Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2016 Oct 14.
Published in final edited form as: Front Biosci (Landmark Ed). 2016 Jun 1;21:1427–1463. doi: 10.2741/4465

Molecular mechanisms and cell signaling of 20-hydroxyeicosatetraenoic acid in vascular pathophysiology

Fan Fan 1, Ying Ge 1, Wenshan Lv 1,2, Matthew R Elliott 1, Yoshikazu Muroya 1,3, Takashi Hirata 1,4, George W Booz 1, Richard J Roman 1,*
PMCID: PMC5064940  NIHMSID: NIHMS778672  PMID: 27100515

Abstract

Cytochrome P450s enzymes catalyze the metabolism of arachidonic acid to epoxyeicosatrienoic acids (EETs), dihydroxyeicosatetraenoic acid and hydroxyeicosatetraeonic acid (HETEs). 20-HETE is a vasoconstrictor that depolarizes vascular smooth muscle cells by blocking K+ channels. EETs serve as endothelial derived hyperpolarizing factors. Inhibition of the formation of 20-HETE impairs the myogenic response and autoregulation of renal and cerebral blood flow. Changes in the formation of EETs and 20-HETE have been reported in hypertension and drugs that target these pathways alter blood pressure in animal models. Sequence variants in CYP4A11 and CYP4F2 that produce 20-HETE, UDP-glucuronosyl transferase involved in the biotransformation of 20-HETE and soluble epoxide hydrolase that inactivates EETs are associated with hypertension in human studies. 20-HETE contributes to the regulation of vascular hypertrophy, restenosis, angiogenesis and inflammation. It also promotes endothelial dysfunction and contributes to cerebral vasospasm and ischemia-reperfusion injury in the brain, kidney and heart. This review will focus on the role of 20-HETE in vascular dysfunction, inflammation, ischemic and hemorrhagic stroke and cardiac and renal ischemia reperfusion injury.

Keywords: 20-HETE, Cytochrome P450, vascular smooth muscle, endothelial dysfunction, vascular remodeling, vascular inflammation, ischemia reperfusion, stroke

2. INTRODUCTION

Eicosanoids are 20-carbon fatty acids produced from the metabolism of arachidonic acid (AA) via cyclooxygenase (COX), lipoxygenase (LOX) and cytochrome P450 (CYP) monooxygenases (Figure 1). It has long been recognized that COX and LOX metabolize AA to 5-, 12- and 15-hydroxyeicosatetraenoic acid (HETE), leukotrienes, prostaglandins (PG), prostacyclin (PGI2) and thromboxane A2 (TXA2) which are involved in the regulation of fever, inflammation, cancer, vascular tone, central nervous system and airway responses (14). The third pathway for the metabolism of AA by CYP enzymes was initially thought to only be involved in the hepatic metabolism of fatty acids (5, 6). However, as summarized in Figure 1, we now know that AA is metabolized by CYP enzymes in a wide variety of tissues to epoxyeicosatrienoic acids (EETs), dihydroxyeicosatetraenoic acids (DiHETEs) and 20-HETE which contribute to the regulation of renal, cerebral, pulmonary, vascular and cardiac function (718), as well as vascular hypertrophy and inflammation (1924).

Figure 1. Pathways for the metabolism of arachidonic acid.

Figure 1

Arachidonic acid is metabolized by enzymes of the cytochrome P450 (CYP) monooxygenase, cyclooxygenase and lipoxygenase pathways to epoxyeicosatrienoic acid (EETs), 20-hydroxytetraenoic acid (20-HETE), prostacyclin (PGI2), thromboxane A2 (TXA2), Prostaglandins (PGE2 and PGF2a), 5-, 12-, and 15-hydroxyeicosatetraenoic acids (HETEs) and leukotrienes. EETs are metabolized by soluble epoxide hydrolase (SEH) to the corresponding dihydroxyeicosatrienoic acids (DHETEs). 20-HETE is metabolized by cyclooxygenase 2 (COX2) to 20 hydroxy-prostaglandins (20-OH-PGE); conjugated by uridine 5′-diphosphoglucuronosyltransferase (UGT) in the liver to form a glucuronide and metabolized by alcohol dehydrogenase (ADH) to 20-carboxy-hydroxyeicosatetraenoic acid (20-COOH-HETE that is converted to shorter chain dicarboxylic acids by β-oxidation. Leukotrienes are metabolized by CYP4F2 enzyme to a less active 20-hydroxy metabolite (20-OH-LTB4).

Cytochrome P450-dependent monooxygenases are heme-containing enzymes that catalyze NADPH-dependent metabolism of a wide variety of substrates. Enzymes of the CYP2C and 2J families metabolize AA to EETs that are subsequently inactivated by soluble and microsomal epoxide hydroxylases (sEH and mEH) to the corresponding DiHETEs (11, 25, 26). Enzymes of the 4A and 4F families catalyze the ω-hydroxylation of AA to produce 20-HETE (11, 25, 27) (Figure 1). The isoforms that produce 20-HETE in man are CYP4A11, 4A22, 4F2 and 4F3 (2831). CYP4F2 exhibits the highest activity followed by 4A11 (2830). CYP4F2 is expressed in the kidney and CYP4F3 is expressed in polymorphonuclear leukocytes (PMNs) (32). CYP4A1, 4A2, 4A3 and 4A8 are the isoforms that produce 20-HETE in rats. They are expressed in the liver, kidney, brain and blood vessels (27, 3335). Enzymes of the CYP4F family (CYP4F1, 4F4, 4F5, 4F6) are also expressed in rats. (3639) CYP4F5 and 4F6 enzymes catalyze the omega-1 and omega-2 hydroxylation of leukotrienes (40). They produce 17-, 18- and 19-HETE rather than 20-HETE when incubated with AA. CYP4A12a is the only isoform that produces 20-HETE in mice (41, 42). CYP4A10 and 4A14 are also expressed in mice but they do not metabolize AA, rather catalyze the omega-hydroxylation of short chain saturated fatty acids (42). 20-HETE is inactivated via metabolism by COX2 to 20-hydroxy-PGE1 (8, 43, 44) and alcohol dehydrogenase to 20-carboxy-eicosatetraenoic acid (20-COOH-HETE) followed by metabolism by beta-oxidation to 18- and 16-carbon dicarboxylic acids (25, 45). Circulating 20-HETE is conjugated in the liver by uridine 5′-diphosphoglucuronosyltransferase (UGT) to a glucuronide (46, 47) that is filtered and excreted in the urine (48).

The expression of CYP4A protein is greater in arterioles than in large arteries, and the production of 20-HETE is inversely proportional to blood vessel diameter (49). 20-HETE is largely synthesized by vascular smooth muscle cells (VSMC) in arterioles (13, 25, 33, 5052). The exceptions are that it has been reported to be produced in the pulmonary endothelium, (18, 53, 54), in the systemic vascular endothelial cells in animals transduced with a CYP4A2 adenovirus (55) or lentivirus (56), and in dihydrotestosterone induced hypertensive rats (5759). 20-HETE is also produced in the neurons (60) and astrocytes in the brain (61), cardiomyocytes (62, 63), pulmonary endothelium, hepatocytes (16, 64, 65), renal proximal tubular, thick ascending loop of Henle (TALH) cells and the glomerulus in the kidney (6668), and in leukocytes and platelets (14, 69, 70). EETs are produced by endothelial cells in the renal (26), cerebral (13, 71), pulmonary (72, 73) and coronary vascular beds (7, 74, 75), in the proximal tubule and collecting duct of the kidney, astrocytes in the brain (7678), cardiac myocytes (79, 80), enterocytes in the gastrointestinal tract and islets of Langerhans cells in the pancreas (81, 82).

20-HETE plays an important role in regulation of a wide variety of normal physiological functions and has been implicated in the pathogenesis of a variety of disease conditions. A summary of these effects is presented in Figure 2. 20-HETE is a potent vasoconstrictor that potentiates the response to angiotensin II (ANG II), endothelin, and ATP. It promotes vascular hypertrophy, endothelial dysfunction, vascular restenosis, angiogenesis, inflammation and apoptosis. In contrast, EETs are vasodilators that have been identified as endothelial-derived hyperpolarizing factors that mediate the response to acetylcholine and bradykinin in the presence of nitric oxide (NO) synthase and COX inhibitors (7, 25, 26). In blood, 20-HETE inhibits platelet aggregation (70). In the lung, 20-HETE contributes to the regulation of airway resistance and pulmonary vascular tone. 20-HETE has been implicated in vascular remodeling in the placenta and with the development of preeclampsia. In the renal vasculature, 20-HETE plays a critical role in the myogenic response of the afferent arteriole (Af-Art), autoregulation of renal blood flow (RBF) and tubuloglomerular feedback (TGF) responses. At the level of the renal tubules, it inhibits Na+, K+-ATPase (83) and sodium transport in the proximal tubule and TALH (11, 25, 84, 85). EETs are also produced in the renal proximal tubule, they inhibit sodium transport and modulate the activity of the epithelial sodium channel in the collecting duct (25, 26). Changes in the renal production of 20-HETE have been implicated in the development of acute and chronic kidney disease (AKD and CKD), polycystic kidney disease (PKD) (8688) and hypertension. Changes in the renal and/or vascular formation of EETs and 20-HETE have been reported in hypertension and drugs that target these pathways have altered blood pressure in various animal models (27, 41, 8997). In the heart, 20-HETE plays an important role in ischemic injury and cardiac hypertrophy. It is also produced in the cerebral vasculature, glial cells and neurons, and is associated with ischemic and hemorrhagic stroke and brain injury. Recent human studies have indicated that sequence variants in CYP4A11 (28, 31, 98110) and CYP4F2 (99, 110115) that produce 20-HETE as well as UGT that is involved in the biotransformation of 20-HETE (46), are all associated with hypertension and/or stroke.

Figure 2. Diverse physiological and pathological effects of 20-HETE.

Figure 2

20-HETE is a potent vasoconstrictor in the renal and cerebral circulation that potentiates the vasoconstrictor response to Ang II, endothelin and other Gq dependent vasoactive agents. It promotes vascular hypertrophy, endothelial dysfunction, vascular restenosis, angiogenesis and vascular inflammation. In blood, 20-HETE inhibits platelet aggregation. In the lung, 20-HETE is an endothelial dependent dilator that reduces pulmonary vascular resistance and lowers airway resistance. 20-HETE plays an important role in the myogenic and tubuloglomerular feedback responses of the renal afferent arteriole. It also inhibits sodium transport in the proximal tubule and thick ascending loop of Henle. Deficiencies in the renal formation of 20-HETE are associated with the development of salt-sensitive forms of hypertension and renal ischemia reperfusion injury. Overproduction of 20-HETE is linked to the development of polycystic kidney disease. 20-HETE plays an important role in the regulation of cerebral vascular tone and elevations in the production of 20-HETE is associated with cerebral vasospasm following subarachnoid hemorrhage and infarct size following ischemic injury. In the heart, elevations in 20-HETE promote the development of cardiac hypertrophy and infarct size following cardiac ischemia.

There has been considerable interest in the role of 20-HETE in the regulation of vascular tone, renal function and blood pressure (25, 8385, 116). This has been the subject of several recent and definitive reviews (11, 26, 117121). The role of 20-HETE in ischemic and hemorrhagic stroke (122125), acute renal failure (126, 127), toxemia of pregnancy (128), hepatorenal syndrome (129), vascular restenosis (23), angiogenesis (24), cardiac hypertrophy (130) and ischemia reperfusion (IR) injury (131), as well as shock (132, 133) is less well known and has received little attention in the literature. This review will focus on the emerging pathogenic role of 20-HETE as an important mediator in vascular dysfunction, inflammation in a variety of diseases.

3. 20-HETE IN THE CONTROL OF VASCULAR FUNCTION

3.1. 20-HETE and the regulation of vascular smooth muscle tone

Cytochrome P450 eicosanoids play an important role in the regulation of vascular function (11, 25). 20-HETE is produced by VSMC in the renal and cerebral microcirculation (11, 25, 26). As summarized in Figure 3, 20-HETE largely serves as an autocrine second messenger similar to diacylglycerol (DAG). There is some evidence that 20-HETE can be produced in astrocytes (61, 134). In the vascular endothelium, 20-HETE can be released and alters vascular function as a paracrine factor (56). 20-HETE increases vascular tone by activating protein kinase C (PKC), mitogen-activated protein kinases (MAPK), tyrosine kinase and Rho kinase which promotes Ca2+ influx by depolarizing the cell secondary to blockade of the large conductance calcium sensitive K+ (BK) channel (13, 25, 52, 119, 135). Activation of PKC also increases the conductance of L-type Ca2+ channels (136) and activates the transient receptor potential canonical 6 (TRPC6) and transient receptor potential vanilloid 1 (TRPV1) channels (137139).

Figure 3. Mechanisms contributing to the vasoconstrictor action of 20-HETE.

Figure 3

Increases in vascular stretch or administration of Gq coupled vasoconstrictor agonists activate phospholipase A (PLA) secondary to inositol triphosphate (IP3) mediated release of intracellular Ca2+. PLA2 catalyzes the releases of arachidonic acid from membrane phospholipids and stimulates the formation of 20-hydroxyeicosatetraenoic acid (20-HETE). 20-HETE activates phosphokinase C to inhibit activity of the large conductance, calcium activated potassium channel (KCa) to depolarize the cell and increases the activities of the L-type calcium and TRPC6 channels to promote Ca2+ entry.

Elevations in transmural pressure increase the production of 20-HETE in cerebral arteries (140). Inhibition of the synthesis of 20-HETE impair the myogenic response of renal and cerebral arteries in vitro (11, 25, 140142) and autoregulation of renal and cerebral blood flow (CBF) in vivo (11, 140, 143). 20-HETE also modulates TGF response mediated constriction of the renal Af-art (141, 144, 145) by potentiating the vasoconstrictor response of adenosine released by the macula densa (141). The formation of 20-HETE in the renal and cerebral vasculature is reduced in Dahl salt-sensitive rats (Dahl SS). These rats exhibit impaired myogenic responses in the renal Af-art and middle cerebral artery (MCA) and autoregulation of RBF and CBF (27, 33, 142, 144, 146). Transfer of the CYP4A genes responsible for the formation of 20-HETE from Brown Norway (BN) to the Dahl SS rat in a chromosome 5 consomic strain or upregulation of the expression of CYP4A1 in a transgenic Dahl SS rat restores the production of 20-HETE. This transfer rescues the myogenic response of the Af-art and autoregulation of RBF and CBF and protects the kidney and brain from hypertension induced injury (27, 33, 144, 146, 147).

In contrast to the vasoconstrictor actions of 20-HETE seen in most vascular beds, 20-HETE dilates bronchiole smooth muscle and the pulmonary artery of humans and experimental animals (18, 53, 148, 149). CYP4A protein is expressed in both VSMC and the endothelium in the pulmonary artery (149). The vasodilator response to 20-HETE in rat pulmonary arteries is mediated by the release of NO and/or carbon monoxide (CO) by the endothelium secondly to an increase in intracellular calcium and activation of eNOS (54, 148). 20-HETE has also been reported to relax human pulmonary arteries and bovine coronary arteries pre-contracted with a thromboxane-mimetic by stimulating the synthesis and release of prostacyclin (53, 150).

The second messenger systems in VSMC activated by 20-HETE resembles the responses produced by ANG II and other Gq-protein coupled vasoconstrictors, and by PGE2, PGF2a and thromboxane acting on their respective receptors to promote vasoconstriction. Thus, there is considerable interest in isolating a 20-HETE receptor and targeting it for drug development. The strongest evidence favoring the existence of a membrane bound 20-HETE receptor is that structure activity studies have indicated that 19-, 15- and other HETEs and several 20-HETE analogs are competitive antagonists of its vasoconstrictor response (151, 152). The existence of inhibitory analogs indicates that there is likely a specific binding site that mediates the action of 20-HETE, but it remains unclear whether 20-HETE binds directly to PKC and other intracellular kinases like DAG or it acts on a membrane bound G protein, MAPK or tyrosine kinase receptor. The search for the receptor has also been made more difficult since 20-HETE is more lipid soluble than other lipid mediators and is avidly taken up by cells, esterified into membrane phospholipids and is highly protein bound. Some of these effects are inhibited by other fatty acids and structural analogs making it difficult to assess specific binding. As such, a membrane bound receptor has yet to be identified, but there are several groups working in this area and a receptor is expected to be identified and isolated in the next few years.

Several compounds have been developed for studying the role of 20-HETE. Originally, 17-octadecynoic acid (17-ODYA) was thought to be a specific suicide substrate inhibitor that blocks the formation of 20-HETE (153), but this compound also inhibits the formation of EETs (154). Dibromo-dodecenyl-methylsulfimide (DDMS) is a more selective inhibitor (155, 156). N-hydroxy-N′-(4-butyl-2 methylphenyl) formamidine (HET0016), and N-(3-chloro-4-morpholin-4-yl) phenyl-N′-hydroxyimido formamide (TS-011) are very potent (EC50 <10 nM) competitive inhibitors of the formation of 20-HETE (116, 157159). They are highly selective when given at concentrations of 0.1 and 1 micromolar, but they will inhibit the formation of EETs at concentrations of 10 micromolar or higher. HET0016 and TS011 can be used to block the formation of 20-HETE in vivo. The effective dose is 1 mg/kg and it can be given i.v., i.m. or s.c. Chronically, these inhibitors are difficult to use as they have a short half-life (<1 hr) and have to be continuously infused or given at high dose (10 mg/kg) at least twice a day (160, 161). Since they are competitive inhibitors, the tissue concentrations of the drugs fall during homogenization; it is difficult to demonstrate that they inhibit 20-HETE production. However, it is possible to show that they reduce tissue concentrations of 20-HETE. 20-HETE analogs, 20-hydroxyeicosa-6(Z), 15(Z)-dienoic acid (6, 15–20-HEDE, WIT002) and N-(20-hydroxyeicosa-6(Z), 15(Z)-dienoyl) glycine (6, 15–20-HEDGE) have been found to antagonize the vasoconstrictor actions of 20-HETE (151, 152). The effective concentrations of these drugs in vitro are 1–10 micromolar. The half-life of 20-HETE agonists and antagonists are also quite short and they have to be given several times a day at high dose (10 mg/kg) when used in vivo (127). More recently, Pandey et al. (162) have described a new water soluble antagonist of 20-HETE, i.e. 2,5,8,11,14,17- hexaoxanonadecan-19-yl-20-hydroxyeicosa 6(Z), 15(Z)-dienoate (20-SOLA). This compound lowered blood pressure when given in the drinking water to a 20-HETE dependent hypertensive mouse model (162, 163). Giving 20-SOLA throughout the day via the drinking water overcomes the problems associated with the need to infuse the 20-HETE antagonists. 20-SOLA may emerge as the preferred method to block the 20-HETE pathway.

3.2. 20-HETE and endothelial function

Endothelial cells play an essential role opposing platelet and white blood cell adhesion, and modulating vascular tone through the release of NO, PGs, EETs and CO. Endothelial dysfunction is associated with aging and numerous disease states, including hypertension, arteriolosclerosis, hyperlipidemia and diabetes (164). The hallmark of endothelial dysfunction is deficiency in the bioavailability of NO due to impaired NO production by the endothelium, and/or increased inactivation of NO by locally generated reactive oxygen species (ROS).

20-HETE produces endothelial dysfunction via several mechanisms as summarized in Figure 4. 20-HETE stimulates the formation of superoxide in endothelial cells by activation of extracellular signal-regulated kinases 1/2 (ERK1/2)/MAPK pathways that stimulates nicotinamide adenine dinucleotide phosphate (NADPH) oxidase activity. It also uncouples eNOS activity to lower the production of NO and increase the formation of superoxide (165, 166). The decrease in the bioavailability of NO impairs the response to acetylcholine (ACH) and other endothelial dependent dilators.

Figure 4. 20-HETE and endothelial dysfunction.

Figure 4

Upregulation of the formation of 20-HETE in endothelial cells increases the formation of superoxide (O2) by stimulating NADPH oxidase activity. It also uncouples endothelial nitric oxide synthase (eNOS) to inhibit the formation of nitric oxide (NO) and increase the formation of superoxide. The rise in superoxide levels reacts with NO to form peroxynitrite and lower the bioavailability of NO and that promotes endothelial dysfunction.

The production of 20-HETE is elevated in the vasculature of hypertensive animal models with endothelial dysfunction. These include: rats treated with an adenovirus to upregulate the expression of CYP4A2 in the endothelium (55); spontaneously hypertensive rats (SHR) (96, 167169); androgen-induced hypertensive rats (57, 58); rats treated with the immunosuppressant drug cyclosporine A which induces CYP4A expression (170); and CYP4A12 transgenic and CYP4A14 KO mice in which the production of 20-HETE is elevated (41, 171, 172). In androgen-induced hypertensive rats, the expression of the gp-91 and gp-47 phox subunits of NADPH oxidase and the production of ROS are elevated (57). Administration of inhibitor of IKB kinase prevented the increase in nuclear factor-kappa beta (NF-KB) expression and ROS production in the vasculature and impaired the vasodilator response to ACH following administration of 20-HETE (58). In SHR rats, administration of the inhibitor of the synthesis of 20-HETE, HET0016, attenuated the elevated levels of superoxide and restored the vasodilator response to ACH in the middle cerebral artery (167, 168). 20-HETE has also been reported to promote endothelial dysfunction in cultured human endothelial cells by stimulating NF-KB and the production of inflammatory cytokines (173). It plays a role in the development of atherosclerotic plaques in vessels (174) and hypertension (175, 176), both of which are associated with increased oxidative stress. More recently, Wang et al. (177) reported that 20-HETE may trigger thrombosis by increasing adhesion of platelets and the secretion of von Willebrand factor (vWF) in endothelial cells by activating ERK and calcium influx (177).

3.3. 20-HETE and vascular remodeling

Vascular remodeling refers to the process of altering the structure and organization of blood vessels through cell proliferation, migration, and alterations in the production of extracellular matrix (ECM). It involves activation of matrix metalloproteinases (MMPs), cytokines and inflammatory mediators (178, 179). This process involves all vascular cell types including endothelial, fibroblasts and VSMC, as well as changes in the ECM and basement membrane (180). Vascular remodeling is essential to angiogenesis and the development of new mature vessels (181), but it also is an adaptive process that responds to changes in hemodynamic and/or humoral factors and the pathophysiology of a variety of vascular disorders (180).

20-HETE has been reported to contribute to the migration and proliferation of VSMC and endothelial cells induced by ANG II (20, 182), epidermal growth factor (EGF) (22), vascular endothelial growth factor (VEGF) (24), and platelet-derived growth factor (PDGF) (183). It stimulates the secretion of the proangiogenic molecules hypoxia-inducible factor-1 alpha (HIF-1alpha), VEGF and stromal cell-derived factor-1alpha (SDF-1alpha), as well as increases the expression of the chemokine receptor type 4 (CXCR4) to promote migration to sites of neovascularization (24, 184187). 20-HETE also uncouples eNOS to reduce the formation of NO and increase the production of superoxide in endothelial cells (165, 166).

Other studies indicate that 20-HETE enhances the apoptotic processes involved in vascular remodeling. 20-HETE inhibits the migration and invasion of human villous trophoblasts and uterine vascular smooth muscle cells (188) indicating that 20-HETE may alter uterine spiral artery remodeling and the development of preeclampsia. In the lung, 20-HETE inhibits the apoptotic response of pulmonary artery smooth muscle cells induced by the activation of the PI3K/Akt pathway by increasing the generation of ROS (189).

