• Non ci sono risultati.

Role of arachidonic acid lipoxygenase

N/A
N/A
Protected

Academic year: 2021

Condividi "Role of arachidonic acid lipoxygenase"

Copied!
14
0
0

Testo completo

(1)

doi:10.1152/ajpheart.00349.2009

You might find this additional information useful...

168 articles, 92 of which you can access free at:

This article cites

http://ajpheart.physiology.org/cgi/content/full/297/2/H495#BIBL

including high-resolution figures, can be found at:

Updated information and services

http://ajpheart.physiology.org/cgi/content/full/297/2/H495

can be found at:

AJP - Heart and Circulatory Physiology

about

Additional material and information

http://www.the-aps.org/publications/ajpheart

This information is current as of July 31, 2009 .

Physiological Society, 9650 Rockville Pike, Bethesda MD 20814-3991. Copyright © 2005 by the American Physiological Society. intact animal to the cellular, subcellular, and molecular levels. It is published 12 times a year (monthly) by the American

lymphatics, including experimental and theoretical studies of cardiovascular function at all levels of organization ranging from the publishes original investigations on the physiology of the heart, blood vessels, and AJP - Heart and Circulatory Physiology

on July 31, 2009

ajpheart.physiology.org

(2)

Role of arachidonic acid lipoxygenase metabolites in the regulation of

vascular tone

Yuttana Chawengsub, Kathryn M. Gauthier, and William B. Campbell

Department of Pharmacology and Toxicology, Medical College of Wisconsin, Milwaukee, Wisconsin Submitted 10 April 2009; accepted in final form 11 June 2009

Chawengsub Y, Gauthier KM, Campbell WB. Role of arachidonic acid lipoxygenase

metabolites in the regulation of vascular tone. Am J Physiol Heart Circ Physiol 297: H495–H507, 2009. First published June 12, 2009; doi:10.1152/ajpheart.00349.2009.— Stimulation of vascular endothelial cells with agonists such as acetylcholine (ACh) or bradykinin or with shear stress activates phospholipases and releases arachidonic acid (AA). AA is metabolized by cyclooxygenases, cytochrome P-450s, and lipoxygenases (LOs) to vasoactive products. In some arteries, a substantial com-ponent of the vasodilator response is dependent on LO metabolites of AA. Nitric oxide (NO)- and prostaglandin (PG)-independent vasodilatory responses to ACh and AA are reduced by inhibitors of LO and by antisense oligonucleotides specifically against 15-LO-1. Vasoactive 15-LO metabolites derived from the vascular endothelium include 15-hydroxy-11,12-epoxyeicosatrienoic acid (15-H-11,12-HEETA) that is hydrolyzed by soluble epoxide hydrolase to 11,12,15-trihydroxyeicosatrienoic acid (11,12,15-THETA). HEETA and THETA are endo-thelium-derived hyperpolarizing factors that induce vascular relaxations by activa-tion of smooth muscle apamin-sensitive, calcium-activated, small-conductance K⫹ channels causing hyperpolarization. In other arteries, the LO metabolite 12-hydroxyeicosatetraenoic acid is synthesized by the vascular endothelium and relaxes smooth muscle by large-conductance, calcium-activated K⫹channel acti-vation. Thus formation of vasodilator eicosanoids derived from LO pathways contributes to the regulation of vascular tone, local blood flow, and blood pressure. endothelium-derived hyperpolarizing factors; vascular relaxation; 15-lipoxygenase; 12-lipoxygenase; trihydroxyeicosatrienoic acids; hydroxyepoxyeicosatrienoic acids

VASCULAR ENDOTHELIAL CELLSregulate the tone of the underlying

vascular smooth muscle by releasing various relaxing and con-tracting factors. This was first demonstrated by the seminal work of Furchgott and Zawadzki (51) describing an endothelium-derived relaxing factor (EDRF). In response to chemical or phys-ical stimuli such as acetylcholine (ACh), thrombin, bradykinin, and fluid sheer stress, the vascular endothelium produces several factors that relax the underlying smooth muscle and thus regulate the vascular tone (25, 72, 97, 112). These relaxing factors are prostacyclin (PGI2), nitric oxide (NO), and

endo-thelium-derived hyperpolarizing factors (EDHFs). Unlike PGI2

and NO, numerous endothelium-derived factors function as EDHFs, and no single molecule or pathway has been identified that exhibits all of the characteristics of EDHF in different vascular beds, species, and diseases (19, 20, 22, 36, 41, 62, 90, 168). Among the compounds and pathways that have been proposed as mediators of EDHF activity are metabolites of arachidonic acid (AA).

Cytochrome P-450 (CYP) epoxygenases metabolize AA to epoxyeicosatrienoic acids (EETs). In the last decade, EETs have been shown by numerous laboratories to function as EDHFs in blood vessels from different species, including humans (19, 20, 44, 83). However, in rabbit aorta and

mesen-teric arteries as well as arteries of other species including dogs, rats, and pigs, EET production is absent under normal condi-tions (33, 93, 117, 166, 168). Yet, AA produces EDHF-like relaxations, suggesting that AA metabolites, other than the EETs, mediate the EDHF activity. These AA metabolites have been identified as products of the lipoxygenase (LO) pathways: the 15-LO-1 metabolites hydroxyepoxyeicosatrienoic acids (HEETAs) and their soluble epoxide hydrolase (sEH) hydro-lytic products, the trihydroxyeicosatrienoic acids (THETAs) (21, 119, 120), and the 12-LO metabolite 12-hydroxyeicosa-tetraenoic acid (12-HETE) (93, 168). These LO pathways also occur in arteries of mice (55, 148), rats (93, 157), rabbits (21, 120), pigs (168), dogs (33, 94, 159), and humans (48, 82) and contribute to relaxation. The purpose of this review is to summa-rize the evidence that LO metabolites function as EDHFs in some arteries. Also, the mechanisms underlying vasorelaxation, the biosynthetic pathway, and the physiological and pathophysio-logical implications of alterations in the expression and activity of vascular 15-LO-1 are reviewed.

Endothelium-Derived Hyperpolarizing Factors

Bolton and colleagues (8, 9) first described endothelium-dependent hyperpolarization of vascular smooth muscle cells by a cholinergic agonist. They suggested that an endothelial factor might be responsible for this activity. Subsequent studies confirmed these findings and showed that this activity was due to an endothelium-derived transferable factor other than NO or

Address for reprint requests and other correspondence: W. B. Campbell, Dept. of Pharmacology and Toxicology, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226 (e-mail: wbcamp@mcw.edu).

on July 31, 2009

ajpheart.physiology.org

(3)

PGI2 (25, 40, 77, 78, 156). The term EDHF was first

intro-duced by Chen et al. (25). They demonstrated the presence of ACh-induced, endothelium-dependent hyperpolarizations of vascular smooth muscle that were associated with relaxation. These hyperpolarizations and relaxations to ACh occurred despite inhibition of NO activity and PGI2 synthesis. They

concluded that ACh stimulated the endothelium to release an endogenous factor, in addition to PGI2 and NO, and they

termed this factor EDHF. Subsequently, a number of investi-gators extended these findings, showing that relaxations to other agonists, cyclic stretch, and flow also were inhibited, but not blocked, by inhibition of NO and/or PGI2production (30,

31, 95, 125). These treatments also induced endothelium-dependent hyperpolarization of smooth muscle cells, implicat-ing EDHF as a mediator in their action.

ACh-induced hyperpolarizations are mediated by K⫹ chan-nel activation. Relaxations and hyperpolarizations to agonists are blocked by high extracellular K⫹ concentrations ([K⫹]o)

and K⫹ channel blockers; however, vessels differ in their sensitivity to blockers (20, 24, 26, 53, 115, 161, 169). Tetra-ethylammonium chloride, iberiotoxin, and charybdotoxin, but not glibenclamide, block relaxations to ACh in the coronary artery, whereas a combination of charybdotoxin and apamin are required for rat mesenteric and hepatic arteries. These data suggest that EDHF acts via Ca2⫹-dependent K(K

Ca)

chan-nels that are blocked by tetraethylammonium and charybdo-toxin or KCachannels sensitive to the combination of

charyb-dotoxin and apamin. ACh-induced hyperpolarizations and re-laxations of the rabbit mesenteric artery are inhibited by apamin alone (100, 165).

The activity termed EDRF is mediated by a single com-pound: NO (112). In contrast, there is no single compound mediating EDHF activity but, rather, a family of endothelial factors that act by a common mechanism of K⫹ channel activation, smooth muscle membrane hyperpolarization, and relaxation (19, 28, 41, 62). Members of this family are chem-ically different and activate different K⫹ channels. Hence, a compound is considered an EDHF when 1) it produces vasore-laxation in the presence of cyclooxygenase (COX) and NO synthase (NOS) inhibitors that is blocked by a high [K⫹]oor by

K⫹ channel inhibitors and 2) it produces endothelium-depen-dent hyperpolarization of vascular smooth muscle.

