Arachidonic acid metabolism in skin health and disease

18
Arachidonic acid metabolism in skin health and disease Lars Iversen*, Knud Kragballe Department of Dermatology, Marselisborg Hospital, University of Aarhus, DK-8000 Aarhus C, Denmark 1. Introduction Arachidonic acid (AA) (eicosa-5,8,11,14-tetraenoic acid, 20:4 v6) is a polyunsaturated fatty acid with 20 carbon atoms and 4 double bounds. It is either derived from dietary sources or synthesized by desaturation and elongation of linoleic acid (C18:2) (Fig. 1). Because the mammalian organism cannot introduce double bounds in the fatty acid structure closer to the v-end than v9 the AA precursors linoleic acid (18:2 v6) and g-linoleic acid (18:3 v3) are considered essential [1]. AA cannot be synthesized locally in the human epidermis because both d-6-desaturase and d-5-desaturase are absent in the epidermis [2]. AA present in the epidermis must therefore come from either dietary sources or it must be transported to the epidermis from other endogenous sources such as the liver, which is capable of elongation and desaturation. In the cell AA is stored in the membrane fraction, primarily esterified to phospholipids at the second carbon (sn-2) of the phospholipid glycerol backbone [3]. It is released from the phospholipids in the cell membrane by the action of phospholipases. Keratinocytes respond to skin irritation and injury by a rapid but transient activation of AA metabolism. An understanding of the enzymatic pathways involved in AA metabolism is therefore important. 2. Phospholipases Esterified AA is not amenable to metabolic transformations. Therefore, AA has to be released from the cell membrane by either the phospholipase A 2 (PLA 2 ) or by a combined action of phospholipase C (PLC) and a diglyceride lipase. PLA 2 catalyzes the hydrolysis of the sn-2 fatty acyl bond of phospholipids to liberate free fatty acids and either lysophos- * Corresponding author. Tel.: 145-89-49-3333; fax: 145-89-49-1870. Prostaglandins & other Lipid Mediators 63 (2000) 25– 42 0090-6980/00/$ – see front matter © 2000 Elsevier Science Inc. All rights reserved. PII: S0090-6980(00)00095-2

Transcript of Arachidonic acid metabolism in skin health and disease

Arachidonic acid metabolism in skin health and disease

Lars Iversen*, Knud Kragballe

Department of Dermatology, Marselisborg Hospital, University of Aarhus, DK-8000 Aarhus C, Denmark

1. Introduction

Arachidonic acid (AA) (eicosa-5,8,11,14-tetraenoic acid, 20:4v6) is a polyunsaturatedfatty acid with 20 carbon atoms and 4 double bounds. It is either derived from dietary sourcesor synthesized by desaturation and elongation of linoleic acid (C18:2) (Fig. 1). Because themammalian organism cannot introduce double bounds in the fatty acid structure closer to thev-end thanv9 the AA precursors linoleic acid (18:2v6) andg-linoleic acid (18:3v3) areconsidered essential [1]. AA cannot be synthesized locally in the human epidermis becauseboth d-6-desaturase andd-5-desaturase are absent in the epidermis [2]. AA present in theepidermis must therefore come from either dietary sources or it must be transported to theepidermis from other endogenous sources such as the liver, which is capable of elongationand desaturation. In the cell AA is stored in the membrane fraction, primarily esterified tophospholipids at the second carbon (sn-2) of the phospholipid glycerol backbone [3]. It isreleased from the phospholipids in the cell membrane by the action of phospholipases.

Keratinocytes respond to skin irritation and injury by a rapid but transient activation ofAA metabolism. An understanding of the enzymatic pathways involved in AA metabolismis therefore important.

2. Phospholipases

Esterified AA is not amenable to metabolic transformations. Therefore, AA has to bereleased from the cell membrane by either the phospholipase A2 (PLA2) or by a combinedaction of phospholipase C (PLC) and a diglyceride lipase. PLA2 catalyzes the hydrolysis ofthe sn-2 fatty acyl bond of phospholipids to liberate free fatty acids and either lysophos-

* Corresponding author. Tel.:145-89-49-3333; fax:145-89-49-1870.

Prostaglandins & other Lipid Mediators 63 (2000) 25–42

0090-6980/00/$ – see front matter © 2000 Elsevier Science Inc. All rights reserved.PII: S0090-6980(00)00095-2

pholipids or lyso-PAF depending on type of binding in the 1-position of the phospholipid(Fig. 2). The lysophospholipid may play a role in modulating or activating certain cells [4,5]and lyso-PAF may act as a precursor of platelet activating factor (PAF).

The PLA2’s are a diverse class of enzymes with regard to function, localization, regula-tion, mechanism, sequence and structure [6]. Thus, they carry out essentially the same

Fig. 1. Arachidonic acid synthesis.

Fig. 2. Phospholipase mediated processes.

26 L. Iversen, K. Kragballe / Prostaglandins & other Lipid Mediators 63 (2000) 25–42

reaction namely the hydrolysis of thesn-2 fatty acyl bond of phospholipids. Traditionallythree different human PLA2 classes have been described and characterized; the pancreaticPLA2 which is classified as digestive [7] and the two regulatory PLA2’s, the secretory (s)PLA2 [8] and the cytoplasmic (c) PLA2 [9]. Although recent evidence has revealed that thePLA2’s may be a much more diverse group of enzymes than previously believed [10] andthat the group designations will undoubtedly need to be adjusted, the designation mentionedabove will be used here.

Both the sPLA2 and the cPLA2 are present in human skin [11,12]. sPLA2’s consist of alarge isozyme family [13]. These 14 kDa isozymes are secretory enzymes and thereforemainly present extracellularly. They are devoid of specificity onsn-2fatty acid and requiremillimolar concentration of Ca21 for activation [14]. They are believed to be a key link inthe inflammatory process, e.g. as an effector system for inflammatory cytokines [15], andboth involved and un-involved psoriatic skin have been shown to contain higher levels ofsPLA2 than normal skin [11]. Several cytokines present in the skin have also been shown tomodulate the expression of sPLA2. IL-1b has been shown to induce expression and secretionof sPLA2 in a variety of cells [15], while IL-6 was shown to elevate sPLA2 levels in a humanhepatoma cell line [16]. In contrast TGF-b inhibits sPLA2 expression in rat mesangial cells[17].

cPLA2 has been purified from monocytic cell lines, U937 [18], and RAW264.7 [19], andthe cDNA coding for the enzyme was cloned from U937 cell cDNA library [20]. Themolecular weight is 85 kDa as determined by sequencing and cloning [20]. The cPLA2 istranslocated from the cytosol to the membrane fraction in a Ca21-dependent fashion [21] atphysiologically relevant Ca21 concentrations. The translocation to the membrane may be aregulatory process augmenting accessibility of the enzyme to the substrate. In contrast to thesPLA2, Ca21 does not play a catalytic role for the cPLA2, it seems only to play a role for theassociation of the enzyme with the membrane phospholipids. An interesting characteristic ofthe cPLA2 with respect to eicosanoid formation is its AA specificity [19,22], and data haveindicated that the cPLA2 is responsible for AA liberation in agonist-stimulated inflammatorycells. The involvement of AA-specific PLA2 in the production of PAF and eicosanoids from1-O-alkyl-glycerophosphocholine was suggested because both PAF and eicosanoid biosyn-thesis was abolished in polymorphonuclear leukocytes [23] and HL-60 cells [24] depleted ofAA.

The calcium requirement of PLA2 has led to speculations that PLC activation and theresulting IP3 and intracellular Ca21 elevation (Fig. 2) precede the PLA2 activation. However,several experiments have shown that PLA2 and PLC activation are regulated independentlyby different G proteins. Agonist induced AA release was shown to be sensitive to pertussistoxin treatment in mouse 3T3 fibroblasts [25] and a rat thyroid cell line, FRTL-5 [26],whereas PIP2 hydrolysis in the same cells were insensitive to toxin treatment.

