KH. Analysis of omega-3 and omega-6 fatty acid-derived lipid metabolite formation in human and mouse blood samples. Prostaglandins Other Lipid Mediat. 2011;94(3-4):81–87. doi: 10.1016/j.prostaglandins.2010.12.006. [DOI] [PubMed] [Google Scholar]
  • [66].von Schacky C, Siess W, Fischer S, Weber PC. A comparative study of eicosapentaenoic acid metabolism by human platelets in vivo and in vitro. J. Lipid Res. 1985;26(4):457–464. [PubMed] [Google Scholar]
  • [67].Morita I, Takahashi R, Saito Y, Murota S. Effects of eicosapentaenoic acid on arachidonic acid metabolism in cultured vascular cells and platelets: species difference. Thromb. Res. 1983;31(2):211–217. doi: 10.1016/0049-3848(83)90323-7. [DOI] [PubMed] [Google Scholar]
  • [68].Schwartzman ML, Balazy M, Masferrer J, Abraham NG, McGiff JC, Murphy RC. 12(R)-hydroxyicosatetraenoic acid: a cytochrome-P450-dependent arachidonate metabolite that inhibits Na+, K+-ATPase in the cornea. Proc. Natl. Acad. Sci. USA. 1987;84(22):8125–8129. doi: 10.1073/pnas.84.22.8125. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [69].Capdevila J, Yadagiri P, Manna S, Falck JR. Absolute configuration of the hydroxyeicosatetraenoic acids (HETEs) formed during catalytic oxygenation of arachidonic acid by microsomal cytochrome P-450. Biochem. Biophys. Res. Commun. 1986;141(3):1007–1011. doi: 10.1016/s0006-291x(86)80144-9. [DOI] [PubMed] [Google Scholar]
  • [70].Sekiya F, Takagi J, Usui T, Kawajiri K, Kobayashi Y, Sato F, Saito Y. 12S-hydroxyeicosatetraenoic acid plays a central role in the regulation of platelet activation. Biochem. Biophys. Res. Commun. 1991;179(1):345–351. doi: 10.1016/0006-291x(91)91376-n. [DOI] [PubMed] [Google Scholar]
  • [71].Setty BN, Werner MH, Hannun YA, Stuart MJ. 15-Hydroxyeicosatetraenoic acid-mediated potentiation of thrombin-induced platelet functions occurs via enhanced production of phosphoinositide-derived second messengers--sn-1,2-diacylglycerol and inositol-1,4,5-trisphosphate. Blood. 1992;80(11):2765–2773. [PubMed] [Google Scholar]
  • [72].Siegel MI, McConnell RT, Porter NA, Cuatrecasas P. Arachidonate metabolism via lipoxygenase and 12L-hydroperoxy-5.;8.;10.;14-icosatetraenoic acid peroxidase sensitive to anti-inflammatory drugs. Proc. Natl. Acad. Sci. USA. 1980;77(1):308–312. doi: 10.1073/pnas.77.1.308. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [73].Chang J, Blazek E, Kreft AF, Lewis AJ. Inhibition of platelet and neutrophil phospholipase A2 by hydroxyeicosatetraenoic acids (HETES). A novel pharmacological mechanism for regulating free fatty acid release. Biochem. Pharmacol. 1985;34(9):1571–1575. doi: 10.1016/0006-2952(85)90701-4. [DOI] [PubMed] [Google Scholar]
  • [74].Vanderhoek JY, Bryant RW, Bailey JM. 15-hydroxy-5,8,11,13-eicosatetraenoic acid: A potent and selective inhibitor of platelet lipoxygenase. J. Biol. Chem. 1980;255(13):5996–5998. [PubMed] [Google Scholar]
  • [75].Takenaga M, Kitagawa H, Hirai A, Tamura Y, Yoshida S. Mechanism of anti-platelet aggregating action of dilazep. J. Pharmacobiodyn. 1985;8(2):77–83. doi: 10.1248/bpb1978.8.77. [DOI] [PubMed] [Google Scholar]
