Download Document 8925410

Document related concepts

Clinical neurochemistry wikipedia , lookup

Two-hybrid screening wikipedia , lookup

Expression vector wikipedia , lookup

Polyclonal B cell response wikipedia , lookup

Lipid signaling wikipedia , lookup

Secreted frizzled-related protein 1 wikipedia , lookup

Gene therapy of the human retina wikipedia , lookup

Endogenous retrovirus wikipedia , lookup

Specialized pro-resolving mediators wikipedia , lookup

5-Hydroxyeicosatetraenoic acid wikipedia , lookup

12-Hydroxyeicosatetraenoic acid wikipedia , lookup

Biochemical cascade wikipedia , lookup

Signal transduction wikipedia , lookup

Paracrine signalling wikipedia , lookup

Transcript
 Linköping University Medical Dissertations No. 1287 The enzymatic machinery of leukotriene biosynthesis: Studies on ontogenic expression, interactions and function Tobias Strid Division of Cell Biology Department of Clinical and Experimental Medicine Faculty of Health Sciences, Linköping University SE-­‐581 85 Linköping, Sweden www.liu.se © Tobias Strid 2012 Cover: Pixel-­‐inverted scan of LTA4H in situ hybridized mouse e18.5 fetus. Printed by LiU-­‐Tryck, Linköping, Sweden 2012 ISBN: 978-­‐91-­‐7519-­‐987-­‐0 ISSN 0345-­‐0082 Published articles were reprinted with permission from the copyright holder Elsevier Ltd. According to Elsevier’s policy on author postings Research is the act of going up alleys to see if they are blind // Plutarch Supervisor: Faculty opponent: Professor Sven Hammarström Professor Ralf Morgenstern Department of Clinical and Experimental Institute of Environmental Medicine Medicine, Faculty of Health Sciences, Karolinska Institute, Stockholm Board committee: Dr. Mats Söderström Dr. David Engblom Department of Clinical and Experimental Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Medicine, Faculty of Health Sciences, Linköping University Linköping University Linköping University Co-­‐supervisor: Professor Jan Ernerudh Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University Professor Ernst Oliw Department of Pharmaceutical Biosciences Uppsala University Professor Christer Tagesson Department of Clinical and Experimental Medicine, Faculty of Health Sciences, Linköping University List of Papers LIST OF PAPERS This thesis is based upon the following papers referred to in the text by roman numerals: I: Strid T., Söderström M., and Hammarström S. (2008) Leukotriene C4 synthase promoter driven expression of GFP reveals cell specificity. Biochem Biophys Res Commun. 366: 80-­‐85. http://www.journals.elsevier.com/biochemical-­‐and-­‐biophysical-­‐research-­‐
communications/ II: Strid T., Svartz J., Franck N., Hallin E., Ingelsson B., Söderström M., and Hammarström S. (2009) Distinct parts of leukotriene C4 synthase interact with 5-­‐lipoxygenase and 5-­‐lipoxygenase activating protein, Biochem Biophys Res Commun. 381: 518-­‐522. III: Strid T., Karlsson C., Söderström M., Zhang J., Qian H., Sigvardsson M., and Hammarström S. (2009) Fetal hepatic expression of 5-­‐lipoxygenase activating protein is confined to colonizing hematopoietic cells, Biochem Biophys Res Commun. 383: 336-­‐339. IV: Strid T., Sigvardsson M., Karlsson C., Söderström M., Qiang H., and Hammarström S. (2012) Expression of leukotriene biosynthesis proteins in fetal and adult hematopoietic cells and its functional effects on hematopoiesis. Manuscript. I Populärvetenskaplig Sammanfattning POPULÄRVETENSKAPLIG SAMMANFATTNING Leukotriener tillhör gruppen eikosanoider som är biologiskt aktiva substanser bildade från fleromättade fettsyror. De verkar proinflammatoriskt och deltar i utvecklandet av inflammatoriska sjukdomar såsom astma, allergi, hjärtinfarkt och stroke. De bildas genom att enzymet 5-­‐lipoxygenas (5-­‐LO) tillsammans med aktiveringsproteinet FLAP, omvandlar arakidonsyra, till leukotrien A4 (LTA4). FLAP saknar egen enzymatisk aktivitet, och dess roll anses vara att överföra arakidonsyra från fosfolipas A2 till 5-­‐LO. LTA4 är en instabil molekyl som spontant bryts ned om den inte omvandlas till LTB4 av enzymet LTA4 hydrolas eller till LTC4 av LTC4 syntas. Leukotriener bildas runt cellens kärnmembran där FLAP och LTC4S är belägna och dit 5-­‐LO förflyttas i samband med inflammatorisk aktivering av cellen. Vi har studerat proteinerna som möjliggör syntes av LTC4 och visat att FLAP och LTC4S kan binda till varandra genom sina membrangenomträngande delar. Proteinerna binder till 5-­‐LO genom sina vattenlösliga delar som sticker upp ur membranet. De proteiner som behövs för att bilda leukotriener finns i vissa typer av vita blodkroppar. LTA4H finns i de flesta celler medan övriga proteiner finns i betydligt färre celltyper. Vi undersökte LTC4S genens uttryck genom att konstruera en vektor som uttrycker det lätt påvisbara proteinet GFP under reglering av promotorn för LTC4S. Denna vektor gav ett cellspecifikt uttryck liknande det naturliga för LTC4S när vektorn fördes in i olika celltyper. GFP uttrycket ökade av ämnen som tidigare visats stimulera LTC4S. Vektorn är alltså lämplig som rapportör för LTC4S uttryck. II Populärvetenskaplig Sammanfattning DNA från vektorn användes för att ta fram genmodifierade möss som uttrycker GFP som LTC4S markör. Leukotrienbiosyntesproteiners uttryck under fosterutveckling undersöktes med två andra tekniker som specifikt påvisar enskilda proteiners mRNA respektive proteinet självt. Resultaten visade att ett komplett maskineri för leukotriensyntes uttrycks i levern under fosterutvecklingen. Under denna tid sker här blodbildning från celler som koloniserar levern. Vi särskilde olika celltyper i fetal lever genom cellsortering och undersökte vilka som uttryckte mRNA kodande för proteiner viktiga för leukotriensyntes. Störst mängd fanns i mogna myeloida celler, men även i omogna blodceller fann vi FLAP. Detta fick oss att spekulera kring leukotrieners roll i att reglera själva blodbildningen. Vi undersökte detta genom att analysera cellsammansättningen i blod och benmärg från möss som saknar FLAP och leukotrien produktion. Det var tidigare känt att sådana möss har dämpade inflammatoriska svar. Våra resultat visade att förhållandet av B-­‐ till T-­‐lymfocyter var lägre hos dessa möss jämfört med kontrolldjur. Resultaten tyder på att leukotriener deltar i reglering av blodcellers differentiering och därmed är möjliga mål vid behandling av sjukdomar som drabbar blodbildningen. II Abstract ABSTRACT Leukotrienes (LTs) are biologically active arachidonic acid (AA) derivatives generated by the 5-­‐lipoxygenase (5-­‐LO) pathway. They are produced by myeloid cells. 5-­‐LO converts AA to LTA4 in cooperation with 5-­‐LO activating protein (FLAP). LTA4 is converted to LTB4, by LTA4-­‐hydrolase (LTA4H) or to LTC4 by LTC4-­‐synthase (LTC4S). LTs act on cells through plasma membrane bound G-­‐protein coupled receptors found on leukocytes, smooth muscle and endothelial cells. We report here protein-­‐protein interactions of proteins involved in LTC4 synthesis. 5-­‐LO interacts with cytosolic domains of the integral membrane proteins FLAP and LTC4S at the nuclear envelope, in addition LTC4S interacts with FLAP through its hydrophobic membrane spanning regions. We constructed an LTC4S promoter controlled GFP reporter vector, displaying cell specific expression and sensitivity to agents known to affect LTC4S expression. The vector was used to create transgenic mice expressing GFP as a reporter for LTC4S. Ontogenic mouse expression studies revealed that the complete LT biosynthesis machinery was present at e11.5 primarily in the hematopoietic cells colonizing the liver. Although mature myeloid cells were the main contributors, a substantial amount of FLAP message was also detected in hematopoietic stem and progenitor cells, indicating possible functions for FLAP in hematopoietic regulation. Functional analyses using FLAP knockout mice suggested fine-­‐tuning roles for LTs during differentiation, primarily along the B-­‐lymphocyte differentiation path. III LSC, leukemic stem cell LSK, lineage-­‐ Sca1+ cKit+ cells LT, leukotriene LTA4H, leukotriene A4 hydrolase LTC4S, leukotriene C4 synthase LT-­‐HSC/LSC: long term HSC/LSC MAPEG, membrane-­‐associated proteins in eicosanoid and glutathione metabolism MAPK, mitogen activated protein kinase MEP, megakaryocyte and erythroid progenitor mGST, microsomal glutathione S-­‐
transferase MPP, multipotential progenitor MRP1, multidrug resistance associated protein 1 MS, multiple sclerosis NE, nuclear envelope NK-­‐cell, natural killer cell OAG, 1-­‐oleoyl-­‐2-­‐acteyl-­‐sn-­‐glycerol PAF, platelet activating factor PG, prostaglandin PI3K, phosphoinositide 3-­‐kinase PIP2, phosphatidylinositol-­‐2-­‐phosphate PKC, protein kinase C PPARα, peroxisome proliferator-­‐
activated receptor-­‐α PTX, pertussis toxin PUFA, polyunsaturated fatty acid RA, retinoic acid ROS, reactive oxygen spices SNP, single nucleotide polymorphism SP1, specificity protein 1 SP3, specificity protein 3 SRS-­‐A, slow reacting substance of anaphylaxis STAT3, signal transducer and activator of transcription 3 ST-­‐HSC/LSC, short term HSC/LSC TGF-­‐ β, transforming growth factor β TPA, 12-­‐O-­‐tetradecanoylphorbol-­‐13-­‐
acetate TX, thromboxane UCB, umbilical cord blood 12-­‐HHT, 12(S)-­‐hydroxyheptadeca-­‐5(Z), 8(E), 10(E)-­‐trienoic acid ABBREVIATIONS AA, arachidonic acid AGM, aorta-­‐gonad-­‐mesonephros AP1, activating protein 1 AP2, activating protein 2 BLT1R, leukotriene B4 receptor 1 BLT2R, leukotriene B4 receptor 2 cAMP, cyclic AMP CLP, common lymphoid progenitors CMP, common myeloid progenitors COX, cyclooxygenase cPLA2, cytosolic phospholipase A2 CysLT, cysteinyl leukotriene CysLT1R, CysLT receptor 1 CysLT2R, CysLT receptor 2 DAG, diacyl glycerol DHGLA, dihomo-­‐γ-­‐linolenic acid eGFP, enhanced green fluorescent protein EPA, eicosapentaenoic acid ER, endoplasmatic reticulum ERK, extracellular regulated kinase EX, eoxin FACS, fluorescence-­‐activated cell sorting FLAP, 5-­‐lipoxygenase activating protein FLIM, fluorescence lifetime imaging microscopy GMLP, granulocyte, macrophage, lymphoid progenitor GMP, granulocyte and macrophage progenitor GPCR, G-­‐protein coupled receptor GSH, reduced glutathione GST, glutathione S-­‐transferase HETE, hydroxyeicosatetraenoic acid HPC, hematopoietic progenitor cell HpETE, hydroperoxyeicosatetraenoic acid HSC, hematopoietic stem cell IP3, Inositol tris-­‐phosphate ISH, in situ hybridization LMPP, lymphoid primed multipotential progenitor LO, lipoxygenase LPS, lipopolysaccharide Abbreviations IV Table of contents TABLE OF CONTENTS LIST OF PAPERS ...................................................................................................... I POPULÄRVETENSKAPLIG SAMMANFATTNING .................................... II ABSTRACT ............................................................................................................. III ABBREVIATIONS ................................................................................................. IV 1. INTRODUCTION ............................................................................................. 13 1.1 Lipids and inflammation .......................................................................... 13 1.2 Biosynthesis of eicosanoids ................................................................... 15 1.2.1 Cyclooxygenases .................................................................................................................. 17 1.2.2 Lipoxygenases ....................................................................................................................... 18 2. LEUKOTRIENES AND THE 5-­‐LO PATHWAY ...................................... 21 2.1 Biosynthesis of leukotrienes ................................................................. 21 2.1.1 Biochemistry of Leukotrienes ........................................................................................ 21 2.1.2 5-­‐LO ..................................................................................................................................... 23 2.1.3 FLAP ..................................................................................................................................... 27 2.1.4 LTC4S ..................................................................................................................................... 29 2.1.5 LTA4H ..................................................................................................................................... 32 2.2 Leukotriene receptors .............................................................................. 35 2.2.1 LTB4-­‐receptors ..................................................................................................................... 36 2.2.2 CysLT receptors. ................................................................................................................... 38 2.3 Leukotriene actions ................................................................................... 41 2.3.1 Modulators of inflammatory responses .................................................................... 41 2.3.2 Regulators of hematopoiesis .......................................................................................... 44 3. AIMS .................................................................................................................... 49 Table of contents 4. RESULTS AND DISCUSSION ...................................................................... 51 4.1 Paper I ............................................................................................................. 51 4.2 Generation of GFP mice (unpublished results) .............................. 53 4.3 Paper II ............................................................................................................ 55 4.4 Paper III .......................................................................................................... 56 4.5 Paper IV ........................................................................................................... 57 5. CONCLUSIONS ............................................................................................... 61 6. GENERAL DISCUSSION AND FUTURE PERSPECTIVES ................. 63 7. ACKNOWLEDGEMENTS .............................................................................. 67 7.1 Financial support ........................................................................................ 67 7.2 Personal thank you / Personligt tack ................................................. 67 8. REFERENCES ................................................................................................... 73 9. REPRINTS OF PUBLISHED ARTICLES AND MANUSCRIPTS .... 105 Introduction 1. INTRODUCTION 1.1 Lipids and inflammation An inflammatory process is initiated as an important and immediate response conducted by our bodies to protect us against infections or injuries. This process typically leads to redness, swelling, heat and pain. These hallmarks of inflammation are caused by increases in blood flow, vascular permeability and leukocyte migration as well as stimulation of pain receptors. The purpose of an inflammatory response is to initiate host defense reactions to eliminate intruders, such as bacteria, viruses and parasites and to initiate repair of injured tissues. Inflammation can be viewed upon as a double-­‐edged sword that needs a strict and precise control to be neither too weak nor too strong, either of which may be detrimental. The responses of cells involved in the inflammatory reaction are directed by a complicated array of signaling molecules (the topic of inflammation has been reviewed in [1-­‐5]). Proinflammatory mediators may be quite diverse at the molecular level. Some of them are bioactive lipids generated from essential fatty acids, and one such family of very potent lipid mediators are the eicosanoids (reviewed in [6, 7]). Essential fatty acids are necessary for survival of mammals and they are found in the phospholipids of most cell membranes. The term “essential” indicates that they cannot be synthesized by the human body and therefore must be obtained by dietary intake. The essential fatty acids are divided into two series; ω-­‐6 fatty acids derived from linoleic acid and ω-­‐3 fatty acids derived from α-­‐linolenic acid (reviewed in [8]). 13 Introduction Essential fatty acids
COOH
COOH
α-Linolenic acid
Linoleic acid
COOH
COOH
Dihomo-γ-linolenic acid
Eicosapentaenoic acid
COOH
Arachidonic acid
Eicosanoids
Fig. 1 Essential fatty acids. The precursors of eicosanoids (dihomo-­‐γ-­‐linolenic acid, arachidonic acid and eicosapentaenoic acid) are 20-­‐carbon fatty acids formed from two 18-­‐carbon essential fatty acids: linoleic acid and α-­‐linolenic acid. 14 Introduction 1.2 Biosynthesis of eicosanoids Eicosanoids are oxygenated derivatives of 20-­‐carbon polyunsaturated fatty acids (PUFAs), which in turn are formed from 18-­‐carbon essential fatty acids (Fig. 1). Both the 20-­‐ and the 18-­‐carbon fatty acids are normal components of phospholipids in the cell membranes. Dihomo-­‐γ-­‐linolenic acid (DHGLA), AA and eicosapentaenoic acid (EPA) give rise to different series of eicosanoids [9, 10]. The conversion of linoleic acid to AA occurs mainly in the liver. From there AA is distributed to cells throughout the body for incorporation into cell membrane phospholipids [11]. AA is formed by desaturation and elongation of linoleic acid, an essential ω-­‐6 fatty acid. These conversions are catalyzed by Δ6-­‐desaturase and fatty acid elongase respectively, giving rise to DHGLA which is further converted by Δ5-­‐desaturase to AA [12]. AA is incorporated into membrane phospholipids by acylation of 2-­‐lysophospholipids with arachidonyl-­‐CoA or by transacylation of existing phospholipids [13]. During evolution, human diet presumably contained an equal ratio οf ω-­‐6 to ω-­‐3 fatty acids. In modern western diets however, this ratio is approximately 16:1[14-­‐16]. This has lead to increased formation of eicosanoids derived from AA and less from EPA [15]. AA constitutes around 20% of membrane phospholipid fatty acids in immune cells from people consuming a western diet [17-­‐23], whereas EPA occupies < 1% [20, 22]. Increased intake of long chain ω-­‐3 PUFAs increases the proportion of these fatty acids in membrane phospholipids [17-­‐23] and leads to lower production of prostaglandin E2 (PGE2), thromboxane A2 (TXA2), leukotriene B4 (LTB4), 5-­‐hydroxyeicosatetraenoic acid (5-­‐HETE) and leukotriene C4 (LTC4) [17-­‐19, 21, 24]. 15 Introduction Instead it increases the production of the 3 series prostaglandins (PGs), thromboxane (TXA3) and the 5 series leukotrienes (LTs) [17, 19, 24, 25] some of which are less potent than the corresponding eicosanoids formed from AA [26, 27] (reviewed in [7, 28]). COOH
Arachidonic acid (20:4)
COX
5-LO
O
OOH
COOH
COOH
O
O
PGG2
5-HpETE
HO
COX
5-LO
O
O
COOH
O
COOH
OH
LTA4
PGH2
Fig. 2 Eicosanoid formation. Eicosanoids are formed from arachidonic acid (AA) primarily by two distinct enzymatic pathways: the cyclooxygenase (COX) pathway by which AA is oxygenated in two-­‐steps to an unstable prostaglandin endoperoxide intermediate (PGH2) and the 5-­‐lipoxygenase (5-­‐LO) pathway, which in two-­‐steps converts AA to the unstable epoxide intermediate LTA4. Enzymes involved in eicosanoid formation use free AA as substrate and the hydrolytic release of AA is a rate limiting step in eicosanoid biosynthesis [29]. The availability of free AA in cells is strictly regulated [30] and the cellular level of free AA is low [31]. AA and other PUFAs are released by phospholipase A2 enzymes [30], most commonly cytosolic phospholipase A2 (cPLA2) [32, 33]. 16 Introduction cPLA2 is activated by an increase in intracellular Ca2+ concentration [34, 35] and by phosphorylation [36-­‐39]. Ca2+ activates cPLA2 directly by interaction with a Ca2+-­‐binding C2 domain in the N-­‐terminal part of the protein and indirectly through protein kinases [40, 41]. Activation results in translocation of the protein from cytosol to the nuclear envelope (NE), endoplasmic reticulum (ER) and Golgi [42-­‐47] which have higher AA content than other cell membranes [48, 49] (reviewed in [30]). Conversion of free AA to eicosanoids takes place via two main enzymatic pathways: the cyclooxygenase (COX) and lipoxygenase (LO) pathways (Fig. 2). A third pathway (the cytochrome P450 pathway) catalyzes conversion of AA to vasoactive hydroxyeicosatetraenoic and epoxyeicosatrienoic acids (reviewed in [50]). 1.2.1 Cyclooxygenases The COX isoenzymes, COX-­‐1[51] and COX-­‐2 [52, 53], metabolize AA to the prostaglandin endoperoxide PGH2. Both isoenzymes are localized to the NE and ER. COX-­‐1 is constitutively expressed while COX-­‐2 expression is inducible [54, 55]. The first steps catalyzed by COX involve dual oxygenation of AA to PGG2. This is followed by reduction of a 15-­‐hydroperoxy group in PGG2 to a 15-­‐hydroxyl in PGH2. PGH2 is the immediate precursor of the primary PGs, prostacyclin (PGI2) and TXA2 and the subsequent products are formed by specific synthase enzymes (Fig. 3). A more detailed description of the COX pathway is beyond the scoop of this thesis. This topic has been reviewed in [56-­‐58]. 17 Introduction OH
COOH
O
COOH
O
OH
O
TXA2
OH
PGD2
O
O
COOH
COOH
O
OH
OH
PGH2
OH
PGE2
OH
CO
OH
O
COOH
OH
OH
OH
PGF2α
PGI2
Fig. 3 The COX pathway. The precursor prostaglandin endoperoxide PGH2 is generated from AA by cyclooxygenases. Specific synthases convert PGH2 to primary prostaglandins (PGD2, PGE2 and PGF2 ), prostacyclin (PGI2) and thromboxane A2 (TXA2). α
1.2.2 Lipoxygenases Lipoxygenases are a group of enzymes, which oxygenate PUFAs containing methylene interrupted cis-­‐double bonds, notably the ω-­‐3 and the ω-­‐6 series essential fatty acids. They are named after the carbon atom where the O2 is introduced. The products from AA are hydroperoxy-­‐eicosatetraenoic-­‐acids (HpETE). One 1,4-­‐cis-­‐cis-­‐diene is transformed into a 1-­‐hydroperoxy-­‐2-­‐
trans-­‐4-­‐cis-­‐diene group [59]. AA contains three 1,4-­‐cis-­‐cis-­‐diene groups and O2 can be introduced at 6 different positions to give 5-­‐, 8-­‐, 9-­‐, 11-­‐, 12-­‐, or 15-­‐HpETE (Fig. 4). HpETEs are reduced to HETEs by peroxidases. 18 Introduction Lipoxygenases occur in fungi, plants, and animals [60]. In addition to the 5-­‐LO pathway, described in detail below, human cells may express 12-­‐ and/or 15-­‐lipoxygenases. Platelet and leukocyte forms of 12-­‐LO and reticulocyte/leukocyte and epidermis types of 15-­‐LO are expressed in humans. Leukocyte 12-­‐LO and reticulocyte 15-­‐LO form the same products and are also referred to as 12/15-­‐LO. Expression of 12-­‐ and 15-­‐LO differs between species; mice do not express 15-­‐LO, but express leukocyte type 12-­‐LO with both 12-­‐LO and 15-­‐LO activity (reviewed in [61]). 12-­‐LO and 15-­‐LO are involved in lipoxin formation (reviewed in [62, 63]). A role in cancer metastasis has been proposed for 12-­‐LO and epidermal 15-­‐LO has been associated with suppression of carcinogenesis (reviewed in [64]). COOH
Arachidonic acid (20:4)
5-LO
15-LO
12-LO
OOH
COOH
COOH
OOH
5-HpETE
COOH
15-HpETE
OOH
12-HpETE