In several studies PLA2 has been suggested to play an important role in inflammatory skindiseases. Intradermal injection of purified sPLA2 has been shown to induce cellular infil-tration, interstitial edema, vascular permeability and hyperemia [27]. Also, in psoriasis PLA2

has been held responsible at least in part for the elevated AA levels found, and elevated PLA2

activity has been demonstrated in psoriatic epidermis [28]. Furthermore, a recent report hasshown an increased lysophosphatidylcholine content in lesional psoriatic skin [29], support-

27L. Iversen, K. Kragballe / Prostaglandins & other Lipid Mediators 63 (2000) 25–42

ing the idea of increased PLA2 activity in inflammatory skin diseases. Investigations on howPLA2 activity is regulated have also been carried out. In the human epidermis PLA2 has beensuggested to be subject to a positive feedback regulation, as its activity was stimulated byPGE2 and PGF2a [30]. PLA2 activity may also be regulated by direct phosphorylatingactivities [31]. Furthermore, PLA2 activity may be regulated indirectly by phosphorylation-dephosphorylation of a soluble 35 kDa PLA2 inhibitory protein termed lipocortin [32]. It hasbeen suggested that upon phosphorylation, lipocortin loses its inhibitory properties, resultingin expression of PLA2 activity, and hyperphosphorylation of lipocortin has been suggestedas the reason for increased PLA2 activity in uninvolved psoriatic epidermis [33]. Also,glucocorticosteroids act by inhibition of PLA2 activity [34] via transcriptional control oflipocortin synthesis [35], and topical glucocorticosteroid application to skin results inreduction of PLA2 activity [36].

A second pathway for AA release is via breakdown of phosphatidylinositol-4,5-biphos-phate (PIP2) by the phosphoinositide-specific PLC. Hydrolysis of PIP2 generates diacylglyc-erol (DAG) and inositol-1,4,5-triphosphate (IP3) [37] (Fig. 2). DAG can be further degradedby diglyceride lipase to glycerol and free fatty acids, including AA [38] (Fig. 2). Also, PLCactivation is a primary event in intracellular signaling [39]. IP3 stimulate Ca21 mobilizationfrom intracellular stores and DAG activates protein kinase C (PKC) which orchestrates asignal transduction cascade that regulate cellular responses such as growth and differentia-tion.

PLC activity is present in most mammalian cells and tissues including human epidermis[39,40]. Epidermal PLC has been demonstrated as a Ca21 dependent enzyme with maximalactivity at pH 7.0. PLC activity in psoriatic plaque has been shown to be increased 188%compared to normal epidermis and may thus contribute to the elevated AA levels observedin this tissue [40].

At least two different mechanisms, phosphorylation of PLC by the tyrosine kinase familyand G protein-mediated PLC activation, appear to be involved in the regulation of PLCactivity [41,42]. Epidermal growth factor has been demonstrated to activate PLC by aug-menting tyrosine kinase activity [41]. The regulatory effect of tyrosine phosphorylation onPLC activity is not yet known. Increased catalytic activity due to tyrosine phosphorylationhas been reported [43], but it has also been suggested that phosphorylation increasesaccessibility of the PLC to substrate [44].

3. Cyclooxygenase

The initial step in AA metabolism by the cyclooxygenase pathway is the transformationof AA into prostaglandin H2 (PGH2) by the cyclooxygenase (COX) or PGH synthase. COXcatalyzes a two step reaction with insertion of two oxygen molecules at C11 and C15resulting in the formation of PGG2 [45]. The second step is a reductive cleavage at the15-hydroperoxy group to yield PGH2 [46]. COX is an iron containing dimer of 70 kDasubunits localized primarily in the endoplasmatic reticulum. COX is inhibited by non-steroidal antiinflammatory drugs such as aspirin and indomethacin. This inhibition largelyaccounts for the antiinflammatory and analgesic effects of these agents [47]. Recently a

28 L. Iversen, K. Kragballe / Prostaglandins & other Lipid Mediators 63 (2000) 25–42

second isoform of the COX has been cloned and sequenced [48,49]. The isoform designatedCOX-1 is constitutively expressed in cells whereas the isoform designated COX-2 seems torequire specific induction. In general, COX-1 regulates prostaglandin synthesis associatedwith cellular homeostasis whereas COX-2 is upregulated in inflammatory conditions andassociated with synthesis of proinflammatory prostaglandins [50]. Much attention has there-fore been paid to develop specific inhibitors of COX-2. Recently, normal murine epidermiswas found to express COX-1 but not COX-2. However, COX-2 could be induced either byacetone treatment or by topical application of the phorbol ester, TPA [51]. This is in contrastto normal human epidermis where COX-2 has been associated with keratinocyte differen-tiation [52]. In normal human skin, COX-1 immunostaining is observed throughout theepidermis whereas COX-2 immunostaining increases in the more differentiated, suprabasilarkeratinocytes [52]. Furthermore, inducing differentiation of cultured human keratinocytes byraising extracellular calcium leads to an increased expression of both COX-2 protein andmRNA. In contrast, no significant alteration in the expression of COX-1 was seen in responseto increased extracellular calcium [52]. COX-2 has also recently been implicated in thedevelopment of skin cancer [53].

PGH2 has been shown to be a central intermediate in prostaglandin biosynthesis byserving as substrate for several competing enzymes (Fig. 3). PGE synthase or PGH-PGEisomerase catalyzes the rearrangement of the endoperoxide group of PGH2 to produce PGE2.PGE synthase activity has been localized to the microsomal fraction and shown to requireglutathione as a co-factor [54]. As early as 1970 PGE2 formation in rat epidermis wasdemonstrated [55], and later rat [56] and human skin [57] were shown to synthesize PGE2.

Fig. 3. Cyclooxygenase pathway.

29L. Iversen, K. Kragballe / Prostaglandins & other Lipid Mediators 63 (2000) 25–42

PGE2 is the main AA cyclooxygenase product in human epidermis homogenates [58]. PGE2

formation has also been demonstrated to be important for keratinocyte differentiation. Incultured human keratinocytes induced to differentiate by increased extracellular calcium, anincreased PGE2 formation was observed 1 h after the calcium concentration was increased[59].

PGD2 is formed from PGH2 either enzymatically or non-enzymatically. Non-enzymati-cally transformation of PGH2 into PGD2 is catalyzed by serum albumin [60]. Also twospecific enzymes, PGD synthases have been demonstrated. PGD synthases have beenlocalized as cytoplasmic enzymes unlike most other enzyme involved in prostaglandinformation which is found predominantly in the microsomal fraction [61]. PGD2 formationhas been demonstrated in human skin [58] mouse epidermis, cultured neonatal mousekeratinocytes, rat skin [62] and in guinea pig skin homogenates [63]. Interestingly PGDsynthase has been shown to be present predominantly in the Langerhans cells in guinea pigand rat epidermis and not in the keratinocytes [62,64]. Furthermore, PGD synthase wasdemonstrated in the dermal macrophages and mast cells, and generally mast cells arebelieved to represent one of the major cellular sources of PGD2.

There are three possible pathways for the synthesis of PGF2a. First by a PGF synthasefrom PGH2 (Fig. 3) [65]. Second by a reduction of PGE2 catalyzed by a NADH-linked15-hydroxy-PG dehydrogenase [66] and third from PGD2 catalyzed by a PGD 11-ketoreductase [67]. PGF2a formation has been demonstrated in both rat and human skin [68].

Thromboxane A2 (TXA2) is formed from PGH2 catalyzed by TX synthase. TXA2 isextremely unstable and therefore, rapidly transformed by a non-enzymatic process into TXB2

[69]. Only very low TXA synthase activity has been found in the skin.Prostacyclin PGI2, like TXA2 is an unstable molecule which is rapidly further metabolized

non-enzymatically into 6-keto-PGF1a. The transformation of PGH2 into PGI2 is catalyzed byPGI synthase [70]. Very low amounts of PGF1a has been found in studies of AA metabolismin guinea pig skin homogenates, indicating that PGI2 formation can take place in at leastguinea pig skin [63].

4. Lipoxygenase

The initial product of AA made by lipoxygenases (LOs) is the (mono)-hydroperoxy-eicosatetraenoic acid (HPETE) (Fig. 4). 5-, 8-, 12- and 15-LOs introduce an oxygen moleculeinto the respective position of the arachidonic acid backbone giving rise to unstable HPETEs.A HPETE undergoes reduction to its corresponding hydroxy-eicosatetraenoic acid (HETE)and under certain circumstances further oxidation to diHETEs. Among the diHETEs of the5-LO pathway are the biologically active leukotrienes (LTs).