  • [76].Srivastava KC. Docosahexaenoic acid (C22:6 omega 3) and linoleic acid are anti-aggregatory.; and alter arachidonic acid metabolism in human platelets. Prostaglandins Leukot. Med. 1985;17(3):319–327. doi: 10.1016/0262-1746(85)90121-0. [DOI] [PubMed] [Google Scholar]
  • [77].Lagarde M, Calzada C, Vericel E. Pathophysiologic role of redox status in blood platelet activation. Influence of docosahexaenoic acid. Lipids. 2003;38(4):465–468. doi: 10.1007/s11745-003-1085-x. [DOI] [PubMed] [Google Scholar]
  • [78].Javouhey-Donzel A, Guenot L, Maupoil V, Rochette L, Rocquelin G. Rat vitamin E status and heart lipid peroxidation: effect of dietary alpha-linolenic acid and marine n-3 fatty acids. Lipids. 1993;28(7):651–655. doi: 10.1007/BF02536061. [DOI] [PubMed] [Google Scholar]
  • [79].Brown JA, Glenn JK, Gold MH. Manganese regulates expression of manganese peroxidase by Phanerochaete chrysosporium. J. Bacteriol. 1990;172(6):3125–3130. doi: 10.1128/jb.172.6.3125-3130.1990. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [80].Kuhn H, Romisch I, Belkner J. The role of lipoxygenase-isoforms in atherogenesis. Mol. Nutr. Food Res. 2005;49(11):1014–1029. doi: 10.1002/mnfr.200500131. [DOI] [PubMed] [Google Scholar]
  • [81].Gonzalez-Nunez D, Claria J, Rivera F, Poch E. Increased levels of 12(S)-HETE in patients with essential hypertension. Hypertension. 2001;37(2):334–338. doi: 10.1161/01.hyp.37.2.334. [DOI] [PubMed] [Google Scholar]
  • [82].Quintana LF, Guzman B, Collado S, Claria J, Poch E. A coding polymorphism in the 12-lipoxygenase gene is associated to essential hypertension and urinary 12(S)-HETE. Kidney Int. 2006;69(3):526–530. doi: 10.1038/sj.ki.5000147. [DOI] [PubMed] [Google Scholar]
  • [83].Bern MM. Platelet functions in diabetes mellitus. Diabetes. 1978;27(3):342–350. doi: 10.2337/diab.27.3.342. [DOI] [PubMed] [Google Scholar]
  • [84].Vericel E, Croset M, Sedivy P, Courpron P, Dechavanne M, Lagarde M. Platelets and aging. I--Aggregation.; arachidonate metabolism and antioxidant status. Thromb. Res. 1988;49(3):331–342. doi: 10.1016/0049-3848(88)90313-1. [DOI] [PubMed] [Google Scholar]
  • [85].Abbate R, Prisco D, Rostagno C, Boddi M, Gensini GF. Age-related changes in the hemostatic system. Int. J. Clin. Lab. Res. 1993;23(1):1–3. doi: 10.1007/BF02592271. [DOI] [PubMed] [Google Scholar]
  • [86].Terres W, Weber K, Kupper W, Bleifeld W. Age, cardiovascular risk factors and coronary heart disease as determinants of platelet function in men. A multivariate approach. Thromb. Res. 1991;62(6):649–661. doi: 10.1016/0049-3848(91)90369-8. [DOI] [PubMed] [Google Scholar]
  • [87].Walton KW. Pathogenetic mechanisms in atherosclerosis. Am. J. Cardiol. 1975;35(4):542–558. doi: 10.1016/0002-9149(75)90839-5. [DOI] [PubMed] [Google Scholar]
  • [88].Lagarde M, Vericel E, Guichardant M, Dechavanne M. Inhibition of thrombin-induced platelet arachidonic acid release by 15-hydroperoxy-arachidonic acid. Biochem. Biophys. Res. C/ommun. 1981;99(4):1398–1402. doi: 10.1016/0006-291x(81)90774-9. [DOI] [PubMed] [Google Scholar]
  • [89].Tohjima T, Honda N, Mochizuki K, Kinoshita J, Watanabe K, Arisaka T, Kawamori R, Nakamura M, Kurahashi Y, Yoshimoto T, Yamamoto S. Decreased activity of arachidonate 12-lipoxygenase in platelets of Japanese patients with non-insulin-dependent diabetes mellitus. Metabolism. 1998;47(3):257–263. doi: 10.1016/s0026-0495(98)90253-0. [DOI] [PubMed] [Google Scholar]