Fig. 4 The LO pathway. Lipoxygenases convert AA to HpETEs. The reaction is stereospecific and the lipoxygenases are named after the carbon atom where molecular oxygen is introduced. Human 5-­‐, 12-­‐ and 15-­‐lipoxygenases have been identified. 19 20 Leukotrienes and the 5-­‐LO pathway 2. LEUKOTRIENES AND THE 5-­‐LO PATHWAY 2.1 Biosynthesis of leukotrienes The 5-­‐LO pathway generates the proinflammatory lipid mediators leukotrienes. The main protein components of this pathway are 5-­‐LO, FLAP, LTA4H and LTC4S. 5-­‐LO, assisted by FLAP, transforms AA to LTA4, which is metabolized either by LTA4H to LTB4, or by LTC4S to LTC4. 2.1.1 Biochemistry of Leukotrienes Leukotrienes of the 4 series are formed from AA following its release from membrane phospholipids by cPLA2 (see above on pages 16-­‐17). Major determinants for the magnitude of LT biosynthesis are the concentration of free AA [65] and its accessibility [66]. Most stimuli of LT synthesis activate both 5-­‐LO and cPLA2 [67]. 5-­‐LO catalyzes a two-­‐step conversion of AA via 5-­‐HpETE to LTA4 (reviewed in [68, 69]). These steps are stereospecific and initiated by abstraction of a hydrogen atom from carbon C-­‐7 of AA and insertion of molecular oxygen at C-­‐5, giving 5-­‐HpETE [70] (Fig. 4). In the second reaction the 10D (pro-­‐R) hydrogen is removed followed by radical migration and formation of the allylic epoxide LTA4 [71-­‐73]1 (Fig. 5). LTA4 is unstable and rapidly metabolized to either LTB4 or LTC4. LTA4H catalyzes a hydrolytic reaction to the dihydroxy acid LTB4 [68, 69] (Fig. 5), which is exported out of the cell by a carrier-­‐mediated, temperature sensitive and energy-­‐dependent mechanism [74, 75]. LTC4S catalyzes a glutathione-­‐S-­‐
transferase reaction yielding the peptidolipid LTC4. 1 In [73] EPA was used instead of AA and the absolute configuration of the 10D-­‐hydrogen in EPA is pro(S) and 5-­‐LO conversion yields LTA5. 21 Leukotrienes and the 5-­‐LO pathway The discovery of LTC4 in 1979 [76, 77] was a result of investigations to find the chemical structure of a factor termed slow reacting substance of anaphylaxis (SRS-­‐A) [78]. LTC4 is formed by conjugation of LTA4 with reduced glutathione (GSH) catalyzed by LTC4S [76, 77]. Shortly after that LTD4 [79-­‐81] and LTE4 [81, 82] were found to be formed by metabolism of the peptide part of LTC4. OH
O
OH
COOH
COOH
LTA4H
LTB4
LTA4
LTC4S + GSH
OH
COOH
Gly
C5H11 γGluCys
LTC4
γ-Glutamyl transpeptidase /
γ-Glutamyl leukotrienase
OH
COOH
C5H11
CysGly
LTD4
Dipeptidase
OH
COOH
C5H11
Cys
LTE4
22 Fig. 5 Leukotriene biochemistry. LTA4, formed from AA by the action of 5-­‐LO, is an unstable epoxide which is converted to either LTB4 or LTC4. LTB4 is formed by hydrolysis catalyzed by LTA4H which introduces an OH group at carbon 12 and converts the epoxide group in LTA4 to an OH group at C-­‐5. LTC4 is formed by the action of LTC4S which conjugates LTA4 with GSH at carbon 6 and converts the epoxide to a C-­‐5 OH group. LTC4 is actively transported out of the cell and converted extracellularly to LTD4 by γ -­‐glutamyl-­‐
transpeptidase or γ-­‐
glutamyl-­‐leukotrienase. LTD4 is metabolized to LTE4 by a dipeptidase. Leukotrienes and the 5-­‐LO pathway LTD4 is generated from LTC4 by cleavage of the isopeptide bond of the GSH moiety by γ-­‐glutamyl transpeptidase [79, 83] or γ-­‐glutamyl leukotrienase [84] and LTE4 is formed from LTD4 by elimination of the glycine residue by a dipeptidase [82, 85] (Fig. 5). LTC4 is actively transported out of the cells by multidrug resistance-­‐associated protein 1 (MRP1) [86-­‐88] and the conversions to LTD4 and LTE4 take place extracellularly. LT production may also occur in cells not expressing all the necessary enzymes. LTB4 was formed when red blood cells, which lack 5-­‐LO, were incubated with LTA4 [89]. Conversion of exogenous LTA4 to LTC4 was demonstrated in bone marrow-­‐derived mast cells [90] and co-­‐incubations of red blood cells with neutrophils provided the first evidence for cooperation between different types of cells in LT synthesis [91]. Transcellular formation of LTC4 was reported soon thereafter [92]. This phenomenon has later been reported for a number of other cell types [93-­‐
100]. Another example of transcellular biosynthesis is the generation of the antiinflammatory lipoxins which requires cooperation between 5-­‐LO and 12/15-­‐LO expressing cells (reviewed in [62, 63]). Resolvins and protectins are generated by cooperation between COX-­‐2 and lipoxygenase enzymes (reviewed in [101]). 2.1.2 5-­‐LO Human 5-­‐LO, is a 674 amino acid protein with a molecular weight of 78 kDa [102, 103] which requires non-­‐heme iron (III) for enzymatic activity [104]. cDNA encoding human 5-­‐LO was cloned in 1988 [102, 103]. 23 Leukotrienes and the 5-­‐LO pathway Sequence comparisons with other lipoxygenases and mutagenesis studies indicated that iron is bound to His367, His372 and His550, Asn554 and Ile673 of the active site in 5-­‐LO [105]. The crystal structure confirmed this binding pattern except that Asn554 is not involved [106]. Resting inflammatory cells harbor 5-­‐LO in the cytosol or in the nucleus, depending on cell type [107-­‐113]. Upon cell activation, cPLA2 and 5-­‐LO translocate to the NE and/or ER, where FLAP resides [114-­‐116]. Most stimuli simultaneously activate 5-­‐LO and cPLA2 (Fig. 6, reviewed in [67]). In the absence of functional FLAP, 5-­‐LO is unable to convert endogenous AA to LTs in cells (reviewed in [117]). However, exogenous AA has been suggested to be converted also by non-­‐translocated cytosolic 5-­‐LO [118]. The crystal structure of 5-­‐LO revealed that the catalytic site is located in the C-­‐terminal part of the protein, while the N-­‐terminal part contains the C2-­‐
like calcium-­‐binding domain [106]. Binding of Ca2+ is reversible [119] and serves as an activator of 5-­‐LO [120]. Asn43, Asp44 and Glu46 located in loop 2 of the C2-­‐like domain, are important for this interaction and for the stimulatory effects of Ca2+ [121]. ATP has been reported as another activator of 5-­‐LO [122] and additional activating factors e.g. microsomal membranes [120, 123] were identified during purification. In addition, glycerides such as 1-­‐oleoyl-­‐2-­‐acteyl-­‐sn-­‐glycerol (OAG) activated 5-­‐LO [124]. 5-­‐LO binding to coactosin-­‐like protein is essential for activation of the enzyme by Ca2+ [125, 126]. The interaction, but not enzyme activity occurs in the absence of Ca2+ [126]. Nitric oxide [127, 128] and certain peroxidases [129, 130] suppress 5-­‐LO activity. 24 Leukotrienes and the 5-­‐LO pathway Another control mechanism is regulation of subcellular distribution: e.g. neutrophils, monocytes and peritoneal macrophages harbor inactive 5-­‐LO in cytosol while alveolar macrophages and Langerhans cells harbor it in the nuclear matrix (reviewed in [67]). These localizations of the protein in resting cells affect their ability to produce LTs. Nuclear localization has been associated with increased LT synthesis, except in eosinophils where the opposite effect was observed (reviewed in [131]). 5-­‐LO has both nuclear import [131, 132] and nuclear export sequences [133]. Nuclear import is induced by glycogen, cytokines and adhesion to cell surfaces (reviewed in [131]). Nuclear import and export are both regulated by phosphorylation [66, 134, 135] of Ser271 by MAPkinase-­‐activated protein kinase 2 [136, 137], Ser663 by extracellular-­‐signal regulated kinases (ERK) [138] and Ser523 by PKA [139]. Ser523 phosphorylation prevented nuclear localization and suppressed 5-­‐LO activity [139, 140]. Control also takes place at the transcriptional level. The human 5-­‐LO gene [141] is located at chromosome 10q11.2. Expression of 5-­‐LO is mainly restricted to granulocytes, monocytes/macrophages, mast cells, dendritic cells, Langerhans cells of the skin and B-­‐lymphocytes (reviewed in [142, 143]). Age-­‐dependent increase of 5-­‐LO expression has also been detected in mouse hippocampus [144]. 5-­‐LO expression was upregulated in differentiated compared to undifferentiated myeloid cells [143], e.g. differentiated tissue macrophages compared to blood monocytes [145, 146]. Elevated levels of 5-­‐LO mRNA were detected in leukocytes from asthmatic patients [147]. 5-­‐LO expression appears to require growth factors since human monocytes lost both 5-­‐LO and FLAP expression when kept in culture for 7 days [148]. 25 Leukotrienes and the 5-­‐LO pathway 5-­‐LO has also been detected in several types of cancer cells (reviewed in [149, 150]), where its products have been suggested to serve as autocrine growth factors [151]. 5-­‐LO products have further been proposed to induce DNA damage, and increase formation of HεdGuo-­‐adducts (a lipid peroxide derived DNA adduct) in Ca2+ ionophore A23187 stimulated cells, whereas treatment with a FLAP inhibitor returned HεdGuo-­‐adducts to basal levels [152]. The human 5-­‐LO gene promoter contains GC rich regions but lacks both TATA and CCAAT boxes [141]. An important factor for silencing 5-­‐LO gene expression appears to be DNA methylation [153]. Transcription factors Sp1 and Egr1 interacted with five of eight tandem arranged GC boxes [154-­‐156]. Naturally occurring mutations in the human 5-­‐LO promoter are either deletions or addition of extra Sp1 or Egr1 sites [155]. Interestingly, the mouse 5-­‐LO promoter lacks tandem GC boxes and contains only one Sp1 or Sp3 site [157]. Promoter analyses have revealed two positive and two negative regulatory regions [154]. A slight increase in promoter activity was observed after treatment with phorbol ester [154]. A positive regulatory region containing several vitamin D3 response elements was detected between positions -­‐779 and -­‐229 [158]. Additional vitamin D3 response elements as well as response elements for the TGF-­‐β effectors, Smad3 and 4 appear further upstream in the promoter as well as in intron 4 and exons 10-­‐14, respectively [159, 160]. Recent findings suggest involvement of male sex hormones in the regulation of 5-­‐LO [161, 162]. Decreased LT formation was detected in male compared to female blood cells due to variations in ERK activation as a result of higher androgen levels. 26 Leukotrienes and the 5-­‐LO pathway Part of the 5-­‐LO pool in male cells reside in the perinuclear region prior to activation, a distribution previously associated with impaired enzyme activity upon Ca2+ ionophore A23187 activation [163] (for reviews on the regulation of 5-­‐LO, see [151, 164]). 2.1.3 FLAP The integral membrane protein, FLAP, localized to NE and ER [109, 112, 165-­‐167] is structurally related to LTC4S (described below). The two proteins belong to the same gene superfamily, referred to as Membrane-­‐
Associated Proteins in Eicosanoid and Glutathione metabolism (MAPEG) [168]. FLAP was discovered in studies using the FLAP inhibitor MK886, at the time known as an inhibitor of 5-­‐LO in intact cells but not in broken cell preparations [169, 170]. Transfection experiments showed that both 5-­‐LO and FLAP were required for Ca2+-­‐ionophore induced LT formation from endogenous AA [171]. FLAP also stimulates conversion of exogenous AA to LTs [172] but in this case the requirement for FLAP is not absolute. Several functions of FLAP have been suggested: 1) It may serve as a scaffold protein assembling 5-­‐LO and cPLA2 at the NE and ER involving important protein-­‐
protein interactions [169, 173, 174], e.g. between FLAP and 5-­‐LO [116]. However, others report that MK886 inhibited LT biosynthesis without affecting translocation and membrane association of 5-­‐LO [112, 163, 175, 176]. 2) FLAP is an AA binding protein, which donates AA to 5-­‐LO [172, 177]. AA and other PUFAs compete with FLAP inhibitors for binding to FLAP (Fig. 6) [178, 179]. Inhibitors such as MK886 bind to the first hydrophilic loop of FLAP as judged by immuneprecipitation of FLAP peptide fragments and mutagenesis studies [180, 181]. 27 Leukotrienes and the 5-­‐LO pathway The crystal structure of inhibitor bound FLAP [182] showed four trans-­‐
membrane α-­‐helices connected by two long cytosolic loops and one short luminal loop. Amino acid residues 38-­‐47 and 102-­‐115 make up cytosolic loops 1 and 2, respectively. The crystal structure also revealed that FLAP is a homotrimer [182] and suggested that its cytosolic loops interact with the C-­‐terminal catalytic domain of 5-­‐LO. The calcium-­‐binding domain of 5-­‐LO anchors it to the nuclear membrane [182]. One FLAP trimer binds one 5-­‐LO monomer [182]. The essential role of FLAP in LT biosynthesis was established using FLAP null mice. Like 5-­‐LO-­‐/-­‐ mice, FLAP-­‐/-­‐ mice lack detectable LT production [183]. It is not known whether FLAP influences AA metabolism by 12-­‐ and 15-­‐LO. FLAP, however, stimulated 5-­‐LO oxygenation of both 12(S)-­‐ and 15(S)-­‐HETE and this effect was abolished by the FLAP inhibitor MK886 as well as when certain FLAP mutant variants were used [184]. A significant reduction of 12-­‐HETE formation was described in FLAP knockout mice [185]. The human FLAP gene [186] is composed of 5 small exons, four large introns, and a promoter region containing a potential TATA box, as well as an AP-­‐2 and several glucocorticoid receptor binding sites. FLAP expression, which occurs mainly in myeloid, 5-­‐LO expressing cells [187], is altered by treatment of cells with TGF-­‐β1, 1,25-­‐dihydroxy-­‐vitamin D3, and phorbol ester [188, 189]. Transcription factors of the C/EBP family were identified as important for FLAP expression modulated by TNF-­‐α [190]. Prolonged exposure to lipopolysaccharide (LPS) induced FLAP mRNA and protein. The effect was mediated by NF-­‐κB and a C/EBP element located within the first 134 bp of the FLAP promoter [191]. 28 Leukotrienes and the 5-­‐LO pathway Increased FLAP expression has been observed in several pathological conditions. Single nucleotide polymorphisms (SNP) identified in the FLAP gene have been correlated with increased risk of developing myocardial infarction and stroke [192]. FLAP was highly upregulated in aorta and adipose tissue in a mouse model of atherosclerosis and the increased FLAP expression was accompanied by elevated LTB4 production [193]. FLAP protein in aorta and adipose tissue was found in infiltrating macrophages, but not in T-­‐cells. Treatment with FLAP inhibitor, however, reduced not only atherosclerotic lesion size but also T-­‐cell content, a finding suggesting FLAP as a ”potential link between innate and adaptive immunity” [193]. 2.1.4 LTC4S Leukotriene A4 is enzymatically converted to LTC4 [194, 195] by a glutathione S transferase (GST) with narrow substrate specificity [196]. LTC4S is a membrane-­‐bound protein [196, 197] located in the microsomal cell fraction [198-­‐201]. LTC4S was purified by two groups in 1992 and 1993 [202, 203] and its molecular weight, 18 kDa, was determined. The amino-­‐
terminal sequence was also reported [203]. The cloning of LTC4S cDNA was reported by two groups in 1994 [204, 205], revealing a 150 amino acid protein. Evidence for “homo-­‐oligomerization of LTC4S in living cells” was published in 2003 [206]. The two-­‐dimensional crystal structure of recombinant LTC4S indicated that it forms homotrimers [207]. This observation was confirmed by the 3D crystal structure reported in 2007 [208, 209]. The enzyme contains 5 α-­‐helices, of which four are membrane spanning, with the N-­‐ and C-­‐termini at the same side of the membrane and an overall structure that resembles that of FLAP. 29 Leukotrienes and the 5-­‐LO pathway Human LTC4S shares 31% amino acid identity with FLAP, and both belong to the MAPEG gene-­‐superfamily [168]. Other closely related MAPEG proteins are the microsomal Glutathione S-­‐transferases (mGSTs) [210]. Helices 1 and 2 in LTC4S are connected by a long cytosolic loop [208, 209] as in FLAP [182]. Helices 3 and 4 are connected by just a short turn in LTC4S compared to a long second cytosolic loop in FLAP. The GSH binding site of LTC4S is formed at the interface between two neighboring monomers close to the protein surface and the top of the binding site is covered by the long cytosolic loop between helices 1 and 2 [208, 209]. Site directed mutagenesis showed that residues important for catalytic activity are located in hydrophilic segments. Arg51 and Tyr93 were suggested as critical residues at the active site [211]. The crystal structure confirmed that Arg30, Arg51, Asn55, Glu58, Tyr59, Tyr93, Tyr97 and Arg104 are important for GSH binding to the protein [208, 209]. LTC4S is distributed in the outer and excluded from the inner part of the nuclear membrane. FLAP, on the other hand, occurs in both the outer and inner segments of the NE [212]. The third hydrophobic region of LTC4S is important for membrane localization [213] and amino acids 114-­‐150 are sufficient for homo-­‐
oligomerization to occur [206]. The human [214, 215] and mouse [216] LTC4S genes have five exons and identical intron /exon boundaries. Murine intron 1 is 1/3 (ca. 500 bp) shorter than its human counterpart. The mouse gene is localized to chromosome 11B1.1-­‐1.2. The promoter region of the mouse gene has consensus sites for AP-­‐2, CCAAT (enhancer binding protein) and polyoma virus enhancer-­‐3 [216]. The human gene is localized to chromosome 5q35 [214, 215] adjacent to a gene cluster implicated in asthma (5q31-­‐33) (reviewed in [217]). 30 Leukotrienes and the 5-­‐LO pathway The human LTC4S and FLAP genes have almost identical intron / exon organization but LTC4S has much shorter introns [186]. The human LTC4S promoter contains a binding motif for the ubiquitously expressed Sp1 and Sp3 transcription factors [218, 219]. Other regulatory elements would therefore be required to explain the limited expression of LTC4S. 5-­‐LO and LTC4S co-­‐express in cells of myeloid origin (basophils, eosinophils, mast cells and monocytes / macrophages) [220]. Platelets contain LTC4S but lack 5-­‐LO [221]. LTC4S expression also occurs in the plexus choroideus and other parts of the brain [222-­‐228]. Treatment of human erythroleukemia cells with the PKC activator TPA induced LTC4S [229] in agreement with the presence of PKC-­‐responsive elements (AP-­‐1 and AP-­‐2) in the promoter [214-­‐216]. Other reports have indicated inhibition of LTC4 formation in cells pre-­‐treated with TPA [230, 231]. LPS [232, 233] and TNF-­‐α [234] down-­‐regulate expression of LTC4S whereas TGF-­‐β [235] upregulates it in mononuclear-­‐like cell lines. Retinoic acid upregulated LTC4S promoter and enzyme activity in rat basophilic leukemia cells [236, 237]. LTC4S expression is thus differentially regulated depending on cell type. This is in agreement with a report suggesting that cell-­‐specific transcription involves Sp1 and a Kruppel-­‐like transcription factor [219]. Several LTC4S promoter polymorphisms have been associated with inflammatory disorders. A Polish study identified a -­‐444 A to C polymorphism with the C allele being more frequent in patients with aspirin-­‐induced asthma [238] but this association was not found in Australia, Japan or the United States [239-­‐241]. In addition, a G/A polymorphism located at -­‐1072 in the LTC4S promoter was associated with an increased risk of developing ischemic brain injury [242]. 31 Leukotrienes and the 5-­‐LO pathway LTC4S knockout mice [243] develop and breed normally but have a reduced inflammatory response in some models. Data obtained from these mice showed that LTC4S accounted for the major part of their LTC4 production, with only a small contribution from other GST enzymes. A few human case reports have indicated individuals lacking LTC4S. These children had no CysLTs in their cerebrospinal fluid and showed impaired CNS development and mental retardation [244] suggesting developmental roles for CysLTs. Eosinophils produce a different spectrum of eicosanoids compared to other granulocytes. When incubated with AA human eosinophils produce a positional isomer of LTC4 with an absorbance maximum at 282 rather than 280 nm and a shorter retention time than LTC4 when analyzed by RP-­‐HPLC. The product was identified as 14,15-­‐LTC4 and named eoxin (EX) C4 [245]. Like LTC4, EXC4 was metabolized to EXD4 and EXE4 by elimination of its γ-­‐
glutamyl followed by its glycine residue, respectively. Incubation of human eosinophils with AA alone favored EX production whereas incubation with AA plus a Ca2+ ionophore resulted in mainly LT products [245]. It is not known whether LTC4S is involved in the biosynthesis of eoxins or other 15-­‐
LO or 12-­‐LO products. 2.1.5 LTA4H LTA4H is a cytosolic 70 kDa zinc-­‐containing metalloprotease [246]. The protein has dual enzyme activities and is responsible for the conversion of LTA4 to LTB4. In contrast to its usual cytosolic localization one report has described a nuclear localization in alveolar macrophages [247]. 32 Leukotrienes and the 5-­‐LO pathway The deduced amino acid sequence [248] revealed similarities to zinc-­‐
containing aminopeptidases [249] an observation, which has been experimentally verified [250]. Inhibition of the peptidase activity also inhibited the conversion of LTA4 to LTB4 [251]. A specific role for the aminopeptidase activity could not be established using LTA4H knockout mice [252]. Cloning of the human cDNA encoding LTA4H [248, 253] identified a 610 amino acid protein. The crystal structure of the protein, bound to a competitive inhibitor, revealed a protein folded into three domains, together creating a deep cleft harboring the catalytic Zn2+ site [254]. LTA4H is self-­‐inactivated during catalysis due to covalent modification of the enzyme by its substrate LTA4 [255]. Phosphorylation of Ser415 has been reported, but present evidence does not support regulation of LTA4H by phosphorylation [256, 257]. The human LTA4H gene is 35 kbp and localized to chromosome position 12q22. It has 19 exons and its 5´-­‐flanking region has several putative transcription factor binding sites including an AP-­‐2 site and two xenobiotic response elements [258]. Promoter studies identified a negative control element between -­‐1702 and -­‐1196, one or more positive elements between -­‐1196 and -­‐123 and a cis-­‐acting cell specific control element between -­‐123 and -­‐40 [259]. Transcriptional regulation of LTA4H is distinct from that of 5-­‐
LO and the other proteins involved in LT synthesis since this enzyme is expressed much more widely [260]. 33 Leukotrienes and the 5-­‐LO pathway LTA4H
LTB4
LTA4
AA
5LO
LTC4
LTC4S
PM
FLAP
cPLA2
5LO
5LO
cPLA2
NM
Fig. 6 Cellular leukotriene synthesis. Activation of cells by inflammatory stimuli results in translocation of cPLA2 and 5-­‐LO to the nuclear membrane. cPLA2 liberates AA from membrane phospholipids. AA is transferred to 5-­‐LO by FLAP and converted to LTA4. LTA4 is either hydrolyzed by LTA4H to LTB4 or conjugated with glutathione by LTC4S to LTC4 depending on the expression of these enzymes. NM: nuclear membrane, PM: plasma membrane 34 Leukotrienes and the 5-­‐LO pathway 2.2 Leukotriene receptors Leukotrienes exert their actions through specific cell surface receptors (reviewed in [261, 262]). Two receptors (BLT1R and BLT2R) mediate intracellular signals in response to LTB4. Cys-­‐LTs mediate their effects through the two cell surface receptors CysLT1R and CysLT2R. Additional membrane proteins may also serve as receptors for CysLTs. The leukotriene receptors are seven-­‐transmembrane G-­‐protein coupled receptors (GPCRs). To date more than 30 GPCRs are known to respond to lipids [261]. The G-­‐proteins involved are heterotrimeric GTP-­‐binding proteins consisting of α-­‐, β-­‐ and γ-­‐subunits. Upon receptor activation the α-­‐
subunit is released and stimulates or inhibits multiple intracellular signaling pathways. The β,γ-­‐subunits are also involved in signaling. GPCR activation may lead to increased concentrations of intracellular calcium ions, increased or decreased cAMP production, Rho activation and modification of ion channels. The heterotrimeric G-­‐proteins are grouped based on their α-­‐subunits (Gαs, Gαi, Gαq and Gα12 reviewed in [263]). Gαs stimulates and Gαi inhibits adenylate cyclase. Gαq stimulates phospholipase C which cleaves phosphatidylinositol-­‐2-­‐phosphate (PIP2) to inositol tris-­‐