4.1. 5-Lipoxygenase

Fig. 4 shows the metabolism of AA by the 5-LO pathway. Once AA is liberated from thephospholipids, the 5-LO is activated in the presence of ATP and Ca21 [71] and translocatedfrom the cytoplasm to the plasma membrane [72] by a Ca21 dependent mechanism.

30 L. Iversen, K. Kragballe / Prostaglandins & other Lipid Mediators 63 (2000) 25–42

Activated 5-LO is always membrane associated [71,73] and recently a novel membraneassociated 5-LO activating protein (FLAP) has been described [74]. FLAP has been purifiedfrom rat neutrophil membranes [74] and cloned from rat basophil leukemia cell and humanHL-60 cDNA libraries [75]. It has been identified as a 18 kDa protein [73]. So far all 5-LOexpressing cells investigated have been shown to contain FLAP. Also, transfection experi-ments have demonstrated that both FLAP and 5-LO must be present in order to transform AAinto 5-HPETE [75]. 5-HPETE is then further metabolized by the 5-LO into LTA4 [71,76] ortransformed, either enzymatically by a glutathione-dependent peroxidase or non-enzymati-cally into 5-HETE [77,78]. The transformation of AA into LTA4 results in suicide inacti-vation of the 5-LO [72].

The end product of the 5-LO activity is LTA4, an unstable allylic intermediate [79,80] thatcan be further metabolized both enzymatically and non-enzymatically. Non-enzymatically,LTA4 is metabolized into 5,6-diHETEs and 5,12-diHETEs [81]. The epoxide hydrolase,LTA4 hydrolase, catalyzes the transformation of LTA4 into LTB4 [82], and this step has beenshown to be the rate-limiting step in LTB4 formation, at least in rat basophilic leukemia cells(RBL-1) and human neutrophils [83,84].

LTA4 may also be conjugated with glutathione by LTC4 synthase to yield LTC4 [79].Successive cleavage byg-glutamyl transferase [85], and a dipeptidase [86] converts LTC4

into LTD4 and LTE4. Together LTC4, LTD4 and LTE4 are termed peptidelukotrienes.Although LTs have been ascribed an important role in inflammatory skin diseases like

psoriasis and atopic dermatitis [87–90] the capacity of the human skin itself to biosynthesizethe LTs has been questioned. It has been reported that freshly isolated human epidermal cells[91,92] and cultured mouse keratinocytes [93] can synthesize low quantities of LTB4 asdetermined by HPLC [93] and by RIA and chemotactic activity [91,92]. This view wasrecently supported by Jassen-Timmen et al. [94]. They reported that undifferentiated kera-tinocytes did not express detectable 5-LO mRNA, 5-LO protein or activity. However,

Fig. 4. 5-, 8-, 12- and 15-lipoxygease pathways.

31L. Iversen, K. Kragballe / Prostaglandins & other Lipid Mediators 63 (2000) 25–42

inducing differentiation by shifting the culture conditions resulted in induction of 5-LO geneexpression. Thus, the induction of 5-LO expression was much more pronounced in keratin-ocytes derived from the HaCat cell line than in normal human keratinocytes. In contrast, inthe study by Brenton et al. [95] measurements of 5-LO protein determined by Western blotsor 5-LO mRNA determined by reverse transcriptase polymerase chain reaction (PCR)analysis in cultured human keratinocytes treated with either IL-1, 1,25-vitamin D3, interfer-on-g, PMA, A23187 or dexamethason did not demonstrate the presence of 5-LO in humankeratinocytes. Furthermore, FLAP was not present in subcellular fractions of these keratin-ocytes consistent with the absence of FLAP mRNA in these cells. The lack of 5-LO is inaccordance with previous reports from our laboratory [96–98] and others [99] showing nodetectable LTB4 formation in normal human cultured keratinocytes stimulated with A23187and in freshly isolated human epidermis. It is therefore likely, that the 5-LO activity in thehuman keratinocytes is very limited.

4.1.1. Transcellular leukotriene synthesisAlthough the epidermis cannot form leukotrienes itself from AA, it can contribute

significantly to leukotriene formation through transcellular leukotriene synthesis. By thismechanism, LTA4 formed in one cell type is released and then further metabolized in anothercell type (Fig. 5). Human cultured keratinocytes and human epidermis has been shown totransform neutrophil derived LTA4 into LTB4 in vitro [96–99]. The key enzyme in trans-cellular LTB4 formation in the epidermis is the LTA4 hydrolase (Fig. 5). The LTA4hydrolase has been localized in the human epidermis by activity determination [96–99], byinhibition of enzyme activity by bestatin and captopril (known LTA4 hydrolase inhibitors)[96–98], by Western blotting [98] and by immunohistochemical staining [100]. Furthermore,the LTA4 hydrolase has been purified and characterized from cultured human keratinocytesand human epidermis [101]. The epidermal LTA4 hydrolase was characterized as a 70 kDaenzyme, with a pI of 5.1–5.4, a pH optimum of 7.5–8.5 and it was localized in thecytoplasmic fraction [98]. The amino acid composition has been determined and all the dataobtained from human epidermis and cultured human keratinocytes have been consistent withdata obtained with LTA4 hydrolase from most other cell types that have been investigated[101]. An interesting characteristic of the LTA4 hydrolase is that it has a dual function

Fig. 5. Transcellular leukotriene synthesis.

32 L. Iversen, K. Kragballe / Prostaglandins & other Lipid Mediators 63 (2000) 25–42

exhibiting both hydrolase activity as well as peptidase activity. Peptidase activity has beenshown against small peptides including opioids [102,103]. Another characteristic feature ofthe LTA4 hydrolase is that it undergoes suicide inactivation when it transforms LTA4 intoLTB4. This has been demonstrated in several cell types [104] including human epidermis[100] and is due to covalent binding of LTA4 to the enzyme [105]. Presumably, as a resultof suicide inactivation, the LTA4 hydrolase has been shown as the rate limiting step in LTB4

formation at least in human neutrophils [84] and rat basophilic leukemia cells [83]. There-fore, transcellular LTA4 metabolism may result in an increased LTB4 formation at aninflammatory site. In inflammatory skin diseases like psoriasis, neutrophils migrate into theepidermis and get into close contact with the keratinocytes. Release of LTA4 into theextracellular space has previously been demonstrated by activated neutrophils, and veryrecently it was demonstrated that more than 50% of the LTA4 formed in neutrophils isreleased from the cell [106]. Transcellular leukotriene synthesis may therefore be an impor-tant mechanism by which the human epidermis can contribute significantly to LTB4 forma-tion in inflammatory skin diseases.

Similar to transcellular LTB4 synthesis, transcellular LTC4 synthesis has been demon-strated as a possible mechanism for LTC4 formation in the epidermis [97] (Fig. 5). LTA4 istransformed into LTC4 by a LTC4 synthase (Fig. 4), which in mouse mastocytoma cells hasbeen shown as a highly specific, membrane bound glutathione-S-transferase (GST) [107]. Inthe skin a specific LTC4 synthase has, however, never been shown. Several isoforms of GSTwith activity toward LTA4 have been identified in human and rodent skin [108], and human,rat and mouse skin has been demonstrated to transform LTA4-methyl ester into LTC4-methylester [109].