Vascular remodeling is a hallmark of hypertension and is associated with an increase in the media-to-lumen ratio of small arteries, which enhances vascular reactivity and peripheral resistance (179). Baumbach et al. (190) first coined the term “remodeling” in 1989 to describe the thickening of the vessel wall and the increase in the media-to-lumen ratio of pial arterioles in stroke-prone spontaneously hypertensive rats. The production of 20-HETE is elevated in the kidney (191, 192), and 20-HETE contributes to increased renal, cerebral and skeletal muscle vascular reactivity in SHR (167, 168, 193, 194). They exhibit profound vascular remodeling leading to the suggestion that elevated 20-HETE levels mediate hypertension induced vascular hypertrophy. In this regard, 5 alpha-dihydrotestosterone has been reported to induce the expression of vascular CYP4A12; increase the synthesis of 20-HETE and is associated with vascular hypertrophy. 20-HETE production has also been reported to contribute to vascular hypertrophy in a CYP4A12 transgenic mouse independent of the rise in arterial pressure (172, 195). In these studies, the rise in systolic blood pressure (SBP) could be prevented by a 20-HETE antagonist, 20-HEDGE, or following blockade of angiotensin I receptors with losartan. However, the vascular remodeling was only prevented by 20-HEDGE (196). These authors concluded that activation of the local vascular renin-angiotensin system (RAS) contributes to 20-HETE mediated hypertension in androgen dependent models of hypertension; however, the vascular remodeling is mediated by elevated levels of 20-HETE rather than locally generated ANG II (196).

3.4. 20-HETE and restenosis

20-HETE increases the migration and proliferation of VSMC. It contributes to the actions of ANG II and a variety of growth factors (2022, 183, 197, 198). Since NO directly binds to enzymes of the CYP4A pathway and inhibits the formation of 20-HETE (155, 199), endothelial damage that reduces the synthesis or bioavailability of NO should increase the production of 20-HETE, which contributes to vascular hypertrophy. This hypothesis is consistent with the results of previous studies indicating that the formation of 20-HETE is elevated in SHR, ANG II and androgen induced hypertensive animals that all exhibit 20-HETE dependent endothelial dysfunction and vascular remodeling (58, 167, 172, 195). Yaghini et al. reported that ANG II and AA increase the expression of CYP4A1 in balloon-injured (BI) rat carotid arteries and enhanced neointimal formation (200). Other investigators have found that the formation of 20-HETE in carotid arteries is elevated following balloon injury; chronic administration of HET0016 or other inhibitors of 20-HETE greatly reduced proliferation and migration of smooth muscle and neointimal formation (23, 200).

3.5. 20-HETE and angiogenesis

Sa et al. (201) provided the first evidence that CYP4A and 20-HETE plays a role in angiogenesis. They reported that fibroblast growth factor 2 (FGF2) activates phospholipase A2 (PLA2) via the p42 MAPK pathway in endothelial cells to increase the release of AA from membrane phospholipids to stimulate CYP4A to produce 20-HETE. Amaral et al. (202) later reported that administration of 20-HETE inhibitor blocked angiogenesis induced by chronic electrical stimulation of skeletal muscle. This was associated with upregulation of the expression of VEGF in the muscle; administration of VEGF neutralizing antibody blocked the rise in vessel density. They suggested that VEGF stimulates angiogenesis secondary to activation of PLA2 to increase release of AA and production of 20-HETE. Jiang et al. (203) reported that smooth muscle specific overexpression of CYP4A1 in VSMCs promotes endothelial sprouting in cultured renal microvessels. This effect was blocked by HET0016 and the effects of HET0016 could be reversed by addition of a 20-HETE agonist. 20-HETE has been reported to increase proliferation and tube formation of human umbilical vein endothelial cells (HUVEC) cells (204). This was associated with stimulation of ROS production, the secretion of HIF-1alpha and activation of the PI3K/AKT pathway (185, 198, 205). In addition, administration of HET0016 has been reported to decrease the expression of a variety of angiogenic growth factors and inhibit growth of triple negative breast cancer tumor in mice (206). In human small cell lung cancer cell line, treatment with the 20-HETE agonist, 20-hydroxyeicosa-5(Z),14(Z)-dienoic acid (WIT003) or upregulation of the expression of CYP4A11, induced expression of VEGF and MMP-9 via PI3K or ERK pathways that significantly increased microvascular density. These effects were attenuated by 20-HETE antagonist, WIT002 or HET0016 (207).

A previous report (Figure 5) demonstrated that 20-HETE induces neovascularization in the rat cornea in vivo; inhibition of the formation of 20-HETE markedly reduced the corneal neovascularization produced by VEGF and FGF (204). These authors also found that inhibition of the formation of 20-HETE blocked neovascularization of the cornea following implantation of glioblastoma cells, suggests that 20-HETE inhibitors might prevent vascularization and proliferation of solid tumors. Subsequent studies indeed confirmed that administration of 20-HETE inhibitor markedly reduced the vascularization and growth of rat 9L and human U251 glioma cells in the brain of nude rats (208, 209). Upregulation of the expression of CYP4A1 in U251 cells enhanced the proliferative phenotype in vitro and the growth of tumors in vivo (210). This sparked considerable interest in the use of 20-HETE inhibitors and antagonists as cancer chemotherapy agents. The findings that the expression of CYP4A/4F enzymes are elevated in human thyroid, ovarian, breast, and colon cancers (211), pancreatic adenocarcinoma (212) further support that 20-HETE may play a role in vascularization and growth of cancerous tumors. More recent studies indicate that inhibition of the synthesis of 20-HETE are effective in reducing the growth of glioblastoma, lung, prostate, renal, breast and colon cancer in experimental animals (206, 213217).

Figure 5. Angiogenic effects of 20-HETE. Panel A and B.

Figure 5

Implantation of a pellet containing angiogenic substances in the rabbit cornea induces growth of blood vessels. Panel C and D indicates that implantation of a 20-HETE mimetic is angiogenic and increases the growth of blood vessels in the cornea of rabbits. Panels E and F indicate that implantation of VEGF, FGF and other growth factors induce angiogenesis; administration of a 20-HETE synthesis inhibitor or a 20-HETE antagonist completely blocks the angiogenic effects of VEGF and other growth factors. Figure redrawn from data presented in Chen et al. (180) with permission.

3.6. 20-HETE and platelet aggregation

Human PMNs express CYP4F3 (32) that produces 20-HETE when incubated with AA (14, 70, 218220). 20-HETE and 16-HETE are released from rabbit PMNs stimulated with a calcium ionophore, thrombin or platelet activating factor (14, 70, 220). 20-HETE inhibits AA- and thromboxane-induced aggregation of human platelets (70). In these studies, 20-HETE was found to inhibit the formation of thromboxane by competing for the metabolism of platelet derived AA by COX. One of the metabolites formed from 20-HETE, 20-hydroxy-thromboxane B2, may serve as a partial agonist and competitive antagonist of the thromboxane receptor. In contrast, Liu et al. (221) recently reported that chronic administration of COX2 inhibitors increased circulating 20-HETE levels in the plasma of mice by 120-fold, this is associated with reduced bleeding time. They further reported that 20-HETE stimulated platelet aggregation in vitro in mice. The reason for the difference in the results between murine and human platelets needs to be further explored. However, Liu et al. (221) concluded that the adverse cardiovascular effects of COX-2 inhibitors may be due to inhibition of the metabolism of 20-HETE and subsequent increase in platelet aggregation leading to increased incidence of stroke and myocardial infarcts.

4. ROLE OF 20-HETE IN VASCULAR INFLAMMATION

Inflammation initially is a protective response in wound healing that involves activation of immune cells, release of cytokines and growth factors, and changes in vascular function and remodeling. The acute inflammatory phase is characterized by vasodilation and increased permeability of the vascular wall. Leukocytes infiltrate the injured site along with plasma contains fibrin and immunoglobulins that promote swelling. Chronic inflammation is characterized by simultaneous destruction and healing of the tissue to form an abscess or scar tissue. A cellular phase sustains the inflammatory response. Cellular-derived mediators are released including: monocyte chemoattractant protein-1 (MCP-1), interleukin 1 (IL-1), IL-6, tumor necrosis factor alpha (TNF-alpha) and interferon gamma (IFN-gamma), adhesion molecules (VCAM-1, E-selectin) and NO, all of which are associated with vascular inflammation and dysfunction.

Eicosanoids play important roles as pro- or anti-inflammatory agents. The roles of LOX or COX-derived eicosanoids in inflammation have been extensively studied (1, 2, 222, 223). Nonsteroidal anti-inflammatory drugs (NSAIDs) exert their anti-inflammatory action by inhibition of COX and the synthesis of prostaglandins and thromboxane. They are widely used to treat inflammation, reduce pain and fever and prevent colorectal cancer. Low dose aspirin is also one of the primary therapies to prevent platelet activation and reduce the risk of heart attack and stroke. Leukotriene inhibitors and receptor antagonists exert their anti-inflammatory effects by blocking the formation and actions of leukotrienes to promote infiltration of leukocytes and inflammation. Much less is known about the role of CYP4A and 4F enzymes in inflammation. LTB4, which plays a key role in recruitment of leukocytes, is metabolized by CYP4F2 enzymes to a less active metabolite, 20-hydroxy-LTB4. This product is further metabolized to 20-carboxy-LTB4 and then degraded by beta-oxidation (40, 224, 225). Thus, inhibition of CYP4F2 may enhance inflammation by increasing LTB4 levels. Moreover, 20-HETE has been reported to play a role in vascular inflammation by regulating the expression and activities of inflammatory mediators, cytokines (63, 173) and adhesion molecules (173, 226) in endothelial cells as well as by altering the levels of NO (54), superoxide and oxidative stress (227).

Induction of sepsis with lipopolysaccharide (LPS) is associated with activation of MEK1/ERK1/2/IKKbeta/IKB-alpha/NF-KB and MyD88/TAK1/IKKbeta/IKB-alpha/NF-KB pathways that increases the levels of inflammatory cytokines (TNF-alpha and IL-8) (133, 228). These changes, along with the associated hypotension and tachycardia, can be prevented by administration of a stable 20-HETE mimetic, 5, 14-HEDGE. The beneficial effects of the 20-HETE agonist in sepsis were reversed by administration of a 20-HETE antagonist (133, 228). Previous studies have shown that NO binds to heme of the CYP4504A enzymes and inhibits the formation of 20-HETE (155, 199, 229231). The protective effect of 5, 14-HEDGE in septic shock is thought to counteract the inhibition of the formation 20-HETE by NO (227). In addition, administration of 20-HETE agonist reduces the upregulation of COX-2 and gp91phox activity suggesting that 20-HETE may have a direct anti-inflammatory effect in septic shock (227). On the other hand, it is possible that this effect maybe simply due to the prevention of hypotension and tissue ischemia normally associated with sepsis.

20-HETE has been reported to promote inflammation by activating ED-1 and monocytes/macrophages induced by acute renal IR injury in uninephrectomized Lewis rats (232). The infiltration of inflammatory cells in the kidney was attenuated by HET0016 and 6, 15-20-HEDE, and enhanced by 5, 14-20-HEDE (232). Interestingly, 20-HETE has the opposite effect and plays a protective role in bilateral renal IR injury in rats (126, 127). An anti-inflammatory effect of 20-HETE was also observed in DOCA-salt hypertensive mice treated with fenofibrate that induces the expression of CYP4A and the production of 20-HETE via stimulation of the peroxisome proliferator-activated receptors (PPAR) -alpha receptor (233).

Toth et al. recently reported that 20-HETE promotes cerebrovascular inflammation in SHR rats by increasing vascular production of ROS and activating NF-KB. Treatment with HET0016 significantly decreased oxidative stress in MCA of SHR; attenuated the activation of vascular NF-KB and the expression of TNFalpha, IL-1beta and IL-6 (167). In addition to increasing cytokine production, 20-HETE stimulates the expression of adhesion molecules in endothelial cells (173, 177, 226). Elevated levels of 20-HETE promote the expression of ICAM-1 and VCAM-1 on B-lymphocytes which interact with receptors on endothelial cells (226, 234). The increased expression of the adhesion molecules enhances the adhesion of macrophages to the vessel wall and triggers a proinflammatory state.

5. 20-HETE IN STROKE AND TRAUMATIC BRAIN INJURY

Each year, 750,000 strokes occur in the United States. There are large numbers of survivors with devastating neurological symptoms that cannot return to work because of physical limitations. The annual cost to treat stroke victims is 51 billion dollars, and rising. Approximately 80% of strokes are ischemic (due to interruption of blood supply) and 20% are hemorrhagic. Recent studies have indicated that 20-HETE contributes to the development of acute and delayed vasospasm, delayed cerebral ischemia and infarct size following ischemic stroke.

5.1. 20-HETE and subarachnoid hemorrhage (SAH)

SAH commonly occurs after rupture of a cerebral aneurysm leading to subarachnoid bleeding and clot formation. It causes acute cerebral ischemia (ACI) lasting several hours due to acute cerebral vasospasm and a rise of cerebral spinal fluid pressure, followed by the later development of delayed vasospasm and cerebral ischemia (DCI) (116). The majority of deaths occurs within the first 2 days of the injury and is associated with extensive ischemic injury to the brain (235237). The later development of DCI is a long-term prognostic factor for outcomes in both SAH and TBI (238240). One-third of patients that develop delayed cerebral vasospasm and DCI die, one-third suffer permanent neurological damage while the remainder recovers (237, 241).

The factors that initiate the acute fall in CBF following SAH remain uncertain. The fall in CBF correlates with the amount of hemoglobin (Hb) in cerebrospinal fluid (CSF) and can be triggered by injection of clotted blood or oxyHb into CSF (242246). There is evidence that acute vasospasm is triggered in part by scavenging of NO (237, 245) by Hb and the release of the vasoconstrictors, serotonin (5-HT), ATP and endothelin (ET) from the clotting blood (237, 242, 245, 247). ET (248, 249), thromboxane (245), isoprostane, (250, 251) and 5-HT (252) levels are elevated in CSF following SAH and the responsiveness of the vasculature to these vasoconstrictors are enhanced (242, 253, 254). There is also release of AA and increased formation of COX and LOX metabolites (237). Acute cerebral vasospasm has been reported to be attenuated by blockade of ET synthesis or the ETA receptor (255257), blockade of the 5-HT1b receptor (252), thromboxane antagonists (258, 259), as well as inhibitors of RAS (260), Rho (261263), MAP kinase (264), PKC (265, 266) and SrC kinase (267).

Delayed vasospasm is associated with activation of PKC (243245, 257, 265, 268), MAP (264), and Rho kinase (263), diminished K+ channel activity and depolarization of VSM cells (269). The vascular responsiveness to ET, 5-HT and other vasoconstrictors is elevated following the development of delayed vasospasm (243, 253, 270) and there is a diminished responsiveness to NO and other endothelial dependent vasodilators (269, 271274). The endothelial dysfunction associated with delayed cerebral vasospasm has been attributed to binding of NO to Hb and increased destruction of NO by superoxide (243, 272274).

The results of previous studies indicate that cerebral arteries produce the potent vasoconstrictor 20-HETE, which has effects on the cerebral vascular tone that mimic those seen following SAH. The level of 20-HETE in CSF was elevated in patients (275278) and experimental animals following SAH (124, 279). Inhibition of the synthesis of 20-HETE prevented the acute fall in CBF following SAH in rats (122) and reversed delayed vasospasm in both dogs and rats (124, 279, 280). These findings demonstrate an important role of 20-HETE in the development of acute and delayed cerebral vasospasm, at least in experimental animal models, and suggest that inhibitors of the synthesis of 20-HETE or 20-HETE antagonists may have therapeutic potential in the treatment of these devastating conditions.

Cortical spreading ischemia (CSI) is another mechanism that contributes to ischemic damage in patients following SAH (281). It is caused by cortical spreading depolarization (CSD), which is a wave of mass neuronal depolarization after SAH that is associated with release of AA, impaired neurovascular coupling and reduced CBF (281, 282). CSD has recently been reported to be associated with an increase in 20-HETE synthesis and reduced CBF in rat model. Administration of 20-HETE inhibitor ameliorated the reduction in CBF, decreased edema and afforded neuroprotection (282, 283).

The proposed mechanism by which 20-HETE contributes to changes in CBF following SAH is presented in Figure 6. Hb that is released following SAH increases the production of 20-HETE in the cerebral vasculature by scavenging NO and CO that chronically inhibit the formation of 20-HETE. AA and vasoconstrictors (ET, ATP and 5-HT) that stimulate the formation of 20-HETE are also released by clotting blood after SAH. 20-HETE plays an important role in the development of acute vasospasm following SAH. CBF, however, rapidly recovers within 12–24 hrs following SAH. This is associated with increased production of NO and CO, both of which inhibit the formation of 20-HETE. However, NO and CO upregulates the expression of CYP4A and 4F enzymes during the recovery phase and this contributes to secondary elevation in the production of 20-HETE and delayed vasospasm after the blood surrounding cerebral arteries is cleared and NO and CO levels return to control.

Figure 6. Role of 20-HETE in hemorrhagic and ischemic stroke.

Figure 6

Subarachnoid hemorrhage (SAH) leads to the release of free hemoglobin (Hb) and serotonin (5-HT) from clotting blood. Serotonin activates phospholipase A2 in the cerebral vasculature to increase the release of arachidonic acid and the formation of 20-hydroxyeicosatetraenoic acid (20-HETE). Free hemoglobin scavenges NO and the fall in NO levels increase the activity of CYP4A enzymes and the formation of 20-HETE. The rise in 20-HETE levels contributes to acute and delayed vasospasm in SAH. 20-HETE levels are also elevated in cerebral tissue, CSF and plasma following ischemic stroke. The increase in 20-HETE levels was initially thought to promote infarct size by reducing blood flow in the penumbral regions, but subsequent work indicates that 20-HETE increases oxidative stress in ischemic neurons and promotes apoptosis and cell death.

5.2. 20-HETE and Traumatic brain injury (TBI)

TBI with intracranial bleeding exhibits many features common with SAH. CBF autoregulation is impaired following TBI (284287) which is associated with CSD in patients and experimental animals (288). TBI patients that exhibit impairments in autoregulation of CBF are more likely to have a worse outcome (289292). Although the mechanisms involved are not yet completely understood, there is some evidence, that 20-HETE may contribute to the impaired autoregulation of CBF after TBI. TBI activates phospholipases, increases the release of fatty acids and AA and the formation of 20-HETE. (282, 293296) There is increased expression of CYP4A and CYP4F in the injured area long term following TBI (225, 297). For example, the expression of CYP4A8 in the hippocampus is increased 6 hours after TBI; the expression of CYP4A1, CYP4F5 and 4F6 remains elevated up to 7 days after TBI (297).

5.3. 20-HETE and ischemic stroke

Approximately 80% of strokes are due to thromboembolic interruption of the blood supply to the brain (298). The most common experimental models involve permanent or transient occlusion of the MCA (MCAo) of rodents (299301). Following occlusion of a cerebral artery, there is depletion of ATP and high-energy phosphates (299, 302, 303), reduced Na+, K+-ATPase activity, cellular acidification (303), accumulation of intracellular Na+ and Ca2+ and loss of intracellular K+ in the ischemic core. With prolonged ischemia (2–6 hrs), there is cell swelling and rupture of intracellular organelles, loss of the integrity of mitochondrial membranes (304), release of proteolytic enzymes, and death of the neurons. This is followed by cellular necrosis and influx of inflammatory cells. Cerebral ischemia is also associated with the release of mediators that augment the degree of ischemic damage. These include: the excitatory amino acids, glycine, glutamate and aspartate (305309), ET, dopamine (307), 5-HT and free fatty acids (310312). The excitatory neurotransmitters contribute to cell death by increasing metabolic rate and oxygen demand, especially in the penumbra after reperfusion (313). The free fatty acids contribute to neuronal damage by serving as substrates for the formation of lipid peroxides that have detrimental effects on cellular metabolism and oxygen utilization (314316). Other fatty acids released during ischemia, such as AA, are converted to HETEs, leukotrienes and eicosanoids (317319) that compromise collateral flow, increase the production of free radicals (208) and recruit leukocytes in the ischemic area. The recruitment of leukocytes and swelling of the brain may also reduce flow to the injured tissue at the margins of the infarct.

There is considerable evidence that 20-HETE plays an important role in determining infarct size following ischemic stroke. The levels of 20-HETE are elevated in cerebral tissue in rat (123, 320) and in the plasma of patients (321) and rats (50) following ischemic stroke. Administration of 20-HETE synthesis inhibitors markedly reduces infarct size and improves neurological outcomes following focal cerebral ischemia in rats (50, 116, 158, 168, 322, 323). The production of 20-HETE is elevated in the cerebral vasculature in SHR stroke-prone rats, and they exhibit an enhanced sensitivity to ischemic stroke (168) along with 20-HETE dependent oxidative stress and endothelial dysfunction in the cerebral circulation. Mutations in CYP4F2 and CYP4A11 have also been linked to an increase in the incidence of ischemic stroke in Chinese, Swedish and Japanese populations (102, 113115). Genetic variants in CYP450 isoforms are also associated with plaque stability in patients with ischemic stroke (125).

The mechanism underlying the neuroprotective effects of blocking 20-HETE synthesis following cerebral ischemia and reperfusion was originally thought to be due to blockade of its vasoconstrictor effects. Thus, blockade of the formation and release of 20-HETE may improve collateral blood flow and oxygen delivery to the penumbra and reduce infarct size. However, subsequent studies indicated that inhibition of 20-HETE reduce infarct size, but had no effect on CBF in the ischemic core or surrounding tissue (324). This leads to speculation that perhaps other mechanisms might be involved. For example, blockade of the synthesis of 20-HETE might reduce the recruitment of PMNs, inflammation and oxidative stress in the at risk tissue or have direct neuroprotective actions. In support of this later possibility, Renic et al. (60) reported that inhibition of 20-HETE markedly reduced necrosis and apoptosis of neonatal brain slices subjected to oxygen and glucose deprivation. The beneficial effect was associated with a reduction in superoxide production and activation of caspase-3. 20-HETE activates a host of cell signaling pathways, such as tyrosine kinase, MAP kinase, PKC, RAS and Rho (20, 325329) involved in apoptosis and cell death. Thus, blockade of the formation of 20-HETE might reduce oxygen consumption or interfere with the signal transduction cascade leading to cell death. Moreover, 20-HETE also inhibits Na+, K+-ATPase (83, 330). It may contribute to the accumulation of Na+, depolarization of neurons that triggers Ca2+ influx, and the release of excitatory amino acids, fatty acids, eicosanoids and other mediators from the brain after reperfusion.

6. ROLE OF 20-HETE IN RENAL AND CARDIAC ISCHEMIA REPERFUSION (IR) INJURY

6.1. 20-HETE and renal ischemia reperfusion injury

Acute kidney injury (AKI) is a common complication following surgery and several medical conditions that significantly increases morbidity and mortality (331, 332). Renal IR injury is the most common cause of AKI (332, 333). AA is released from membrane phospholipids in response to ischemia and can be metabolized to 20-HETE (334). Indeed, we recently reported that 20-HETE levels are markedly increased in the renal outer medulla following IR (126, 127). In the kidney, 20-HETE enhances vascular tone and reduces tubular sodium transport (8, 11, 25, 84, 85). 20-HETE increases vascular responsiveness to vasoconstrictors and would be expected to reduce tissue blood flow and exacerbate IR injury as has been reported both in the heart and the brain (50, 334). Several reports have addressed the role of 20-HETE on renal IR injury. Regner et al. demonstrated that blockade of 20-HETE with HET0016 increased bilateral renal IR injury; administration of 20-HETE agonists (5,14-20-HEDGE and 5,14-20-HEDE) reduced bilateral renal IR injury by preventing the secondary fall in medullary blood flow and prolonged renal ischemia (126, 127). On the other hand, Hoff et al. (232) reported that 20-HETE inhibitor (HET0016), or a 20-HETE antagonist (6, 15-20-HEDE), protected the kidney against renal IR following uninephrectomy. The reasons for the divergent results remain unresolved, but appear due to differences in the experimental models in these two studies since in a follow up study Roman et al. (335) confirmed that blockade of the formation of 20-HETE opposes renal IR injury in bilateral ischemic model, but enhances injury in the uninephrectomized rats. A recent study in Dahl SS rats, which have a genetic deficiency in the expression of CYP4A enzymes and the formation of 20-HETE relative to other strains (27, 336), indicated that they are much more susceptible to renal IR injury (337). Transferring of the CYP4A genes on chromosome 5 from BN rats into the Dahl SS genetic background in a consomic strain normalized the production of 20-HETE and the susceptibility to renal IR injury (126).