phosphate (IP3) and diacylglycerol (DAG) resulting in increased Ca2+ concentrations and PKC activation. Gα12 activates Rho GDP / GTP exchange factor resulting in actin rearrangements (reviewed in [261]). 35 Leukotrienes and the 5-­‐LO pathway 2.2.1 LTB4-­‐receptors BLT1R, was discovered in 1997 [264] and BLT2R, in 2000 [265]. Expression of BLT1R occurs mainly in inflammatory cells e.g. neutrophils, eosinophils, dendritic cells, macrophages, B-­‐lymphocytes and certain T-­‐lymphocytes [266-­‐272] but low expression has also been detected in spleen and thymus [264]. The human BLT1R gene is located on chromosome 14, (14q11.2-­‐q12) [273]. The precise mechanism behind the leukocyte-­‐restricted expression is not clear. Sp1 binds to the promoter but, due to a GC rich sequence, the gene is heavily methylated in non-­‐expressing cells and this may explain its restricted expression [274]. BLT2R is a low affinity receptor for LTB4 [275, 276]. In man it is expressed mainly in skeletal muscle, heart, lung, spleen, liver, ovaries, and leukocytes, but low expression levels are detected in most tissues [261, 265]. The coding region for BLT2R was identified as a putative intronless open reading frame in close proximity to the BLT1R gene [275, 276]. Intracellular signaling evoked by LTB4 receptor activation is cell type specific depending on the type of G-­‐protein involved. BLT1R signaling in granulocytes is commonly mediated by Gαi2, while Gαi1 and Gα0 has been found to mediate BLT1R signaling in the nervous system. Signaling through Gαq has also been reported there (reviewed in [261, 262]). Downstream effects of BLT1R signaling involve activation of kinases [277], MAPKs [278, 279], phosphatidylinositol 3-­‐kinase (PI3-­‐K) [280, 281] and tyrosine kinases [281, 282]. LTB4 signaling also increases expression of inflammatory genes e.g. IL-­‐6 and monocyte chemoattractant protein-­‐1 (MCP-­‐1, see Fig. 7) [283-­‐285] (reviewed in [261, 262, 286]). BLT2R activation inhibits adenylate-­‐cyclase and increases intracellular Ca2+, but less potently than BLT1R [265]. Gαi-­‐mediated chemotaxis has been described [265, 275]. 36 Leukotrienes and the 5-­‐LO pathway Generation of reactive oxygen spices (ROS) mediated by activation of Rac-­‐
ERK has also been proposed to occur through BLT2R [287]. As mentioned above, BLT2R is a low affinity LTB4 receptor, consequently higher LTB4 concentrations are needed to inhibit adenylate-­‐cyclase and to induce chemotaxis than when BLT1R is activated [265, 275]. BLT1R
G αi1
G αq G α16
Gαi2 Gα0
BLT2R
Gα0
cAMP
2+
Ca
PKC
cAMP
2+
Ca
Gαi
Rac-ERK
MAPKs
PI3-K
Tyrosine kinases
IL-5
IL-6
IL-10
Fig. 7 LTB4 receptors. LTB4 interacts with two GPCRs; BLT1R and BLT2R. The receptors couple to different G-­‐
proteins in different cell types. LTB4 signaling activates several kinases and induces expression of inflammatory genes e.g. the IL-­‐5, IL-­‐6 and IL-­‐10 genes (revived in [261, 262, 286]). The ligand potency ranking for BLT1R is LTB4 > 20-­‐OH-­‐LTB4 = 12-­‐oxo-­‐LTB4 > 12(R)-­‐HETE > 20-­‐COOH-­‐LTB4 [264] while that of BLT2R is LTB4 > 12-­‐epi-­‐
LTB4 > 12(S)HETE > 12(S)HpETE > 12(R)HETE > 20-­‐OH-­‐LTB4 [288]. 12(S)-­‐
hydroxyheptadeca-­‐5(Z), 8(E), 10(E)-­‐trienoic acid (12-­‐HHT) generated from PGH2 during its conversion by thromboxane A2 synthase [289] was recently identified as a high affinity ligand for BLT2R having higher affinity than LTB4 [290]. In addition to BLT1R and BLT2R, LTB4 also binds to and activates the nuclear orphan receptor peroxisome proliferator-­‐activated receptor-­‐α (PPARα) in vitro [291] and in cells [292]. 37 Leukotrienes and the 5-­‐LO pathway 2.2.2 CysLT receptors. Pharmacological evidence for the existence of CysLT (SRS-­‐A) receptors was reported 6 years before the structure of LTC4 was reported [293]. Subsequent studies on venous preparations where LTC4 and LTD4 antagonists failed to block the response indicated the presence of a second CysLT receptor [294]. Cysteinyl leukotriene receptors 1(CysLT1R) and 2 (CysLT2R) have been cloned and characterized [295-­‐298]. Human CysLT1R is a 337 amino acid protein [295, 297] expressed in adipose tissue, brain, bronchus, colon, heart, kidney, liver, lung, pancreas, peripheral blood leukocytes, placenta, prostate gland, skeletal muscle, small intestine and spleen [295, 297, 299]. At the cell level CysLT1R occurs in B-­‐lymphocytes, endothelial cells, eosinophils, fibroblasts, fibrocytes, mast cells, monocytes/macrophages, neutrophils, pregranulocytic CD34+ cells and smooth muscle cells [300-­‐305]. CysLT2R, a 346 amino acid protein, is expressed in adrenal glands, brain, heart, leukocytes, lymph nodes, peripheral blood, placenta and spleen [296, 298, 306]. At the cell level expression occurs in macrophages and in smooth muscle cells of the human lung [296] (reviewed in [261]). It also occurs in neurons of the myenteric plexus where lack of expression in knockout mice dampened experimental colitis progression [307]. CysLT2R dominates over CysLT1R in heart and eosinophils [296, 298, 308] while only CysLT1R is expressed in trachea [298]. Similar CysLT2R patterns occur in mice but the pharmacological characteristics of the mouse receptor differ from those of the human receptor [309, 310]. The CysLT1R gene is located on the X chromosome both in man (Xq13.3-­‐
q21.1, [295]) and mouse [311]. The human gene has 5 variably spliced exons and a promoter region with multiple transcription start sites [299]. 38 Leukotrienes and the 5-­‐LO pathway The human CysLT2R gene is located on chromosome 13 (13q14.12-­‐q21.1) in a region with several markers of asthma or atopi [312, 313]. The gene has 6 variably spliced exons and the whole coding sequence is located in exon 6 [314]. CysLT1R signaling employs multiple second messengers (e.g. DAG, inositol phosphates and Ca2+) and PKC and involves differentiated G-­‐protein coupling, including pertussis toxin (PTX)-­‐sensitive (Gαi/o) and insensitive (Gαq/ii) ones [315, 316] (Reviewed in [261]). MAPK pathways are activated in several cell types (Fig. 8) [317-­‐324]. CysLT effects on mouse embryonic stem cells involve STAT3, PI3-­‐K/Akt, glycogen synthase kinase-­‐3β/β-­‐
catenin and Ca2+-­‐calcineurin pathways [325]. Like LTB4, CysLTs, induce expression of inflammation-­‐associated genes [326, 327] (reviewed in [261, 262, 286]). GPCRs, including CysLT1R, have recently been localized in the cell nucleus in contrast to the usual plasma membrane localization and a nuclear localization signal was recognized in CysLT1R [328]. Evidence for intracrine signaling activating ERK 1/2 and vesicular transport mediated IL-­‐4 secretion has been reported [328]. Detailed information on CysLT2R signal transduction is not available due to a lack of selective antagonists: coupling to both PTX sensitive and insensitive G proteins has been suggested (Fig. 8, [296, 329] reviewed in [261, 262]). 39 Leukotrienes and the 5-­‐LO pathway CysLT1R
CysLT2R
Gα11
Gα0
G αq
Gαi
Gαi
DAG
2+
Ca
cAMP
PKC
Rho GEF
2+
Ca
cAMP
MAPK
PI3-K
STAT3
AA-release
Actin reorganization
Gα11
Gαq
Gαo
NO
IL-8
IL-4
VEGF IL-13
Integrins
Fig. 8 CysLT receptors. CysLTs interact with two GPCRs; CysLT1R and CysLT2R. Additional receptors have been reported to respond to CysLTs. G-­‐protein coupling of the receptor depends on the expressing cell type. Signaling primarily involves kinases including MAPK. Signaling also induces expression of inflammatory genes (revived in [261, 262, 286]). The ligand affinity order for CysLT1R is LTD4 > LTC4 > LTE4 [297] and for CysLT2R LTD4 = LTC4 > LTE4 [296]. Short-­‐hairpin (sh)RNA-­‐mediated knockdown of CysLT2R enhanced the surface expression of CysLT1R, suggesting a coherent regulation of the two receptors [330]. Additional CysLT receptors with specificity for LTC4 [331-­‐333] and LTE4 [334] have been postulated (reviewed in [261]) and recently an orphan GPCR (GPR17 [335]) was recognized as a phylogenetic intermediate between purinergic P2Y and CysLT receptors [336]. GPR17 is a dual receptor for uracil nucleotides and CysLTs [336, 337] and negatively regulates CysLT1R signaling in a ligand-­‐independent manner [338, 339]. 40 Leukotrienes and the 5-­‐LO pathway 2.3 Leukotriene actions 2.3.1 Modulators of inflammatory responses At the site of inflammation, recruited leukocytes produce LTs in response to microbes interacting via cell surface receptors e.g. Fc-­‐γ receptors [340, 341], Toll-­‐like receptors [342-­‐344] and β-­‐glycan receptors [345] (reviewed in [346]). LTs facilitate recruitment of additional leukocytes in several ways: LTB4 is a potent chemoattractant for neutrophils and monocytes [347-­‐349] and upregulates the expression of cell surface adhesion glycoproteins. Interactions with blood vessel endothelial cells slow down and stop the circulating leukocytes so they can migrate into surrounding tissues [350]. LTB4 also aids leukocyte accumulation by attenuation of leukocytes apoptosis [351, 352]. CysLTs contribute to leukocyte accumulation by increasing vascular permeability and by attracting subsets of T-­‐cells [353-­‐355]. CysLT receptor expression is upregulated by activation of CD4+ T-­‐cells and is responsible for their chemotaxis to LTD4 [355, 356]. CysLT signaling on eosinophils induces adhesion, IL-­‐4 release, migration, recruitment, and increases eosinophil survival [352, 357-­‐359]. It activates dendritic cells and their cytokine release [360, 361], mast-­‐cell cytokine production [362] and activation of basophils [363]. CysLT receptor signaling also enhances IgG and IgE production in IL-­‐4 / CD40-­‐activated B-­‐
cells [364]. These proinflammatory actions of LTs on the immune system are a major contributor to the pathogenesis of several diseases. 41 Leukotrienes and the 5-­‐LO pathway Elevated levels of LTB4 (LTB4-­‐like immunereactivity) has been reported in ulcerative colitis and other types of inflammatory bowel disease [365-­‐367], psoriasis [368], cystic fibrosis [369], rheumatoid arthritis [370, 371] and in neutrophils from patients treated surgically for aortic abdominal aneurysm [372, 373]. The main symptoms of asthma come from CysLT formation. These compounds are potent stimulators of bronchial smooth muscle [374] and induce airway mucus secretion [375]. CysLTs also constrict coronary (but not pulmonary) artery and pulmonary vein [376, 377]. They potently increase vascular permeability throughout the body [353, 354, 378]. Th2-­‐
biased allergic responses due to CysLT signaling affect both acute and chronic phases of asthmatic inflammation. The effects are primarily mediated by CysLT1R signaling but CysLT2R may be involved in certain cases [379, 380] (reviewed in [381]). CysLTs have also been implicated in the pathogenesis of allergic rhinitis [382] and atopic dermatitis [383, 384]. Clinical studies using inhibitors of LT biosynthesis or antagonists of LT receptors in asthmatic patients blocked both the early and the late phase of an allergic response [385, 386] (reviewed in [261]). The CyLT1R antagonists montelukast2, pranlukast and safirlukast and the 5-­‐LO inhibitor zileuton are approved drugs for the treatment of asthma (reviewed in [387]). Some features of asthma may also be mediated by LTB4 [388], e.g. proliferative effects on airway smooth muscle cells has been described [279]. Leukotrienes are also important players of the inflammatory component of cardiovascular diseases (reviewed in [389-­‐391]). 2 Montelukast under the trade name Singulair is one of Merk Sharp & Dohme´s best selling drugs. 42 Leukotrienes and the 5-­‐LO pathway Effects mediated by CysLT1R [392-­‐396] and CysLT2R [397] were shown to be involved in ischemic brain injury. The proposed CysLT receptor GPR17 was upregulated in damaged tissues, and knockdown reduced neuronal injury after ischemia [337, 398]. Elevated LT levels were detected in cerebrospinal fluid of MS patients [399]. In spite of detrimental roles in inflammatory diseases, LTs are also important players in host defense reactions against microbial infections (reviewed in [286]). LTs stimulate phagocytosis [341, 400-­‐402] and microbe killing [346, 403, 404] the latter by induction of NO synthesis and production of ROS [404-­‐408]. LTs induce release of α-­‐defensin [409] and antimicrobial peptide LL-­‐37 [410, 411]. In malnutrition [412-­‐414], vitamin D deficiency [415], HIV infection [416, 417], liver cirrhosis [418], diabetes [419] and among cigarette smokers [420, 421] there is correlation between the susceptibility to infectious diseases and attenuated LT synthesis.
Several studies have postulated LTs as mediators in different types of cancer. For example: LTB4 increases proliferation and survival of a colon cancer cell line and proliferation of pancreatic cancer cells. CysLT signaling increased proliferation of colorectal cancer cell lines and CysLT1R antagonists induced apoptosis in a pancreatic cancer cell line. Effects have also been seen on metastasis: BLTR antagonists reduced metastasis in a mouse model of pancreatic cancer (reviewed in [422]). 43 Leukotrienes and the 5-­‐LO pathway 2.3.2 Regulators of hematopoiesis The hematopoietic system is one of the largest and most proliferative cell systems in the body. Studies in mice revealed that the first hematopoietic cells develop in the yolk sac at around day 8 of gestation. Around day 9 hematopoietic stem cells (HSC) appear in the aorta-­‐gonad-­‐mesonephros (AGM) region [423] and somewhat later in the placenta and liver [424]. HSC from yolk sac, AGM region and placenta all appear to contribute to the colonization of the liver, the main hematopoietic organ during fetal life [423, 425] (reviewed in [426]). Shortly before birth, bone marrow starts to take over this role and becomes the main hematopoietic organ for the remaining life [427, 428]. All types of blood cells derive from a common HSC (reviewed in [429]). This cell can a), self-­‐renew and b), differentiate into the different blood cell lineages (Fig. 9). Flow cytometry is widely used to fractionate hematopoietic cells into different cell types. Stem cell activity is found in the so-­‐called LSK population, i.e. cells lacking lineage specific surface markers (B220, Mac1, Gr1, IL7Rα, Ter119, CD3, CD4, and CD8) and amply expressing the markers Sca1 and cKit [430, 431] (reviewed in [432]). LSK cells include both long term (LT)-­‐HSC, short term (ST)-­‐HSC, multipotent progenitors (MPPs), lymphoid primed multipotent progenitors (LMPPs) and granulocyte-­‐macrophage-­‐lymphocyte progenitors (GMLPs). Further compartmentalization of the LSK population is achieved using antibodies against CD34 [433] and Flt3 [434, 435]. LSK cells lacking expression of CD34 and Flt3 are LT-­‐HSC. ST-­‐HSC and MPPs express CD34 while LMPPs and GMLPs express both CD34 and Flt3 [436, 437] (reviewed in [438]). Lymphoid, natural killer (NK) and dendritic cell potential lies within the Lin-­‐/Flt3+/IL7R+/cKitlow/Sca1+ common lymphoid progenitor (CLP) population [438-­‐440]. 44 Leukotrienes and the 5-­‐LO pathway The cKithigh/Sca1-­‐ myeloid, megakaryocyte and erythroid progenitor population contains three subpopulations: megakaryocyte and erythroid progenitors (MEP; CD34low/CD16/CD32low), common myeloid progenitors (CMP; CD34low/CD16/CD32+), and granulocyte and macrophage progenitors (GMP; CD34high/CD16/CD32high) [441]. Only MEP has megakaryocyte potential while erythroid potential is found in both MEP and CMP cells [438] (Fig. 9). The CLP population has B-­‐ and T-­‐lymphoid potential [439], but is considered to develop mainly into B-­‐lymphocytes in vivo. T-­‐lymphocytes primarily mature from early thymic progenitors seeding the thymus [442-­‐444] (Fig. 9, reviewed in [445]). 45 Leukotrienes and the 5-­‐LO pathway LSK
LT-HSC
MPP
ST-HSC
LMPP/
GMLP
ELP
CMP
MEP
CLP
ETP
MkP
PreCFU-E
Pre-GMP
CFU-E
GMP
Granulocytes
Erythrocytes
Megakaryocytes
T-lymphocytes
Dendritic cells/
NK-cells
Monocytes/
Macrophages
B-lymphocytes
Fig. 9 Hematopoietic development. All hematopoietic cells are derived from a single ancestor stem cell with potential of regeneration as well as differentiation. During proliferation, hematopoietic cells gradually become specialized and concomitantly lose Zandi eal PMID: 20969584
potential to differentiate into other cell types (reviewed in [438, 445]). LT-­‐HSC, long term hematopoietic stem cell; ST-­‐HSC, short term HSC; MPP, multipotent progenitor cell; LMPP, lymphoid primed MPP; GMLP, granulocyte, macrophage and lymphoid progenitor; CMP, common myeloid progenitor; MEP, megakaryocyte and erythroid progenitor; MkP, megakaryocyte progenitor; GMP, granulocyte and macrophage progenitor; CFU-­‐E, colony forming unit erythrocyte; ELP, early lymphoid progenitor; CLP, common lymphoid progenitor; ETP, early thymic progenitor. 46 Leukotrienes and the 5-­‐LO pathway The mechanisms regulating the hematopoietic system are complex and both intrinsic (involving transcription factor modulation) and extrinsic (cytokine-­‐mediated) pathways are employed [446]. It is not yet understood to which extent eicosanoids are involved in this system or their precise roles. There is an excellent review on the topic; “Eicosanoid regulation of hematopoiesis and hematopoietic stem and progenitor trafficking” [447]. PGE2 inhibits in vitro proliferation of mouse and human cells of the myeloid lineage and an abnormal inhibitory response to PGE2 has been reported in cells derived from leukemia patients [448-­‐455]. There is a prognostic connection between disordered CFU-­‐GM inhibition by PGE2 and myelodysplastic syndromes [456]. However other groups did not observe inhibition by PGE2 on hematopoiesis; PGE2 treatment of murine bone marrow increased proliferation of hematopoietic cells and increased production of HSC [457-­‐459]. LT effects on hematopoietic regulation have also been reported. The expression of 5-­‐LO and FLAP in human CD34+ cells is low, but increases when the cells are differentiated in vitro to dendritic cells [460]. In vitro differentiation of CD34+ cells also leads to increased expression of BLT1R [269]. Other in vitro studies suggest that LTs promote hematopoietic progenitor cell (HPC) proliferation [461-­‐463]. These proliferative effects were reduced by 5-­‐LO inhibitors [461, 464]. LTB4 and LTC4 decreased erythroid and granulocyte-­‐macrophage colony formation [465] whereas a 5-­‐LO inhibitor had the opposite effect. LTB4 increased HPC proliferation in umbilical cord blood (UCB) and decreased the number of CD34+ cells whereas a BLTR antagonist had opposite effects (increased the number of CD34+ cells and blocked HPC proliferation) [466]. LTD4 induced chemotaxis and transendothelial migration of CD34+ progenitor cells and affected their trafficking [467]. 47 Leukotrienes and the 5-­‐LO pathway LTD4 promoted hematopoietic progenitor cell expansion and stimulated integrin-­‐dependent adhesion by upregulating α4β1 and α5β1 integrins [468]. LTD4 also increased growth of eosinophil progenitor cells from atopic patients [462]. The 5-­‐LO gene has been identified as a critical regulator of leukemia stem cells (LSC) in a mouse model of chronic myeloid leukemia, since induction of the disease failed in 5-­‐LO-­‐/-­‐ mice [469]. Lack of 5-­‐LO impaired differentiation, cell division and survival of long term LSC, but not of normal HSC. BM cell compositions were not altered in 5-­‐LO knockout mice compared to wild type animals [469] but reduced numbers of memory B-­‐cells and mature germinal centers in spleen after immunization were reported in knockout compared to wild type mice [470]. A reduction was also seen in spleen follicular B-­‐cells and the ability to generate a sub-­‐
population of helper T-­‐cells (Tfh) in the spleen was nearly lost in 5-­‐LO deficient mice [470]. 5-­‐LO deficient mouse spleen dendritic cells have altered cytokine production [471]. Other LO products have also been proposed to influence hematopoietic regulation. 12-­‐HETE and 15-­‐HETE stimulated proliferation and accelerated differentiation of CD34+ cells toward the erythroid lineage [472]. 48 Aims 3. AIMS The aims of this thesis were to investigate subcellular localization and protein-­‐protein interactions taking place within the LT machinery, to investigate the expression pattern of the LT machinery during mouse fetal development compared to the expression pattern in adult animals and to identify functional roles of LTs in the hematopoietic system. 49 50 Results and Discussion 4. RESULTS AND DISCUSSION 4.1 Paper I To investigate the mechanisms behind LTC4S gene expression we constructed a vector expressing enhanced green fluorescent protein (eGFP) under the control of the mouse leukotriene C4 synthase promoter (Fig. 1A, Paper I). This vector was used to study promoter activity in various cell lines. GFP expression was observed in human monocytic leukemia (THP-­‐1) and rat basophilic leukemia (RBL-­‐1) myeloid cells which both normally express leukotriene C4 synthase, but not in human embryonic kidney (HEK-­‐293T) epithelial cells which do not express this enzyme (Fig. 1, Paper I). In the myeloid cells, but not in the epithelial cells, we observed that LTC4S promoter activity was stimulated by 12-­‐O-­‐tetradecanoylphorbol-­‐13-­‐
acetate (TPA) in both RBL-­‐1 and THP-­‐1 cells and by all-­‐trans-­‐retinoic acid (RA) in RBL-­‐1 cells. In contrast dimethyl sulfoxide did not affect promoter activity (Figs. 2 and 3, Paper I). In a previous study TPA stimulated leukotriene C4 synthase activity in human erythroleukemia cells whereas dimethyl sulfoxide inhibited the activity and retinoic acid had no effect [229]. The stimulatory effect of TPA was confirmed in this study suggesting that it occurs at the promoter level. The lack of effect of the various treatments in HEK-­‐293T control cells and in cells transfected with control plasmid further supported this notion. The 5´-­‐flanking region of the mouse leukotriene C4 synthase gene contains consensus sequences for AP-­‐2, CCAAT (enhancer binding protein) and polyoma virus enhancer-­‐3 [216]. 51 Results and Discussion AP-­‐1 and AP-­‐2 sites, known to be affected by TPA, are found in the human LTC4S promoter [214, 215]. Sp1 and a Kruppel-­‐like transcription factor have been suggested to determine cell-­‐specific LTC4S transcription [219]. TPA induced leukotriene C4 synthesis in certain cell lines [229], while it decreased leukotriene C4 production in HL-­‐60 cells, an effect that was blocked by PKC specific inhibitors [230]. Furthermore, retinoic acid upregulated leukotriene C4 synthase in rat basophilic leukemia cells [236, 237]. Thus it appears that leukotriene C4 synthase gene expression is differentially regulated depending on the cellular environment, in agreement with the results presented in Paper I. Our recombinant vector is regulated in an LTC4S specific manner suggesting that the promoter sequence and intron 2 are sufficient for LTC4S regulation and furthermore that the vector should be useful as a reporter of LTC4S expression. 52 Results and Discussion 4.2 Generation of GFP mice (unpublished results) The vector expressing GFP under control of the regulatory regions of the mouse LTC4S promoter and intron 2 (described in Paper I) and an analogous vector where intron 2 was replaced by LTC4S intron 1, were used to produce transgenic mice. The intron 1 vector was constructed by PCR amplification of a sequence containing mouse LTC4S intron 1 (primer 1: 5´-­‐
CACCCTCGTGGGAGTTCTGTTGC-­‐3´, primer 2: 5´-­‐CAGAGATCACCTGTCTCGA-­‐ GAAGTAGGC-­‐3´) and subsequently subcloned into the vector described in Paper I after excision of its intron 2 sequence by Nhe I / Xho I digestion. Transgenic mice were created by pronuclear injection of cassettes containing LTC4S regulatory regions, eGFP and a poly A tail, excised from the recombinant vectors. Injected mouse oocytes were implanted in the uteri of pseudopregnant mice and transgenic litters were identified by PCR screening using one primer matching a sequence in the LTC4S promoter and a second primer matching the eGFP region (primer 1: 5´-­‐AAGTGGCTCTTCT-­‐