The chemical identity of EDHFs remains the focus of intense investigation. Several compounds and pathways medi-ate EDHF activity, including K⫹, AA metabolites, residual NO, hydrogen peroxide, C-type natriuretic peptide, and myoendothelial cell gap junctions (20 –22, 27, 36, 62, 90). The presence of these compounds and pathways is not exclusive but can coexist in certain vascular beds (165).

Endothelial Signaling Pathways

An increase in free intracellular Ca2⫹ concentration ([Ca2⫹]

i) in the endothelial cells is a crucial step in the

production and release of NO, PGI2, and EDHFs (74, 107).

Upon stimulation of muscarinic receptors, membrane-associ-ated phospholipase C is activmembrane-associ-ated, resulting in the production of inositol 1,4,5-trisphosphate (IP3) and diacylglycerol (DAG)

(68, 153). Binding of IP3to its receptor releases Ca2⫹from the

endoplasmic reticulum. The increase in the [Ca2⫹]i from the

intracellular stores opens KCachannels, causing K⫹efflux and

hyperpolarization of the cell membrane, which increases the electrochemical driving force for Ca2⫹ influx through nonse-lective, Ca2⫹-permeable cation channels (68, 76, 87, 107).

Endothelial intracellular free Ca2⫹ binds to calmodulin, which activates the constitutively expressed endothelial NOS (eNOS). NOS catalyzes the conversion of L-arginine to L

-citrulline and NO (15, 86). Once formed, NO diffuses to the underlying smooth muscle and causes relaxation.

Elevated endothelial cell [Ca2⫹]i also activates cytosolic

phospholipase A2to release free AA from the cell membrane

by cleavage of the sn-2 bond of phosphatidylcholine and phosphatidylethanolamine (11). Free AA also can be derived from the phospholipase C pathway. DAG lipase converts DAG to 2-arachidonoylglycerol, from which free AA is released by monoacylglycerol lipase or fatty acid amidohydrolase (153). Free AA is metabolized by COX to PGI2or by LOs and CYPs

to AA-derived EDHFs. AA metabolites are then released, diffuse to the smooth muscle, and cause hyperpolarizations and relaxations (Fig. 1).

Role of Lipoxygenase Metabolites of AA in Endothelium-Dependent Relaxation

ACh induces potent endothelium-dependent vascular relax-ations in most species, including rats, mice, cows, pigs, guinea pigs, and rabbits (28, 41). Several factors and pathways are proposed to mediate these relaxations to ACh. NO mediates part of the ACh-induced vascular relaxation, since the vascular responses are reduced, but not blocked, by NOS inhibitors. The contribution of PGI2 to ACh relaxation is variable. Several

lines of evidence indicate that AA metabolites mediate endo-thelium-dependent vascular hyperpolarizations and a compo-nent of the endothelium-dependent relaxations to ACh and other stimuli. Furchgott and Zawadzki (51) first suggested that relaxations to ACh in the rabbit aorta were mediated by LO metabolites of AA. Endothelium-dependent relaxations to ACh were inhibited by the LO inhibitor eicosatetraynoic acid and the phospholipase A2 inhibitor meparcrine but not by COX

inhibitors, aspirin, or indomethacin. The involvement of AA and its LO metabolites in ACh-induced relaxations was con-firmed in arteries from a variety of vascular beds and species [skeletal muscle arterioles and mesenteric arteries of mice (55, 148), aorta and mesenteric arteries of rats (93, 157), aorta and mesenteric arteries of rabbits (21, 120, 165), coronary arteries of pigs (168), femoral and coronary arteries of dogs (33, 94, 133), and coronary arteries of humans (48, 82)]. All of these studies used pharmacological inhibitors to determine the con-tribution of AA metabolites. A large number of structurally different LO inhibitors, including nordihydroguaiaretic acid (NDGA), phenidone, BW755c, caffeic acid, diethylcarbam-azine, AA861, TKM777, cinnanyl-dihydroxy-cyanocinnamate, ebselen, and others (21, 33, 45, 47, 50, 94, 116, 146), as well as inhibitors of phospholipases A2and C (21, 33, 46, 85, 146,

153), inhibited relaxations to ACh. Interestingly, inhibitors of CPY also inhibited the endothelium-dependent relaxations to ACh (21, 45, 116, 147). Bradykinin, histamine, substance P, thrombin, and other agonists caused endothelium-dependent relaxations that were inhibited or blocked by inhibitors of phospholipases and LOs (33, 45, 47, 94, 158).

Moncada et al. (98) and Fo¨rstermann et al. (45) questioned the mechanism of inhibition by LO inhibitors and thus the role

on July 31, 2009

ajpheart.physiology.org

(4)

of LO metabolites as mediators of vascular relaxations. Endo-thelium-dependent relaxations to several agonists were inhib-ited by LO inhibitors; however, this inhibition was reversed by superoxide dismutase and oxidized cytochrome c, which inac-tivate or bind superoxide anion, respectively (98). Moncada et al. suggested that LO inhibitors block LO activity but also generate superoxide anion, which inactivates EDRF. This latter action was proposed as the primary mechanism for LO inhib-itors for reduced endothelium-dependent relaxations. In a sub-sequent study, superoxide dismutase did not reverse the activ-ity of all LO inhibitors that block ACh-induced relaxations (94). Despite the studies to the contrary, these findings ques-tioned the specificity of LO inhibitors and the role of LO metabolites as mediators of ACh-induced relaxations. These concerns slowed the search and delayed the discovery of endothelium-derived LO metabolites that act as EDHFs.

Furchgott and Zawadzki (51) first showed that endothelial cells produce a diffusible, transferable relaxing factor. A de-tector vessel without endothelium did not relax to ACh; how-ever, if a donor vessel with endothelium was placed next to the detector vessel, the detector relaxed. Other investigators have developed variations on this bioassay method by using either superfused cultured endothelial cells on beads or perfused arteries with an intact endothelium as the relaxing factor source, i.e., the donor (12, 56, 63, 69, 88, 116, 132, 133). The donor perfusate was used to superfuse or perfuse a detector artery without endothelium. Changes in tension or diameter were measured in the detector artery. In all cases, ACh or

bradykinin added to the donor caused relaxation of the detector artery, but the agonists were without effect when added to detector directly or when added to a donor artery without endothelium. These results confirmed the existence of an endothelium-derived, transferable relaxing factor. Addition of NDGA to inhibit LO, metyrapone to inhibit CYP, or mepacrine to inhibit phospholipase to the donor inhibited agonist-induced relaxation of the detector artery (63, 88, 133). These studies also provided evidence for the existence of other endothelial factors (12, 69, 133, 160). ACh when added to the donor caused a biphasic concentration-related relaxation of the de-tector artery. Inhibitors of LO, CYP, and phospholipases in-hibited the first phase of relaxations, occurring at low ACh concentrations, but not the second phase, occurring at high concentrations. Subsequent studies indicated that this factor was an EDHF, since ACh relaxations were inhibited by addi-tion of high [K⫹]o to the detector artery (88). Addition of

bradykinin to the donor caused membrane hyperpolarization of the smooth muscle cells of the detector artery (58, 96, 124). These studies suggest that endothelial cells release a transfer-able, hyperpolarizing factor that is a LO and/or CYP metabo-lite of AA.

Further evidence for a role of LO and CYP metabolites of AA was the ability of AA to induce endothelium-dependent vascular relaxations in arteries from various vascular beds of rabbits, dogs, rats, and pigs (33, 39, 50, 116, 131, 145, 147, 153, 166, 168). Relaxations to AA were inhibited by LO inhibitors and CYP inhibitors but not by phospholipase

inhib-Endothelium PLA2 AA COXs LOs PGI2 CYPs EETs AA Ca2+ Hyperpolarization Ca2+ CaM L-citrulline + NO L-Arg R IP3 ER K+ K+ Ca2+ CaM/ eNOS Acetylcholine Bradykinin (Agonists) DAG MAG HEETA THETA 12-HETE K+ Smooth muscle IP ? BKCa K+ ? SKCa K+ K+ KIR ATPase Na+ K+ sGC cGMP GTP AC ATP cAMP Hyperpolarization Relaxation K+ KCa Ca2+ KCa