4.2. 8-Lipoxygenase

The 8-LO is one of the most recent discovered LOs in the skin. 8-LO activity is bestcharacterized in tissue samples from marine sources [110]. Only brief reports have beenpublished on the enzymatic production of 8-HETE in human tissues and cell types such ashuman leukocytes [111], human tracheal cells [112] and psoriatic skin [113]. However,recently 8-LO activity in mouse skin has been determined and characterized [114,115]. Theepidermal 8-LO was shown as a specific 8(S)-LO catalyzing the formation of only 8(S)-HPETE and 8(S)-HETE [114]. However, recently varying S to R ratios of 8-HETE in miceskin has been shown [116] indicating that in addition to 8(S)-LO other enzymes yet to bedefined may be involved in 8-HETE production. Enzyme activity was localized in thecytosolic fraction of the cells in the suprabasal compartment of the epidermis [115] andactivity was stimulated by phosphatidylcholine and lecithin whereas no requirement of ATP,calcium or NADPH was found [114,115]. In contrast to the 5-LO, 8-LO is not translocatedto the membrane when it is activated. Interestingly, only very low 8-LO activity has beendetermined in normal mouse epidermis indicating that the enzyme is not constitutivelyexpressed in the epidermis. However, a prominent dose-dependent induction of 8-LO activityhas been demonstrated in mouse epidermis after topical treatment with TPA [115]. Inter-estingly, mouse skin 8-LO has also been shown to exhibits leukotriene A synthase activitywhen incubated with 5-HPETE [117], and in contrast to a linear time course of the 8-LO

33L. Iversen, K. Kragballe / Prostaglandins & other Lipid Mediators 63 (2000) 25–42

reaction with AA, leukotriene A synthase activity seems to undergo suicide inactivation[117]. These data together with the fact that 8-HETE is present in psoriatic skin [113] suggestsome pathophysiological function of the 8-LO pathway in the skin. Interestingly, it hasrecently been reported that 8(S)-HETE is a high affinity ligand for the peroxisome prolif-erator-activated receptora (PPARa) whereas 8(R)-HETE was much less potent [118,119].PPARs are members of the nuclear receptor superfamily that includes receptors for thesteroid, thyroid and retinoid hormones. Through dimerization of the PPAR with the 9-cis-retinoic acid receptor (RXR) and activation of a PPAR response element, transcription ofspecific genes are regulated. It can be speculated that this is a possible pathway by which8(S)-HETE exerts some of its possible pathophysiological effects in the skin. It is certainlyan area which deserves further investigation in the future.

4.3. 12-Lipoxygenase

12-HETE has been shown as one of the main eicosanoids formed by the epidermis [58],and with the discovery of large quantities of 12-HETE in human psoriatic lesions [58,120]the epidermal 12-LO has gained considerable interest. 12-LO activity results in the formationof 12-HPETE, which is reduced to 12-HETE. The reduction of 12-HPETE involves aglutathione-dependent peroxidase [121]. The 12-LO has been partially characterized inmouse [122,123], human [58,124], rat [124,125] and guinea pig [126] epidermis andkeratinocytes [93]. Furthermore, recently cDNA cloning of mouse epidermal 12-LOs hasbeen carried out [123]. Both a cytosolic and a microsomal 12-LO was identified [123,127].The cytoplasmic 12-LO is also termed the leukocyte type lipoxygenase whereas the micro-somal 12-LO is termed the platelet type. The platelet-type 12-LO metabolizes AA exclu-sively into 12-HETE whereas the leukocyte-type 12-LO transforms AA into both 12-HETEand 15-HETE [123]. In normal human and mouse epidermis as well as involved psoriaticepidermis and cultured human keratinocytes only the platelet-type 12-LO is detectable(mRNA and antibodies against the protein) [123,128,129]. However, in mouse epidermisboth isozymes were induced transiently by phorbol esters [123]. The leukocyte-type 12-LOthat is induced by phorbol ester treatment has been suggested to originate from Langerhanscells [123]. In mice the platelet-type 12-LO has been localized to the stratum granulosum byimmunohistochemical analysis [130]. Both the platelet-type 12-LO and the leukocyte-type12-LO specifically result in the formation of 12(S)-HETE [123,131]. It is therefore of interestthat the 12-HETE in psoriatic scale is not the expected lipoxygenase derived 12(S)-HETE butinstead consists predominantly of 12(R)-HETE [132]. This stereochemical difference mayindicate that 12-HETE is synthesized by different enzymes. For example, cytochrome P-450monooxygenases have been reported to produce predominantly 12(R)-HETE rather than12(S)-HETE from AA [133,134]. Whether 12(R)-HETE recovered from psoriatic lesions isformed by the cytochrome P-450 system has not yet been clarified.

4.4. 15-Lipoxygenase

The initial step of the 15-LO pathway is oxygenation of AA at C-15 resulting in theformation of 15-HPETE, which can either be reduced by glutathione peroxidase to the

34 L. Iversen, K. Kragballe / Prostaglandins & other Lipid Mediators 63 (2000) 25–42

corresponding 15-HETE or converted to 14,15-LTA4 by the action of LTA synthase.14,15-LTA4 is then transformed into 5,15-diHETE, isomers of 8,15-diHETE and isomers of14,15-diHETE [135,136]. Alternatively, 5,15-diHETE can be formed in a reaction catalyzedby 5-LO, because 15-HETE can act as substrate for 5-LO leading to the formation of5,15-diHETE [137]. The 15-LO has been shown to be a Ca21-dependent enzyme localizedpredominantly in the cytosolic fraction [138]. 15-HETE formation may be increased byendogenous HETEs [139] and 15-HETE itself has been shown as a potent activator of itsown formation. Because 15-HETE is a potent 5-LO inhibitor [140], the increased formationof 15-HETE is most likely caused by inhibition of 5-LO resulting in increased substrateavailability for the 15-LO pathway.

The 15-LO has been shown to be present in a variety of tissues including human skin[141] and human keratinocytes [142].

However, several careful studies have revealed rather conflicting data regarding 15-LOactivity in human epidermis. 15-HETE was identified as the only mono-HETE formed bycultured human keratinocytes by several different groups [142–144] whereas others havefound 12-HETE as the prominent AA-metabolite produced by homogenous suspensions offreshly isolated epidermal cells, with only low amounts of 15-HETE present [93,145].Recently these differences have been explained by the various techniques used to isolate thekeratinocytes and the epidermis. It has been demonstrated that AA-metabolism by humankeratinocytes depends upon their functional differentiation. 12-HETE is mainly formed bythe upper epidermal layers whereas 15-HETE is formed predominantly by the basal kera-tinocytes [146].

5. Catabolism of leukotrienes

For cellular homeostasis and for limiting the pro-inflammatory effects of leukotrienes it isessential that the generated leukotrienes in the skin are catabolized to less biologically activecompounds. The metabolism of LTB4 differs among tissues. In human epidermis LTB4 ismetabolized by anv-oxidation pathway into 20-hydroxy-LTB4 and 20-carboxy-LTB4 [147,148]. The first step of inactivation is the conversion of LTB4 into 20-hydroxy-LTB4 throughthe action of a NADPH-dependent microsomal cytochrome p-450 [147,149]. The secondstep is oxidation of 20-hydroxy-LTB4 into 20-carboxy-LTB4 by an aldehyde dehydrogenase.Both 20-hydroxy-LTB4 and 20-carboxy-LTB4 are at least 10-times less potent than LTB4 ininducing chemotaxis of guinea pig peritoneal eosinophils and neutrophils [150]. In culturedhuman keratinocytes LTB4 has also been shown to be metabolized by a 12-hydroxydehy-drogenase resulting in the formation of 12-oxo-LTB4 which is further metabolized intovarious dihydro-LTB4 metabolites [151]. The LTB4 12-hydroxydehydrogenase has beenpurified to homogeneity from porcine kidney cytosol [152] and very recently cDNA cloningof porcine and human LTB4 12-hydroxydehydrogenase has been carried out [153]. 12-oxo-LTB4 has been shown to be at least 100-times less potent than LTB4 in increasing intracel-lular calcium [154].

Metabolic inactivation of the peptideleukotrienes are thought to result from the transfor-

35L. Iversen, K. Kragballe / Prostaglandins & other Lipid Mediators 63 (2000) 25–42

mation of LTC4 to LTD4 and LTE4. However, both LTD4 and LTE4 have potent biologicactivities depending upon the biologic system studied.

References

[1] Willis AL. Nutritional and pharmacological factors in eicosanoid biology. Nutr Rev 1981;39:289.[2] Chapkin RS, Ziboh VA, Marcelo CL, Voorhees JJ. Enzyme preparation from human epidermis lack the

capacity to transform linoleic acid (18:2, n6) and gammahomolinoleic acid (20:3, n6) into arachidonic acid(20:4, n6). J Lipid Res 1985:348.

[3] Irvine RF. How is the level of free arachidonic acid controlled in mammalian cells? Biochem J 1982;204:3.[4] Ryborg AK, Deleuran B, Thestrup-Pedersen K, Kragballe K. Lysophosphatidylcholine: a chemotactic

factor for human T-lymphocytes. Arch Dermatol Res 1994;286:6447.[5] Asaoka Y, Yoshida K, Sasaki Y, et al. Possible role of mammalian secretory group II phospholipase A2

in T-lymphocyte activation: implication in propagation of inflammatory reaction. Proc Natl Acad Sci USA1993;90:176.

[6] Dennis EA. Diversity of group types, regulation and function of phospholipase A2. J Biol Chem 1994;269:13057.