  • [90].Antonipillai I, Nadler J, Vu EJ, Bughi S, Natarajan R, Horton R. A 12-lipoxygenase product.; 12-hydroxyeicosatetraenoic acid.; is increased in diabetics with incipient and early renal disease. J. Clin.Endocrinol. Metab. 1996;81(5):1940–1945. doi: 10.1210/jcem.81.5.8626861. [DOI] [PubMed] [Google Scholar]
  • [91].Cho H, Ueda M, Tamaoka M, Hamaguchi M, Aisaka K, Kiso Y, Inoue T, Ogino R, Tatsuoka T, Ishihara T, Noguchi T, Morita I, Murota S. Novel caffeic acid derivatives: extremely potent inhibitors of 12-lipoxygenase. J. Med. Chem. 1991;34(4):1503–1505. doi: 10.1021/jm00108a039. [DOI] [PubMed] [Google Scholar]
  • [92].Kalvegren H, Andersson J, Grenegard M, Bengtsson T. Platelet activation triggered by Chlamydia pneumoniae is antagonized by 12-lipoxygenase inhibitors but not cyclooxygenase inhibitors. Eur. J. Pharmacol. 2007;566(1-3):20–27. doi: 10.1016/j.ejphar.2007.03.024. [DOI] [PubMed] [Google Scholar]
  • [93].Pergola C, Jazzar B, Rossi A, Buehring U, Luderer S, Dehm F, Northoff H, Sautebin L, Werz O. Cinnamyl-3,4-dihydroxy-{alpha}-cyanocinnamate (CDC) is a potent inhibitor of 5-lipoxygenase. J. Pharmacol. Exp. Ther. 2011;338(1):205–213. doi: 10.1124/jpet.111.180794. [DOI] [PubMed] [Google Scholar]
  • [94].Radmark O, Werz O, Steinhilber D, Samuelsson B. 5-Lipoxygenase: regulation of expression and enzyme activity. Trends Biochem. Sci. 2007;32(7):332–341. doi: 10.1016/j.tibs.2007.06.002. [DOI] [PubMed] [Google Scholar]
  • [95].Deschamps JD, Kenyon VA, Holman TR. Baicalein is a potent in vitro inhibitor against both reticulocyte 15-human and platelet 12-human lipoxygenases. Bioorg. Med. Chem. 2006;14(12):4295–4301. doi: 10.1016/j.bmc.2006.01.057. [DOI] [PubMed] [Google Scholar]
  • [96].Sekiya K, Okuda H. Selective inhibition of platelet lipoxygenase by baicalein. Biochem. Biophys. Res. Commun. 1982;105(3):1090–1095. doi: 10.1016/0006-291x(82)91081-6. [DOI] [PubMed] [Google Scholar]
  • [97].Choi HJ, Song BJ, Gong YD, Gwak WJ, Soh Y. Rapid degradation of hypoxia-inducible factor-1alpha by KRH102053.; a new activator of prolyl hydroxylase 2. Br. J. Pharmacol. 2008;154(1):114–125. doi: 10.1038/bjp.2008.70. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [98].Nakahata N, Tsuchiya C, Nakatani K, Ohizumi Y, Ohkubo S. Baicalein inhibits Raf-1-mediated phosphorylation of MEK-1 in C6 rat glioma cells. Eur. J. Pharmacol. 2003;461(1):1–7. doi: 10.1016/s0014-2999(02)02950-3. [DOI] [PubMed] [Google Scholar]
  • [99].Kong D, Yamazaki K, Yamori T. Discovery of phosphatidylinositol 3-kinase inhibitory compounds from the Screening Committee of Anticancer Drugs (SCADS) library. Biol. Pharm. Bull. 2010;33(9):1600–1604. doi: 10.1248/bpb.33.1600. [DOI] [PubMed] [Google Scholar]
  • [100].Rhee BG, Hall ER, McIntire LV. Platelet modulation of polymorphonuclear leukocyte shear induced aggregation. Blood. 1986;67(1):240–246. [PubMed] [Google Scholar]
  • [101].Zavodovskaya M, Campbell MJ, Maddux BA, Shiry L, Allan G, Hodges L, Kushner P, Kerner JA, Youngren JF, Goldfine ID. Nordihydroguaiaretic acid (NDGA), an inhibitor of the HER2 and IGF-1 receptor tyrosine kinases, blocks the growth of HER2-overexpressing human breast cancer cells. J. Cell. Biochem. 2008;103(2):624–635. doi: 10.1002/jcb.21435. [DOI] [PubMed] [Google Scholar]