Fig. 1. Schematic of the role of Ca2⫹ on relaxing factor production in vascular endothelial cells. Activation of mus-carinic receptors leads to the production of inositol 1,4,5-trisphosphate (IP3), which releases Ca2⫹from the

endoplas-mic reticulum (ER). Extracellular Ca2⫹can enter the endo-thelial cell through nonselective Ca2⫹-permeable cation channels or store-operated Ca2⫹channels, which are regu-lated by the ER Ca2⫹store. An increase in Ca2⫹ concentra-tion promotes K⫹efflux through activated Ca2⫹-dependent K⫹ (KCa) channels, causing membrane hyperpolarization,

which in turn increases Ca2⫹influx. Intracellular Ca2⫹binds to calmodulin (CaM) and activates nitric oxide synthase (NOS) and the production of nitric oxide (NO) or stimulates phospholipase A2(PLA2), which results in the release of

arachidonic acid (AA) from the cell plasma membrane. Free AA also can be obtained from the phospholipase C (PLC) pathway, in which diacylglycerol (DAG) is hydrolyzed at the sn-1 position by DAG lipase to monoacylglycerol (MAG), specifically 2-arachidonylglycerol (2-AG). MAG lipase or fatty acid amidohydrolase (FAAH) releases free AA from 2-AG. Free AA can be metabolized through cyclooxygenase (COX), cytochrome P-450 (CYP), and/or lipoxygenase (LO) pathways to produce relaxing factors. PGI2, prostacyclin; EET, epoxyeicosatrienoic acid; HEETA,

hydroxyepoxyeicosatrienoic acid; THETA, trihydroxyeico-satrienoic acid; HETE, hydroxyeicosatetraenoic acid; BKCa,

large-conductance KCachannel; SKCa, apamin-sensitive KCa

channel; Kir, inward rectifying K⫹ channel; AC, adenylyl

cyclase; sGC, soluble guanylate cyclase.

on July 31, 2009

ajpheart.physiology.org

(5)

itors, since AA bypasses the phospholipase step. Like ACh and bradykinin, addition of AA to a donor artery with endothelium caused relaxation of a detector artery without endothelium (116). This indicated that AA released a transferable relaxing factor. In rabbit aorta, AA also stimulated endothelium-depen-dent hyperpolarization of vascular smooth muscle that was blocked by LO inhibitors (21, 23, 57). Relaxations to AA also were inhibited by high [K⫹]o and blockade of K⫹ channels.

Together, these data emphasize the role of LO metabolites of AA in endothelium-dependent relaxations and EDHF activity of some arteries (50, 116, 145, 166).

Lipoxygenases

LOs are a family of non-heme iron-containing enzymes that dioxygenate polyunsaturated fatty acids to hydroperoxyl me-tabolites. Three major LO isoforms include 5-LO (127), 12-LO (163), and 15-LO (79), which correspond to the carbon posi-tion of AA oxygenaposi-tion. Depending on the isoform, each LO oxygenates AA to form a regio- and stereospecific hydroper-oxyeicosatetraenoic acid (HPETE). HPETE is unstable and can be reduced by peroxidases to the corresponding hydroxyeico-satetraenoic acid (HETE). All three LO isoforms are found in vascular endothelial cells, and their products have various vasoactive properties, including both vasodilation and vaso-constriction (89, 93, 168). The 5-LO pathway is involved in the regulation of inflammation, and 5-LO is the initial enzyme in the synthesis of leukotrienes. Few reports have evaluated the role of 5-LOs in vascular tone regulation. 5-LO products were involved in the endothelium-dependent, ACh-induced constric-tion of the hypertensive rat aorta through the activaconstric-tion of cysteinyl leukotriene receptors (84). Also, 5-HETE inhibits the production of PGI2in coronary artery endothelial cells,

indi-rectly contributing to overall vascular constriction (60). 5-LO activity was not observed in rabbit or mouse arteries, and 5-HETE and leukotrienes were not detected when these arteries were incubated with AA (50, 118). In contrast, the 12-LO and 15-LO pathways are important in vasorelaxation in various blood vessels of pigs, rats, rabbits, dogs, mice, and humans (39, 50, 55, 82, 93, 120, 121, 145, 148, 157, 168).

The 12-LO pathway metabolizes AA to a variety of products with numerous biological activities. Major products of this pathway are 12(S)-HETE, hydroxyepoxy-containing hepoxi-lins, and trihydroxy-containing trioxilins (109). 12(S)-HETE is produced by arteries and the vascular endothelium (93, 99, 117, 120, 168). The role of 12(S)-HETE as an EDHF was reported in rat mesenteric arteries (93), rat basilar arteries (39), porcine coronary microvessels (168), and human coronary arteries (82). In these vessels, 12(S)-HETE induced vascular smooth muscle hyperpolarization through activation of large-conductance KCa (BKCa) channels, resulting in relaxation. In

contrast, 12-HETE is a vasoconstrictor in dog renal arcuate arteries (89). 12(S)-HETE can induce vascular smooth muscle cell growth, matrix protein production (128), and migration (102), which may contribute to atherosclerosis, hypertension, and restenosis. Treatment of endothelial cells with 12(S)-HETE leads to increased ICAM-1 expression and monocyte binding (114, 129), suggesting this metabolite contributes to inflammation. 12(S)-HETE also may play a role in angiogen-esis by inducing endothelial cell proliferation and migration (105, 152).

12(S)-HPETE is converted to the bioactive 8-hydroxy-11,12-epoxyeicosatrienoic acid (hepoxilin A3) and inactive

10-hydroxy-11,12-epoxyeicosatrienoic acid (hepoxilin B3) by

hepoxilin A3 synthase activity of 12-LO (106, 111). The

biological actions of hepoxilin A3have been intensely studied

in neutrophils (35), pancreatic islets (110), and neuronal syn-apses (123). However, little is known about the effect of hepoxilins on the regulation of vascular tone. Hepoxilin A3

stimulates intracellular Ca2⫹ release (35) and increases Ca2⫹ transport across the cell membrane (34); therefore, biological actions of hepoxilin A3are, for the most part, Ca2⫹dependent.

In rat aorta and portal vein, hepoxilin A3did not have a direct

effect on the vascular tone but potentiated norepinephrine-induced vascular contraction in a Ca2⫹-dependent manner (80, 81). Rabbit aorta converted AA to 8,9,12-trihydroxyeicosatrie-noic acid (trioxilin C3) when 12-LO was supplied exogenously,

and trioxilin C3produced concentration-dependent relaxations

of rabbit aorta (121). The vascular actions of trioxilin A3or B3

are unknown.

In humans, there are two types of 15-LO: reticulocyte type (15-LO-1) (144) and epidermis type (15-LO-2) (14). 15-LO-1 was first described in rabbit reticulocytes (139). The enzyme is a 75-kDa single-polypeptide chain with a two-domain structure (59). It contains one non-heme iron per molecule. 15-LO-2 was subsequently identified, and its expression was reported in human prostate, skin, and cornea (14). Both enzymes convert AA to 15(S)-HPETE. They share only 40% amino acid homol-ogy. There are two major differences between enzymes:

1) 15-LO-1 converts AA to 15(S)-HPETE (90%) and lesser

amounts of 12(S)-HPETE (10%), whereas 15-LO-2 produces exclusively 15(S)-HPETE (14), and 2) although both AA and linoleic acid are preferred substrates for 15-LO-1 (73, 137), only AA is a substrate for 15-LO-2 (14).

Tang et al. (154) demonstrated the expression of 15-LO-1, but not 12-LO, mRNA and protein in rabbit aorta. Histologi-cally, 15-LO is localized to the vascular endothelium (2, 155), and there is no metabolism of AA to LO products in arteries with the endothelium removed (2, 4). The kinetic characteris-tics of aortic 15-LO-1 are similar to those of purified reticulo-cyte 15-LO, supporting the conclusion that 15-LO-1 is respon-sible for the 15-LO activity in arteries (154). 15-LO-1 metab-olizes AA to the chemically unstable 15-HPETE, which is reduced to its stable metabolite, 15-HETE. Supporting a vaso-dilatory role of 15-LO metabolites of AA, Uotila et al. (157) reported that endothelium-denuded rat aorta incubated with soybean 15-LO relaxes to AA. AA alone does not relax the aorta in the absence of the endothelium (33, 116, 145). On basal tone, 15-HETE and 15-HPETE cause slight relaxations in lower concentrations but contractions in higher concentrations (91, 157, 159). Thromboxane A2receptor antagonists block the

contractions to these LO metabolites. 15-HETE also causes concentration-dependent contraction of pulmonary arterial rings from rabbits exposed to hypoxia but not normoxia (167). In norepinephrine- or PGF2␣-precontracted vessels, 15-HPETE

causes endothelium-dependent relaxations that are not affected by COX inhibition. 15-HPETE does not relax arteries precon-tracted with KCl. However, 15-HETE does not cause relax-ation of precontracted arteries (49, 117, 166). These observa-tions that 15-HPETE causes relaxation of precontracted arter-ies, but 15-HETE does not, suggest that some other metabolite

on July 31, 2009

ajpheart.physiology.org

(6)

of 15-HPETE and the 15-LO pathway must mediate the relax-ations to AA under these conditions.