GGCTACCG-­‐3´, primer 2: 5´-­‐GCTTCTCGTTGGGGTCTTTGC-­‐3´). To establish the new strains, positive offspring was mated with C57BL/6 mice. eGFP expression was analyzed by fluorescence microscopy using 478-­‐495 nm blue light. Analysis of blood from transgenic litter revealed eGFP expression in nucleated white blood cells. Prior to analysis the cells were counterstained with DAPI to visualize the cell nuclei (Fig. 10). Tissues and embryos from sacrificed transgenic animals were analyzed for the expression of GFP by in situ hybridization using a short GFP anti sense probe created by primer 1: 5´-­‐CTCGTGAGGATCCTGACCTACGGCG-­‐3´, primer 2: 5´-­‐GATGTTGTGGCGGATCTAGAAGTTCACCTT-­‐3 ´or a long probe using same primers as in Paper I to amplify eGFP. 53 Results and Discussion GFP
DAPI
pLTC4S-intron2-GFP
pLTC4S-intron1-GFP
Fig. 10 eGFP expression in leukocytes. Blood from transgenic mice carrying a cassette expressing eGFP regulated by the LTC4S promoter plus intron 2 or intron 1 were examined under 478-­‐495 nm blue light in a fluorescence microscope. Prior to mounting, cells were treated with DAPI nuclear stain. DAPI was visualized in UV-­‐light Analysis of tissues or embryos from transgenic mice by in situ hybridization revealed expression of eGFP message in the fetal and neonatal liver (Fig. 11). Tissues from non-­‐transgenic littermates were used as negative controls. The results from fluorescence microscopy and in situ hybridization confirmed that GFP was expressed at the mRNA and protein levels under control of the LTC4S promoter combined with either intron 1 or 2, with a localization resembling that of wild type LTC4S expression. +
GFP P0 tissue
GFP P0 tissue
Fig. 11 In situ hybridization using eGFP antisense probes. Tissues from mice carrying a transgenic cassette expressing eGFP under control of pLTC4S + intron 1 or tissues from non-­‐transgenic mice were examined using in situ hybridization with antisense probes for eGFP. Expression was detected in liver. AS, anti-­‐sense. long AS GFP probe
short AS GFP probe
54 Results and Discussion 4.3 Paper II In this paper we report interactions between proteins involved in LTC4 biosynthesis. Fluorescent FLAP-­‐ and LTC4S-­‐fusion proteins co-­‐localized at the ER and nuclear envelope (NE) in transfected cells (Fig. 2, Paper II). Upon activation with Ca2+ ionophore A23187, 5-­‐LO translocated from cytosol/nucleus to ER and NE (Fig. 3, Paper II). Both FLAP and LTC4S interacted with 5-­‐LO in vitro (Fig. 1, Paper II). 5-­‐LO and FLAP have previously been shown to interact by fluorescence lifetime imaging microscopy (FLIM) and immunoprecipitation [116] but no interaction between 5-­‐LO and LTC4S was reported. Using GST-­‐pulldown technique, we showed preferential interaction between 5-­‐LO and the second hydrophilic segment of LTC4S in vitro (Fig. 1, Paper II). In contrast, both hydrophilic loops of FLAP bound 5-­‐LO (Fig. 1, Paper II). Thus the binding of 5-­‐LO to FLAP and LTC4S, respectively, appears to be different, even though FLAP and LTC4S have a very similar 3-­‐D structure. FLAP and LTC4S interacted via hydrophobic regions of LTC4S (Fig. 1, Paper II), suggesting that membrane-­‐
spanning regions of one LTC4S trimer contacts membrane-­‐spanning regions of a neighboring FLAP trimer. Alternatively, heterotrimers or higher multimers might form (see below). The interaction between LTC4S and FLAP was more efficient with an N-­‐terminal part of LTC4S than with the full length protein and interestingly this N-­‐terminal part did not bind 5-­‐LO (Fig. 1, Paper II). It is not known if formation of LTC4S and FLAP heterooligomers competes with formation of homotrimers of each protein or if larger protein complexes are formed. The in vitro finding of 5-­‐LO/ LTC4S interaction was supported by BRET experiments, which showed a similar interaction in transfected cells following activation with calcium ionophore A23187 (Fig. 4, Paper II). 55 Results and Discussion 4.4 Paper III The expression of FLAP message was investigated in mouse fetuses at different stages of development. High levels of expression was detected in liver by in situ hybridization (ISH) and fluorescence-­‐activated cell sorting (FACS) plus qRT-­‐PCR analyses. FLAP expression in liver was detected as early as e11.5 (Fig. 1, Paper III). The ISH-­‐images obtained revealed a non-­‐
homogenous patchy expression pattern. Sorting of e15.5 fetal liver hematopoietic cells and CD45-­‐ stromal cells (Fig. 2, Paper III) followed by qPCR analyses, showed that hematopoietic cells colonizing the liver during embryogenesis express FLAP (Fig. 4, Paper III). The liver is formed as a diverticulum from foregut epithelium between e8.5 and e9.5 in the mouse [473]. Hematopoietic cells are not generated in the liver but migrate there from other tissues notably the yolk sac and AGM region [426, 474]. By e10.5 the colonized liver is the major hematopoietic organ of the developing mouse embryo. The hematopoietic cells from embryonic liver were sorted as mature (CD45+/Lin+) and immature (CD45+/Lin-­‐) cells (Fig. 2, Paper III). The highest FLAP expression was found in mature hematopoietic cells, but also immature cells expressed FLAP (Fig. 4, Paper III). These results demonstrated that hematopoietic but not stromal cells expressed the FLAP gene. Of the adult hematopoietic cells that are known to express FLAP [117], neutrophils and lymphocytes are present in fetal mouse liver from e12.5 [475]. These cell types may thus be the source of, or part of the FLAP expression in the mature (CD45+, Lin+) cell population. Interestingly, significant FLAP expression was also observed in the immature hematopoietic cell (CD45+, Lin-­‐) population suggesting that FLAP may have roles prior to or during the maturation of hematopoietic cells. 56 Results and Discussion 4.5 Paper IV Additional studies of LT biosynthesis protein expression in fetal tissues revealed that in addition to FLAP (Paper III), 5-­‐LO, LTC4S and LTA4H were also expressed during fetal development (Figs. 1 & 2, Paper IV). Co-­‐
expression of the four proteins was seen in the hematopoietic cells of fetal liver (Fig. 2, Paper IV). Expression was also analyzed in adult bone marrow. LTC4S expression was very low, while higher expression was observed for 5-­‐LO, FLAP, and LTA4H. The relative expression pattern of these genes was similar in fetal liver and adult bone marrow. However LTA4H mRNA, relative to the other messages examined, was higher in fetal liver than in adult bone marrow, probably since also liver stromal cells express LTA4H. 5-­‐LO expression has been reported in granulocytes, monocytes / macrophages, mast cells, dendritic cells, Langerhans cells of the skin and B-­‐
lymphocytes while platelets, endothelial cells, T-­‐lymphocytes and erythrocytes lack expression of 5-­‐LO message [142, 143]. Expression of FLAP was previously found only in 5-­‐LO expressing cells but not in cells lacking 5-­‐LO [187]. Co-­‐expression of LTC4S and 5-­‐LO has been found in cells of myeloid lineage e.g. basophils, eosinophils, mast cells and monocytes / macrophages [220]. LTC4S has also been found in platelets, which lack 5-­‐LO, but express 12-­‐LO [221]. Platelets can form LTC4 by transcellular synthesis using LTA4 provided by other cells [95]. In mouse embryos LTA4H expression was detected in several tissues compared to the other LT biosynthetic enzymes. Also FLAP was detected at sites lacking detectable 5-­‐
LO expression. For example, thymus and spleen expressed FLAP and LTA4H but not 5-­‐LO (Fig. S1, Paper IV). A more refined sorting of hematopoietic cells revealed a similar expression pattern for cells from fetal liver and adult bone marrow. 57 Results and Discussion The highest expression was found in mature myeloid cells (Figs. 3 & 4, Paper IV). 5-­‐LO expression was low in most of the isolated cell populations whereas FLAP expression was high in the LSK cell compartment (Fig. 4, Paper IV). Interestingly, FLAP expression in LSK cells was the second highest, after myeloid cells. This population contains the hematopoietic stem cells. Although 5-­‐LO expression in this compartment was very low (Fig 4, Paper IV) increased expression of both 5-­‐LO and FLAP has been reported after differentiation of human CD34+ cells into dendritic cells in vitro [460]. Our results suggested a role for leukotrienes in hematopoietic regulation. To probe the functional importance of LTs, or LT-­‐independent FLAP effects in the hematopoietic system, we used FLAP-­‐/-­‐ mice. HPLC analysis confirmed that these mice did not form LTs from AA. Interestingly, a significant reduction of 5(S)-­‐HETE formation after incubations of bone marrow cells with AA + Ca2+ ionophore accompanied the FLAP deficiency (Figs. 5 & S2, Paper IV). We analyzed peripheral blood and bone marrow from FLAP deficient mice and control animals using flow cytometry and detected an increased frequency of T-­‐lymphocytes and a decreased frequency of B-­‐lymphocytes in peripheral blood (Fig. 6, Paper IV). A slight, but not statistically significant, reduction of total white blood cells was also observed. When bone marrow was investigated, no difference was seen between FLAP knockouts and controls for B-­‐cells or B-­‐cell progenitors (Fig. 8 and table III, Paper IV), but a statistically significant increase of common lymphoid progenitors (CLP) was found in the knockout animals (Fig. 7 and table II, Paper IV). 58 Results and Discussion Interestingly, our experiments with FLAP knockout mice showed significant differences between these animals and control mice in bone marrow and peripheral blood cell compositions. No such effects were observed in another study using 5-­‐LO knockout mice [469]. However, effects on spleen B-­‐ and helper T-­‐cell composition have been reported in 5-­‐LO-­‐/-­‐ mice [470]. In summary our results suggest a mild block of B-­‐cell differentiation at an early stage of lineage commitment in FLAP knockout mice. The CLP population of bone marrow is considered to develop in vivo into mainly B-­‐
lymphocytes (reviewed in [445]). Interestingly, 5-­‐LO knockout mice had a tendency to reduce the CLP compartment in BM [469]. The distinctly different results obtained with FLAP-­‐/-­‐ compared to 5-­‐LO-­‐/-­‐ mice may suggest important LT-­‐independent functions of FLAP in hematopoiesis. 59 60 Conclusions 5. CONCLUSIONS The key enzyme in CysLT biosynthesis, LTC4S has a restricted expression. This may be regulated differently depending on cell type. A transfected sequence containing 1230 bp of the upstream promoter region and LTC4S intron 2 regulated GFP expression in a LTC4S-­‐specific manner. This included upregulated expression in THP-­‐1 and RBL-­‐1 cells after treatment with TPA and in RBL-­‐1 after RA treatment. The LTC4S promoter plus intron 2 or intron 1 also drove expression of GFP in a LTC4S specific manner in transgenic mice as judged by GFP expression in white blood cells and fetal liver. The leukotriene synthesis protein machinery is arranged around the nuclear envelope and ER where FLAP and LTC4S reside as integral membrane proteins. LTC4S interacted with FLAP in vitro through hydrophobic, membrane spanning regions. FLAP and LTC4S also interacted with 5-­‐LO, which translocates from cytosol and nuclear matrix to the ER and nuclear envelope upon cell activation. The complete protein machinery for production of proinflammatory leukotrienes was expressed already at stage e11.5 during mouse embryogenesis and is restricted to the hematopoietic cells of the liver. 61 Conclusions Expression of LT synthesis proteins was similar in various hematopoietic populations of fetal liver and adult bone marrow. FLAP, but not 5-­‐LO mRNA was upregulated in hematopoietic stem and progenitor cells and there was a decreased ratio of B to T-­‐lymphocytes in peripheral blood and an increased ratio of common lymphoid progenitor cells in FLAP-­‐/-­‐ animals, suggesting a block in lymphocyte maturation in the absence of LT-­‐
biosynthesis. 62 General discussion and future perspectives 6. GENERAL DISCUSSION AND FUTURE PERSPECTIVES Following the discovery of leukotrienes they were considered as mediators of allergic and “classical” inflammatory disorders. More recently however, it has been realized that LTs are important for a much wider range of diseases such as cardiovascular disease [389-­‐391] including ischemic brain injury [242, 337, 392-­‐398] and in cancer (reviewed in [149, 150, 422]). A few case studies have suggested CysLT roles during fetal development of the central nervous system, where congenital lack of LTC4S was associated with developmental CNS abnormalities [244]. Several reports, including our work in this thesis, suggest a possible role for LTs in hematopoietic regulation [269, 460-­‐470] (Papers III & IV). Other reports suggest LTs as important factors for cancer cells, possibly implying direct effects on regulation of cell differentiation and proliferation. Further insights to roles of leukotrienes have been provided by studies of gene knockout animals. However, generation of these mice gave somewhat surprising results. Animals lacking functional genes for 5-­‐LO, FLAP or LTC4S did not give profound phenotypes [183, 243, 476]. This could mean that LTs are not essential regulators of embryonic development and cell differentiation, or that redundant regulating systems exist. The results are however, compatible with LT roles as fine-­‐tuning regulators of cell differentiation, as suggested in Paper IV, as well as with roles in induction or maintenance of malignant cell states. 63 General discussion and future perspectives The impaired LT synthesis in knockout mice is associated with a reduction of certain inflammatory reactions [183, 243, 476], which in many cases may be favorable. However, recent reports have shown also negative effects of impaired LT synthesis (reviewed in [286]). Thus, production of LTs needs to be well balanced and a shift from normal LT synthesis, in either direction, may have adverse effects. Too much LTs will lead to hyperreactive inflammatory disorders like asthma and allergy. Todays diet consumed in the western world is rich in ω-­‐6 PUFAs and leads to an increased cell content of AA [14-­‐23]. Hence, this could lead to an increased potential of leukotriene production that may in part explain the increased frequency of asthma and other allergic disorders observed in several parts of primarily the industrialized world [477]. Too little LTs on the other hand might lead to impaired inflammatory responses and a reduced defense capacity to microbe infections. Changes in LT production in either direction may also have mild effects on differentiation of e.g. hematopoietic cells, and possibly also other tissues where the outcome still needs to be explored. The effects observed in FLAP null mice on the lymphocyte lineage differentiation are interesting and should be investigated further. FLAP is essential for LT production from endogenous AA. In addition, FLAP affected the yield of metabolites formed by 12-­‐lipoxygenase [185]. This suggests that studies of 5-­‐LO-­‐ and LT-­‐ independent effects of FLAP may be another interesting field for investigations. Pharmaceutical research has been directed to the development of LT synthesis inhibitors and antagonists of LT receptors. The medical indications to use such drugs are primarily asthma and allergic rhinitis, but the involvement of LTs in other inflammatory conditions suggests that LT modulating drugs could be beneficial also in other diseases. 64 General discussion and future perspectives The ability to pharmacologically increase LT effects with receptor agonists might also be favorable in certain cases, such as malnutrition, vitamin D deficiency, HIV infection, liver cirrhosis or diabetes [412-­‐419], where endogenous production is low. To allow a broader use of drugs affecting the LT system drugs of greater specificity will probably be needed. Highly specific agonists or receptor-­‐blockers may be of great clinical importance since the effects mediated by the different subclasses of receptors in certain cells have been reported to oppose each other. Detailed information regarding protein-­‐protein interactions taking place between the proteins of the LT machinery (cf. Paper II) may provide information which will aid the development of new more specific modulators of the LT synthesis machinery, such as peptides designed to compete at interaction sites between LT synthesis proteins. Studies regarding expression and function of LTs (Papers III and IV), contribute to better understanding of where beneficial or detrimental LT synthesis can be expected. Hence, indicate when and where there may be a gain to pharmacologically modulate production or actions of LTs. In the future, pharmacological modulation of the LT system will probably be used for an increasing number of disorders. The data presented in this thesis may indicate possibilities for new therapeutic approaches to diseases such as malignancies of the blood forming system using agents directed against the leukotriene system. 65 66 Acknowledgements 7. ACKNOWLEDGEMENTS 7.1 Financial support The work of this thesis was financially supported by: The Swedish Research Council, projects 31X-­‐5914 & 31BI-­‐14751 Östergötlands County Council Research Fund Märtha Lundqvists stiftelse Stiftelsen Lars Hiertas minne Fredrik och Ingrid Thurings stiftelse Stiftelsen för Gamla Tjänarinnor The Swedish Foundation for Strategic Research Lions forskningsfond mot folksjukdomar The Swedish Cancer Society (to Mikael Sigvardsson) 7.2 Personal thank you/Personligt tack The work behind a thesis like this is not solely the work of a single person. In addition to myself, several others have, directly or indirectly, contributed so that this book was ever written. Some of you were directly involved in the research work. Others were there as colleagues and friends and as parts of our nice milieu in the Division of cell biology in general and at floor 12 in particular. I would also like to acknowledge friends and family outside the university. All of you deserve a thank you… First of all, my supervisors Sven Hammarström; Thank you for giving me the opportunity to do the work, which has now resulted in this thesis and for sharing your great knowledge and amazing proof reading skills. 67 Acknowledgements Mats Söderström; Thank you for sharing your great methodological knowledge in the lab. Thanks to you a lot of trial and error experiments did not need to be done. Thank you also for being, not only a supervisor, but also a friend at work and outside of work and for sharing the great interest in Ice hockey. Former members of the Sven and Mats lab, either as undergraduate students or PhD students: Jesper, my predecessor as a PhD student working for Sven and Mats. I started as an undergraduate student when you were still there but we did not get to work that much together and you left before I started my PhD studies. However you still made a lasting impression on me and in the lab and I guess on everybody working on floor 12 (not until recently people dare to eat directly out of their lunch boxes again J). Elisabeth, when I started you were already there as a student from “forskarskolan”. During our time together in the lab we worked a lot together on similar projects and had a lot of fun (at least I did, I´m not sure about you since you first moved to the next room and later across the Atlantic ocean). Kristina, you were also a part of the lab when I started. Even though you did not work in the LT field you did not hesitate to help me with my questions. That you have left the lab is very obvious; I have not heard E-­‐type on the stereo since. Björn and Cissi started off as undergraduate students in our lab when I had started my run towards the PhD degree. We had a lot of fun working together, it is always more fun than working alone. But after a while Björn climbed to floor 13 to work with trees and Cissi moved to “Strålforskningsgruppen” and left me doing the DIPPING alone J. 68 Acknowledgements Mikael Sigvardsson and Hong Qiang for a lot of help with Papers III and IV and the hematopoietic connection in this work. A lot of the work behind these papers could not have been made without your help. Micke; thanks for letting me work with this in your lab and thanks also for taking the time to talk science, always in a very enthusiastic way. Hong thanks for taking the time to help me perform a lot of work using FACS and also for introducing me to the field of hematopoiesis. Thanks also for being a patient teacher regarding questions of hematopoiesis and cell sorting. Michael G Rosenfeld and Christopher K Glass at UCSD for giving me the opportunity to learn important molecular techniques and Jie Zhang in the Rosenfeld lab for being an excellent teacher of in situ hybridization. Johan Ruud in the Blomqvist lab for pointers on DAB immuno, even though I abandoned your floating method. Thanks also for many hours of teaching histology together, to more or less motivated students. Åsa and Anette in ”cellkärnan” for help with numerous sequences and questions over the years and for always being friendly and nice. All other PhD student colleagues and friends at floor 12: Sebastian “Tysken” Schultz, my current room-­‐mate. Thanks for all our good talks in the office. Those could deal with scientific questions as well as completely trivial things, sometimes just moaning about stuff together. It has been great to share office with a friend who understands the situation you are in. In many things we are very alike in others we are not that alike. In this part of YOUR thesis you suggested that I might join you on a camping trip. We will see, but may you first explain to me what that is?? J. 69 Acknowledgements My former roommates; Johan, Sofia and Veronica. It was great fun sharing office with all of you. Since you left the room it is bit quieter and (for some reason) seems to have a less active phone. All other present or former PhD student colleagues at floor 12; Åsa, Siri, Ia: it has been nice to always be able to stop by your office for a chat. I have missed you all during your times away from work. Marie, too bad that you left for Uppsala, you were great fun to have around, always one comment away from a laugh. Niclas thanks for nice discussions both on science and other stuff and for great “kräftskivor” at your “cottage”. Thanks also to everybody who has worked or works at “tolvan” for being a part of the nice atmosphere on our floor over the last 5-­‐6 years. Other group leaders at floor 12 during my time here. It would have been a very different place without you: Gunilla Westermark: Thanks for always being helpful and for the spirit you created here and also for providing me with a lot of teaching jobs. Peter Strålfors: Thanks for many interesting and fun talks in the Lunch room and for being a big part of the nice working atmosphere at our floor. PhD student from other parts of HU whom I have got to know during my time here: Anders C, Jonas U, Johan J, Jocke H, and many more. Thanks for great fun and all great parties together. The rest of the staff in the cell biology building and in entire IKE for making it such a nice place to work in. 70 Acknowledgements Tack också till släkt och vänner utanför jobbet som också varit ett viktigt stöd för att kunna koppla bort jobbet ibland och tänka på andra saker. Mamma, Pappa, Syrran Sofia och hennes familj Pontus och Theo. Tack för att ni alltid är uppmuntrande även om ni inte har en aning om vad det är jag sysslat med på jobbet. And last but not least: Victoria, ÄNTLIGEN tror jag är ett ord som kan beskriva dina känslor kring att den här boken blivit klar. Trots att du stundtals varit less på att “det där doktorerandet” tar sån tid så har du ändå alltid stöttat och peppat och bara varit där.