  • [102].Gonzales M, Bowden GT. Nordihydroguaiaretic acid-mediated inhibition of ultraviolet B-induced activator protein-1 activation in human keratinocytes. Mol. Carcinog. 2002;34(2):102–111. doi: 10.1002/mc.10052. [DOI] [PubMed] [Google Scholar]
  • [103].Lanni C, Becker EL. Inhibition of neutrophil phospholipase A2 by p-bromophenylacyl bromide.; nordihydroguaiaretic acid, 5,8,11,14-eicosatetraynoic acid and quercetin. Int. Arch. Allergy Appl. Im/munol. 1985;76(3):214–217. doi: 10.1159/000233694. [DOI] [PubMed] [Google Scholar]
  • [104].Provost P, Merhi Y. BW755C, a dual lipoxygenase/cyclooxygenase inhibitor, reduces mural platelet and neutrophil deposition and vasoconstriction after angioplasty injury in pigs. J. Pharmacol. Exp. Ther. 1996;277(1):17–21. [PubMed] [Google Scholar]
  • [105].Higgs GA, Flower RJ, Vane JR. A new approach to anti-inflammatory drugs. Biochem. Pharmacol. 1979;28(12):1959–1961. doi: 10.1016/0006-2952(79)90651-8. [DOI] [PubMed] [Google Scholar]
  • [106].Mita H, Yui Y, Shida T. Effect of AA-861, a 5-lipoxygenase inhibitor, on leukotriene synthesis in human polymorphonuclear leukocytes and on cyclooxygenase and 12-lipoxygenase activities in human platelets. Allergy. 1986;41(7):493–498. doi: 10.1111/j.1398-9995.1986.tb00334.x. [DOI] [PubMed] [Google Scholar]
  • [107].Nakadate T, Yamamoto S, Aizu E, Kato R. Inhibition of mouse epidermal 12-lipoxygenase by 2,3,4-trimethyl-6-(12-hydroxy-5,10-dodecadiynyl)-1,4-benzoquinon e (AA861) J. Pharm. Pharmacol. 1985;37(1):71–73. doi: 10.1111/j.2042-7158.1985.tb04938.x. [DOI] [PubMed] [Google Scholar]
  • [108].Yoshimoto T, Yokoyama C, Ochi K, Yamamoto S, Maki Y, Ashida Y, Terao S, Shiraishi M. 2,3,5-Trimethyl-6-(12-hydroxy-5,10-dodecadiynyl)-1,4-benzoquinone (AA861), a selective inhibitor of the 5-lipoxygenase reaction and the biosynthesis of slow-reacting substance of anaphylaxis. Biochim. Biophys. Acta. 1982;713(2):470–473. [PubMed] [Google Scholar]
  • [109].Conrad M, Sandin A, Forster H, Seiler A, Frijhoff J, Dagnell M, Bornkamm GW, Radmark O, van Huijsduijnen R. Hooft, Aspenstrom P, Bohmer F, Ostman A. 12/15-lipoxygenase-derived lipid peroxides control receptor tyrosine kinase signaling through oxidation of protein tyrosine phosphatases. Proc. Natl. Acad. Sci. USA. 2010;107(36):15774–15779. doi: 10.1073/pnas.1007909107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [110].Ahnfelt-Ronne I, Arrigoni-Martelli E. A new antiinflammatory compound.; timegadine (N-cyclohexyl-N”-4-[2-methylquinolyl]-N’-2-thiazolylguanidine), which inhibits both prostaglandin and 12-hydroxyeicosatetraenoic acid (12-HETE) formation. Biochem. Pharmacol. 1980;29(24):3265–3269. doi: 10.1016/0006-2952(80)90301-9. [DOI] [PubMed] [Google Scholar]
  • [111].Ahnfelt-Ronne I, Arrigoni-Martelli E. Multiple effects of a new anti-inflammatory agent.; timegadine.; on arachidonic acid release and metabolism in neutrophils and platelets. Biochem. Pharmacol. 1982;31(16):2619–2624. doi: 10.1016/0006-2952(82)90708-0. [DOI] [PubMed] [Google Scholar]
  • [112].Bramm E, Binderup L, Arrigoni-Martelli E. An unusual profile of activity of a new basic anti-inflammatory drug.; timegadine. Agents Actions. 1981;11(4):402–409. doi: 10.1007/BF01982478. [DOI] [PubMed] [Google Scholar]