Pharmacological LO inhibitors are nonspecific and block 5-LO, 12-LO, 15-LO, and possibly other pathways of AA metabolism (136). Many of the LO inhibitors also function as antioxidants (135). A more specific method of reducing 15-LO activity involved decreasing 15-LO-1 expression with anti-sense oligonucleotides (154). This decreased the production of LO metabolites by rabbit aorta and decreased the NO- and PGI2-independent relaxations to ACh (154). On the other hand,

overexpression of 15-LO-1 with adenoviruses containing hu-man 15-LO-1 cDNA increased the endothelial expression of the enzyme, increased the arterial production of LO metabo-lites, and enhanced relaxations to AA (3, 4). Together, these data suggest an important role of 15-LO-1 in the regulation of vascular tone.

Identification of 15-LO Metabolites

After the incubation of arteries with AA, metabolites were resolved by reverse-phase HPLC and coeluted with authentic THETAs, HEETAs, and 15-HETE (118, 120, 166) (Fig. 2). A similar pattern of AA metabolism was observed in arteries from rabbits, mice, dogs, and guinea pigs. No metabolism of AA was observed in arteries without endothelium. The THETA, HEETA, and HETE production in rabbit arteries was blocked by LO inhibitors and enhanced by indomethacin (119, 120). Biological THETA and HEETA peaks that were col-lected from HPLC relaxed preconstricted rabbit aortic rings (120). The biological HETE peak was without activity. Also, 15-HETE had no vasodilatory effect (49, 117, 166).

The THETAs produced by rabbit aorta were identified as 11,12,15- and 11,14,15-THETA (120). 11,12,15-THETA re-laxed preconstricted mesenteric arteries and aorta in an endo-thelium-independent manner, and the relaxations were blocked by high [K⫹]o and by apamin (21, 165). 11,12,15-THETA

relaxed rabbit aorta and hyperpolarized rabbit aortic smooth muscle cells through the opening of apamin-sensitive KCa

(SKCa) channels (21, 57). In contrast, 11,14,15-THETA was

inactive. The stereochemical configuration of 11,12,15-THETA was identified by comparing the vascular activities and chromatographic migration times of the biologically pro-duced 11,12,15-THETA to eight chemically synthesized ste-reoisomers of 11,12,15-THETA (54). 11(R),12(S),15(S)-TH-5(Z),8(Z),13(E)-ETA was the only stereoisomer that comi-grated with the biological 11,12,15-THETA in several chromatographic systems. In addition, it was the only stereo-isomer that relaxed the rabbit aorta and activated SKCa

chan-nels. These findings indicate that 11(R),12(S),15(S)-TH-5(Z),8(Z),13(E)-ETA is the endothelium-derived active THETA. It also suggests that 11,12,15-THETA is the active isomer that mediates NO- and PGI2-independent relaxations to AA and

ACh (21, 57).

15-Hydroxy-11,12-epoxyeicosatrienoic acid (15-H-11,12-EETA) was proposed as a biologically active precursor of 11,12,15-THETA (120). 15-H-11,12-EETA has an epoxide group in conjugation with a double bond and is rapidly con-verted to 11,12,15- and 11,14,15-THETA under acidic condi-tions. The half-life of 15-H-11,12-EETA in aqueous solution at pH 3.0 and pH 7.4 is ⬃50 s and 33 h, respectively (66). To access the formation of the acid labile 15-H-11,12-EETA, aortic incubations with AA were terminated by acidification in an excess of methanol to trap the 15-H-11,12-EETA as the methoxy derivatives 12-methoxy-11,15-dihydroxy- and 14-methoxy-11,15-dihydroxyeicosatrienoic acids (MDHEs) (23). Pretreatment of aortic tissue with the sEH inhibitor 12-(3-adamantan-1-yl-ureido)-dodecanoic acid (AUDA) increased the formation of 15-H-11,12-EETA, measured as MDHEs, and decreased THETA production (23). Thus 15-H-11,12-EETA is a LO metabolite of AA and is metabolized by sEH to 11,12,15-THETA. 0 200 400 600 800 0 1000 2000 3000 4000 0 1000 2000 3000 4000 5000 0 5 10 15 20 25 30 35 0 5 10 15 20 25 30 35 0 5 10 15 20 25 30 35 0 5 10 15 20 25 30 35 40 0 2000 4000 6000 8000 Dog Guinea Pig Mouse Rabbit M P C M P C Fraction Fraction Fraction Fraction THETA THETA THETA THETA HEETA HEETA HEETA HEETA HETEs HETEs HETEs HETEs AA AA AA AA EETs EETs EETs EETs

Fig. 2. Metabolism of [14C]AA by rabbit aorta, mouse

aorta, dog mesenteric arteries, and guinea pig carotid arteries. Arterial rings with an intact endothelium were incubated with indomethacin and [14C]AA.

Metabo-lites were extracted and resolved by reverse-phase HPLC (21). Migration times of known standards are indicated by arrows above the chromatogram. CPM, counts per minute.

on July 31, 2009

ajpheart.physiology.org

(7)

15-LO Biosynthetic Pathways

Pace-Asciak et al. (109) first reported that the rat lung produced 12-LO HEETA metabolites from AA. 12(S)-HPETE, formed by oxygenation of AA by 12-LO, undergoes reduction that is catalyzed by peroxidases to 12(S)-HETE or isomeriza-tion by hepoxilin synthase to hepoxilin A3and B3. Hepoxilins

are hydrolyzed by an epoxide hydrolase to the corresponding 8,11,12-THETA (trioxilin A3) and 10,11,12-THETA (trioxilin

B3) (109). 15-LO-1 is the major LO expressed in rabbit aorta

and other arteries (154), and it does not produce hepoxilins or trioxilins (121). Based on the biosynthetic pathway of hepoxi-lins and trioxihepoxi-lins, the 15-LO pathway in rabbit vascular endothelium was postulated (120). AA is first converted to HPETE by 15-LO-1. HPETE is reduced to 15(S)-HETE or metabolized to 15-H-11,12-EETA by a hydroperox-ide isomerase. The epoxhydroperox-ide group of 15-H-11,12-EETA is hydrolyzed by sEH and/or acid to 11,12,15-THETA.

Bifurcation of the 12-LO pathway occurs at the metabolism of HPETEs to either 12-HETE or hepoxilins. Cytosolic gluta-thione peroxidase (GPx) and membrane-bound phospholipid hydroperoxide GPx prevented the production of hepoxilins by shifting 12(S)-HPETE conversion to 12(S)-HETE (150). Inac-tivation of the GPx activity inhibited the formation of 12(S)-HETE by 80%, increased the accumulation of 12(S)-HPETE by two orders of magnitude, and increased hepoxilin produc-tion (150). The impact of GPx inhibiproduc-tion on the vascular synthesis or vascular effects of 12-HETE has not been inves-tigated. This concept was tested in rabbit aorta to examine the role of GPxs on the regulation of HEETA production in 15-LO pathway. When GPxs were inhibited by depleting cellular gluta-thione with diethylmaleate, an increase in THETA and HEETA production was accompanied by a decrease in 15(S)-HETE pro-duction. This suggests a role of GPxs in regulating HEETA and THETA production.

In addition to the reduction of HPETEs to HETEs, several enzymes, collectively named hydroperoxide isomerases, me-tabolize fatty acid hydroperoxides to an array of products, including epoxy alcohols such as HEETAs. Transformation of an unsaturated fatty acid hydroperoxide containing the 1-hy-droperoxy-2(E),4(Z)-pentadiene partial structure like that of HPETEs into epoxy alcohols occurs by two distinct pathways. These pathways differ with respect to the mode of formation and the origin of the epoxide group. One pathway involves homolytic cleavage of the hydroperoxide oxygen-oxygen bond, producing an alkoxyl radical that undergoes cyclization to an epoxide. The hydroxyl oxygen is derived from the terminal hydroperoxide oxygen, from O2, or from the solvent

(52). The other pathway involves heterolytic cleavage of the hydroperoxide oxygen-oxygen bond and formation of an ep-oxide group by epoxidation of the double bonds. In this pathway, the epoxide group is derived from the terminal hydroperoxide oxygen, which can be transferred to the double bonds intra- or intermolecularly (65). The identity of hydroper-oxide isomerase(s) that converts 15(S)-HPETE to HEETAs in arteries is unknown. However, from the chemical structures of the HEETAs, the mechanism of the synthesis and the enzymes involved can be speculated. The hydroperoxyl oxygen-oxygen bond of 15-HPETE can be cleaved heterolytically by a per-oxygenase to form a hydroxyl group at the C-15 position. An epoxide group can then be formed by epoxidation of the 11,12

cis-double bonds, resulting in 15-H-11,12-EETA formation.

The fact that 15-H-13,14-EETA has not been detected in arteries supports the role of a peroxygenase, since peroxygen-ases only epoxidize cis-, but not trans-, double bonds (7). CYP2J2, CYP2J5, CYP2J9, and CYP2C8 have been shown to be hydroperoxide isomerases in 11,12,15-THETA synthesis (122). Both CYP2J2 and CYP2C8 are present in rabbit aorta. Inhibitors of CYP2J, but not CYP2C, decrease THETA syn-thesis. In addition, other CYPs also have peroxygenase activity (92). Therefore, a CYP2J isoform in rabbit aorta may mediate the peroxygenase activity and 15-H-11,12-EETA formation. On the other hand, homolytic cleavage of the hydroperoxyl oxygen-oxygen bond will produce an alkoxyl radical, which can cyclize to form the 14,15-epoxide. Hydroxylation at the C-11 position results in 11-H-14,15-EETA formation. How-ever, direct evidence of 11-H-14,15-EETA formation has not been observed in rabbit aorta.