71 72 References 8. REFERENCES 1. 2. 3. 4. 5. 6. 7. 8. 9. 10. 11. 12. 13. 14. 15. 16. 17. 18. 19. Cohen S. The role of cell-­‐mediated immunity in the induction of inflammatory responses. Parke-­‐Davis Award Lecture, 1977. Am J Pathol 1977, 88:502-­‐28. Koj A. Cytokines regulating acute inflammation and synthesis of acute phase proteins. Blut 1985, 51:267-­‐74. Paukov VS, Kaufman O. [The interrelation of the "local" and the "general" in inflammation]. Arkh Patol 1988, 50:7-­‐16. de Weck AL, Stadler BM, Dahinden CA. New perspectives in the modulation of allergic inflammation. Int Arch Allergy Appl Immunol 1989, 90 Suppl 1:17-­‐21. van der Poll T, van Zoelen MA, Wiersinga WJ. Regulation of pro-­‐and anti-­‐
inflammatory host responses. Contrib Microbiol 2011, 17:125-­‐36. Murakami M. Lipid mediators in life science. Exp Anim 2011, 60:7-­‐20. Calder PC. Polyunsaturated fatty acids and inflammation. Prostaglandins Leukot Essent Fatty Acids 2006, 75:197-­‐202. Das UN. Essential fatty acids: biochemistry, physiology and pathology. Biotechnol J 2006, 1:420-­‐39. Corey EJ, Niwa H, Falck JR, Mioskowski C, Arai Y, Marfat A. Recent studies on the chemical synthesis of eicosanoids. Adv Prostaglandin Thromboxane Res 1980, 6:19-­‐25. Crawford MA. Background to essential fatty acids and their prostanoid derivatives. Br Med Bull 1983, 39:210-­‐3. Leslie CC. Regulation of arachidonic acid availability for eicosanoid production. Biochem Cell Biol 2004, 82:1-­‐17. Nakamura MT, Nara TY. Essential fatty acid synthesis and its regulation in mammals. Prostaglandins Leukot Essent Fatty Acids 2003, 68:145-­‐50. Yamashita A, Sugiura T, Waku K. Acyltransferases and transacylases involved in fatty acid remodeling of phospholipids and metabolism of bioactive lipids in mammalian cells. J Biochem 1997, 122:1-­‐16. Eaton SB, Konner M. Paleolithic nutrition. A consideration of its nature and current implications. N Engl J Med 1985, 312:283-­‐9. Simopoulos AP. Omega-­‐3 fatty acids in health and disease and in growth and development. Am J Clin Nutr 1991, 54:438-­‐63. Simopoulos AP. Is insulin resistance influenced by dietary linoleic acid and trans fatty acids? Free Radic Biol Med 1994, 17:367-­‐72. Lee TH, Hoover RL, Williams JD, Sperling RI, Ravalese J 3rd, Spur BW, Robinson DR, Corey EJ, Lewis RA, Austen KF. Effect of dietary enrichment with eicosapentaenoic and docosahexaenoic acids on in vitro neutrophil and monocyte leukotriene generation and neutrophil function. N Engl J Med 1985, 312:1217-­‐24. Endres S, Ghorbani R, Kelley VE, Georgilis K, Lonnemann G, van der Meer JW, Cannon JG, Rogers TS, Klempner MS, Weber PC et al. The effect of dietary supplementation with n-­‐3 polyunsaturated fatty acids on the synthesis of interleukin-­‐1 and tumor necrosis factor by mononuclear cells. N Engl J Med 1989, 320:265-­‐71. Sperling RI, Benincaso AI, Knoell CT, Larkin JK, Austen KF, Robinson DR. Dietary omega-­‐3 polyunsaturated fatty acids inhibit phosphoinositide formation and chemotaxis in neutrophils. J Clin Invest 1993, 91:651-­‐60. 73 20. 21. 22. 23. 24. 25. 26. 27. 28. 29. 30. 31. 32. 33. 34. 35. 36. Gibney MJ, Hunter B. The effects of short-­‐ and long-­‐term supplementation with fish oil on the incorporation of n-­‐3 polyunsaturated fatty acids into cells of the immune system in healthy volunteers. Eur J Clin Nutr 1993, 47:255-­‐9. Caughey GE, Mantzioris E, Gibson RA, Cleland LG, James MJ. The effect on human tumor necrosis factor alpha and interleukin 1 beta production of diets enriched in n-­‐3 fatty acids from vegetable oil or fish oil. Am J Clin Nutr 1996, 63:116-­‐22. Yaqoob P, Pala HS, Cortina-­‐Borja M, Newsholme EA, Calder PC. Encapsulated fish oil enriched in alpha-­‐tocopherol alters plasma phospholipid and mononuclear cell fatty acid compositions but not mononuclear cell functions. Eur J Clin Invest 2000, 30:260-­‐74. Healy DA, Wallace FA, Miles EA, Calder PC, Newsholm P. Effect of low-­‐to-­‐
moderate amounts of dietary fish oil on neutrophil lipid composition and function. Lipids 2000, 35:763-­‐8. von Schacky C, Kiefl R, Jendraschak E, Kaminski WE. n-­‐3 fatty acids and cysteinyl-­‐
leukotriene formation in humans in vitro, ex vivo, and in vivo. J Lab Clin Med 1993, 121:302-­‐9. Hawkes JS, James MJ, Cleland LG. Separation and quantification of PGE3 following derivatization with panacyl bromide by high pressure liquid chromatography with fluorometric detection. Prostaglandins 1991, 42:355-­‐68. Goldman DW, Pickett WC, Goetzl EJ. Human neutrophil chemotactic and degranulating activities of leukotriene B5 (LTB5) derived from eicosapentaenoic acid. Biochem Biophys Res Commun 1983, 117:282-­‐8. Lee TH, Menica-­‐Huerta JM, Shih C, Corey EJ, Lewis RA, Austen KF. Characterization and biologic properties of 5,12-­‐dihydroxy derivatives of eicosapentaenoic acid, including leukotriene B5 and the double lipoxygenase product. J Biol Chem 1984, 259:2383-­‐9. Simopoulos AP. Evolutionary aspects of diet, the omega-­‐6/omega-­‐3 ratio and genetic variation: nutritional implications for chronic diseases. Biomed Pharmacother 2006, 60:502-­‐7. Irvine RF. How is the level of free arachidonic acid controlled in mammalian cells? Biochem J 1982, 204:3-­‐16. Leslie CC. Regulation of arachidonic acid availability for eicosanoid production. Biochem Cell Biol 2004, 82:1-­‐17. Hagenfeldt L. Turnover of individual free fatty acids in man. Fed Proc 1975, 34:2246-­‐9. Uozumi N, Kume K, Nagase T, Nakatani N, Ishii S, Tashiro F, Komagata Y, Maki K, Ikuta K, Ouchi Y et al. Role of cytosolic phospholipase A2 in allergic response and parturition. Nature 1997, 390:618-­‐22. Bonventre JV, Huang Z, Taheri MR, O'Leary E, Li E, Moskowitz MA, Sapirstein A. Reduced fertility and postischaemic brain injury in mice deficient in cytosolic phospholipase A2. Nature 1997, 390:622-­‐5. Clark JD, Schievella AR, Nalefski EA, Lin LL. Cytosolic phospholipase A2. J Lipid Mediat Cell Signal 1995, 12:83-­‐117. Leslie CC. Properties and regulation of cytosolic phospholipase A2. J Biol Chem 1997, 272:16709-­‐12. Lin LL, Wartmann M, Lin AY, Knopf JL, Seth A, Davis RJ. cPLA2 is phosphorylated and activated by MAP kinase. Cell 1993, 72:269-­‐78. References 74 37. 38. 39. 40. 41. 42. 43. 44. 45. 46. 47. 48. 49. 50. Nemenoff RA, Winitz S, Qian NX, Van Putten V, Johnson GL, Heasley LE. Phosphorylation and activation of a high molecular weight form of phospholipase A2 by p42 microtubule-­‐associated protein 2 kinase and protein kinase C. J Biol Chem 1993, 268:1960-­‐4. Hefner Y, Borsch-­‐Haubold AG, Murakami M, Wilde JI, Pasquet S, Schieltz D, Ghomashchi F, Yates JR 3rd, Armstrong CG, Paterson A et al. Serine 727 phosphorylation and activation of cytosolic phospholipase A2 by MNK1-­‐related protein kinases. J Biol Chem 2000, 275:37542-­‐51. Muthalif MM, Hefner Y, Canaan S, Harper J, Zhou H, Parmentier JH, Aebersold R, Gelb MH, Malik KU. Functional interaction of calcium-­‐/calmodulin-­‐dependent protein kinase II and cytosolic phospholipase A2. J Biol Chem 2001, 276:39653-­‐
60. Dessen A, Tang J, Schmidt H, Stahl M, Clark JD, Seehra J, Somers WS. Crystal structure of human cytosolic phospholipase A2 reveals a novel topology and catalytic mechanism. Cell 1999, 97:349-­‐60. Gijon MA, Spencer DM, Kaiser AL, Leslie CC. Role of phosphorylation sites and the C2 domain in regulation of cytosolic phospholipase A2. J Cell Biol 1999, 145:1219-­‐
32. Channon JY, Leslie CC. A calcium-­‐dependent mechanism for associating a soluble arachidonoyl-­‐hydrolyzing phospholipase A2 with membrane in the macrophage cell line RAW 264.7. J Biol Chem 1990, 265:5409-­‐13. Clark JD, Lin LL, Kriz RW, Ramesha CS, Sultzman LA, Lin AY, Milona N, Knopf JL. A novel arachidonic acid-­‐selective cytosolic PLA2 contains a Ca2+-­‐dependent translocation domain with homology to PKC and GAP. Cell 1991, 65:1043-­‐51. Glover S, de Carvalho MS, Bayburt T, Jonas M, Chi E, Leslie CC, Gelb MH. Translocation of the 85-­‐kDa phospholipase A2 from cytosol to the nuclear envelope in rat basophilic leukemia cells stimulated with calcium ionophore or IgE/antigen. J Biol Chem 1995, 270:15359-­‐67. Hirabayashi T, Kume K, Hirose K, Yokomizo T, Iino M, Itoh H, Shimizu T. Critical duration of intracellular Ca2+ response required for continuous translocation and activation of cytosolic phospholipase A2. J Biol Chem 1999, 274:5163-­‐9. Perisic O, Paterson HF, Mosedale G, Lara-­‐Gonzalez S, Williams RL. Mapping the phospholipid-­‐binding surface and translocation determinants of the C2 domain from cytosolic phospholipase A2. J Biol Chem 1999, 274:14979-­‐87. Evans JH, Spencer DM, Zweifach A, Leslie CC. Intracellular calcium signals regulating cytosolic phospholipase A2 translocation to internal membranes. J Biol Chem 2001, 276:30150-­‐60. Neufeld EJ, Majerus PW, Krueger CM, Saffitz JE. Uptake and subcellular distribution of [3H]arachidonic acid in murine fibrosarcoma cells measured by electron microscope autoradiography. J Cell Biol 1985, 101:573-­‐81. Capriotti AM, Furth EE, Arrasmith ME, Laposata M. Arachidonate released upon agonist stimulation preferentially originates from arachidonate most recently incorporated into nuclear membrane phospholipids. J Biol Chem 1988, 263:10029-­‐34. Makita K, Falck JR, Capdevila JH. Cytochrome P450, the arachidonic acid cascade, and hypertension: new vistas for an old enzyme system. Faseb J 1996, 10:1456-­‐
63. References 75 51. 52. 53. 54. 55. 56. 57. 58. 59. 60. 61. 62. 63. 64. 65. 66. 67. 68. 69. References Merlie JP, Fagan D, Mudd J, Needleman P. Isolation and characterization of the complementary DNA for sheep seminal vesicle prostaglandin endoperoxide synthase (cyclooxygenase). J Biol Chem 1988, 263:3550-­‐3. Simmons DL, Levy DB, Yannoni Y, Erikson RL. Identification of a phorbol ester-­‐
repressible v-­‐src-­‐inducible gene. Proc Natl Acad Sci U S A 1989, 86:1178-­‐82. Xie WL, Chipman JG, Robertson DL, Erikson RL, Simmons DL. Expression of a mitogen-­‐responsive gene encoding prostaglandin synthase is regulated by mRNA splicing. Proc Natl Acad Sci U S A 1991, 88:2692-­‐6. Rollins TE, Smith WL. Subcellular localization of prostaglandin-­‐forming cyclooxygenase in Swiss mouse 3T3 fibroblasts by electron microscopic immunocytochemistry. J Biol Chem 1980, 255:4872-­‐5. Li Y, Smith T, Grabski S, DeWitt DL. The membrane association sequences of the prostaglandin endoperoxide synthases-­‐1 and -­‐2 isozymes. J Biol Chem 1998, 273:29830-­‐7. Zurier RB. Prostaglandins: then and now and next. Semin Arthritis Rheum 2003, 33:137-­‐9. Ricciotti E, Fitzgerald GA. Prostaglandins and inflammation. Arterioscler Thromb Vasc Biol 2011, 31:986-­‐1000. Wolfe LS. Eicosanoids: prostaglandins, thromboxanes, leukotrienes, and other derivatives of carbon-­‐20 unsaturated fatty acids. J Neurochem 1982, 38:1-­‐14. Hamberg M, Samuelsson B. On the specificity of the oxygenation of unsaturated fatty acids catalyzed by soybean lipoxidase. J Biol Chem 1967, 242:5329-­‐35. Brash AR. Lipoxygenases: occurrence, functions, catalysis, and acquisition of substrate. J Biol Chem 1999, 274:23679-­‐82. Dobrian AD, Lieb DC, Cole BK, Taylor-­‐Fishwick DA, Chakrabarti SK, Nadler JL. Functional and pathological roles of the 12-­‐ and 15-­‐lipoxygenases. Prog Lipid Res 2011, 50:115-­‐31. Serhan CN, Samuelsson B. Lipoxins: a new series of eicosanoids (biosynthesis, stereochemistry, and biological activities). Adv Exp Med Biol 1988, 229:1-­‐14. Ryan A, Godson C. Lipoxins: regulators of resolution. Curr Opin Pharmacol 2010, 10:166-­‐72. Pidgeon GP, Lysaght J, Krishnamoorthy S, Reynolds JV, O'Byrne K, Nie D, Honn KV. Lipoxygenase metabolism: roles in tumor progression and survival. Cancer Metastasis Rev 2007, 26:503-­‐24. Zarini S, Gijon MA, Folco G, Murphy RC. Effect of arachidonic acid reacylation on leukotriene biosynthesis in human neutrophils stimulated with granulocyte-­‐
macrophage colony-­‐stimulating factor and formyl-­‐methionyl-­‐leucyl-­‐
phenylalanine. J Biol Chem 2006, 281:10134-­‐42. Luo M, Jones SM, Peters-­‐Golden M, Brock TG. Nuclear localization of 5-­‐
lipoxygenase as a determinant of leukotriene B4 synthetic capacity. Proc Natl Acad Sci U S A 2003, 100:12165-­‐70. Werz O. 5-­‐lipoxygenase: cellular biology and molecular pharmacology. Curr Drug Targets Inflamm Allergy 2002, 1:23-­‐44. Samuelsson B, Funk CD. Enzymes involved in the biosynthesis of leukotriene B4. J Biol Chem 1989, 264:19469-­‐72. Hammarström S. Leukotrienes. Annu Rev Biochem 1983, 52:355-­‐77. 76 70. 71. 72. 73. 74. 75. 76. 77. 78. 79. 80. 81. 82. 83. 84. 85. 86. References Borgeat P, Hamberg M, Samuelsson B. Transformation of arachidonic acid and homo-­‐γ-­‐linolenic acid by rabbit polymorphonuclear leukocytes. Monohydroxy acids from novel lipoxygenases. J Biol Chem 1976, 251:7816-­‐20. Maas RL, Ingram CD, Taber DF, Oates JA, Brash AR. Stereospecific removal of the DR hydrogen atom at the 10-­‐carbon of arachidonic acid in the biosynthesis of leukotriene A4 by human leukocytes. J Biol Chem 1982, 257:13515-­‐9. Panossian A, Hamberg M, Samuelsson B. On the mechanism of biosynthesis of leukotrienes and related compounds. FEBS Lett 1982, 150:511-­‐3. Hammarström S. Stereospecific elimination of hydrogen at C-­‐10 in eicosapentaenoic acid during the conversion to leukotriene C5. J Biol Chem 1983, 258:1427-­‐30. Mori M, Izumi T, Shimizu T. Energy-­‐dependent export of leukotriene B4 in Xenopus oocytes. Biochim Biophys Acta 1993, 1168:23-­‐9. Lam BK, Gagnon L, Austen KF, Soberman RJ. The mechanism of leukotriene B4 export from human polymorphonuclear leukocytes. J Biol Chem 1990, 265:13438-­‐41. Murphy RC, Hammarström S, Samuelsson B. Leukotriene C: a slow-­‐reacting substance from murine mastocytoma cells. Proc Natl Acad Sci U S A 1979, 76:4275-­‐9. Hammarström S, Murphy RC, Samuelsson B, Clark DA, Mioskowski C, Corey EJ. Structure of leukotriene C. Identification of the amino acid part. Biochem Biophys Res Commun 1979, 91:1266-­‐72. Brocklehurst WE. "SRS-­‐A" The slow reacting substance of anaphylaxis. Biochem Pharmacol 1963, 12:431-­‐5. Örning L, Hammarström S, Samuelsson B. Leukotriene D: a slow reacting substance from rat basophilic leukemia cells. Proc Natl Acad Sci U S A 1980, 77:2014-­‐7. Lewis RA, Austen KF, Drazen JM, Clark DA, Marfat A, Corey EJ. Slow reacting substances of anaphylaxis: identification of leukotrienes C-­‐1 and D from human and rat sources. Proc Natl Acad Sci U S A 1980, 77:3710-­‐4. Lewis RA, Drazen JM, Austen KF, Clark DA, Corey EJ. Identification of the C6-­‐S-­‐
conjugate of leukotriene A with cysteine as a naturally occurring slow reacting substance of anaphylaxis (SRS-­‐A). Importance of the 11-­‐cis-­‐geometry for biological activity. Biochem Biophys Res Commun 1980, 96:271-­‐7. Bernström K, Hammarström S. Metabolism of leukotriene D by porcine kidney. J Biol Chem 1981, 256:9579-­‐82. Anderson ME, Allison RD, Meister A. Interconversion of leukotrienes catalyzed by purified gamma-­‐glutamyl transpeptidase: concomitant formation of leukotriene D4 and gamma-­‐glutamyl amino acids. Proc Natl Acad Sci U S A 1982, 79:1088-­‐91. Carter BZ, Shi ZZ, Barrios R, Lieberman MW. γ -­‐glutamyl leukotrienase, a γ -­‐
glutamyl transpeptidase gene family member, is expressed primarily in spleen. J Biol Chem 1998, 273:28277-­‐85. Lee CW, Lewis RA, Corey EJ, Austen KF. Conversion of leukotriene D4 to leukotriene E4 by a dipeptidase released from the specific granule of human polymorphonuclear leucocytes. Immunology 1983, 48:27-­‐35. Lam BK, Owen WF, Jr., Austen KF, Soberman RJ. The identification of a distinct export step following the biosynthesis of leukotriene C4 by human eosinophils. J Biol Chem 1989, 264:12885-­‐9. 77 87. References Lam BK, Xu K, Atkins MB, Austen KF. Leukotriene C4 uses a probenecid-­‐sensitive export carrier that does not recognize leukotriene B4. Proc Natl Acad Sci U S A 1992, 89:11598-­‐602. 88. Leier I, Jedlitschky G, Buchholz U, Cole SP, Deeley RG, Keppler D. The MRP gene encodes an ATP-­‐dependent export pump for leukotriene C4 and structurally related conjugates. J Biol Chem 1994, 269:27807-­‐10. 89. Fitzpatrick F, Liggett W, McGee J, Bunting S, Morton D, Samuelsson B. Metabolism of leukotriene A4 by human erythrocytes. A novel cellular source of leukotriene B4. J Biol Chem 1984, 259:11403-­‐7. 90. Dahinden CA, Clancy RM, Gross M, Chiller JM, Hugli TE. Leukotriene C4 production by murine mast cells: evidence of a role for extracellular leukotriene A4. Proc Natl Acad Sci U S A 1985, 82:6632-­‐6. 91. McGee JE, Fitzpatrick FA. Erythrocyte-­‐neutrophil interactions: formation of leukotriene B4 by transcellular biosynthesis. Proc Natl Acad Sci U S A 1986, 83:1349-­‐53. 92. Feinmark SJ, Cannon PJ. Endothelial cell leukotriene C4 synthesis results from intercellular transfer of leukotriene A4 synthesized by polymorphonuclear leukocytes. J Biol Chem 1986, 261:16466-­‐72. 93. Edenius C, Heidvall K, Lindgren JÅ. Novel transcellular interaction: conversion of granulocyte-­‐derived leukotriene A4 to cysteinyl-­‐containing leukotrienes by human platelets. Eur J Biochem 1988, 178:81-­‐6. 94. Maclouf J, Murphy RC, Henson PM. Transcellular biosynthesis of sulfidopeptide leukotrienes during receptor-­‐mediated stimulation of human neutrophil/platelet mixtures. Blood 1990, 76:1838-­‐44. 95. Maclouf JA, Murphy RC. Transcellular metabolism of neutrophil-­‐derived leukotriene A4 by human platelets. A potential cellular source of leukotriene C4. J Biol Chem 1988, 263:174-­‐81. 96. Brady HR, Serhan CN. Adhesion promotes transcellular leukotriene biosynthesis during neutrophil-­‐glomerular endothelial cell interactions: inhibition by antibodies against CD18 and L-­‐selectin. Biochem Biophys Res Commun 1992, 186:1307-­‐14. 97. Iversen L, Kristensen P, Grøn B, Ziboh VA, Kragballe K. Human epidermis transforms exogenous leukotriene A4 into peptide leukotrienes: possible role in transcellular metabolism. Arch Dermatol Res 1994, 286:261-­‐6. 98. Amat M, Diaz C, Vila L. Leukotriene A4 hydrolase and leukotriene C4 synthase activities in human chondrocytes: transcellular biosynthesis of Leukotrienes during granulocyte-­‐chondrocyte interaction. Arthritis Rheum 1998, 41:1645-­‐51. 99. Farias SE, Zarini S, Precht T, Murphy RC, Heidenreich KA. Transcellular biosynthesis of cysteinyl leukotrienes in rat neuronal and glial cells. J Neurochem 2007, 103:1310-­‐8. 100. Bigby TD, Meslier N. Transcellular lipoxygenase metabolism between monocytes and platelets. J Immunol 1989, 143:1948-­‐54. 101. Levy BD. Resolvins and protectins: natural pharmacophores for resolution biology. Prostaglandins Leukot Essent Fatty Acids 2010, 82:327-­‐32. 102. Matsumoto T, Funk CD, Rådmark O, Höög JO, Jörnvall H, Samuelsson B. Molecular cloning and amino acid sequence of human 5-­‐lipoxygenase. Proc Natl Acad Sci U S A 1988, 85:26-­‐30. 78 References 103. Dixon RA, Jones RE, Diehl RE, Bennett CD, Kargman S, Rouzer CA. Cloning of the cDNA for human 5-­‐lipoxygenase. Proc Natl Acad Sci U S A 1988, 85:416-­‐20. 104. Schilstra MJ, Veldink GA, Vliegenthart JF. The dioxygenation rate in lipoxygenase catalysis is determined by the amount of iron (III) lipoxygenase in solution. Biochemistry 1994, 33:3974-­‐9. 105. Hammarberg T, Kuprin S, Rådmark O, Holmgren A. EPR investigation of the active site of recombinant human 5-­‐lipoxygenase: inhibition by selenide. Biochemistry 2001, 40:6371-­‐8. 106. Gilbert NC, Bartlett SG, Waight MT, Neau DB, Boeglin WE, Brash AR, Newcomer ME. The structure of human 5-­‐lipoxygenase. Science 2011, 331:217-­‐9. 107. Luo M, Flamand N, Brock TG. Metabolism of arachidonic acid to eicosanoids within the nucleus. Biochim Biophys Acta 2006, 1761:618-­‐25. 108. Brock TG, Paine R 3rd, Peters-­‐Golden M. Localization of 5-­‐lipoxygenase to the nucleus of unstimulated rat basophilic leukemia cells. J Biol Chem 1994, 269:22059-­‐66. 109. Brock TG, McNish RW, Peters-­‐Golden M. Translocation and leukotriene synthetic capacity of nuclear 5-­‐lipoxygenase in rat basophilic leukemia cells and alveolar macrophages. J Biol Chem 1995, 270:21652-­‐8. 110. Brock TG, Anderson JA, Fries FP, Peters-­‐Golden M, Sporn PH. Decreased leukotriene C4 synthesis accompanies adherence-­‐dependent nuclear import of 5-­‐
lipoxygenase in human blood eosinophils. J Immunol 1999, 162:1669-­‐76. 111. Chen XS, Naumann TA, Kurre U, Jenkins NA, Copeland NG, Funk CD. cDNA cloning, expression, mutagenesis, intracellular localization, and gene chromosomal assignment of mouse 5-­‐lipoxygenase. J Biol Chem 1995, 270:17993-­‐9. 112. Woods JW, Coffey MJ, Brock TG, Singer, II, Peters-­‐Golden M. 5-­‐Lipoxygenase is located in the euchromatin of the nucleus in resting human alveolar macrophages and translocates to the nuclear envelope upon cell activation. J Clin Invest 1995, 95:2035-­‐46. 113. Healy AM, Peters-­‐Golden M, Yao JP, Brock TG. Identification of a bipartite nuclear localization sequence necessary for nuclear import of 5-­‐lipoxygenase. J Biol Chem 1999, 274:29812-­‐8. 114. Rouzer CA, Kargman S. Translocation of 5-­‐lipoxygenase to the membrane in human leukocytes challenged with ionophore A23187. J Biol Chem 1988, 263:10980-­‐8. 115. Wong A, Hwang SM, Cook MN, Hogaboom GK, Crooke ST. Interactions of 5-­‐
lipoxygenase with membranes: studies on the association of soluble enzyme with membranes and alterations in enzyme activity. Biochemistry 1988, 27:6763-­‐9. 116. Mandal AK, Jones PB, Bair AM, Christmas P, Miller D, Yamin TT, Wisniewski D, Menke J, Evans JF, Hyman BT et al. The nuclear membrane organization of leukotriene synthesis. Proc Natl Acad Sci U S A 2008, 105:20434-­‐9. 117. Peters-­‐Golden M, Brock TG. 5-­‐lipoxygenase and FLAP. Prostaglandins Leukot Essent Fatty Acids 2003, 69:99-­‐109. 118. Werz O, Klemm J, Samuelsson B, Rådmark O. Phorbol ester up-­‐regulates capacities for nuclear translocation and phosphorylation of 5-­‐lipoxygenase in Mono Mac 6 cells and human polymorphonuclear leukocytes. Blood 2001, 97:2487-­‐95. 119. Hammarberg T, Rådmark O. 5-­‐lipoxygenase binds calcium. Biochemistry 1999, 38:4441-­‐7. 79 References 120. Rouzer CA, Samuelsson B. On the nature of the 5-­‐lipoxygenase reaction in human leukocytes: enzyme purification and requirement for multiple stimulatory factors. Proc Natl Acad Sci U S A 1985, 82:6040-­‐4. 121. Hammarberg T, Provost P, Persson B, Rådmark O. The N-­‐terminal domain of 5-­‐
lipoxygenase binds calcium and mediates calcium stimulation of enzyme activity. J Biol Chem 2000, 275:38787-­‐93. 122. Ochi K, Yoshimoto T, Yamamoto S, Taniguchi K, Miyamoto T. Arachidonate 5-­‐
lipoxygenase of guinea pig peritoneal polymorphonuclear leukocytes. Activation by adenosine 5'-­‐triphosphate. J Biol Chem 1983, 258:5754-­‐8. 123. Rådmark O, Samuelsson B. Regulation of 5-­‐lipoxygenase enzyme activity. Biochem Biophys Res Commun 2005, 338:102-­‐10. 124. Hörnig C, Albert D, Fischer L, Hörnig M, Rådmark O, Steinhilber D, Werz O. 1-­‐
Oleoyl-­‐2-­‐acetylglycerol stimulates 5-­‐lipoxygenase activity via a putative (phospho)lipid binding site within the N-­‐terminal C2-­‐like domain. J Biol Chem 2005, 280:26913-­‐21. 125. Provost P, Samuelsson B, Rådmark O. Interaction of 5-­‐lipoxygenase with cellular proteins. Proc Natl Acad Sci U S A 1999, 96:1881-­‐5. 126. Provost P, Doucet J, Hammarberg T, Gerisch G, Samuelsson B, Rådmark O. 5-­‐
Lipoxygenase interacts with coactosin-­‐like protein. J Biol Chem 2001, 276:16520-­‐