20-HETE has differing effects on renal tubular and vascular function, thus the mechanism of its renoprotective effects in IR injury remains to be determined. It activates a number of signaling pathways that could promote or attenuate renal IR injury. In this regard, 20-HETE is a potent constrictor of preglomerular arterioles that may exacerbate reductions in RBF after initial ischemic injury, leading to secondary injury (25). Prolonged exposure to high concentrations of 20-HETE may also potentiate renal IR injury by increasing the generation of reactive oxygen species both by stimulating of NADPH oxidase directly and/or through uncoupling of nitric oxide synthase (338). On the other hand, 20-HETE may oppose renal IR injury since it has been shown to prevent the secondary fall in medullary blood flow and the prolonged medullary ischemia (126, 127). It also inhibits sodium transport and could reduce oxygen consumption in this region of the kidney (8, 25, 84, 85, 126, 339). In addition, 20-HETE and 5,14-20-HEDE activates the Raf–MEK–ERK and phosphatidylinositol-3′-kinase/AKT pathways in renal tubular epithelial cells (340). Activation of the AKT pathway has been reported to enhance renal epithelial-cell survival in models of renal IR injury (341, 342).

6.2. 20-HETE and allograft function in kidney transplantation

IR is an unavoidable process in kidney transplantation. It determines both early and long-term allograft function that affects morbidity and mortality. Previous studies have suggested that vascular dysfunction, tubular injury, and inflammation all contribute to renal IR injury after kidney transplantation (343347). Following cold ischemia, intracellular Ca2+ concentration increases which activates PLA2 to promote the release of AA from membrane phospholipid pools. As discussed earlier, AA can be metabolized into prostaglandins, leukotrienes and 5-, 12-, 15-HETE by COX and LOX, and 20-HETE and EETs via cytochrome P450 pathways (25). Dolegowska et al. (348) reported that during the first 5 minutes following allograft reperfusion, the concentration of 20-HETE in a blood sample is significantly elevated in patients with good graft function (serum creatinine less than 3 mg/dl in 5 days). It was unchanged in patients with delayed graft function (serum creatinine greater than 3 mg/dl); and very low in patients with poor graft function that requires dialysis. Moreover, perioperative 20-HETE levels in plasma are strongly associated with improved graft function following renal transplant (349). These results suggest that 20-HETE is protective in renal transplant or at least may serve as an early clinical biomarker of transplant allograft function.

6.3. 20-HETE and cardiac ischemia reperfusion injury

Recent studies have indicated that 20-HETE may contribute to myocardial injury following IR. Nithipatikom et al. reported that CYP4A and CYP4F enzymes are expressed in the heart (334, 350). The synthesis and release of 20-HETE from dog heart is increased following IR injury. Inhibition of the synthesis of 20-HETE with HET0016 or blockade of the actions of 20-HETE with 5, 14 20-HEDE attenuated cardiac dysfunction and infarct size after IR (334, 350, 351). Moreover, inhibition of the formation of 20-HETE have a beneficial effect on the recovery of cardiac function following IR injury in diabetic rats (62). Conversely, exogenous administration of 20-HETE increased infarct size following IR in the heart (334, 350).

Ischemic preconditioning reduces infarct size following cardiac IR. The magnitude of the effect is similar to that seen following pretreatment of animals with an inhibitor of the synthesis of 20-HETE (300). Recent studies have indicated that ischemic preconditioning is associated with a reduction in the concentration of 20-HETE in coronary venous blood (352). Moreover, blockade of the synthesis of 20-HETE or administration of a 20-HETE antagonist potentiates the cardio protective effects of ischemia preconditioning (352). These findings indicate that 20-HETE plays an important role in the pathogenesis of myocardial injury following IR. They also indicate that inhibitors of 20-HETE or receptor antagonists have therapeutic potential to protect against myocardial injury following IR.

Gross et al. (350) reported that the cardio protective effects of 20-HETE inhibitors is associated with an increase in K+ channel activity that is abolished by administration of the KATP channel blocker HMR-1098. While the mechanism by which 20-HETE contributes to myocardial injury remains to be fully determined, 20-HETE has been shown to increase the formation of ROS and myocardial dysfunction in isolated perfused rat heart following IR injury. The increase in oxidative stress associated with administration of 20-HETE was blocked by the NAPDH oxidase inhibitor apocynin. Moreover, the cardioprotective action of HET0016 is associated with a reduction in oxidative stress following IR (131). These findings indicate that 20-HETE stimulates NADPH oxidase-derived ROS production, which aggravates IR-induced myocardial injury.

Bao et al. (353) reported that 20-HETE induces apoptosis of cultured cardiomyocytes by activation of mitochondria-dependent pathways. However, 20-HETE induced increase in inflammation as discussed earlier may also contribute to the adverse effects of 20-HETE on myocardial injury following IR (116, 173). Several studies have reported that sequence variants in the CYP4A11 gene that produce 20-HETE are linked with the incidence of myocardial infarction (MI) in human population studies (101, 104, 105). A haplotype based case control study that looked at three different SNPs in CYP4A11 gene (rs2269231, rs1126742, and rs9333025) found that the T-T-A haplotype was a protective genetic marker for MI in male Japanese (104).

There is also compelling evidence that 20-HETE may contribute to left ventricular hypertrophy, which is an important risk factor for heart failure, cardiac arrhythmias, myocardial infarction, and sudden cardiac death (44, 121, 153, 156, 354). In both patients and animal models, enzymes of the CYP4A and 4F families are upregulated in association with maladaptive cardiac hypertrophy associated with heart failure (96). For example, the expression of CYP4A11 mRNA is elevated in hypertrophic human hearts (169) and the expression of CYP4A3 and 4F4, and the production of 20-HETE was found to be elevated in isoproterenol-induced cardiac hypertrophy in rats (130, 355). Treatment of SD rats with fenofibrate was found to decrease CYP4A3 cardiac gene expression and protein levels and 20-HETE formation cardiac microsomes (356). Moreover, fenofibrate attenuated isoproterenol-induced cardiac hypertrophy, as determined by the increase in the expression ANP and BNP in the heart and the increase in the heart weight (356). Aryl hydrocarbon receptor (AhR) ligands, that promote cardiac hypertrophy, increased the expression of several CYP omega-hydroxylases, including CYP4F4, and increased 20-HETE production (357, 358). Inhibition of the synthesis of 20-HETE prevented AhR ligand-induced cardiac hypertrophy (358). In another study, 20-HETE levels in the heart were markedly elevated in ANG II-induced cardiac hypertrophy in rats (359). Recently, cardiac hypertrophy in this model was found to be inhibited by the antibiotic isoniazid, a well-known inducer of CYP2E1 which increases the formation of 19-HETE which serves as an endogenous competitive inhibitor of 20-HETE (25, 359). Overall, the results of these studies not only highlight the contribution of 20-HETE to cardiac hypertrophy, but the potential of using inhibitors of this pathway to prevent adverse cardiac remodeling.

The expression of CYP4A and 4F enzymes and 20-HETE production has recently been shown to be elevated in an animal model of heart failure (360). Moreover, the cancer chemotherapy agent doxorubicin (DOX), that produces cardiomyopathy, upregulates the expression of CYP4A1, CYP4A3, CYP4F1 and CYP4F4 in the heart, and enhances the formation of 20-HETE (361). Thus, DOX-induced cardiotoxicity is associated with early disturbances in the formation of 20-HETE that may contribute to the increase in oxidative stress. The cardiotoxic effects may limit the usefulness of this important cancer therapy.

The signaling events linking 20-HETE to cardiac hypertrophy remain to be determined. Alsaad et al. suggested that 20-HETE may involve in activation of Rho kinase, ERK1/2, NF-KB, and NFAT pathways (96)in pathological cardiac hypertrophy as seen in other cell types. In adult rat cardiomyocytes, 20-HETE has been reported to activate PKC, which increases NADPH oxidase and ROS production, and in turn increases the activity of the L-type Ca2+ channel (362). Of note, PKC, NADH oxidase, ROS, and increased intracellular Ca2+ have been linked to pathological cardiac hypertrophy (363365). It remains to be determined whether 20-HETE activates PKC via a receptor or directly interacts with PKC similar to the action of DAG (366). More recently, 20-HETE-mediated mitochondrial ROS production was implicated in ANG II-induced apoptosis of neonatal rat cardiac myocytes (367). Enhanced apoptosis of cardiac myocytes by 20-HETE (353) may have significance for the etiology of heart failure, which is associated with increased apoptosis of cardiac myocytes, as well as other forms of cell death.

7. SUMMARY

There is growing evidence that CYP450 of the 4A and 4F pathways and 20-HETE play an important role in a wide array of vascular dysfunction. The production of 20-HETE is altered in a variety of diseases. It acts as vasoconstrictor in vascular smooth muscle and promotes the myogenic response and autoregulation of renal and cerebral blood flow. 20-HETE also serves as a second messenger in the regulation of endothelial dysfunction, inflammation, angiogenesis, restenosis and IR injury in the brain, heart and kidney. These available data clearly indicate that 20-HETE is a potential novel pharmacological target and biomarker in a variety of diseases associated with vascular dysfunction and hypertrophy, and in ischemia-reperfusion injury in the brain, kidney and heart.

Acknowledgments

This study was supported in part by grants from the National Institutes of Health, HL36279 and DK104184 (Roman), AG050049 and a Pilot grant from P20GM104357 (Fan).