[7] Seilhammer JJ, Randall TL, Yamanaka M, Johnson LK. Pancreatic phospholipase A2: isolation of the geneand cDNAs from porcine pancrease and human lung. DNA 1986;5:519.

[8] Kramer RM, Hession C, Johansen B, et al. Structure and properties of a human non-pancreatic phospho-lipase A2. J Biol Chem 1989;264:5768.

[9] Clark JD, Milona N, Knopf JL. Purification of a 110-kilo-dalton cytosolic phospholipase A2 from thehuman monocytic cell line U937. Proc Natl Acad Sci USA 1990;87:7708.

[10] Kudo I, Murakami M, Hara S, Inoue K. Mammalian non-pancreatic phospholipase A2. Biochim BiophysActa 1993;1170:217.

[11] Andersen S, Sjursen W, Lægreid A, Volden G, Johansen B. Elevated expression of human nonpancreaticphospholipase A2 in psoriatic tissue. Inflammation 1994;18:1.

[12] McCord M, Chabot-Fletcher M, Breton, Marshall LA. Human keratinocytes possess ansn-2 acylhydrolasethat is biochemically similar to the U937-derived 85-kDa phospholipase A2. J Invest Dermatol 1994;102:980.

[13] Davidson FF, Dennis EA. Evolutionary relationships and implications for the regulation of phospholipaseA2 from snake venom to human secreted forms. J Mol Evol 1990;31:228.

[14] Hara S, Kudo I, Chang HW, Matsuta K, Miyamoto T, Inoue K. Purification and characterization ofextracellular phospholipase A2 from human synovial fluid in rhumatoid arthritis. J Biochem 1989;105:395.

[15] Pruzanski W, Vadas P. Phospholipase A2 - a mediator between proximal and distal effectors of inflam-mation. Immunol Today 1991;12:143.

[16] Crowl RM, Stoller TJ, Conroy RR, Stoner CR. Induction of phospholipase A2 gene expression in humanhepatoma cells by mediators of the acute phase response. J Biol Chem 1991;266:2647.

[17] Muhl H, Geiger T, Pignat W, et al. Transforming growth factors type-b and dexamethasone attenuategroup II phospholipase A2 gene expression by interleukin-1 (IL-1) and forskolin in rat mesangial cells.FEBS Lett 1992;301:190.

[18] Clark JD, Milona N, Knopf JL. Purification of a 110-kilodalton cytosolic phospholipase A2 from thehuman monocytic cell line U937. Proc Natl Acad Sci USA 1990;87:7708.

[19] Leslie CC, Voelker DR, Channon JY, Wall MW, Zelarney PT. Properties and purification of an arachi-donyl-hydrolysing phospholipase A2 from a macrophage cell line RAW 264.7. Biochim Biophys Acta1988;963:476.

[20] Clark JD, Lin LL, Kriz WR, et al. A novel arachidonic acid-selective cytosolic PLA2 contains aCa21-dependent translocation domain with homology to PKC and GAP. Cell 1991;65:1043.

36 L. Iversen, K. Kragballe / Prostaglandins & other Lipid Mediators 63 (2000) 25–42

[21] Channon JY, Leslie CC. A calcium-dependent mechanism for associating a soluble arachidonyl-hydrol-ysing phospholipase A2 with membrane in the macrophage cell line RAW 264.7. J Biol Chem 1990;265:5409.

[22] Kim DK, Kudo I, Inoue K. Detection in human platelets of phospholipase A2 activity which preferentiallyhydrolyses an arachidonyl residue. J Biochem (Tokyo) 1988;104:492.

[23] Ramesha CS, Pickett WC. Platelet activating factor and leukotriene biosynthesis is inhibited in polymor-phonuclear leucocytes depleted of arachidonic acid. J Biol Chem 1986;261:7592.

[24] Suga K, Kawasaki T, Blamk ML, Snyder F. An arachidonoyl (polyenoic)-specific phospholipase A2

activity regulates the synthesis of platelet activating factor in granulocytic HL-60 cells. J Biol Chem1990;265:12363.

[25] Murayama T, Ui M. Receptor-mediated inhibition of adenylate cyclase and stimulation of arachidonic acidrelease in 3T3 fibroblasts. Selective susceptibility to islet-activating protein, pertussis toxin. J Biol Chem1985;260:7226.

[26] Burch RM, Luini A, Axelrod J. Phospholipase A2 and phospholipase C are activated by distinct GTP-binding proteins in response tob-adrenergic stimulation in FRTL5 thyroid cells. Proc Natl Acad Sci USA1986;83:7201.

[27] Pruzanski W, Vadas P, Fornasier V. Inflammatory effect of intradermal administration of soluble phos-pholipase A2 in rabbits. J Invest Dermatol 1986;86:380.

[28] Forster S, Ilderton E, Summerly R, Yardly HJ. The level of phospholipase A2 activity is raised in theuninvolved epidermis of psoriasis. Br J Dermatol 1983;108:103.

[29] Ryborg AK, Grøn B, Kragballe K. Increased lysophosphatidylcholine content in lesional psoriatic skin.Br J Dermatol 1995;134:398.

[30] Ziboh VA, Lord JT. Phospholipase A activity in the skin. Modulators of arachidonic acid release fromphosphatidylcholine. Biochem J 1979;184:283.

[31] Kvedar J, Levine L. Modulation of arachidonic acid metabolism in a cultured newborn keratinocyte cellline. J Invest Dermatol 1987;88:124.

[32] Hirata F. The regulation of lipomodulin, a phospholipase inhibitory protein, in rabbit neutrophils byphosphorylation. J Biol Chem 1981;256:7730.

[33] Ilchysyn A, Ilderton E, Kingsbury JA, Yardley HJ. Evidence that raised levels of phospholipase A2 inuninvolved epidermis of psoriasis are caused by hyperphosphorylation of an inhibitor. Br J Dermatol1984;111:721.

[34] Hong LS, Levine L. Inhibition of arachidonic acid release from cells as the biochemical action ofanti-inflammatory corticosteroids. Proc Natl Acad Sci USA 1976;73:1730.

[35] Hirata F, Schiffermann E, Venkatasubramanian K, Salomon D, Axelrod J. A phospholipase A2 inhibitoryprotein in rabbit neutrophils induced by glucocorticoids. Proc Natl Acad Sci USA 1980;77:2533.

[36] Norris JFB, Ilderton E, Yardley HJ, Summerly R, Forster S. Utilization of epidermal phospholipase A2

inhibition to monitor topical steroid action. Br J Dermatol 1984;111(suppl. 27):195.[37] Berridge MJ. Cell signaling through phospholipid metabolism. J Cell Suppl 1986;4:137.[38] Hirata F, Axelrod J. Phospholipid methylation and biological signal transmission. Science 1980;209:1082.[39] Rhee SG, Suh PG, Ryu SH, Lee SY. Studies of inositol phospholipid-specific phospholipase. Science

1989;244:546.[40] Bartel RL, Marcelo CL, Voorhees JJ. Partial characterization of phospholipase C activity in normal,

psoriatic uninvolved, and lesional epidermis. J Invest Dermatol 1987;88:447.[41] Cantley LL, Auger KR, Carpenter C, et al. Oncogenes and signal transduction. Cell 1990;64:281.[42] Cockroft S. Phosphoinositide phosphodiesterase: regulation by a novel guanine nucleotide binding protein

Gp. Trends Pharmacol Sci 1987;12:75.[43] Nishibe S, Wahl MI, Hernandes-Sotomayor SMT, Tonks NK, Rhee SG, Carpenter G. Increase of the

catalytic activity of phospholipase C-g1 by tyrosine phosphorylation. Science 1990;250:1253.[44] Goldschmidt-Clermont PJ, Kim JW, Machesky LM, Rhee SG, Pollard TD. Regulation of phospholipase

C-g1 by profilin and tyrosine phosphorylation. Science 1991;251:1231.

37L. Iversen, K. Kragballe / Prostaglandins & other Lipid Mediators 63 (2000) 25–42

[45] Hamberg M, Svensson J, Wakabayashi T, Samuelsson B. Isolation and structure of two prostaglandinendoperoxides that cause platelet aggregation. Proc Natl Acad Sci USA 1974;71:345.