7. 127. Brunn G, Hey C, Wessler I, Racke K. Endogenous nitric oxide inhibits leukotriene B4 release from rat alveolar macrophages. Eur J Pharmacol 1997, 326:53-­‐60. 128. Brock TG, McNish RW, Mancuso P, Coffey MJ, Peters-­‐Golden M. Prolonged lipopolysaccharide inhibits leukotriene synthesis in peritoneal macrophages: mediation by nitric oxide and prostaglandins. Prostaglandins Other Lipid Mediat 2003, 71:131-­‐45. 129. Werz O, Steinhilber D. Selenium-­‐dependent peroxidases suppress 5-­‐lipoxygenase activity in B-­‐lymphocytes and immature myeloid cells. The presence of peroxidase-­‐insensitive 5-­‐lipoxygenase activity in differentiated myeloid cells. Eur J Biochem 1996, 242:90-­‐7. 130. Straif D, Werz O, Kellner R, Bahr U, Steinhilber D. Glutathione peroxidase-­‐1 but not -­‐4 is involved in the regulation of cellular 5-­‐lipoxygenase activity in monocytic cells. Biochem J 2000, 349:455-­‐61. 131. Brock TG. Regulating leukotriene synthesis: the role of nuclear 5-­‐lipoxygenase. J Cell Biochem 2005, 96:1203-­‐11. 132. Luo M, Pang CW, Gerken AE, Brock TG. Multiple nuclear localization sequences allow modulation of 5-­‐lipoxygenase nuclear import. Traffic 2004, 5:847-­‐54. 133. Hanaka H, Shimizu T, Izumi T. Nuclear-­‐localization-­‐signal-­‐dependent and nuclear-­‐export-­‐signal-­‐dependent mechanisms determine the localization of 5-­‐
lipoxygenase. Biochem J 2002, 361:505-­‐14. 134. Lepley RA, Muskardin DT, Fitzpatrick FA. Tyrosine kinase activity modulates catalysis and translocation of cellular 5-­‐lipoxygenase. J Biol Chem 1996, 271:6179-­‐84. 135. Hanaka H, Shimizu T, Izumi T. Stress-­‐induced nuclear export of 5-­‐lipoxygenase. Biochem Biophys Res Commun 2005, 338:111-­‐6. 136. Werz O, Klemm J, Samuelsson B, Rådmark O. 5-­‐lipoxygenase is phosphorylated by p38 kinase-­‐dependent MAPKAP kinases. Proc Natl Acad Sci U S A 2000, 97:5261-­‐6. 80 References 137. Werz O, Szellas D, Steinhilber D, Rådmark O. Arachidonic acid promotes phosphorylation of 5-­‐lipoxygenase at Ser-­‐271 by MAPK-­‐activated protein kinase 2 (MK2). J Biol Chem 2002, 277:14793-­‐800. 138. Werz O, Burkert E, Fischer L, Szellas D, Dishart D, Samuelsson B, Rådmark O, Steinhilber D. Extracellular signal-­‐regulated kinases phosphorylate 5-­‐
lipoxygenase and stimulate 5-­‐lipoxygenase product formation in leukocytes. Faseb J 2002, 16:1441-­‐3. 139. Luo M, Jones SM, Phare SM, Coffey MJ, Peters-­‐Golden M, Brock TG. Protein kinase A inhibits leukotriene synthesis by phosphorylation of 5-­‐lipoxygenase on serine 523. J Biol Chem 2004, 279:41512-­‐20. 140. Luo M, Jones SM, Flamand N, Aronoff DM, Peters-­‐Golden M, Brock TG. Phosphorylation by protein kinase a inhibits nuclear import of 5-­‐lipoxygenase. J Biol Chem 2005, 280:40609-­‐16. 141. Funk CD, Hoshiko S, Matsumoto T, Rådmark O, Samuelsson B. Characterization of the human 5-­‐lipoxygenase gene. Proc Natl Acad Sci U S A 1989, 86:2587-­‐91. 142. Rådmark O, Werz O, Steinhilber D, Samuelsson B. 5-­‐Lipoxygenase: regulation of expression and enzyme activity. Trends Biochem Sci 2007, 32:332-­‐41. 143. Steinhilber D. 5-­‐Lipoxygenase: a target for antiinflammatory drugs revisited. Curr Med Chem 1999, 6:71-­‐85. 144. Chinnici CM, Yao Y, Pratico D. The 5-­‐lipoxygenase enzymatic pathway in the mouse brain: young versus old. Neurobiol Aging 2007, 28:1457-­‐62. 145. Bigby TD, Holtzman MJ. Enhanced 5-­‐lipoxygenase activity in lung macrophages compared to monocytes from normal subjects. J Immunol 1987, 138:1546-­‐50. 146. Pueringer RJ, Bahns CC, Hunninghake GW. Alveolar macrophages have greater amounts of the enzyme 5-­‐lipoxygenase than do monocytes. J Appl Physiol 1992, 73:781-­‐6. 147. Koshino T, Takano S, Houjo T, Sano Y, Kudo K, Kihara H, Kitani S, Takaishi T, Hirai K, Ito K et al. Expression of 5-­‐lipoxygenase and 5-­‐lipoxygenase-­‐activating protein mRNAs in the peripheral blood leukocytes of asthmatics. Biochem Biophys Res Commun 1998, 247:510-­‐3. 148. Ring WL, Riddick CA, Baker JR, Munafo DA, Bigby TD. Human monocytes lose 5-­‐
lipoxygenase and FLAP as they mature into monocyte-­‐derived macrophages in vitro. Am J Physiol 1996, 271:C372-­‐7. 149. Fürstenberger G, Krieg P, Müller-­‐Decker K, Habenicht AJ. What are cyclooxygenases and lipoxygenases doing in the driver's seat of carcinogenesis? Int J Cancer 2006, 119:2247-­‐54. 150. Werz O, Steinhilber D. Therapeutic options for 5-­‐lipoxygenase inhibitors. Pharmacol Ther 2006, 112:701-­‐18. 151. Rådmark O, Werz O, Steinhilber D, Samuelsson B. 5-­‐Lipoxygenase: regulation of expression and enzyme activity. Trends Biochem Sci 2007, 32:332-­‐41. 152. Jian W, Lee SH, Williams MV, Blair IA. 5-­‐Lipoxygenase-­‐mediated endogenous DNA damage. J Biol Chem 2009, 284:16799-­‐807. 153. Uhl J, Klan N, Rose M, Entian KD, Werz O, Steinhilber D. The 5-­‐lipoxygenase promoter is regulated by DNA methylation. J Biol Chem 2002, 277:4374-­‐9. 154. Hoshiko S, Rådmark O, Samuelsson B. Characterization of the human 5-­‐
lipoxygenase gene promoter. Proc Natl Acad Sci U S A 1990, 87:9073-­‐7. 81 References 155. In KH, Silverman ES, Asano K, Beier D, Fischer AR, Keith TP, Serino K, Yandava C, De Sanctis GT, Drazen JM. Mutations in the human 5-­‐lipoxygenase gene. Clin Rev Allergy Immunol 1999, 17:59-­‐69. 156. Silverman ES, Du J, De Sanctis GT, Rådmark O, Samuelsson B, Drazen JM, Collins T. Egr-­‐1 and Sp1 interact functionally with the 5-­‐lipoxygenase promoter and its naturally occurring mutants. Am J Respir Cell Mol Biol 1998, 19:316-­‐23. 157. Silverman ES, Le L, Baron RM, Hallock A, Hjoberg J, Shikanai T, Storm van's Gravesande K, Auron PE, Lu W. Cloning and functional analysis of the mouse 5-­‐
lipoxygenase promoter. Am J Respir Cell Mol Biol 2002, 26:475-­‐83. 158. Sorg BL, Klan N, Seuter S, Dishart D, Rådmark O, Habenicht A, Carlberg C, Werz O, Steinhilber D. Analysis of the 5-­‐lipoxygenase promoter and characterization of a vitamin D receptor binding site. Biochim Biophys Acta 2006, 1761:686-­‐97. 159. Seuter S, Vaisanen S, Rådmark O, Carlberg C, Steinhilber D. Functional characterization of vitamin D responding regions in the human 5-­‐Lipoxygenase gene. Biochim Biophys Acta 2007, 1771:864-­‐72. 160. Seuter S, Sorg BL, Steinhilber D. The coding sequence mediates induction of 5-­‐
lipoxygenase expression by Smads3/4. Biochem Biophys Res Commun 2006, 348:1403-­‐10. 161. Pergola C, Dodt G, Rossi A, Neunhoeffer E, Lawrenz B, Northoff H, Samuelsson B, Rådmark O, Sautebin L, Werz O. ERK-­‐mediated regulation of leukotriene biosynthesis by androgens: a molecular basis for gender differences in inflammation and asthma. Proc Natl Acad Sci U S A 2008, 105:19881-­‐6. 162. Pergola C, Rogge A, Dodt G, Northoff H, Weinigel C, Barz D, Rådmark O, Sautebin L, Werz O. Testosterone suppresses phospholipase D, causing sex differences in leukotriene biosynthesis in human monocytes. Faseb J 2011, 25:3377-­‐87. 163. Brock TG, McNish RW, Peters-­‐Golden M. Capacity for repeatable leukotriene generation after transient stimulation of mast cells and macrophages. Biochem J 1998, 329 ( Pt 3):519-­‐25. 164. Rådmark O, Samuelsson B. 5-­‐Lipoxygenase: mechanisms of regulation. J Lipid Res 2009, 50 Suppl:S40-­‐5. 165. Peters-­‐Golden M, McNish RW. Redistribution of 5-­‐lipoxygenase and cytosolic phospholipase A2 to the nuclear fraction upon macrophage activation. Biochem Biophys Res Commun 1993, 196:147-­‐53. 166. Woods JW, Evans JF, Ethier D, Scott S, Vickers PJ, Hearn L, Heibein JA, Charleson S, Singer, II. 5-­‐lipoxygenase and 5-­‐lipoxygenase-­‐activating protein are localized in the nuclear envelope of activated human leukocytes. J Exp Med 1993, 178:1935-­‐
46. 167. Pouliot M, McDonald PP, Krump E, Mancini JA, McColl SR, Weech PK, Borgeat P. Colocalization of cytosolic phospholipase A2, 5-­‐lipoxygenase, and 5-­‐lipoxygenase-­‐
activating protein at the nuclear membrane of A23187-­‐stimulated human neutrophils. Eur J Biochem 1996, 238:250-­‐8. 168. Jakobsson PJ, Morgenstern R, Mancini J, Ford-­‐Hutchinson A, Persson B. Common structural features of MAPEG -­‐-­‐ a widespread superfamily of membrane associated proteins with highly divergent functions in eicosanoid and glutathione metabolism. Protein Sci 1999, 8:689-­‐92. 82 References 169. Rouzer CA, Ford-­‐Hutchinson AW, Morton HE, Gillard JW. MK886, a potent and specific leukotriene biosynthesis inhibitor blocks and reverses the membrane association of 5-­‐lipoxygenase in ionophore-­‐challenged leukocytes. J Biol Chem 1990, 265:1436-­‐42. 170. Miller DK, Gillard JW, Vickers PJ, Sadowski S, Leveille C, Mancini JA, Charleson P, Dixon RA, Ford-­‐Hutchinson AW, Fortin R et al. Identification and isolation of a membrane protein necessary for leukotriene production. Nature 1990, 343:278-­‐
81. 171. Dixon RA, Diehl RE, Opas E, Rands E, Vickers PJ, Evans JF, Gillard JW, Miller DK. Requirement of a 5-­‐lipoxygenase-­‐activating protein for leukotriene synthesis. Nature 1990, 343:282-­‐4. 172. Abramovitz M, Wong E, Cox ME, Richardson CD, Li C, Vickers PJ. 5-­‐lipoxygenase-­‐
activating protein stimulates the utilization of arachidonic acid by 5-­‐
lipoxygenase. Eur J Biochem 1993, 215:105-­‐11. 173. Mandal AK, Skoch J, Bacskai BJ, Hyman BT, Christmas P, Miller D, Yamin TT, Xu S, Wisniewski D, Evans JF et al. The membrane organization of leukotriene synthesis. Proc Natl Acad Sci U S A 2004, 101:6587-­‐92. 174. Plante H, Picard S, Mancini J, Borgeat P. 5-­‐Lipoxygenase-­‐activating protein homodimer in human neutrophils: evidence for a role in leukotriene biosynthesis. Biochem J 2006, 393:211-­‐8. 175. Kargman S, Vickers PJ, Evans JF. A23187-­‐induced translocation of 5-­‐lipoxygenase in osteosarcoma cells. J Cell Biol 1992, 119:1701-­‐9. 176. Coffey M, Peters-­‐Golden M, Fantone JC 3rd, Sporn PH. Membrane association of active 5-­‐lipoxygenase in resting cells. Evidence for novel regulation of the enzyme in the rat alveolar macrophage. J Biol Chem 1992, 267:570-­‐6. 177. Mancini JA, Abramovitz M, Cox ME, Wong E, Charleson S, Perrier H, Wang Z, Prasit P, Vickers PJ. 5-­‐lipoxygenase-­‐activating protein is an arachidonate binding protein. FEBS Lett 1993, 318:277-­‐81. 178. Hatzelmann A, Goossens J, Fruchtmann R, Mohrs KH, Raddatz S, Müller-­‐
Peddinghaus R. Inversely-­‐correlated inhibition of human 5-­‐lipoxygenase activity by BAY X1005 and other quinoline derivatives in intact cells and a cell-­‐free system-­‐-­‐implications for the function of 5-­‐lipoxygenase activating protein. Biochem Pharmacol 1994, 47:2259-­‐68. 179. Charleson S, Evans JF, Leger S, Perrier H, Prasit P, Wang Z, Vickers PJ. Structural requirements for the binding of fatty acids to 5-­‐lipoxygenase-­‐activating protein. Eur J Pharmacol 1994, 267:275-­‐80. 180. Vickers PJ, Adam M, Charleson S, Abramovitz M, O'Neill G, Mancini JA. Amino acid residues of 5-­‐lipoxygenase-­‐activating protein critical for the binding of leukotriene biosynthesis inhibitors. J Lipid Mediat 1993, 6:31-­‐42. 181. Vickers PJ, Adam M, Charleson S, Coppolino MG, Evans JF, Mancini JA. Identification of amino acid residues of 5-­‐lipoxygenase-­‐activating protein essential for the binding of leukotriene biosynthesis inhibitors. Mol Pharmacol 1992, 42:94-­‐102. 182. Ferguson AD, McKeever BM, Xu S, Wisniewski D, Miller DK, Yamin TT, Spencer RH, Chu L, Ujjainwalla F, Cunningham BR et al. Crystal structure of inhibitor-­‐
bound human 5-­‐lipoxygenase-­‐activating protein. Science 2007, 317:510-­‐2. 83 References 183. Byrum RS, Goulet JL, Griffiths RJ, Koller BH. Role of the 5-­‐lipoxygenase-­‐activating protein (FLAP) in murine acute inflammatory responses. J Exp Med 1997, 185:1065-­‐75. 184. Mancini JA, Waterman H, Riendeau D. Cellular oxygenation of 12-­‐
hydroxyeicosatetraenoic acid and 15-­‐hydroxyeicosatetraenoic acid by 5-­‐
lipoxygenase is stimulated by 5-­‐lipoxygenase-­‐activating protein. J Biol Chem 1998, 273:32842-­‐7. 185. Griffiths RJ, Smith MA, Roach ML, Stock JL, Stam EJ, Milici AJ, Scampoli DN, Eskra JD, Byrum RS, Koller BH et al. Collagen-­‐induced arthritis is reduced in 5-­‐
lipoxygenase-­‐activating protein-­‐deficient mice. J Exp Med 1997, 185:1123-­‐9. 186. Kennedy BP, Diehl RE, Boie Y, Adam M, Dixon RA. Gene characterization and promoter analysis of the human 5-­‐lipoxygenase-­‐activating protein (FLAP). J Biol Chem 1991, 266:8511-­‐6. 187. Reid GK, Kargman S, Vickers PJ, Mancini JA, Leveille C, Ethier D, Miller DK, Gillard JW, Dixon RA, Evans JF. Correlation between expression of 5-­‐lipoxygenase-­‐
activating protein, 5-­‐lipoxygenase, and cellular leukotriene synthesis. J Biol Chem 1990, 265:19818-­‐23. 188. Bennett CF, Chiang MY, Monia BP, Crooke ST. Regulation of 5-­‐lipoxygenase and 5-­‐
lipoxygenase-­‐activating protein expression in HL-­‐60 cells. Biochem J 1993, 289 ( Pt 1):33-­‐9. 189. Martel-­‐Pelletier J, Mineau F, Fahmi H, Laufer S, Reboul P, Boileau C, Lavigne M, Pelletier JP. Regulation of the expression of 5-­‐lipoxygenase-­‐activating protein/5-­‐
lipoxygenase and the synthesis of leukotriene B4 in osteoarthritic chondrocytes: role of transforming growth factor beta and eicosanoids. Arthritis Rheum 2004, 50:3925-­‐33. 190. Reddy KV, Serio KJ, Hodulik CR, Bigby TD. 5-­‐lipoxygenase-­‐activating protein gene expression. Key role of CCAAT/enhancer-­‐binding proteins (C/EBP) in constitutive and tumor necrosis factor (TNF) alpha-­‐induced expression in THP-­‐1 cells. J Biol Chem 2003, 278:13810-­‐8. 191. Serio KJ, Reddy KV, Bigby TD. Lipopolysaccharide induces 5-­‐lipoxygenase-­‐
activating protein gene expression in THP-­‐1 cells via a NF-­‐κB and C/EBP-­‐
mediated mechanism. Am J Physiol Cell Physiol 2005, 288:C1125-­‐33. 192. Helgadottir A, Manolescu A, Thorleifsson G, Gretarsdottir S, Jonsdottir H, Thorsteinsdottir U, Samani NJ, Gudmundsson G, Grant SF, Thorgeirsson G et al. The gene encoding 5-­‐lipoxygenase activating protein confers risk of myocardial infarction and stroke. Nat Genet 2004, 36:233-­‐9. 193. Bäck M, Sultan A, Ovchinnikova O, Hansson GK. 5-­‐Lipoxygenase-­‐activating protein: a potential link between innate and adaptive immunity in atherosclerosis and adipose tissue inflammation. Circ Res 2007, 100:946-­‐9. 194. Rådmark O, Malmsten C, Samuelsson B. Leukotriene A4: enzymatic conversion to leukotriene C4. Biochem Biophys Res Commun 1980, 96:1679-­‐87. 195. Jakschik BA, Lee LH. Enzymatic assembly of slow reacting substance. Nature 1980, 287:51-­‐2. 196. Bach MK, Brashler JR, Morton DR, Jr. Solubilization and characterization of the leukotriene C4 synthetase of rat basophil leukemia cells: a novel, particulate glutathione S-­‐transferase. Arch Biochem Biophys 1984, 230:455-­‐65. 197. Jakschik BA, Harper T, Murphy RC. Leukotriene C4 and D4 formation by particulate enzymes. J Biol Chem 1982, 257:5346-­‐9. 84 References 198. Yoshimoto T, Soberman RJ, Lewis RA, Austen KF. Isolation and characterization of leukotriene C4 synthetase of rat basophilic leukemia cells. Proc Natl Acad Sci U S A 1985, 82:8399-­‐403. 199. Abe M, Hugli TE. Characterization of leukotriene C4 synthetase in mouse peritoneal exudate cells. Biochim Biophys Acta 1988, 959:386-­‐98. 200. Izumi T, Honda Z, Ohishi N, Kitamura S, Tsuchida S, Sato K, Shimizu T, Seyama Y. Solubilization and partial purification of leukotriene C4 synthase from guinea-­‐pig lung: a microsomal enzyme with high specificity towards 5,6-­‐epoxide leukotriene A4. Biochim Biophys Acta 1988, 959:305-­‐15. 201. Söderström M, Hammarström S, Mannervik B. Leukotriene C synthase in mouse mastocytoma cells. An enzyme distinct from cytosolic and microsomal glutathione transferases. Biochem J 1988, 250:713-­‐8. 202. Penrose JF, Gagnon L, Goppelt-­‐Struebe M, Myers P, Lam BK, Jack RM, Austen KF, Soberman RJ. Purification of human leukotriene C4 synthase. Proc Natl Acad Sci U S A 1992, 89:11603-­‐6. 203. Nicholson DW, Ali A, Vaillancourt JP, Calaycay JR, Mumford RA, Zamboni RJ, Ford-­‐
Hutchinson AW. Purification to homogeneity and the N-­‐terminal sequence of human leukotriene C4 synthase: a homodimeric glutathione S-­‐transferase composed of 18-­‐kDa subunits. Proc Natl Acad Sci U S A 1993, 90:2015-­‐9. 204. Lam BK, Penrose JF, Freeman GJ, Austen KF. Expression cloning of a cDNA for human leukotriene C4 synthase, an integral membrane protein conjugating reduced glutathione to leukotriene A4. Proc Natl Acad Sci U S A 1994, 91:7663-­‐7. 205. Welsch DJ, Creely DP, Hauser SD, Mathis KJ, Krivi GG, Isakson PC. Molecular cloning and expression of human leukotriene-­‐C4 synthase. Proc Natl Acad Sci U S A 1994, 91:9745-­‐9. 206. Svartz J, Blomgran R, Hammarström S, Söderström M. Leukotriene C4 synthase homo-­‐oligomers detected in living cells by bioluminescence resonance energy transfer. Biochim Biophys Acta 2003, 1633:90-­‐5. 207. Schmidt-­‐Krey I, Kanaoka Y, Mills DJ, Irikura D, Haase W, Lam BK, Austen KF, Kuhlbrandt W. Human leukotriene C4 synthase at 4.5 Å resolution in projection. Structure 2004, 12:2009-­‐14. 208. Martinez Molina D, Wetterholm A, Kohl A, McCarthy AA, Niegowski D, Ohlson E, Hammarberg T, Eshaghi S, Haeggström JZ, Nordlund P. Structural basis for synthesis of inflammatory mediators by human leukotriene C4 synthase. Nature 2007, 448:613-­‐6. 209. Ago H, Kanaoka Y, Irikura D, Lam BK, Shimamura T, Austen KF, Miyano M. Crystal structure of a human membrane protein involved in cysteinyl leukotriene biosynthesis. Nature 2007, 448:609-­‐12. 210. Morgenstern R, Guthenberg C, dePierre JW. Microsomal glutathione S-­‐
transferase. Purification, initial characterization and demonstration that it is not identical to the cytosolic glutathione S-­‐transferases A, B and C. Eur J Biochem 1982, 128:243-­‐8. 211. Lam BK, Penrose JF, Xu K, Baldasaro MH, Austen KF. Site-­‐directed mutagenesis of human leukotriene C4 synthase. J Biol Chem 1997, 272:13923-­‐8. 212. Christmas P, Weber BM, McKee M, Brown D, Soberman RJ. Membrane localization and topology of leukotriene C4 synthase. J Biol Chem 2002, 277:28902-­‐8. 85 References 213. Svartz J, Hallin E, Shi Y, Söderström M, Hammarström S. Identification of regions of leukotriene C4 synthase which direct the enzyme to its nuclear envelope localization. J Cell Biochem 2006, 98:1517-­‐27. 214. Bigby TD, Hodulik CR, Arden KC, Fu L. Molecular cloning of the human leukotriene C4 synthase gene and assignment to chromosome 5q35. Mol Med 1996, 2:637-­‐46. 215. Penrose JF, Spector J, Baldasaro M, Xu K, Boyce J, Arm JP, Austen KF, Lam BK. Molecular cloning of the gene for human leukotriene C4 synthase. Organization, nucleotide sequence, and chromosomal localization to 5q35. J Biol Chem 1996, 271:11356-­‐61. 216. Penrose JF, Baldasaro MH, Webster M, Xu K, Austen KF, Lam BK. Molecular cloning of the gene for mouse leukotriene-­‐C4 synthase. Eur J Biochem 1997, 248:807-­‐13. 217. Bigby TD. The leukotriene C4 synthase gene and asthma. Am J Respir Cell Mol Biol 2000, 23:273-­‐6. 218. Serio KJ, Hodulik CR, Bigby TD. Sp1 and Sp3 function as key regulators of leukotriene C4 synthase gene expression in the monocyte-­‐like cell line, THP-­‐1. Am J Respir Cell Mol Biol 2000, 23:234-­‐40. 219. Zhao JL, Austen KF, Lam BK. Cell-­‐specific transcription of leukotriene C4 synthase involves a Kruppel-­‐like transcription factor and Sp1. J Biol Chem 2000, 275:8903-­‐
10. 220. Lam BK. Leukotriene C4 synthase. Prostaglandins Leukot Essent Fatty Acids 2003, 69:111-­‐6. 221. Tornhamre S, Sjölinder M, Lindberg Å, Ericsson I, Näsman-­‐Glaser B, Griffiths WJ, Lindgren JÅ. Demonstration of leukotriene-­‐C4 synthase in platelets and species distribution of the enzyme activity. Eur J Biochem 1998, 251:227-­‐35. 222. Söderström M, Engblom D, Blomqvist A, Hammarström S. Expression of leukotriene C4 synthase mRNA by the choroid plexus in mouse brain suggests novel neurohormone functions of cysteinyl leukotrienes. Biochem Biophys Res Commun 2003, 307:987-­‐90. 223. Lindgren JÅ, Hökfelt T, Dahlén SE, Patrono C, Samuelsson B. Leukotrienes in the rat central nervous system. Proc Natl Acad Sci U S A 1984, 81:6212-­‐6. 224. Lindgren JÅ, Miyamoto T, Schalling M, Hökfelt T, Samuelsson B. Production and binding of leukotrienes in the rat brain. Adv Prostaglandin Thromboxane Leukot Res 1987, 17B:923-­‐8. 225. Dray F, Wisner A, Bommelaer-­‐Bayet MC, Tiberghien C, Gerozissis K, Saadi M, Junier MP, Rougeot C. Prostaglandin E2, leukotriene C4, and platelet-­‐activating factor receptor sites in the brain. Binding parameters and pharmacological studies. Ann N Y Acad Sci 1989, 559:100-­‐11. 226. Simmet T, Luck W, Winking M, Delank WK, Peskar BA. Identification and characterization of cysteinyl-­‐leukotriene formation in tissue slices from human intracranial tumors: evidence for their biosynthesis under in vivo conditions. J Neurochem 1990, 54:2091-­‐9. 227. Morris AA, Rodger IW. Leukotrienes and the brain. Lancet 1998, 352:1487-­‐8. 228. Winking M, Deinsberger W, Joedicke A, Boeker DK. Cysteinyl-­‐leukotriene levels in intracerebral hemorrhage: an edema-­‐promoting factor? Cerebrovasc Dis 1998, 8:318-­‐26. 86 References 229. Söderström M, Bolling A, Hammarström S. Induction of leukotriene C4 synthase activity in differentiating human erythroleukemia cells. Biochem Biophys Res Commun 1992, 189:1043-­‐9. 230. Ali A, Ford-­‐Hutchinson AW, Nicholson DW. Activation of protein kinase C down-­‐
regulates leukotriene C4 synthase activity and attenuates cysteinyl leukotriene production in an eosinophilic substrain of HL-­‐60 cells. J Immunol 1994, 153:776-­‐
88. 231. Kargman S, Ali A, Vaillancourt JP, Evans JF, Nicholson DW. Protein kinase C-­‐
dependent regulation of sulfidopeptide leukotriene biosynthesis and leukotriene C4 synthase in neutrophilic HL-­‐60 cells. Mol Pharmacol 1994, 45:1043-­‐9. 232. Serio KJ, Johns SC, Luo L, Hodulik CR, Bigby TD. Lipopolysaccharide down-­‐
regulates leukotriene C4 synthase gene expression in a cell-­‐specific manner in the monocyte-­‐like cell line, THP-­‐1. Chest 2003, 123:426S. 233. Serio KJ, Johns SC, Luo L, Hodulik CR, Bigby TD. Lipopolysaccharide down-­‐
regulates the leukotriene C4 synthase gene in the monocyte-­‐like cell line, THP-­‐1. J Immunol 2003, 170:2121-­‐8. 234. Serio KJ, Luo C, Luo L, Mao JT. TNF-­‐α downregulates the leukotriene C4 synthase gene in mononuclear phagocytes. Am J Physiol Lung Cell Mol Physiol 2007, 292:L215-­‐22. 235. Riddick CA, Serio KJ, Hodulik CR, Ring WL, Regan MS, Bigby TD. TGF-­‐β increases leukotriene C4 synthase expression in the monocyte-­‐like cell line, THP-­‐1. J Immunol 1999, 162:1101-­‐7. 236. Abe M, Shibata K, Saruwatar S, Soeda S, Shimeno H, Katsuragi T. cDNA cloning and expression of rat leukotriene C4 synthase: elevated expression in rat basophilic leukemia-­‐1 cells after treatment with retinoic acid. Prostaglandins Leukot Essent Fatty Acids 2002, 67:319-­‐26. 237. Abe M, Shibata K, Urata H, Sakata N, Katsuragi T. Induction of leukotriene C4 synthase after the differentiation of rat basophilic leukemia cells with retinoic acid and a low dose of actinomycin D and its suppression with methylprednisolone. J Cell Physiol 2003, 196:154-­‐64. 238. Sanak M, Simon HU, Szczeklik A. Leukotriene C4 synthase promoter polymorphism and risk of aspirin-­‐induced asthma. Lancet 1997, 350:1599-­‐600. 239. Kedda MA, Shi J, Duffy D, Phelps S, Yang I, O'Hara K, Fong K, Thompson PJ. Characterization of two polymorphisms in the leukotriene C4 synthase gene in an Australian population of subjects with mild, moderate, and severe asthma. J Allergy Clin Immunol 2004, 113:889-­‐95. 240. Kawagishi Y, Mita H, Taniguchi M, Maruyama M, Oosaki R, Higashi N, Kashii T, Kobayashi M, Akiyama K. Leukotriene C4 synthase promoter polymorphism in Japanese patients with aspirin-­‐induced asthma. J Allergy Clin Immunol 2002, 109:936-­‐42. 241. Van Sambeek R, Stevenson DD, Baldasaro M, Lam BK, Zhao J, Yoshida S, Yandora C, Drazen JM, Penrose JF. 5'-­‐Flanking region polymorphism of the gene encoding leukotriene C4 synthase does not correlate with the aspirin-­‐intolerant asthma phenotype in the United States. J Allergy Clin Immunol 2000, 106:72-­‐6. 242. Freiberg JJ, Tybjaerg-­‐Hansen A, Sillesen H, Jensen GB, Nordestgaard BG. Promotor polymorphisms in leukotriene C4 synthase and risk of ischemic cerebrovascular disease. Arterioscler Thromb Vasc Biol 2008, 28:990-­‐6. 87 References 243. Kanaoka Y, Maekawa A, Penrose JF, Austen KF, Lam BK. Attenuated zymosan-­‐