Abbreviations

17-ODYA

17-octadecynoic acid

20-COOH-HETE

20-carboxy-eicosatatraenoic acid

6, 15, 20-HEDE

20-hydroxyeicosa-6(Z), 15(Z)-dienoic acid

6, 15–20-HEDGE

N-(20-hydroxyeicosa-6(Z), 15(Z)-dienoyl) glycine

20-SOLA

2,5,8,11,14,17- hexaoxanonadecan-19-yl-20-hydroxyeicosa 6(Z), 15(Z)-dienoate

AA

arachidonic acid

ACI

acute cerebral ischemia

ACH

acetylcholine

Af-art

afferent arteriole

AKD

acute kidney disease

AKI

acute kidney injury

ANG II

angiotensin II

BI

balloon-injured

BK

the large conductance calcium sensitive potassium channel

BN

Brown Norway

CBF

cerebral blood flow

CKD

chronic kidney disease

CO

carbon monoxide

CSD

cortical spreading depolarization

CSF

cerebrospinal fluid

CSI

cortical spreading ischemia

COX

cyclooxygenase

COX2

cyclooxygenase 2

CXCR4

chemokine Receptor Type 4

CYP

cytochrome P450

Dahl SS

Dahl salt-sensitive

DAG

diacylglycerol

DCI

delayed vasospasm and cerebral ischemia

DDMS

dibromo-dodecenyl-methylsulfimide

DiHETE

dihydroxyeicosatetraenoic acid

DOX

doxorubicin

ECM

extracellular matrix

EETs

epoxyeicosatrienoic acids

EGF

epidermal growth factor

ERK1/2

extracellular signal-regulated kinases ½

ET

endothelin

FGF2

fibroblast growth factor 2

Hb

hemoglobin

HETE

hydroxyeicosatetraeonic acid

HET0016

N-hydroxy-N′-(4-butyl-2 methylphenyl)formamidine

HIF-1 alpha

hypoxia-inducible factor-1 alpha

HUVEC

human umbilical vein endothelial cell

IFN-gamma

interferon gamma

IL-1

interleukin 1

IR

ischemia reperfusion

LOX

lipoxygenase

MAPK

mitogen-activated protein kinases

MCA

middle cerebral artery

MCAo

occlusion of middle cerebral artery

MCP-1

monocyte chemoattractant protein-1

MMPs

metalloproteinases

NADPH

nicotinamide adenine dinucleotide phosphate

NF-KB

nuclear factor-kappa beta

NO

nitric oxide

NSAID

nonsteroidal anti-inflammatory drugs

PDGF

platelet-derived growth factor

PG

prostaglandins

PGI2

prostacyclin

PKC

protein kinase C

PKD

polycystic kidney disease

PLA2

phospholipase A2

PPAR

peroxisome proliferator-activated receptors

RBF

renal blood flow

RAS

renin-angiotensin system

ROS

reactive oxygen species

SAH

subarachnoid hemorrhage

SBP

systolic blood pressure

SDF-1alpha

stromal cell-derived factor-1alpha

SHR

spontaneously hypertensive rat

TALH

thick ascending loop of henle

TGF

tubuloglomerular feedback

TNF alpha

tumor necrosis factor alpha

TRPC6

the transient receptor potential canonical 6 channel

TRPV1

the transient receptor potential vanilloid 1 channel

TS-011

N-(3-chloro-4-morpholin-4-yl) phenyl-N′-hydroxyimido formamide

TXA2

thromboxane A2

UGT

uridine 5′-diohosphoglucuronosyltransferase

VEGF

vascular endothelial growth factor

VSMC

vascular smooth muscle cells

vWF

von Willebrand factor

WIT003

20-hydroxyeicosa-5(Z),14(Z)-dienoic acid

References

  • 1.Claar D, Hartert TV, Peebles RS., Jr The role of prostaglandins in allergic lung inflammation and asthma. Expert Rev Respir Med. 2015;9:55–72. doi: 10.1586/17476348.2015.992783. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2.Shimizu T. Lipid mediators in health and disease: enzymes and receptors as therapeutic targets for the regulation of immunity and inflammation. Annu Rev Pharmacol Toxicol. 2009;49:123–50. doi: 10.1146/annurev.pharmtox.011008.145616. [DOI] [PubMed] [Google Scholar]
  • 3.Harris RC, Zhang MZ. Cyclooxygenase metabolites in the kidney. Compr Physiol. 2011;1:1729–58. doi: 10.1002/cphy.c100077. [DOI] [PubMed] [Google Scholar]
  • 4.Capra V, Back M, Barbieri SS, Camera M, Tremoli E, Rovati GE. Eicosanoids and their drugs in cardiovascular diseases: focus on atherosclerosis and stroke. Med Res Rev. 2013;33:364–438. doi: 10.1002/med.21251. [DOI] [PubMed] [Google Scholar]
  • 5.Morrison AR, Pascoe N. Metabolism of arachidonate through NADPH-dependent oxygenase of renal cortex. Proc Natl Acad Sci U S A. 1981;78:7375–8. doi: 10.1073/pnas.78.12.7375. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6.Oliw EH, Lawson JA, Brash AR, Oates JA. Arachidonic acid metabolism in rabbit renal cortex. Formation of two novel dihydroxyeicosatrienoic acids. J Biol Chem. 1981;256:9924–31. [PubMed] [Google Scholar]
  • 7.Campbell WB, Gebremedhin D, Pratt PF, Harder DR. Identification of epoxyeicosatrienoic acids as endothelium-derived hyperpolarizing factors. Circ Res. 1996;78:415–23. doi: 10.1161/01.res.78.3.415. [DOI] [PubMed] [Google Scholar]
  • 8.McGiff JC, Quilley J. 20-HETE and the kidney: resolution of old problems and new beginnings. Am J Physiol. 1999;277:R607–23. doi: 10.1152/ajpregu.1999.277.3.R607. [DOI] [PubMed] [Google Scholar]
  • 9.Rahman M, Wright JT, Jr, Douglas JG. The role of the cytochrome P450-dependent metabolites of arachidonic acid in blood pressure regulation and renal function: a review. Am J Hypertens. 1997;10:356–65. doi: 10.1016/s0895-7061(96)00381-0. [DOI] [PubMed] [Google Scholar]
  • 10.Roman RJ, Alonso-Galicia M. P-450 Eicosanoids: A Novel Signaling Pathway Regulating Renal Function. News Physiol Sci. 1999;14:238–242. doi: 10.1152/physiologyonline.1999.14.6.238. [DOI] [PubMed] [Google Scholar]
  • 11.Fan F, Muroya Y, Roman RJ. Cytochrome P450 eicosanoids in hypertension and renal disease. Curr Opin Nephrol Hypertens. 2015;24:37–46. doi: 10.1097/MNH.0000000000000088. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Harder DR, Alkayed NJ, Lange AR, Gebremedhin D, Roman RJ. Functional hyperemia in the brain: hypothesis for astrocyte-derived vasodilator metabolites. Stroke. 1998;29:229–34. doi: 10.1161/01.str.29.1.229. [DOI] [PubMed] [Google Scholar]
  • 13.Harder DR, Gebremedhin D, Narayanan J, Jefcoat C, Falck JR, Campbell WB, Roman R. Formation and action of a P-450 4A metabolite of arachidonic acid in cat cerebral microvessels. Am J Physiol. 1994;266:H2098–2107. doi: 10.1152/ajpheart.1994.266.5.H2098. [DOI] [PubMed] [Google Scholar]
  • 14.Hill E, Murphy RC. Quantitation of 20-hydroxy-5,8,11,14-eicosatetraenoic acid (20-HETE) produced by human polymorphonuclear leukocytes using electron capture ionization gas chromatography/mass spectrometry. Biol Mass Spectrom. 1992;21:249–53. doi: 10.1002/bms.1200210505. [DOI] [PubMed] [Google Scholar]
  • 15.Imig JD, Zou AP, Stec DE, Harder DR, Falck JR, Roman RJ. Formation and actions of 20-hydroxyeicosatetraenoic acid in rat renal arterioles. Am J Physiol. 1996;270:R217–27. doi: 10.1152/ajpregu.1996.270.1.R217. [DOI] [PubMed] [Google Scholar]
  • 16.Ito O, Nakamura Y, Tan L, Ishizuka T, Sasaki Y, Minami N, Kanazawa M, Ito S, Sasano H, Kohzuki M. Expression of cytochrome P-450 4 enzymes in the kidney and liver: regulation by PPAR and species-difference between rat and human. Mol Cell Biochem. 2006;284:141–8. doi: 10.1007/s11010-005-9038-x. [DOI] [PubMed] [Google Scholar]
  • 17.Roman RJ, Maier KG, Sun CW, Harder DR, Alonso-Galicia M. Renal and cardiovascular actions of 20-hydroxyeicosatetraenoic acid and epoxyeicosatrienoic acids. Clin Exp Pharmacol Physiol. 2000;27:855–65. doi: 10.1046/j.1440-1681.2000.03349.x. [DOI] [PubMed] [Google Scholar]
  • 18.Zhu D, Effros RM, Harder DR, Roman RJ, Jacobs ER. Tissue sources of cytochrome P450 4A and 20-HETE synthesis in rabbit lungs. Am J Respir Cell Mol Biol. 1998;19:121–8. doi: 10.1165/ajrcmb.19.1.3145. [DOI] [PubMed] [Google Scholar]
  • 19.Node K, Huo Y, Ruan X, Yang B, Spiecker M, Ley K, Zeldin DC, Liao JK. Anti-inflammatory properties of cytochrome P450 epoxygenase-derived eicosanoids. Science. 1999;285:1276–9. doi: 10.1126/science.285.5431.1276. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Muthalif MM, Benter IF, Karzoun N, Fatima S, Harper J, Uddin MR, Malik KU. 20-Hydroxyeicosatetraenoic acid mediates calcium/calmodulin-dependent protein kinase II-induced mitogen-activated protein kinase activation in vascular smooth muscle cells. Proc Natl Acad Sci U S A. 1998;95:12701–6. doi: 10.1073/pnas.95.21.12701. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Uddin MR, Muthalif MM, Karzoun NA, Benter IF, Malik KU. Cytochrome P-450 metabolites mediate norepinephrine-induced mitogenic signaling. Hypertension. 1998;31:242–7. doi: 10.1161/01.hyp.31.1.242. [DOI] [PubMed] [Google Scholar]
  • 22.Lin F, Rios A, Falck JR, Belosludtsev Y, Schwartzman ML. 20-Hydroxyeicosatetraenoic acid is formed in response to EGF and is a mitogen in rat proximal tubule. Am J Physiol. 1995;269:F806–16. doi: 10.1152/ajprenal.1995.269.6.F806. [DOI] [PubMed] [Google Scholar]
  • 23.Orozco LD, Liu H, Perkins E, Johnson DA, Chen BB, Fan F, Baker RC, Roman RJ. 20-Hydroxyeicosatetraenoic acid inhibition attenuates balloon injury-induced neointima formation and vascular remodeling in rat carotid arteries. J Pharmacol Exp Ther. 2013;346:67–74. doi: 10.1124/jpet.113.203844. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 24.Guo AM, Arbab AS, Falck JR, Chen P, Edwards PA, Roman RJ, Scicli AG. Activation of vascular endothelial growth factor through reactive oxygen species mediates 20-hydroxyeicosatetraenoic acid-induced endothelial cell proliferation. J Pharmacol Exp Ther. 2007;321:18–27. doi: 10.1124/jpet.106.115360. [DOI] [PubMed] [Google Scholar]
  • 25.Roman RJ. P-450 metabolites of arachidonic acid in the control of cardiovascular function. Physiol Rev. 2002;82:131–85. doi: 10.1152/physrev.00021.2001. [DOI] [PubMed] [Google Scholar]
  • 26.Imig JD. Epoxyeicosatrienoic acids, 20-hydroxyeicosatetraenoic acid, and renal microvascular function. Prostaglandins Other Lipid Mediat. 2013;104–105:2–7. doi: 10.1016/j.prostaglandins.2013.01.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 27.Williams JM, Fan F, Murphy S, Schreck C, Lazar J, Jacob HJ, Roman RJ. Role of 20-HETE in the antihypertensive effect of transfer of chromosome 5 from Brown Norway to Dahl salt-sensitive rats. Am J Physiol Regul Integr Comp Physiol. 2012;302:R1209–18. doi: 10.1152/ajpregu.00604.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Lasker JM, Chen WB, Wolf I, Bloswick BP, Wilson PD, Powell PK. Formation of 20-hydroxyeicosatetraenoic acid, a vasoactive and natriuretic eicosanoid, in human kidney. Role of Cyp4F2 and Cyp4A11. J Biol Chem. 2000;275:4118–26. doi: 10.1074/jbc.275.6.4118. [DOI] [PubMed] [Google Scholar]
  • 29.Powell PK, Wolf I, Jin R, Lasker JM. Metabolism of arachidonic acid to 20-hydroxy-5,8,11, 14-eicosatetraenoic acid by P450 enzymes in human liver: involvement of CYP4F2 and CYP4A11. J Pharmacol Exp Ther. 1998;285:1327–36. [PubMed] [Google Scholar]
  • 30.Hirani V, Yarovoy A, Kozeska A, Magnusson RP, Lasker JM. Expression of CYP4F2 in human liver and kidney: assessment using targeted peptide antibodies. Arch Biochem Biophys. 2008;478:59–68. doi: 10.1016/j.abb.2008.06.025. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Gainer JV, Bellamine A, Dawson EP, Womble KE, Grant SW, Wang Y, Cupples LA, Guo CY, Demissie S, O’Donnell CJ, Brown NJ, Waterman MR, Capdevila JH. Functional variant of CYP4A11 20-hydroxyeicosatetraenoic acid synthase is associated with essential hypertension. Circulation. 2005;111:63–9. doi: 10.1161/01.CIR.0000151309.82473.59. [DOI] [PubMed] [Google Scholar]
  • 32.Kikuta Y, Kusunose E, Endo K, Yamamoto S, Sogawa K, Fujii-Kuriyama Y, Kusunose M. A novel form of cytochrome P-450 family 4 in human polymorphonuclear leukocytes. cDNA cloning and expression of leukotriene B4 omega-hydroxylase. J Biol Chem. 1993;268:9376–80. [PubMed] [Google Scholar]
  • 33.Fan F, Geurts AM, Murphy SR, Pabbidi MR, Jacob HJ, Roman RJ. Impaired myogenic response and autoregulation of cerebral blood flow is rescued in CYP4A1 transgenic Dahl salt-sensitive rat. Am J Physiol Regul Integr Comp Physiol. 2015;308:R379–90. doi: 10.1152/ajpregu.00256.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Hardwick JP, Song BJ, Huberman E, Gonzalez FJ. Isolation, complementary DNA sequence, and regulation of rat hepatic lauric acid omega-hydroxylase (cytochrome P-450LA omega). Identification of a new cytochrome P-450 gene family. J Biol Chem. 1987;262:801–10. [PubMed] [Google Scholar]
  • 35.Nguyen X, Wang MH, Reddy KM, Falck JR, Schwartzman ML. Kinetic profile of the rat CYP4A isoforms: arachidonic acid metabolism and isoform-specific inhibitors. Am J Physiol. 1999;276:R1691–700. doi: 10.1152/ajpregu.1999.276.6.R1691. [DOI] [PubMed] [Google Scholar]
  • 36.Kawashima H, Kusunose E, Thompson CM, Strobel HW. Protein expression, characterization, and regulation of CYP4F4 and CYP4F5 cloned from rat brain. Arch Biochem Biophys. 1997;347:148–54. doi: 10.1006/abbi.1997.0342. [DOI] [PubMed] [Google Scholar]
  • 37.Kawashima H, Strobel HW. cDNA cloning of three new forms of rat brain cytochrome P450 belonging to the CYP4F subfamily. Biochem Biophys Res Commun. 1995;217:1137–44. doi: 10.1006/bbrc.1995.2887. [DOI] [PubMed] [Google Scholar]
  • 38.Kikuta Y, Kusunose E, Ito M, Kusunose M. Purification and characterization of recombinant rat hepatic CYP4F1. Arch Biochem Biophys. 1999;369:193–6. doi: 10.1006/abbi.1999.1271. [DOI] [PubMed] [Google Scholar]
  • 39.Xu F, Falck JR, Ortiz de Montellano PR, Kroetz DL. Catalytic activity and isoform-specific inhibition of rat cytochrome p450 4F enzymes. J Pharmacol Exp Ther. 2004;308:887–95. doi: 10.1124/jpet.103.059626. [DOI] [PubMed] [Google Scholar]
  • 40.Bylund J, Harder AG, Maier KG, Roman RJ, Harder DR. Leukotriene B4 omega-side chain hydroxylation by CYP4F5 and CYP4F6. Arch Biochem Biophys. 2003;412:34–41. doi: 10.1016/s0003-9861(03)00030-4. [DOI] [PubMed] [Google Scholar]
  • 41.Holla VR, Adas F, Imig JD, Zhao X, Price E, Jr, Olsen N, Kovacs WJ, Magnuson MA, Keeney DS, Breyer MD, Falck JR, Waterman MR, Capdevila JH. Alterations in the regulation of androgen-sensitive Cyp 4a monooxygenases cause hypertension. Proc Natl Acad Sci U S A. 2001;98:5211–6. doi: 10.1073/pnas.081627898. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Muller DN, Schmidt C, Barbosa-Sicard E, Wellner M, Gross V, Hercule H, Markovic M, Honeck H, Luft FC, Schunck WH. Mouse Cyp4a isoforms: enzymatic properties, gender- and strain-specific expression, and role in renal 20-hydroxyeicosatetraenoic acid formation. Biochem J. 2007;403:109–18. doi: 10.1042/BJ20061328. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Schwartzman ML, Falck JR, Yadagiri P, Escalante B. Metabolism of 20-hydroxyeicosatetraenoic acid by cyclooxygenase. Formation and identification of novel endothelium-dependent vasoconstrictor metabolites. J Biol Chem. 1989;264:11658–62. [PubMed] [Google Scholar]
  • 44.Escalante B, Sessa WC, Falck JR, Yadagiri P, Schwartzman ML. Vasoactivity of 20-hydroxyeicosatetraenoic acid is dependent on metabolism by cyclooxygenase. J Pharmacol Exp Ther. 1989;248:229–32. [PubMed] [Google Scholar]
  • 45.Collins XH, Harmon SD, Kaduce TL, Berst KB, Fang X, Moore SA, Raju TV, Falck JR, Weintraub NL, Duester G, Plapp BV, Spector AA. Omega-oxidation of 20-hydroxyeicosatetraenoic acid (20-HETE) in cerebral microvascular smooth muscle and endothelium by alcohol dehydrogenase 4. J Biol Chem. 2005;280:33157–64. doi: 10.1074/jbc.M504055200. [DOI] [PubMed] [Google Scholar]
  • 46.Jarrar YB, Cha EY, Seo KA, Ghim JL, Kim HJ, Kim DH, Lee SJ, Shin JG. Determination of major UDP-glucuronosyltransferase enzymes and their genotypes responsible for 20-HETE glucuronidation. J Lipid Res. 2014;55:2334–42. doi: 10.1194/jlr.M051169. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Knights KM, Rowland A, Miners JO. Renal drug metabolism in humans: the potential for drug-endobiotic interactions involving cytochrome P450 (CYP) and UDP-glucuronosyltransferase (UGT) Br J Clin Pharmacol. 2013;76:587–602. doi: 10.1111/bcp.12086. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Dreisbach AW, Smith SV, Kyle PB, Ramaiah M, Amenuke M, Garrett MR, Lirette ST, Griswold ME, Roman RJ. Urinary CYP eicosanoid excretion correlates with glomerular filtration in African-Americans with chronic kidney disease. Prostaglandins Other Lipid Mediat. 2014;113–115:45–51. doi: 10.1016/j.prostaglandins.2014.08.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Marji JS, Wang MH, Laniado-Schwartzman M. Cytochrome P-450 4A isoform expression and 20-HETE synthesis in renal preglomerular arteries. Am J Physiol Renal Physiol. 2002;283:F60–7. doi: 10.1152/ajprenal.00265.2001. [DOI] [PubMed] [Google Scholar]
  • 50.Renic M, Klaus JA, Omura T, Kawashima N, Onishi M, Miyata N, Koehler RC, Harder DR, Roman RJ. Effect of 20-HETE inhibition on infarct volume and cerebral blood flow after transient middle cerebral artery occlusion. J Cereb Blood Flow Metab. 2009;29:629–39. doi: 10.1038/jcbfm.2008.156. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 51.Carroll MA, Kemp R, Cheng MK, McGiff JC. Regulation of preglomerular microvascular 20-hydroxyeicosatetraenoic acid levels by salt depletion. Med Sci Monit. 2001;7:567–72. [PubMed] [Google Scholar]
  • 52.Fan F, Sun CW, Maier KG, Williams JM, Pabbidi MR, Didion SP, Falck JR, Zhuo J, Roman RJ. 20-Hydroxyeicosatetraenoic acid contributes to the inhibition of K+ channel activity and vasoconstrictor response to angiotensin II in rat renal microvessels. PLoS One. 2013;8:e82482. doi: 10.1371/journal.pone.0082482. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Birks EK, Bousamra M, Presberg K, Marsh JA, Effros RM, Jacobs ER. Human pulmonary arteries dilate to 20-HETE, an endogenous eicosanoid of lung tissue. Am J Physiol. 1997;272:L823–9. doi: 10.1152/ajplung.1997.272.5.L823. [DOI] [PubMed] [Google Scholar]
  • 54.Yaghi A, Webb CD, Scott JA, Mehta S, Bend JR, McCormack DG. Cytochrome P450 metabolites of arachidonic acid but not cyclooxygenase-2 metabolites contribute to the pulmonary vascular hyporeactivity in rats with acute Pseudomonas pneumonia. J Pharmacol Exp Ther. 2001;297:479–88. [PubMed] [Google Scholar]
  • 55.Wang JS, Singh H, Zhang F, Ishizuka T, Deng H, Kemp R, Wolin MS, Hintze TH, Abraham NG, Nasjletti A, Laniado-Schwartzman M. Endothelial dysfunction and hypertension in rats transduced with CYP4A2 adenovirus. Circ Res. 2006;98:962–9. doi: 10.1161/01.RES.0000217283.98806.a6. [DOI] [PubMed] [Google Scholar]
  • 56.Inoue K, Sodhi K, Puri N, Gotlinger KH, Cao J, Rezzani R, Falck JR, Abraham NG, Laniado-Schwartzman M. Endothelial-specific CYP4A2 overexpression leads to renal injury and hypertension via increased production of 20-HETE. Am J Physiol Renal Physiol. 2009;297:F875–84. doi: 10.1152/ajprenal.00364.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 57.Singh H, Cheng J, Deng H, Kemp R, Ishizuka T, Nasjletti A, Schwartzman ML. Vascular cytochrome P450 4A expression and 20-hydroxyeicosatetraenoic acid synthesis contribute to endothelial dysfunction in androgen-induced hypertension. Hypertension. 2007;50:123–9. doi: 10.1161/HYPERTENSIONAHA.107.089599. [DOI] [PubMed] [Google Scholar]
  • 58.Wu CC, Cheng J, Zhang FF, Gotlinger KH, Kelkar M, Zhang Y, Jat JL, Falck JR, Schwartzman ML. Androgen-dependent hypertension is mediated by 20-hydroxy-5,8,11,14-eicosatetraenoic acid-induced vascular dysfunction: role of inhibitor of kappaB Kinase. Hypertension. 2011;57:788–94. doi: 10.1161/HYPERTENSIONAHA.110.161570. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Singh H, Schwartzman ML. Renal vascular cytochrome P450-derived eicosanoids in androgen-induced hypertension. Pharmacol Rep. 2008;60:29–37. [PubMed] [Google Scholar]
  • 60.Renic M, Kumar SN, Gebremedhin D, Florence MA, Gerges NZ, Falck JR, Harder DR, Roman RJ. Protective effect of 20-HETE inhibition in a model of oxygen-glucose deprivation in hippocampal slice cultures. Am J Physiol Heart Circ Physiol. 2012;302:H1285–93. doi: 10.1152/ajpheart.00340.2011. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.MacVicar BA, Newman EA. Astrocyte regulation of blood flow in the brain. Cold Spring Harb Perspect Biol. 2015;7 doi: 10.1101/cshperspect.a020388. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62.Yousif MH, Benter IF, Roman RJ. Cytochrome P450 metabolites of arachidonic acid play a role in the enhanced cardiac dysfunction in diabetic rats following ischaemic reperfusion injury. Auton Autacoid Pharmacol. 2009;29:33–41. doi: 10.1111/j.1474-8673.2009.00429.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63.Anwar-mohamed A, Zordoky BN, Aboutabl ME, El-Kadi AO. Alteration of cardiac cytochrome P450-mediated arachidonic acid metabolism in response to lipopolysaccharide-induced acute systemic inflammation. Pharmacol Res. 2010;61:410–8. doi: 10.1016/j.phrs.2009.12.015. [DOI] [PubMed] [Google Scholar]
  • 64.Antoun J, Goulitquer S, Amet Y, Dreano Y, Salaun JP, Corcos L, Plee-Gautier E. CYP4F3B is induced by PGA1 in human liver cells: a regulation of the 20-HETE synthesis. J Lipid Res. 2008;49:2135–41. doi: 10.1194/jlr.M800043-JLR200. [DOI] [PubMed] [Google Scholar]
  • 65.Wilson TW, Alonso-Galicia M, Roman RJ. Effects of lipid-lowering agents in the Dahl salt-sensitive rat. Hypertension. 1998;31:225–31. doi: 10.1161/01.hyp.31.1.225. [DOI] [PubMed] [Google Scholar]
  • 66.Ito O, Alonso-Galicia M, Hopp KA, Roman RJ. Localization of cytochrome P-450 4A isoforms along the rat nephron. Am J Physiol. 1998;274:F395–404. doi: 10.1152/ajprenal.1998.274.2.F395. [DOI] [PubMed] [Google Scholar]
  • 67.Ito O, Roman RJ. Regulation of P-450 4A activity in the glomerulus of the rat. Am J Physiol. 1999;276:R1749–57. doi: 10.1152/ajpregu.1999.276.6.R1749. [DOI] [PubMed] [Google Scholar]
