.,~:.;...:nr~ ‘q. CIA—.1 at.“ é3w¢m¢4 #931 ... ,1“ - u : 1‘ “13"!" g l . 3} u’, j 33’ 1'3 J I” "f . u - in :-!’J’ nil-7 ' t ,2“ ‘ (r 4.31.}. an a" :23}; ‘33,: 3133, $5523}, 7 ‘l’ '- . .- i f; This is to certify that the dissertation entitled STRUCTURAL AND CATALYTIC DETERMINANTS OF INTRACELLULAR CYCLOOXYGENASE PROTEIN DEGRADATION presented by URI R. MBONYE has been accepted towards fulfillment of the requirements for the Doctoral degree in Biochemistry /%/@g_ z . (524% Major Professor’s Signature rayon/a7 Date MSU is an affinnative—action, equal-opportunity employer PLACE IN RETURN BOX to remove this checkout from your record. TO AVOID FINES return on or before date due. MAY BE RECALLED with earlier due date if requested. DATE DUE DATE DUE DATE DUE 6/07 p:/ClRC/DateDue.indd-p.1 STRUCTURAL AND CATALYTIC DETERNIINANT S OF INTRACELLULAR CYCLOOXYGENASE PROTEIN DEGRADATION By Uri R. Mbonye A DISSERTATION Submitted to Michigan State Universtiy in partial fulfillment of the requirements for the degree of DOCTOR OF PHILOSOPHY Department of Biochemistry and Molecular Biology 2007 ABSTRACT STRUCTURAL AND CATALYTIC DETERMINANTS OF INTRACELLULAR CYCLOOXYGEN ASE PROTEIN DEGRADATION By Uri R. Mbonye Cyclooxygenases (COX-1 and COX-2) are ER-resident, membrane-bound glycoproteins that catalyze the committed step in the synthesis of biologically active lipid hormones called prostanoids. COX-1 is constitutively expressed in many mammalian cells, whereas COX-2 is usually expressed inducibly and transiently. In murine NIH/3T3 fibroblasts, COX-2 protein is degraded by a proteasome-dependent process with a half- life (ha) of about 2 h whereas COX-1 is reasonably stable (t1/2> 12 h). The mature forms of the COX isoforms are very similar in structure except that COX-2 has a unique 19- amino acid (19-aa) segment of unknown function located just near the C-terminus. Here, we provide evidence that the major role of the l9-aa cassette is to mediate entry of COX- 2 into the ER-associated degradation (ERAD) system that transports ER proteins to the cytoplasm for degradation by the 26S proteasome. COX-l expressed heterologously in HEK293 cells is quite stable (t1,2> 24 h) while COX-2 expressed heterologously is degraded with a tug of ~5 h and its degradation is blocked by proteasome inhibitors. A deletion mutant of COX-2 lacking 18 residues of the 19-aa cassette retains native COX-2 enzymatic activity and subcellular localization but, unlike native COX-2, is stable in HEK293 cells (t1,2> 24 h). Conversely, inserting the COX-2 cassette near the C-terminus of COX-1 yields a mutant ins594-612 COX-1 that is unstable (ti/2 ~ 3 h). A mannosidase inhibitor that can block entry of ER proteins into the ER-associated degradation system, retards the degradation of COX-2 and ins594-612 COX-l. Scanning mutagenesis of the l9-aa cassette has revealed that Asn-594, an N-glycosylation site at the beginning of the l9-aa cassette, is required, albeit insufficient to enable ERAD entry of COX-2 and ins594-612 COX-1. The remaining 16 amino acids of the l9-aa cassette are not only essential for COX-2 degradation but are also required for the enzyme to become glycosylated at Asn-594. Structural and biochemical analysis of the region immediately upstream of Asn-594 has revealed an 8-residue a—helical region that appears to impede Asn-594 glycosylation. We propose that a structural conformational change of the or- helical structure mediated by the C-terminal l6-residue sequence of the l9-aa cassette is needed for Asn—594 glycosylation. We show that the helical region upstream of Asn-594 and the l9-aa cassette actually constitute a C-terminal 27 instability element (27 IE) that dictates the intracellular stability of COX-2 by regulating glycosylation at Asn-594. We have also identified a second distinct mechanism of COX-2 protein turnover that is induced by arachidonic acid (AA), a major cylooxygenase (COX) fatty acid substrate. AA-dependent degradation of COX-2 is blocked by COX inhibitors, is not affected by inhibitors of proteasomal or lysosomal degradation, and occurs independently of the C- terminal l9-aa cassette of the enzyme. A COX-inactive point mutant of COX-2 (G533A COX-2) is resistant to AA-induced degradation even though it undergoes substrate- independent degradation at the same rate as native COX-2. Loss of COX-2 specific activity and prostanoid product formation is also observed in COX—2 expressing cells treated with AA. We propose that substrate-dependent inactivation of COX-2 causes structural damage to the enzyme that subsequently leads to its degradation. To Baba, my Mom, and the loving memory of my grandfather Reuben Rwabusizoni iv ACKNOWLEDGMENTS I would like to express my utmost gratitude to my advisor Dr. Bill Smith for his wonderful mentorship. He has done an excellent job of guiding me through a very challenging and thought-provoking thesis project. I have relied on his remarkable guidance and intellectual and technical expertise to be able to successfully complete my graduate career. I also sincerely acknowledge my advisory committee for providing me with very helpful advice and constantly ensuring that my thesis research is on the right track. I extend my appreciation to members of our lab, both present and past, for all the collaboration, advice, technical help, and friendship that I have received throughout my graduate career. Much thanks goes to Dr. Rana Sidhu for teaching me how to use the PyMOL software for protein structure analysis. He has also been a very cool and approachable guy that I have constantly turned to for suggestions and help on wide- ranging issues. Much thanks also goes to: Dr. Chong Yuan for teaching me, among other things, the very useful technique of overlap-extension PCR; Dr. Masayuki Wada and Jill Rieke for collaborative research related to examining the biological function of the COX- 2 C-terminal l9-amino acid cassette; Drs. Cindy DeLong and Inseok Song for helpful discussions and teaching me how to perform TLC assays; and Drs. Jiayan Liu, Yeon-Joo Kang, and Christine Harman for intellectually stimulating discussions. Thank you all for providing me with a conducive and friendly atmosphere to pursue my thesis research. Finally, I take this opportunity to also thank my family for their constant love and support. TABLE OF CONTENTS LIST OF TABLES ............................................................................... viii LIST OF FIGURES ................................................................................. ix ABBREVIATIONS ................................................................................ xii CHAPTER I: LITERATURE REVIEW Introduction ................................................................. l Mobilization of Endogenous AA for Prostanoid Synthesis .......... 6 Coupling between Cyclooxygenases and PLAzs ...................... 9 Functional Coupling between Cyclooxygenases and Terminal Prostanoid Synthases ........................................ 10 Regulation of Cyclooxygenase Gene Expression .................... 13 CHAPTER II: Cyclooxygenase Protein Structure and Subcellular Compartmentation ......................................... 20 Cyclooxygenase Catalysis, Substrate Specificity, and Suicide Inactivation ................................................. 28 Proteolytic Degradation of Intracellular Proteins .................... 33 Proteolytic Degradation of Endoplasmic Reticulum- associated Proteins ....................................................... 35 THE 19 AMINO CASSETTE OF CYCLOOXYGENASE-2 MEDIATES THE ENTRY OF THE PROTEIN INTO THE ER- ASSOCIATED DEGRADATION SYSTEM Summary .................................................................. 46 Introduction ............................................................... 47 Experimental Procedures ................................................ 49 Results ..................................................................... 53 vi Discussion ................................................................. 67 Acknowledgments ....................................................... 70 CHAPTER III: ASN-594 GLYCOSYLATION AND PROTEIN DEGRADATION OF COX-2 IS REGULATED BY ITS C-TERMINAL 27 AMINO ACID INSTABILITY ELEMENT Summary .................................................................. 71 Introduction ............................................................... 72 Experimental procedures ................................................ 75 Results ..................................................................... 79 Discussion ............................................................... 108 Acknowledgments ...................................................... l 16 CHAPTER IV: SUBSTRATE DEPENDENT COX-2 PROTEIN DEGRADATION Summary ................................................................. 1 17 Introduction .............................................................. 1 18 Experimental procedures .............................................. 121 Results .................................................................... 126 Discussion ............................................................... 153 CONCLUSION .................................................................................... 160 APPENDIX ......................................................................................... 162 BIBLIOGRAPHY ................................................................................. 170 vii Table 1. Table 2. Table 3. Table 4. LIST OF TABLES Participants of glycoprotein ERAD- L in yeast and mammalian cells .................................................................. 45 Relationship between the extent of Asn-594 glycosylation and overall protein degradation ................................................ 107 C-terminal mutants designed by QuickChangeTM site-directed mutagenesis ....................................................... 162 C-terminal mutants designed by overlap-extension PCR mutagenesis ...................................................................... 165 viii Figure 1. Figure 2. Figure 3. Figure 4. Figure 5a. Figure 5b. Figure 6. Figure 7a. Figure 7b. Figure 8. Figure 9a. Figure 9b. Figure 10. Figure 11. LIST OF FIGURES Synthesis and incorporation of AA into membrane glycerophospholipids ............................................................... 2 Stimulus-induced synthesis of bioactive prostanoids .......................... 5 Domain alignment of COX-1 and COX-2 highlighting major differences in their primary structures .................................... 6 Ribbons diagrams of the three dimensional structures of ovine COX-1 and murine COX-222 A view from the membrane plane showing the opening into the cyclooxygenase active site of ovine COX-l created by the membrane binding domain .................... 23 A view from the top showing the peroxidase active site of ovine COX-1. Proximal His-388 coordinates the heme prosthetic group (red) at the active site by bonding to the ferric heme ....................................................... 24 Ribbon diagrams showing two views (A and B) of the C-terminal structure of murine COX-2 ................. _ ................... 27 Mechanism for bis-oxygenation of AA and subsequent reduction of PGGz to form PGH; ............................................... 29 Proposed catalytic mechanism of the COX isoforms ........................ 3O Mechanism for the polyubiquitination of proteins prior to their degradation by the 26S proteasome .................. 37 Role of the calnexin/calreticulin cycle in ER quality control ............... 43 Processing of N-linked oligosaccharides in the ER lumen during proten quality control .................................................... 44 Cyclooxygenase protein degradation in serum—treated NIH/3T3 cells ...................................................................... 55 Stability of heterologously expressed native and C-terminal cyclooxygenase mutants in HEK293 cells .................. 58 ix Figure 12. Figure 13. Figure 14. Figure 15. Figure 16. Figure 17. Figure 18. Figure 19. Figure 20. Figure 21. Figure 22. Figure 23. Figure 24. Figure 25. Figure 26. Subcellular localization of native huCOX-2, delS95-612 huCOX-2, native ovCOX-l and ins594-612 ovCOX-l in HEK293 cells ................................................................... 60 Kinetic properties of purified native muCOX- 2 and de1595 -612 muCOX- 2 ............................................................ 61 Stability of cyclooxygenase mutants in HEK293 cells having various modifications of the 19 amino acid cassette ................ 63 Glycosylation of ins594-612 ovCOX-l and native huCOX-2 at Asn-594 ......................................................................... 66 Deletion and scrambled mutations of the 19-aa cassette disrupt COX-2 degradation ...................................................... 83 The C-terminal end of l9-aa is essential for COX-2 protein degradation ............................................................... 87 KIF treatment stabilizes the Asn-594 glycosylated form of COX-2 ........................................................................... 90 COX-2 stabilizing mutations of the l9-aa prevent glycosylation of Asn-59493 Structure of the C-terminal region of COX-1 and COX-2 .................. 98 Degradation of the UPSTRM8 huCOX-2 mutants .......................... 101 Asn-594 glycosylation of the UPSTRM8 huCOX-2 mutants and their degradation by the ERAD pathway ...................... 106 Schemes illustrating how the C-terminus of COX-2 may regulate Asn—594 glycosylation .......................................... 112 Two possibilities of how Asn-594 of native huCOX-2 may become accessible to glycosylation ......................... 113 A model of how Asn-594 glycosylation mediates the entry of COX-2 into the ERAD pathway ..................................... 114 Cyclooxygenase inhibitors prevent the rapid degradation of COX-2 in serum-treated NIH/3T3, but not HEK293, cells ........................................................... 130 Figure 27. COX-2 degradation in HEK293 cells is enhanced by exogenous AA treatment .................................................... 136 Figure 28. COX-2 degradation is selectively induced by PUFA COX substrates for the enzyme ................................................ 139 Figure 29. The G533A cyclooxygenase-inactive COX-2 mutant is not susceptible to substrate-dependent degradation ...................... 140 Figure 30. The enhancement of COX-2 degradation in NIH/3T3 cells by exogenous and endogenous AA is inhibited by NSAIDS ....................................................................... 145 Figure 31. COX-1 stability is not significantly affected by exogenous AA .................................................................... 146 Figure 32. Inhibitors of proteasomal and lysosomal degradation do not prevent substrate-dependent COX-2 protein turnover ......................................................... 150 Figure 33. COX activity and prostaglandin product formation of His ovCOX-l and His muCOX-2 cells pretreated with exogenous AA .............................................................. 152 Figure 34. Model for the basal and COX substrate-initiated degradation of COX-2 in HEK293 and serum-treated NIH/3T3 cells ..................................................................... 156 xi 2-AG ether 3’-UTR 19-aa AA BFA CHX COX CPLAza DHLA EDA Endo H EPA Epox ER ERAD ER Man I FB FBS GI GH Glc GlcNAc hu IFN-Y KIF LA Leup LPS Man mu NSAID OA OST ov PBS POX SPLAZ tet TLC UGGT UPSTRM8 ABBREVIATIONS 2-arachidonyl glycerol ether 3’-untranslated region C-terminal l9-amino acid cassette of COX-2 arachidonic acid ' brefeldin A cycloheximide cyclooxygenase cytosolic phospholipase Aza dihomo-y-linolenic acid eicosadienoic acid endoglycosidase H eicosapentaenoic acid epoxomicin endoplasmic reticulum ER-associated degradation ER Ot-l,2 mannosidase I flurbiprofen fetal bovine serum glucosidase I glucosidase 11 glucose N-acetyl glucosamine human interferon-y kifunensine linoleic acid leupeptin bacterial lipopolysaccharide mannose murine non-steroidal anti-inflammatory drug oleic acid oligosaccharyltransferase ovine phosphate buffered saline peroxidase secretory phospholipase A2 tetracycline thin layer chromatography UDP-glucose glucosyltransferase the 8 amino acid segment immediately upstream of Asn-594 in COX-2 xii CHAPTER I LITERATURE REVIEW Introduction Prostanoids, namely prostaglandins, prostacyclin and thromboxanes, are potent lipid signaling molecules that are synthesized in most mammalian tissues. These lipid hormones belong to the family of twenty carbon fatty acid derivatives that are collectively known as eicosanoids. Prostanoids act locally in an autocrine or paracrine manner through transmembrane G-protein coupled receptors to elicit a wide range of physiological and pathological responses (1-7). The major fatty acid precursor for prostanoids is widely believed to be arachidonic acid (AA), a twenty carbon fatty acid that is a metabolite of the dietary essential fatty acid linoleic acid. Once synthesized de novo AA is preferentially esterified to the sn-2 position of glycerophospholipids in lipid bilayers of cell membranes during phospholipid remodeling (Fig. 1) (8). The release of free AA from membrane phospholipids by phospholipase A2 is tightly regulated and represents the initial and first rate-limiting step of prostanoid synthesis (8-10). A second key point of control of prostanoid synthesis is the subsequent step involving the conversion of free AA to prostaglandin H2 (PGHz) by prostaglandin endoperoxide H2 synthase (PGHS) also known as cyclooxygenase (COX). This step is also rate-limiting for prostanoid formation and is considered to be the committed step in the pathway (1,3). Cell specific prostanoid synthases then catalyze the isomerization or reduction of PGHz to form various bioactive prostanoids (Fig. 2). Dietary linoleic Acid (18:2A”n(1)6) ATP + CoA AMP + PPi linoleoyl-CoA A6 desaturase y-linolenoyl CoA (18:2A6’9'12w6) malonyI-CoA dihomo-y-linolenoyl—CoA (20:3A8’11'Mw6) 0 R2__.