induced peritoneal vascular permeability and IgE-­‐dependent passive cutaneous anaphylaxis in mice lacking leukotriene C4 synthase. J Biol Chem 2001, 276:22608-­‐13. 244. Mayatepek E. Leukotriene C4 synthesis deficiency: a member of a probably underdiagnosed new group of neurometabolic diseases. Eur J Pediatr 2000, 159:811-­‐8. 245. Feltenmark S, Gautam N, Brunnström Å, Griffiths W, Backman L, Edenius C, Lindbom L, Björkholm 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 U S A 2008, 105:680-­‐5. 246. Rådmark O, Shimizu T, Jörnvall H, Samuelsson B. Leukotriene A4 hydrolase in human leukocytes. Purification and properties. J Biol Chem 1984, 259:12339-­‐45. 247. Brock TG, Maydanski E, McNish RW, Peters-­‐Golden M. Co-­‐localization of leukotriene A4 hydrolase with 5-­‐lipoxygenase in nuclei of alveolar macrophages and rat basophilic leukemia cells but not neutrophils. J Biol Chem 2001, 276:35071-­‐7. 248. Funk CD, Rådmark O, Fu JY, Matsumoto T, Jörnvall H, Shimizu T, Samuelsson B. Molecular cloning and amino acid sequence of leukotriene A4 hydrolase. Proc Natl Acad Sci U S A 1987, 84:6677-­‐81. 249. Vallee BL, Auld DS. Zinc coordination, function, and structure of zinc enzymes and other proteins. Biochemistry 1990, 29:5647-­‐59. 250. Haeggström JZ, Wetterholm A, Vallee BL, Samuelsson B. Leukotriene A4 hydrolase: an epoxide hydrolase with peptidase activity. Biochem Biophys Res Commun 1990, 173:431-­‐7. 251. Örning L, Krivi G, Fitzpatrick FA. Leukotriene A4 hydrolase. Inhibition by bestatin and intrinsic aminopeptidase activity establish its functional resemblance to metallohydrolase enzymes. J Biol Chem 1991, 266:1375-­‐8. 252. Byrum RS, Goulet JL, Snouwaert JN, Griffiths RJ, Koller BH. Determination of the contribution of cysteinyl leukotrienes and leukotriene B4 in acute inflammatory responses using 5-­‐lipoxygenase-­‐ and leukotriene A4 hydrolase-­‐deficient mice. J Immunol 1999, 163:6810-­‐9. 253. Minami M, Ohno S, Kawasaki H, Rådmark O, Samuelsson B, Jörnvall H, Shimizu T, Seyama Y, Suzuki K. Molecular cloning of a cDNA coding for human leukotriene A4 hydrolase. Complete primary structure of an enzyme involved in eicosanoid synthesis. J Biol Chem 1987, 262:13873-­‐6. 254. Thunnissen MM, Nordlund P, Haeggström JZ. Crystal structure of human leukotriene A4 hydrolase, a bifunctional enzyme in inflammation. Nat Struct Biol 2001, 8:131-­‐5. 255. Mueller MJ, Wetterholm A, Blomster M, Jörnvall H, Samuelsson B, Haeggström JZ. Leukotriene A4 hydrolase: mapping of a henicosapeptide involved in mechanism-­‐
based inactivation. Proc Natl Acad Sci U S A 1995, 92:8383-­‐7. 256. Haeggström JZ. Leukotriene A4 hydrolase/aminopeptidase, the gatekeeper of chemotactic leukotriene B4 biosynthesis. J Biol Chem 2004, 279:50639-­‐42. 257. Murphy RC, Gijon MA. Biosynthesis and metabolism of leukotrienes. Biochem J 2007, 405:379-­‐95. 258. Mancini JA, Evans JF. Cloning and characterization of the human leukotriene A4 hydrolase gene. Eur J Biochem 1995, 231:65-­‐71. 88 References 259. Arguello M, Paz S, Hernandez E, Corriveau-­‐Bourque C, Fawaz LM, Hiscott J, Lin R. Leukotriene A4 hydrolase expression in PEL cells is regulated at the transcriptional level and leads to increased leukotriene B4 production. J Immunol 2006, 176:7051-­‐61. 260. Izumi T, Shimizu T, Seyama Y, Ohishi N, Takaku F. Tissue distribution of leukotriene A4 hydrolase activity in guinea pig. Biochem Biophys Res Commun 1986, 135:139-­‐45. 261. Nakamura M, Shimizu T. Leukotriene receptors. Chem Rev 2011, 111:6231-­‐98. 262. Bäck M, Dahlén SE, Drazen JM, Evans JF, Serhan CN, Shimizu T, Yokomizo T, Rovati GE. International union of basic and clinical pharmacology. LXXXIV: leukotriene receptor nomenclature, distribution, and pathophysiological functions. Pharmacol Rev 2011, 63:539-­‐84. 263. Simon MI, Strathmann MP, Gautam N. Diversity of G proteins in signal transduction. Science 1991, 252:802-­‐8. 264. Yokomizo T, Izumi T, Chang K, Takuwa Y, Shimizu T. A G-­‐protein-­‐coupled receptor for leukotriene B4 that mediates chemotaxis. Nature 1997, 387:620-­‐4. 265. Yokomizo T, Kato K, Terawaki K, Izumi T, Shimizu T. A second leukotriene B4 receptor, BLT2. A new therapeutic target in inflammation and immunological disorders. J Exp Med 2000, 192:421-­‐32. 266. Ohnishi H, Miyahara N, Gelfand EW. The role of leukotriene B4 in allergic diseases. Allergol Int 2008, 57:291-­‐8. 267. Huang WW, Garcia-­‐Zepeda EA, Sauty A, Oettgen HC, Rothenberg ME, Luster AD. Molecular and biological characterization of the murine leukotriene B4 receptor expressed on eosinophils. J Exp Med 1998, 188:1063-­‐74. 268. Peres CM, Aronoff DM, Serezani CH, Flamand N, Faccioli LH, Peters-­‐Golden M. Specific leukotriene receptors couple to distinct G proteins to effect stimulation of alveolar macrophage host defense functions. J Immunol 2007, 179:5454-­‐61. 269. Pettersson A, Richter J, Owman C. Flow cytometric mapping of the leukotriene B4 receptor, BLT1, in human bone marrow and peripheral blood using specific monoclonal antibodies. Int Immunopharmacol 2003, 3:1467-­‐75. 270. Goodarzi K, Goodarzi M, Tager AM, Luster AD, von Andrian UH. Leukotriene B4 and BLT1 control cytotoxic effector T cell recruitment to inflamed tissues. Nat Immunol 2003, 4:965-­‐73. 271. Ott VL, Cambier JC, Kappler J, Marrack P, Swanson BJ. Mast cell-­‐dependent migration of effector CD8+ T cells through production of leukotriene B4. Nat Immunol 2003, 4:974-­‐81. 272. Tager AM, Bromley SK, Medoff BD, Islam SA, Bercury SD, Friedrich EB, Carafone AD, Gerszten RE, Luster AD. Leukotriene B4 receptor BLT1 mediates early effector T cell recruitment. Nat Immunol 2003, 4:982-­‐90. 273. Owman C, Nilsson C, Lolait SJ. Cloning of cDNA encoding a putative chemoattractant receptor. Genomics 1996, 37:187-­‐94. 274. Kato K, Yokomizo T, Izumi T, Shimizu T. Cell-­‐specific transcriptional regulation of human leukotriene B4 receptor gene. J Exp Med 2000, 192:413-­‐20. 275. Kamohara M, Takasaki J, Matsumoto M, Saito T, Ohishi T, Ishii H, Furuichi K. Molecular cloning and characterization of another leukotriene B4 receptor. J Biol Chem 2000, 275:27000-­‐4. 89 References 276. Tryselius Y, Nilsson NE, Kotarsky K, Olde B, Owman C. Cloning and characterization of cDNA encoding a novel human leukotriene B4 receptor. Biochem Biophys Res Commun 2000, 274:377-­‐82. 277. Brink C, Dahlén SE, Drazen J, Evans JF, Hay DW, Nicosia S, Serhan CN, Shimizu T, Yokomizo T. International Union of Pharmacology XXXVII. Nomenclature for leukotriene and lipoxin receptors. Pharmacol Rev 2003, 55:195-­‐227. 278. Nieves D, Moreno JJ. Role of 5-­‐lipoxygenase pathway in the regulation of RAW 264.7 macrophage proliferation. Biochem Pharmacol 2006, 72:1022-­‐30. 279. Watanabe S, Yamasaki A, Hashimoto K, Shigeoka Y, Chikumi H, Hasegawa Y, Sumikawa T, Takata M, Okazaki R, Watanabe M et al. Expression of functional leukotriene B4 receptors on human airway smooth muscle cells. J Allergy Clin Immunol 2009, 124:59-­‐65 e1-­‐3. 280. Haribabu B, Zhelev DV, Pridgen BC, Richardson RM, Ali H, Snyderman R. Chemoattractant receptors activate distinct pathways for chemotaxis and secretion. Role of G-­‐protein usage. J Biol Chem 1999, 274:37087-­‐92. 281. Sabirsh A, Bristulf J, Owman C. Exploring the pharmacology of the leukotriene B4 receptor BLT1, without the confounding effects of BLT2. Eur J Pharmacol 2004, 499:53-­‐65. 282. Dryden P, Duronio V, Martin L, Hudson AT, Salari H. Inhibition of human neutrophil responses by alpha-­‐cyano-­‐3,4-­‐dihydroxythiocinnamamide; a protein-­‐
tyrosine kinase inhibitor. Br J Pharmacol 1992, 106:656-­‐64. 283. Brach MA, de Vos S, Arnold C, Gruss HJ, Mertelsmann R, Herrmann F. Leukotriene B4 transcriptionally activates interleukin-­‐6 expression involving NK-­‐chi B and NF-­‐IL6. Eur J Immunol 1992, 22:2705-­‐11. 284. Huang L, Zhao A, Wong F, Ayala JM, Struthers M, Ujjainwalla F, Wright SD, Springer MS, Evans J, Cui J. Leukotriene B4 strongly increases monocyte chemoattractant protein-­‐1 in human monocytes. Arterioscler Thromb Vasc Biol 2004, 24:1783-­‐8. 285. Poubelle PE, Stankova J, Grassi J, Rola-­‐Pleszczynski M. Leukotriene B4 up-­‐
regulates IL-­‐6 rather than IL-­‐1 synthesis in human monocytes. Agents Actions 1991, 34:42-­‐5. 286. Flamand N, Mancuso P, Serezani CH, Brock TG. Leukotrienes: mediators that have been typecast as villains. Cell Mol Life Sci 2007, 64:2657-­‐70. 287. Woo CH, You HJ, Cho SH, Eom YW, Chun JS, Yoo YJ, Kim JH. Leukotriene B4 stimulates Rac-­‐ERK cascade to generate reactive oxygen species that mediates chemotaxis. J Biol Chem 2002, 277:8572-­‐8. 288. Yokomizo T, Kato K, Hagiya H, Izumi T, Shimizu T. Hydroxyeicosanoids bind to and activate the low affinity leukotriene B4 receptor, BLT2. J Biol Chem 2001, 276:12454-­‐9. 289. Diczfalusy U, Falardeau P, Hammarström S. Conversion of prostaglandin endoperoxides to C17-­‐hydroxy acids catalyzed by human platelet thromboxane synthase. FEBS Lett 1977, 84:271-­‐4. 290. Okuno T, Iizuka Y, Okazaki H, Yokomizo T, Taguchi R, Shimizu T. 12(S)-­‐
Hydroxyheptadeca-­‐5Z, 8E, 10E-­‐trienoic acid is a natural ligand for leukotriene B4 receptor 2. J Exp Med 2008, 205:759-­‐66. 291. Lin Q, Ruuska SE, Shaw NS, Dong D, Noy N. Ligand selectivity of the peroxisome proliferator-­‐activated receptor α. Biochemistry 1999, 38:185-­‐90. 90 References 292. Devchand PR, Keller H, Peters JM, Vazquez M, Gonzalez FJ, Wahli W. The PPARα-­‐
leukotriene B4 pathway to inflammation control. Nature 1996, 384:39-­‐43. 293. Augstein J, Farmer JB, Lee TB, Sheard P, Tattersall ML. Selective inhibitor of slow reacting substance of anaphylaxis. Nat New Biol 1973, 245:215-­‐7. 294. Labat C, Ortiz JL, Norel X, Gorenne I, Verley J, Abram TS, Cuthbert NJ, Tudhope SR, Norman P, Gardiner P et al. A second cysteinyl leukotriene receptor in human lung. J Pharmacol Exp Ther 1992, 263:800-­‐5. 295. Lynch KR, O'Neill GP, Liu Q, Im DS, Sawyer N, Metters KM, Coulombe N, Abramovitz M, Figueroa DJ, Zeng Z et al. Characterization of the human cysteinyl leukotriene CysLT1 receptor. Nature 1999, 399:789-­‐93. 296. Heise CE, O'Dowd BF, Figueroa DJ, Sawyer N, Nguyen T, Im DS, Stocco R, Bellefeuille JN, Abramovitz M, Cheng R et al. Characterization of the human cysteinyl leukotriene 2 receptor. J Biol Chem 2000, 275:30531-­‐6. 297. Sarau HM, Ames RS, Chambers J, Ellis C, Elshourbagy N, Foley JJ, Schmidt DB, Muccitelli RM, Jenkins O, Murdock PR et al. Identification, molecular cloning, expression, and characterization of a cysteinyl leukotriene receptor. Mol Pharmacol 1999, 56:657-­‐63. 298. Takasaki J, Kamohara M, Matsumoto M, Saito T, Sugimoto T, Ohishi T, Ishii H, Ota T, Nishikawa T, Kawai Y et al. The molecular characterization and tissue distribution of the human cysteinyl leukotriene CysLT2 receptor. Biochem Biophys Res Commun 2000, 274:316-­‐22. 299. Woszczek G, Pawliczak R, Qi HY, Nagineni S, Alsaaty S, Logun C, Shelhamer JH. Functional characterization of human cysteinyl leukotriene 1 receptor gene structure. J Immunol 2005, 175:5152-­‐9. 300. Zhu J, Qiu YS, Figueroa DJ, Bandi V, Galczenski H, Hamada K, Guntupalli KK, Evans JF, Jeffery PK. Localization and upregulation of cysteinyl leukotriene-­‐1 receptor in asthmatic bronchial mucosa. Am J Respir Cell Mol Biol 2005, 33:531-­‐40. 301. Figueroa DJ, Breyer RM, Defoe SK, Kargman S, Daugherty BL, Waldburger K, Liu Q, Clements M, Zeng Z, O'Neill GP et al. Expression of the cysteinyl leukotriene 1 receptor in normal human lung and peripheral blood leukocytes. Am J Respir Crit Care Med 2001, 163:226-­‐33. 302. Holgate ST, Peters-­‐Golden M, Panettieri RA, Henderson WR, Jr. Roles of cysteinyl leukotrienes in airway inflammation, smooth muscle function, and remodeling. J Allergy Clin Immunol 2003, 111:S18-­‐34; discussion S-­‐6. 303. James AJ, Penrose JF, Cazaly AM, Holgate ST, Sampson AP. Human bronchial fibroblasts express the 5-­‐lipoxygenase pathway. Respir Res 2006, 7:102. 304. Vannella KM, McMillan TR, Charbeneau RP, Wilke CA, Thomas PE, Toews GB, Peters-­‐Golden M, Moore BB. Cysteinyl leukotrienes are autocrine and paracrine regulators of fibrocyte function. J Immunol 2007, 179:7883-­‐90. 305. Shirasaki H, Kanaizumi E, Watanabe K, Matsui T, Sato J, Narita S, Rautiainen M, Himi T. Expression and localization of the cysteinyl leukotriene 1 receptor in human nasal mucosa. Clin Exp Allergy 2002, 32:1007-­‐12. 306. Nothacker HP, Wang Z, Zhu Y, Reinscheid RK, Lin SH, Civelli O. Molecular cloning and characterization of a second human cysteinyl leukotriene receptor: discovery of a subtype selective agonist. Mol Pharmacol 2000, 58:1601-­‐8. 91 References 307. Barajas-­‐Espinosa A, Ochoa-­‐Cortes F, Moos MP, Daniel Ramirez F, Vanner SJ, Funk CD. Characterization of the cysteinyl leukotriene 2 receptor in novel expression sites of the gastrointestinal tract. Am J Pathol 2011, 178:2682-­‐9. 308. Mita H, Hasegawa M, Saito H, Akiyama K. Levels of cysteinyl leukotriene receptor mRNA in human peripheral leucocytes: significantly higher expression of cysteinyl leukotriene receptor 2 mRNA in eosinophils. Clin Exp Allergy 2001, 31:1714-­‐23. 309. Ogasawara H, Ishii S, Yokomizo T, Kakinuma T, Komine M, Tamaki K, Shimizu T, Izumi T. Characterization of mouse cysteinyl leukotriene receptors mCysLT1 and mCysLT2: differential pharmacological properties and tissue distribution. J Biol Chem 2002, 277:18763-­‐8. 310. Hui Y, Yang G, Galczenski H, Figueroa DJ, Austin CP, Copeland NG, Gilbert DJ, Jenkins NA, Funk CD. The murine cysteinyl leukotriene 2 (CysLT2) receptor. cDNA and genomic cloning, alternative splicing, and in vitro characterization. J Biol Chem 2001, 276:47489-­‐95. 311. Maekawa A, Austen KF, Kanaoka Y. Targeted gene disruption reveals the role of cysteinyl leukotriene 1 receptor in the enhanced vascular permeability of mice undergoing acute inflammatory responses. J Biol Chem 2002, 277:20820-­‐4. 312. Daniels SE, Bhattacharrya S, James A, Leaves NI, Young A, Hill MR, Faux JA, Ryan GF, le Souef PN, Lathrop GM et al. A genome-­‐wide search for quantitative trait loci underlying asthma. Nature 1996, 383:247-­‐50. 313. Kimura K, Noguchi E, Shibasaki M, Arinami T, Yokouchi Y, Takeda K, Yamakawa-­‐
Kobayashi K, Matsui A, Hamaguchi H. Linkage and association of atopic asthma to markers on chromosome 13 in the Japanese population. Hum Mol Genet 1999, 8:1487-­‐90. 314. Woszczek G, Chen LY, Nagineni S, Alsaaty S, Harry A, Logun C, Pawliczak R, Shelhamer JH. IFN-­‐γ induces cysteinyl leukotriene receptor 2 expression and enhances the responsiveness of human endothelial cells to cysteinyl leukotrienes. J Immunol 2007, 178:5262-­‐70. 315. Capra V, Accomazzo MR, Ravasi S, Parenti M, Macchia M, Nicosia S, Rovati GE. Involvement of prenylated proteins in calcium signaling induced by LTD4 in differentiated U937 cells. Prostaglandins Other Lipid Mediat 2003, 71:235-­‐51. 316. Sjölander A, Grönroos E, Hammarström S, Andersson T. Leukotriene D4 and E4 induce transmembrane signaling in human epithelial cells. Single cell analysis reveals diverse pathways at the G-­‐protein level for the influx and the intracellular mobilization of Ca2+. J Biol Chem 1990, 265:20976-­‐81. 317. Hoshino M, Izumi T, Shimizu T. Leukotriene D4 activates mitogen-­‐activated protein kinase through a protein kinase Cα-­‐Raf-­‐1-­‐dependent pathway in human monocytic leukemia THP-­‐1 cells. J Biol Chem 1998, 273:4878-­‐82. 318. McMahon B, Stenson C, McPhillips F, Fanning A, Brady HR, Godson C. Lipoxin A4 antagonizes the mitogenic effects of leukotriene D4 in human renal mesangial cells. Differential activation of MAP kinases through distinct receptors. J Biol Chem 2000, 275:27566-­‐75. 319. Paruchuri S, Hallberg B, Juhas M, Larsson C, Sjölander A. Leukotriene D4 activates MAPK through a Ras-­‐independent but PKCε-­‐dependent pathway in intestinal epithelial cells. J Cell Sci 2002, 115:1883-­‐93. 92 References 320. Capra V, Ravasi S, Accomazzo MR, Parenti M, Rovati GE. CysLT1 signal transduction in differentiated U937 cells involves the activation of the small GTP-­‐
binding protein Ras. Biochem Pharmacol 2004, 67:1569-­‐77. 321. Ciccarelli R, D'Alimonte I, Santavenere C, D'Auro M, Ballerini P, Nargi E, Buccella S, Nicosia S, Folco G, Caciagli F et al. Cysteinyl-­‐leukotrienes are released from astrocytes and increase astrocyte proliferation and glial fibrillary acidic protein via cys-­‐LT1 receptors and mitogen-­‐activated protein kinase pathway. Eur J Neurosci 2004, 20:1514-­‐24. 322. Parhamifar L, Jeppsson B, Sjölander A. Activation of cPLA2 is required for leukotriene D4-­‐induced proliferation in colon cancer cells. Carcinogenesis 2005, 26:1988-­‐98. 323. Jiang Y, Kanaoka Y, Feng C, Nocka K, Rao S, Boyce JA. Cutting edge: Interleukin 4-­‐
dependent mast cell proliferation requires autocrine/intracrine cysteinyl leukotriene-­‐induced signaling. J Immunol 2006, 177:2755-­‐9. 324. Ravasi S, Citro S, Viviani B, Capra V, Rovati GE. CysLT1 receptor-­‐induced human airway smooth muscle cells proliferation requires ROS generation, EGF receptor transactivation and ERK1/2 phosphorylation. Respir Res 2006, 7:42. 325. Kim MH, Lee YJ, Kim MO, Kim JS, Han HJ. Effect of leukotriene D4 on mouse embryonic stem cell migration and proliferation: involvement of PI3K/Akt as well as GSK-­‐3β/ β -­‐catenin signaling pathways. J Cell Biochem 2010, 111:686-­‐98. 326. Thompson C, Cloutier A, Bosse Y, Thivierge M, Gouill CL, Larivee P, McDonald PP, Stankova J, Rola-­‐Pleszczynski M. CysLT1 receptor engagement induces activator protein-­‐1-­‐ and NF-­‐κB-­‐dependent IL-­‐8 expression. Am J Respir Cell Mol Biol 2006, 35:697-­‐704. 327. Lötzer K, Jahn S, Kramer C, Hildner M, Nusing R, Funk CD, Habenicht AJ. 5-­‐
Lipoxygenase/cyclooxygenase-­‐2 cross-­‐talk through cysteinyl leukotriene receptor 2 in endothelial cells. Prostaglandins Other Lipid Mediat 2007, 84:108-­‐
15. 328. Nielsen CK, Campbell JI, Öhd JF, Morgelin M, Riesbeck K, Landberg G, Sjölander A. A novel localization of the G-­‐protein-­‐coupled CysLT1 receptor in the nucleus of colorectal adenocarcinoma cells. Cancer Res 2005, 65:732-­‐42. 329. Mellor EA, Frank N, Soler D, Hodge MR, Lora JM, Austen KF, Boyce JA. Expression of the type 2 receptor for cysteinyl leukotrienes (CysLT2R) by human mast cells: Functional distinction from CysLT1R. Proc Natl Acad Sci U S A 2003, 100:11589-­‐
93. 330. Jiang Y, Borrelli LA, Kanaoka Y, Bacskai BJ, Boyce JA. CysLT2 receptors interact with CysLT1 receptors and down-­‐modulate cysteinyl leukotriene dependent mitogenic responses of mast cells. Blood 2007, 110:3263-­‐70. 331. Ravasi S, Capra V, Mezzetti M, Nicosia S, Rovati GE. A kinetic binding study to evaluate the pharmacological profile of a specific leukotriene C4 binding site not coupled to contraction in human lung parenchyma. Mol Pharmacol 2000, 57:1182-­‐9. 332. Capra V, Bolla M, Belloni PA, Mezzetti M, Folco GC, Nicosia S, Rovati GE. Pharmacological characterization of the cysteinyl-­‐leukotriene antagonists CGP 45715A (iralukast) and CGP 57698 in human airways in vitro. Br J Pharmacol 1998, 123:590-­‐8. 93 References 333. Ravasi S, Capra V, Panigalli T, Rovati GE, Nicosia S. Pharmacological differences among CysLT1 receptor antagonists with respect to LTC4 and LTD4 in human lung parenchyma. Biochem Pharmacol 2002, 63:1537-­‐46. 334. Maekawa A, Kanaoka Y, Xing W, Austen KF. Functional recognition of a distinct receptor preferential for leukotriene E4 in mice lacking the cysteinyl leukotriene 1 and 2 receptors. Proc Natl Acad Sci U S A 2008, 105:16695-­‐700. 335. Raport CJ, Schweickart VL, Chantry D, Eddy RL, Jr., Shows TB, Godiska R, Gray PW. New members of the chemokine receptor gene family. J Leukoc Biol 1996, 59:18-­‐23. 336. Ciana P, Fumagalli M, Trincavelli ML, Verderio C, Rosa P, Lecca D, Ferrario S, Parravicini C, Capra V, Gelosa P et al. The orphan receptor GPR17 identified as a new dual uracil nucleotides/cysteinyl-­‐leukotrienes receptor. Embo J 2006, 25:4615-­‐27. 337. Lecca D, Trincavelli ML, Gelosa P, Sironi L, Ciana P, Fumagalli M, Villa G, Verderio C, Grumelli C, Guerrini U et al. The recently identified P2Y-­‐like receptor GPR17 is a sensor of brain damage and a new target for brain repair. PLoS One 2008, 3:e3579. 338. Maekawa A, Balestrieri B, Austen KF, Kanaoka Y. GPR17 is a negative regulator of the cysteinyl leukotriene 1 receptor response to leukotriene D4. Proc Natl Acad Sci U S A 2009, 106:11685-­‐90. 339. Maekawa A, Xing W, Austen KF, Kanaoka Y. GPR17 regulates immune pulmonary inflammation induced by house dust mites. J Immunol 2010, 185:1846-­‐54. 340. Claesson HE, Lindgren JÅ, Gustafsson B. Opsonized bacteria stimulate leukotriene synthesis in human leukocytes. Biochim Biophys Acta 1985, 836:361-­‐7. 341. Mancuso P, Standiford TJ, Marshall T, Peters-­‐Golden M. 5-­‐Lipoxygenase reaction products modulate alveolar macrophage phagocytosis of Klebsiella pneumoniae. Infect Immun 1998, 66:5140-­‐6. 342. McCurdy JD, Olynych TJ, Maher LH, Marshall JS. Cutting edge: distinct Toll-­‐like receptor 2 activators selectively induce different classes of mediator production from human mast cells. J Immunol 2003, 170:1625-­‐9. 343. Surette ME, Palmantier R, Gosselin J, Borgeat P. Lipopolysaccharides prime whole human blood and isolated neutrophils for the increased synthesis of 5-­‐
lipoxygenase products by enhancing arachidonic acid availability: involvement of the CD14 antigen. J Exp Med 1993, 178:1347-­‐55. 344. Hattermann K, Picard S, Borgeat M, Leclerc P, Pouliot M, Borgeat P. The Toll-­‐like receptor 7/8-­‐ligand resiquimod (R-­‐848) primes human neutrophils for leukotriene B4, prostaglandin E2 and platelet-­‐activating factor biosynthesis. Faseb J 2007, 21:1575-­‐85. 345. Olsson S, Sundler R. The macrophage beta-­‐glucan receptor mediates arachidonate release induced by zymosan: essential role for Src family kinases. Mol Immunol 2007, 44:1509-­‐15. 346. Peters-­‐Golden M, Canetti C, Mancuso P, Coffey MJ. Leukotrienes: underappreciated mediators of innate immune responses. J Immunol 2005, 174:589-­‐94. 347. Ford-­‐Hutchinson AW, Bray MA, Doig MV, Shipley ME, Smith MJ. Leukotriene B, a potent chemokinetic and aggregating substance released from polymorphonuclear leukocytes. Nature 1980, 286:264-­‐5. 94 References 348. Kim ND, Chou RC, Seung E, Tager AM, Luster AD. A unique requirement for the leukotriene B4 receptor BLT1 for neutrophil recruitment in inflammatory arthritis. J Exp Med 2006, 203:829-­‐35. 349. Matsukawa A, Hogaboam CM, Lukacs NW, Lincoln PM, Strieter RM, Kunkel SL. Endogenous monocyte chemoattractant protein-­‐1 (MCP-­‐1) protects mice in a model of acute septic peritonitis: cross-­‐talk between MCP-­‐1 and leukotriene B4. J Immunol 1999, 163:6148-­‐54. 350. Zimmerman BJ, Holt JW, Paulson JC, Anderson DC, Miyasaka M, Tamatani T, Todd RF 3rd, Rusche JR, Granger DN. Molecular determinants of lipid mediator-­‐
induced leukocyte adherence and emigration in rat mesenteric venules. Am J Physiol 1994, 266:H847-­‐53. 351. Hébert MJ, Takano T, Holthöfer H, Brady HR. Sequential morphologic events during apoptosis of human neutrophils. Modulation by lipoxygenase-­‐derived eicosanoids. J Immunol 1996, 157:3105-­‐15. 352. Lee E, Robertson T, Smith J, Kilfeather S. Leukotriene receptor antagonists and synthesis inhibitors reverse survival in eosinophils of asthmatic individuals. Am J Respir Crit Care Med 2000, 161:1881-­‐6. 353. Drazen JM, Austen KF, Lewis RA, Clark DA, Goto G, Marfat A, Corey EJ. Comparative airway and vascular activities of leukotrienes C-­‐1 and D in vivo and in vitro. Proc Natl Acad Sci U S A 1980, 77:4354-­‐8. 354. Dahlén SE, Björk J, Hedqvist P, Arfors KE, Hammarström S, Lindgren JÅ, Samuelsson B. Leukotrienes promote plasma leakage and leukocyte adhesion in postcapillary venules: in vivo effects with relevance to the acute inflammatory response. Proc Natl Acad Sci U S A 1981, 78:3887-­‐91. 355. Prinz I, Gregoire C, Mollenkopf H, Aguado E, Wang Y, Malissen M, Kaufmann SH, Malissen B. The type 1 cysteinyl leukotriene receptor triggers calcium influx and chemotaxis in mouse αβ-­‐ and γδ effector T cells. J Immunol 2005, 175:713-­‐9. 356. Spinozzi F, Russano AM, Piattoni S, Agea E, Bistoni O, de Benedictis D, de Benedictis FM. Biological effects of montelukast, a cysteinyl-­‐leukotriene receptor-­‐