[46] Ohki S, Ogino N, Yamamoto S, Hayaishi O. Prostaglandin hydroperoxidase, an integral part of prosta-glandin endoperoxide synthase from bovine vesicular gland microsomes. J Biol Chem 1979;254:829.

[47] Vane JR. Inhibition of prostaglandin synthesis as a mechanism of action for aspirin-like drugs. Nat NewBiol 1971;231:232.

[48] O’Banion MK, Sadowski HB, Winn V, Young DA. A serum- and glucocorticoid-regulated 4-kilobasemRNA encodes a cyclooxygenase-related protein. J Biol Chem 1991;266:23261.

[49] Kujubu DA, Fletcher BS, Varnum BC, Lim RW, Hirschmann HR. TIS10, a phorbol ester tumor promotor-inducible mRNA from Swiss 3T3 cells, encodes a novel prostaglandin synthase/cyclooxygenase homo-logue. J Biol Chem 1991;266:12866.

[50] Vane JR, Mitchell JA, Appleton I, et al. Inducible isoforms of cyclooxygenase and nitric-oxide synthasein inflammation. 1994;91:2046.

[51] Scholz K, Furstenberger G, Muller-Decker K, Marks F. Differential expression of prostaglandin Hsynthase isozymes in normal and activated keratinocytes in vivo and in vitro. Biochem J 1995;309:263.

[52] Leong J, Hughes-Fulford M, Rakhlin N, Habib A, Maclouf J, Goldyne ME. Cyclooxygenase in human andmouse skin and cultured human keratinocytes: association of COX-2 expression with human keratinocytedifferentiation. Exp Cell Res 1996;224:79.

[53] Buckman SY, Gresham A, Hale P, et al. COX-2 expression is induced by UVB exposure in human skin:implications for the development of skin cancer. Carcinogenesis 1998;19:723.

[54] Ogino N, Miyamoto T, Yamamoto S, Hayaishi O. Prostaglandin endoperoxide E isomerase from bovinevesicular gland microsomes, a glutathione-requiring enzyme. J Biol Chem 1977;252:890.

[55] Jouvenaz GH, Nugteren DH, Beerthuis RK, van Dorp DA. A sensitive method for the determination ofprostaglandins by gas chromatography with electron-capture detection. Biochim Biophys Acta 1970;202:231.

[56] Ziboh VA, Hsia SL. Prostaglandin E2: biosynthesis and effects of glucose and lipid metabolism in rat skin.Arch Biochem Biophys 1971;146:100.

[57] Jonsson CE, Anggard E. Biosynthesis and metabolism of prostaglandin E2 in human skin. Scand J Clin LabInvest 1972;29:289.

[58] Hammarstro¨m S, Lindgren JA, Marcello C, Duell EA, Anderson TE, Voorhees JJ. Arachidonic acidtransformations in normal and psoriatic skin. J Invest Dermatol 1979;73:180.

[59] Evans CB, Pillai SK, Goldyne ME. Endogenous prostaglandin E2 modulates calcium-induced differenti-ation in human skin keratinocytes. Prostaglandins Leukotrienes Essent Fatty Acids 1993;49:777.

[60] Christ-Hazelhof E, Nugteren DH, van Dorp DA. Conversion of prostaglandin endoperoxides by gluta-thionew-S-transferases and serum albumins. Biochim Biophys Acta 1976;450:450.

[61] Christ-Hazelhof E, Nugteren DH. Purification and characterization of prostaglandin endoperoxide D-isomerase, a cytoplasmic, gluthathione-requiring enzyme. Biochim Biophys Acta 1979;572:43.

[62] Ujihara M, Horiguchi Y, Ikai K, Urade Y. Characterization and distribution of prostaglandin D synthetasein rat skin. J Invest Dermatol 1988;90:448.

[63] Ruzicka T, Printz MP. Arachidonic acid metabolism in guinea pig skin. Biochim Biophys Acta 1981;711:391.

[64] Ruzicka T, Aubo¨ck J. Arachidonic acid metabolism in guinea pig Langerhans cells: Studies on cycloox-ygenase and lipoxygenase pathway. J Immunol 1987;138:539.

[65] Watanabe K, Iguchi Y, Iguchi S, Arai Y, Hayaishi O, Roberts LJ. Stereospecific conversion of prosta-glandin D2 to 9a, 11b-prostaglandin F2 and prostaglandin H2 to prostaglandin F2a by PGF synthase. AdvProstaglandin Thromboxane Leukotriene Res 1987;17A:44.

[66] Chang DGB, Sun M, Tai HH. Prostaglandin 9-ketoreductase and type II 15-hydroxyprostaglandin dehy-drogenase from swine kidney are alternate activities of a single enzyme protein. Biochem Biophys ResCommun 1981;99:745.

[67] Reingold DF, Kawasaki A, Needleman P. A novel prostaglandin 11-keto reductase found in rabbit liver.Biochim Biophys Acta 1981;659:179.

38 L. Iversen, K. Kragballe / Prostaglandins & other Lipid Mediators 63 (2000) 25–42

[68] Ziboh VA, Lord JT, Penneys NS. Alterations of prostaglandin E2-9-ketoreductase activity in proliferatingskin. J Lipid Res 1977;18:37.

[69] Anderson MW, Crutchley DJ, Tainer BE, Eling TE. Kinetic studies on the conversion of prostaglandinendoperoxide PGH2 by thromboxane synthase. Prostaglandins 1978;16:563.

[70] Salmon JA, Smith DR, Flower RJ, Moncada S, Vane JR. Further studies on the enzymatic conversion ofprostaglandin endoperoxide into prostacyclin by procine aorta microsomes. Biochim Biophys Acta 1978;523:250.

[71] Rouzer CA, Matsumoto T, Samuelsson B. Single protein from human leukocytes possesses 5-lipoxygenaseand LTA4 synthase activities. Proc Natl Acad Sci USA 1986;83:857.

[72] Rouzer CA, Kargman S. Translocation of 5-lipoxygenase to the membrane in human leukocytes challengedwith ionophore A23187. J Biol Chem 1988;263:10980.

[73] Wong A, Hwang SM, Cook MN, Hogaboom GK, Crooke ST. Interactions of 5-lipoxygenase withmembranes: studies on the associations of soluble enzyme with membranes and alterations in enzymeactivity. Biochemistry 1988;27:6763.

[74] Miller DK, Gillard JW, Vickers PJ, et al. Identification and isolation of a membrane protein necessary forleukotriene production. Nature 1990;343:278.

[75] Dixon RAF, Diehl RE, Opas E, et al. Requirement of a 5-lipoxygenase-activating protein for leukotrienesynthesis. Nature 1990;343:282.

[76] Shimizu T, Rådmark O, Samuelsson B. Enzyme with dual lipoxygenase activities catalyzes leukotriene A4

synthesis from arachidonic acid. Proc Natl Acad Sci USA 1984;81:689.[77] Borgeat P, Hamberg M, Samuelsson B. Transformation of arachidonic acid and homo-g-linolenic acid by

rabbit polymorphonuclear leukocytes: monohydroxy acids from novel lipoxygenases. J Biol Chem 1976;251:7816.

[78] Lewis RA, Austen KF, Soberman RJ. Leukotrienes and other products of the 5-lipoxygenase pathway.Biochemistry and relation to pathobiology in human diseases. N Engl J Med 1990;323:645.

[79] Rådmark O, Malmsten C, Samuelsson B. Leukotriene A4: enzymatic conversion to leukotriene C4.Biochem Biophys Res Commun 1980;96:1679.

[80] Rådmark O, Malmsten C, Samuelsson B, Goto G, Marfat A, Corey EJ. Leukotriene A. Isolation fromhuman polymorphonuclear leukocytes. J Biol Chem 1980;255:11828.

[81] Maycock AL, Anderson MS, DeSousa DM, Kuehl FA. Leukotriene A4: preparation and enzymaticconversion in a cell-free system to leukotriene B4. J Biol Chem 1982;257:13911.

[82] Rådmark O, Shimizu T, Jo¨rnvall H, Samuelsson B. Leukotriene A4 hydrolase in human leukocytes.Purification and properties. J Biol Chem 1984;259:12339.

[83] Jakschik BA, Kuo CG. Characterization of leukotriene A4 and B4 biosynthesis. Prostaglandins 1983;25:767.

[84] Sun FF, McGuire JC. Metabolism of arachidonic acid by human neutrophils. Characterization of theenzymatic reactions that lead to the synthesis of leukotriene B4. Biochim Biophys Acta 1984;794:56.