  • 68.Omata K, Abraham NG, Schwartzman ML. Renal cytochrome P-450-arachidonic acid metabolism: localization and hormonal regulation in SHR. Am J Physiol. 1992;262:F591–9. doi: 10.1152/ajprenal.1992.262.4.F591. [DOI] [PubMed] [Google Scholar]
  • 69.Tsai IJ, Croft KD, Puddey IB, Beilin LJ, Barden A. 20-Hydroxyeicosatetraenoic acid synthesis is increased in human neutrophils and platelets by angiotensin II and endothelin-1. Am J Physiol Heart Circ Physiol. 2011;300:H1194–200. doi: 10.1152/ajpheart.00733.2010. [DOI] [PubMed] [Google Scholar]
  • 70.Hill E, Fitzpatrick F, Murphy RC. Biological activity and metabolism of 20-hydroxyeicosatetraenoic acid in the human platelet. Br J Pharmacol. 1992;106:267–74. doi: 10.1111/j.1476-5381.1992.tb14327.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71.Harder DR, Roman RJ, Gebremedhin D, Birks EK, Lange AR. A common pathway for regulation of nutritive blood flow to the brain: arterial muscle membrane potential and cytochrome P450 metabolites. Acta Physiol Scand. 1998;164:527–32. doi: 10.1111/j.1365-201x.1998.tb10702.x. [DOI] [PubMed] [Google Scholar]
  • 72.Zhu D, Bousamra M, 2nd, Zeldin DC, Falck JR, Townsley M, Harder DR, Roman RJ, Jacobs ER. Epoxyeicosatrienoic acids constrict isolated pressurized rabbit pulmonary arteries. Am J Physiol Lung Cell Mol Physiol. 2000;278:L335–43. doi: 10.1152/ajplung.2000.278.2.L335. [DOI] [PubMed] [Google Scholar]
  • 73.Zeldin DC, Plitman JD, Kobayashi J, Miller RF, Snapper JR, Falck JR, Szarek JL, Philpot RM, Capdevila JH. The rabbit pulmonary cytochrome P450 arachidonic acid metabolic pathway: characterization and significance. J Clin Invest. 1995;95:2150–60. doi: 10.1172/JCI117904. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Rosolowsky M, Campbell WB. Role of PGI2 and epoxyeicosatrienoic acids in relaxation of bovine coronary arteries to arachidonic acid. Am J Physiol. 1993;264:H327–35. doi: 10.1152/ajpheart.1993.264.2.H327. [DOI] [PubMed] [Google Scholar]
  • 75.Rosolowsky M, Falck JR, Willerson JT, Campbell WB. Synthesis of lipoxygenase and epoxygenase products of arachidonic acid by normal and stenosed canine coronary arteries. Circ Res. 1990;66:608–21. doi: 10.1161/01.res.66.3.608. [DOI] [PubMed] [Google Scholar]
  • 76.Munzenmaier DH, Harder DR. Cerebral microvascular endothelial cell tube formation: role of astrocytic epoxyeicosatrienoic acid release. Am J Physiol Heart Circ Physiol. 2000;278:H1163–7. doi: 10.1152/ajpheart.2000.278.4.H1163. [DOI] [PubMed] [Google Scholar]
  • 77.Alkayed NJ, Narayanan J, Gebremedhin D, Medhora M, Roman RJ, Harder DR. Molecular characterization of an arachidonic acid epoxygenase in rat brain astrocytes. Stroke. 1996;27:971–9. doi: 10.1161/01.str.27.5.971. [DOI] [PubMed] [Google Scholar]
  • 78.Amruthesh SC, Boerschel MF, McKinney JS, Willoughby KA, Ellis EF. Metabolism of arachidonic acid to epoxyeicosatrienoic acids, hydroxyeicosatetraenoic acids, and prostaglandins in cultured rat hippocampal astrocytes. J Neurochem. 1993;61:150–9. doi: 10.1111/j.1471-4159.1993.tb03550.x. [DOI] [PubMed] [Google Scholar]
  • 79.Wu S, Chen W, Murphy E, Gabel S, Tomer KB, Foley J, Steenbergen C, Falck JR, Moomaw CR, Zeldin DC. Molecular cloning, expression, and functional significance of a cytochrome P450 highly expressed in rat heart myocytes. J Biol Chem. 1997;272:12551–9. doi: 10.1074/jbc.272.19.12551. [DOI] [PubMed] [Google Scholar]
  • 80.Wu S, Moomaw CR, Tomer KB, Falck JR, Zeldin DC. Molecular cloning and expression of CYP2J2, a human cytochrome P450 arachidonic acid epoxygenase highly expressed in heart. J Biol Chem. 1996;271:3460–8. doi: 10.1074/jbc.271.7.3460. [DOI] [PubMed] [Google Scholar]
  • 81.Zhang QY, Raner G, Ding X, Dunbar D, Coon MJ, Kaminsky LS. Characterization of the cytochrome P450 CYP2J4: expression in rat small intestine and role in retinoic acid biotransformation from retinal. Arch Biochem Biophys. 1998;353:257–64. doi: 10.1006/abbi.1998.0654. [DOI] [PubMed] [Google Scholar]
  • 82.Zeldin DC, Foley J, Boyle JE, Moomaw CR, Tomer KB, Parker C, Steenbergen C, Wu S. Predominant expression of an arachidonate epoxygenase in islets of Langerhans cells in human and rat pancreas. Endocrinology. 1997;138:1338–46. doi: 10.1210/endo.138.3.4970. [DOI] [PubMed] [Google Scholar]
  • 83.Schwartzman M, Ferreri NR, Carroll MA, Songu-Mize E, McGiff JC. Renal cytochrome P450-related arachidonate metabolite inhibits (Na+ + K+)ATPase. Nature. 1985;314:620–2. doi: 10.1038/314620a0. [DOI] [PubMed] [Google Scholar]
  • 84.Quigley R, Baum M, Reddy KM, Griener JC, Falck JR. Effects of 20-HETE and 19(S)-HETE on rabbit proximal straight tubule volume transport. Am J Physiol. 2000;278:F949–53. doi: 10.1152/ajprenal.2000.278.6.F949. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85.Ito O, Roman RJ. Role of 20-HETE in elevating chloride transport in the thick ascending limb of Dahl SS/Jr rats. Hypertension. 1999;33:419–23. doi: 10.1161/01.hyp.33.1.419. [DOI] [PubMed] [Google Scholar]
  • 86.Park F, Sweeney WE, Jia G, Akbulut T, Mueller B, Falck JR, Birudaraju S, Roman RJ, Avner ED. Chronic blockade of 20-HETE synthesis reduces polycystic kidney disease in an orthologous rat model of ARPKD. Am J Physiol. 2009;296:F575–F582. doi: 10.1152/ajprenal.90705.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 87.Park F, Sweeney WE, Jia G, Roman RJ, Avner ED. 20-HETE mediates proliferation of renal epithelial cells in polycystic kidney disease. JASN. 2008;19:1929–1939. doi: 10.1681/ASN.2007070771. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88.Klawitter J, Klawitter J, McFann K, Pennington AT, Abebe KZ, Brosnahan G, Cadnapaphornchai MA, Chonchol M, Gitomer B, Christians U, Schrier RW. Bioactive lipid mediators in polycystic kidney disease. J Lipid Res. 2013;55:1139–1149. doi: 10.1194/jlr.P042176. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 89.Ma YH, Schwartzman ML, Roman RJ. Altered renal P-450 metabolism of arachidonic acid in Dahl salt-sensitive rats. Am J Physiol. 1994;267:R579–89. doi: 10.1152/ajpregu.1994.267.2.R579. [DOI] [PubMed] [Google Scholar]
  • 90.Honeck H, Gross V, Erdmann B, Kargel E, Neunaber R, Milia AF, Schneider W, Luft FC, Schunck WH. Cytochrome P450-dependent renal arachidonic acid metabolism in desoxycorticosterone acetate-salt hypertensive mice. Hypertension. 2000;36:610–6. doi: 10.1161/01.hyp.36.4.610. [DOI] [PubMed] [Google Scholar]
  • 91.Imig JD, Falck JR, Gebremedhin D, Harder DR, Roman RJ. Elevated renovascular tone in young spontaneously hypertensive rats. Role of cytochrome P-450. Hypertension. 1993;22:357–64. doi: 10.1161/01.hyp.22.3.357. [DOI] [PubMed] [Google Scholar]
  • 92.Kroetz DL, Huse LM, Thuresson A, Grillo MP. Developmentally regulated expression of the CYP4A genes in the spontaneously hypertensive rat kidney. Mol Pharmacol. 1997;52:362–72. doi: 10.1124/mol.52.3.362. [DOI] [PubMed] [Google Scholar]
  • 93.Laffer CL, Laniado-Schwartzman M, Wang MH, Nasjletti A, Elijovich F. 20-HETE and furosemide-induced natriuresis in salt-sensitive essential hypertension. Hypertension. 2003;41:703–8. doi: 10.1161/01.HYP.0000051888.91497.47. [DOI] [PubMed] [Google Scholar]
  • 94.Messer-Letienne I, Bernard N, Roman RJ, Sassard J, Benzoni D. Cytochrome P-450 arachidonate metabolite inhibition improves renal function in Lyon hypertensive rats. Am J Hypertens. 1999;12:398–404. doi: 10.1016/s0895-7061(98)00256-8. [DOI] [PubMed] [Google Scholar]
  • 95.Messer-Letienne I, Bernard N, Roman RJ, Sassard J, Benzoni D. 20-Hydroxyeicosatetraenoic acid and renal function in Lyon hypertensive rats. Eur J Pharmacol. 1999;378:291–7. doi: 10.1016/s0014-2999(99)00470-7. [DOI] [PubMed] [Google Scholar]
  • 96.Sacerdoti D, Abraham NG, McGiff JC, Schwartzman ML. Renal cytochrome P-450-dependent metabolism of arachidonic acid in spontaneously hypertensive rats. Biochem Pharmacol. 1988;37:521–7. doi: 10.1016/0006-2952(88)90223-7. [DOI] [PubMed] [Google Scholar]
  • 97.Nakagawa K, Marji JS, Schwartzman ML, Waterman MR, Capdevila JH. Androgen-mediated induction of the kidney arachidonate hydroxylases is associated with the development of hypertension. Am J Physiol Regul Integr Comp Physiol. 2003;284:R1055–62. doi: 10.1152/ajpregu.00459.2002. [DOI] [PubMed] [Google Scholar]
  • 98.Zhang B, Yi X, Wang C, Liao D, Lin J, Chi L. Cytochrome 4A11 Genetic Polymorphisms Increase Susceptibility to Ischemic Stroke and Associate with Atherothrombotic Events After Stroke in Chinese. Genet Test Mol Biomarkers. 2015;19:235–41. doi: 10.1089/gtmb.2014.0305. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Ding H, Cui G, Zhang L, Xu Y, Bao X, Tu Y, Wu B, Wang Q, Hui R, Wang W, Dackor RT, Kissling GE, Zeldin DC, Wang DW. Association of common variants of CYP4A11 and CYP4F2 with stroke in the Han Chinese population. Pharmacogenet Genomics. 2010;20:187–94. doi: 10.1097/FPC.0b013e328336eefe. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100.Elijovich F, Laffer CL. The relationship between CYP4A11 and human hypertension. J Hypertens. 2008;26:1712–4. doi: 10.1097/HJH.0b013e3283000504. [DOI] [PubMed] [Google Scholar]
  • 101.Fu Z, Ma Y, Xie X, Huang D, Yang H, Nakayama T, Sato N. A novel polymorphism of the CYP4A11 gene is associated with coronary artery disease. Clin Appl Thromb Hemost. 2013;19:60–5. doi: 10.1177/1076029611436197. [DOI] [PubMed] [Google Scholar]
  • 102.Fu Z, Nakayama T, Sato N, Izumi Y, Kasamaki Y, Shindo A, Ohta M, Soma M, Aoi N, Sato M, Matsumoto K, Ozawa Y, Ma Y. Haplotype-based case study of human CYP4A11 gene and cerebral infarction in Japanese subject. Endocrine. 2008;33:215–22. doi: 10.1007/s12020-008-9078-6. [DOI] [PubMed] [Google Scholar]
  • 103.Fu Z, Nakayama T, Sato N, Izumi Y, Kasamaki Y, Shindo A, Ohta M, Soma M, Aoi N, Sato M, Ozawa Y, Ma Y. A haplotype of the CYP4A11 gene associated with essential hypertension in Japanese men. J Hypertens. 2008;26:453–61. doi: 10.1097/HJH.0b013e3282f2f10c. [DOI] [PubMed] [Google Scholar]
  • 104.Fu Z, Nakayama T, Sato N, Izumi Y, Kasamaki Y, Shindo A, Ohta M, Soma M, Aoi N, Sato M, Ozawa Y, Ma Y. Haplotype-based case-control study of CYP4A11 gene and myocardial infarction. Hereditas. 2012;149:91–8. doi: 10.1111/j.1601-5223.2012.02247.x. [DOI] [PubMed] [Google Scholar]
  • 105.Fu Z, Yang H, Ma Y, Huang D, Xie X, Zheng Y, Zhu Q, Nakayama T. Haplotype study of the CYP4A11 gene and coronary artery disease in Han and Uygur populations in China. Gene. 2013;512:510–6. doi: 10.1016/j.gene.2012.10.007. [DOI] [PubMed] [Google Scholar]
  • 106.Gainer JV, Lipkowitz MS, Yu C, Waterman MR, Dawson EP, Capdevila JH, Brown NJ, Group AS. Association of a CYP4A11 variant and blood pressure in black men. J Am Soc Nephrol. 2008;19:1606–12. doi: 10.1681/ASN.2008010063. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 107.Laffer CL, Gainer JV, Waterman MR, Capdevila JH, Laniado-Schwartzman M, Nasjletti A, Brown NJ, Elijovich F. The T8590C polymorphism of CYP4A11 and 20-hydroxyeicosatetraenoic acid in essential hypertension. Hypertension. 2008;51:767–72. doi: 10.1161/HYPERTENSIONAHA.107.102921. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 108.Liang JQ, Yan MR, Yang L, Suyila Q, Cui HW, Su XL. Association of a CYP4A11 polymorphism and hypertension in the Mongolian and Han populations of China. Genet Mol Res. 2014;13:508–17. doi: 10.4238/2014.January.21.20. [DOI] [PubMed] [Google Scholar]
  • 109.Sugimoto K, Akasaka H, Katsuya T, Node K, Fujisawa T, Shimaoka I, Yasuda O, Ohishi M, Ogihara T, Shimamoto K, Rakugi H. A polymorphism regulates CYP4A11 transcriptional activity and is associated with hypertension in a Japanese population. Hypertension. 2008;52:1142–8. doi: 10.1161/HYPERTENSIONAHA.108.114082. [DOI] [PubMed] [Google Scholar]
  • 110.Ward NC, Tsai IJ, Barden A, van Bockxmeer FM, Puddey IB, Hodgson JM, Croft KD. A single nucleotide polymorphism in the CYP4F2 but not CYP4A11 gene is associated with increased 20-HETE excretion and blood pressure. Hypertension. 2008;51:1393–8. doi: 10.1161/HYPERTENSIONAHA.107.104463. [DOI] [PubMed] [Google Scholar]
  • 111.Stec DE, Roman RJ, Flasch A, Rieder MJ. Functional polymorphism in human CYP4F2 decreases 20-HETE production. Physiol Genomics. 2007;30:74–81. doi: 10.1152/physiolgenomics.00003.2007. [DOI] [PubMed] [Google Scholar]
  • 112.Ward NC, Croft KD, Puddey IB, Phillips M, van Bockxmeer F, Beilin LJ, Barden AE. The effect of a single nucleotide polymorphism of the CYP4F2 gene on blood pressure and 20-hydroxyeicosatetraenoic acid excretion after weight loss. J Hypertens. 2014;32:1495–502. doi: 10.1097/HJH.0000000000000208. [DOI] [PubMed] [Google Scholar]
  • 113.Deng S, Zhu G, Liu F, Zhang H, Qin X, Li L, Zhiyi H. CYP4F2 gene V433M polymorphism is associated with ischemic stroke in the male Northern Chinese Han population. Prog Neuropsychopharmacol Biol Psychiatry. 2010;34:664–8. doi: 10.1016/j.pnpbp.2010.03.009. [DOI] [PubMed] [Google Scholar]
  • 114.Fava C, Montagnana M, Almgren P, Rosberg L, Lippi G, Hedblad B, Engstrom G, Berglund G, Minuz P, Melander O. The V433M variant of the CYP4F2 is associated with ischemic stroke in male Swedes beyond its effect on blood pressure. Hypertension. 2008;52:373–80. doi: 10.1161/HYPERTENSIONAHA.108.114199. [DOI] [PubMed] [Google Scholar]
  • 115.Fu Z, Nakayama T, Sato N, Izumi Y, Kasamaki Y, Shindo A, Ohta M, Soma M, Aoi N, Sato M, Matsumoto K, Ozawa Y, Ma Y. A haplotype of the CYP4F2 gene is associated with cerebral infarction in Japanese men. Am J Hypertens. 2008;21:1216–23. doi: 10.1038/ajh.2008.276. [DOI] [PubMed] [Google Scholar]
  • 116.Miyata N, Roman RJ. Role of 20-hydroxyeicosatetraenoic acid (20-HETE) in vascular system. J Smooth Muscle Res. 2005;41:175–93. doi: 10.1540/jsmr.41.175. [DOI] [PubMed] [Google Scholar]
  • 117.Wu CC, Gupta T, Garcia V, Ding Y, Schwartzman ML. 20-HETE and blood pressure regulation: clinical implications. Cardiol Rev. 2014;22:1–12. doi: 10.1097/CRD.0b013e3182961659. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118.Hoopes SL, Garcia V, Edin ML, Schwartzman ML, Zeldin DC. Vascular actions of 20-HETE. Prostaglandins Other Lipid Mediat. 2015;120:9–16. doi: 10.1016/j.prostaglandins.2015.03.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Williams JM, Murphy S, Burke M, Roman RJ. 20-hydroxyeicosatetraeonic acid: a new target for the treatment of hypertension. J Cardiovasc Pharmacol. 2010;56:336–44. doi: 10.1097/FJC.0b013e3181f04b1c. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 120.Khan AH, Falck JR, Manthati VL, Campbell WB, Imig JD. Epoxyeicosatrienoic acid analog attenuates angiotensin II hypertension and kidney injury. Front Pharmacol. 2014;5:216. doi: 10.3389/fphar.2014.00216. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Imig JD, Hye Khan MA. Cytochrome P450 and Lipoxygenase Metabolites on Renal Function. Compr Physiol. 2015;6:423–41. doi: 10.1002/cphy.c150009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122.Kehl F, Cambj-Sapunar L, Maier KG, Miyata N, Kametani S, Okamoto H, Hudetz AG, Schulte ML, Zagorac D, Harder DR, Roman RJ. 20-HETE contributes to the acute fall in cerebral blood flow after subarachnoid hemorrhage in the rat. Am J Physiol Heart Circ Physiol. 2002;282:H1556–65. doi: 10.1152/ajpheart.00924.2001. [DOI] [PubMed] [Google Scholar]
  • 123.Kawasaki T, Marumo T, Shirakami K, Mori T, Doi H, Suzuki M, Watanabe Y, Chaki S, Nakazato A, Ago Y, Hashimoto H, Matsuda T, Baba A, Onoe H. Increase of 20-HETE synthase after brain ischemia in rats revealed by PET study with 11C-labeled 20-HETE synthase-specific inhibitor. J Cereb Blood Flow Metab. 2012;32:1737–46. doi: 10.1038/jcbfm.2012.68. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124.Hacein-Bey L, Harder DR, Meier HT, Varelas PN, Miyata N, Lauer KK, Cusick JF, Roman RJ. Reversal of delayed vasospasm by TS-011 in the dual hemorrhage dog model of subarachnoid hemorrhage. AJNR Am J Neuroradiol. 2006;27:1350–4. [PMC free article] [PubMed] [Google Scholar]
  • 125.Yi XY, Liao DX, Wang C, Cheng W, Fu XQ, Zhang B. Cytochrome P450 Genetic Variants and Their Metabolite Levels Associated with Plaque Stability in Patients with Ischemic Stroke. J Atheroscler Thromb. 2015 doi: 10.5551/jat.31120. [DOI] [PubMed] [Google Scholar]
  • 126.Muroya Y, Fan F, Regner KR, Falck JR, Garrett MR, Juncos LA, Roman RJ. Deficiency in the Formation of 20-Hydroxyeicosatetraenoic Acid Enhances Renal Ischemia-Reperfusion Injury. J Am Soc Nephrol. 2015;26:2460–9. doi: 10.1681/ASN.2014090868. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127.Regner KR, Zuk A, Van Why SK, Shames BD, Ryan RP, Falck JR, Manthati VL, McMullen ME, Ledbetter SR, Roman RJ. Protective effect of 20-HETE analogues in experimental renal ischemia reperfusion injury. Kidney Int. 2009;75:511–7. doi: 10.1038/ki.2008.600. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128.Catella F, Lawson JA, Fitzgerald DJ, FitzGerald GA. Endogenous biosynthesis of arachidonic acid epoxides in humans: increased formation in pregnancy-induced hypertension. Proc Natl Acad Sci U S A. 1990;87:5893–7. doi: 10.1073/pnas.87.15.5893. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129.Sacerdoti D, Balazy M, Angeli P, Gatta A, McGiff JC. Eicosanoid excretion in hepatic cirrhosis. Predominance of 20-HETE. J Clin Invest. 1997;100:1264–70. doi: 10.1172/JCI119640. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Althurwi HN, Maayah ZH, Elshenawy OH, El-Kadi AO. Early Changes in Cytochrome P450s and Their Associated Arachidonic Acid Metabolites Play a Crucial Role in the Initiation of Cardiac Hypertrophy Induced by Isoproterenol. Drug Metab Dispos. 2015;43:1254–66. doi: 10.1124/dmd.115.063776. [DOI] [PubMed] [Google Scholar]
  • 131.Han Y, Zhao H, Tang H, Li X, Tan J, Zeng Q, Sun C. 20-Hydroxyeicosatetraenoic acid mediates isolated heart ischemia/reperfusion injury by increasing NADPH oxidase-derived reactive oxygen species production. Circ J. 2013;77:1807–16. doi: 10.1253/circj.cj-12-1211. [DOI] [PubMed] [Google Scholar]
  • 132.Tunctan B, Korkmaz B, Buharalioglu CK, Firat SS, Anjaiah S, Falck J, Roman RJ, Malik KU. A 20-hydroxyeicosatetraenoic acid agonist, N-[20-hydroxyeicosa-5(Z),14(Z)-dienoyl]glycine, opposes the fall in blood pressure and vascular reactivity in endotoxin-treated rats. Shock. 2008;30:329–35. doi: 10.1097/SHK.0b013e31816471c6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 133.Sari AN, Korkmaz B, Serin MS, Kacan M, Unsal D, Buharalioglu CK, Sahan Firat S, Manthati VL, Falck JR, Malik KU, Tunctan B. Effects of 5,14-HEDGE, a 20-HETE mimetic, on lipopolysaccharide-induced changes in MyD88/TAK1/IKKbeta/IkappaB-alpha/NF-kappaB pathway and circulating miR-150, miR-223, and miR-297 levels in a rat model of septic shock. Inflamm Res. 2014;63:741–56. doi: 10.1007/s00011-014-0747-z. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 134.Metea MR, Newman EA. Glial cells dilate and constrict blood vessels: a mechanism of neurovascular coupling. J Neurosci. 2006;26:2862–70. doi: 10.1523/JNEUROSCI.4048-05.2006. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135.Zou AP, Fleming JT, Falck JR, Jacobs ER, Gebremedhin D, Harder DR, Roman RJ. 20-HETE is an endogenous inhibitor of the large-conductance Ca(2+)-activated K+ channel in renal arterioles. Am J Physiol. 1996;270:R228–37. doi: 10.1152/ajpregu.1996.270.1.R228. [DOI] [PubMed] [Google Scholar]
  • 136.Gebremedhin D, Lange AR, Narayanan J, Aebly MR, Jacobs ER, Harder DR. Cat cerebral arterial smooth muscle cells express cytochrome P450 4A2 enzyme and produce the vasoconstrictor 20-HETE which enhances L-type Ca2+ current. J Physiol. 1998;507:771–81. doi: 10.1111/j.1469-7793.1998.771bs.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 137.Basora N, Boulay G, Bilodeau L, Rousseau E, Payet MD. 20-hydroxyeicosatetraenoic acid (20-HETE) activates mouse TRPC6 channels expressed in HEK293 cells. J Biol Chem. 2003;278:31709–16. doi: 10.1074/jbc.M304437200. [DOI] [PubMed] [Google Scholar]
  • 138.Brayden JE, Earley S, Nelson MT, Reading S. Transient receptor potential (TRP) channels, vascular tone and autoregulation of cerebral blood flow. Clin Exp Pharmacol Physiol. 2008;35:1116–20. doi: 10.1111/j.1440-1681.2007.04855.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139.Wen H, Ostman J, Bubb KJ, Panayiotou C, Priestley JV, Baker MD, Ahluwalia A. 20-Hydroxyeicosatetraenoic acid (20-HETE) is a novel activator of transient receptor potential vanilloid 1 (TRPV1) channel. J Biol Chem. 2012;287:13868–76. doi: 10.1074/jbc.M111.334896. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 140.Gebremedhin D, Lange AR, Lowry TF, Taheri MR, Birks EK, Hudetz AG, Narayanan J, Falck JR, Okamoto H, Roman RJ, Nithipatikom K, Campbell WB, Harder DR. Production of 20-HETE and its role in autoregulation of cerebral blood flow. Circ Res. 2000;87:60–5. doi: 10.1161/01.res.87.1.60. [DOI] [PubMed] [Google Scholar]
  • 141.Ge Y, Murphy SR, Lu Y, Falck J, Liu R, Roman RJ. Endogenously produced 20-HETE modulates myogenic and TGF response in microperfused afferent arterioles. Prostaglandins Other Lipid Mediat. 2013;102–103:42–8. doi: 10.1016/j.prostaglandins.2013.03.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 142.Ren Y, D’Ambrosio MA, Garvin JL, Peterson EL, Carretero OA. Mechanism of impaired afferent arteriole myogenic response in Dahl salt-sensitive rats: role of 20-HETE. Am J Physiol Renal Physiol. 2014;307:F533–8. doi: 10.1152/ajprenal.00283.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Zou AP, Imig JD, Kaldunski M, Ortiz de Montellano PR, Sui Z, Roman RJ. Inhibition of renal vascular 20-HETE production impairs autoregulation of renal blood flow. Am J Physiol. 1994;266:F275–82. doi: 10.1152/ajprenal.1994.266.2.F275. [DOI] [PubMed] [Google Scholar]