  • [113].Sun FF, McGuire JC, Morton DR, Pike JE, Sprecher H, Kunau WH. Inhibition of platelet arachidonic acid 12-lipoxygenase by acetylenic acid compounds. Prostaglandins. 1981;21(2):333–343. doi: 10.1016/0090-6980(81)90151-9. [DOI] [PubMed] [Google Scholar]
  • [114].Salari H, Braquet P, Borgeat P. Comparative effects of indomethacin.; acetylenic acids.; 15-HETE.; nordihydroguaiaretic acid and BW755C on the metabolism of arachidonic acid in human leukocytes and platelets. Prostaglandins Leukot. Med. 1984;13(1):53–60. doi: 10.1016/0262-1746(84)90102-1. [DOI] [PubMed] [Google Scholar]
  • [115].Schafer AI, Turner NA, Handin RI. Platelet lipoxygenase-dependent oxygen burst. Evidence for differential activation of lipoxygenase in intact and disrupted human platelets. Biochim. Biophys. Acta. 1982;712(3):535–541. doi: 10.1016/0005-2760(82)90281-8. [DOI] [PubMed] [Google Scholar]
  • [116].Wilhelm TE, Sankarappa SK, VanRollins M, Sprecher H. Selective inhibitors of platelet lipoxygenase: 4,7,10,13-eicosatetraynoic acid and 5,8,11,14-henicosatetraynoic acid. Prostaglandins. 1981;21(2):323–332. doi: 10.1016/0090-6980(81)90150-7. [DOI] [PubMed] [Google Scholar]
  • [117].Ekokoski E, Tornquist K. Effects of 5,8,11,14-eicosatetraynoic acid on thapsigargin-induced calcium entry.; and intracellular pH in thyroid FRTL-5 cells. Biochim. Biophys. Acta. 1994;1223(2):274–278. doi: 10.1016/0167-4889(94)90237-2. [DOI] [PubMed] [Google Scholar]
  • [118].Nakamura MT, Cho HP, Clarke SD. Regulation of hepatic delta-6 desaturase expression and its role in the polyunsaturated fatty acid inhibition of fatty acid synthase gene expression in mice. J. Nutr. 2000;130(6):1561–1565. doi: 10.1093/jn/130.6.1561. [DOI] [PubMed] [Google Scholar]
  • [119].Sekiya K, Okuda H, Arichi S. Selective inhibition of platelet lipoxygenase by esculetin. Biochim. Biophys. Acta. 1982;713(1):68–72. [PubMed] [Google Scholar]
  • [120].Noguchi M, Earashi M, Minami M, Miyazaki I, Tanaka M, Sasaki T. Effects of piroxicam and esculetin on the MDA-MB-231 human breast cancer cell line. Prostaglandins Leukot. Essent. Fatty Acids. 1995;53(5):325–329. doi: 10.1016/0952-3278(95)90051-9. [DOI] [PubMed] [Google Scholar]
  • [121].Pidgeon GP, Kandouz M, Meram A, Honn KV. Mechanisms controlling cell cycle arrest and induction of apoptosis after 12-lipoxygenase inhibition in prostate cancer cells. Cancer Res. 2002;62(9):2721–2727. [PubMed] [Google Scholar]
  • [122].Alanko J, Kurahashi Y, Yoshimoto T, Yamamoto S, Baba K. Panaxynol, a polyacetylene compound isolated from oriental medicines.; inhibits mammalian lipoxygenases. Biochem. Pharmacol. 1994;48(10):1979–1981. doi: 10.1016/0006-2952(94)90598-3. [DOI] [PubMed] [Google Scholar]
  • [123].Kwon BM, Ro SH, Kim MK, Nam JY, Jung HJ, Lee IR, Kim YK, Bok SH. Polyacetylene analogs.; isolated from hairy roots of Panax ginseng.; inhibit Acyl-CoA : cholesterol acyltransferase. Planta. Med. 1997;63(6):552–553. doi: 10.1055/s-2006-957763. [DOI] [PubMed] [Google Scholar]