[85] Orning L, Hammarstro¨m S. Inhibition of leukotriene C and leukotriene D biosynthesis. J Biol Chem1980;255:8023.

[86] Lee CW, Lewis RA, Corey EJ, Austen KF. Conversion of leukotriene D4 to leukotriene E4 by dipeptidasereleased from specific granule of human polymorphonuclear leukocytes. Immunology 1983;48:27.

[87] Brain S, Camp R, Dowd P, Black AK, Greaves M. The release of leukotriene B4-like material inbiologically active amounts from lesional skin of psoriasis. J Invest Dermatol 1984;83:70.

[88] Grabbe J, Czarnetzki MB, Rosenbach T, Mardin M. Identification of chemotactic lipoxygenase productsof arachidonate metabolism in psoriasis. J Invest Dermatol 1984;82:477.

[89] Brain SD, Camp RDR, Kobza Black A. Leukotriene C4 and D4 in psoriatic skin lesions. Prostaglandins1985;29:611.

[90] Sampson AP, Thomas RU, Costello JF. Enhanced leukotriene synthesis in leukocytes of atopic dermatisand asthma subjects. Br J Clin Pharmacol 1992;33:423.

[91] Grabbe J, Rosenbach T, Czarnetzki BM. Production of LTB4-like chemotactic arachidonate metabolitesfrom human keratinocytes. J Invest Dermatol 1985;85:527.

39L. Iversen, K. Kragballe / Prostaglandins & other Lipid Mediators 63 (2000) 25–42

[92] Rosenbach T, Grabbe J, Moller A, Schwanitz HJ, Czarnetski BM. Generation of leukotrienes from normalepidermis and their demonstration in cutaneous disease. Br J Dermatol 1985;113(suppl 28):157.

[93] Ziboh VA, Casebolt TL, Marcelo CL, Voorhees JJ. Lipoxygenation of arachidonic acid by subcellularpreparations from murine keratinocytes. J Invest Dermatol 1984;83:248.

[94] Janssen-Timmen U, Vickers PJ, Wittig U. Expression of 5-lipoxygenase in differentiating human skinkeratinocytes. Proc Natl Acad Sci USA 1995;92:6966.

[95] Brenton J, Woof D, Young P, Chabot-Fletcher M. Human keratinocytes lack the components to produceleukotriene B4. J Invest Dermatol 1996;106:162.

[96] Iversen L, Fogh K, Ziboh VA, Kristensen P, Schmedes A, Kragballe K. Leukotriene B4 formation duringhuman neutrophil keratinocyte interactions: evidence for transformation of leukotriene A4 by putativekeratinocyte leukotriene A4 hydrolase. J Invest Dermatol 1993;100:293.

[97] Iversen L, Kristensen P, Grøn B, Ziboh VA, Kragballe K. Human epidermis transforms exogenousleukotriene A4 into peptide leukotrienes: possible role in transcellular metabolism. Arch Dermatol Res1994;286:261.

[98] Iversen L, Ziboh VA, Shimizu T, et al. Identification and subcellular localization of leukotriene A4-hydrolase activity in human epidermis. J Dermatol Sci 1994;7:191.

[99] Sola J, Godessart N, Vila L, Puig L, de Moragas JM. Epidermal cell-polymorphonuclear leukocytecoopration in the formation of leukotriene B4 by transcellular biosynthesis. J Invest Dermatol 1992;98:333.

[100] Iversen L, Deleuran B, Hoberg AM, Kragballe K. LTA4 hydrolase in human skin: decreased activity, butnormal concentration in lesional psoriatic skin. Evidence for different LTA4 hydrolase activity in humanlymphocytes and human skin. Arch Dermatol Res 1996;288:217.

[101] Iversen L, Kristensen P, Nissen JB, Merrick WC, Kragballe K. Purification and characterization ofleukotriene A4 hydrolase from human epidermis. FEBS Lett 1995;358:316.

[102] Nissen JB, Iversen L, Kragballe K. Characterization of the aminopeptidase activity of epidermal leuko-triene A4 hydrolase against the opioid dynorphin fragment 1–7. Br J Dermatol 1995;133:742.

[103] Griffin KJ, Gierse J, Krivi G, Fitzpatrick FA. Opioid peptides are substrates for the bifunctional enzymeLTA4 hydrolase/aminopeptidase. Prostaglandins 1992;44:251.

[104] Haeggstro¨m J, Bergman T, Jo¨rnvall H, Rådmark O. Guinea-pig liver leukotriene A4 hydrolase. Purifica-tion, characterization and structural properties. Eur J Biochem 1988;174:717.

[105] Orning L, Jones DA, Fitzpatrick FA. Mechanism-based inactivation of leukotriene A4 hydrolase duringleukotriene B4 formation by human erythrocytes. J Biol Chem 1990;265:14911.

[106] Sala A, Bolla M, Zarini S, Mu¨ller-Peddinghaus R, Folco G. Release of leukotriene A4 versus leukotrieneB4 from human polymorphonuclear leukocytes. J Biol Chem 1996;271:17944.

[107] Soderstrom M, Hammarstro¨m S, Mannervik B. Leukotriene C synthase in mouse mastocytoma cells. Anenzyme distinct from cytosolic and microsomal glutathione transferases. Biochem J 1988;250:713.

[108] Raza H, Awasthi YC, Zaim MT, Eckert RL, Mukhtar H. Glutathione S-transferases in human and rodentskin: multiple forms and species-specific expression. J Invest Dermatol 1991;96:463.

[109] Agarwal R, Raza H, Allyn DL, Bickers DR, Mukhtar H. Glutathione S-transferase-dependent conjugationof leukotriene A4-methyl ester to leukotriene C4-methyl ester in mammalian skin. Biochem Pharmacol1992;44:2047.

[110] Meijer L, Brash AR, Bryant RW, Machlouf J, Sprecher H. Stereospecific induction of starfish oocytematuration by (8R)-hydroxyeicosatetraenoic acid. J Biol Chem 1986;261:17040.

[111] Goetzl EJ, Sun FF. Generation of unique mono-hydroxy-eicosatetraenoic acids from arachidonic acid byhuman neutrophils. J Exp Med 1979;150:406.

[112] Hunter JA, Finkbeiner WE, Nadel JA, Goetzl EJ. Predominant generation of 15-lipoxygenase metabolitesof arachidonic acid by epithelial cells from human trachea. Proc Natl Acad Sci USA 1985;82:4633.

[113] Camp RDR, Mallet AJ, Woollard PM, Brain SD, Black AK, Greaves MW. The identification of hydroxyfatty acids in psoriatic skin. Prostaglandins 1983;26:431.

[114] Hughes MA, Brash AR. Investigation of the mechanism of biosynthesis of 8-hydroxyeicosatetraenoic acidin mouse skin. Biochim Biophys Acta 1991;1081:347.

40 L. Iversen, K. Kragballe / Prostaglandins & other Lipid Mediators 63 (2000) 25–42

[115] Furstenberger G, Hagedorn H, Jacobi T, et al. Characterization of an 8-lipoxygenase activity induced bythe phorbol ester tumor promoter 12-O-tetradecanoylphorbol-13-acetate in mouse skinin vivo. J BiolChem 1991;266:15738.

[116] Burger F, Krieg P, Kinzig A, Schurich B, Marks F, Furstenberger G. Constitutive expression of 8-lipoxy-genase in papillomas and clastogenic effects of lipoxygenase-derived arachidonic acid metabolites inkeratinocytes. Mol Carcinog 1999;24:108.

[117] Qiao N, Takahashi Y, Takamatsu H, Yoshimoto T. Leukotriene A synthase activity of purified mouse skinarachidonate 8-lipoxygenase expressed in Escherichia coli. Biochim Bioiphys Acta 1999;19:131.

[118] Forman BM, Chen J, Evans RM. Hypolipidemic drugs, polyunsaturated fatty acids, and eicosanoids areligands for peroxisome proliferator-activated receptorsa andd. Proc Natl Acad Sci USA 1997;94:4312.

[119] Kliewer SA, Sundseth SS, Jones SA, et al. Fatty acids and eicosanoids regulate gene expression throughdirect interactions with peroxisome proliferator-actiated receptorsa and g. Proc Natl Acad Sci USA1997;94:4318.