  • 144.Ge Y, Murphy SR, Fan F, Williams JM, Falck JR, Liu R, Roman RJ. Role of 20-HETE in the impaired myogenic and TGF responses of the Af-Art of Dahl salt-sensitive rats. Am J Physiol. 2014;307:F509–15. doi: 10.1152/ajprenal.00273.2014. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 145.Zou AP, Imig JD, Ortiz de Montellano PR, Sui Z, Falck JR, Roman RJ. Effect of P-450 omega-hydroxylase metabolites of arachidonic acid on tubuloglomerular feedback. Am J Physiol. 1994;266:F934–41. doi: 10.1152/ajprenal.1994.266.6.F934. [DOI] [PubMed] [Google Scholar]
  • 146.Fan F, Ge Y, Murphy S, Geurts A, Jacob H, Roman R. CYP4A1 transgenic rats generated using Sleeping Beauty transposon system restores the impaired myogenic responses in the afferent arteriole of Dahl S rats. Hypertens. 2013;62:A39. Abstract. [Google Scholar]
  • 147.Murphy S, Fan F, Baker R, Guerts A, Jacob H, Roman RJ. Upregulation of Renal Medullary 20-HETE Production Opposes the Development of Hypertension in Sleeping Beauty Transposon CYP4A1 Transgenic Dahl S rats. FASEB J. 2012 [Google Scholar]
  • 148.Chen Y, Medhora M, Falck JR, Pritchard KA, Jr, Jacobs ER. Mechanisms of activation of eNOS by 20-HETE and VEGF in bovine pulmonary artery endothelial cells. Am J Physiol Lung Cell Mol Physiol. 2006;291:L378–85. doi: 10.1152/ajplung.00424.2005. [DOI] [PubMed] [Google Scholar]
  • 149.Zhu D, Zhang C, Medhora M, Jacobs ER. CYP4A mRNA, protein, and product in rat lungs: novel localization in vascular endothelium. J Appl Physiol. 2002;93:330–7. doi: 10.1152/japplphysiol.01159.2001. [DOI] [PubMed] [Google Scholar]
  • 150.Pratt PF, Falck JR, Reddy KM, Kurian JB, Campbell WB. 20-HETE relaxes bovine coronary arteries through the release of prostacyclin. Hypertension. 1998;31:237–41. doi: 10.1161/01.hyp.31.1.237. [DOI] [PubMed] [Google Scholar]
  • 151.Alonso-Galicia M, Falck JR, Reddy KM, Roman RJ. 20-HETE agonists and antagonists in the renal circulation. Am J Physiol. 1999;277:F790–F796. doi: 10.1152/ajprenal.1999.277.5.F790. [DOI] [PubMed] [Google Scholar]
  • 152.Yu M, Alonso-Galicia M, Sun CW, Roman RJ, Ono N, Hirano H, Ishimoto T, Reddy YK, Katipally KR, Reddy KM, Gopal VR, Yu J, Takhi M, Falck JR. 20-hydroxyeicosatetraenoic acid (20-HETE): structural determinants for renal vasoconstriction. Bioorg Med Chem. 2003;11:2803–21. doi: 10.1016/s0968-0896(03)00192-5. [DOI] [PubMed] [Google Scholar]
  • 153.Ortiz de Montellano PR, Reich NO. Specific inactivation of hepatic fatty acid hydroxylases by acetylenic fatty acids. J Biol Chem. 1984;259:4136–41. [PubMed] [Google Scholar]
  • 154.Zou AP, Ma YH, Sui ZH, De Montellano PO, Clark JE, Masters BS, Roman RJ. Effects of 17-octadecynoic acid, a suicide-substrate inhibitor of cytochrome P450 fatty acid omega-hydroxylase, on renal function in rats. J JPharmacol Exp Ther. 1994;268:474–481. [PubMed] [Google Scholar]
  • 155.Alonso-Galicia M, Drummond HA, Reddy KK, Falck JR, Roman RJ. Inhibition of 20-HETE production contributes to the vascular responses to nitric oxide. Hypertension. 1997;29:320–5. doi: 10.1161/01.hyp.29.1.320. [DOI] [PubMed] [Google Scholar]
  • 156.Wang MH, Brand-Schieber E, Zand BA, Nguyen X, Falck JR, Balu N, Schwartzman ML. Cytochrome P450-derived arachidonic acid metabolism in the rat kidney: characterization of selective inhibitors. J Pharmacol Exp Ther. 1998;284:966–73. [PubMed] [Google Scholar]
  • 157.Miyata N, Taniguchi K, Seki T, Ishimoto T, Sato-Watanabe M, Yasuda Y, Doi M, Kametani S, Tomishima Y, Ueki T, Sato M, Kameo K. HET0016, a potent and selective inhibitor of 20-HETE synthesizing enzyme. Br J Pharmacol. 2001;133:325–9. doi: 10.1038/sj.bjp.0704101. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 158.Omura T, Tanaka Y, Miyata N, Koizumi C, Sakurai T, Fukasawa M, Hachiuma K, Minagawa T, Susumu T, Yoshida S, Nakaike S, Okuyama S, Harder DR, Roman RJ. Effect of a new inhibitor of the synthesis of 20-HETE on cerebral ischemia reperfusion injury. Stroke. 2006;37:1307–13. doi: 10.1161/01.STR.0000217398.37075.07. [DOI] [PubMed] [Google Scholar]
  • 159.Sato M, Ishii T, Kobayashi-Matsunaga Y, Amada H, Taniguchi K, Miyata N, Kameo K. Discovery of a N′-hydroxyphenylformamidine derivative HET0016 as a potent and selective 20-HETE synthase inhibitor. Bioorg Med Chem Lett. 2001;11:2993–5. doi: 10.1016/s0960-894x(01)00614-x. [DOI] [PubMed] [Google Scholar]
  • 160.Hoagland KM, Flasch AK, Roman RJ. Inhibitors of 20-HETE formation promote salt-sensitive hypertension in rats. Hypertension. 2003;42:669–673. doi: 10.1161/01.HYP.0000084634.97353.1A. [DOI] [PubMed] [Google Scholar]
  • 161.Williams JM, Sarkis A, Lopez B, Ryan RP, Flasch AK, Roman RJ. Elevations in renal interstitial hydrostatic pressure and 20-hydroxyeicosatetraenoic acid contribute to pressure natriuresis. Hypertension. 2007;49:687–94. doi: 10.1161/01.HYP.0000255753.89363.47. [DOI] [PubMed] [Google Scholar]
  • 162.Pandey VG, Garcia V, Joseph G, Zhang FF, Shkolnik B, Mishra P, Falck JR, Capdevila J, Schwartzman ML. 20-SOLA, a novel water-soluble 20-HETE antagonist, elicits natriuresis and abrogates hypertension in CYP4A14 knockout mice. Hypertension. 2014;64:A047. [Google Scholar]
  • 163.Garcia V, Shkolnik B, Pandey V, Capdevila J, Falck J, Schwartzman M. 20-SOLA, a Novel Water Soluble 20-HETE Antagonist, Reduces Blood Pressure Through Regulation of Vascular ACE Expression via an IKK Dependent Pathway. FASEB J. 2015;29:647.9. [Google Scholar]
  • 164.Deanfield J, Donald A, Ferri C, Giannattasio C, Halcox J, Halligan S, Lerman A, Mancia G, Oliver JJ, Pessina AC, Rizzoni D, Rossi GP, Salvetti A, Schiffrin EL, Taddei S, Webb DJ E. Working Group on and H. Endothelial Factors of the European Society of. Endothelial function and dysfunction. Part I: Methodological issues for assessment in the different vascular beds: a statement by the Working Group on Endothelin and Endothelial Factors of the European Society of Hypertension. J Hypertens. 2005;23:7–17. doi: 10.1097/00004872-200501000-00004. [DOI] [PubMed] [Google Scholar]
  • 165.Cheng J, Ou JS, Singh H, Falck JR, Narsimhaswamy D, Pritchard KA, Jr, Schwartzman ML. 20-hydroxyeicosatetraenoic acid causes endothelial dysfunction via eNOS uncoupling. Am J Physiol Heart Circ Physiol. 2008;294:H1018–26. doi: 10.1152/ajpheart.01172.2007. [DOI] [PubMed] [Google Scholar]
  • 166.Cheng J, Wu CC, Gotlinger KH, Zhang F, Falck JR, Narsimhaswamy D, Schwartzman ML. 20-hydroxy-5,8,11,14-eicosatetraenoic acid mediates endothelial dysfunction via IkappaB kinase-dependent endothelial nitric-oxide synthase uncoupling. J Pharmacol Exp Ther. 2010;332:57–65. doi: 10.1124/jpet.109.159863. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 167.Toth P, Csiszar A, Sosnowska D, Tucsek Z, Cseplo P, Springo Z, Tarantini S, Sonntag WE, Ungvari Z, Koller A. Treatment with the cytochrome P450 omega-hydroxylase inhibitor HET0016 attenuates cerebrovascular inflammation, oxidative stress and improves vasomotor function in spontaneously hypertensive rats. Br J Pharmacol. 2013;168:1878–88. doi: 10.1111/bph.12079. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 168.Dunn KM, Renic M, Flasch AK, Harder DR, Falck J, Roman RJ. Elevated production of 20-HETE in the cerebral vasculature contributes to severity of ischemic stroke and oxidative stress in spontaneously hypertensive rats. Am J Physiol Heart Circ Physiol. 2008;295:H2455–65. doi: 10.1152/ajpheart.00512.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 169.Schwartzman ML, da Silva JL, Lin F, Nishimura M, Abraham NG. Cytochrome P450 4A expression and arachidonic acid omega-hydroxylation in the kidney of the spontaneously hypertensive rat. Nephron. 1996;73:652–63. doi: 10.1159/000189154. [DOI] [PubMed] [Google Scholar]
  • 170.Blanton A, Nsaif R, Hercule H, Oyekan A. Nitric oxide/cytochrome P450 interactions in cyclosporin A-induced effects in the rat. J Hypertens. 2006;24:1865–72. doi: 10.1097/01.hjh.0000242412.88653.f2. [DOI] [PubMed] [Google Scholar]
  • 171.Fidelis P, Wilson L, Thomas K, Villalobos M, Oyekan AO. Renal function and vasomotor activity in mice lacking the Cyp4a14 gene. Exp Biol Med. 2010;235:1365–74. doi: 10.1258/ebm.2010.009233. [DOI] [PubMed] [Google Scholar]
  • 172.Wu CC, Mei S, Cheng J, Ding Y, Weidenhammer A, Garcia V, Zhang F, Gotlinger K, Manthati VL, Falck JR, Capdevila JH, Schwartzman ML. Androgen-sensitive hypertension associates with upregulated vascular CYP4A12-20-HETE synthase. J Am Soc Nephrol. 2013;24:1288–96. doi: 10.1681/ASN.2012070714. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 173.Ishizuka T, Cheng J, Singh H, Vitto MD, Manthati VL, Falck JR, Laniado-Schwartzman M. 20-Hydroxyeicosatetraenoic acid stimulates nuclear factor-kappaB activation and the production of inflammatory cytokines in human endothelial cells. J Pharmacol Exp Ther. 2008;324:103–10. doi: 10.1124/jpet.107.130336. [DOI] [PubMed] [Google Scholar]
  • 174.Schuck RN, Theken KN, Edin ML, Caughey M, Bass A, Ellis K, Tran B, Steele S, Simmons BP, Lih FB, Tomer KB, Wu MC, Hinderliter AL, Stouffer GA, Zeldin DC, Lee CR. Cytochrome P450-derived eicosanoids and vascular dysfunction in coronary artery disease patients. Atherosclerosis. 2013;227:442–8. doi: 10.1016/j.atherosclerosis.2013.01.034. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Ward NC, Puddey IB, Hodgson JM, Beilin LJ, Croft KD. Urinary 20-hydroxyeicosatetraenoic acid excretion is associated with oxidative stress in hypertensive subjects. Free Radic Biol Med. 2005;38:1032–6. doi: 10.1016/j.freeradbiomed.2004.12.024. [DOI] [PubMed] [Google Scholar]
  • 176.Ward NC, Rivera J, Hodgson J, Puddey IB, Beilin LJ, Falck JR, Croft KD. Urinary 20-hydroxyeicosatetraenoic acid is associated with endothelial dysfunction in humans. Circulation. 2004;110:438–43. doi: 10.1161/01.CIR.0000136808.72912.D9. [DOI] [PubMed] [Google Scholar]
  • 177.Wang J, Li H, He J, Li B, Bao Q, Zhang X, Lv Z, Zhang Y, Han J, Ai D, Zhu Y. 20-Hydroxyeicosatetraenoic acid involved in endothelial activation and thrombosis. Am J Physiol Heart Circ Physiol. 2015;308:H1359–67. doi: 10.1152/ajpheart.00802.2014. [DOI] [PubMed] [Google Scholar]
  • 178.Renna NF. Oxidative stress, vascular remodeling, and vascular inflammation in hypertension. Int J Hypertens. 2013:710136. doi: 10.1155/2013/710136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 179.Renna NF, de Las Heras N, Miatello RM. Pathophysiology of vascular remodeling in hypertension. Int J Hypertens. 2013:808353. doi: 10.1155/2013/808353. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.McGrath JC, Deighan C, Briones AM, Shafaroudi MM, McBride M, Adler J, Arribas SM, Vila E, Daly CJ. New aspects of vascular remodelling: the involvement of all vascular cell types. Exp Physiol. 2005;90:469–75. doi: 10.1113/expphysiol.2005.030130. [DOI] [PubMed] [Google Scholar]
  • 181.Langille BL. Remodeling of developing and mature arteries: endothelium, smooth muscle, and matrix. J Cardiovasc Pharmacol. 1993;21:S11–7. doi: 10.1097/00005344-199321001-00003. [DOI] [PubMed] [Google Scholar]
  • 182.Parmentier JH, Muthalif MM, Nishimoto AT, Malik KU. 20-Hydroxyeicosatetraenoic acid mediates angiotensin ii-induced phospholipase d activation in vascular smooth muscle cells. Hypertension. 2001;37:623–9. doi: 10.1161/01.hyp.37.2.623. [DOI] [PubMed] [Google Scholar]
  • 183.Stec DE, Gannon KP, Beaird JS, Drummond HA. 20-Hydroxyeicosatetraenoic acid (20-HETE) stimulates migration of vascular smooth muscle cells. Cell Physiol Biochem. 2007;19:121–8. doi: 10.1159/000099200. [DOI] [PubMed] [Google Scholar]
  • 184.Guo AM, Janic B, Sheng J, Falck JR, Roman RJ, Edwards PA, Arbab AS, Scicli AG. The cytochrome P450 4A/F-20-hydroxyeicosatetraenoic acid system: a regulator of endothelial precursor cells derived from human umbilical cord blood. J Pharmacol Exp Ther. 2011;338:421–9. doi: 10.1124/jpet.111.179036. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 185.Guo AM, Scicli G, Sheng J, Falck JC, Edwards PA, Scicli AG. 20-HETE can act as a nonhypoxic regulator of HIF-1alpha in human microvascular endothelial cells. Am J Physiol Heart Circ Physiol. 2009;297:H602–13. doi: 10.1152/ajpheart.00874.2008. [DOI] [PubMed] [Google Scholar]
  • 186.Cheng J, Edin ML, Hoopes SL, Li H, Bradbury JA, Graves JP, DeGraff LM, Lih FB, Garcia V, Shaik JS, Tomer KB, Flake GP, Falck JR, Lee CR, Poloyac SM, Schwartzman ML, Zeldin DC. Vascular characterization of mice with endothelial expression of cytochrome P450 4F2. FASEB J. 2014;28:2915–31. doi: 10.1096/fj.13-241927. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 187.Chen L, Ackerman R, Saleh M, Gotlinger KH, Kessler M, Mendelowitz LG, Falck JR, Arbab AS, Scicli AG, Schwartzman ML, Yang J, Guo AM. 20-HETE regulates the angiogenic functions of human endothelial progenitor cells and contributes to angiogenesis in vivo. J Pharmacol Exp Ther. 2014;348:442–51. doi: 10.1124/jpet.113.210120. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 188.Lu S, Zhu C, Long A, Tan L, Li Q, Zhu Y. Effect of 20-hydroxyeicosatetraenoic acid on biological behavior of human villous trophoblasts and uterine vascular smooth muscle cells. Mol Med Rep. 2014;9:1889–94. doi: 10.3892/mmr.2014.2017. [DOI] [PubMed] [Google Scholar]
  • 189.Wang Z, Tang X, Li Y, Leu C, Guo L, Zheng X, Zhu D. 20-Hydroxyeicosatetraenoic acid inhibits the apoptotic responses in pulmonary artery smooth muscle cells. Eur J Pharmacol. 2008;588:9–17. doi: 10.1016/j.ejphar.2008.03.045. [DOI] [PubMed] [Google Scholar]
  • 190.Baumbach GL, Heistad DD. Remodeling of cerebral arterioles in chronic hypertension. Hypertension. 1989;13:968–72. doi: 10.1161/01.hyp.13.6.968. [DOI] [PubMed] [Google Scholar]
  • 191.Levere RD, Martasek P, Escalante B, Schwartzman ML, Abraham NG. Effect of heme arginate administration on blood pressure in spontaneously hypertensive rats. J Clin Invest. 1990;86:213–9. doi: 10.1172/JCI114686. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Schwartzman ML, Omata K, Lin FM, Bhatt RK, Falck JR, Abraham NG. Detection of 20-hydroxyeicosatetraenoic acid in rat urine. Biochem Biophys Res Commun. 1991;180:445–9. doi: 10.1016/s0006-291x(05)81313-0. [DOI] [PubMed] [Google Scholar]
  • 193.Gebremedhin D, Ma YH, Imig JD, Harder DR, Roman RJ. Role of cytochrome P-450 in elevating renal vascular tone in spontaneously hypertensive rats. J Vasc Res. 1993;30:53–60. doi: 10.1159/000158975. [DOI] [PubMed] [Google Scholar]
  • 194.Frisbee JC, Roman RJ, Falck JR, Linderman JR, Lombard JH. Impairment of flow-induced dilation of skeletal muscle arterioles with elevated oxygen in normotensive and hypertensive rats. Microvasc Res. 2000;60:37–48. doi: 10.1006/mvre.2000.2245. [DOI] [PubMed] [Google Scholar]
  • 195.Ding Y, Wu CC, Garcia V, Dimitrova I, Weidenhammer A, Joseph G, Zhang F, Manthati VL, Falck JR, Capdevila JH, Schwartzman ML. 20-HETE induces remodeling of renal resistance arteries independent of blood pressure elevation in hypertension. Am J Physiol Renal Physiol. 2013;305:F753–63. doi: 10.1152/ajprenal.00292.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 196.Garcia V, Joseph G, Shkolnik B, Ding Y, Zhang FF, Gotlinger K, Falck JR, Dakarapu R, Capdevila JH, Bernstein KE, Schwartzman ML. Angiotensin II receptor blockade or deletion of vascular endothelial ACE does not prevent vascular dysfunction and remodeling in 20-HETE-dependent hypertension. Am J Physiol Regul Integr Comp Physiol. 2015;309:R71–8. doi: 10.1152/ajpregu.00039.2015. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Ljuca F, Drevensek G. Endothelin-1 induced vascular smooth muscle cell proliferation is mediated by cytochrome p-450 arachidonic acid metabolites. Bosn J Basic Med Sci. 2010;10:223–6. doi: 10.17305/bjbms.2010.2691. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Chen L, Ackerman R, Guo AM. 20-HETE in neovascularization. Prostaglandins Other Lipid Mediat. 2012;98:63–8. doi: 10.1016/j.prostaglandins.2011.12.005. do. [DOI] [PubMed] [Google Scholar]
  • 199.Sewer MB, Koop DR, Morgan ET. Endotoxemia in rats is associated with induction of the P4504A subfamily and suppression of several other forms of cytochrome P450. Drug Metab Dispos. 1996;24:401–7. [PubMed] [Google Scholar]
  • 200.Yaghini FA, Zhang C, Parmentier JH, Estes AM, Jafari N, Schaefer SA, Malik KU. Contribution of arachidonic acid metabolites derived via cytochrome P4504A to angiotensin II-induced neointimal growth. Hypertension. 2005;45:1182–7. doi: 10.1161/01.HYP.0000168051.04275.ea. [DOI] [PubMed] [Google Scholar]
  • 201.Sa G, Murugesan G, Jaye M, Ivashchenko Y, Fox PL. Activation of cytosolic phospholipase A2 by basic fibroblast growth factor via a p42 mitogen-activated protein kinase-dependent phosphorylation pathway in endothelial cells. J Biol Chem. 1995;270:2360–6. doi: 10.1074/jbc.270.5.2360. [DOI] [PubMed] [Google Scholar]
  • 202.Amaral SL, Maier KG, Schippers DN, Roman RJ, Greene AS. CYP4A metabolites of arachidonic acid and VEGF are mediators of skeletal muscle angiogenesis. Am J Physiol Heart Circ Physiol. 2003;284:H1528–35. doi: 10.1152/ajpheart.00406.2002. [DOI] [PubMed] [Google Scholar]
  • 203.Jiang M, Mezentsev A, Kemp R, Byun K, Falck JR, Miano JM, Nasjletti A, Abraham NG, Laniado-Schwartzman M. Smooth muscle--specific expression of CYP4A1 induces endothelial sprouting in renal arterial microvessels. Circ Res. 2004;94:167–74. doi: 10.1161/01.RES.0000111523.12842.FC. [DOI] [PubMed] [Google Scholar]
  • 204.Chen P, Guo M, Wygle D, Edwards PA, Falck JR, Roman RJ, Scicli AG. Inhibitors of cytochrome P450 4A suppress angiogenic responses. Am J Pathol. 2005;166:615–24. doi: 10.1016/S0002-9440(10)62282-1. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 205.Dhanasekaran A, Bodiga S, Gruenloh S, Gao Y, Dunn L, Falck JR, Buonaccorsi JN, Medhora M, Jacobs ER. 20-HETE increases survival and decreases apoptosis in pulmonary arteries and pulmonary artery endothelial cells. Am J Physiol Heart Circ Physiol. 2009;296:H777–86. doi: 10.1152/ajpheart.01087.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 206.Borin TF, Zuccari DA, Jardim-Perassi BV, Ferreira LC, Iskander AS, Varma NR, Shankar A, Guo AM, Scicli G, Arbab AS. HET0016, a selective inhibitor of 20-HETE synthesis, decreases pro-angiogenic factors and inhibits growth of triple negative breast cancer in mice. PLoS One. 2014;9:e116247. doi: 10.1371/journal.pone.0116247. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 207.Yu W, Chen L, Yang YQ, Falck JR, Guo AM, Li Y, Yang J. Cytochrome P450 omega-hydroxylase promotes angiogenesis and metastasis by upregulation of VEGF and MMP-9 in non-small cell lung cancer. Cancer Chemother Pharmacol. 2011;68:619–29. doi: 10.1007/s00280-010-1521-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 208.Guo M, Roman RJ, Fenstermacher JD, Brown SL, Falck JR, Arbab AS, Edwards PA, Scicli AG. 9L gliosarcoma cell proliferation and tumor growth in rats are suppressed by N-hydroxy-N′-(4-butyl-2-methylphenol) formamidine (HET0016), a selective inhibitor of CYP4A. J Pharmacol Exp Ther. 2006;317:97–108. doi: 10.1124/jpet.105.097782. [DOI] [PubMed] [Google Scholar]
  • 209.Guo M, Roman RJ, Falck JR, Edwards PA, Scicli AG. Human U251 glioma cell proliferation is suppressed by HET0016 [N-hydroxy-N′-(4-butyl-2-methylphenyl)formamidine], a selective inhibitor of CYP4A. J Pharmacol Exp Ther. 2005;315:526–33. doi: 10.1124/jpet.105.088567. [DOI] [PubMed] [Google Scholar]
  • 210.Guo AM, Sheng J, Scicli GM, Arbab AS, Lehman NL, Edwards PA, Falck JR, Roman RJ, Scicli AG. Expression of CYP4A1 in U251 human glioma cell induces hyperproliferative phenotype in vitro and rapidly growing tumors in vivo. J Pharmacol Exp Ther. 2008;327:10–9. doi: 10.1124/jpet.108.140889. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 211.Alexanian A, Miller B, Roman RJ, Sorokin A. 20-HETE-producing enzymes are up-regulated in human cancers. Cancer Genomics Proteomics. 2012;9:163–9. [PMC free article] [PubMed] [Google Scholar]
  • 212.Mozzillo N, Pennacchioli E, Gandini S, Caraco C, Crispo A, Botti G, Lastoria S, Barberis M, Verrecchia F, Testori A. Sentinel node biopsy in thin and thick melanoma. Ann Surg Oncol. 2013;20:2780–6. doi: 10.1245/s10434-012-2826-0. [DOI] [PubMed] [Google Scholar]
  • 213.Alexanian A, Sorokin A. Targeting 20-HETE producing enzymes in cancer - rationale, pharmacology, and clinical potential. Onco Targets Ther. 2013;6:243–55. doi: 10.2147/OTT.S31586. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 214.Johnson AL, Edson KZ, Totah RA, Rettie AE. Cytochrome P450 omega-Hydroxylases in Inflammation and Cancer. Adv Pharmacol. 2015;74:223–62. doi: 10.1016/bs.apha.2015.05.002. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 215.Yu XY, Glantz CS, Yao J, He H, Petrocchi AJ, Craig DK, Ciolek JT, Booth AE. Enhancing the chemical mixture methodology in emergency preparedness and consequence assessment analysis. Toxicology. 2013;313:174–84. doi: 10.1016/j.tox.2012.10.011. [DOI] [PubMed] [Google Scholar]