antagonist, on T lymphocytes. Clin Exp Allergy 2004, 34:1876-­‐82. 357. Busse W, Kraft M. Cysteinyl leukotrienes in allergic inflammation: strategic target for therapy. Chest 2005, 127:1312-­‐26. 358. Bandeira-­‐Melo C, Hall JC, Penrose JF, Weller PF. Cysteinyl leukotrienes induce IL-­‐
4 release from cord blood-­‐derived human eosinophils. J Allergy Clin Immunol 2002, 109:975-­‐9. 359. Bandeira-­‐Melo C, Woods LJ, Phoofolo M, Weller PF. Intracrine cysteinyl leukotriene receptor-­‐mediated signaling of eosinophil vesicular transport-­‐
mediated interleukin-­‐4 secretion. J Exp Med 2002, 196:841-­‐50. 360. Okunishi K, Dohi M, Nakagome K, Tanaka R, Yamamoto K. A novel role of cysteinyl leukotrienes to promote dendritic cell activation in the antigen-­‐induced immune responses in the lung. J Immunol 2004, 173:6393-­‐402. 361. Alvarez C, Amaral MM, Langellotti C, Vermeulen M. Leukotriene C4 prevents the complete maturation of murine dendritic cells and modifies interleukin-­‐
12/interleukin-­‐23 balance. Immunology 2011, 134:185-­‐97. 95 References 362. Mellor EA, Austen KF, Boyce JA. Cysteinyl leukotrienes and uridine diphosphate induce cytokine generation by human mast cells through an interleukin 4-­‐
regulated pathway that is inhibited by leukotriene receptor antagonists. J Exp Med 2002, 195:583-­‐92. 363. Gauvreau GM, Plitt JR, Baatjes A, MacGlashan DW. Expression of functional cysteinyl leukotriene receptors by human basophils. J Allergy Clin Immunol 2005, 116:80-­‐7. 364. Lamoureux J, Stankova J, Rola-­‐Pleszczynski M. Leukotriene D4 enhances immunoglobulin production in CD40-­‐activated human B lymphocytes. J Allergy Clin Immunol 2006, 117:924-­‐30. 365. Lauritsen K, Laursen LS, Bukhave K, Rask-­‐Madsen J. Effects of topical 5-­‐
aminosalicylic acid and prednisolone on prostaglandin E2 and leukotriene B4 levels determined by equilibrium in vivo dialysis of rectum in relapsing ulcerative colitis. Gastroenterology 1986, 91:837-­‐44. 366. Cole AT, Pilkington BJ, McLaughlan J, Smith C, Balsitis M, Hawkey CJ. Mucosal factors inducing neutrophil movement in ulcerative colitis: the role of interleukin 8 and leukotriene B4. Gut 1996, 39:248-­‐54. 367. Sharon P, Stenson WF. Enhanced synthesis of leukotriene B4 by colonic mucosa in inflammatory bowel disease. Gastroenterology 1984, 86:453-­‐60. 368. Ruzicka T, Simmet T, Peskar BA, Ring J. Skin levels of arachidonic acid-­‐derived inflammatory mediators and histamine in atopic dermatitis and psoriasis. J Invest Dermatol 1986, 86:105-­‐8. 369. Cromwell O, Walport MJ, Morris HR, Taylor GW, Hodson ME, Batten J, Kay AB. Identification of leukotrienes D and B in sputum from cystic fibrosis patients. Lancet 1981, 2:164-­‐5. 370. Klickstein LB, Shapleigh C, Goetzl EJ. Lipoxygenation of arachidonic acid as a source of polymorphonuclear leukocyte chemotactic factors in synovial fluid and tissue in rheumatoid arthritis and spondyloarthritis. J Clin Invest 1980, 66:1166-­‐
70. 371. Elmgreen J, Nielsen OH, Ahnfelt-­‐Rønne I. Enhanced capacity for release of leucotriene B4 by neutrophils in rheumatoid arthritis. Ann Rheum Dis 1987, 46:501-­‐5. 372. Gadaleta D, Fantini GA, Silane MF, Davis JM. Neutrophil leukotriene generation and pulmonary dysfunction after abdominal aortic aneurysm repair. Surgery 1994, 116:847-­‐52. 373. Houard X, Ollivier V, Louedec L, Michel JB, Bäck M. Differential inflammatory activity across human abdominal aortic aneurysms reveals neutrophil-­‐derived leukotriene B4 as a major chemotactic factor released from the intraluminal thrombus. Faseb J 2009, 23:1376-­‐83. 374. Dahlén SE, Hedqvist P, Hammarström S, Samuelsson B. Leukotrienes are potent constrictors of human bronchi. Nature 1980, 288:484-­‐6. 375. Marom Z, Shelhamer JH, Bach MK, Morton DR, Kaliner M. Slow-­‐reacting substances, leukotrienes C4 and D4, increase the release of mucus from human airways in vitro. Am Rev Respir Dis 1982, 126:449-­‐51. 376. Michelassi F, Landa L, Hill RD, Lowenstein E, Watkins WD, Petkau AJ, Zapol WM. Leukotriene D4: a potent coronary artery vasoconstrictor associated with impaired ventricular contraction. Science 1982, 217:841-­‐3. 96 References 377. Schellenberg RR, Foster A. Differential activity of leukotrienes upon human pulmonary vein and artery. Prostaglandins 1984, 27:475-­‐82. 378. Hua XY, Dahlén SE, Lundberg JM, Hammarström S, Hedqvist P. Leukotrienes C4, D4 and E4 cause widespread and extensive plasma extravasation in the guinea pig. Naunyn Schmiedebergs Arch Pharmacol 1985, 330:136-­‐41. 379. Panettieri RA, Tan EM, Ciocca V, Luttmann MA, Leonard TB, Hay DW. Effects of LTD4 on human airway smooth muscle cell proliferation, matrix expression, and contraction In vitro: differential sensitivity to cysteinyl leukotriene receptor antagonists. Am J Respir Cell Mol Biol 1998, 19:453-­‐61. 380. Espinosa K, Bosse Y, Stankova J, Rola-­‐Pleszczynski M. CysLT1 receptor upregulation by TGF-­‐β and IL-­‐13 is associated with bronchial smooth muscle cell proliferation in response to LTD4. J Allergy Clin Immunol 2003, 111:1032-­‐40. 381. Peters-­‐Golden M, Henderson WR, Jr. Leukotrienes. N Engl J Med 2007, 357:1841-­‐
54. 382. Peters-­‐Golden M, Gleason MM, Togias A. Cysteinyl leukotrienes: multi-­‐functional mediators in allergic rhinitis. Clin Exp Allergy 2006, 36:689-­‐703. 383. Carucci JA, Washenik K, Weinstein A, Shupack J, Cohen DE. The leukotriene antagonist zafirlukast as a therapeutic agent for atopic dermatitis. Arch Dermatol 1998, 134:785-­‐6. 384. Yanase DJ, David-­‐Bajar K. The leukotriene antagonist montelukast as a therapeutic agent for atopic dermatitis. J Am Acad Dermatol 2001, 44:89-­‐93. 385. Lipworth BJ. Leukotriene-­‐receptor antagonists. Lancet 1999, 353:57-­‐62. 386. Drazen JM, Israel E, O'Byrne PM. Treatment of asthma with drugs modifying the leukotriene pathway. N Engl J Med 1999, 340:197-­‐206. 387. Montuschi P, Peters-­‐Golden ML. Leukotriene modifiers for asthma treatment. Clin Exp Allergy 2010, 40:1732-­‐41. 388. Hallstrand TS, Henderson WR, Jr. An update on the role of leukotrienes in asthma. Curr Opin Allergy Clin Immunol 2010, 10:60-­‐6. 389. Funk CD. Leukotriene modifiers as potential therapeutics for cardiovascular disease. Nat Rev Drug Discov 2005, 4:664-­‐72. 390. Lötzer K, Funk CD, Habenicht AJ. The 5-­‐lipoxygenase pathway in arterial wall biology and atherosclerosis. Biochim Biophys Acta 2005, 1736:30-­‐7. 391. Rådmark O, Samuelsson B. 5-­‐lipoxygenase: regulation and possible involvement in atherosclerosis. Prostaglandins Other Lipid Mediat 2007, 83:162-­‐74. 392. Fang SH, Wei EQ, Zhou Y, Wang ML, Zhang WP, Yu GL, Chu LS, Chen Z. Increased expression of cysteinyl leukotriene receptor-­‐1 in the brain mediates neuronal damage and astrogliosis after focal cerebral ischemia in rats. Neuroscience 2006, 140:969-­‐79. 393. Chu LS, Wei EQ, Yu GL, Fang SH, Zhou Y, Wang ML, Zhang WP. Pranlukast reduces neutrophil but not macrophage/microglial accumulation in brain after focal cerebral ischemia in mice. Acta Pharmacol Sin 2006, 27:282-­‐8. 394. Yu GL, Wei EQ, Wang ML, Zhang WP, Zhang SH, Weng JQ, Chu LS, Fang SH, Zhou Y, Chen Z et al. Pranlukast, a cysteinyl leukotriene receptor-­‐1 antagonist, protects against chronic ischemic brain injury and inhibits the glial scar formation in mice. Brain Res 2005, 1053:116-­‐25. 395. Zhang WP, Wei EQ, Mei RH, Zhu CY, Zhao MH. Neuroprotective effect of ONO-­‐
1078, a leukotriene receptor antagonist, on focal cerebral ischemia in rats. Acta Pharmacol Sin 2002, 23:871-­‐7. 97 References 396. Zhao R, Shi WZ, Zhang YM, Fang SH, Wei EQ. Montelukast, a cysteinyl leukotriene receptor-­‐1 antagonist, attenuates chronic brain injury after focal cerebral ischaemia in mice and rats. J Pharm Pharmacol 2011, 63:550-­‐7. 397. Zhao CZ, Zhao B, Zhang XY, Huang XQ, Shi WZ, Liu HL, Fang SH, Lu YB, Zhang WP, Tang FD et al. Cysteinyl leukotriene receptor 2 is spatiotemporally involved in neuron injury, astrocytosis and microgliosis after focal cerebral ischemia in rats. Neuroscience 2011, 189:1-­‐11. 398. Zhao B, Zhao CZ, Zhang XY, Huang XQ, Shi WZ, Fang SH, Lu YB, Zhang WP, Xia Q, Wei EQ. The new P2Y-­‐like receptor GPR17 mediates acute neuronal injury and late microgliosis after focal cerebral ischemia in rats. Neuroscience 2012, 202:42-­‐
57. 399. Neu I, Mallinger J, Wildfeuer A, Mehlber L. Leukotrienes in the cerebrospinal fluid of multiple sclerosis patients. Acta Neurol Scand 1992, 86:586-­‐7. 400. Wirth JJ, Kierszenbaum F. Stimulatory effects of leukotriene B4 on macrophage association with and intracellular destruction of Trypanosoma cruzi. J Immunol 1985, 134:1989-­‐93. 401. Wirth JJ, Kierszenbaum F. Effects of leukotriene C4 on macrophage association with and intracellular fate of Trypanosoma cruzi. Mol Biochem Parasitol 1985, 15:1-­‐10. 402. Demitsu T, Katayama H, Saito-­‐Taki T, Yaoita H, Nakano M. Phagocytosis and bactericidal action of mouse peritoneal macrophages treated with leukotriene B4. Int J Immunopharmacol 1989, 11:801-­‐8. 403. Bailie MB, Standiford TJ, Laichalk LL, Coffey MJ, Strieter R, Peters-­‐Golden M. Leukotriene-­‐deficient mice manifest enhanced lethality from Klebsiella pneumonia in association with decreased alveolar macrophage phagocytic and bactericidal activities. J Immunol 1996, 157:5221-­‐4. 404. Serezani CH, Aronoff DM, Jancar S, Mancuso P, Peters-­‐Golden M. Leukotrienes enhance the bactericidal activity of alveolar macrophages against Klebsiella pneumoniae through the activation of NADPH oxidase. Blood 2005, 106:1067-­‐75. 405. Serezani CH, Perrela JH, Russo M, Peters-­‐Golden M, Jancar S. Leukotrienes are essential for the control of Leishmania amazonensis infection and contribute to strain variation in susceptibility. J Immunol 2006, 177:3201-­‐8. 406. Talvani A, Machado FS, Santana GC, Klein A, Barcelos L, Silva JS, Teixeira MM. Leukotriene B4 induces nitric oxide synthesis in Trypanosoma cruzi-­‐infected murine macrophages and mediates resistance to infection. Infect Immun 2002, 70:4247-­‐53. 407. Dewald B, Baggiolini M. Activation of NADPH oxidase in human neutrophils. Synergism between fMLP and the neutrophil products PAF and LTB4. Biochem Biophys Res Commun 1985, 128:297-­‐304. 408. Larfars G, Lantoine F, Devynck MA, Palmblad J, Gyllenhammar H. Activation of nitric oxide release and oxidative metabolism by leukotrienes B4, C4, and D4 in human polymorphonuclear leukocytes. Blood 1999, 93:1399-­‐405. 409. Flamand L, Borgeat P, Lalonde R, Gosselin J. Release of anti-­‐HIV mediators after administration of leukotriene B4 to humans. J Infect Dis 2004, 189:2001-­‐9. 410. Wan M, Sabirsh A, Wetterholm A, Agerberth B, Haeggström JZ. Leukotriene B4 triggers release of the cathelicidin LL-­‐37 from human neutrophils: novel lipid-­‐
peptide interactions in innate immune responses. Faseb J 2007, 21:2897-­‐905. 98 References 411. Wan M, Godson C, Guiry PJ, Agerberth B, Haeggström JZ. Leukotriene B4/antimicrobial peptide LL-­‐37 proinflammatory circuits are mediated by BLT1 and FPR2/ALX and are counterregulated by lipoxin A4 and resolvin E1. Faseb J 2011, 25:1697-­‐705. 412. Skerrett SJ, Henderson WR, Martin TR. Alveolar macrophage function in rats with severe protein calorie malnutrition. Arachidonic acid metabolism, cytokine release, and antimicrobial activity. J Immunol 1990, 144:1052-­‐61. 413. Cederholm T, Lindgren JÅ, Palmblad J. Impaired leukotriene C4 generation in granulocytes from protein-­‐energy malnourished chronically ill elderly. J Intern Med 2000, 247:715-­‐22. 414. Mancuso P, Huffnagle GB, Olszewski MA, Phipps J, Peters-­‐Golden M. Leptin corrects host defense defects after acute starvation in murine pneumococcal pneumonia. Am J Respir Crit Care Med 2006, 173:212-­‐8. 415. Coffey MJ, Wilcoxen SE, Phare SM, Simpson RU, Gyetko MR, Peters-­‐Golden M. Reduced 5-­‐lipoxygenase metabolism of arachidonic acid in macrophages from 1,25-­‐dihydroxyvitamin D3-­‐deficient rats. Prostaglandins 1994, 48:313-­‐29. 416. Coffey MJ, Phare SM, Kazanjian PH, Peters-­‐Golden M. 5-­‐Lipoxygenase metabolism in alveolar macrophages from subjects infected with the human immunodeficiency virus. J Immunol 1996, 157:393-­‐9. 417. Mayatepek E, Flock B, Zelezny R, Kreutzer K, von Giesen HJ. LTB4 and LTC4 are absent in the cerebrospinal fluid of human immunodeficiency virus type 1-­‐
seropositive persons with toxoplasmic encephalitis: evidence for inhibition of 5-­‐
lipoxygenase by Toxoplasma gondii. J Infect Dis 1999, 179:714-­‐6. 418. Claria J, Titos E, Jimenez W, Ros J, Gines P, Arroyo V, Rivera F, Rodes J. Altered biosynthesis of leukotrienes and lipoxins and host defense disorders in patients with cirrhosis and ascites. Gastroenterology 1998, 115:147-­‐56. 419. Jubiz W, Draper RE, Gale J, Nolan G. Decreased leukotriene B4 synthesis by polymorphonuclear leukocytes from male patients with diabetes mellitus. Prostaglandins Leukot Med 1984, 14:305-­‐11. 420. Laviolette M, Coulombe R, Picard S, Braquet P, Borgeat P. Decreased leukotriene B4 synthesis in smokers' alveolar macrophages in vitro. J Clin Invest 1986, 77:54-­‐
60. 421. Balter MS, Toews GB, Peters-­‐Golden M. Multiple defects in arachidonate metabolism in alveolar macrophages from young asymptomatic smokers. J Lab Clin Med 1989, 114:662-­‐73. 422. Wang D, Dubois RN. Eicosanoids and cancer. Nat Rev Cancer 2010, 10:181-­‐93. 423. Medvinsky A, Dzierzak E. Definitive hematopoiesis is autonomously initiated by the AGM region. Cell 1996, 86:897-­‐906. 424. Gekas C, Dieterlen-­‐Lievre F, Orkin SH, Mikkola HK. The placenta is a niche for hematopoietic stem cells. Dev Cell 2005, 8:365-­‐75. 425. Müller AM, Medvinsky A, Strouboulis J, Grosveld F, Dzierzak E. Development of hematopoietic stem cell activity in the mouse embryo. Immunity 1994, 1:291-­‐
301. 426. Dzierzak E, Speck NA. Of lineage and legacy: the development of mammalian hematopoietic stem cells. Nat Immunol 2008, 9:129-­‐36. 427. Mikkola HK, Orkin SH. The journey of developing hematopoietic stem cells. Development 2006, 133:3733-­‐44. 99 References 428. Zanjani ED, Ascensao JL, Flake AW, Harrison MR, Tavassoli M. The fetus as an optimal donor and recipient of hemopoietic stem cells. Bone Marrow Transplant 1992, 10 Suppl 1:107-­‐14. 429. Reya T, Morrison SJ, Clarke MF, Weissman IL. Stem cells, cancer, and cancer stem cells. Nature 2001, 414:105-­‐11. 430. Ikuta K, Weissman IL. Evidence that hematopoietic stem cells express mouse c-­‐Kit but do not depend on steel factor for their generation. Proc Natl Acad Sci U S A 1992, 89:1502-­‐6. 431. Li CL, Johnson GR. Murine hematopoietic stem and progenitor cells: I. Enrichment and biologic characterization. Blood 1995, 85:1472-­‐9. 432. Bryder D, Rossi DJ, Weissman IL. Hematopoietic stem cells: the paradigmatic tissue-­‐specific stem cell. Am J Pathol 2006, 169:338-­‐46. 433. Osawa M, Hanada K, Hamada H, Nakauchi H. Long-­‐term lymphohematopoietic reconstitution by a single CD34-­‐low/negative hematopoietic stem cell. Science 1996, 273:242-­‐5. 434. Christensen JL, Weissman IL. Flk-­‐2 is a marker in hematopoietic stem cell differentiation: a simple method to isolate long-­‐term stem cells. Proc Natl Acad Sci U S A 2001, 98:14541-­‐6. 435. Adolfsson J, Borge OJ, Bryder D, Theilgaard-­‐Mönch K, Åstrand-­‐Grundström I, Sitnicka E, Sasaki Y, Jacobsen SE. Upregulation of Flt3 expression within the bone marrow Lin-­‐ Sca1+ c-­‐Kit+ stem cell compartment is accompanied by loss of self-­‐
renewal capacity. Immunity 2001, 15:659-­‐69. 436. Yang L, Bryder D, Adolfsson J, Nygren J, Månsson R, Sigvardsson M, Jacobsen SE. Identification of Lin-­‐ Sca1+ Kit+ CD34+ Flt3-­‐ short-­‐term hematopoietic stem cells capable of rapidly reconstituting and rescuing myeloablated transplant recipients. Blood 2005, 105:2717-­‐23. 437. Adolfsson J, Månsson R, Buza-­‐Vidas N, Hultquist A, Liuba K, Jensen CT, Bryder D, Yang L, Borge OJ, Thorén LA, Anderson K, Sitnicka E, Sasaki Y, Sigvardsson M, Jacobsen SE. Identification of Flt3+ lympho-­‐myeloid stem cells lacking erythro-­‐
megakaryocytic potential a revised road map for adult blood lineage commitment. Cell 2005, 121:295-­‐306. 438. Zandi S, Bryder D, Sigvardsson M. Load and lock: the molecular mechanisms of B-­‐
lymphocyte commitment. Immunol Rev 2010, 238:47-­‐62. 439. Kondo M, Weissman IL, Akashi K. Identification of clonogenic common lymphoid progenitors in mouse bone marrow. Cell 1997, 91:661-­‐72. 440. Månsson R, Zandi S, Welinder E, Tsapogas P, Sakaguchi N, Bryder D, Sigvardsson M. Single-­‐cell analysis of the common lymphoid progenitor compartment reveals functional and molecular heterogeneity. Blood 2010, 115:2601-­‐9. 441. Pronk CJ, Rossi DJ, Månsson R, Attema JL, Norddahl GL, Chan CK, Sigvardsson M, Weissman IL, Bryder D. Elucidation of the phenotypic, functional, and molecular topography of a myeloerythroid progenitor cell hierarchy. Cell Stem Cell 2007, 1:428-­‐42. 442. Allman D, Sambandam A, Kim S, Miller JP, Pagan A, Well D, Meraz A, Bhandoola A. Thymopoiesis independent of common lymphoid progenitors. Nat Immunol 2003, 4:168-­‐74. 443. Benz C, Martins VC, Radtke F, Bleul CC. The stream of precursors that colonizes the thymus proceeds selectively through the early T lineage precursor stage of T cell development. J Exp Med 2008, 205:1187-­‐99. 100 References 444. Benz C, Bleul CC. A multipotent precursor in the thymus maps to the branching point of the T versus B lineage decision. J Exp Med 2005, 202:21-­‐31. 445. Bryder D, Sigvardsson M. Shaping up a lineage-­‐-­‐lessons from B lymphopoesis. Curr Opin Immunol 2010, 22:148-­‐53. 446. Laiosa CV, Stadtfeld M, Graf T. Determinants of lymphoid-­‐myeloid lineage diversification. Annu Rev Immunol 2006, 24:705-­‐38. 447. Hoggatt J, Pelus LM. Eicosanoid regulation of hematopoiesis and hematopoietic stem and progenitor trafficking. Leukemia 2010, 24:1993-­‐2002. 448. Pelus LM, Broxmeyer HE, Kurland JI, Moore MA. Regulation of macrophage and granulocyte proliferation. Specificities of prostaglandin E and lactoferrin. J Exp Med 1979, 150:277-­‐92. 449. Pelus LM, Broxmeyer HE, Moore MA. Regulation of human myelopoiesis by prostaglandin E and lactoferrin. Cell Tissue Kinet 1981, 14:515-­‐26. 450. Aglietta M, Piacibello W, Gavosto F. Insensitivity of chronic myeloid leukemia cells to inhibition of growth by prostaglandin E1. Cancer Res 1980, 40:2507-­‐11. 451. Pelus LM, Broxmeyer HE, Clarkson BD, Moore MA. Abnormal responsiveness of granulocyte-­‐macrophage committed colony-­‐forming cells from patients with chronic myeloid leukemia to inhibition by prostaglandin E1. Cancer Res 1980, 40:2512-­‐5. 452. Taetle R, Guittard JP, Mendelsohn JM. Abnormal modulation of granulocyte/macrophage progenitor proliferation by prostaglandin E in chronic myeloproliferative disorders. Exp Hematol 1980, 8:1190-­‐201. 453. Taetle R, Koessler A. Effects of cyclic nucleotides and prostaglandins on normal and abnormal human myeloid progenitor proliferation. Cancer Res 1980, 40:1223-­‐9. 454. Pelus LM, Gold E, Saletan S, Coleman M. Restoration of responsiveness of chronic myeloid leukemia granulocyte-­‐macrophage colony-­‐forming cells to growth regulation in vitro following preincubation with prostaglandin E. Blood 1983, 62:158-­‐65. 455. Moore MA, Mertelsmann R, Pelus LM. Phenotypic evaluation of chronic myeloid leukemia. Blood Cells 1981, 7:217-­‐36. 456. Gold EJ, Conjalka M, Pelus LM, Jhanwar SC, Broxmeyer H, Middleton AB, Clarkson BD, Moore MA. Marrow cytogenetic and cell-­‐culture analyses of the myelodysplastic syndromes: insights to pathophysiology and prognosis. J Clin Oncol 1983, 1:627-­‐34. 457. Feher I, Gidali J. Prostaglandin E2 as stimulator of haemopoietic stem cell proliferation. Nature 1974, 247:550-­‐1. 458. Verma DS, Spitzer G, Zander AR, McCredie KB, Dicke KA. Prostaglandin E1-­‐
mediated augmentation of human granulocyte-­‐macrophage progenitor cell growth in vitro. Leuk Res 1981, 5:65-­‐71. 459. North TE, Goessling W, Walkley CR, Lengerke C, Kopani KR, Lord AM, Weber GJ, Bowman TV, Jang IH, Grosser T et al. Prostaglandin E2 regulates vertebrate haematopoietic stem cell homeostasis. Nature 2007, 447:1007-­‐11. 460. Spanbroek R, Hildner M, Steinhilber D, Fusenig N, Yoneda K, Rådmark O, Samuelsson B, Habenicht AJ. 5-­‐lipoxygenase expression in dendritic cells generated from CD34+ hematopoietic progenitors and in lymphoid organs. Blood 2000, 96:3857-­‐65. 101 References 461. Vore SJ, Eling TE, Danilowicz M, Tucker AN, Luster MI. Regulation of murine hematopoiesis by arachidonic acid metabolites. Int J Immunopharmacol 1989, 11:435-­‐42. 462. Braccioni F, Dorman SC, O'Byrne P M, Inman MD, Denburg JA, Parameswaran K, Baatjes AJ, Foley R, Gauvreau GM. The effect of cysteinyl leukotrienes on growth of eosinophil progenitors from peripheral blood and bone marrow of atopic subjects. J Allergy Clin Immunol 2002, 110:96-­‐101. 463. Elsas PX, Queto T, Mendonca-­‐Sales SC, Elsas MI, Kanaoka Y, Lam BK. Cysteinyl leukotrienes mediate the enhancing effects of indomethacin and aspirin on eosinophil production in murine bone marrow cultures. Br J Pharmacol 2008, 153:528-­‐35. 464. Kozubik A, Hofmanova J, Pospisil M, Netikova J, Hola J, Lojek A. Effects of drugs inhibiting prostaglandin or leukotriene biosynthesis on postirradiation haematopoiesis in mouse. Int J Radiat Biol 1994, 65:369-­‐77. 465. Estrov Z, Halperin DS, Coceani F, Freedman MH. Modulation of human marrow haematopoiesis by leucotrienes in vitro. Br J Haematol 1988, 69:321-­‐7. 466. Chung JW, Kim GY, Mun YC, Ahn JY, Seong CM, Kim JH. Leukotriene B4 pathway regulates the fate of the hematopoietic stem cells. Exp Mol Med 2005, 37:45-­‐50. 467. Bautz F, Denzlinger C, Kanz L, Möhle R. Chemotaxis and transendothelial migration of CD34+ hematopoietic progenitor cells induced by the inflammatory mediator leukotriene D4 are mediated by the 7-­‐transmembrane receptor CysLT1. Blood 2001, 97:3433-­‐40. 468. Boehmler AM, Drost A, Jaggy L, Seitz G, Wiesner T, Denzlinger C, Kanz L, Möhle R. The CysLT1 ligand leukotriene D4 supports α4β1-­‐ and α5β1-­‐mediated adhesion and proliferation of CD34+ hematopoietic progenitor cells. J Immunol 2009, 182:6789-­‐98. 469. Chen Y, Hu Y, Zhang H, Peng C, Li S. Loss of the Alox5 gene impairs leukemia stem cells and prevents chronic myeloid leukemia. Nat Genet 2009, 41:783-­‐92. 470. Nagashima T, Ichimiya S, Kikuchi T, Saito Y, Matsumiya H, Ara S, Koshiba S, Zhang J, Hatate C, Tonooka A et al. Arachidonate 5-­‐lipoxygenase establishes adaptive humoral immunity by controlling primary B cells and their cognate T-­‐cell help. Am J Pathol 2011, 178:222-­‐32. 471. DiMeo D, Tian J, Zhang J, Narushima S, Berg DJ. Increased interleukin-­‐10 production and Th2 skewing in the absence of 5-­‐lipoxygenase. Immunology 2008, 123:250-­‐62. 472. Desplat V, Ivanovic Z, Dupuis F, Faucher JL, Denizot Y, Praloran V. Effects of lipoxygenase metabolites of arachidonic acid on the growth of human blood CD34+ progenitors. Blood Cells Mol Dis 2000, 26:427-­‐36. 473. Cascio S, Zaret KS. Hepatocyte differentiation initiates during endodermal-­‐
mesenchymal interactions prior to liver formation. Development 1991, 113:217-­‐
25. 474. Houssaint E. Differentiation of the mouse hepatic primordium. II. Extrinsic origin of the haemopoietic cell line. Cell Differ 1981, 10:243-­‐52. 475. Tada T, Widayati DT, Fukuta K. Morphological study of the transition of haematopoietic sites in the developing mouse during the peri-­‐natal period. Anat Histol Embryol 2006, 35:235-­‐40. 102 References 476. Goulet JL, Snouwaert JN, Latour AM, Coffman TM, Koller BH. Altered inflammatory responses in leukotriene-­‐deficient mice. Proc Natl Acad Sci U S A 1994, 91:12852-­‐6. 477. Dennis RJ, Solarte I, Rodrigo G. Asthma in adults. Clin Evid (Online) 2011, 2011. 103