| |__ 0— Co A As desaturase Arachidonoyl-CoA (20:4A5’3’ll’1‘ufi) l o o o—P—o—X R2 | | o l (I) o—lT-o-X 0 Figure 1. Synthesis and incorporation of AA into membrane glycerophospholipids. All mammalian cells except erythrocytes synthesize AA from the essential fatty acid linoleic acid (LA). As shown in the above scheme, AA synthesis from LA involves formation of two additional double bonds (A5 and A8) and two-carbon chain elongation. Once synthesized, AA is specifically incorporated into the sn-2 position of glycerophospholipids by acyltransferase during phospholipid remodeling. There are two known isoforms of cyclooxygenase: COX-1 and COX-2. Both are membrane-bound heme-containing glycoproteins that possess two sequential catalytic activities: a cyclooxygenase activity that bis-oxygenates AA to form the hydroperoxide prostaglandin G2 (PGGz) and a peroxidase activity that reduces the 15-hydroperoxyl group of PGGz to form PGHZ (Fig. 2) (1,3,7). COX-1 and COX-2 are structural and functional homodimers and are known to associate monotopically with the luminal faces of the endoplasmic reticulum (ER) membrane and the nuclear envelope (11-17). Their membrane association is such that the opening of the cyclooxygenase active site is in the lipid bilayer to allow the uptake of mobilized free AA. Despite being monotopically associated with the membrane these isoenzymes are considered to be integral membrane proteins because they can only be removed from the membrane with detergent and not with chaotropic salts (15). Thus, by associating with one leaflet of the lipid bilayer, the COXs constitute a unique class of integral membrane binding proteins. COX-1 and -2 are encoded by different genes; nonetheless, they share ~60% identity in primary structure. Both isoforms have a cleavable N-temiinal signal peptide sequence, an epidermal growth factor-like domain, a membrane binding domain, and a globular catalytic domain. At the C-termini of COX-1 and -2 are KDEL-like motifs that have been shown to be important for retention of the enzymes in the ER (18,19). Adjacent to the ER retention signal of COX-2 is a l9-amino acid insertion that is lacking in COX-l (Fig. 3). This C-terminal insertion also confers COX-2 with an additional consensus site for N-glycosylation. The biological role of this COX-2 C-terminal insertion will be explored in detail in Chapters 11 and 111. Although COX-1 and COX-2 are structural and functional isoforms, they have different expression patterns and biological functions. COX-1 is constitutively expressed in resting cells of most tissues (1,20). In contrast, COX—2 expression is usually absent in resting cells and can be induced in fibroblast, epithelial, endothelial, macrophage, and smooth muscle cells in response to growth factors, cytokines, mitogenic, and pro- inflammatory stimuli (1,20-24). Induced COX-2 expression is usually observed to be transient (21-24). It is possible that the differential expression profiles of COX-1 and -2 may contribute to their contrasting biological roles. In general, prostanoids generated as a result of COX-1 activity are believed to play housekeeping roles (25) while those generated by COX-2 are not only important physiologically but have also been implicated in various pathophysiological processes such as inflammation (26), pain (27), fever (28), angiogenesis (29), and tumorigenesis (30). The COXs are the best known cellular target for non-steroidal anti inflammatory drugs (NSAIDS) such as aspirin, ibuprofen, indomethacin, and flurbiprofen (1). O mass: A 333.53? 5383:. «not... 368:... 38—953 II.— _I|~. / >> 4 IX. 7on 2.? _ o IIIIIIIIIIII 3m. . .3 W o I o: Qasewgmoaoamo w my \ o:u TO I I t». I OI OI 30 $3998 (4 r 019 z» k n 3.83323 IO 4.3.0360qu OI $559.8 $5598 81? 3 ‘IE/ \.\ ..o D I I0 \\ m " .32! m: I0 01 mam—:6 N. mange—smbuacnaa mus—98mm 3. 38596 enema—seam. .25 mag B8 magnum find em Roman—Boa mew—demo: mm 9o mange—swinging Bog—masses 0». >> ~89 505335 uremusosuam 3. 2.8232630 >~ Got/NV. 38 >> mm 8:333 8 wmmw m: a 25-3% Boone: 90x08 OBS—Ema 3 En owaooxwmoammo 805 3a Rain—ewe QOVO coming cm OOXL Ba -N. womim m 983 ©8953 mew $503 ESSA—cam man So: mosoammoz mm 3855- BE oo=éfio mwoommo. Zm>~Um :55: 9o 00X waning e». OOXL Ba OOX-~. A5068 Mn“ 0 i” ii: “l’ 7 _.J Signal EGF MBD Globular-Catalytic . COX-1 Asn67 Asn144 Asn410 Asn594 T W W) Ii) .\ ERSTELCCC" Signal ~—~ EGF MBD Globular-Catalytic E :: COX-2 // L\ / \ / \ 594 / \612 §5§SSRSGLDDINPTVLLK human §§§ASHSRLDDINPTVLIK mouse §§§SSHSGLDDINPTVLLK ovine §§§ASHSRLDDINPTVLIK rat Figure 3. Domain alignment of COX-1 and COX-2 highlighting major differences in their primary structures. The cleavable ER signal sequence of COX-2 is shorter (18 residues) than that of COX-1 (25 residues). COX-1 is found to be glycosylated at three sites whereas COX-2 has four functional N-glycosylation sites. The last glycosylation site of COX-2 (Asn-594) is variably glycosylated and is part of a unique C-terminal l9-amino acid insertion (19-aa). Numbering for COX-1 begins with the Met at the translation start site. Numbering for COX-2 parallels the COX-1 numbering in which the start of the mature, processed COX-2 protein has the same number as the start of the mature, processed COX-1 (12). The Asn-594 glycosylation site is underlined, and 19-aa residues that are conserved in humans, mouse, sheep, and rat have been colored green. Images in this dissertation are presented in color. Mobilization of Endogenous AA for Prostanoid Synthesis Membrane glycerophospholipids can be deacylated at the sn-2 position by phospholipase A2 (PLA2) enzymes leading to the production of free polyunsaturated fatty acids, usually AA (31), and lysophospholipids. The mobilization of AA from the cell membrane for prostanoid synthesis depends on stimuli that will activate catalysis and/or induce the expression of the PLA2s involved. Therefore, AA liberation from the membrane is rate limiting for prostanoid metabolism. There is overwhelming evidence to indicate that of the nearly 22 mammalian PLA2 enzymes that have been cloned and characterized, cytosolic PLA20t (cPLA20L) and certain members of the family of secretory PLA2s (SPLA2) are largely responsible for the mobilization of AA for prostanoid metabolism (32-34). Of these two categories of CaZ+-dependent PLA2s, a consensus is emerging that cPLA201 is critical for stimulus-dependent liberation of AA from membrane phospholipids. cPLA20t is expressed constitutively in most mammalian cell types (35). The expression of cPLA20t can be elevated by various stimuli such as growth factors, cytokines and proinflammatory agonists (36-40). It is tightly regulated by multiple signaling pathways that control intracellular Ca2+ concentration and the phosphorylation state of the enzyme. Under the appropriate stimuli that will raise the intracellular cytosolic Ca2+ concentration to rnicromolar levels (eg. bradykinin and thrombin) (35,41- 43), cPLA20t will bind Ca2+ at its N-terminal C2 domain and, consequently, translocate to the ER membrane and nuclear envelope (44-46). It is thought that if the Ca2+ flux is low and transient, phosphorylation of the C-terminal catalytic domain of cPLA201 by mitogen activated protein kinases (MAPKs) may increase the affinity of the Ca2+-bound lipase for membrane binding (10,47). Caz+-dependent membrane association enables cPLA20L to preferentially cleave AA from the sn-2 position of phospholipid substrates. That CPLA20t is the phospholipase that is essential for mobilizing free AA from cellular membranes for eicosanoid metabolism is unequivocally demonstrated by studies using genetically deficient cPLA201 mice. These mice are generally reported to be healthy and fertile (48- 50). However, they are resistant to pathologies that require the actions of prostanoids and other eicosanoids generated from the agonist-induced mobilization of AA (48,49,51-57). These models include collagen-induced rheumatoid arthritis, bleomycin-induced lung fibrosis, brain ischemia reperfusion injury, acute respiratory distress syndrome, autoimmune diabetes, multiple sclerosis, and intestinal polyposis (48,49,51-57). sPLA2s, particularly those belonging to Groups IIA, V, and X are thought to complement cPLA20t activity by augmenting the release of cellular AA under certain pathophysiological settings such as those of an inflammatory response where the stimulus is robust and prolonged (58-60). Since sPLA2s are usually expressed extracellularly it is reasonable to expect that these lipases would have a significant role in transcellular prostanoid biosynthesis. sPLA2s require micromolar to millimolar concentrations of Ca2+ for catalytic activity and will indiscriminately hydrolyze cell surface phospholipids at the sn-2 position (10,61). Recent evidence has shown that prior to their secretion sPLA2s could also act on intracellular membranes to mobilize endogenous AA (33,34). The expression of sPLA2s, which is usually low in unstimulated immune effector cells (ie. macrophages, neutrophils, and mast cells) and in tissues of normal healthy individuals, is induced by proinflammatory stimuli such as IL-lB, TNFOL, interferon-y, phorbol ester, and bacterial lipopolysaccharide (62-66). Abnormally high levels of sPLA2-IIA can be detected in synovial fluid of patients with rheumatoid arthritis (67,68) or in plasma of patients experiencing systemic inflammation (69-71). Not surprisingly, the marked elevation of sPLA2-IIA during an inflammatory response is often accompanied by an induction of COX-2 expression and elevation of prostaglandins (10,72). The possible functional coupling between sPLA2-IIA and COX-2 to generate prostaglandins in inflammatory cells such as macrophages and mast cells appears to absolutely require cPLA201 activity (34,73,74). These observations have implied that the activation of cPLA20t precedes the mobilization of AA by sPLA2-HA for COX-2-mediated eicosanoid formation. However, the mechanistic basis for this crosstalk between cPLA20t and sPLA2-HA is not understood. Coupling between Cyclooxygenases and PLA2s Free AA that is mobilized by cPLA201 and sPLA2 is acted upon by cyclooxygenases to form the intermediate PGH2 that is a substrate for terminal prostanoid synthases. It is still not clear whether there is preferential coupling between cyclooxygenases and the PLA2s. Several research groups have reported that there exists two distinct phases of prostaglandin synthesis that are to a significant extent dictated by the stimulus involved in activating PLA2, the endogenous levels of free AA, and the temporal expression of COX-2 (75-77). The immediate phase usually occurs within minutes after stimulation with bradykinin or calcium ionophore (A23187) and is characterized by a rapid and transient increase in cytoplasmic Ca2+ concentration (76). Since COX-1 is constitutively expressed, it is reasonable to expect that during the immediate phase of prostaglandin and thromboxane synthesis cPLA20t is functionally coupled to COX-1. The delayed phase of prostaglandin synthesis which is induced by proinflammatory stimuli such as IL-IB occurs over several hours of cell stimulation and is largely mediated by inducible COX-2 activity (76). The delayed phase also appears to be dependent on cPLA2a activity even though this phase is not concomitant with the cytoplasmic elevation of Ca2+ (36,78,79). It is well known that COX-1 and COX-2 are differentially sensitive to the levels of endogenous AA. During the immediate phase the levels of liberated AA reach a level that is sufficiently high enough to be utilized by COX-l (76,77). However, the burst of AA release during the immediate phase is transient and as cells progress into the delayed phase AA release becomes limiting for prostaglandin synthesis. That COX-2 has been shown to be preferentially capable of utilizing AA at low concentrations (1,76,77) would reasonably explain why this COX isoform is the predominant cyclooxygenase during the delayed phase. It has been suggested that differences in the enzymatic properties of COX- 1 and COX—2 may account for their segregated utilization of AA in cells that coexpress both enzymes (1). Functional Coupling Between Cyclooxygenases and Terminal Prostanoid Synthases Cyclooxygenases are thought to exhibit distinct functional coupling with the downstream prostanoid synthases, which include prostaglandin E2, prostaglandin D2, prostaglandin F2a, prostacyclin, and thromboxane synthases. It is well established that in platelet cells, which do not express COX-2, thromboxane A2 (T XA2) is synthesized due to functional coupling between COX-1 and thromboxane synthase (TxAS), both of which are constitutively expressed (80). Platelet-synthesized TXA2 acts in an autocrine fashion to induce platelet aggregation and in a paracrine fashion to contract vascular smooth muscle cells (81). The beneficial effect of low dose aspirin in reducing the risk of myocardial infarction and other cardiovascular diseases is due to the suppression of platelet TXA2 synthesis. Stimulation of macrophage cells with Ca2+ ionophore leads to formation of TXA2 during the immediate phase (82). In this case, constitutively 10 expressed COX-1 is functionally linked to TxAS. Eliciting the delayed phase in macrophage cells by stimulation with bacterial lipopolysaccharide (LPS) can also result in TXA2 synthesis due to coupling between inducible COX-2 and TxAS (82). The anti-thrombotic and vasodilatory activities. of prostacylin (PGI2) (83) counteract the effects of TXA2 to achieve a balance that is crucial for maintaining vascular homeostasis. Vascular endothelial cells synthesize prostacyclin under normal physiological conditions (84) and during stress (85,86). Very low basal levels of PGI2 can be detected in resting endothelial cells attributable to COX-1/PGIsynthase coupling (87). In these cells mechanical stimuli, such as shear stress, and inflammatory stimuli, such as LPS or TNFOL, result in coordinate induction of COX-2 expression and a dramatic rise in the levels of PGI2 (85-87). That COX-2 is functionally coupled to PGI synthase in vascular endothelial cells provides an explanation for the negative side effects of anti- inflammatory COX-2 selective inhibitors on the cardiovascular system. Recent clinical trials have shown that these inhibitors are associated with an increased risk of myocardial infarction and stroke (88,89). Vascular smooth muscle cells, which express high basal levels of PGI synthase (90), are reported to generate PGI2 under stress and/or inflammatory conditions (91). In LPS- or TNFOL- stimulated macrophage cells, COX-2- derived PGI2 is thought to be important for the resolution of the inflammatory response through suppression of the synthesis of proinflammatory cytokines (92). PGD synthase (PGDS) catalyzes the isomerization of the endoperoxide PGH2 to form PGD2. Notably, PGD2 is produced in brain where it is involved in sleep induction and regulation of pain (93,94). PGD2 is also synthesized by mast cells in response to allergens and is believed to mediate allergic and inflammatory responses (95-97). The 11 chemical dehydration of PGD2 yields 15-deoxy-A'2’l4-PGJ2 which is thought to be the endogenous ligand for the nuclear receptor PPARy (98-100). Acting via PPARy this cyclopentenone prostaglandin has been shown to have anti-inflammatory activity and is thought to be formed during the resolution phase to counter the proinflammatory effects of other prostaglandins, including PGD2 (100-103). Two distinct forms of PGDS exist: lipocalin-type PGDS (L-PGDS), a secreted protein that is highly expressed in the central nervous system (CNS), and hematopoietic PGDS (H-PGDS) expressed in mast cells, megakaryocytes, and macrophage cells (104). In a model of systemic inflammation, PGD2 synthesis in the CNS by L-PGDS is dependent on COX-2 activity (105). In mast cells, the generation of PGD2 upon challenge with IgF/antigen or A23187 is COX-1 dependent (40,82). However, stimulating mast cells with cytokines in the presence of IgF/antigen not only induces COX-2 expression but also results in a six-fold increase in PGD2 synthesis (106). Prostaglandin E2 (PGE2) is the major prostanoid that has been implicated in the pathophysiology of inflammation, pain, and fever. Genetic inactivation of COX-2 or selective inhibition of the enzyme suppresses inflammation, pain, and fever concomitant with a marked decrease in PGE2 generation (107-109). This observation is consistent with the idea that PGE2 synthesis under these pathophysiological conditions is mainly COX-2-derived. Three PGE synthase enzymes that catalyze the conversion of PGH2 to PGE2 have been identified. These are cytosolic PGE synthase (cPGES), and membrane- bound PGE synthases-1 and -2 (mPGES-l and -2). cPGES is constitutively expressed in a wide variety of cell types (110) and its expression is usually not enhanced by proinflammatory stimuli. This enzyme has been shown to be capable of forming PGE2 12 from COX-1-, but not COX-2-, derived PGH2, particularly during the immediate Ca2+- dependent phase of prostaglandin synthesis (110). Of the three PGE synthases, mPGES- 1 is believed to be crucial for PGE2 generation under various pathological conditions. mPGES-l is coordinately induced with COX-2 in cells and tissues in which COX-2- derived PGE2 is believed to exert physiological and/or pathological actions (109). Just like COX-2, mPGES-l expression can be inhibited by anti-inflammatory glucocorticoids (111). Macrophage cells isolated from mPGES-l null mice generate much smaller amounts of PGE2 relative to wild type upon stimulation with LPS (112). Just like cPLA20L and COX-2 deficient mice (51,113), mPGES-l nulls have been reported to be resistant to collagen-induced arthritis (114), a model of chronic inflammation. Moreover, similar to COX-2-deficient mice (108), these mPGES-l null mice have an impaired LPS—induced febrile response that is accompanied by a significant reduction of PGE2 levels in the central nervous system (115). These observations suggest that functional coupling between cPLA201, COX-2, and mPGES-l is important for stimulus-induced PGE2 formation under various stress or pathological conditions. Regulation of Cyclooxygenase Gene Expression Transcriptional Regulation Although COX-1 and COX-2 exhibit a high degree of structural homology they are the products of two distinct genes that are regulated differently. COX-l is constitutively expressed in most mammalian tissues (1,20). The COX-1 gene is approximately 22 kb in length, is located on chromosome 9, and is transcribed as a 2.8 kb mRNA (20). The COX-1 gene promoter is TATA-less, CAAT—less, GC-rich, and 13 contains multiple transcription start sites (116). These promoter features are usually characteristic of housekeeping genes that are constitutively expressed under basal conditions. The COX-l promoter also contains three potential SP1 binding sites at -610, - 111, and -89 relative to the ATG start codon. Reporter gene assays have demonstrated that the SP1 sites at -610 and -111 are functionally important in maintaining basal constitutive expression of COX-1 (117). Inducible COX-1 expression has been observed in a few cell types. For instance, differentiation of megakaryoblasts into megakaryocytes in vitro upon stimulation with phorbol ester (PMA) is usually accompanied by at least a 5-fold increase in COX-1 mRNA and protein (118,119). Megakaryocytes are the precursors of platelets which lack nuclei and constitutively express high basal levels of COX-1. Recently, it has been reported that the SP1 site at -l 11 works in conjunction with an intronic cis-acting AP-l site to facilitate PMA-induced expression of COX-1 in megakaryoblasts (119). Inducible COX-l is also observed in vascular endothelial cells that have been subjected to shear stress (86). Upon induction, COX—1 mRNA expression is usually prolonged in these cells. In contrast, induction of COX-2 mRNA by shear loading of endothelial cells is short-lived (86,120). COX—2 was initially identified as a rnitogen-responsive immediate early gene in fibroblast cells (121-124). Since then inducible COX-2 expression has been reported in a wide variety of cell types, including smooth muscle cells, macrophages, endothelial cells, synoviocytes, chondrocytes, pancreatic islet cells, and epithelial cells lining the airways, stomach and intestine. Irnportantly, induced COX-2 is reported to be short-lived in endothelial, smooth muscle, fibroblast, and epithelial cells (21-24). However, there are a few exceptions to the inducible and transient expression of COX-2; constitutive l4 expression of COX-2 has been reported in kidney, neuronal cells, and lung epithelial cells under physiological conditions (125-127). COX-2 expression is notably absent in erythrocytes, lymphocytes, and platelets. Various stimuli will induce COX-2 depending on the cell type. These stimuli include mitogens, inflammatory cytokines (IL-1 B, IL-IOL, TNFOL, and interferon-y), pro-inflammatory factors (eg. gram negative bacterial LPS), growth factors (FGF, PDGF, VEGF, and EGF), hormones (estrogen, luteinizing hormone, follicle stimulating