  • 216.Panigrahy D, Kaipainen A, Greene ER, Huang S. Cytochrome P450-derived eicosanoids: the neglected pathway in cancer. Cancer Metastasis Rev. 2010;29:723–35. doi: 10.1007/s10555-010-9264-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 217.Zhang Y, Hoda MN, Zheng X, Li W, Luo P, Maddipati KR, Seki T, Ergul A, Wang MH. Combined therapy with COX-2 inhibitor and 20-HETE inhibitor reduces colon tumor growth and the adverse effects of ischemic stroke associated with COX-2 inhibition. Am J Physiol Regul Integr Comp Physiol. 2014;307:R693–703. doi: 10.1152/ajpregu.00422.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 218.Bednar MM, Gross CE, Balazy MK, Belosludtsev Y, Colella DT, Falck JR, Balazy M. 16(R)-hydroxy-5,8,11,14-eicosatetraenoic acid, a new arachidonate metabolite in human polymorphonuclear leukocytes. Biochem Pharmacol. 2000;60:447–55. doi: 10.1016/s0006-2952(00)00345-2. [DOI] [PubMed] [Google Scholar]
  • 219.Rosolowsky M, Falck JR, Campbell WB. Metabolism of arachidonic acid by canine polymorphonuclear leukocytes synthesis of lipoxygenase and omega-oxidized metabolites. Biochim Biophys Acta. 1996;1300:143–50. doi: 10.1016/0005-2760(95)00238-3. [DOI] [PubMed] [Google Scholar]
  • 220.Zhu Y, Schieber EB, McGiff JC, Balazy M. Identification of arachidonate P-450 metabolites in human platelet phospholipids. Hypertension. 1995;25:854–9. doi: 10.1161/01.hyp.25.4.854. [DOI] [PubMed] [Google Scholar]
  • 221.Liu JY, Li N, Yang J, Li N, Qiu H, Ai D, Chiamvimonvat N, Zhu Y, Hammock BD. Metabolic profiling of murine plasma reveals an unexpected biomarker in rofecoxib-mediated cardiovascular events. Proc Natl Acad Sci U S A. 2010;107:17017–22. doi: 10.1073/pnas.1011278107. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 222.Bos CL, Richel DJ, Ritsema T, Peppelenbosch MP, Versteeg HH. Prostanoids and prostanoid receptors in signal transduction. Int J Biochem Cell Biol. 2004;36:1187–205. doi: 10.1016/j.biocel.2003.08.006. [DOI] [PubMed] [Google Scholar]
  • 223.Kowal-Bielecka O, Kowal K, Distler O, Rojewska J, Bodzenta-Lukaszyk A, Michel BA, Gay RE, Gay S, Sierakowski S. Cyclooxygenase- and lipoxygenase-derived eicosanoids in bronchoalveolar lavage fluid from patients with scleroderma lung disease: an imbalance between proinflammatory and antiinflammatory lipid mediators. Arthritis Rheum. 2005;52:3783–91. doi: 10.1002/art.21432. [DOI] [PubMed] [Google Scholar]
  • 224.Sehgal N, Agarwal V, Valli RK, Joshi SD, Antonovic L, Strobel HW, Ravindranath V. Cytochrome P4504f, a potential therapeutic target limiting neuroinflammation. Biochem Pharmacol. 2011;82:53–64. doi: 10.1016/j.bcp.2011.03.025. [DOI] [PubMed] [Google Scholar]
  • 225.Wang Y, Zhao J, Kalsotra A, Turman CM, Grill RJ, Dash PK, Strobel HW. CYP4Fs expression in rat brain correlates with changes in LTB4 levels after traumatic brain injury. J Neurotrauma. 2008;25:1187–94. doi: 10.1089/neu.2008.0542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 226.Liu X, Zhu P, Freedman BD. Multiple eicosanoid-activated nonselective cation channels regulate B-lymphocyte adhesion to integrin ligands. Am J Physiol Cell Physiol. 2006;290:C873–82. doi: 10.1152/ajpcell.00229.2005. [DOI] [PubMed] [Google Scholar]
  • 227.Tunctan B, Korkmaz B, Sari AN, Kacan M, Unsal D, Serin MS, Buharalioglu CK, Sahan-Firat S, Cuez T, Schunck WH, Manthati VL, Falck JR, Malik KU. Contribution of iNOS/sGC/PKG pathway, COX-2, CYP4A1, and gp91(phox) to the protective effect of 5,14-HEDGE, a 20-HETE mimetic, against vasodilation, hypotension, tachycardia, and inflammation in a rat model of septic shock. Nitric Oxide. 2013;33:18–41. doi: 10.1016/j.niox.2013.05.001. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 228.Tunctan B, Korkmaz B, Sari AN, Kacan M, Unsal D, Serin MS, Buharalioglu CK, Sahan-Firat S, Cuez T, Schunck WH, Falck JR, Malik KU. 5,14-HEDGE, a 20-HETE mimetic, reverses hypotension and improves survival in a rodent model of septic shock: contribution of soluble epoxide hydrolase, CYP2C23, MEK1/ERK1/2/IKKbeta/IkappaB-alpha/NF-kappaB pathway, and proinflammatory cytokine formation. Prostaglandins Other Lipid Mediat. 2013;102–103:31–41. doi: 10.1016/j.prostaglandins.2013.01.005. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 229.Sun CW, Alonso-Galicia M, Taheri MR, Falck JR, Harder DR, Roman RJ. Nitric oxide-20-hydroxyeicosatetraenoic acid interaction in the regulation of K+ channel activity and vascular tone in renal arterioles. Circ Res. 1998;83:1069–79. doi: 10.1161/01.res.83.11.1069. [DOI] [PubMed] [Google Scholar]
  • 230.Alonso-Galicia M, Hudetz AG, Shen H, Harder DR, Roman RJ. Contribution of 20-HETE to vasodilator actions of nitric oxide in the cerebral microcirculation. Stroke. 1999;30:2727–34. doi: 10.1161/01.str.30.12.2727. discussion 2734. [DOI] [PubMed] [Google Scholar]
  • 231.Wang MH, Wang J, Chang HH, Zand BA, Jiang M, Nasjletti A, Laniado-Schwartzman M. Regulation of renal CYP4A expression and 20-HETE synthesis by nitric oxide in pregnant rats. Am J Physiol Renal Physiol. 2003;285:F295–302. doi: 10.1152/ajprenal.00065.2003. [DOI] [PubMed] [Google Scholar]
  • 232.Hoff U, Lukitsch I, Chaykovska L, Ladwig M, Arnold C, Manthati VL, Fuller TF, Schneider W, Gollasch M, Muller DN, Flemming B, Seeliger E, Luft FC, Falck JR, Dragun D, Schunck WH. Inhibition of 20-HETE synthesis and action protects the kidney from ischemia/reperfusion injury. Kidney Int. 2011;79:57–65. doi: 10.1038/ki.2010.377. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 233.Lee DL, Wilson JL, Duan R, Hudson T, El-Marakby A. Peroxisome Proliferator-Activated Receptor-alpha Activation Decreases Mean Arterial Pressure, Plasma Interleukin-6, and COX-2 While Increasing Renal CYP4A Expression in an Acute Model of DOCA-Salt Hypertension. PPAR Res. 2011:502631. doi: 10.1155/2011/502631. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 234.Zhu P, Liu X, Labelle EF, Freedman BD. Mechanisms of hypotonicity-induced calcium signaling and integrin activation by arachidonic acid-derived inflammatory mediators in B cells. J Immunol. 2005;175:4981–9. doi: 10.4049/jimmunol.175.8.4981. [DOI] [PubMed] [Google Scholar]
  • 235.Broderick JP, Brott TG, Duldner JE, Tomsick T, Leach A. Initial and recurrent bleeding are the major causes of death following subarachnoid hemorrhage. Stroke. 1994;25:1342–7. doi: 10.1161/01.str.25.7.1342. [DOI] [PubMed] [Google Scholar]
  • 236.Hop JW, Rinkel GJ, Algra A, van Gijn J. Case-fatality rates and functional outcome after subarachnoid hemorrhage: a systematic review. Stroke. 1997;28:660–4. doi: 10.1161/01.str.28.3.660. [DOI] [PubMed] [Google Scholar]
  • 237.Adams HP, Jr, Kassell NF, Torner JC, Nibbelink DW, Sahs AL. Early management of aneurysmal subarachnoid hemorrhage. A report of the Cooperative Aneurysm Study. J Neurosurg. 1981;54:141–5. doi: 10.3171/jns.1981.54.2.0141. [DOI] [PubMed] [Google Scholar]
  • 238.Tian HL, Geng Z, Cui YH, Hu J, Xu T, Cao HL, Chen SW, Chen H. Risk factors for posttraumatic cerebral infarction in patients with moderate or severe head trauma. Neurosurg Rev. 2008d;31:431–6. doi: 10.1007/s10143-008-0153-5. discussion 436–7. [DOI] [PubMed] [Google Scholar]
  • 239.Tawil I, Stein DM, Mirvis SE, Scalea TM. Posttraumatic cerebral infarction: incidence, outcome, and risk factors. J Trauma. 2008;64:849–53. doi: 10.1097/TA.0b013e318160c08a. [DOI] [PubMed] [Google Scholar]
  • 240.Vergouwen MD, Ilodigwe D, Macdonald RL. Cerebral infarction after subarachnoid hemorrhage contributes to poor outcome by vasospasm-dependent and -independent effects. Stroke. 2011;42:924–9. doi: 10.1161/STROKEAHA.110.597914. [DOI] [PubMed] [Google Scholar]
  • 241.Weir B, Grace M, Hansen J, Rothberg C. Time course of vasospasm in man. J Neurosurg. 1978;48:173–8. doi: 10.3171/jns.1978.48.2.0173. [DOI] [PubMed] [Google Scholar]
  • 242.Megyesi JF, Vollrath B, Cook DA, Findlay JM. In vivo animal models of cerebral vasospasm: a review. Neurosurgery. 2000;46:448–60. [PubMed] [Google Scholar]
  • 243.Sobey CG, Faraci FM. Subarachnoid haemorrhage: what happens to the cerebral arteries? Clin Exp Pharmacol Physiol. 1998;25:867–76. doi: 10.1111/j.1440-1681.1998.tb02337.x. [DOI] [PubMed] [Google Scholar]
  • 244.Bederson JB, Levy AL, Ding WH, Kahn R, DiPerna CA, Jenkins AL, 3rd, Vallabhajosyula P. Acute vasoconstriction after subarachnoid hemorrhage. Neurosurgery. 1998;42:352–60. doi: 10.1097/00006123-199802000-00091. [DOI] [PubMed] [Google Scholar]
  • 245.Macdonald RL, Weir BK. A review of hemoglobin and the pathogenesis of cerebral vasospasm. Stroke. 1991;22:971–82. doi: 10.1161/01.str.22.8.971. [DOI] [PubMed] [Google Scholar]
  • 246.Pluta RM, Afshar JK, Boock RJ, Oldfield EH. Temporal changes in perivascular concentrations of oxyhemoglobin, deoxyhemoglobin, and methemoglobin after subarachnoid hemorrhage. J Neurosurg. 1998;88:557–61. doi: 10.3171/jns.1998.88.3.0557. [DOI] [PubMed] [Google Scholar]
  • 247.Macdonald RL, Weir BK, Marton LS, Zhang ZD, Sajdak M, Johns LM, Kowalczuk A, Borsody M. Role of adenosine 5′-triphosphate in vasospasm after subarachnoid hemorrhage: human investigations. Neurosurgery. 2001;48:854–62. doi: 10.1097/00006123-200104000-00033. [DOI] [PubMed] [Google Scholar]
  • 248.Armstead WM. Endothelins and the role of endothelin antagonists in the management of posttraumatic vasospasm. Curr Pharm Des. 2004;10:2185–92. doi: 10.2174/1381612043384178. [DOI] [PubMed] [Google Scholar]
  • 249.Juvela S. Plasma endothelin concentrations after aneurysmal subarachnoid hemorrhage. J Neurosurg. 2000;92:390–400. doi: 10.3171/jns.2000.92.3.0390. [DOI] [PubMed] [Google Scholar]
  • 250.Hoffman SW, Rzigalinski BA, Willoughby KA, Ellis EF. Astrocytes generate isoprostanes in response to trauma or oxygen radicals. J Neurotrauma. 2000;17:415–20. doi: 10.1089/neu.2000.17.415. [DOI] [PubMed] [Google Scholar]
  • 251.Hoffman SW, Moore S, Ellis EF. Isoprostanes: free radical-generated prostaglandins with constrictor effects on cerebral arterioles. Stroke. 1997;28:844–9. doi: 10.1161/01.str.28.4.844. [DOI] [PubMed] [Google Scholar]
  • 252.Cambj-Sapunar L, Yu M, Harder DR, Roman RJ. Contribution of 5-hydroxytryptamine1B receptors and 20-hydroxyeiscosatetraenoic acid to fall in cerebral blood flow after subarachnoid hemorrhage. Stroke. 2003;34:1269–75. doi: 10.1161/01.STR.0000065829.45234.69. [DOI] [PubMed] [Google Scholar]
  • 253.Harder DR. Increased sensitivity of cat cerebral arteries to serotonin upon elevation of transmural pressure. Pflugers Arch. 1988;411:698–700. doi: 10.1007/BF00580870. [DOI] [PubMed] [Google Scholar]
  • 254.Mendelow AD, McCalden TA, Hattingh J, Coull A, Rosendorff C, Eidelman BH. Cerebrovascular reactivity and metabolism after subarachnoid hemorrhage in baboons. Stroke. 1981;12:58–65. doi: 10.1161/01.str.12.1.58. [DOI] [PubMed] [Google Scholar]
  • 255.Clozel M, Watanabe H. BQ-123, a peptidic endothelin ETA receptor antagonist, prevents the early cerebral vasospasm following subarachnoid hemorrhage after intracisternal but not intravenous injection. Life Sci. 1993;52:825–34. doi: 10.1016/0024-3205(93)90081-d. [DOI] [PubMed] [Google Scholar]
  • 256.Foley PL, Caner HH, Kassell NF, Lee KS. Reversal of subarachnoid hemorrhage-induced vasoconstriction with an endothelin receptor antagonist. Neurosurgery. 1994;34:108–12. [PubMed] [Google Scholar]
  • 257.Josko J, Hendryk S, Jedrzejowska-Szypula H, Gwozdz B, Herman ZS, Gawlik R. Influence endothelin ETA receptor antagonist--BQ-123--on changes of endothelin-1 level in plasma of rats with acute vasospasm following subarachnoid hemorrhage. J Physiol Pharmacol. 1998;49:367–75. [PubMed] [Google Scholar]
  • 258.Kaminogo M, Yonekura M, Onizuka M, Yasunaga A, Shibata S. Combination of serine protease inhibitor FUT-175 and thromboxane synthetase inhibitor OKY-046 decreases cerebral vasospasm in patients with subarachnoid hemorrhage. Neurol Med Chir. 1998;38:704–8. doi: 10.2176/nmc.38.704. [DOI] [PubMed] [Google Scholar]
  • 259.Takeuchi H, Tanabe M, Okamoto H, Yamazaki M. Effects of thromboxane synthetase inhibitor (RS-5186) on experimentally-induced cerebral vasospasm. Neurol Res. 1999;21:513–6. [PubMed] [Google Scholar]
  • 260.Yamaguchi M, Zhou C, Nanda A, Zhang JH. Ras protein contributes to cerebral vasospasm in a canine double-hemorrhage model. Stroke. 2004;35:1750–5. doi: 10.1161/01.STR.0000129898.68350.9f. [DOI] [PubMed] [Google Scholar]
  • 261.Wickman G, Lan C, Vollrath B. Functional roles of the rho/rho kinase pathway and protein kinase C in the regulation of cerebrovascular constriction mediated by hemoglobin: relevance to subarachnoid hemorrhage and vasospasm. Circ Res. 2003;92:809–16. doi: 10.1161/01.RES.0000066663.12256.B2. [DOI] [PubMed] [Google Scholar]
  • 262.Obara K, Nishizawa S, Koide M, Nozawa K, Mitate A, Ishikawa T, Nakayama K. Interactive role of protein kinase C-delta with rho-kinase in the development of cerebral vasospasm in a canine two-hemorrhage model. J Vasc Res. 2005;42:67–76. doi: 10.1159/000083093. [DOI] [PubMed] [Google Scholar]
  • 263.Miyagi Y, Carpenter RC, Meguro T, Parent AD, Zhang JH. Upregulation of rho A and rho kinase messenger RNAs in the basilar artery of a rat model of subarachnoid hemorrhage. J Neurosurg. 2000;93:471–6. doi: 10.3171/jns.2000.93.3.0471. [DOI] [PubMed] [Google Scholar]
  • 264.Sasaki T, Kasuya H, Onda H, Sasahara A, Goto S, Hori T, Inoue I. Role of p38 mitogen-activated protein kinase on cerebral vasospasm after subarachnoid hemorrhage. Stroke. 2004;35:1466–70. doi: 10.1161/01.STR.0000127425.47266.20. [DOI] [PubMed] [Google Scholar]
  • 265.Laher I, Zhang JH. Protein kinase C and cerebral vasospasm. J Cereb Blood Flow Metab. 2001;21:887–906. doi: 10.1097/00004647-200108000-00001. [DOI] [PubMed] [Google Scholar]
  • 266.Matsui T, Sugawa M, Johshita H, Takuwa Y, Asano T. Activation of the protein kinase C-mediated contractile system in canine basilar artery undergoing chronic vasospasm. Stroke. 1991;22:1183–7. doi: 10.1161/01.str.22.9.1183. [DOI] [PubMed] [Google Scholar]
  • 267.Kusaka G, Kimura H, Kusaka I, Perkins E, Nanda A, Zhang JH. Contribution of Src tyrosine kinase to cerebral vasospasm after subarachnoid hemorrhage. J Neurosurg. 2003;99:383–90. doi: 10.3171/jns.2003.99.2.0383. [DOI] [PubMed] [Google Scholar]
  • 268.Nishizawa S, Yamamoto S, Uemura K. Interrelation between protein kinase C and nitric oxide in the development of vasospasm after subarachnoid hemorrhage. Neurol Res. 1996;18:89–95. doi: 10.1080/01616412.1996.11740384. [DOI] [PubMed] [Google Scholar]
  • 269.Harder DR, Dernbach P, Waters A. Possible cellular mechanism for cerebral vasospasm after experimental subarachnoid hemorrhage in the dog. J Clin Invest. 1987;80:875–80. doi: 10.1172/JCI113146. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 270.Murray MA, Faraci FM, Heistad DD. Effect of protein kinase C inhibitors on endothelin- and vasopressin-induced constriction of the rat basilar artery. Am J Physiol. 1992:263H1643–9. doi: 10.1152/ajpheart.1992.263.6.H1643. [DOI] [PubMed] [Google Scholar]
  • 271.Kim P, Schini VB, Sundt TM, Jr, Vanhoutte PM. Reduced production of cGMP underlies the loss of endothelium-dependent relaxations in the canine basilar artery after subarachnoid hemorrhage. Circ Res. 1992;70:248–56. doi: 10.1161/01.res.70.2.248. [DOI] [PubMed] [Google Scholar]
  • 272.Sobey CG, Heistad DD, Faraci FM. Effect of subarachnoid hemorrhage on dilatation of rat basilar artery in vivo. Am J Physiol. 1996;271:H126–32. doi: 10.1152/ajpheart.1996.271.1.H126. [DOI] [PubMed] [Google Scholar]
  • 273.Yamamoto S, Nishizawa S, Yokoyama T, Ryu H, Uemura K. Subarachnoid hemorrhage impairs cerebral blood flow response to nitric oxide but not to cyclic GMP in large cerebral arteries. Brain Res. 1997;757:1–9. doi: 10.1016/s0006-8993(96)01433-3. [DOI] [PubMed] [Google Scholar]
  • 274.Schwartz AY, Sehba FA, Bederson JB. Decreased nitric oxide availability contributes to acute cerebral ischemia after subarachnoid hemorrhage. Neurosurgery. 2000;47:208–14. doi: 10.1097/00006123-200007000-00042. [DOI] [PubMed] [Google Scholar]
  • 275.Poloyac SM, Reynolds RB, Yonas H, Kerr ME. Identification and quantification of the hydroxyeicosatetraenoic acids, 20-HETE and 12-HETE, in the cerebrospinal fluid after subarachnoid hemorrhage. J Neurosci Methods. 2005;144:257–63. doi: 10.1016/j.jneumeth.2004.11.015. [DOI] [PubMed] [Google Scholar]
  • 276.Donnelly MK, Crago EA, Conley YP, Balzer JR, Ren D, Ducruet AF, Kochanek PM, Sherwood PR, Poloyac SM. 20-HETE is associated with unfavorable outcomes in subarachnoid hemorrhage patients. J Cereb Blood Flow Metab. 2015;35:1515–22. doi: 10.1038/jcbfm.2015.75. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 277.Siler DA, Martini RP, Ward JP, Nelson JW, Borkar RN, Zuloaga KL, Liu JJ, Fairbanks SL, Raskin JS, Anderson VC, Dogan A, Wang RK, Alkayed NJ, Cetas JS. Protective role of p450 epoxyeicosanoids in subarachnoid hemorrhage. Neurocrit Care. 2015;22:306–19. doi: 10.1007/s12028-014-0011-y. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 278.Pilitsis JG, Coplin WM, O’Regan MH, Wellwood JM, Diaz FG, Fairfax MR, Michael DB, Phillis JW. Measurement of free fatty acids in cerebrospinal fluid from patients with hemorrhagic and ischemic stroke. Brain Res. 2003;985:198–201. doi: 10.1016/s0006-8993(03)03044-0. [DOI] [PubMed] [Google Scholar]
  • 279.Takeuchi K, Renic M, Bohman QC, Harder DR, Miyata N, Roman RJ. Reversal of delayed vasospasm by an inhibitor of the synthesis of 20-HETE. Am J Physiol Heart Circ Physiol. 2005;289:H2203–11. doi: 10.1152/ajpheart.00556.2005. [DOI] [PubMed] [Google Scholar]
  • 280.Miyata N, Seki T, Tanaka Y, Omura T, Taniguchi K, Doi M, Bandou K, Kametani S, Sato M, Okuyama S, Cambj-Sapunar L, Harder DR, Roman RJ. Beneficial effects of a new 20-hydroxyeicosatetraenoic acid synthesis inhibitor, TS-011 [N-(3-chloro-4-morpholin-4-yl) phenyl-N′-hydroxyimido formamide], on hemorrhagic and ischemic stroke. J Pharmacol Exp Ther. 2005;314:77–85. doi: 10.1124/jpet.105.083964. [DOI] [PubMed] [Google Scholar]
  • 281.Dreier JP, Major S, Manning A, Woitzik J, Drenckhahn C, Steinbrink J, Tolias C, Oliveira-Ferreira AI, Fabricius M, Hartings JA, Vajkoczy P, Lauritzen M, Dirnagl U, Bohner G, Strong AJ C. s. group. Cortical spreading ischaemia is a novel process involved in ischaemic damage in patients with aneurysmal subarachnoid haemorrhage. Brain. 2009;132:1866–81. doi: 10.1093/brain/awp102. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 282.Fordsmann JC, Ko RW, Choi HB, Thomsen K, Witgen BM, Mathiesen C, Lonstrup M, Piilgaard H, MacVicar BA, Lauritzen M. Increased 20-HETE synthesis explains reduced cerebral blood flow but not impaired neurovascular coupling after cortical spreading depression in rat cerebral cortex. J Neurosci. 2013;33:2562–70. doi: 10.1523/JNEUROSCI.2308-12.2013. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 283.Shaik JS, Poloyac SM, Kochanek PM, Alexander H, Tudorascu DL, Clark RS, Manole MD. 20-Hydroxyeicosatetraenoic Acid Inhibition by HET0016 Offers Neuroprotection, Decreases Edema, and Increases Cortical Cerebral Blood Flow in a Pediatric Asphyxial Cardiac Arrest Model in Rats. J Cereb Blood Flow Metab. 2015;35:1757–63. doi: 10.1038/jcbfm.2015.117. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 284.Cold GE. Cerebral blood flow in the acute phase after head injury. Part 2: Correlation to intraventricular pressure (IVP), cerebral perfusion pressure (CPP), PaCO2, ventricular fluid lactate, lactate/pyruvate ratio and pH. Acta Anaesthesiol Scand. 1981;25:332–5. doi: 10.1111/j.1399-6576.1981.tb01662.x. [DOI] [PubMed] [Google Scholar]
  • 285.Cold GE, Jensen FT. Cerebral autoregulation in unconscious patients with brain injury. Acta Anaesthesiol Scand. 1978;22:270–80. doi: 10.1111/j.1399-6576.1978.tb01301.x. [DOI] [PubMed] [Google Scholar]
  • 286.Overgaard J, Tweed WA. Cerebral circulation after head injury. 1. Cerebral blood flow and its regulation after closed head injury with emphasis on clinical correlations. J Neurosurg. 1974;41:531–41. doi: 10.3171/jns.1974.41.5.0531. [DOI] [PubMed] [Google Scholar]
  • 287.Paulson OB, Strandgaard S, Edvinsson L. Cerebral autoregulation. Cerebrovasc Brain Metab Rev. 1990;2:161–92. [PubMed] [Google Scholar]
  • 288.Strong AJ, Fabricius M, Boutelle MG, Hibbins SJ, Hopwood SE, Jones R, Parkin MC, Lauritzen M. Spreading and synchronous depressions of cortical activity in acutely injured human brain. Stroke. 2002;33:2738–43. doi: 10.1161/01.str.0000043073.69602.09. [DOI] [PubMed] [Google Scholar]
  • 289.Hiler M, Czosnyka M, Hutchinson P, Balestreri M, Smielewski P, Matta B, Pickard JD. Predictive value of initial computerized tomography scan, intracranial pressure, and state of autoregulation in patients with traumatic brain injury. J Neurosurg. 2006;104:731–7. doi: 10.3171/jns.2006.104.5.731. [DOI] [PubMed] [Google Scholar]
  • 290.Schmidt EA, Czosnyka M, Steiner LA, Balestreri M, Smielewski P, Piechnik SK, Matta BF, Pickard JD. Asymmetry of pressure autoregulation after traumatic brain injury. J Neurosurg. 2003;99:991–8. doi: 10.3171/jns.2003.99.6.0991. [DOI] [PubMed] [Google Scholar]
  • 291.Steiner LA, Czosnyka M, Piechnik SK, Smielewski P, Chatfield D, Menon DK, Pickard JD. Continuous monitoring of cerebrovascular pressure reactivity allows determination of optimal cerebral perfusion pressure in patients with traumatic brain injury. Crit Care Med. 2002;30:733–8. doi: 10.1097/00003246-200204000-00002. [DOI] [PubMed] [Google Scholar]