  • [124].Fujimoto Y, Sakuma S, Komatsu S, Sato D, Nishida H, Xiao YQ, Baba K, Fujita T. Inhibition of 15-hydroxyprostaglandin dehydrogenase activity in rabbit gastric antral mucosa by panaxynol isolated from oriental medicines. J. Pharm. Pharmacol. 1998;50(9):1075–1078. doi: 10.1111/j.2042-7158.1998.tb06925.x. [DOI] [PubMed] [Google Scholar]
  • [125].Choi SY, Ahn EM, Song MC, Kim DW, Kang JH, Kwon OS, Kang TC, Baek NI. In vitro GABA-transaminase inhibitory compounds from the root of Angelica dahurica. Phytother. Res. 2005;19(10):839–845. doi: 10.1002/ptr.1424. [DOI] [PubMed] [Google Scholar]
  • [126].Liminga M, Hornsten L, Sprecher HW, Oliw EH. Arachidonate 15-lipoxygenase in human corneal epithelium and 12- and 15-lipoxygenases in bovine corneal epithelium: comparison with other bovine 12-lipoxygenases. Biochim. Biophys. Acta. 1994;1210(3):288–296. doi: 10.1016/0005-2760(94)90231-3. [DOI] [PubMed] [Google Scholar]
  • [127].Vindlacheruvu RR, Rink TJ, Sage SO. Lack of evidence for a role for the lipoxygenase pathway in increases in cytosolic calcium evoked by ADP and arachidonic acid in human platelets. FEBS Lett. 1991;292(1-2):196–200. doi: 10.1016/0014-5793(91)80866-2. [DOI] [PubMed] [Google Scholar]
  • [128].El Azher M. Alaoui, Havet N, Singer M, Dumarey C, Touqui L. Inhibition by unsaturated fatty acids of type II secretory phospholipase A2 synthesis in guinea-pig alveolar macrophages evidence for the eicosanoid-independent pathway. Eur. J. Biochem. 2000;267(12):3633–3639. doi: 10.1046/j.1432-1327.2000.01392.x. [DOI] [PubMed] [Google Scholar]
  • [129].Suzuki H, Ueda T, Juranek I, Yamamoto S, Katoh T, Node M, Suzuki T. Hinokitiol, a selective inhibitor of the platelet-type isozyme of arachidonate 12-lipoxygenase. Biochem. Biophys. Res. Commun. 2000;275(3):885–889. doi: 10.1006/bbrc.2000.3390. [DOI] [PubMed] [Google Scholar]
  • [130].Yamamoto S, Katsukawa M, Nakano A, Hiraki E, Nishimura K, Jisaka M, Yokota K, Ueda N. Arachidonate 12-lipoxygenases with reference to their selective inhibitors. Biochem. Biophys. Res. Commun. 2005;338(1):122–127. doi: 10.1016/j.bbrc.2005.08.214. [DOI] [PubMed] [Google Scholar]
  • [131].Lee MJ, Kim JW, Yang EG. Hinokitiol activates the hypoxia-inducible factor (HIF) pathway through inhibition of HIF hydroxylases. Biochem. Biophys. Res. Commun. 2010;396(2):370–375. doi: 10.1016/j.bbrc.2010.04.099. [DOI] [PubMed] [Google Scholar]
  • [132].Morita Y, Matsumura E, Okabe T, Shibata M, Sugiura M, Ohe T, Tsujibo H, Ishida N, Inamori Y. Biological activity of tropolone. Biol. Pharm. Bull. 2003;26(10):1487–1490. doi: 10.1248/bpb.26.1487. [DOI] [PubMed] [Google Scholar]
  • [133].Kitamura S, Iida T, Shirahata K, Kase H. Studies on lipoxygenase inhibitors. I. MY3-469 (3-methoxytropolone).; a potent and selective inhibitor of 12-lipoxygenase.; produced by Streptoverticillium hadanonense KY11449. J. Antibiot. (Tokyo) 1986;39(4):589–593. doi: 10.7164/antibiotics.39.589. [DOI] [PubMed] [Google Scholar]
  • [134].Hamasaki Y, Miyazaki S, Tai HH. Rat basophilic leukemia-1 cell possesses 12-lipoxygenase and 5-lipoxygenase activities which are specifically inhibited by gossypol acetic acid. Arerugi. 1984;33(12):1040–1046. [PubMed] [Google Scholar]
  • [135].Hamasaki Y, Tai HH. Gossypol, a potent inhibitor of arachidonate 5- and 12-lipoxygenases. Biochim. Biophys. Acta. 1985;834(1):37–41. doi: 10.1016/0005-2760(85)90173-0. [DOI] [PubMed] [Google Scholar]