[120] Hammarstro¨m S, Hamberg M, Samuelsson B, Duell EA, Stawiski M, Voorhees JJ. Increased concentra-tions of nonesterified arachidonic acid, 12L-hydroxy-5.8.10.14-eicosatetraenoic acid, prostaglandin E2,prostaglandin F2a in epidermis of psoriasis. Proc Natl Acad Sci USA 1975;72:5130.

[121] Chang WC, Nakao J, Orimo H, Murota SI. Effects of reduced glutathione on the 12-lipoxygenase pathwaysin platelets. Biochem J 1982;202:771.

[122] Henke D, Danilowicz R, Eling T. Arachidonic acid metabolism by isolated epidermal basal and differ-entiated keratinocytes from the hairless mouse. Biochim Biophys Acta 1986;876:271.

[123] Krieg P, Kinzig A, Ress-Lo¨schke M, et al. 12-lipoxygenase isozymes in mouse skin tumor development.Mol Carcinog 1995;14:118.

[124] Nugteren DH, Kivits GAA. Conversion of linoleic acid and arachidonic acid by skin epidermal lipoxy-genases. Biochim Biophys Acta 1987;921:135.

[125] Wang MM, Reynaud D, Pace-Asciak CR. In vivo stimulation of 12-(S)-lipoxygenase in the rat skin bybradykinin and platelet activating factor: formation of 12(S)-HETE and hepoxilins, and actions on vascularpermeability. Biochim Biophys Acta 1999;4:354.

[126] Ruzicka T, Vitto A, Printz MP. Epidermal arachidonate lipoxygenase. Biochim Biophys Acta 1983;751:369.

[127] Nakadate T, Aizu E, Yamamoto S, Kato R. Some properties of lipoxygenase activities in cytosol andmicrosomal fractions of mouse epidermal homogenate. Prostagl Leukotr Med 1986;21:305.

[128] Hussain H, Shornick LP, Shannon VR, et al. Epidermis contains platelet-type 12-lipoxygenase that isoverexpressed in germinal layer keratinocytes in psoriasis. Am J Physiol 1994;266:C243.

[129] Takahashi Y, Reddy GR, Ueda N, Yamamoto S, Arase S. Arachidonate 12-lipoxygenase of platelet-typein human epidermal cells. J Biol Chem 1993;268:16443.

[130] Johnson EN, Nanney LB, Virmani J, Lawson JA, Funk CD. Basal transepidermal water loss is increasedin platelet-type 12-lipoxygenase deficient mice. J Invest Dermatol 1999;112:861.

[131] Hamberg M, Samuelsson B. Prostaglandin endoperoxides. Novel transformations of arachidonic acid inhuman platelets. Proc Natl Acad Sci USA 1974;71:3400.

[132] Wollard PM. Stereochemical difference between 12-hydroxy-5,8,10,14-eicosatetraenoic acid in plateletsand psoriatic lesions. Biochem Biophys Res Commun 1986;136:169.

[133] Schwartzman ML, Balazy M, Masferrer J, Abraham NG, McGiff JC, Murphy RC. 12(R)-Hydroxyeico-satetraenoic acid: a cytochrome P450-dependent arachidonate metabolite that inhibits Na,K-ATPase in thecornea. Proc Natl Acad Sci USA 1987;84:8125.

[134] Capdevila J, Yadagiri P, Manna S, Falck JR. Absolute configuration of the hydroxyeicosatetraenoic acids(HETEs) formed during catalytic oxygenation of arachidonic acid by microsomal cytochrome P-450.Biochem Biophys Res Commun 1986;141:1007.

[135] Maas RL, Turk J, Oates JA, Brash A. Formation of a novel dihydroxy acid from arachidonic acid bylipoxygenase-catalyzed double oxygenation in rat mononuclear cells and human leukocytes. J Biol Chem1982;257:7056.

41L. Iversen, K. Kragballe / Prostaglandins & other Lipid Mediators 63 (2000) 25–42

[136] Turk J, Maas RL, Brash AR, Roberts LJ, Oates JA. Arachidonic acid 15-lipoxygenase products fromhuman eosinophils. J Biol Chem 1982;257:7068.

[137] Wilkinson D, Hallam C, Hamsley PE, Lord GH, Mitchell PD. 15-L(S)-hydroxy-5z,8z,11z,13e-eicosatet-raenoic acid is a substrate for 5-lipoxygenase. Biochem Soc Trans 1985;13:180.

[138] Burrall BA, Wintroub BU, Goetzl EJ. Cytosolic localization of the 15-lipoxygenase of human neonatalforeskin keratinocytes. J Invest Dermatol 1986;86:466.

[139] Vanderhoek JY, Karmin HT, Ekborg SL. Endogenous hydroxyeicosatetraenoic acids stimulate the humanpolymorphonuclear leukocyte 15-lipoxygenase pathway. J Biol Chem 1985;260:14482.

[140] Vanderhoek JY, Bryant RW, Baily JM. Inhibition of leukotriene biosynthesis by the leukocyte product15-hydroxy-5,8,11,13-eicosatetraenoic acid. J Biol Chem 1980;255:10064.

[141] Kragballe K, Pinnamaneni G, Desjarlais L, Duell EA, Voorhess JJ. Dermis-derived 15-hydroxy-eicosa-tetraenoic acid inhibits epidermal 12-lipoxygenase activity. J Invest Dermatol 1986;87:494.

[142] Burrall BA, Wintroub BU, Goetzl EJ. Selective expression of 15-lipoxygenase activity by cultured humankeratinocytes. Biochem Biophys Res Commun 1985;133:208.

[143] Burall BA, Cheung M, Chiu A, Goetzl EJ. Enzymatic properties of the 15-lipoxygenase of human culturedkeratinocytes. J Invest Dermatol 1988;71:157.

[144] Green FA. Generation of lipoxygenase products in normal and membrane-damaged cultured humankeratinocytes. J Invest Dermatol 1989;93:486.

[145] Holtzman MJ, Turk J, Pentland A. A regiospecific monooxygenase with novel stereopreference is themajor pathway for arachidonic acid oxygenation in isolated epidermal cells. J Clin Invest 1989;84:1446.

[146] von Zepelin HHH, Schro¨der JM, Smid P, Reusch MK, Christophers E. Metabolism of arachidonic acid byhuman epidermal cells depends upon maturational stage. J Invest Dermatol 1991;97:291.

[147] Mukhtar H, Bik DP, Ruzicka T, Merk HF, Bickers DR. Cytochrome P-450-dependent omega-oxidation ofleukotriene B4 in rodent and human epidermis. J Invest Dermatol 1989;93:231.

[148] Morelli JG, Norris DA, Lyons MB, Murphy RC. Metabolism of exogenous leukotrienes by cultured humankeratinocytes and melanocytes. J Invest Dermatol 1988;90:590.

[149] Sumimoto J, Takeshige K, Minakami S. Characterization of human neutrophil leukotriene B4 omega-hydroxylase as a system involving a unique cytochrome P-450 and NADPH-cytochrome P-450 reductase.Eur J Biochem 1988;172:315.

[150] Ford-Hutchinson AW, Rackham A, Zamboni R, Rokach J, Roy S. Comparative biological activities ofsynthetic leukotriene B4 and its omega-oxidation products. Prostaglandins 1983;25:29.

[151] Wheelan P, Zirolli JA, Morelli JG, Murphy RC. Metabolism of leukotriene B4 by cultured keratinocytes.J Biol Chem 1993;268:25439.

[152] Yokomizo T, Izumi T, Takahashi T, et al. Enzymatic inactivation of leukotriene B4 by a novel enzymefound in the porcine kidney. Purification and properties of leukotriene B4 12-hydroxydehydrogenase.J Biol Chem 1993;268:18128.

[153] Yokomizo T, Ogawa Y, Uozumi N, Kume K, Izumi T, Shimizu T. cDNA cloning, expression, andmutagenesis study of leukotriene B4 12-hydroxydehydrogenase. J Biol Chem 1996;271:2844.

[154] Yokomizo T, Uozumi N, Takahashi T, Kume K, Izumi T, Shimizu T. Leukotriene A4 and leukotriene B4metabolism. J Lipid Cell Signaling 1995;12:321.

42 L. Iversen, K. Kragballe / Prostaglandins & other Lipid Mediators 63 (2000) 25–42