Once formed, HEETAs can be hydrolyzed to their corre-sponding THETAs. The epoxide group of 15-H-11,12-EETA is hydrolyzed by sEH and/or acid to 11,12,15-THETA (23). sEH is present in rabbit aorta and other arteries (18, 23, 38). The nonspecific hydrolysis of 15-H-11,12-EETA by acidic condi-tions will produce both 11,12,15-THETA and 11,14,15-THETA.

There is no information about the metabolism of 11,12,15-THETA by vascular cells. Because there are strict structural and stereochemical requirements for vascular activity (54), metabolism of 11,12,15-THETA would result in a loss of activity. Many eicosanoids and prostanoids with a 15-hydroxyl group are inactivated by 15-hydroxyprostaglandin dehydroge-nase, which converts the hydroxyl to a ketone, and subsequent reduction of the ⌬13,14-olefin by a reductase (6, 67). ␤- and

␻-oxidation are other inactivation pathways (29, 75). It is not known whether 11,12,15-THETA is a substrate for these en-zymes. The proposed endothelial biosynthetic pathway of HEETAs and THETAs is described in Fig. 3.

Roles of THETA and HEETAs in the Regulation of Vascular Tone

The roles of these metabolites as EDHFs were studied using synthetic analogs. 11(R),12(S),15(S)-TH-5(Z),8(Z),13(E)-ETA re-laxed the preconstricted arteries of rabbits and mice (54, 55). These relaxations were blocked by apamin. With the use of whole cell patch clamp, 11(R),12(S),15(S)-TH-5(Z),8(Z),13(E)-ETA in-creased outward K⫹ current in isolated rabbit aortic smooth muscle cells (54). This current was also blocked by apamin. These studies suggest that 11(R),12(S),15(S)-TH-5(Z),8(Z),13(E)-ETA is an EDHF. The requirements for a specific structure and stere-ochemical configuration for 11,12,15-THETA suggest that a spe-cific binding site or receptor is involved in mediating the SKCa

channel activation, hyperpolarization, and vascular relaxations. In this regard, there are examples of membrane receptors for other trihydroxylated AA metabolites. Lipoxin A4, a trihydroxy

metab-olite of AA, also has strict structural requirements for activity, and its membrane receptor has been identified (43, 64).

The biological activity of 15-H-11,12-EETA requires further study. Because 15-H-11,12-EETA is easily hydrolyzed (23, 66), a stable analog is required to directly study its biological activity. At this time, such an analog is not available; therefore, there are no direct studies of its biological activity or

on July 31, 2009

ajpheart.physiology.org

(8)

isons of its activity to 11,12,15-THETA. However, by using the sEH inhibitor AUDA to prevent the hydrolysis of HEETAs to THETAs, the role of endogenous 15-H-11,12-EETA on the vascular tone has been indirectly tested. ACh- and AA-induced relaxations of rabbit aorta were enhanced by pretreatment with AUDA, and these enhanced responses to both agonists were blocked by the LO inhibitor NDGA and by high [K⫹]o(23).

AA caused hyperpolarization of smooth muscle in endothelium-intact segments of aorta. The AA-induced hyperpolarizations also were enhanced by AUDA, and the enhanced hyperpolar-izations were inhibited by NDGA. Thus the enhanced relax-ations by sEH inhibition were mediated by a LO metabolite, K⫹channel activation, and membrane hyperpolarization. This vasoactive HEETA is probably 15-H-11,12-EETA, since its production by rabbit aorta is enhanced by sEH inhibition. The HEETA causes relaxation by K⫹ channel activation, K⫹ ef-flux, and membrane hyperpolarization, since increasing [K⫹]o

inhibits the AUDA-enhanced relaxations to AA and ACh (23). The ability of AUDA to increase vascular relaxation and hyperpolarization of smooth muscle suggests that the HEETA is more potent than the THETA. Nevertheless, these data must be interpreted with caution, since it is not known whether AUDA completely inhibited sEH under the experimental con-ditions used. The presence of THETA could complicate this interpretation.

In rabbit arteries, 11,12,15-THETA and 15-H-11,12-EETA both are produced by the endothelium and activate smooth muscle apamin-sensitive SKCa channels to cause membrane

hyperpolarization and relaxation (23, 54, 57, 165, 166). This raises the question of how two structurally different AA me-tabolites activate the same SKCa channel, particularly when

there is a strict structural requirement for 11,12,15-THETA (54). SKCachannels are voltage independent and activated by

submicromolar concentrations of intracellular Ca2⫹(10). The

gating of SKCa channels is induced by Ca2⫹ binding to

cal-modulin, which is constitutively bound to each channel sub-unit. Ca2⫹ binding to calmodulin induces a conformational change, which leads to the opening of the channels (162). Therefore, 11,12,15-THETA and 15-H-11,12-EETA may not activate the channel directly but, instead, bind to their own receptors/binding sites and activate the same or different sig-naling pathways that open the SKCachannel.

SKCaand intermediate-conductance KCa(IKCa) channels are

expressed on the vascular endothelium (16, 17, 37, 41, 104, 134). Specifically, endothelial cells express the SK2 and SK3 channel subtypes along with IK channels. SKCachannels of the

SK2 subtype and BKCachannels are found on smooth muscle

cells and are likely involved in smooth muscle hyperpolariza-tion. Murphy and Brayden (100) reported in rabbit mesenteric arteries that the EDHF-mediated hyperpolarization was blocked by the SKCa inhibitor apamin and by other inhibitors

of the apamin-sensitive K⫹ channel but not by the BKCa

inhibitor iberiotoxin. Electrophysiological studies on vascular smooth muscle cells from rabbit aortas characterized a 24-pS-conductance K⫹ channel that is voltage and Ca2⫹ dependent and inhibited selectively by apamin (57). However, the subtype of this SKCa channel was not determined. Therefore, further

characterization of the K⫹channels, their regulation, the sub-types, and their cellular localization in the vasculature is needed.

LO Metabolites and Other EDHFs

In addition to AA metabolites, a small increase in [K⫹]o

hyperpolarizes the smooth muscle cell membrane and causes relaxation (36, 104, 126). As discussed above, a crucial step in the EDHF pathway is a rise in endothelial [Ca2⫹]

i, which is

followed by the activation of endothelial IKCa and/or SKCa

channels, allowing K⫹ efflux and hyperpolarization of the endothelial cell membrane. The K⫹ released by the endothe-lium into the myoendothelial intercellular space activates smooth muscle inward rectifier K⫹(Kir) channels and/or Na⫹

-K⫹-ATPase, causing smooth muscle hyperpolarization and relaxation in rat hepatic arteries (36). This observation is counterintuitive, since increasing [K⫹]owill change the

equi-librium potential of K⫹, as predicted by the Nernst equation, and will favor smooth muscle membrane depolarization and contraction. However, the activation of Kir and the Na⫹-K⫹

ATPase by small increases in [K⫹]o(1 to 15 mM) overcomes

the minor depolarizing effects caused by the increase in [K⫹]o.

The net result is hyperpolarization and thus relaxation of the smooth muscle cells. Therefore, endothelium-derived K⫹may mediate the EDHF response (36).

In rabbit mesenteric arteries, the corelease of endothelial cell K⫹and AA metabolites of the 15-LO pathway synergistically function as EDHFs (165) (Fig. 4). With the use of whole cell patch clamp, ACh increased the outward K⫹currents in endo-thelial cells. This outward current was blocked by charybdo-toxin, an IKCachannel blocker. Thus ACh increases endothe-OH OH O O OH O AA OH O OOH 15(S)-HPETE 15(S)-HETE OH O OH OH O OH HO OH 11,12,15-THETA OH O HO HO OH 11,14,15-THETA 15-LO-I sEH / H+ peroxidase Hydroperoxide isomerase 15-H-11,12-EETA H+

Fig. 3. Proposed biosynthesis pathway of HEETAs and THETAs. AA is con-verted to 15(S)-hydroperoxyeicosatetraenoic acid [15(S)-HPETE] by 15-LO-1. 15(S)-HPETE is converted by hydroperoxide isomerases to the HEETAs. 15-Hydroxy-11,12-epoxyeicosatrienoic acid (15-H-11,12-EETA) can be hydro-lyzed by either soluble epoxide hydrolase (sEH) or acid to THETAs. It is hydrolyzed to 11,12,15-THETA by sEH. However, with acid hydrolysis, 15-H-11,12-EETA is hydrolyzed to both 11,12,15- and 11,14,15-THETA. The exact stereochemistry of 15-H-11,12-EETAs is not known.