hormone), oncogenes (v-Src and v-Ras), and mechanical stimuli such as fluid shear stress (1,20). Many of these COX-2-inducing stimuli will also activate the mobilization of AA from the cell membrane. More recently, it has been shown that some prostaglandins, particularly PGE2, which would be formed during the immediate phase due to COX-l activity, may activate their respective G protein coupled receptors to induce COX-2 in a cyclic AMP- and protein kinase A-dependent fashion (128). The 8.3 kb COX-2 gene is located on chromosome 1 and is mainly transcribed as a 4.5 kb mRNA (20). The last exon of the COX-2 gene encodes the 3’-untranslated region (3’-UTR), which contains multiple copies of the ‘AUUUA’ RNA instability elements that have a crucial role in the post-transcriptional regulation of COX-2 (20,129). Unlike COX-l, the COX-2 promoter is replete with putative cis-acting regulatory elements suggesting tight and complex regulation of the gene by numerous signaling pathways. Of these cis elements, those that have been identified to have a regulatory role in COX-2 transcription include E-Box, CAMP response element (CRE), NFKB, AP-l, CAAT enhancer binding protein (C/EBP), SP1, serum response element (SRE), and peroxisome proliferator response element (PPRE) (1,20,130,131). Subtle differences exist 15 in the COX-2 promoter sequence between the human and mouse gene. For instance, the human gene has one CRE, one NFKB, and two C/EBP elements while the mouse gene has one C/EBP, two CRE, and two NFKB sites. Depending on the cell type and the stimulus, distinct combinations of cis- regulatory elements will be utilized to activate COX-2 transcription. This is most evident during septic shock, a systemic inflammatory response of the host to infection by gram negative bacteria. The initial recognition of the LPS component of the bacterial cell wall by monocytes and macrophages that comprise the host innate immune system will lead to the activation of COX-2 gene transcription (130,132). LPS is recognized by toll-like receptor 4 (TLR4) resulting in signaling cascades that lead to the simultaneous activation of the NFKB, and the three MAP kinase (ERK, p38, and JNK) pathways (131,133). LPS- induced MAP kinase signaling activates C/EBP, CREB, and the AP-l transcription factor complex (130,131,133). These activated trans-acting factors localize to the nucleus and bind the COX-2 promoter upon which they act in concert to stabilize the binding of coactivator complexes that are responsible for recruiting the basal transcriptional machinery (ie. RNA polymerase II and its associated factors) needed to initiate transcription. COX-2 inducible gene expression can be strongly retarded by glucocorticoids. There is evidence indicating that these anti-inflammatory steroids may exert part of their inhibitory action by suppressing COX-2 transcription. However, it is not clear how glucocorticoids negatively regulate COX-2 transcription; moreover, the COX-2 promoter lacks a glucocorticoid response element suggesting that the regulation may be indirect. Several research groups have independently shown that dexamethasone, a glucocorticoid 16 analog, prevents the activation of NFKB and its subsequent nuclear localization by upregulating the expression of IkBoc (134,135), an NFKB-interacting protein that sequesters the transcription factor in the cytosol. Therefore, it is highly likely that glucocorticoids may inhibit COX-2 transcription by interfering with the activation of NFKB. The anti-inflammatory cyclopentenone prostaglandin 15-deoxy-A'2‘14-PGJ2 (15-d PGJ2), the dehydration product of PGD2, is known to suppress COX-2 transcription (136,137). 15-d PGJ2 binding to its nuclear receptor PPARy inhibits PMA-mediated induction of COX-2 in human epithelial cells by preventing binding of the AP-l trans- acting complex to the promoter and the subsequent recruitment of the CBP/p300 coactivator complex (138). Inhibition of AP-l transactivation of COX-2 can be rescued by a PPAR response element decoy oligonucleotide suggesting that ligand-bound PPARy may bind the PPRE site of the COX-2 promoter to repress transcription. Post-transcriptional regulation COX-2 mRNA has a very short half-life. This can be explained by the presence of multiple copies of the AUUUA motif within the 3’-UTR of COX-2 mRNA that are known to direct mRNA decay (20,129). Deletion of these cis-acting decay motifs from the 3’-UTR of COX-2 stabilizes the transcript (20,22). Unlike COX-2, COX-1 mRNA is very stable and its 3’-UTR lacks these AU-rich elements (ARES) (22). The mechanism by which ARES promote mRNA degradation in mammalian cells is not clearly understood. There is general agreement that ARES promote deadenylation of the polyA tail which precedes 5’ to 3’ or 3’ to 5’ exonuclease cleavage of the message (139-142). Most 17 recently, Stoecklin et al. have reported that ARE-mediated deadenylation leads to decapping of the 5’ 7-methyl guanosine cap causing the message to be degraded S’to 3’ by the exonuclease Xml (142). Although numerous ARE binding proteins have been shown to interact with COX-2 ARES, the ARE binding protein(s) responsible for initiating ARE-mediated decay of COX-2 mRNA is yet to be identified. The activation of the p38 MAPK pathway by pro-inflammatory stimuli has been implicated in stabilizing COX-2 mRNA (143). Stabilization of the COX-2 transcript by p38 MAPK signaling is inhibited by the glucocorticoid dexamethasone and is believed to require a l23-nucleotide region within the 3’-UTR and immediately downstream of the termination codon that has six ARE copies (144). It is not clear what ARE binding proteins become activated by p38 MAPK signaling to effect ARE-dependent COX-2 stabilization. However, various ARE binding proteins such as CUGBP2 and HuR have been shown to bind the COX-2 3’-UTR and stabilize the transcript. HuR-mediated COX-2 transcript stabilization has been reported in colon cancer cells where COX-2 is aberrantly over-expressed (145). In contrast, CUGBP2-mediated COX-2 transcript stabilization is observed in epithelial cells undergoing apoptosis due to radiation exposure (146,147). In these cells, CUGBP2 has also been shown to have the paradoxical role of inhibiting translation of the COX-2 transcript. This is believed to be the mechanism by which radiation-induced expression of CUGBP2 in epithelial cells inhibits COX-2-mediated formation of PGE2 that is known to have anti-apoptotic and mitogenic activities. The translational silencing of COX-1 has been reported to occur in megakaryocytes. During the PMA-induced differentiation of the megakaryocytic cell line 18 MEG-01 COX-1 mRNA is upregulated within a day of PMA treatment while an increase in COX-1 protein is not observed for several days (118). Duquette and Laneuville have reported a correlation between the occupancy of a putative 20-nucleotide cis element within the COX-1 3’-UTR by a protein complex and the inhibition of COX-1 protein synthesis (118). This complex of COX-1 mRNA binding proteins is yet to be characterized. Cyclooxyge’nase protein turnover It is well established that the distinct profiles of expression of COX-1 and COX-2 observed in some mammalian cells could at least be partly attributed to differences in regulation of the isoforms at the transcriptional and post-transcriptional levels. Although it is not clear whether these enzymes are also regulated differently at the post- translational level by proteolysis, their distinct patterns of expression suggest that this may be the case. Certainly, rapid turnover of COX-2 in tissues where the enzyme is transiently expressed may serve a significant physiological role in regulating the levels of prostanoids whose synthesis is attributed to this COX isoform. Consistent with this idea Rockwell and co-workers have reported observing the accumulation of COX-2 in its native form and as polyubiquitin conjugates in HT4 neuronal-like cells treated with inhibitors or disruptors of proteasomal degradation (148,149). In contrast, COX-1 protein levels were unaffected by this treatment. These experimental results suggest that COX-2 may be selectively regulated by the ubiquitin-proteasome pathway, which has been implicated in the proteolysis of intracellular proteins with short half-lives. My thesis will 19 attempt to identify and effectively address the molecular basis for the selective regulation of COX-2 by proteolytic degradation. Cyclooxygenase Protein Structure and Subcellular Compartmentation Cyclooxygenases are membrane-bound, heme-containing glycoproteins that are resident in the ER lumen and the contiguous lumen of the nuclear envelope (1 1-17). They are believed to exist and function as homodimers with each monomeric subunit possessing cyclooxygenase and peroxidase active sites (11,17,150). X-ray crystal structures of mature COX-1 and COX-2 from several mammalian species have already been determined. Their three dimensional structures exhibit a high degree of homology which would be expected since both enzymes are ~60% identical in primary structure. The maturation of COX-1 and COX-2 in the ER lumen involves cleavage of the N-terminal ER targeting signal sequence, N-glycosylation at multiple sites, disulfide bond formation, heme incorporation, membrane insertion, and dimerization. These co- translational and post-translational modifications yield COX-1 and COX-2 mature glycoproteins with monomeric masses of ~70 and ~72-74 kDa, respectively (151). There are three major folding domains in mature COX-1 and -2: an epidermal growth factor (EGF) domain, a membrane binding domain (MBD), and a globular catalytic domain (Figs. 3 and 4). The EGF domain forms a significant portion of the dimer interface; however, it is not clear whether it is critical for homodimerization. It has been proposed that the EGF domain may have an important role in enabling the monotopic insertion of the maturing cyclooxygenase into the membrane (152). The MBD is the only membrane anchor for cyclooxygenases that has been identified thus far. It is made up of four 20 consecutive amphipathic or-helices that surround the opening of the cyclooxygenase active site (Figs. 4 and 5). Hydrophobic and aromatic side chains protrude downwards from these amphipathic helices and become inserted into one leaflet of the lipid bilayer. Hydropathy analysis of the MBD of both isoforms indicates that the COX-1 MBD is significantly more hydrophobic than that of COX-2 (16). Nonetheless, in both cases the MBD-mediated monotopic insertion creates an unusually tight membrane association that cannot even be disrupted with a high salt wash (15,16). Therefore, the cyclooxygenases constitute a unique class of monotopic integral membrane binding proteins. The cyclooxygenase catalytic domain is globular and is mostly comprised of 01- helical secondary structure. The cyclooxygenase (COX) active site is an ~25 A long hydrophobic channel that begins at the MBD and extends upwards into the protein hydrophobic core. In order to be utilized as substrate, unesterified AA has to enter the COX active site through the mouth formed by the MBD. The productive conformation of AA at the COX active site is such that its (1) end extends into the hydrophobic core while the A carboxyl end is stabilized at the mouth of the active site through electrostatic interactions with the guanidium group of Arg-120 (153,154). The peroxidase (POX) active site is solvent-exposed and contains a ferric-protoporphyrin IX heme group that is bonded to the proximal histidine, His-388 (Fig. 5b). Both COX isoforms can be purified as apo-enzymes indicating that the ferric heme is reversibly bound to the POX site. Although the POX and COX active sites are spatially distinct they are functionally interdependent. The COX oxygenation reaction requires activation by POX activity (155,156). In turn, PGG2 formed at the COX active site diffuses to the POX active site where it is reduced to form PGH2. 21 Figure 4. Ribbons diagrams of the three dimensional structures of ovine COX-1 (top) and murine COX-2 (bottom). The COX-1 and COX-2 structures are virtually superimposable. One monomer of each enzyme is colored orange; the other monomer is colored to show the N-terminal epidermal growth factor-like domain (green) at the dimer interface, the four amphipathic helices that make up the membrane binding domain (gray), and the globular catalytic domain (cyan). N-linked oligosaccharide groups (blue) are also shown. 22 Figure 5a. A view from the membrane plane showing the opening into the cyclooxygenase active site of ovine COX-1 created by the membrane binding domain (gray). The carboxylate group of bound AA (yellow) forms a salt bridge with the guadinium group of Arg-120 thereby stabilizing the substrate at the active site. Shown in the background is the catalytic residue Tyr—385 which is suitably positioned to abstract the l3pr0S hydrogen of AA. Subtle differences exist between the primary structures of COX-1 and COX-2. The N- terminal signal sequence of COX-1 is longer (25 residues) and more hydrophobic than that of COX-2 (18 residues) (Fig. 3). After cleavage of the signal sequence the N- terrninal most residue in both enzymes is usually an alanine. For convenience in structural comparisons between the isoforms the sequence of mature COX-2 is numbered such that it parallels the COX-1 numbering in which the start of the mature, processed COX-2 protein has the same number as the start of the mature, processed COX-1 (12). COX-2 numbering parallels that of COX-1 up to the C-terminus, where a 19-amino acid insertion occurs in COX-2 six residues in from the C-terminal end. This C-terminal 23 Figure 5b. A view from the top showing the peroxidase active site of ovine COX-1. Proximal His-388 coordinates the heme prosthetic group (red) at the active site by bonding to the ferric heme. Mutagenesis studies have shown that both His-388 and distal His-207 are important for POX catalysis. Although the role of distal His-207 in POX catalysis has not been lucidly defined, it’s proposed to be important for deprotonation of the hydroperoxide substrate and reprotonation of the alkyloxide ion intermediate to form an alcohol (151). Tyr-385 and the membrane binding domain (gray) are shown to indicate the position of the COX active site relative to the POX active site. insertion imparts COX-2 with a consensus N—glycosylation site at Asn-594 (Fig. 3) that has no counterpart in COX-l. As a result COX-1 is found to be N-glycosylated at three sites, while COX-2 is glycosylated at its first three N-glycosylation sites and variably glycosylated at Asn—594 (151). It has been hypothesized that the C-terminal COX-2 insertion may serve as a signal to regulate protein turnover or the subcellular compartmentation of COX-2 (1,152). In view of the short-lived expression of COX-2 protein in cellular contexts 24 where COX-1 is constitutive (21-24), it is possible that the insertion could mediate the selective and rapid degradation of COX-2. The C-terminus of COX-2 has not been successfully resolved in its entirety by X-ray crystallography analysis. The last resolved residue in the highest resolution murine COX-2 crystal structure attained thus far (the human COX-2 structure is not available in the PDB even though it has already been solved) is Ser-596 (157), the final amino acid in the Asn-594 N-glycosylation consensus sequence at the beginning of the insertion. This N—glycosylation site is part of a two- tum helix (TKTATIlm) that is linked to an upstream helix by a long 15-residue loop that includes 11 resolved residues (KGCPFI‘SFNVQ) and an unresolved 4-residue sequence (DPQP) (Fig. 6). Asn-594 is not glycosylated in the murine structure presumably because the amide group of its side chain is pointed upwards towards the upstream helix that is at a distance of ~4.5 A. This space is barely sufficient to accommodate an N-glycan group. Moreover, a disulfide bridge between Cys-569 of the upstream helix and Cys-575 of the loop may further hinder Asn-594 glycosylation. Therefore, in order for Asn-594 to be glycosylated a local conformational change would have to occur, probably involving breakage of the disulfide bond and movement of the long and inherently flexible intervening loop. The inability to resolve the remaining 16 amino acids of the C-terminal l9-amino acid insert in COX-2 crystal structures may be an indication that this portion is largely disordered and lacks any degree of secondary structure. The hydroxyl side chain of Ser- 596 is solvent-exposed and is situated close to the surface of the membrane (157). The KDEL-like C-terminal ER retention signal of COX-2 also has to be solvent-exposed and near the membrane in order to be bound by membrane proteins involved in ER retention 25 (18,19). Therefore, the unresolved portion of the COX-2 insert may occur on the surface of the protein close to the membrane surface. My thesis project has tested the hypothesis that the biological role of the C-terminal 19-amino acid insertion is to selectively regulate COX-2 protein turnover. I have also attempted to decipher the molecular mechanism by which the COX-2 insertion may initiate and promote protein degradation. My findings are presented and discussed in detail in Chapters 11 and III. Ser-596 26 Figure 6. Ribbon diagrams showing two views (A and B) of the C-terminal structure of murine COX-2. The last resolved residue in this structure is Ser-596 which is the final amino acid of the Asn-594 consensus glycosylation site. Not surprisingly, Asa-594 is not glycosylated most likely because its amide side chain is pointed away from the surface and in the direction of a nearby helix. Both helices are separated by a long 15-residue loop; the last four residues of this loop are not resolved in the structure. A disulfide bond is formed between the thiols of Cys-569 and Cys-575 which may prevent glycosylation of Asn-594. Futhermore, the distance between Asn-594 and Cys-569 is too small to accommodate an N-glycan group. For Asn-594 to become glycosylated the disulfide bond may have to be reduced to increase the flexibility of the loop. The catalytic domain is colored green and the MBD is colored gray. Cyclooxygenase Catalysis, Substrate Specificity, and Suicide Inactivation The committed step in the prostanoid synthesis pathway, catalyzed by COX-1 and COX-2, is actually a two-reaction step that involves: a) the incorporation of two molecules of O2 to AA (5,8,11,14-eicosatetraenoic acid) to form the hydroperoxy endoperoxide PGG2, and b) the two electron reduction of PGG2 to form the hydroxy endoperoxide PGH2(1,3). The endoperoxide bridge of PGH2 is then acted upon by distinct terminal prostanoid synthases to form different prostanoids. The proposed catalytic mechanism of the cyclooxygenases is shown in Fig. 7 (156,158). Both the peroxidase (POX) and cyclooxygenase (COX) reactions are dependent on the ferric- protoporphyrin IX (Fe3+-IX) heme prosthetic group which is coordinated at the active site by proximal His-388. The POX activity is required to initiate the COX activity. Cyclooxygenase catalysis commences at the POX active site where an initiator hydroperoxide that is yet to be identified oxidizes the heme group by removal of two electrons yielding an oxyferryl heme radical intermediate called compound I. Neutralization of this radical intermediate requires an electron provided by a neighboring tyrosine residue (T yr-385) in an intramolecular reaction that leads to the formation of intermediate 11 comprising an oxyferryl heme and a tyrosyl radical. Thereafter, the reactive tyrosyl radical initiates COX catalysis by abstracting the 13-proS hydrogen from AA at the COX active site yielding a radical at the 13-carbon (C-13). The AA radical undergoes an isomerization that eventually transfers the radical to C-ll. Subsequently, an endoperoxide bridge is formed between C-9 and C-11 after reaction with one molecule of 02. As this happens, a second molecule of O2 is added to the C-15 position forming the hydroperoxy] PGG2. 