  • [136].Ma X, Lian QQ, Dong Q, Ge RS. Environmental inhibitors of 11beta-hydroxysteroid dehydrogenase type 2. Toxicology. 2011;285(3):83–89. doi: 10.1016/j.tox.2011.04.007. [DOI] [PubMed] [Google Scholar]
  • [137].Wang J, Zhou JY, Zhang L, Wu GS. Involvement of MKP-1 and Bcl-2 in acquired cisplatin resistance in ovarian cancer cells. Cell Cycle. 2009;8(19):3191–3198. doi: 10.4161/cc.8.19.9751. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [138].Morre DM, Morre DJ. Catechin-vanilloid synergies with potential clinical applications in cancer. Rejuvenation Res. 2006;9(1):45–55. doi: 10.1089/rej.2006.9.45. [DOI] [PubMed] [Google Scholar]
  • [139].Hope WC, Welton AF, Fiedler-Nagy C, Batula-Bernardo C, Coffey JW. In vitro inhibition of the biosynthesis of slow reacting substance of anaphylaxis (SRS-A) and lipoxygenase activity by quercetin. Biochem. Pharmacol. 1983;32(2):367–371. doi: 10.1016/0006-2952(83)90569-5. [DOI] [PubMed] [Google Scholar]
  • [140].Choi JS, Piao YJ, Kang KW. Effects of quercetin on the bioavailability of doxorubicin in rats: role of CYP3A4 and P-gp inhibition by quercetin. Arch. Pharm. Res. 2011;34(4):607–613. doi: 10.1007/s12272-011-0411-x. [DOI] [PubMed] [Google Scholar]
  • [141].Shirfule AL, Sangamwar AT, Khobragade CN. Exploring glycolate oxidase (GOX) as an antiurolithic drug target: Molecular modeling and in vitro inhibitor study. Int. J. Biol. Macromol. 2011;49(1):62–70. doi: 10.1016/j.ijbiomac.2011.03.016. [DOI] [PubMed] [Google Scholar]
  • [142].Cotrim CA, de Oliveira SC, Filho E.B. Diz, Fonseca FV, Baldissera L, Jr., Antunes E, Ximenes RM, Monteiro HS, Rabello MM, Hernandes MZ, de Oliveira Toyama D, Toyama MH. Quercetin as an inhibitor of snake venom secretory phospholipase A2. Chem. Biol. Interact. 2011;189(1-2):9–16. doi: 10.1016/j.cbi.2010.10.016. [DOI] [PubMed] [Google Scholar]
  • [143].Young PR, Bell RL, Lanni C, Summers JB, Brooks DW, Carter GW. Inhibition of leukotriene biosynthesis in the rat peritoneal cavity. Eur. J. Pharmacol. 1991;205(3):259–266. doi: 10.1016/0014-2999(91)90907-8. [DOI] [PubMed] [Google Scholar]
  • [144].Keshavarzian A, Sedghi S, Kanofsky J, List T, Robinson C, Ibrahim C, Winship D. Excessive production of reactive oxygen metabolites by inflamed colon: analysis by chemiluminescence probe. Gastroenterology. 1992;103(1):177–185. doi: 10.1016/0016-5085(92)91111-g. [DOI] [PubMed] [Google Scholar]
  • [145].Kenyon V, Rai G, Jadhav A, Schultz L, Armstrong M, Jameson JB, Perry S, Joshi N, Bougie JM, Leister W, Taylor-Fishwick DA, Nadler JL, Holinstat M, Simeonov A, Maloney DJ, Holman TR. Discovery of Potent and Selective Inhibitors of Human Platelet-Type 12- Lipoxygenase. J. Med. Chem. 2011 doi: 10.1021/jm2005089. [Epub ahead of print] [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [146].Morita I, Saito Y, Chang WC, Murota S. Effects of purified eicosapentaenoic acid on arachidonic acid metabolism in cultured murine aortic smooth muscle cells.; vessel walls and platelets. Lipids. 1983;18(1):42–49. doi: 10.1007/BF02534689. [DOI] [PubMed] [Google Scholar]
  • [147].Daret D, Blin P, Larrue J. Synthesis of hydroxy fatty acids from linoleic acid by human blood platelets. Prostaglandins. 1989;38(2):203–214. doi: 10.1016/0090-6980(89)90083-x. [DOI] [PubMed] [Google Scholar]