on July 31, 2009

ajpheart.physiology.org

(9)

lial K⫹ release by activating charybdotoxin-sensitive IKCa

channels. The relaxations to ACh were partially inhibited by apamin alone or charybdotoxin alone but were blocked by the combination. Barium blocked the smooth muscle cell Kir

channels that were activated by K⫹and inhibited a portion of ACh-induced relaxations. The combination of barium and apamin, like the combination of charybdotoxin and apamin, blocked the relaxations, indicating that they act in parallel on different pathways. In contrast, the combination of barium and charybdotoxin was only as effective as barium alone and charybdotoxin alone, indicating that these inhibitors act in series on the same pathway. These studies indicate that ACh stimulates endothelial IKCa channels to release K⫹ that

acti-vates Kir channels on smooth muscle cells. This mediates a

portion of the hyperpolarization and relaxation response to the agonist. In parallel, ACh also stimulates the endothelial syn-thesis of 11,12,15-THETA (21). The THETA activates apamin-sensitive SKCachannels on smooth muscle to mediate

a portion of the hyperpolarization and relaxation to ACh. Increasing the [K⫹]ofrom 5 to 10.9 mM caused partial

relax-ation of the artery and potentiated the relaxrelax-ations to AA and 11,12,15-THETA (165). These studies indicate that ACh stim-ulates the corelease of K⫹ and 11,12,15-THETA, and these mediators act synergistically via different mechanisms to cause hyperpolarization and relaxation. It is not known whether a similar interaction occurs with other EDHFs such as 12-HETE or EETs.

Role of LO Metabolites in Regulating Vascular Tone In Vivo

The role of LO metabolites on blood pressure regulation has been studied in normotensive and hypertensive animals. In normotensive rats and cholesterol-fed, normotensive rabbits, inhibition of LOs with phenidone, baicalein, or BW755c did not affect basal blood pressure or heart rate (2, 149). Similarly, the basal blood pressure was identical in wild-type and 12/ 15-LO knockout mice (5). Treatment of rabbits in vivo with an

adenovirus containing the human 15-LO-1 cDNA increased the expression of the enzyme in the vascular endothelium and the vascular synthesis of 15-LO metabolites of AA (3). However, overexpression of 15-LO did not alter basal blood pressure or heart rate. These studies indicate that augmenting or inhibiting endothelial LOs does not change basal blood pressure.

LO metabolites contribute to ACh-mediated hypotension. In normal rabbits, ACh decreased blood pressure in a concentra-tion-related manner (1–3). Treatment with inhibitors of COX and NOS reduced, but did not block, the ACh-induced hypo-tension (1). The non-PG, non-NO-mediated hypohypo-tension was inhibited by apamin and blocked by the combination of charyb-dotoxin and apamin. Thus ACh-induced hypotension has PG, NO, and EDHF components as described in isolated arteries (1, 21, 165). These hypotensive responses to ACh were signifi-cantly enhanced in rabbits overexpressing endothelial 15-LO-1 due to adenovirus treatment, age, or cholesterol treatment (1–3). ACh-induced hypotension was attributed to 15-LO me-tabolites of AA acting as EDHFs, since the LO inhibitor BW755c and K⫹ channel blocker apamin inhibited the re-sponses to a similar extent. Similar results were obtained with wild-type mice and mice with genetic hypercholesterolemia (148). Although LO metabolites of AA do not contribute to basal blood pressure, they mediate a component of the hypo-tension to ACh.

In hypertensive models, the effects of LO inhibitors are complex. Stern et al. (149) first showed that the LO inhibitors phenidone, esculetin, and baicalein attenuate the contractile effects of angiotensin II in femoral arterial rings of rats and decrease the pressor response to angiotensin II. Responses to phenylephrine were unaltered by LO inhibition. These in vivo and in vitro findings were confirmed in a study using wild-type and 12/15-LO knockout mice (5). These data indicate that a vascular LO metabolite must mediate or modulate specifically angiotensin-induced vasoconstriction. This role of vascular smooth muscle LO differs from the role of endothelial LO and the synthesis of EDHFs. Similarly, LO inhibitors reduce blood pressure in rats with renovascular hypertension (103, 108), aortic coarctation-induced hypertension (32), spontaneous hy-pertension (138), and fructose-induced hyhy-pertension (61). However, in DOCA-salt hypertension, in which the renin-angiotensin system is suppressed, the LO inhibitor phenidone was without effect on blood pressure (108). Two mechanisms have been proposed for LO mediating or regulating angioten-sin-induced vasoconstriction. Angiotensin II increases 12-HETE production by smooth muscle, and 12-12-HETE increases [Ca2⫹]

i in smooth muscle (108, 138, 149). These findings

imply that 12-HETE is an intracellular mediator of angiotensin vasoconstriction. In contrast, Nasjletti and colleagues (32, 103) have shown that 12-HPETE inhibits PGI2synthase, decreasing

the production of PGI2. As a result, less PGI2 is available to

antagonize angiotensin-induced vasoconstriction. Inhibition of 12-HPETE production with LO inhibitors restores PGI2

syn-thase activity, and PGI2antagonizes the action of angiotensin.

Despite the mechanism or combination of mechanisms in-volved, these studies indicate that LO inhibitors reduce blood pressure in renin-dependent hypertension by reducing angio-tensin-induced vasoconstriction. Additional studies are needed to determine the role of LO-derived EDHFs in hypertension.

EC Hyperpolarization and Relaxation AA 11,12,15-THETA SMC IKCa SKCa Kir ACh K+ PL Phospho-lipids 15-LO Apamin ChTX Barium Mepacrine AA861, BW755c, caffeic acid, CDC, diethylcarbamazine, ebselen, ETYA, NDGA, phenidone, TKM777

Fig. 4. Endothelium-derived hyperpolarizing factor (EDHF)-mediated re-sponses to acetylcholine (ACh) involve 2 mechanisms acting in parallel. ACh activates intermediate-conductance KCa(IKCa) channels on endothelial cells

(EC), releasing K⫹that activates Kirchannels on smooth muscle. ACh also

releases AA in endothelial cells that is metabolized by 15-LO to 15-H-11,12-EETA and 11,12,15-THETA. THETA activates SKCa channels on smooth

muscle. These 2 pathways cause smooth muscle cell membrane hyperpolar-ization and relaxation. Inhibitors of these enzymes and channels are indicated. PL, phospholipase; CDC, cinnamyl-3,4-dihydroxy-␣-cyanocinnamate; ETYA, 5,8,11,14-eicosatetraynoic acid; NDGA, nordihydroguaiaretic acid; ChTX, charybdotoxin; SMC, smooth muscle cells.

on July 31, 2009

ajpheart.physiology.org

(10)

Summary, Perspective, and Future Direction

15-LO-1 is expressed in a wide variety of human cells. The biological role of 15-LO-1, however, is unclear and sometimes contradictory. 15-LO-1 mediates inflammatory monocyte/en-dothelial interactions and atherosclerosis (129). On the other hand, the 15-LO-1-derived lipoxins are anti-inflammatory (142). The major AA metabolite from 15-LO-1 pathway is 15(S)-HETE. However, in some experimental conditions, mi-cromolar concentrations of 15(S)-HETE are required to ob-serve its activities (91, 159, 167). This concentration is un-likely a physiological concentration and suggests that other metabolites mediate the biological actions of 15-LO-1. In addition to 15(S)-HETE, the 15-LO-1 pathway also produces lipoxins (141), eoxins (42), 8(S),15(S)-diHETE (143), 5-oxo-15-hydroxy-6,8,11,13-eicosatetraenoic acid (140), HEETAs (23), and THETAs (21, 120), which possess several biological activities. The physiological or pathophysiological importance of 15-LO-1 has been shown by knockdown or overexpression studies (4, 5, 154). The HEETAs and THETAs may represent potential mediators of the actions of 15-LO-1.

15-LO metabolites function as novel EDHFs in both conduit arteries and resistance arteries (3, 21, 120, 165, 166). It is generally accepted that in large conduit arteries, such as the aorta, the contribution of EDHF to vascular relaxation is small (41, 71, 101). In contrast, in resistance arterioles (diameter ⬍100 ␮m), which play a critical role in blood pressure control, EDHF is a predominant relaxing influence. The findings reviewed above are in agreement with this concept. PGI2 and PGE2 are

active in mesenteric arteries (166), whereas in the aorta, they do not contribute to vasodilation (116). In conduit arteries such as rabbit aorta, 11(R),12(S),15(S)-TH-5(Z),8(Z),13(E)-ETA is the major EDHF that mediates non-NO, non-PGI2, ACh-induced

relaxations and hyperpolarizations (21). In contrast, 11,12,15-THETA, possibly 15-H-11,12-EETA, and K⫹ all function as EDHFs in resistance arteries such as rabbit mesenteric arteries (165). 11,12,15-THETA and K⫹act synergistically in this vascu-lar bed to cause relaxation.