28 CYCLOOXYGENASE PEROXIDASE Figure 7a. Mechanism for bis-oxygenation of AA and subsequent reduction of PGG2 NIT—W COOH \=/<-_—/\/V ARACHIDONIC ACID to form PGH2. Reproduced with permission from Ref. 156. 29 POX suicide 0“ R01] R00“ 0 0" Inactivation Ai-h / ‘ Compound I Resting enzyme Tyr-385 . Tyr-385 POX Intermediate ll Tyr-385 Tyr-385 0 0H % AA COX 2 02 I COX suicide AA inactivation Tyr-385 Figure 7b. Proposed catalytic mechanism of the COX isoforms. Cyclooxygenase catalysis is initiated by hydroperoxide which oxidizes the ferric heme to form compound I. This enzyme intermediate undergoes an intramolecular rearrangement of electrons resulting in a shift of the radical from the heme to Tyr-3 85. Both radical intermediates are very reactive and may cause modifications in the protein that lead to suicide inactivation. The reactive tyrosyl radical at Tyr-385 will abstract the l3pr0S hydrogen of AA as shown in Fig. 7a leaving a radical at C-13. This initiates bis-oxygenation of AA to form PGG2 which is subsequently reduced at the POX site to form PGH2. The reduction of PGG2 is coupled with formation of compound I and the initiation of a second catalytic cycle. There is no direct channel connecting the COX and POX active sites through which PGG2 can diffuse. It has been proposed that PGG2 must exit the COX site by first diffusing through the opening in the MBD and then traveling around the protein surface in order to reach the POX site (156). The two electron reduction of PGG2 at the POX site has the dual role of forming PGH2 and regenerating compound I for procession through 30 another catalytic cycle. The kinetic constants for the COX activity of COX-1 and -2 are very similar. When AA is utilized as substrate, both enzymes have similar COX specific activities (~25-30 umol/min of substrate per mg of enzyme) and the K1m of the purified or microsomal (membrane-bound) forms of these enzymes is 7~ 5 11M (1). The catalytic activities of the cyclooxygenase isoforms are distinctly regulated. This became initially apparent when Reddy and Herschman demonstrated that stimulus- induced prostaglandin synthesis in mitogen-stimulated 3T3 fibroblasts could almost entirely be attributed to inducible COX-2 activity, even though COX-1 is constitutively expressed at detectable levels in these cells (77). A year later, it was reported that the COX activities of COX-1 and COX-2 are initiated by different levels of hydroperoxide (155). Using varying amounts of the hydroperoxide scavenger, glutathione peroxidase, to regulate the levels of hydroperoxide in the reaction mixture needed to initiate COX activity, Kumalcz and Wang were able to show that both COX-1 and COX-2 COX activities could be stimulated by nanomolar concentrations of hydroperoxide, with the latter activity requiring 10-fold less hydroperoxide. Interestingly, they also noted that addition of COX-2 to a COX reaction mixture containing COX-1 did not significantly improve COX-1 activity at low hydroperoxide concentrations, suggesting that the amount of PGG2 generated by COX-2 was not sufficient to initiate COX-1 COX activity. Therefore, it is conceivable that in cells coexpressing both isoforms COX-2 could be the functionally active isoform with COX-1 remaining in its latent or resting form at low intracellular hydroperoxide levels. That COX-1 is mostly active during the immediate phase of prostanoid synthesis would suggest the possibility of a peroxide flux that would be sufficiently high to activate COX-1 during this phase but insufficient to activate COX- 31 1 during the delayed phase. Alternatively, the inability of COX-1 to utilize AA at low concentrations as is observed during the delayed phase could also contribute to the temporal segregation of COX-1 and COX-2 activities. Although the hydroperoxide responsible for initiating COX activity in vivo is not known, COX-1 and -2 can utilize a broad variety of hydroperoxides as substrates. In addition to PGG2, COX-1 and -2 can reduce hydrogen peroxide, t-butyl hydroperoxide, ethyl hydroperoxide, lS-hydroperoxy eicosatetraenoic acid (lS-HPETE), and cumene hydroperoxide (156). Despite their broad substrate specificity, the COXs tend to prefer secondary alkyl hydroperoxides such as 15-HPETE and PGG2 (156,159,160) and (Liu and Smith, unpublished results). With respect to COX activity, AA, the physiological substrate of both COX isoforms, is the most preferred substrate as determined by kinetic measurements of oxygenation efficiency (156,161). Unlike COX-1, COX-2 can also oxygenate a variety other substrates in vitro albeit not as efficiently as AA (156). These substrates include, 2-arachidonyl glycerol (2-AG), an ester derivative of AA, anandamide, an amide derivative of AA, eicosapentaenoic acid (EPA), and dihomo-y- linolenic acid (DHLA). COX-2 is able to selectively utilize 2-AG and anandamide as substrates because it has an active site side pocket extension that creates a larger active site compared to that of COX-1 that can accommodate these arachidonyl derivatives (3). This side pocket extension has been exploited by the pharmaceutical industry to design selective COX-2 NSAIDS. The POX and COX activities of the COXs are well known to undergo self inactivation after an unknown number of rounds of catalytic turnover. POX and COX self inactivation is irreversible and is also mechanism-based since it is induced by POX or 32 COX substrate, and proceeds from heme and tyrosyl radical intermediates formed during the POX and COX reactions, respectively (3,156). However, the specific structural changes in the holoenzyme that lead to suicide inactivation remain unknown. Mevkh et al. noted that COX self inactivation is accompanied by dramatic changes in protein structure as is manifested by the increased susceptibility of the inactive enzyme to trypsin cleavage and the increased number of exposed histidine residues subject to chemical covalent modification (162,163). POX and COX suicide inactivation have only been observed in vitra and it is still not clear whether the COXs can undergo self-inactivation in viva. Suicide inactivation could indeed serve as an additional control mechanism for regulating prostanoid synthesis in viva. In this regard it would be interesting to investigate whether the inactive forms of COX-1 and -2 are more susceptible to degradation compared to the native forms. It is well established that misfolded or structurally damaged proteins present in the ER can be targeted for degradation by a process known as ER-associated degradation (164-166). It is possible that COX-2 might selectively undergo suicide inactivation at endogenous hydroperoxide or AA levels that would be insufficient to inactivate COX-1. If indeed the inactive protein gets degraded, this could explain the short-lived nature of COX-2. Chapter IV of my thesis explores inactivation-induced COX-2 protein turnover as a potential mechanism for regulating COX-2 protein levels in viva. Proteolytic Degradation of Intracellular Proteins Lysosomal hydrolysis and ubiquitin-proteasome degradation are the two major pathways for the degradation of intracellular proteins. Lysosomal protein degradation is a 33 non-selective process that is catalyzed by cysteine hydrolases called cathepsins which function optimally at the low acidic pH of the lysosome (167). In addition to digesting exogenous proteins that have been internalized through phagocytosis or receptor- mediated endocytosis, lysosomes are also known to engulf and indiscriminately degrade cytosolic and organelle proteins in bulk (168,169). This process, known as autophagy, is usually activated for the clearance of cytosolic protein aggregates and damaged organelles (168). Autophagy is also induced by cell starvation and can be inhibited by the mTOR (mammalian target of rapamycin)/phosphatidylinositol-3 kinase signaling pathway (170). The selective degradation of intracellular proteins is largely non-lysosomal and appears to be mediated mainly by the ubiquitin-proteasome pathway. The 268 proteasome is a ~2 MDa cytosolic and nuclear multi-protein complex that catalyzes the selective degradation of three classes of protein substrates: a) normal short-lived proteins; b) structurally defective proteins; and c) misfolded proteins (171). The 26S proteasome consists of a barrel shaped, channel-like 2OS proteolytic core that is capped on each end with a 198 ATPase subunit (172). The proteolytic core comprises three known catalytic activities, namely trypsin-like, chymotrypsin-like, and peptidyl-glutarnyl activities (173). For most proteasome substrates the prerequisite for degradation is polyubiquitination, a process that involves the sequential attachment of molecules of the highly conserved 76-amino acid ubiquitin to the e-amino group of one or more internal lysine residues of the protein to form a long polymer of ubiquitin (174). Conjugation of the protein substrate with a polyubiquitin tag is catalyzed by three different enzymes (Fig. 8) (174,175). Firstly, the carboxyl end of ubiquitin is conjugated to a thiol group at the 34 active site of a ubiquiting activating enzyme (E1) in an ATP-dependent manner. Formation of the thioester linkage will activate ubiquitin for transfer to an active site cysteine residue of ubiquitin conjugating enzyme (E2). Finally, ubiquitin protein ligase (E3) will specifically recognize the protein substrate and aid in the transfer of the ubiquitin moiety to the amide side chain of an exposed lysine residue. The successive addition of ubiquitin molecules to one another will occur in a similar three-step reaction where the carboxyl end of one ubiquitin molecule is conjugated to a specific lysine (Lys- 48) of the previously conjugated ubiquitin. The 19S subunit of the 26S proteasome will specifically recognize the polyubiquitin tag on the substrate (174,175). Thereafter, the polyubiquitin tag will be removed by deubiquitinating enzymes associated with the proteasome (176). Meanwhile, the 19S subunit will employ its ATP-dependent chaperone-like activity to unfold the protein and deliver it through the narrow pore of the 208 core for proteolytic cleavage (177). Some normally short-lived proteins like omithine decarboxylase and cyclin D1 are known to undergo ubiquitin-independent proteasome degradation (178,179). These proteins become targeted for proteasome destruction upon conjugation with the molecule antizyme which has been shown to facilitate recognition of the substrate by the 19S subunit (180,181). Proteolytic Degradation of Endoplasmic Reticulum-associated Proteins It has long been known that most substrates for the 26S proteasome complex are present in the nucleus and cytosol. However, evidence has been accumulating within the last decade to show that the 26S proteasome has a pivotal role in ER quality control involving the elimination of misfolded, structurally damaged, and unassembled proteins that are 35 generated in the ER (l64-166,182-186). During the synthesis of membrane and secretory proteins in the ER, these proteins undergo various co-translational modifications, such as N-glycosylation and disulfide bond formation, that facilitate proper folding. The ER possesses a surveillance or quality control system that ensures that newly synthesized proteins are properly folded, N-glycosylated, and correctly assembled prior to exiting into the secretory pathway (186). Since the native state of a protein is considered to possess its lowest free energy (187), the surveillance system may selectively identify proteins with non-native structure based on their lower thermodynamic stabilities. Non-native proteins will be retained in the ER until they have achieved their native state. Otherwise, those proteins that are irreversibly defective will be unfolded and exported to the cytosol where they are degraded by the 26S proteasome. The pathway for the proteasomal degradation of ER-associated proteins has been termed ER-associated degradation (ERAD) (l64- 166,182-186). It has been recently proposed that the topology of the ERAD substrate relative to the ER membrane and the subcellular location of its misfolded or defective lesion will determine the manner by which the protein is selected and targeted for cytosolic proteasomal degradation (188-190). ERAD glycoprotein substrates that are wholly luminal or that are membrane-bound with substantially large luminal domains will be selected for degradation in the ER lumen (189,190). The key mediators of substrate selection in the ER lumen include the ER-resident molecular chaperone Hsp70 heavy chain binding protein (BiP), the ER-resident lectin binding chaperone calnexin and its homologue calreticulin, and ER-resident oxidoreductases such as ERp57 and protein 36 Figure 8. Mechanism for the polyubiquitination of proteins prior to their degradation by the 26S proteasome. Ubiquitin is activated for conjugation by E1 in an ATP-dependent manner. Activated ubiquitin is then transferred to the ubiquitin conjugating enyzme E2. After selecting the protein substrate, E3 ligase in conjunction with E2 will catalyze ubiquitin transfer to an exposed lysine residue on the protein. Polyubiquitination results due to the successive addition of ubiquitin molecules to one another where the carboxyl end of one ubiquitin molecule is conjugated to a specific lysine (Lys-48) of the previously conjugated ubiquitin. disulfide isomerase (PDI) (164-166,l83,186,190). Mueller et al. have recently demonstrated that SELlL, a mammalian homologue of the yeast Hrdlp/Hrd3p E3 ligase complex component Hrd3p is essential of the cytomegalovirus—mediated removal of MHC class I heavy chains from the ER lumen (191). Their observations suggest that SELlL may be a component of the ER luminal surveillance system and would therefore 37 have an important role in the recognition of lurrrinal ERAD glycoprotein substrates. The putative role of SELlL in glycoprotein ERAD is consistent with its topology and that of its yeast homologue Hrd3p; both are ER membrane proteins with relatively large luminal domains (191). ER-resident chaperones will retain improperly folded glycoproteins in the ER and assist them to fold properly. If the glycoprotein cannot be folded into its native conformation, or if its native structure has been disrupted, it becomes selected for ERAD (Fig. 9). The process is thought to begin with the recognition of exposed hydrophobic sequences on the glycoprotein by BiP and other molecular chaperones. These hydrophobic sequences are more likely to be exposed if the protein is unfolded, misfolded, or if its native structure has been compromised. By binding to these hydrophobic regions BiP prevents the aggregation of the misfolded protein in the ER (192). A specific N-linked glycan on the surface of the misfolded protein is processed to an oligosaccharide containing two molecules of N-acetyl glucosamine, nine mannoses, and one glucose molecule (GlclMangGlcNAC2) by glucosidase I and glucosidase II. This monoglucosylated oligosaccharide is specifically recognized by the membrane-bound lectin calnexin and its soluble ER luminal homologue calreticulin. Upon binding, these lectins in conjuction with BiP and the oxidoreductase ERp57 will assist in the productive folding of the glycoprotein (193-195). If the protein becomes properly folded, cleavage of the terminal glucose of the N-linked oligosaccharide by glucosidase U will release the bound calnexin or calreticulin. If the protein is not properly folded, MangGlcNAC2 will be reglucosylated by UDP-glucose glycosyltransferase (UGGT) so that the improperly folded protein is reentered into the calnexin/calreticulin cycle. UGGT is a folding sensor that will preferentially glucosylate ER proteins that have solvent-exposed hydrophobic 38 sequences (196,197). Repeated alterations of deglucosylation and reglucosylation will retain the glycoprotein in the calnexin/calreticulin cycle. If the glycoprotein spends significant time in this folding cycle, it will be considered to be terminally defective. In this case, the 0t1,2-linked terminal mannose residue of the N-linked oligosaccharide is irreversibly cleaved by ER 0L1,2 mannosidase I to form MangGlcNAC2 (198-200). A Mans-binding lectin such as ER-degradation mannosidase I-like protein (EDEM) or Yos9p will bind MangGlcNAC2 and facilitate the delivery of the terminally defective glycoprotein to the export machinery on the ER membrane (201-205). At this juncture, the ERAD substrate will have been unfolded by the combined action of PDI, which breaks disulfide bonds, and other chaperones in order to be exported as an extended polypeptide chain (206-209). The molecular mechanism for the vectorial export or retrotranslocation of the ERAD glycoprotein substrate across the ER membrane is yet to be clarified. The export process requires energy and there is some evidence to indicate that the Sec6l translocon, which is involved in ER protein import, may also be involved in protein export (206,210- 213). The recent X-ray crystal structure of the archeal SecY complex (214), a homolog of the eukaryotic Sec6l translocon, shows that the protein-conducting channel can only allow the passage of a molecule with a maximum diameter of 10-12 A (215). This pore size of the translocon is barely large enough to permit the retrotranslocation of ER polypeptides with their bulky N—glycan groups intact. However, there is evidence to suggest that the Sec6l protein-conducting channel may be very flexible (216). Indeed, while mediating cotranslational protein translocation the translocon pore size has been measured to be about 40-60 A in diameter (217). Recently, a mammalian ER multi- 39 spanning membrane protein called Derlin-l was identified that shows weak resemblance to the Derlp, a yeast transmembrane protein required for the degradation of yeast ERAD luminal substrates. Derlin-l is proposed to be the putative retrotranslocation channel in mammalian cells based on several studies showing its requirement for ERAD and its ability to simultaneously associate with lurrrinal ERAD substrates and accessory cytosolic components such as the p97(Cdc48)/del/Npl4 complex that have been shown to facilitate export or removal of the ERAD substrates from the ER (164,218-221). After the identification of Derlin-l two additional mammalian orthologues of Derlp, Derlin-2 and Derlin-3, were later identified during a microarray screen for ER-stress inducible genes (222). Derlin-2 and Derlin-3 exhibit ~75% homology and are ~30% identical to Derlin-l (222). In the same study overexpression of Derlins-2 or -3 facilitated the degradation of a misfolded mutant of the glycoprotein al-antitrypsin (222). While the mechanism of ERAD is yet to be clearly defined in mammalian cells, it has been extensively studied in yeast S. cerevisiae. Screening yeast genetic mutants that are defective in ERAD, has enabled the functional components involved to be easily characterized. By conducting homology database searches it was then possible to identify homologs or functional equivalents of these yeast ERAD components in mammalian cells. In both yeast and mammals the vectorial transport of the luminal ERAD substrate across the putative export channel is ATP-dependent and is believed to be facilitated by a cytosolic ATPase complex p97(Cdc48 in yeast)/de1/Npl4 that specifically recognizes and binds polyubiquitin chains (164,220,223). As the substrate appears in the cytosol it is polyubiquitinated by a series of membrane-bound ubiquitin conjugating enzymes such as ch6p (UBC6) and ch7p (UBC7), and membrane-bound E3 ubiquitin ligases, that 40 include DoalO, Hrdlp (HRD), and gp78 (l64,188,l89,221,224,225). The mammalian putative export channel Derlin-l can form membrane complexes with Hrdlp (226). p97/del/Npl4 is recruited to the membrane by interacting with Derlin-l through an accessory protein VIMP (p97-interacting membrane protein) (219). Recently, this ATPase complex has also been shown to interact with the E3 ligases gp78 (227) and Hrdlp (228) which may also contribute to its recruitment to the membrane. Membrane- associated p97/del/Npl4 will bind the nascent polyubiquitin chain on the protein and employ its ATPase activity to pull the polypeptide out of the ER (229). As the glycoprotein substrate appears on the cytosolic side of the membrane it is deglycosylated by peptidezN-glycanase (230,231). The deglycosylated substrate is then delivered to the 26S proteasome for degradation. ER membrane proteins with defective lesions on their cytoplasmic or membrane binding domains will be degraded by an ERAD pathway that is distinct from the one described above (188,190). Degradation of these substrates does not appear to involve recognition by the ER lunrinal surveillance system and is independent of their N- glycosylation status. Thus, the targeting of ER luminal proteins for proteasomal degradation has been designated ERAD-L to distinguish this ERAD pathway from ERAD-C and ERAD-M which degrade ER membrane proteins with defective cytosolic or transmembrane portions, respectively (188). A few mammalian ER membrane-associated proteins are known to be regulated by ERAD under physiological conditions. These include, HMG-CoA reductase, hepatic microsomal cytochrome p450 CYP3A4, and inositol 1,4,5-triphosphate (IP3) receptor (232-234). Even though all three membrane proteins have luminal portions, they are 41 likely to be degraded by an ERAD pathway other than ERAD-L since their turnover does not appear to require the ER lurrrinal quality control system. 