  • 292.Brady KM, Shaffner DH, Lee JK, Easley RB, Smielewski P, Czosnyka M, Jallo GI, Guerguerian AM. Continuous monitoring of cerebrovascular pressure reactivity after traumatic brain injury in children. Pediatrics. 2009;124:e1205–12. doi: 10.1542/peds.2009-0550. [DOI] [PubMed] [Google Scholar]
  • 293.Wei EP, Lamb RG, Kontos HA. Increased phospholipase C activity after experimental brain injury. J Neurosurg. 1982;56:695–8. doi: 10.3171/jns.1982.56.5.0695. [DOI] [PubMed] [Google Scholar]
  • 294.Homayoun P, Rodriguez de Turco EB, Parkins NE, Lane DC, Soblosky J, Carey ME, Bazan NG. Delayed phospholipid degradation in rat brain after traumatic brain injury. J Neurochem. 1997;69:199–205. doi: 10.1046/j.1471-4159.1997.69010199.x. [DOI] [PubMed] [Google Scholar]
  • 295.Phillis JW, O’Regan MH. A potentially critical role of phospholipases in central nervous system ischemic, traumatic, and neurodegenerative disorders. Brain Res Brain Res Rev. 2004;44:13–47. doi: 10.1016/j.brainresrev.2003.10.002. [DOI] [PubMed] [Google Scholar]
  • 296.Pilitsis JG, Coplin WM, O’Regan MH, Wellwood JM, Diaz FG, Fairfax MR, Michael DB, Phillis JW. Free fatty acids in cerebrospinal fluids from patients with traumatic brain injury. Neurosci Lett. 2003;349:136–8. doi: 10.1016/s0304-3940(03)00803-6. [DOI] [PubMed] [Google Scholar]
  • 297.Birnie M, Morrison R, Camara R, Strauss KI. Temporal changes of cytochrome P450 (Cyp) and eicosanoid-related gene expression in the rat brain after traumatic brain injury. BMC Genomics. 2013;14:303. doi: 10.1186/1471-2164-14-303. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 298.Stoll G, Kleinschnitz C, Nieswandt B. Molecular mechanisms of thrombus formation in ischemic stroke: novel insights and targets for treatment. Blood. 2008;112:3555–62. doi: 10.1182/blood-2008-04-144758. [DOI] [PubMed] [Google Scholar]
  • 299.Ginsberg MD. Adventures in the pathophysiology of brain ischemia: penumbra, gene expression, neuroprotection: the 2002 Thomas Willis Lecture. Stroke. 2003;34:214–23. doi: 10.1161/01.str.0000048846.09677.62. [DOI] [PubMed] [Google Scholar]
  • 300.Carmichael ST. Rodent models of focal stroke: size, mechanism, and purpose. NeuroRx. 2005;2:396–409. doi: 10.1602/neurorx.2.3.396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 301.Hossmann KA. Experimental models for the investigation of brain ischemia. Cardiovasc Res. 1998;39:106–20. doi: 10.1016/s0008-6363(98)00075-3. [DOI] [PubMed] [Google Scholar]
  • 302.Folbergrova J, Memezawa H, Smith ML, Siesjo BK. Focal and perifocal changes in tissue energy state during middle cerebral artery occlusion in normo- and hyperglycemic rats. J Cereb Blood Flow Metab. 1992;12:25–33. doi: 10.1038/jcbfm.1992.4. [DOI] [PubMed] [Google Scholar]
  • 303.Sarvary E, Halsey JH, Conger KA, Garcia JH, Kovach AG. ATP and pH predictors of histologic damage following global cerebral ischemia in the rat. Acta Physiol Hung. 1994;82:109–24. [PubMed] [Google Scholar]
  • 304.Chen M, Won DJ, Krajewski S, Gottlieb RA. Calpain and mitochondria in ischemia/reperfusion injury. J Biol Chem. 2002;277:29181–6. doi: 10.1074/jbc.M204951200. [DOI] [PubMed] [Google Scholar]
  • 305.MacKay KB, Patel TR, Galbraith SL, Woodruff GN, McCulloch J. The relationship between glutamate release and cerebral blood flow after focal cerebral ischaemia in the cat: effect of pretreatment with enadoline (a kappa receptor agonist) Brain Res. 1996;712:329–34. doi: 10.1016/0006-8993(95)01559-0. [DOI] [PubMed] [Google Scholar]
  • 306.Tseng EE, Brock MV, Kwon CC, Annanata M, Lange MS, Troncoso JC, Johnston MV, Baumgartner WA. Increased intracerebral excitatory amino acids and nitric oxide after hypothermic circulatory arrest. Ann Thorac Surg. 1999;67:371–6. doi: 10.1016/s0003-4975(99)00033-8. [DOI] [PubMed] [Google Scholar]
  • 307.Werling LL, Jacocks HM, 3rd, Rosenthal RE, Fiskum G. Dopamine release from canine striatum following global cerebral ischemia/reperfusion. Brain Res. 1993;606:99–105. doi: 10.1016/0006-8993(93)91575-d. [DOI] [PubMed] [Google Scholar]
  • 308.Guyot LL, Diaz FG, O’Regan MH, McLeod S, Park H, Phillis JW. Real-time measurement of glutamate release from the ischemic penumbra of the rat cerebral cortex using a focal middle cerebral artery occlusion model. Neurosci Lett. 2001;299:37–40. doi: 10.1016/s0304-3940(01)01510-5. [DOI] [PubMed] [Google Scholar]
  • 309.Mauler F, Fahrig T, Horvath E, Jork R. Inhibition of evoked glutamate release by the neuroprotective 5-HT(1A) receptor agonist BAY x 3702 in vitro and in vivo. Brain Res. 2001;888:150–157. doi: 10.1016/s0006-8993(00)03074-2. [DOI] [PubMed] [Google Scholar]
  • 310.Pilitsis JG, Diaz FG, O’Regan MH, Phillis JW. Differential effects of phospholipase inhibitors on free fatty acid efflux in rat cerebral cortex during ischemia-reperfusion injury. Brain Res. 2002;951:96–106. doi: 10.1016/s0006-8993(02)03142-6. [DOI] [PubMed] [Google Scholar]
  • 311.Narita K, Kubota M, Nakane M, Kitahara S, Nakagomi T, Tamura A, Hisaki H, Shimasaki H, Ueta N. Therapeutic time window in the penumbra during permanent focal ischemia in rats: changes of free fatty acids and glycerophospholipids. Neurol Res. 2000;22:393–400. doi: 10.1080/01616412.2000.11740689. [DOI] [PubMed] [Google Scholar]
  • 312.Sun D, Kintner D, Fitzpatrick JH, Emoto SE, Braquet PG, Bazan NG, Gilboe DD. The effect of a free radical scavenger and platelet-activating factor antagonist on FFA accumulation in post-ischemic canine brain. Neurochem Res. 1994;19:525–8. doi: 10.1007/BF00967333. [DOI] [PubMed] [Google Scholar]
  • 313.Liu Y, Mituska S, Hashizume K, Hosaka T, Nukui H. The time course of glucose metabolism in rat cerebral ischemia with middle cerebral artery occlusion-reperfusion model and the effect of MK-801. Neurol Res. 1996;18(6):505–8. doi: 10.1080/01616412.1996.11740462. [DOI] [PubMed] [Google Scholar]
  • 314.Hall ED. Inhibition of lipid peroxidation in central nervous system trauma and ischemia. J Neurol Sci. 1995;134:79–83. doi: 10.1016/0022-510x(95)00211-j. [DOI] [PubMed] [Google Scholar]
  • 315.Murin R, Drgova A, Kaplan P, Dobrota D, Lehotsky J. Ischemia/Reperfusion-induced oxidative stress causes structural changes of brain membrane proteins and lipids. Gen Physiol Biophys. 2001;20:431–8. [PubMed] [Google Scholar]
  • 316.McCracken E, Valeriani V, Simpson C, Jover T, McCulloch J, Dewar D. The lipid peroxidation by-product 4-hydroxynonenal is toxic to axons and oligodendrocytes. J Cereb Blood Flow Metab. 2000;20:1529–36. doi: 10.1097/00004647-200011000-00002. [DOI] [PubMed] [Google Scholar]
  • 317.Ciceri P, Rabuffetti M, Monopoli A, Nicosia S. Production of leukotrienes in a model of focal cerebral ischaemia in the rat. Br J Pharmacol. 2001;133:1323–9. doi: 10.1038/sj.bjp.0704189. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 318.Tegtmeier F, Weber C, Heister U, Haker I, Scheller D, Nikolov R, Holler M. Eicosanoids in rat brain during ischemia and reperfusion--correlation to DC depolarization. J Cereb Blood Flow Metab. 1990;10:358–64. doi: 10.1038/jcbfm.1990.65. [DOI] [PubMed] [Google Scholar]
  • 319.Usui M, Asano T, Takakura K. Identification and quantitative analysis of hydroxy-eicosatetraenoic acids in rat brains exposed to regional ischemia. Stroke. 1987;18:490–4. doi: 10.1161/01.str.18.2.490. [DOI] [PubMed] [Google Scholar]
  • 320.Tanaka Y, Omura T, Fukasawa M, Horiuchi N, Miyata N, Minagawa T, Yoshida S, Nakaike S. Continuous inhibition of 20-HETE synthesis by TS-011 improves neurological and functional outcomes after transient focal cerebral ischemia in rats. Neurosci Res. 2007;59:475–80. doi: 10.1016/j.neures.2007.08.018. [DOI] [PubMed] [Google Scholar]
  • 321.Ward NC, Croft KD, Blacker D, Hankey GJ, Barden A, Mori TA, Puddey IB, Beer CD. Cytochrome P450 metabolites of arachidonic acid are elevated in stroke patients compared with healthy controls. Clin Sci. 2011;121:501–7. doi: 10.1042/CS20110215. [DOI] [PubMed] [Google Scholar]
  • 322.Tanaka Y, Koizumi C, Tashiro T, Susumu T, Fukasawa M, Horiuchi N, Minagawa T, Omura T, Miyata N, Yoshida S. TS-011 improves neurological and functional outcomes after focal cerebral ischemia in rats. Stroke. 2007 doi: 10.1016/j.neures.2007.08.018. [DOI] [PubMed] [Google Scholar]
  • 323.Marumo T, Eto K, Wake H, Omura T, Nabekura J. The inhibitor of 20-HETE synthesis, TS-011, improves cerebral microcirculatory autoregulation impaired by middle cerebral artery occlusion in mice. Br J Pharmacol. 2010;161:1391–402. doi: 10.1111/j.1476-5381.2010.00973.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 324.Poloyac SM, Zhang Y, Bies RR, Kochanek PM, Graham SH. Protective effect of the 20-HETE inhibitor HET0016 on brain damage after temporary focal ischemia. J Cereb Blood Flow Metab. 2006;26:1551–61. doi: 10.1038/sj.jcbfm.9600309. [DOI] [PubMed] [Google Scholar]
  • 325.Randriamboavonjy V, Busse R, Fleming I. 20-HETE-induced contraction of small coronary arteries depends on the activation of Rho-kinase. Hypertension. 2003;41:801–6. doi: 10.1161/01.HYP.0000047240.33861.6B. [DOI] [PubMed] [Google Scholar]
  • 326.Sun CW, Falck JR, Harder DR, Roman RJ. Role of tyrosine kinase and PKC in the vasoconstrictor response to 20-HETE in renal arterioles. Hypertension. 1999;33:414–8. doi: 10.1161/01.hyp.33.1.414. [DOI] [PubMed] [Google Scholar]
  • 327.Muthalif MM, Benter IF, Khandekar Z, Gaber L, Estes A, Malik S, Parmentier JH, Manne V, Malik KU. Contribution of Ras GTPase/MAP kinase and cytochrome P450 metabolites to deoxycorticosterone-salt-induced hypertension. Hypertension. 2000;35:457–63. doi: 10.1161/01.hyp.35.1.457. [DOI] [PubMed] [Google Scholar]
  • 328.Muthalif MM, Karzoun NA, Gaber L, Khandekar Z, Benter IF, Saeed AE, Parmentier JH, Estes A, Malik KU. Angiotensin II-induced hypertension: contribution of Ras GTPase/Mitogen-activated protein kinase and cytochrome P450 metabolites. Hypertension. 2000;36:604–9. doi: 10.1161/01.hyp.36.4.604. [DOI] [PubMed] [Google Scholar]
  • 329.Muthalif MM, Uddin MR, Fatima S, Parmentier J, Khandekar Z, Malik KU. Small GTP binding protein Ras contributes to norepinephrine-induced mitogenesis of vascular smooth muscle cells(1) Prostaglandins. 2001;65:33–43. doi: 10.1016/s0090-6980(01)00112-5. [DOI] [PubMed] [Google Scholar]
  • 330.Nowicki S, Chen SL, Aizman O, Cheng XJ, Li D, Nowicki C, Nairn A, Greengard P, Aperia A. 20-Hydroxyeicosa-tetraenoic acid (20 HETE) activates protein kinase C. Role in regulation of rat renal Na+, K+-ATPase. J Clin Invest. 1997;99:1224–30. doi: 10.1172/JCI119279. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 331.Chertow GM, Burdick E, Honour M, Bonventre JV, Bates DW. Acute kidney injury, mortality, length of stay, and costs in hospitalized patients. J Am Soc Nephrol. 2005;16:3365–70. doi: 10.1681/ASN.2004090740. [DOI] [PubMed] [Google Scholar]
  • 332.Lameire N, Van Biesen W, Vanholder R. The changing epidemiology of acute renal failure. Nat Clin Pract Nephrol. 2006;2:364–77. doi: 10.1038/ncpneph0218. [DOI] [PubMed] [Google Scholar]
  • 333.Lameire N, Vanholder R. Pathophysiologic features and prevention of human and experimental acute tubular necrosis. J Am Soc Nephrol. 2001;12(Suppl 17):S20–32. [PubMed] [Google Scholar]
  • 334.Nithipatikom K, Gross ER, Endsley MP, Moore JM, Isbell MA, Falck JR, Campbell WB, Gross GJ. Inhibition of cytochrome P450omega-hydroxylase: a novel endogenous cardioprotective pathway. Circ Res. 2004;95:e65–71. doi: 10.1161/01.RES.0000146277.62128.6f. [DOI] [PubMed] [Google Scholar]
  • 335.Roman RJ, Akbulut T, Park F, Regner KR. 20-HETE in acute kidney injury. Kidney Int. 2011;79:10–3. doi: 10.1038/ki.2010.396. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 336.Williams JM, Sarkis A, Hoagland KM, Fredrich K, Ryan RP, Moreno C, Lopez B, Lazar J, Fenoy FJ, Sharma M, Garrett MR, Jacob HJ, Roman RJ. Transfer of the CYP4A region of chromosome 5 from Lewis to Dahl S rats attenuates renal injury. Am J Physiol Renal Physiol. 2008;295:F1764–77. doi: 10.1152/ajprenal.90525.2008. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 337.Basile DP, Donohoe D, Cao X, Van Why SK. Resistance to ischemic acute renal failure in the Brown Norway rat: a new model to study cytoprotection. Kidney Int. 2004;65:2201–11. doi: 10.1111/j.1523-1755.2004.00637.x. [DOI] [PubMed] [Google Scholar]
  • 338.Nilakantan V, Maenpaa C, Jia G, Roman RJ, Park F. 20-HETE-mediated cytotoxicity and apoptosis in ischemic kidney epithelial cells. Am J Physiol Renal Physiol. 2008;294:F562–70. doi: 10.1152/ajprenal.00387.2007. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 339.Oyekan AO. Differential effects of 20-hydroxyeicosatetraenoic acid on intrarenal blood flow in the rat. J Pharmacol Exp Ther. 2005;313:1289–95. doi: 10.1124/jpet.104.080218. [DOI] [PubMed] [Google Scholar]
  • 340.Akbulut T, Regner KR, Roman RJ, Avner ED, Falck JR, Park F. 20-HETE activates the Raf/MEK/ERK pathway in renal epithelial cells through an EGFR-and c-Src-dependent mechanism. Am J Physiol. 2009;297:F662–F670. doi: 10.1152/ajprenal.00146.2009. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 341.di Mari JF, Davis R, Safirstein RL. MAPK activation determines renal epithelial cell survival during oxidative injury. Am J Physiol. 1999;277:F195–203. doi: 10.1152/ajprenal.1999.277.2.F195. [DOI] [PubMed] [Google Scholar]
  • 342.Park KM, Chen A, Bonventre JV. Prevention of kidney ischemia/reperfusion-induced functional injury and JNK, p38, and MAPK kinase activation by remote ischemic pretreatment. J Biol Chem. 2001;276:11870–6. doi: 10.1074/jbc.M007518200. [DOI] [PubMed] [Google Scholar]
  • 343.Waller HL, Harper SJ, Hosgood SA, Bagul A, Kay MD, Kaushik M, Yang B, Bicknell GR, Nicholson ML. Differential expression of cytoprotective and apoptotic genes in an ischaemia-reperfusion isolated organ perfusion model of the transplanted kidney. Transpl Int. 2007;20:625–31. doi: 10.1111/j.1432-2277.2007.00489.x. [DOI] [PubMed] [Google Scholar]
  • 344.Pratschke J, Weiss S, Neuhaus P, Pascher A. Review of nonimmunological causes for deteriorated graft function and graft loss after transplantation. Transpl Int. 2008;21:512–22. doi: 10.1111/j.1432-2277.2008.00643.x. [DOI] [PubMed] [Google Scholar]
  • 345.Toledo-Pereyra LH, Toledo AH, Walsh J, Lopez-Neblina F. Molecular signaling pathways in ischemia/reperfusion. Exp Clin Transplant. 2004;2:174–7. [PubMed] [Google Scholar]
  • 346.Hernandez D, Estupinan S, Perez G, Rufino M, Gonzalez-Posada JM, Luis D, Delgado P, Rodriguez A, Marrero D, Porrini E, Torres A. Impact of cold ischemia time on renal allograft outcome using kidneys from young donors. Transpl Int. 2008;21:955–62. doi: 10.1111/j.1432-2277.2008.00708.x. [DOI] [PubMed] [Google Scholar]
  • 347.Weiss AS, Gralla J, Chan L, Klem P, Wiseman AC. Aggressive immunosuppression minimization reduces graft loss following diagnosis of BK virus-associated nephropathy: a comparison of two reduction strategies. Clin J Am Soc Nephrol. 2008;3:1812–9. doi: 10.2215/CJN.05691207. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 348.Dolegowska B, Blogowski W, Domanski L. Is it possible to predict the early post-transplant allograft function using 20-HETE measurements? A preliminary report. Transpl Int. 2009;22:546–53. doi: 10.1111/j.1432-2277.2008.00829.x. [DOI] [PubMed] [Google Scholar]
  • 349.Dolegowska B, Blogowski W, Safranow K, Domanski L, Jakubowska K, Olszewska M. Lipoxygenase-derived hydroxyeicosatetraenoic acids--novel perioperative markers of early post-transplant allograft function? Nephrol Dial Transplant. 2010;25:4061–7. doi: 10.1093/ndt/gfq320. [DOI] [PubMed] [Google Scholar]
  • 350.Gross ER, Nithipatikom K, Hsu AK, Peart JN, Falck JR, Campbell WB, Gross GJ. Cytochrome P450 omega-hydroxylase inhibition reduces infarct size during reperfusion via the sarcolemmal KATP channel. J Mol Cell Cardiol. 2004;37:1245–9. doi: 10.1016/j.yjmcc.2004.10.008. [DOI] [PubMed] [Google Scholar]
  • 351.Nithipatikom K, DiCamelli RF, Kohler S, Gumina RJ, Falck JR, Campbell WB, Gross GJ. Determination of cytochrome P450 metabolites of arachidonic acid in coronary venous plasma during ischemia and reperfusion in dogs. Anal Biochem. 2001;292:115–24. doi: 10.1006/abio.2001.5044. [DOI] [PubMed] [Google Scholar]
  • 352.Nithipatikom K, Endsley MP, Moore JM, Isbell MA, Falck JR, Campbell WB, Gross GJ. Effects of selective inhibition of cytochrome P-450 omega-hydroxylases and ischemic preconditioning in myocardial protection. Am J Physiol Heart Circ Physiol. 2006;290:H500–5. doi: 10.1152/ajpheart.00918.2005. [DOI] [PubMed] [Google Scholar]
  • 353.Bao Y, Wang X, Li W, Huo D, Shen X, Han Y, Tan J, Zeng Q, Sun C. 20-Hydroxyeicosatetraenoic acid induces apoptosis in neonatal rat cardiomyocytes through mitochondrial-dependent pathways. J Cardiovasc Pharmacol. 2011;57:294–301. doi: 10.1097/FJC.0b013e3182073c78. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 354.Mozaffarian D, Benjamin EJ, Go AS, Arnett DK, Blaha MJ, Cushman M, de Ferranti S, Despres JP, Fullerton HJ, Howard VJ, Huffman MD, Judd SE, Kissela BM, Lackland DT, Lichtman JH, Lisabeth LD, Liu S, Mackey RH, Matchar DB, McGuire DK, Mohler ER, 3rd, Moy CS, Muntner P, Mussolino ME, Nasir K, Neumar RW, Nichol G, Palaniappan L, Pandey DK, Reeves MJ, Rodriguez CJ, Sorlie PD, Stein J, Towfighi A, Turan TN, Virani SS, Willey JZ, Woo D, Yeh RW, Turner MB C. American Heart Association Statistics and S. Stroke Statistics: Heart disease and stroke statistics--2015 update: a report from the American Heart Association. Circulation. 2015;131:e29–322. doi: 10.1161/CIR.0000000000000152. [DOI] [PubMed] [Google Scholar]
  • 355.Zordoky BN, Aboutabl ME, El-Kadi AO. Modulation of cytochrome P450 gene expression and arachidonic acid metabolism during isoproterenol-induced cardiac hypertrophy in rats. Drug Metab Dispos. 2008d;36:2277–86. doi: 10.1124/dmd.108.023077. [DOI] [PubMed] [Google Scholar]
  • 356.Althurwi HN, Elshenawy OH, El-Kadi AO. Fenofibrate modulates cytochrome P450 and arachidonic acid metabolism in the heart and protects against isoproterenol-induced cardiac hypertrophy. J Cardiovasc Pharmacol. 2014;63:167–77. doi: 10.1097/FJC.0000000000000036. [DOI] [PubMed] [Google Scholar]
  • 357.Aboutabl ME, Zordoky BN, Hammock BD, El-Kadi AO. Inhibition of soluble epoxide hydrolase confers cardioprotection and prevents cardiac cytochrome P450 induction by benzo(a)pyrene. J Cardiovasc Pharmacol. 2011;57:273–81. doi: 10.1097/FJC.0b013e3182055baf. [DOI] [PubMed] [Google Scholar]
  • 358.Aboutabl ME, Zordoky BN, El-Kadi AO. 3-methylcholanthrene and benzo(a)pyrene modulate cardiac cytochrome P450 gene expression and arachidonic acid metabolism in male Sprague Dawley rats. Br J Pharmacol. 2009;158:1808–19. doi: 10.1111/j.1476-5381.2009.00461.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 359.Elkhatali S, El-Sherbeni AA, Elshenawy OH, Abdelhamid G, El-Kadi AO. 19-Hydroxyeicosatetraenoic acid and isoniazid protect against angiotensin II-induced cardiac hypertrophy. Toxicol Appl Pharmacol. 2015;289:550–9. doi: 10.1016/j.taap.2015.10.003. [DOI] [PubMed] [Google Scholar]
  • 360.Zordoky BN, El-Kadi AO. Modulation of cardiac and hepatic cytochrome P450 enzymes during heart failure. Curr Drug Metab. 2008;9:122–8. doi: 10.2174/138920008783571792. [DOI] [PubMed] [Google Scholar]
  • 361.Zordoky BN, Anwar-Mohamed A, Aboutabl ME, El-Kadi AO. Acute doxorubicin cardiotoxicity alters cardiac cytochrome P450 expression and arachidonic acid metabolism in rats. Toxicol Appl Pharmacol. 2010;242:38–46. doi: 10.1016/j.taap.2009.09.012. [DOI] [PubMed] [Google Scholar]
  • 362.Zeng Q, Han Y, Bao Y, Li W, Li X, Shen X, Wang X, Yao F, O’Rourke ST, Sun C. 20-HETE increases NADPH oxidase-derived ROS production and stimulates the L-type Ca2+ channel via a PKC-dependent mechanism in cardiomyocytes. Am J Physiol Heart Circ Physiol. 2010;299:H1109–17. doi: 10.1152/ajpheart.00067.2010. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 363.Moreno MU, San Jose G, Pejenaute A, Landecho MF, Diez J, Beloqui O, Fortuno A, Zalba G. Association of phagocytic NADPH oxidase activity with hypertensive heart disease: a role for cardiotrophin-1? Hypertension. 2014;63:468–74. doi: 10.1161/HYPERTENSIONAHA.113.01470. [DOI] [PubMed] [Google Scholar]
  • 364.Steinberg SF. Cardiac actions of protein kinase C isoforms. Physiology. 2012;27:130–9. doi: 10.1152/physiol.00009.2012. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 365.Dorn GW, 2nd, Force T. Protein kinase cascades in the regulation of cardiac hypertrophy. J Clin Invest. 2005;115:527–37. doi: 10.1172/JCI24178. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 366.Shinomura T, Asaoka Y, Oka M, Yoshida K, Nishizuka Y. Synergistic action of diacylglycerol and unsaturated fatty acid for protein kinase C activation: its possible implications. Proc Natl Acad Sci U S A. 1991;88:5149–53. doi: 10.1073/pnas.88.12.5149. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 367.Zhao H, Qi G, Han Y, Shen X, Yao F, Xuan C, Gu Y, Qian SY, Zeng Q, O’Rourke ST, Sun C. 20-Hydroxyeicosatetraenoic Acid Is a Key Mediator of Angiotensin II-induced Apoptosis in Cardiac Myocytes. J Cardiovasc Pharmacol. 2015;66:86–95. doi: 10.1097/FJC.0000000000000248. [DOI] [PubMed] [Google Scholar]

RESOURCES