It is not uncommon for both HEETAs and their hydrolysis products, the THETAs, to have similar biological effects. Hep-oxilin and its four triHep-oxilin hydrolysis products activate peroxi-some proliferator-activated receptor-␣ (PPAR␣) with a compara-ble efficiency (164). Therefore, the redundancy of vasodilatory functions of both 15-H-11,12-EETA and 11,12,15-THETA was not unexpected. Although the vascular activity of these 15-LO metabolites is the first to be characterized, it is likely that other biological functions of HEETAs and THETAs have yet to be discovered. In fact, we observed that 11(R),12(S),15(S)-THETA inhibits migration of aortic smooth muscle cells.

Cardiovascular disease is the major cause of death in the United States (130). EDHF plays an important role in cardio-vascular physiology and pathology in various animal models as well as in humans (28, 41). For example, in patients with essential hypertension, the EDHF system becomes active when there is a defect in the endothelium-derived NO (113, 151), suggesting its role as an important mechanism to maintain endothelium-dependent vasodilation. In isolated aorta, mesen-teric arteries, and renal arteries of hypercholesterolemic rab-bits, an enhanced contribution of EDHF compensates for the decrease in NO-mediated relaxation (13, 70). Given this bio-logical importance of EDHF, characterization of new members

of the EDHF family will broaden our understanding, set a new direction for the research, and may lead to the development a new therapeutic target for the treatment of cardiovascular diseases.

ACKNOWLEDGMENTS

We thank Gretchen Barg for secretarial assistance.

GRANTS

These studies were supported by National Heart, Lung, and Blood Institute Grant HL-37981. Y. Chawengsub was supported by a predoctoral fellowship from the American Heart Association (Greater Midwest Affiliate), and K. M. Gauthier was supported by an American Heart Association Grant-in-Aid.

REFERENCES

1. Aggarwal N, Gauthier KM, Campbell WB. 15-Lipoxygenase metab-olites contribute to age-related reductions in acetylcholine-induced hy-potension in rabbits. Am J Physiol Heart Circ Physiol 295: H89 –H96, 2008.

2. Aggarwal N, Pfister SL, Campbell WB. Hypercholesterolemia en-hances 15-lipoxygenase mediated vasorelaxation and acetylcholine-in-duced hypotension. Arterioscler Thromb Vasc Biol 28: 2209 –2215, 2008.

3. Aggarwal NT, Chawengsub Y, Gauthier KM, Viita H, Yla-Herttuala

S, Campbell WB. Endothelial 15-lipoxygenase-1 overexpression

in-creases acetylcholine-induced hypotension and vasorelaxation in rabbits. Hypertension 51: 246 –251, 2008.

4. Aggarwal NT, Holmes BB, Cui L, Viita H, Yla-Herttuala S,

Camp-bell WB. Adenoviral expression of 15-lipoxygenase-1 in rabbit aortic

endothelium: role in arachidonic acid-induced relaxation. Am J Physiol Heart Circ Physiol 292: H1033–H1041, 2007.

5. Anning PB, Coles B, Bermudez-Fajardo A, Martin PWM, Levison

BS, Hazen SL, Funk CD, Kuhn H, O’Donnell VB. Elevated

endothe-lial nitric oxide bioactivity and resistance to angiotensin-dependent hypertension in 12/15-lipoxygenase knockout mice. Am J Pathol: 653– 662, 2005.

6. Bergholte JM, Soberman RJ, Hayes R, Murphy RC, Okita RT. Oxidation of 15-hydroxyeicosatetraenoic acids and other hydroxy fatty acids by lung prostaglandin dehydrogenase. Arch Biochem Biophys 257: 444 – 450, 1987.

7. Blee E, Wilcox AL, Marnett LJ, Schuber F. Mechanism of reaction of fatty acid hydroperoxides with soybean peroxygenase. J Biol Chem 268: 1708 –1715, 1993.

8. Bolton TB, Clapp LH. Endothelial-dependent relaxant actions of car-bachol and substance P in arterial smooth muscle. Br J Pharmacol 87: 713–723, 1986.

9. Bolton TB, Lang RJ, Takewaki T. Mechanisms of action of noradren-aline and carbachol on smooth muscle of guinea-pig anterior mesenteric artery. J Physiol 351: 549 –572, 1984.

10. Bond CT, Maylie J, Adelman JP. Small-conductance calcium-activated potassium channels. Ann NY Acad Sci 868: 370 –378, 1999.

11. Bonventre JV. Phospholipase A2and signal transduction. J Am Soc

Nephrol 3: 128 –150, 1992.

12. Boulanger C, Hendrickson H, Lorenz RR, Vanhoutte PM. Release of different relaxing factors by cultured porcine endothelial cells. Circ Res 64: 1070 –1078, 1989.

13. Brandes RP, Behra A, Lebherz C, Boger RH, Bode-Boger SM,

Phivthong-Ngam L, Mugge A. NG-nitro-L-arginine- and

indomethacin-resistant endothelium-dependent relaxation in the rabbit renal artery: effect of hypercholesterolemia. Atherosclerosis 135: 49 –55, 1997. 14. Brash AR, Beoglin WE, Chang MS. Discovery of a second

15S-lipoxygenase in humans. Proc Natl Acad Sci USA 94: 6148 – 6152, 1997. 15. Bredt DS, Snyder SH. Isolation of nitric oxide synthetase, a

calmodulin-requiring enzyme. Proc Natl Acad Sci USA 87: 682– 685, 1990. 16. Burnham MP, Bychkov R, Feletou M, Richards GR, Vanhoutte PM,

Weston AH, Edwards G. Characterization of an apamin-sensitive

small-conductance Ca2⫹-activated K⫹ channel in porcine coronary artery endothelium: relevance to EDHF. Br J Pharmacol 135: 1133–1143, 2002.

17. Bychkov R, Burnham MP, Richards GR, Edwards G, Weston AH,

Feletou M, Vanhoutte PM. Characterization of a

charybdotoxin-sensi-tive intermediate conductance Ca2⫹-activated K⫹ channel in porcine

on July 31, 2009

ajpheart.physiology.org

(11)

coronary endothelium: relevance to EDHF. Br J Pharmacol 137: 1346 – 1354, 2002.

18. Campbell WB, Deeter C, Gauthier KM, Ingraham RH, Falck JR, Li

PL. 14,15-Dihydroxyeicosatrienoic acid relaxes bovine coronary arteries

by activation of KCachannels. Am J Physiol Heart Circ Physiol 282:

H1656 –H1664, 2002.

19. Campbell WB, Falck JR. Arachidonic acid metabolites as endothelium-derived hyperpolarizing factors. Hypertension 49: 590 –596, 2007. 20. Campbell WB, Gebremedhin D, Pratt PF, Harder DR. Identification

of epoxyeicosatrienoic acids as endothelium-derived hyperpolarizing factors. Circ Res 78: 415– 423, 1996.

21. Campbell WB, Spitzbarth N, Gauthier KM, Pfister SL. 11,12,15-Trihydroxyeicosatrienoic acid mediates acetylcholine-induced relax-ations in the rabbit aorta. Am J Physiol Heart Circ Physiol 285: H2648 – H2656, 2003.

22. Chauhan SD, Nilsson H, Ahluwalia A, Hobbs AJ. Release of C-type natriuretic peptide accounts for the biological activity of endothelium-derived hyperpolarizing factor. Proc Natl Acad Sci USA 100: 1426 – 1431, 2003.

23. Chawengsub Y, Aggarwal NT, Nithipatikom K, Gauthier KM,

An-jaiah S, Hammock BD, Falck JR, Campbell WB. Identification of

15-hydroxy-11,12-epoxyeicosatrienoic acid as a vasoactive 15-lipoxy-genase metabolite in rabbit aorta. Am J Physiol Heart Circ Physiol 294: H1348 –H1356, 2008.

24. Chen G, Suzuki H. Some electrical properties of the endothelium-dependent hyperpolarization recorded from rat arterial smooth muscle cells. J Physiol 410: 91–106, 1989.

25. Chen G, Suzuki H, Weston AH. Acetylcholine released endothelium-derived hyperpolarizing factor and EDRF from rat blood vessels. Br J Pharmacol 95: 1165–1174, 1988.

26. Chen G, Yamamoto Y, Miwa K, Suzuki H. Hyperpolarization of arterial smooth muscle induced by endothelial humoral substances. Am J Physiol Heart Circ Physiol 260: H1888 –H1892, 1991.

27. Cohen RA, Plane F, Najibi S, Huk I, Malinski T, Garland CJ. Nitric oxide is the mediator of both endothelium-dependent relaxation and hyperpolarization of the rabbit carotid artery. Proc Natl Acad Sci USA 94: 4193– 4198, 1997.