1P3 receptors, which form channels that control Ca2+ release from the ER, are degraded in response to steady state elevated intracellular levels of IP3 (234). ERAD of 1P3 receptors has been shown to be mediated by ch7 and p97/de1/Npl4 (234,235). HMG-CoA reductase is a tightly regulated enzyme that catalyzes the rate-limiting step in cholesterol biosynthesis involving the conversion of 3-hydroxy-3-methylglutaryl CoA to mevalonate. It is a multi- spanning ER membrane protein with a large cytosolic catalytic domain. Mevalonate or sterols induce proteasomal degradation of HMG-CoA reductase by stimulating the binding of the membrane protein Insig-l or Insig-2 to the membrane domain of the reductase (236,237). Formation of the reductase-insig complex probably disrupts the native structure of the enzyme because it is immediately polyubiquitinated by gp78 and degraded by the proteasome (237-239). CYP3A4 undergoes substrate-induced suicide inactivation, a process that irreversibly damages the active site heme prosthetic group and makes the protein a substrate for ERAD (233). Just like the cytochrome p450 CYP3A4, it is possible that COX-2 may undergo irreversible substrate-induced suicide inactivation in viva leading to its clearance from the cell by ERAD. However, since COX-2 is an ER-resident luminal glycoprotein, its degradation would likely proceed through ERAD-L. 42 (MangGlcNAc2) Unfolding and refolding I K m _ Calreticulin (GlclMangGlcNAc2) GII ERp57 Properly folding glycovrotcin (MangGIcNAc2) 8.3.; Misfolded : [fig]. glycoprotein EDEP ( ° “‘3 “m” GI and an (MangGlcNAc2) i? ER Man I Terminally misfolded " glycoprotein Proteasomal degradation Figure 9a. Role of the calnexin/calreticulin cycle in ER quality control. N—glycosylation usually takes place co-translationally as the elongating polypeptide is translocated into the ER through the Sec6l translocon. Oligosaccharyltransferase (OST) transfers an intact Glc3Man9GlcNAc2 from membrane-associated dolichyl pyrophosphate to an Asn-X-Ser/Thr site on the polypeptide (where X is any amino acid except proline). As ER-resident molecular chaperones such as BiP assist the Asn (N)-linked glycoprotein to properly fold, glucosidases I and II (GI and GH) cleave off the two terminal glucose residues. If the glycoprotein is not properly folded it will be brought into the calnexin/calreticulin cycle via the binding of calnexin/calreticulin to the monoglucosylated N-glycan group of the protein. The heterodimer complex of calnexin/calreticulin and ERp57 will unfold and refold the protein. The properly folding glycoprotein will be released from the calnexin/calreticulin cycle by GII cleavage of the remaining glucose residue. If the glycoprotein is not properly folded it will be maintained in the cycle by the UGGT which reglucosylates the MangGlcNAc2 moiety. Misfolded glycoproteins that remain in the cycle for too long are eventually released through the action of (11,2 ER mannosidase I (ER Man I) which cleaves off the al.2-linked mannose to form MangGlcNAc2. This initiates ERAD of the terminally misfolded glycoprotein. The protein is unfolded with the help of BiP and PDI and exported out of the ER into the cytoplasm for proteasomal degradation. 43 0.00?.3300_02>0~ ”. 0.0.2»: 0.02.8. m .0 .> 052050.52?” 20:302.»: 20:032.»: 0/». e. be 00 00 H mm. ”nu.“ ”a. on C O O .u :03 u u I i _ COP—010.. 1:20.452 _|O|_IV |.|.IV .qude III..I1V— $20452 — IV — “50.3.2 Om... 0: - 2.0:. . 53.8302 D 0.0 _ a.» a." z. 0 m: / 0...... I 0.022" \ 3.0 Enid e... 500000.:m 0.. 233.83 0..m00300..3...30m .3 :8 EN .530: 33...:m .308: 333.3. 00380.. .....0 0.50030030130 0:30.88 8.. 2.3.83 088.: «€003.38: .m 3000.30.03 0:8 30.83.. 34003000338 0. :8 3033830 0m .30 m” 00 0.0073330Q.0Z>0~. >m8~ 23:08.. 0.. 3.0 0..m00000..m...30 0:8 .30 388.: 3. 0..m00000.8Q.8:m..0800 AOmd. .. .0 9.000803 8 : .30:0m.:000<.083 was: AQ.0_7\.0300.0Z>0~. 3. 3.0 0033300 0.. E88888 . 3:3 .. Am. 0:: DE. 33... 0:00.00 .30 «750.088.: 8 .00 03883 .38 .30 8.300.380.8305? 8.0.0. .8308. 0.. 3.0 833.30. £30000 80.9.0 3. 0.. 8.03000 3.0 £80388? #0.: 3.0 03.39.380.885; 8.0.0. W0-m.:0omva.330: 3. CUw-m.:0000 «80035808800 Adam... 33.38.:0 3.0 388.: .3 3.0 8.0.0. .m .30 m.<00.3.08.: .0 833.30.? 3.08.30... mw 333833000 . 933: 5 0.0300 3.0 8.43.30. 0.. .N :38: 833.33. 3333000 80.330. .230 08: 23.0.. .m 3.3.0.83 3. 58:03.30 A53 .0 3.03m... 8 30 .30 00:33.83 08.: :8. .33080 mid 0.. .30 3080.78 m.v.00.3.08.:. 44 Table 1. Participants of glycoprotein ERAD-L in yeast and mammalian cells Yeast Lamina! Kar2p Cuelp UGGT (11,2 Man I PDI Htmlp Yos9p Membrane Hrd3p Sec61p Derlp ch6p and ch7p Hrdlp Doa10 Cuelp 99 ee Cytosolic Cdc48 del N pl4 Mammals BiP Calnexin/ Calreticulin/ERp57 UGGT (11,2 Man I PDI EDEMS (EDEM 1,2, and 3) 08-9? SELlL Sec61p Derlins (Derlin l, 2, and 3) UBC6 and UBC7 gp78 and HRD 99 I. ?? VIMP p97 de1 Npl4 peptidezN-glycanase Role prevents aggregation of ERAD substrates glycoprotein folding detects misfolded proteins and maintains them in the calnexin/calreticulin cycle initiates ERAD by irreversibly releasing terminally misfolded glycoproteins from the calnexin/calretican cycle unfolds the terminally misfolded glycoprotein in preparation for export to cytosol escorts the ERAD substrate to the export/retrotranslocation channel similar function as Htmlp/EDEM role not clear; both have a large luminal domain that may be involved in selective recognition of ERAD substrates retrotranslocation retrotranslocation E2 ubuquitin conjugating enzymes E3 ubiquitin ligase E3 ubiquitin ligase anchors ch7p to the membrane recniits p97 to the membrane facilitates retrotranlocation Cdc48/p97 cofactor Cdc48lp97 cofactor deglycosylates ERAD substrates prior to their proteolytic cleavage 45 References 206,207 193-195 196,197 198-200 209,210 203-205 201,202 188,191 206, 210-213 218,219, 222 164,189 164,188, 189,224, 225 164,188, 189 189 219 164,188, 218-221 223 223 230,231 CHAPTER II THE 19 AMINO ACID CASSETTE OF CYCLOOXYGENASE-2 MEDIATES ENTRY OF THE PROTEIN INTO THE ER-ASSOCIATED DEGRADATION SYSTEM Summary Cyclooxygenase (COX) isoforms catalyze the committed step in prostaglandin biosynthesis. The primary structures of COX-1 and COX-2 are very similar except that COX-2 has a 19-amino acid (19-aa) segment of unknown function located just inside its C-terminus. Here we provide evidence that the major role of the 19-aa cassette is to mediate entry of COX—2 into the ER-associated degradation system that transports ER proteins to the cytoplasm. COX-1 is constitutively expressed in many cells, whereas COX-2 is expressed inducibly and transiently. In murine NIH/3T3 fibroblasts COX-2 protein is degraded by a proteasome-dependent process with a half-life (ti/2) of about 2 h whereas COX—1 is reasonably stable (t1/2> 12 h). Similarly, COX-l expressed heterologously in HEK293 cells is quite stable (II/2 >24 h) while COX-2 expressed heterologously is degraded with a t1/2 of ~5 h and its degradation is blocked by proteasome inhibitors. A deletion mutant of COX-2 was prepared lacking 18 residues of the l9-aa cassette. This mutant retains native COX-2 enzymatic activity but, unlike native COX-2, is stable in HEK293 cells (mp 24 h). Conversely, inserting the COX-2 cassette near the C-terminus of COX-1 yields a mutant ins594-612 COX-1 that is unstable (tm ~3 h). Mutation of Asn-594, an N-glycosylation site at the beginning of the l9-aa cassette, stabilizes both COX-2 and ins594-612 COX-1; nonetheless, COX mutants that have the Asn-594 consensus glycosylation site but lack the remainder of the 19-aa cassette (ie. 46 de1597-612 COX-2 and ins594-596 COX-l) are stable. Thus, although the Asn-594 glycosylation site is necessary for COX-2 degradation, at least part of the remainder of the 19-aa insert is also required. Finally, kifunensine, a mannosidase inhibitor that can block entry of ER proteins into the ER-associated degradation system, retards COX-2 degradation. Introduction Prostanoids are an important class of lipid mediators that are synthesized in almost all mammalian tissues. Prostanoids act in an autocrine and paracrine fashion through G protein-coupled receptors to elicit a variety of physiological and pathological responses (1). The committed step in the prostanoid synthesis pathway is catalyzed by the cyclooxygenase isozymes, COX-1 and COX-2 (1,3,7,17). Within a species, COX-1 and COX-2 exhibit ~60% amino acid sequence identity. Both isoforms are membrane-bound, ER-resident, heme-containing glycoproteins that function as homodimers (l,3,7,17,150). Structural and biochemical studies have shown that COX-1 and COX-2 are unusual integral membrane proteins that associate monotopically with the luminal face of the ER membrane and the contiguous inner membrane of the nuclear envelope (11,12,15,l6,151). Despite their close similarities in structure, catalytic function, and subcellular localization, COX-1 and COX-2 differ remarkably in their profiles of expression. COX-l is constitutively expressed in resting cells of many tissues in the absence of COX-2 ( 1,20). Inducible expression of COX-2, which is usually short-lived, occurs in fibroblast, epithelial, endothelial, macrophage, and smooth muscle cells in response to growth 47 factors, cytokines, and proinflammatory stimuli and expression is usually transient ( 1,20— 24). Differences in the patterns of expression of COX-1 and COX-2 genes have been quite clearly delineated in serum-stimulated NIH/3T3 fibroblasts (22,124,240). COX-1 mRNA and protein levels remain unchanged before and after serum stimulation of quiescent NIH/3T3 cells. In contrast, COX-2 mRNA and protein are barely detectable prior to stimulation, become significantly up-regulated shortly after stimulation and then rapidly decline to basal levels. It is well established that the different profiles of COX-1 and COX-2 expression as observed in serum-stimulated NIH/3T3 fibroblasts are partly attributable to differences in regulation of the isoforms at the transcriptional and post- transcriptional levels (l,20,240,241). It is still not clear the degree to which COX-1 and COX-2 may also be regulated differently at the post translational level. However, rapid degradation of COX-2 protein may serve a significant physiological role in regulating the levels of prostanoids whose syntheses occur via this isoform. Previous studies have indicated that COX-2 can be ubiquitinated and degraded by the 26S proteasome in the cytoplasm (148,149,242), suggesting that COX-2 degradation can involve exit from the ER via ER-associated degradation (ERAD) system(s) followed by proteolysis by the proteasome (165,183,206,243,244). In the present study, we establish that COX-2 protein is preferentially and rapidly degraded in NIH/3T3 and HEK293 cells under conditions in which COX-1 is very stable. We also confirm that even though COX-2 is an ER-resident enzyme its degradation is proteasome-dependent. In investigating the molecular basis for the different rates of COX-1 and COX-2 degradation, we found that the 19-amino acid segment unique to COX-2 (Asn594-Lys612) and located six residues in from the carboxyl terminal end 48 targets the protein for rapid degradation through the ER-associated degradation (ERAD) pathway (5) (164-166). Experimental Procedures . Materials. Dulbecco’s modified Eagle medium (DMEM), fetal bovine serum (FBS), ponasterone A, tetracycline, and goat serum were from Invitrogen/Gibco. Bovine calf serum was from Hyclone. Cycloheximide, puromycin, and bacterial lipopolysaccharide (LPS) were obtained from Sigma-Aldrich. Kifunensine, M0132, epoxomicin, E64, and leupeptin were purchased from Calbiochem. Endoglycosidase H (Endo H) was purchased from Roche. Unlabeled arachidonic acid was purchased from Cayman Chemicals, while radioactive l4C-arachidonic acid and l4C-eicosapentaenoic acid were from American Radiolabeled Chemicals. Construction of Pla_smids for Transfection. Recombinant ovine (ov) COX-1 cDNA and human (hu) COX-2 cDNA were subcloned into pIND (Invitrogen) and pcDNAS/FRT/TO (Invitrogen), respectively. pIND is ecdysone-inducible while pcDNAS/FRT/TO is tetracycline- inducible. After subcloning, the Quick-ChangeTM site- directed mutagenesis kit (Stratagene) was used to create the following C-terminal mutants: N594A huCOX-Z, delS95-612 huCOX-2, de1597-612 huCOX-2, and insS94- 596 ovCOX-l. The COX-1 insertion mutants, insS94-6l2 ovCOX-l, insS94-612(N 594A) ovCOX-l, and insS94-612 ovCOX-l were created from the cDNA template for native ovCOX-l by overlap extension PCR then subcloned into pIND using HindIII restriction sites. Correct cDNA orientation and mutations were confirmed by sequencing. The primers and the PCR conditions used to design the above mutants are shown in the ‘Appendix’ section. 49 Cell Culture and Transfection. NIH/3T3 fibroblasts at early passage (<6 passages) were cultured in DMEM supplemented with 10% bovine calf serum and 100 u/ml penicillin/streptomycin. To induce COX-2 expression, the cells were first made quiescent by serum starvation for 48 h in DMEM containing 0.2% bovine calf serum and thereafter treated with DMEM supplemented with 20% FBS for 4 h. RAW 264.7 macrophage-like cells were cultured in DMEM supplemented with 10% FBS and 100 u/ml penicillin/streptomycin. To stimulate COX-2 expression, the cells were challenged with 200 ng/ml LPS. HEK293-derived cell lines stably expressing native or mutant COX constructs were generated using either the tetracycline-inducible and ecdysone-inducible mammalian expression systems (Invitrogen) according to the manufacturer’s protocol. Constructs that were expressed under the control of a tetracycline-inducible promoter were native huCOX-2, delS95-612 huCOX-2, N594A huCOX-2, and delS97-612 huCOX-2; those expressed under the control of the ecdysone-inducible promoter were native ovCOX-l, ins594-612 ovCOX-l, insS94-612(N594A) ovCOX-l, and insS94-596 ovCOX-l. Stably transfected HEK293 cells were cultured in DMEM supplemented with 10% FBS, 100 u/ml penicillin/streptomycin, and the appropriate pharmacological selective reagents. Inducible expression was achieved by a 24 h serum starvation in serum-free medium followed by treatment with 10 ug/ml tetracycline or 10 M ponasterone A (ecdysone analog) for the appropriate times in normal culture medium. Protein Degradation and Drug Treatments. A cycloheximide (CI-IX) decay experiment was performed to measure the protein stabilities of COX-1 and COX-2 in serum-treated NIH/3T3 cells. Briefly, quiescent cells were serum-stimulated for 4 h then 50 incubated for different times with 50 M CHX in DMEM supplemented with 10% FBS. The protein degradation experiment was performed in the absence or presence of MG132 (20 11M and 50 11M), epoxomicin (3 M and 5 uM), 25 MM leupeptin, and 25 M E64. HEK293 cells inducibly expressing wild-type or mutant cyclooxygenase constructs were grown to 80% confluency, serum-starved for 24 h, and then treated with 10 rig/ml tetracycline or 10 M ponasterone A in complete culture medium to induce expression. Afterwards, the cells were incubated for various times with 50 MM puromycin to block translation in the absence or presence of 20 MM MG132, 25 M KIF, or 50 11M KIF. Envzmatic Deglvcosvlation. For complete deglycosylation, HEK293 whole cell lysates were denatured by boiling in NuPAGE SDS sample loading buffer (Invitrogen) and then treated for at least 12 h with endoglycosidase H (Endo H) at a concentration of 0.4 mU/ul. Western Transfer Blottig. After the appropriate treatments, NIH/3T3, RAW 264.7, and HEK293 cells were scraped into ice-cold PBS (phosphate-buffered saline), pH 7.4 containing 5 mM EDTA and a cocktail of protease inhibitors (Roche) and lysed by sonication. RIPA lysis buffer (10 mM Tris, pH 7.2, 150 mM NaCl, SmM EDTA, 0.1% SDS, 1.0% Triton X-100, 1% deoxycholate) containing a protease inhibitor cocktail (Roche Applied Science) was also used for cell lysis. Protein concentrations were determined using the BCA protein assay kit (Pierce). The NuPAGE system (Invitrogen) was used to resolve the proteins in the whole cell lysates on a 7% tris-acetate polyacrylamide gel. After transfer to a nitrocellulose membrane, immunoblotting was performed with the appropriate primary antibody. Horseradish peroxidase-conjugated 51 anti-rabbit or anti-mouse IgG antibodies (Bio-Rad) were used as secondary antibodies. Immunodetection was performed using the Western Lighting Chemilurninescent kit (Amersham Biosciences) followed by exposure to X-ray film. Densitometry analysis was performed using ImageQuant TL software (Amersham Biosciences). Antibodies Specific for COX-1 or COX-2. A previously generated (13), peptide- specific, polyclonal primary antibody for murine (mu) COX—2 against the epitope Ser598-Ly3612 was used in the current study to detect muCOX-2 by immunoblotting. A polyclonal antibody raised against whole recombinant ovCOX-l was used to detect muCOX-l. Peptide-specific, polyclonal primary antibodies for ovCOX-l and huCOX-2 were synthesized by Covance Research Products against the following epitopes: Leu272- Gln283 of ovCOX-l and ProS83-Asn594 of huCOX—2. Immunocflofluorescence. Stably transfected HEK293 cells were grown on poly L-lysine-coated cover slips and induced with either 10 ug/ml tetracycline or 10 M ponasterone A for the appropriate times. After fixation in 3.7% formaldehyde and perrneabilization with 0.2% Triton X-100, the cover slips were blocked in 0.1% Triton- X-100 solution supplemented with 10% goat serum. Thereafter, the cover slips were incubated with the appropriate primary antibody against either ovCOX—l or huCOX-2, extensively washed with PBS, and incubated with Alexa Fluor 488 fluorophore- conjugated secondary antibody (Invitrogen). After three additional extensive washes with PBS the cover slips were dried and mounted onto glass slides using Prolong antifade kit (Invitrogen). Microscopy was conducted on a Zeiss Atto Arc® 2 HBO 100W fluorescence microscope at a magnification of 200X. 52 Results Rapid, Protea_some—dependent Dggra_d§tion of cox-g. COX-1 is expressed constitutively in murine NIH/3T3 fibroblasts, whereas COX-2 is expressed inducibly and transiently(22,124,240). This raises the possibility that the protein stabilities of the COX isoforms are different with COX-2 being more susceptible to degradation. A cycloheximide (CHX) decay experiment in NIH/3T3 cells was performed to compare the stabilities of COX-1 and COX-2 (Figs. 10a and b). COX-2 was degraded with a short half-life (11/2) of ~2 h while COX-l was more stable and did not appear to undergo degradation during the experiment (111/2 >12 h). Although COX-2 expression is typically transient, a few tissues and cell types are reported to express COX-2 in a prolonged and/or constitutive manner (125,127,242,245,246). It is possible that COX-2 protein is stable under these conditions. To address this, we examined COX-2 protein degradation in LPS-stimulated murine RAW264.7 macrophage cells where COX-2 protein expression continues to increase up to 24 h after LPS treatment. COX-2 was also degraded rapidly (t.,2~2 h) in LPS- challenged RAW 264.7 cells (data not shown) indicating that increased COX-2 stability is not responsible for prolonged expression of the enzyme at least in this cell type. We performed experiments with several classes of protease inhibitors to determine the pathway responsible for the selective degradation of COX-2 in NIH/3T3 cells. The selective 26S proteasome inhibitors, epoxomicin and M6132 (Figs. 10c and d), significantly slowed COX-2 protein degradation at 4 and 8 h after CHX treatment 53 10A. 0 1 2 4 6 12 Timewith CHX(h) cox-2 5-3 in... .