  • [148].You KM, Jong HG, Kim HP. Inhibition of cyclooxygenase/lipoxygenase from human platelets by polyhydroxylated/methoxylated flavonoids isolated from medicinal plants. Arch. Pharm. Res. 1999;22(1):18–24. doi: 10.1007/BF02976430. [DOI] [PubMed] [Google Scholar]
  • [149].Si D, Wang Y, Zhou YH, Guo Y, Wang J, Zhou H, Li ZS, Fawcett JP. Mechanism of CYP2C9 inhibition by flavones and flavonols. Drug Metab. Dispos. 2009;37(3):629–634. doi: 10.1124/dmd.108.023416. [DOI] [PubMed] [Google Scholar]
  • [150].Berger ME, Golub MS, Chang CT, al-Kharouf JA, Nyby MD, Hori M, Brickman AS, Tuck ML. Flavonoid potentiation of contractile responses in rat blood vessels. J. Pharmacol. Exp. Ther. 1992;263(1):78–83. [PubMed] [Google Scholar]
  • [151].Guichardant M, Michel M, Borel C, Fay L, Crozier G, Magnolato D, Finot PA. Effects of 9.;12.;15-octadecatrien-6-ynoic acid on the metabolism of arachidonic acid in platelets and on the platelet aggregation. Agents Actions Suppl. 1992;37:215–221. doi: 10.1007/978-3-0348-7262-1_30. [DOI] [PubMed] [Google Scholar]
  • [152].Wube AA, Bucar F, Asres K, Gibbons S, Adams M, Streit B, Bodensieck A, Bauer R. Knipholone, a selective inhibitor of leukotriene metabolism. Phytomedicine. 2006;13(6):452–456. doi: 10.1016/j.phymed.2005.01.012. [DOI] [PubMed] [Google Scholar]
  • [153].Ozeki Y, Nagamura Y, Ito H, Unemi F, Kimura Y, Igawa T, Kambayashi J, Takahashi Y, Yoshimoto T. An anti-platelet agent.; OPC-29030.; inhibits translocation of 12-lipoxygenase and 12-hydroxyeicosatetraenoic acid production in human platelets. Br. J. Pharmacol. 1999;128(8):1699–1704. doi: 10.1038/sj.bjp.0702976. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [154].Inamori Y, Tsujibo H, Ohishi H, Ishii F, Mizugaki M, Aso H, Ishida N. Cytotoxic effect of hinokitiol and tropolone on the growth of mammalian cells and on blastogenesis of mouse splenic T cells. Biol. Pharm. Bull. 1993;16(5):521–523. doi: 10.1248/bpb.16.521. [DOI] [PubMed] [Google Scholar]
  • [155].Deschamps JD, Gautschi JT, Whitman S, Johnson TA, Gassner NC, Crews P, Holman TR. Discovery of platelet-type 12-human lipoxygenase selective inhibitors by high-throughput screening of structurally diverse libraries. Bioorg. Med. Chem. 2007;15(22):6900–6908. doi: 10.1016/j.bmc.2007.08.015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • [156].A randomised, blinded, trial of clopidogrel versus aspirin in patients at risk of ischaemic events (CAPRIE). CAPRIE Steering Committee. Lancet. 1996;348(9038):1329–1339. doi: 10.1016/s0140-6736(96)09457-3. [DOI] [PubMed] [Google Scholar]
  • [157].Inhibition of platelet glycoprotein IIb/IIIa with eptifibatide in patients with acute coronary syndromes. The PURSUIT Trial Investigators. Platelet Glycoprotein IIb/IIIa in Unstable Angina: Receptor Suppression Using Integrilin Therapy. N. Engl. J. Med. 1998;339(7):436–443. doi: 10.1056/NEJM199808133390704. [DOI] [PubMed] [Google Scholar]
  • [158].Hamberg M, Samuelsson B. Prostaglandin endoperoxides. Novel transformations of arachidonic acid in human platelets. Proc. Natl. Acad. Sci. USA. 1974;71(9):3400–3404. doi: 10.1073/pnas.71.9.3400. [DOI] [PMC free article] [PubMed] [Google Scholar]

RESOURCES