28. Cohen RA, Vanhoutte PM. Endothelium-dependent hyperpolarization: beyond nitric oxide and cyclic GMP. Circulation 92: 3337–3349, 1995. 29. 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 de-hydrogenase 4. J Biol Chem 280: 33157–33164, 2005.

30. Cowan CL, Cohen RA. Two mechanisms mediate relaxation by brady-kinin of pig coronary artery: NO-dependent and -independent responses. Am J Physiol Heart Circ Physiol 261: H830 –H835, 1991.

31. Cowan CL, Palacino JJ, Najibi S, Cohen RA. Potassium channel-mediated relaxation to acetylcholine in rabbit arteries. J Pharmacol Exp Ther 266: 1482–1489, 1993.

32. DelliPizzi A, Guan H, Tong X, Takizawa H, Nasjletti A. Lipoxygen-ase-dependent mechanisms in hypertension. Clin Exp Hypertens 22: 181–192, 2000.

33. DeMey JG, Claeys M, Vanhoutte PM. Endothelium-dependent inhib-itory effects of acetylcholine, adenosine triphosphate, thrombin and arachidonic acid in the canine femoral artery. J Pharmacol Exp Ther 222: 166 –173, 1982.

34. Derewlany LO, Pace-Asciak CR, Radde IC. Hepoxilin A, hydroxye-poxide metabolite of arachidonic acid, stimulates transport of45Ca across

the guinea pig visceral yolk sac. Can J Physiol Pharmacol 62: 1466 – 1469, 1984.

35. Dho S, Grinstein S, Corey EJ, Su WG, Pace-Asciak CR. Hepoxilin A3

induced changes in cytosolic calcium, intracellular pH and membrane potential in human neutrophils. Biochem J 266: 63– 68, 1990. 36. Edwards G, Dora KA, Gardener MJ, Garland CJ, Weston AH. K is

an endothelium-derived hyperpolarizing factor in rat arteries. Nature 396: 269 –272, 1998.

37. Eichler I, Wibawa J, Grgic I, Korr A, Brakemeier S, Priest AR,

Hoyer J, Kohler R. Selective blockade of endothelial Ca2⫹-activated small- and intermediate-conductance K⫹-channels suppresses EDHF-mediated vasodilation. Br J Pharmacol 138: 594 – 601, 2003.

38. Fang X, Kaduce TL, Weintraub NL, Harmon S, Teesch LM,

Moris-seau C, Thompson DA, Hammock BD, Spector AA. Pathways of

epoxyeicosatrienoic acid metabolism in endothelial cells: implications

for the vascular effects of soluble epoxide hydrolase inhibition. J Biol Chem 276: 14867–14874, 2001.

39. Faraci FM, Sobey CG, Chrissobolis S, Lund DD, Heistad DD,

Weintraub NL. Arachidonate dilates basilar artery by

lipoxygenase-dependent mechanism and activation of K⫹ channels. Am J Physiol

Regul Integr Comp Physiol 281: R246 –R253, 2001.

40. Feletou M, Vanhoutte PM. Endothelium-dependent hyperpolarization of canine coronary smooth muscle. Br J Pharmacol 93: 515–524, 1988. 41. Feletou M, Vanhoutte PM. Endothelium-derived hyperpolarizing fac-tor: where are we now? Arterioscler Thromb Vasc Biol 26: 1215–1225, 2006.

42. Feltenmark S, Gautam N, Brunnstrom A, Griffiths W, Backman L,

Edenius C, Lindbom L, Bjorkholm M, Claesson HE. Eoxins are

proinflammatory arachidonic acid metabolites produced via the 15-lipoxygenase-1 pathway in human eosinophils and mast cells. Proc Natl Acad Sci USA 105: 680 – 685, 2008.

43. Fiore S, Maddox JF, Perez HD, Serhan CN. Identification of a human cDNA encoding a functional high affinity lipoxin A4 receptor. J Exp Med 180: 253–260, 1994.

44. Fisslthaler B, Popp R, Kiss L, Potente M, Harder DR, Fleming I,

Busse R. Cytochrome P450 2C is an EDHF synthase in coronary arteries.

Nature 401: 493– 497, 1999.

45. Fo¨rstermann U, Alheid U, Frolich JG, Mulsch A. Mechanisms of action of lipoxygenase and cytochrome P-450-mono-oxygenase inhibi-tors in blocking endothelium-dependent vasodilation. Br J Pharmacol 93: 569 –578, 1988.

46. Forstermann U, Burgwitz K, Frolich JC. Effect of nonsteroidal phos-pholipase inhibitors and glucocorticoids on endothelium-dependent re-laxations of rabbit aorta induced by different agents. J Cardiovasc Pharmacol 10: 356 –364, 1987.

47. Fo¨rstermann U, Hertting G, Neufang B. The role of endothelial and nonendothelial prostaglandins in the relaxation of isolated blood vessels of the rabbit induced by bradykinin and acetylcholine. Br J Pharmacol 87: 521–532, 1986.

48. Forstermann U, Mugge A, Frolich JC. Endothelium-dependent relax-ations of human epicardial coronary arteries: frequent lack of effect of acetylcholine. Eur J Pharmacol 128: 277–281, 1986.

49. Forstermann U, Neufang B. The endothelium-dependent relaxation of rabbit aorta: effects of antioxidants and hydroxylated eicosatetraenoic acids. Br J Pharmacol 82: 765–767, 1984.

50. Fo¨rstermann U, Neufang B. The endothelium-dependent vasodilator effect of acetylcholine: characterization of the endothelial relaxing factor with inhibitors of arachidonic acid metabolism. Eur J Pharmacol 103: 65–70, 1984.

51. Furchgott RF, Zawadzki JW. The obligatory role of endothelial cells in the relaxation of arterial smooth muscle by acetylcholine. Nature 288: 373–376, 1980.

52. Gardner HW. Oxygen radical chemistry of polyunsaturated fatty acids. Free Radic Biol Med 7: 65– 86, 1989.

53. Garland CJ, McPherson GA. Evidence that nitric oxide does not mediate the hyperpolarization and relaxation to acetylcholine in the rat small mesenteric artery. Br J Pharmacol 105: 429 – 435, 1992. 54. Gauthier KM, Chawengsub Y, Goldman DH, Conrow RE, Anjaiah

S, Falck JR, Campbell WB.

11(R),12(S),15(S)-trihydroxyeicosa-5(Z),8(Z),13(E)-trienoic acid: an endothelium-derived 15-lipoxygenase metabolite that relaxes rabbit aorta. Am J Physiol Heart Circ Physiol 294: H1467–H1472, 2008.

55. Gauthier KM, Christian S, Goldman DH, Aggarwal N, Campbell

WB. Lipoxygenase metabolites of arachidonic acid mediate

acetylcho-line-induced relaxations of mouse arteries (Abstract). Hypertension 52: e55, 2008.

56. Gauthier KM, Edwards EM, Falck JR, Reddy DS, Campbell WB. 14,15-Epoxyeicosatrienoic acid represents a transferable endothelium-dependent relaxing factor in bovine coronary arteries. Hypertension 45: 666 – 671, 2005.

57. Gauthier KM, Spitzbarth N, Edwards EM, Campbell WB. Apamin-sensitive K⫹ currents mediate arachidonic acid-induced relaxations of rabbit aorta. Hypertension 43: 413– 419, 2004.

58. Gebremedhin D, Harder DR, Pratt PF, Campbell WB. Bioassay of an endothelium-derived hyperpolarizing factor from bovine coronary arter-ies: role of a cytochrome P450 metabolite. J Vasc Res 35: 274 –284, 1998.

59. Gillmor SA, Villasenor A, Fletterick R, Sigal E, Browner MF. The structure of mammalian 15-lipoxygenase reveals similarity to the lipases

on July 31, 2009

ajpheart.physiology.org

Riferimenti

Documenti correlati

The study shows how quantitative spatial planning methods may support the selection of the sites of potential interest for the marine renewable energy sector in the perspective

compounds, catechin, chlorogenic acid, ferulic acid, vanillic acid, gallic acid, p-coumaric 57.. acid, 3,4-dihydroxyphenylacetic acid and tannic acid were purchased from Sigma (Milan,

Based on modal mineralogy and texture of the Aeolian Bronze Age pottery we recognised four statistically sep- arated groups (AI, AIV, AVIII and AX) among the vol- canic

Black curves passed successfully the selection test; red lines failed the selection test; black dashed line represents the reference regression curve.. qNMR analysis of

The primary end point was: the learning curve analysis for TAVI procedures performed by a single interven- tional team, aimed at detecting a correlation between experience and

The binding portion of our ITs is represented by a single-chain antibody fragment (scFv) directed against CD38 antigen, a surface molecule highly expressed by B

two study sites differing for tree age, stand density, slope and soil characteristics, on the basis of tree-ring growth and stable isotope ratio variations, (2) analyze the

In primo luogo, avendo a disposizione alcune conoscenze di base sia sullo sviluppo tipico che su quello atipico, le educatrici possono, come abbiamo sottolineato