-.... .. e ,. cox-1 F‘Hflfifi Actin w- “A an; L was wad §§§ f—O—W“ A" fic'oxa 5 j_.fl 1cox-2j % Protein Remaining s s 8 \ E‘F- Time after CHX (h) No protease inhibitor 5 uM Epox 25 pM Leup _0 2 4 8 2 4 8 2 4 8 TimewithCHX(h) 54 10C' N o protease (cont’d) inhibitor 50 “M MG132 3 pM Epox . 0_ 2 .11.. 8 2 4 >8 2_ 74 8 TimewithCHX(h) I-Ii and un- « ~ ‘c-fl u-d' -~‘ a—a u—a «it Actin W m m D. _ N O E —0 - CHX only +cux + M6132 \L\ “ i-——§ % Protein Remaining s 0 2 4 6 8 10 Time with CHX (b) Figure 10. Cyclooxygenase protein degradation in serum-treated NIH/3T3 cells. A, Quiescent 3T3 cells were stimulated with 20% FBS for 4 h to induce COX-2 expression. Cycloheximide (CHX; 50 [2M final concentration) was added to the medium and the cells were incubated for the indicated times. Cell lysates were then prepared and analyzed by Western blotting for COX-1, COX-2 and actin as described in the Methods section. B, Densitometry was performed to quantify the levels of COX-1 and COX—2 which were normalized to those of actin. Error bars denote +/- SE of the mean for three independent experiments. C, COX—2 expression was induced in murine NIH/3T3 cells and 50 11M CHX added to the medium as described in A. The cells were then treated for the indicated times with or without 3 11M or 5 uM epoxomicin (Epox), 25 “M leupeptin (Leup) or 50 uM MG132. COX-2 protein levels were examined by Western blotting and densitometry. D, Quantitative analysis of the effect of 20 11M MG132 on COX-2 protein stability in 3T3 cells. Densitometry values for each time point were from six independent experiments performed as described above and represent the mean +/- SE. Asterisks denote that a p value < 0.05 based on a Student’s paired t-Test. 55 while two different cysteine protease inhibitors of lysosomal degradation, leupeptin (Fig. 100) and E64 (data not shown), did not. At the 2-h time point in six independent experiments with 20 uM M0132, there was a trend but not a statistically significant inhibition (Fig. 10d). Accordingly, it is not clear whether a protease other than the 26S proteasome can also be involved in COX-2 degradation. It should be noted that both 26S proteasome inhibitors prevented the degradation of the two alternatively glycosylated (ie. 72 and 74 kDa) forms of COX-2 (Figs. 10c and d). Overall, our findings and those of others (148,149,242) indicate that COX-2 can be degraded in the cytosol by the 26S proteasome. Because COX-2 is located on the luminal surface of the ER (13,15,16), our results imply that its degradation must involve transport across the ER membrane to the cytosol. The Unique C-terminal l9—amino Acid Segment is Involved in COX-2 Degradation. Despite the high degree of structural similarity between the COX isoforms, COX-2 is distinct from COX-1 in possessing a unique l9-amino acid '(19-aa) cassette that is located six residues in from the carboxyl-terminal end (Fig. 11a). The N-terminal most residue of the 19-aa insert is Asn-594, which is one of four functional N-glycosylation sites of COX-2. COX-l has the other three N—glycosylation sites in common with COX-2 We used site-directed mutagenesis to test the hypothesis that the unique C-terminal 19-aa of COX-2 affects its degradation. HEK293 cells, which do not have detectable levels of COX-1 or COX-2, were used to stably and inducibly express native or C-terminal mutant versions of the cyclooxygenase enzymes (Fig. 11b). Native human (hu) COX-2 inducibly expressed in HEK293 cells was degraded with a t”; of ~5 h while native ovine (ov) 56 11A. Asn68 Asn l 44 1% l L | Signal | EGF ] MBD lGlobular-Catalytic l Asn67 Asn l 44 Asn410 Asn4l0 RPPTEL C00“ COX-l Am594 I ‘ 1 ERS'rELC00H I Signal j EGF I MBD lGlobular-Catalytic - COX-2 B. 5‘14 -(~ 1 3 53°FSVPDPELIK'I'VTI“hisSSIRSGLDDI.‘li'lP'.l.'VI-Iaml7{S'I‘EL618 Sa"FSVPDPELIKTVTIN EIRS‘I‘EL600 S“IE'SVIE’DPELIKIV‘IT ‘ .. iEL“8 58"FSVPDPELIK‘I V I INKS ---------------- ERSTEL‘SOZ 58°FHVPDPRQEDR“CV/E RPPI‘EL600 Sa”FI'IVPDPRQEDRPGVMAS8SRSGIJJDIN'P'I'VIIZJLKERPP’I‘EL519 580IE‘HVPDPRQEIJR. v . "“1VLLKERPPTEL‘19 58°FHVPDPRQEDRPGVENAS ---------------- RPP‘TEL603 native huCOX—Z de1595-612 huCOX—2 N594A huCOX—Z delS97—612 huCOX—2 native ovCOX—l insS94—612 ovCOX-l in5594—612(N594A) ovCOX—l insS94-596 ovCOX—l C. native ovCOX—l 0 4 s 12 24 (h) AAA—ya Actin ‘3'! Illlqlla Clint—II 11,, > 24 h delS95-612 huCOX-2 0 4 3 12 24 (h) lira-nuflhiliuu-y Actin mac-vac. t],2 > 24 h 57 native huCOX-2 0 2 4 s 24 (h) r“ - d“. W 1 F ins594-612 ovCOX-l 0 2 4 8 24 (h) ~a‘ fine-bud 1 ID' native huCOX—2 No protease inhibitor 20 1.1M MG132 0 4 812 24 4 8 12 24 (h) ‘ manure: . u. - . .‘r,"r --.-'. . .’ 3", r; ' “_ ”,1: ',‘~_‘~."-rfl" :1! at ”IV 'm¢ .9 r a. - , l M-‘ia‘JcflQ-l. ht. _ A. I‘“m‘dwl‘0.l'H-J‘MV §\l~- I. .' .'.I." «hHMI-N'fdf‘gfl-‘m'i‘lfl. 5-5. .a Vanna insS94-612 ovCOX-l No protease inhibitor 20 “M MG132 2 4 8 2 4 8 (h) "‘F'"_r‘-‘-T'TQT" ‘ " ”.Tm’ "'"— ' ' ~‘ " I "" ' ‘ '_“‘-"-‘ * r We“ 2”“ ”*5 E Fig. 11. Stability of heterologously expressed native and C-terminal cyclooxygenase mutants in HEK293 cells. A, Alignment of major folding domains of COX-1 and COX- 2 showing consensus N-glycosylation sites and the relative position of the unique 19- amino acid (l9-aa) insert located near the C-terminus of COX-2. Numbering for COX-l begins with the Met at the translation start site. Numbering for COX-2 parallels the COX- 1 numbering in which the start of the mature, processed COX-2 protein has the same number as the start of the mature, processed COX-1(12). B, Amino acid sequences of the C-termini of cyclooxygenase constructs that were stably transfected into HEK293 cells. The underlined sequence is the consensus N-glycosylation site at the start of the unique COX-2 l9-aa cassette. Proteins that were expressed under the control of a tetracycline- inducible promoter were native huCOX-Z, delS95-612 huCOX-2, N594A huCOX-2, and delS97-612 huCOX-2; those expressed under the control of the ecdysone-inducible promoter were native ovCOX-l, ins594-612 ovCOX-l, ins594-612(N594A) ovCOX-l, and insS94-596 ovCOX-l. C, HEK293 cell lines stably and inducibly expressing the constructs shown in B were grown to ~80% confluency, subjected to serum starvation for 24 h, and then treated with the appropriate inducing agent (10 ug/ml tetracycline or 10 “M ponasterone A) for 24 h (native huCOX-2, delS95-612 huCOX-Z, and insS94-612 ovCOX-l) or 12 h (native ovCOX-l). Puromycin (50 uM) was then added to block translation and protein levels were analyzed at different times by Western blotting. Densitometry was performed as described in the legend to Fig. l. The Western blotting results are representative of at least three separate experiments from which protein half- life measurements were made. D, Following serum-starvation and COX protein induction of HEK293 cells expressing huCOX-2 and ins594-612 ovCOX-l as described in C, 50 11M puromycin was added to inhibit translation in the presence or absence of 20 uM MG132. At the indicated times, cells were lysed and subjected to Western blotting. The results shown for each protein are representative of two independent experiments. 58 COX-1 was not degraded at a detectable rate (tug >24 h) (Fig. 11c). A deletion mutant delS95-612 huCOX-2 was prepared that lacks 18 amino acids of the 19-aa; additionally, Asn-594 cannot be glycosylated in this mutant as the consensus N-glycosylation is no longer present. This deletion mutant was stable with a t1}; comparable to that of native ovCOX-l (>24 h) (Fig. 11c). Inserting l9-aa of huCOX-2 near the C-terminus of ovCOX-l yielded a mutant ins594-6l2 ovCOX—l that had a relatively short “/2 of ~3 h (Fig. 11c). Degradation of both native COX-2 and ins594—612 ovCOX-l was retarded by treatment with the proteasome inhibitor M6132 (Fig. 11d). These observations suggest that the l9-aa segment of COX-2 is involved in the rapid degradation of this COX isoform, perhaps by targeting it for proteolysis by the 26S proteasome. The native and mutant proteins used in the studies depicted in Fig. 11 and in subsequent figures are inducibly expressed in HEK293 cells with different time courses. To the extent possible, we performed protein degradation experiments with HEK293 cells expressing comparable amounts of COX protein to achieve a more direct comparison of half-lives. The subcellular distribution of the native and mutant proteins heterologously expressed in HEK293 cells was determined by immunofluorescent staining. Both mutant enzymes exhibited a perinuclear and diffuse pattern characteristic of COX-1 and COX-2 (Fig. 12) (13,14,19); we also determined by Western blotting that delS95-6l2 huCOX-2 and insS94-612 ovCOX-l are present in a high speed, microsomal membrane fraction prepared from HEK293 cells (data not shown). Dr. Masayuki Wada measured and compared the Km and Vmx values of native murine (mu) COX-2 and delS95-612 muCOX-2 for oxygenation of arachidonic acid (Fig 13a). He also analyzed the products 59 formed from [1-14C] arachidonic acid and [1-14C] eicosapentanoic acid by both enzymes (Fig. 13b). The kinetic constants and product distributions were the same for the native and mutant enzymes. The specific cyclooxygenase activity of purified ins594-612 ovCOX-l was also comparable to that of the purified native ovCOX-l (38.2 and 40 pmol/min/mg, respectively; Dr. Rana Sidhu, unpublished results). A- (I) native huCOX-Z (11) delS95-612 huCOX—2 (I) native ovCOX-l (II) native ovCOX-l Fig. 12. Subcellular localization of native huCOX-Z, delS95-612 huCOX-Z, native ovCOX—l and ins594-612 ovCOX-l in HEK293 cells. HEK293 cells stably expressing the various COX variants were grown on poly L-lysine-coated cover slips and induced with either 10 ug/ml tetracycline or 10 uM ponasterone A as described in the legend to Fig. 11. The cells were then fixed with formaldehyde, permeabilized with Triton X-100 and incubated with an appropriate primary antibody. A, For huCOX-2 (I) and delS95-612 huCOX-2 (II) a rabbit anti-peptide antibody to residues 583—594 of huCOX-2 was used. B, For native ovCOX-l (I and II) and insS94-612 ovCOX-l (III) a rabbit anti-peptide antibody to residues 272-283 of ovCOX-l was used for (I) while an antibody against whole ovCOX-l was used for (11) and (111). After washing with 1X PBS the cells were incubated with fluorophore-conjugated anti-rabbit IgG secondary antibody, washed with 1X PBS, mounted using an anti-fade reagent, and examined by fluorescence microscopy at a magnification of 200X. 60 E‘ A. €53 35, .2 9 30‘ H 4 < a,25+ .2 E 20~ ’5 E ‘ Natlve muCOX-2 - _. . 0 E 15* Km=7.6 9"“ ‘ VM=32unltslmg (D o 10‘ X _ ‘ del595-612 muCOX-2 o ------- o O 0 5: Km=9.9iiM U E Vm=28 unltslmg 1 0 v ' l l I v r , 0 so 100 150 200 [Arachidonic Acid] (pM) B. E "i' fi'i' ” 0,! {:63 e >< 3* E at 3x 2 is es 7‘3 25" Se , a , '«EPA MS we. "i“ . “C-AA(20 iiM) _ ' "C-EPA (20 pM) . Figure 13. Kinetic properties of purified native muCOX-2 and delS95-612 muCOX- 2. A, Hexahistidine-tagged versions of muCOX-2 and delS95-612 muCOX-2 were expressed in Sf21 insect cells and purified. Oxygen electrode assays were performed to compare the specific cyclooxygenase activities of the wild type and mutant enzyme. B, Similar amounts of purified hexahistidine—tagged muCOX-2 or delS95—612 muCOX-2 (~23 mU) were incubated with 20 W [1-I4C]arachidonic acid (AA) or [1- l4C]eicosapentaenoic acid (EPA) for 30 sec. The products were extracted, separated by thin-layer chromatography, and visualized by autoradiography. The radioactive band between EPA and PGH3 has chromatographic properties of the PGH3 degradation product 17-hydroxy-5,12,15—heptadecatrienoic acid. The experiments in A and B were carried out by Dr. Masayuki Wada. 61 Collectively, these results suggest that effects on substrate turnover or subcellular localization do not account for the large difference in the rates of degradation between the native and mutant enzymes. Asn-594 Glycosylation Site is Necessary but not Sufficient to Effect COX-2 Degradation. A point mutation of the Asn-594 N—glycosylation site at the beginning of the l9-aa segment was sufficient to extend the [”2 of the mutated COX-2 (N594A huCOX-2) to >24 h (Fig. 14a). Consistent with this, ins594-6l2 ovCOX-l was also stabilized by mutating the Asn-594 glycosylation site (Fig. 14a). However, a deletion mutant delS97-612 huCOX-2 carrying the Asn-594 glycosylation site but lacking the rest of the l9-AA insert was as stable as delS95-612 huCOX-2 (Fig. 14b). Similarly, insertion of a consensus N-glycosylation site near the C-terminus of ovCOX-l (ins594—596 ovCOX-l) did not destabilize this COX isoform (Fig. 14b), although this mutant is at least partly glycosylated at Asn-594 (Fig. 15a, panel I). In evaluating the glycosylation status of Asn-594 in various forms of COX-1 and COX-2, we compared the molecular mass of ins594-612 ovCOX-l with that of ins594- 612(N594A) ovCOX-l and the mass of native huCOX-2 to N594A huCOX-2. The approximate mass of an N-linked oligosaccharide moiety is 2 kDa (151). Both native huCOX-2 and insS94-612 ovCOX-l would be expected to have a total of four N- glycosylation sites (151,247). As anticipated, the electrophoretic mobility of insS94-612 ovCOX-l was less than that of insS94-612(N594A) ovCOX-l by a difference corresponding to about 2 kDa (Fig. 15a, panel I); moreover, deglycosylation of ins594- 612 ovCOX-l and ins594-612(N594A) ovCOX—l by Endo H increased the mobilities of 62 N594A huCOX-2 ins594-612(N594A) ovCOX-l o241224(h) 0241224(h) . 4t .‘C'f' ’T".f first“ ' fig W““""”"i mun-inn!" Actin ”II-5 in» we at 'v ’ all. iii—vuln- A: "I .- t,,2 > 24 h t1,2 > 24 h B. delS97-612 huCOX-2 ins594-596 ovCOX-l 024824(h) 024122401) m _ 1"; A w in- if“ r’ Q Q; i a v- Actin “your“ “kw—e. “Mi“ and: .....~._. .31! t1/2>24h tm>24h Figure 14. Stability of cyclooxygenase mutants in HEK293 cells having various modifications of the 19 amino acid cassette. A and B, Amino acid sequences of the C- terrnini of cyclooxygenase constructs that were stably transfected in HEK293 cells are shown in Fig. 118. Proteins that were expressed under the control of the tetracycline- inducible promoter are N594A huCOX-2 and delS97-612 huCOX—2; those expressed under the control of the ecdysone-inducible promoter were ins594-612(N594A) ovCOX- l and ins594-596 ovCOX-l. Expression was induced for 12 h (N594A huCOX-2 and delS97-612 huCOX-2) or 24 h (ins594-612(N594A) ovCOX-l and ins594-596 ovCOX- 1). Thereafter, the time course of degradation of the proteins was determined as described in the legend to Fig. 11. The results shown in A are representative of at least three independent experiments from which protein half-life measurements were made. The results shown in B are representative of two independent experiments. 63 both mutants such that they had apparently identical molecular masses (Fig. 15a, panel 11). These results suggest that the difference in molecular mass between the two COX—l insertion mutants results from glycosylation of Asn-594. In contrast to the results obtained with the COX-1 mutants, we found no difference between the electrophoretic mobilities of native huCOX-2 and N594A huCOX-2 expressed in HEK293 cells (Fig. 15b, panel I). This is somewhat similar to what is seen with murine COX-2 expressed in cos-1 cells (151) where the major bands for native muCOX-2 and N594Q muCOX—2 have the same electrophoretic mobilities; however in the case of muCOX-2 expressed in cos-1 cells, a more slowly moving band was also observed. A second, less mobile band was observed with huCOX-2, but not N594A huCOX-2, when HEK293 cells were treated with KIF, an al,2 ER mannosidase I inhibitor (Fig. 15b, panel H). Moreover, Endo H treatment led to deglycosylated forms of native COX-2 and N594A COX-2 that had the same molecular mass (Fig. 15b, panel 11). In addition to causing the appearance of a second, alternatively glycosylated form of huCOX-2, KIF treatment of HEK293 cells expressing native huCOX-2 inhibits COX-2 degradation (Fig. 15b, panel II). Degradation of ins594-612 ovCOX-l is also inhibited by KIF (Fig. 15c). This is a diagnostic feature of N-glycosylated proteins whose degradation involves transport to the cytoplasm by the ERAD system (199,200,248—251). These results are consistent with those obtained with 26S proteasome inhibitors in indicating that COX—2 degradation involves entry into the ERAD system prior to degradation by the 26S proteasome. 64 15A. (0 l 2 3 4 l. native ovCOX—l 2. insS94-596 ovCOX-l 3. ins594-612 ovCOX-l 4. ins594-612 (N 594A) ovCOX-l (lo 4. I “12 :3" fl . w insS94-612 ovCOX—l insS94-612(N 594A) ovCOX-l Endo H cleavage of l Endo H cleavage of 2 95”.“? 1. native huCOX—2 2. N594A huCOX-Z (ll) native huCOX-z N594A huCOX-Z l 2 3 4 1 2 3 4 3 an H c- .- 1. tet-induced 2. tet-induced + 12 h KIF 3. Endo H cleavage of l 4. Endo H cleavage of 2 65 15C. 25 uM KIF 0 2 4 8 24 2 4 8 24 Time with puromycin (h) "'7‘?“ .42... a A: A A Actin ‘l‘?’ " 3" a “a “‘3 Wig-taut 1‘ Figure 15. Glycosylation of ins594-612 ovCOX-l and native huCOX—Z at Asn-594. A (I) Native ovCOX-l (1), ins594-596 ovCOX-l (2), insS94-612 ovCOX-l (3), or ins594- 612(N594A) ovCOX-l (4) expression was induced with 10 M ponasterone A in HEK293 cells for 24 h. (II) After protein induction of insS94-612 ovCOX-l and in3594- 612(N594A) ovCOX-l cell lysates were prepared and boiled in SDS sample loading buffer for 10 min and then treated with or without Endo H (0.4 mU/ul) for at least 12 h. Proteins were resolved by SDS-PAGE on a 7% tris-acetate polyacrylamide gel and subjected to Western blotting. B (I), native huCOX-2 (1) or N594A huCOX-2 (2) expression was induced with 10 ug/ml tetracycline in HEK293 cells for 24 h and 3 h, respectively. ( II ), after protein induction as in (I) cells were rinsed in PBS and incubated for an additional 12 h in normal culture medium with or without 50 M KIF. Thereafter, cell lysates were prepared and boiled in SDS sample loading buffer for 10 min and then treated with or without Endo H (0.4 mU/ul) for at least 12 h. Proteins were resolved and subjected to Western blotting as in A. Arrows indicate differences in molecular mass due to N-glycosylation. C. insS94-612 ovCOX-l was induced in HEK293 cells as described in the legend to Fig. 2. The cells were then incubated with puromycin (50 M) in the presence or absence of KIF (25 M) for the indicated times. Cell lysates were prepared and subjected to SDS-PAGE and Western blotting as described in the legend to Fig. 11. The results shown are representative of two independent experiments. 66 Discussion Substitution of the COX-1 coding region for COX-2 in transgenic mice partially rescues the typical phenotype of COX-2 null mice (252). Thus, a key difference between COX-1 and COX-2 is in their patterns of expression and not simply in their enzymatic properties. The difference in the profiles of COX-1 and COX-2 protein expression are at least partly attributable to differences in the regulation of transcription of the two genes (reviewed in (1,20,241)). In general, COX-1 gene expression is constitutive, and COX-2 expression is inducible. In addition, COX-2 mRNA with its repetitive AUUUA degradation sequences is subject to post-transcriptional control (145,253,254). In this report, we confirm and extend previous findings indicating that COX-l is a stable protein, whereas COX-2 is degraded relatively rapidly, and that COX-2 can be ubiquitinated and degraded by the 26S proteasome (148,149,242). COX-2 is localized to the luminal surface of the ER (13-16), and so to be degraded, it must first be transported to the cytosol from the ER. In the studies reported here, we have focused on the molecular basis for the relatively rapid rate of COX-2 degradation when compared with that of COX-1. Specifically, we evaluated the 19-aa cassette (Asn-594—Lys-612) that is unique to COX-2 for its potential role in degradation. This 19-aa insert is located 6 residues in from the COX-2 C terminus. It has only one recognizable consensus sequence, an N-glycosylation sequence involving Asn-594—Ser—596. We have demonstrated that the 19-aa cassette of COX-2, with an intact Asn-594 N-glycosylation site, targets COX-2 for proteasomal degradation. This is based on the following observations. a) A deletion mutant of huCOX-2 lacking 18 amino acids of the l9-aa insert (delS95—612 huCOX-2) and a 67 functional N—glycosylation site at Asn-594 has native COX activity and the same subcellular location as native enzyme but is refractory to protein degradation. b) Point mutation of the Asn-594 N-glycosylation site (N594A huCOX-2) also stabilizes COX-2. c) Inserting the COX-2 l9-aa cassette at the C terminus of ovCOX-l (insS94—612 ovCOX-1)destabilizes ovCOX-l. d) Point mutation of Asn-594 in insS94—612 ovCOX-l stabilizes this insertion mutant. e) The degradation of both native huCOX-2 and insS94— 612 ovCOX-l can be retarded by M6132, a 268 proteasome inhibitor. Although the Asn- 594 N-glycosylation site is required for degradation, we found evidence that all or part of the remainder of the l9-aa insert is also necessary. Our results suggest that glycosylation of Asn-594 of COX—2 is critical for initiating entry of the enzyme into the ERAD system for transport to the cytoplasm. The ERAD system is thought to involve unfolding of N-glycosylated proteins in the ER lumen and ATP-dependent retrograde transport across the ER membrane, and frequently, this is followed by ubiquitination of the transported protein by ubiquitin-prOtein isopeptide (E3) ligases associated with the cytosolic face of the ER membrane and subsequent degradation by the 26 S proteasome (165,183,206,243,244). The ERAD system is usually regarded as a quality control pathway for the clearance of misfolded, unassembled, or toxic proteins from the ER lumen (165,206,243,244,255). It has also been implicated in the degradation of native ER-associated proteins such as hydroxymethylglutaryl-CoA reductase, hepatic microsomal cytochrome P450 CYP3A4, and inositol 1,4,5-triphosphate receptor (232-234), all of which are ER transmembrane proteins. An ER luminal glyc0protein is selected for the ERAD pathway by a complex process that involves enzymatic processing of specific N-linked oligosaccharide groups on the surface of 68 glycoproteins (166,204,255,256). This requires the action of 0L1,2 ER mannosidase I whose inhibition by KIF leads to retention and stabilization of glycoproteins in the ER (199,200,248-251). In the current study, we show that KIF retards the degradation of COX-2 and a COX-l mutant containing the 19-aa cassette of COX-2. Spear and Ng (256) have recently shown that in a multiply glycosylated glycoprotein a single, specific N-linked oligosaccharide group serves as the determinant for entry into the ERAD pathway. In the present study, we have provided evidence suggesting that only one of the four N-glycosylation sites of COX-2 (Le. Asn-594) is involved in COX-2 degradation; however, degradation also requires at least some amino acids downstream of the consensus N-glycosylation sequence. We presume that a carbohydrate moiety linked to Asn-594 along with adjacent amino acids of the 19-aa insert interact with the complex of proteins that provide entry into the ERAD system. Very little of the l9-aa cassette can be resolved in the COX-2 crystal structure (12), and therefore, the l9-aa insert is thought to lack secondary structure. The last resolved residue in the murine COX-2 crystal structure is Ser—596, the final amino acid in the N- glycosylation consensus sequence. Ser-596 is solvent-exposed and appears to be oriented in such a way that its C terminus would point toward the lipid bilayer. The COX-2 KDEL-like ER retention motif at the C terminus (19) also has to be on the protein surface to be recognized (18). Thus, the 19-aa cassette of COX-2 is likely to be solvent-exposed and situated close to the membrane surface. In summary, we have identified a 19-aa cassette having a consensus N- glycosylation site at its N terminus that can target a protein to the ERAD system. Scanning mutagenesis should be helpful in identifying the critical residues in the l9-aa 69 cassette that work in conjunction with the Asn-594 N-glycosylation site to target COX-2 for degradation. It will also be important to characterize the oligosaccharide group linked to Asn-594. Acknowledgments We thank Dr. Anne Vojtek and Jennifer Taylor for their kind help with immunocytofluorescence. We are also grateful to Dr. Billy Tsai for his thoughtful suggestions relevant to this work. This work was supported in part by NIH Grant GM68848. 7O CHAPTER III ASN-594 GLYCOSYLATION AND PROTEIN DEGRADATION OF COX-2 IS REGULATED BY ITS C-TERMINAL 27 AMINO ACID INSTABILITY ELEMENT Summary The C-terminal 19 amino acid cassette (19-aa) of COX-2 is required for the entry of the protein into the ER-associated degradation (ERAD) system. We have previously shown that the Asn—594 glycosylation site at the start of the 19—aa cassette is necessary, but not sufficient, to mediate COX—2 ERAD. Here, we demonstrate that the C-terminal 16 amino acid portion of l9-aa is also essential for COX-2 degradation. We also show a positive correlation between the extent of glycosylation at Asn-594 and the overall degradation of COX-2. Evidence is provided to demonstrate that the 19-aa cassette is part of a larger 27 amino acid sequence that dictates the intracellular stability of COX-2 by regulating glycosylation at Asn-594. Analysis of the region immediately upstream of Asn—594 suggests that an 8-residue sequence with OL-helical structure impedes Asn-594 glycosylation, and in order for COX-2 to become glycosylated at Asn-594 there must be a change in the structure or conformation of this a—helix. We propose that this is elicited by the C-terminal l6-residue sequence of l9-aa. The upstream 8-residue sequence also appears to play a role in the processing of the N-glycan group at Asn-594 that preceeds translocation of COX-2 to the cytoplasm for proteasomal degradation. Therefore, the 71 interplay of the upstream 8-residue segment and the C-terminal 16 amino acid portion of l9-aa to regulate Asn—594 glycosylation and its N-glycan processing may control the timing and the extent of COX-2 protein degradation. Introduction Cyclooxygenases-l and -2 (COX-1 and COX-2) catalyze the key committed step in prostanoid synthesis (l,3,7,17). These enzymes are multiply N-glycosylated ER resident proteins that exist as homodimers and exhibit ~60% identity in primary structure (l,3,7,17,150). They are also integral membrane proteins that monotopically insert into the lipid bilayer so that they are largely compartmentalized in the ER lumen or the contiguous lumen of the nuclear envelope (14-16). The mature forms of the COX isoforms are very similar in structure except that COX-2 has a unique l9-residue insertion (l9-aa) near its C-terminus. Previously, we have shown that the 19-aa is essential for ER-associated degradation (ERAD) of COX-2 by the 26S proteasome (257). We have also demonstrated that the Asn-594 glycosylation site at the N-terminus of 19- aa is required for COX-2 ERAD (257). Immunoblotting of a variety of cell lines for COX-2 usually detects 72 and 74 kDa variably glycosylated forms of the enzyme due at least in part to alternative glycosylation at Asn-594 (148,151,258). Inhibition of co- translational N-glycosylation of COX-2 with tunicamycin at its four consensus glycosylation sites eliminates the cyclooxygenase (COX) and peroxidase (POX) activities of the enzyme (151). However, point mutation of the Asn-594 glycosylation site actually enhances COX—2 COX specific activity (151) most likely due to the increased stability of 72 the mutant enzyme (257). These results imply that while the Asn-594 glycosylation site is dispensable for proper co—translational protein folding, it is critical for the degradation of the enzyme. In HEK293 cells stably expressing recombinant native human COX-2 the Asn- 594 glycosylated form of the protein can only be detected upon addition of kifunensine, an ER (X-l,2 mannosidase I inhibitor (257). This is an indication that Asn-594 glycosylated COX-2 is rapidly degraded by ERAD. It is well established that the ER possesses a quality control or surveillance system that selectively identifies glycoproteins with non-native structure so that they are retained in the ER long enough to achieve their native state (l64—l66,182—186). Glycoproteins that are irreversibly defective are degraded by ERAD, a process that involves unfolding of the protein and its export to the cytosol where it undergoes proteolysis by the 26S proteasome (164-166,182-186,188,189). COX— 2 is the only ER luminal integral membrane protein that is known to be degraded via ERAD in its native or properly folded form. This raises the question as to why and how a properly folded glycoprotein is targeted for ERAD by the ER protein quality control system. We believe that a thorough structural and biochemical analysis of the C-terminal 19-aa of COX-2 may provide an answer to this conundrum. Very little of the l9-aa can be resolved in the highest resolution mouse COX-2 X- ray crystal structure attained thus far (157). The last resolved residue in this 2.4 A structure is Ser-596, the final amino acid in the Asn-594 N-glycosylation consensus sequence at the N-terminal end of 19-aa. The inability to resolve the remaining 16 residues of the 19-aa by X-ray crystallography may be an indication that this segment is largely disordered and lacks any degree of secondary structure. The Asn-594 73 glycosylation site is at the end of a two-tum helix that is linked to an upstream helix by a long lS-residue loop (Fig. 6). Asn-594 is not glycosylated in this structure presumably because the amide of its side chain is pointed upwards toward the upstream helix that is a distance of ~4.5 A. Since the space between the helices is barely sufficient to accommodate an N-glycan group, a local conformational change, probably mediated by movement of the long and inherently flexible intervening loop, would have to occur to expose Asn-594 for N-glycosylation. In our previous study, a HEK293 heterologous system was utilized to uncover the role of 19-aa in directing COX-2 protein degradation. In a different study, Dr. Masayuki Wada prepared fibroblast primary cultures from skin tissue of wild-type mice or a knock- in mouse carrying the mutant delS95-612 murine (mu) COX-2 in place of the wild-type gene (W ada and Smith, unpublished results). To induce COX-2 expression, these primary cultures were first made quiescent by 24 h serum starvation followed by challenge with fetal bovine serum. NIH/3T3 fibroblasts that have been growth—arrested by serum starvation lack detectable COX-2 expression but constitutively express COX-1 (22). By immunoblotting analysis, Dr. Masayuki Wada made the interesting finding that while COX-l was the only isoform that could be detected in growth-arrested primary cultures of wild-type mice, quiescent cultures prepared from delS95-612 COX-2 mutant (-/-) mice constitutively expressed both COX isoforms. Fibroblasts from heterozygous (+/-) mice also constitutively expressed COX-2; however, the COX-2 protein levels were lower compared to the -l- cells indicating that protein expression was gene dose-dependent. Serum-stimulation of these quiescent primary cells resulted in the time-dependent accumulation of COX-2 protein in ~/- cells in deep contrast to the more transient 74 expression of COX-2 in serum-treated wild-type cells. This is a confirmation that the 19- aa of COX-2 is essential for the degradation of the protein. More importantly, these results strongly argue that COX-2 ERAD that is mediated by l9-aa could represent a significant physiological mechanism for the regulation of COX-2 gene expression. In the present study we set out to examine what specific features of the C- terrninal l9-aa, in addition to Asn-594 glycosylation, are critical for targeting the enzyme to the ERAD system. The role of the helical region upstream of the Asn-594 glycosylation site in regulating Asn-594 glycosylation and COX-2 protein degradation was also investigated. Our findings have enabled us to identify a C-terminal 27 instability element of COX-2 that regulates the glycosylation of the enzyme at Asn-594, and subsequently, controls the timing and extent of protein degradation. Experimental Procedures Materials. Dulbecco’s modified Eagle’s medium (DMEM), fetal bovine serum (FBS), ponasterone A, and tetracycline were from Invitrogen. Bovine calf serum was from Hyclone. Cycloheximide, puromycin, and bacterial lipopolysaccharide (LPS) were obtained from Sigma. Kifunensine and MG132 were purchased from Calbiochem. Endoglycosidase H (Endo H) was purchased from Roche Applied Science. gastruction of Flam for Trflfection. Recombinant human (hu) COX-2 cDNA was subcloned into the tetracycline-inducible vector pcDNAS/FRT/TO (Invitrogen). After subcloning, the QuickChangeTM site-directed mutagenesis kit (Strategene) was used to create the following C-terminal mutants: del602-612 huCOX-2, del607-612 huCOX—Z, P607A T608A huCOX-2, V609A huCOX-Z, L610A huCOX-Z, 75 L611A huCOX-2, K612A huCOX-2, V591P T5920 huCOX-2, ovine (ov) COX-l UPSTRM8 huCOX-2, and murine (mu) COX-2 UPSTRM8 huCOX-2. The COX-1 insertion mutant insS94-60l ovCOX-l was created by QuikChangeTM site-directed mutagenesis using insS94-612 ovCOX-l as template. In3594-612 ovCOX-l mutant was previously made by overlap extension PCR and subcloned into the ecdysone-inducible pIND vector (Invitrogen). The COX-2 mutants, scrambled insert (Sins) 595-612 huCOX- 2 and Sins597-6l2 huCOX-2 were created from the cDNA template for native huCOX-2 by overlap extension PCR and then subcloned into pcDNAS/FRT/TO using BamHI and XhoI sites. Correct cDN A orientation and mutations were confirmed by sequencing. The primers and the PCR conditions used to design the above mutants are shown in the ‘Appendix’ section. Cell Culture and Transfection. NIH/3T 3 fibroblasts at early passage (<6 passages) were cultured in DMEM supplemented with 10% bovine calf serum and 100 u/ml penicillin/streptomycin. To induce COX—2 expression, the cells were first made quiescent by serum starvation for 48 h in DMEM containing 0.2% bovine calf serum and thereafter treated with DMEM supplemented with 20% FBS for 4 h. RAW 264.7 macrophage-like cells were cultured in DMEM supplemented with 10% FBS and 100 u/ml penicillin/streptomycin. To stimulate COX-2 expression, the cells were challenged with 200 ng/ml LPS for 12 h. HEK293-derived cell lines stably expressing native or mutant COX constructs were generated using either the tetracycline-inducible and ecdysone-inducible mammalian expression systems (Invitrogen) according to the manufacturer’s protocol. Constructs that were expressed under the control of a tetracycline-inducible promoter 76 were native huCOX-2, del602-612 huCOX-2, del607-6l2 huCOX-2, P607A T608A huCOX-2, V609A huCOX-Z, L610A huCOX-2, L611A huCOX-2, K612A huCOX-2, V591P T5920 huCOX-2, ovCOX—l UPSTRM8 huCOX-2, muCOX-2 UPSTRM8 huCOX-2, SinsS95-612 huCOX-2, and Sins597-612 huCOX—2. The insS94-60l ovCOX- 1 construct was expressed under the control of the ecdysone-inducible promoter. Stably transfected HEK293 cells were cultured in DMEM supplemented with 10% FBS, 100 u/ml penicillin/streptomycin, and the appropriate pharmacological selective reagents. Inducible expression was achieved by by treatment with 10 jig/ml tetracycline or 10 MM ponasterone A (ecdysone analog) for 24 h in normal culture medium. Protein Degradation and Drug Treatments. Quiescent 3T3 cells were serum- stimulated for 4 h then treated with 50 M cycloheximide (CHX) for different times in the presence or absence of 25 M kifunensine (KIF). RAW264.7 macrophage cells were challenged with 200 ng/ml LPS for 12 h then treated with 50 M CHX in the presence or absence of treatment with 50 M KIF. HEK293 cells stably and inducibly expressing wild-type or mutant cyclooxygenase constructs were grown to ~80% confluency, serum- starved for 24 h, and then treated with 10 ug/ml tetracycline or 10 uM ponasterone A in complete culture medium to induce expression. Afterwards, the cells were incubated for various times with 50 MM puromycin to block translation in the absence or presence of 20 MM M0132 or 25 “M KIF. Alternatively, stable transfectants of HEK293 cells were grown to ~80% confluency, treated with 10 ug/ml tetracycline for 24 h to induce expression, and thereafter treated for 12 h with 25 M KIF. Enyzmatic Deglycosylation. For complete deglycosylation, HEK293 whole cell lysates were denatured by boiling in NuPAGE SDS sample loading buffer (Invitrogen) 77 and then treated for at least 12 h with endoglycosidase H (Endo H) at a concentration of 0.4 mU/ul. Western Transfer Blotting. After the appropriate treatments, NIH/3T3, RAW 264.7, and HEK293 cells were scraped into ice-cold PBS (phosphate-buffered Saline), pH 7.4 containing 5 mM EDTA and a cocktail of protease inhibitors (Roche) and lysed by sonication. RIPA lysis buffer (10 mM Tris, pH 7.2, 150 mM NaCl, SmM EDTA, 0.1% SDS, 1.0% Triton X-100, 1% deoxycholate) containing a protease inhibitor cocktail (Roche Applied Science) was also used for cell lysis. Protein concentrations were determined using the BCA protein assay kit (Pierce). The NuPAGE system (Invitrogen) was used to resolve the proteins in the whole cell lysates on a 7% his-acetate polyacrylamide gel. After transfer to a nitrocellulose membrane, immunoblotting was performed with the appropriate primary antibody. Horseradish peroxidase-conjugated anti-rabbit or anti-mouse IgG antibodies (Bio-Rad) were used as secondary antibodies. Immunodetection was performed using the Western Lighting Chemiluminescent kit (Amersham Biosciences) followed by exposure to X-ray film. Densitometry analysis was performed using Quantity One software (Bio-Rad). Antibodies for Western Analysis. A previously generated (13), peptide-specific, polyclonal primary antibody for murine (mu) COX-2 against the epitope Ser598—Lys612 was used in the current study to detect muCOX-2 expressed in NIH/3T3 and RAW264.7 cells. Peptide-specific, polyclonal primary antibodies for ovCOX-l and huCOX-2 were synthesized by Covance Research Products against the following epitopes: Leu272- 01n283 of ovCOX-l and Pr0583-Asn594 of huCOX-2. A polyclonal antibody raised against whole muCOX-2, which also specifically detects native huCOX-2, was used to 78 immunoblot for the mutants V591P T5920 huCOX-2, ovCOX-l UPSTRM8 huCOX-2, and muCOX-2 UPSTRM8 huCOX-2. Results The C-terminal region of 19-& is important for COX-2 degradation. It has previously been shown that the Asn-594 glycosylation site is necessary, but not sufficient, for enabling the proteasomal degradation of COX-2 (257). Therefore, an additional portion of l9-aa is required to mediate COX-2 degradation. We embarked on determining which amino acids of the C-terminal l9-aa are needed for this process. Initially, two C-terminal deletion mutants of human (hu) COX-2 were prepared, namely del602-612 huCOX-2 and del607-612 huCOX-2 (Fig. 16a, panel I). We also prepared an insertion mutant of ovine (ov) COX-l, ins594-601 ovCOX-l, that carried an identical truncated region of l9-aa as del602-612 huCOX-2 (Fig. 16a, panel I). These mutants were stably transfected into HEK293 cells and their degradation profiles examined. All three mutants, including del607-612 huCOX-2, were stable and did not degrade within the 24 h time frame of the experiment (Fig. 16a, panel 11). These results indicated that removal of the last six residues of 19-aa was sufficient to eliminate COX-2 degradation. Preventing COX-2 degradation by trimming the l9-aa raised the possibility that a correct length for this insertion, rather than its primary structure, was needed to mediate protein turnover. To eliminate this possibility, we prepared two mutants of COX-2 that carried scrambled versions of the 19-aa (Fig. 16b, panel I). Sins597-612 huCOX-2 has an intact Asn-594 glycosylation site but the rest of the l9-aa has been scrambled. SinsS95-612 huCOX-2 is not only mutated at Asn-594, but it also has a randomized version of the rest 79 of the l9-AA. After stably transfecting these scrambled mutants into HEK293 cells their protein stabilities were analyzed. Both mutants were much more stable than native huCOX-2 because they degraded very slowly with a half-life (tug) > 24 h (Fig. 16b, panels 11 and 111). Overall, these results suggest that there is primary structure information within the 19-aa cassette, in particular its last six residues, that is essential for dictating COX-2 protein degradation. To identify which of the last six amino acids of 19-aa were important for enabling COX-2 degradation we performed alanine scanning mutagenesis of this region (Fig. 17a). The mutant P607A T608A huCOX-2 had a tug of ~8 h which was not far removed from that of the native huCOX-2 (ti/2 ~5 h) (Figs. 17b and c). Its degradation kinetics was also similar to that of the native enzyme (Fig. 17c). The V609A huCOX-2 mutant also degraded with a Ill/2 of ~8 h (Figs. 17b and c). However, careful examination of its overall degradation profile revealed that V609A huCOX-2 was degraded less efficiently than both the native and P607A T608A huCOX-2 (Fig 17c). There seemed to be a significant loss of V609A huCOX-2 protein at the earlier time points after inhibiting protein translation. Thereafter, at the 12 h and 24 h time points, this mutant appeared to be relatively stable compared to native and P607A T608A huCOX-2 (Figs. 17b and c). Although the mutations L610A huCOX-2, L611A huCOX-2, and K612A huCOX-2 did not completely prevent COX-2 degradation, they significantly stabilized the enzyme; the 11/2 for the degradation of all three mutants was >24 h (Figs. 17b and c). Collectively, these results point to a three amino acid sequence at the C-terminal end of 19-aa, namely Leu-6lO, Len-611, and Lys-612, as being essential for enabling COX-2 degradation. 80 16A. (1) 5BOFSVPDPELIKTVTInnisssasC4Llanrnp-r'vnracERSTEL618 native huCOX—2 53°FSVPDPELIKTVTINA_ ERSTEL607 del602—612 hucox—2 58°FSVPDPELIKTVTINASSSR8GLDDIN —————— ERSTELGlz de1607-612 hucox-z SBOFHVPDPRQEDRPGV ERPPTEL5°° native ovCOX—l 58°FHVPDPRQEDRPGVNASSSRSGLDDINPTV'LLKERPPTELG19 insS94—612 ovCOX-l SBOFHVPDPRQEDRPGVNA ERPPTEL6°8 insS94—601 ovCOX—l A. (II) del602-612 huCOX-Z del607—612 huCOX-Z 0 2 4 12 24 (h) 0 2 4 12 24 (h) ‘ r 7i ' ’T” " , ’T! if"? ”7‘ h ” " " —_” ' 71‘ z: . a in " ' I ‘N ~'.‘. . . .. . , Actin? fl-n-nq'v I Actinii v" ”A " * Q rm > 24 h rm > 24 h iu5594-601 ovCOX-l o 2 4 12 24 (h) Actin t,,, > 24 h 16B. (1) 58°FSVPDPELIKTVTINASSSRSGIDDIN’PTVLLKERSTELSIB native huCOX-Z SBOFSVPDPELIKTV'I‘INASIRGPLTKLSDNSLVSDERSTELGlB Sins597—612 hucox-2 SBOFSVPDPELIKTVTIAASIRGPLTKLSDNSLVSDERSTELEle SinsS95—612 hucox—z B. (11) Sins595-612 huCOX-Z Sins597-612 huCOX—Z 0 2 4 12 24 (h) 0 2 4 12 24 (h) "'r‘r'xA—x “all-Arr.“ Acfinwflflad Actin “'.w (M > 24 h t” > 24 h 82 16B. (111) c» C .E —- .5 —-___-— E __~—— - 2 —_ .5 8 2 (L 32 ‘5 0 . . e . 0 5 10 15 20 25 Time after puromycin (h) . O native nucox_2 ‘- — — — i SinsS97-612 “cox-2 '- — — — '- SinsS95—612 huCOX-Z Figure 16. Deletion and scrambled mutations of the l9-AA cassette disrupt COX-2 degradation. A (I) and B (1), Amino acid sequences of the C-termini of cyclooxygenase constructs that were stably transfected into HEK293 cells. The underlined sequence is the consensus N-glycosylation site at the start of the COX-2 l9-aa cassette. The C-terminal sequences of native huCOX-2, native ovCOX-l, and ins594—612 ovCOX-l are included for comparison. A (II) and B (11), HEK293 cells stably and inducibly expressing the constructs shown in (I) were grown to ~80% confluency, subjected to serum-starvation for 24 h, and then treated with the appropriate inducing agent (10 ug/ml tetracycline or 10 [1M ponasterone A) for 24 h. Puromycin (50 M) was then added to block translation and COX-2 and actin protein levels were analyzed at different times by Western blotting. The Western blotting results in A(II) and EU!) are representative of two and three separate experiments, respectively, from which protein half-lives were determined. B (III), Densitometry analysis of the degradation of native huCOX-Z, Sins595-6l2 huCOX- 2, and Sins597-612 huCOX-Z. The results are based on at least three different experiments for each protein. The error bars denote +/- SE of the mean. 83 17A. 58017SVPDPELIKTVTIEAjSSRSGmDINPTVLMERSTELMB 5SOFSVPDPELIKTVTIESSRSGLDDINAAVLIKERSTELMB 58°FSVPDPELIKTVTIflhjSSRSGLDDINPTAI-LKERSTEL“8 58°FSVPDPELI KTVTI_NA_SSSRSGLDDINPTVALKERSTEL618 58°1='svapiz:LIKTV'I‘I_1~m_sssitsGm!):tmvrvniutfi:RsTEL618 53°FSVPDPELIKTVT1&8s8118mm)ImuvrvnnnERsTEL618 native huCOX—2 . P607A T608A huCOX—2 V609A huCOX—2 L610A huCOX-2 L61 1A huCOX-Z K612A huCOX-2 B. P607A T608A huCOX-Z V609A huCOX-2 0 2 4122401) 0 2 4122401) n “ A fi H .- 2.. Actin rc r. m m are; Aetin v- v n w. ~ tr2"8h L610A huCOX—Z L61 1A huCOX-2 0 2 4 12 24(h) ‘III-If-|‘-"‘ Actin I'- F- i- O '- Actin tm > 24 b K612A huCOX-Z 0 2 4 8 24 (h) Actin Actin “have m 6’3w «”4. 55¢ 3? 1503 é‘ufl it? " .4- g1>24h 84 'Ili- ' “WAA " ~ 0 2 412 24 (h) iqw ,. .. (“an .9 - ‘ w finfii‘ifiq t1,2 > 24 h native huCOX-Z 0 2 4 3‘24 '(h) {‘1' are: new we. a“. if.» in ~ 5 h 17 C. P607A T608A huCOX—2 100 a: 80 - .E .E 2 60 ‘ 2 .E 6 . § 40 a .\’ 20 ‘ O . . f . 0 5 10 15 20 25 Time (h) V609A huCOX-2 100 g 80 . .E 3 so - 2 5 3 4o ‘ 2 :1. ,\° 20 + 0 r . O 5 10 15 20 25 Time (h) ¢—0 native .______.. mutant 85 17C. (cont’d) L610A huCOX-Z 100 \ ‘ ’ t. 804 \ \ D \ \ .5 ‘ x ----------- 2 5 J ‘3 4o - E at 20 ~ 0 . . r . 0 5 10 15 20 25 Time (h) L611A huCOX-Z 100 a 80 " C E (I E 60 ‘ 2 .E 3 40 ~ 2 n. =2 20 j 0 . . . . 0 5 1O 15 20 25 Time (h) ¢——9 native .. ..... _. mutant 86 K612A huCOX—Z 100 00 O \‘I---{ ______ _ —————— i O) O 4h 0 °/o Protein remaining N O O 10 15 20 25 Time (h) C m . ‘—9 native '- ..... .. mutant Figure 17. The C-terminal end of l9-aa is essential for COX-2 protein degradation. A, Alanine scanning mutants of the C-terminal portion of l9-aa that were stably transfected into HEK293 cells. The underlined sequence is the Asn-594 glycosylation site. B, HEK293 cells stably and inducibly expressing the constructs shown in (A) were grown to ~80% confluency, subjected to serum-starvation for 24 h, and then treated with 10 ug/ml tetracycline for 24 h. Thereafter, the time course of degradation of the proteins was determined as described in the legend to Fig. 16. Protein half-lives of all proteins except P607A T608A huCOX-Z were determined from at least three separate experiments. C, Densitometry analysis of the degradation of the mutants shown in A and B. The results for native huCOX-2 and for all mutants except P607A T608A huCOX-Z are based on at least three independent experiments. Densitometry analysis of P607A T608A huCOX-2 is based on a single experiment. The degradation profile of huCOX-Z has been included for comparison. The error bars denote +/— SE of the mean. 87 Glycosylation of Asn-594 correlates with COX-2 protein degwon. We previously reported that kifunensine, which inhibits ERAD by preventing N—glycan processing by ER 0L1,2 mannosidase I, stabilizes huCOX-Z stably expressed in HEK293 cells (257). At the same time kifunesine (KIF) leads to the appearance of a less mobile, alternatively glycosylated form of the native enzyme in HEK293, NIH/3T3, and RAW 264.7 cells even after protein translation has been inhibited (Figs. 180, b, and c). This higher glycosylated form is not observed after KIF treatment of HEK293 cells expressing N594A huCOX-2 (257). Endo H treatment of native huCOX-2 expressing cells collapses both alternatively glycosylated forms to the 66 kDa fully deglycosylated form of the enzyme (Fig. 18a). Therefore, we believe that KIF stabilizes the 74 kDa Asn-594 glycosylated form of the native enzyme. The enhanced levels of the 74 kDa form upon KIF treatment could be used to assess Asn-594 glycosylation of COX-2 in HEK293 cells. Experiments were performed to determine if the deletion mutants delS97-612 huCOX-Z, del602-612 huCOX-2 and del607-612 huCOX-Z, and the scrambled mutant Sins597-612 huCOX-2 are glycosylated at Asn-594. The deletion mutant delS97-612 huCOX-2 has the same electrophoretic mobility as delS95-612 huCOX-Z, which lacks the Asn—594 glycosylation site (Fig. 19a, panel I). KIF treatment of cells expressing delS97-612 huCOX-2 did not affect the mobility of this mutant suggesting that it is not glycosylated at Asn-594 (Fi g. 19a, panel II). Similarly, del602-612 huCOX-2 and del607-612 huCOX- 2 did not appear to be glycosylated at Asn-594 (Fig. 19a, panel 11). The scrambled mutant SinsS97-612 huCOX—Z carrying an intact Asn-594 glycosylation site had the same electrophoretic mobility as SinsS95-612 huCOX—2 with or without KIF treatment 88 18A. 74 kDa 72 kDa 66 kDa —* 1. tet-induced 2. tet-induced + 12 h KIF 3. EndoH cleavage of l 4. EndoH cleavage of 2 B. 25 EM KIF 0 2 4 8 24 2 4 8 24 Time with puromycin (h) HEK293 pan-i.“ put-grit“: 120 .E’ .E g ._. No inhibitor :3 L". 25 uM KIF .5 3 L ...... A 50 “M KIF O. 32 0 Time with puromycin (h) 89 18C. 25 pM KIF 50 pM KIF 0 2 4 8 2 4 8 4 8 Time with CHX (h) 1:: NIH/3T3 II'_‘___‘____,;_;....:.... d------""" Actin so KIF o 2 4 2 4 Time with CHXOI) RAW 264.7 {,figifl-Wtwgz Figure 18. KIF treatment stabilizes the Asn-594 glycosylated form of COX-2. A, Native huCOX-2 expression in HEK293 cells was induced with 10 ug/ml tetracycline for 24 h . After protein induction cells were incubated for an additional 12 h in normal culture medium with or without 25 “M KIF. Thereafter, cell lysates were prepared and boiled in SDS sample loading buffer and then treated with or without Endo H (0.4 mU/ul) for at least 12 h. Proteins were resolved by SDS-PAGE on a 7% tris-acetate polyacrylamide gel and subjected to Western blotting. B, Native huCOX-2 expression was induced as described in the legend to Fig. 16. Puromycin (50 M) was then added to block translation in the absence or presence of 25 51M KIF. Thereafter, COX-2 protein levels were analyzed at different times by Western blotting and densitometry. The densitometry analysis is based on three different experiments that were performed to examine the effect of 25 “M and 50 M KIF on COX-2 degradation. The error bars denote +/- SE of the mean. C, Quiescent NIH/3T3 cells were stimulated with 20% FBS for 4 h to induce COX-2 expression. Cycloheximide (CI-IX; 50 11M final concentration) was added to the medium and the cells were incubated for the indicated times with or without additional treatment with 25 11M or 50 11M KIF. RAW264.7 cells were treated with 200 ng/ml LPS for 12 h to induce COX-2 expression. Thereafter, cells were treated with 50 ”M CHX for the indicated times in the presence or absence of 50 M KIF. Cell lysates were prepared and analyzed for COX-2 protein levels. Arrows in B and C show the 72 and 74 alternatively glycosylated forms of COX-2 detected by immunoblotting. 9O (Fig. 19b). Just like delS97-612 huCOX-2, treatment with KIF did not result in the detection of a less mobile glycosylated form of SinsS97—612 huCOX-2 (Fig. 19b). Collectively, these results indicate that mutations of the 19-aa that confer stability to COX-2 also prevent glycosylation of Asn-594. Consistent with this observation, the mutant V609A huCOX-2, which is degraded to a lesser extent compared to native huCOX-2, is also glycosylated less efficiently at Asn-594 in KIF-treated cells (Fig. 19c). Therefore, the C—terminal 16 amino acids of 19-aa appear to play an important role in effecting glycosylation of Asn-594. This could be one mechanism by which the 19-aa cassette mediates COX-2 protein degradation. The helical region upstream of Asn-594 negatively regu_lates Nglvcosvlation. Previously, we showed that inserting the COX-2 19-aa near the C-terminus of ovCOX-l yielded an unstable mutant ins594-612 ovCOX—l that was degraded with a tm of ~3 h (257). A comparison of the molecular masses of ins594-612 ovCOX—l with that of its stable, Asn-594-mutated counterpart ins594-612 (N594A) ovCOX-l suggested that the former was fully glycosylated at Asn-594 (257). Unlike native huCOX-Z, KIF treatment of HEK293 cells expressing ins594-612 ovCOX-l failed to elicit the appearance of a higher glycosylated form of this ovCOX-l mutant; instead, KIF significantly stabilized insS94-612 ovCOX-l (257). The apparent disparity between native huCOX-2 and insS94-612 ovCOX-l with respect to Asn-594 glycosylation suggested that the ease with which the two proteins became glycosylated at this site was different. The fact that the “/2 of ins594-612 ovCOX-l is ~2 h shorter than that of native huCOX-2, also suggested that the efficiency of Asn-594 glycosylation appears to positively correlate with the rate of 91 19A. (1) 1 2 5 ~ . 1. delS95-612 hucox-z “ fl 2. delS97-612 huCOX-Z A. (II) (191597-512 huCOX—Z del602-612 huCOX-Z del607-612 huCOX-Z 1 2 3 4 1 2 3 4 l 2 3 4 fl ‘9‘} “It U W I -~,’ wtfi w~ ‘V l. tet-induced 2. tet-induced + 12 h KIF 3. EndoH cleavage of 1 4. EndoH cleavage of 2 B. l 2 3 4 . SinsS95-612 huCOX-Z Sins597-612 huCOX-Z Sins595-612 huCOX-Z + 12 h KIF SinsS97-612 huCOX-2 + 12 h KIF AWN—- 92 19C' native V609A hucox-z l 2 l 2 3 4 ' “'1':- 1"?“ 1. tet-induced 2. tet-induced + 12 h KIF 3. EndoH cleavage of l 4. EndoH cleavage of 2 Figure 19. COX-2 stabilizing mutations of the 19-aa prevent glycosylation of Asn- 594. A (I), The expression of delS95-6l2 huCOX-2 or delS97-612 huCOX-2 was induced with 10 jig/ml tetracycline for 24 h. (11), After protein induction as in (1) cells were incubated for an additional 12 h in normal culture medium with or without 25 M KIF (delS97-612 huCOX—2) or 50 M KIF (del602-6l2 huCOX-2 and del607-612 huCOX-2). Thereafter, cell lysates were prepared and boiled in SDS sample loading buffer for 10 min and then treated with or without Endo H (0.4 mU/iil) for at least 12 h. Proteins were resolved by SDS-PAGE and subjected to Western blotting as in 18A. B, Expression of SinsS95-612 huCOX-2 or Sins597-612 huCOX-2 was induced as in A(I). After protein induction cells were incubated for an additional 12 h with or without 25 11M KIF. Proteins were resolved by SDS-PAGE and subjected to Western blotting as in 18.4. C, Expression of native huCOX-2 or V609A huCOX-2 was induced as in A( I). After protein induction cells were incubated for an additional 12 h with or without 25 uM KIF. Cell lysates were then prepared and boiled in SDS sample loading buffer for 10 min and then treated with or without Endo H (0.4 mU/ttl) for at least 12 h. Proteins were resolved by SDS-PAGE and subjected to Western blotting as in 18A. Arrows indicate differences in molecular mass due to N—glycosylation. 93 COX degradation. Native huCOX-2 and ins594-612 ovCOX-l have very similar sequences downstream of Asn-594; therefore, we decided to probe the region upstream of Asn-594 in huCOX-2 for its ability to negatively regulate N-glycosylation. In the muCOX-2 X-ray crystal structure, the Asn-594 glycosylation site is at the C-terminal end of a 9-residue (it-helical region (Figs. 20a and b). This helix (helix A) is connected to a nearby helix (helix B) by a long 15-residue loop; four of these residues are presumed to be part of the loop because they cannot be resolved in the crystal structure (Figs. 20a and b). The amide side chain of Asn-594 appears to be pointed upwards in the direction of helix B which is at a distance of ~4.5 A (Fig. 20c). The length of two GlcNAc residues of an N-glycan group is ~8 A. Therefore, helix B is situated such that it may impede the transfer of an N-glycan group to the amine group of the amide chain of Asn-594. Furthermore, a nearby disulfide bridge formed between Cys-569 of helix B and Cys-575 of the loop may also impede Asn-594 glycosylation because it is situated ~4.7 A away (Fig. 20c). Moreover, the disulfide bridge may limit the flexibility of the loop, thereby, constraining helix A into a conformation in which Asn-594 cannot become glycosylated. The loop and the disulfide bridge can both be seen in the X-ray crystal structure of ovCOX-l, indicating that they are conserved in both isoforms (Fig. 20d). The last resolved residue in the ovCOX-l structure is Pro-583, which is part of the loop; therefore, the region that would be immediately upstream of Asn-594 in ins594-612 ovCOX-l (ie. Asp-584 to Val-593) cannot be resolved, suggesting a lack of secondary structure. Furthermore the presence of proline and glycine at two consecutive positions, 591 and 592, respectively, makes it highly improbable that this region would possess any secondary structure. It is therefore likely that ins594—612 ovCOX-l is fully glycosylated 94 at Asn-594 because this glycosylation site is situated in a loop that is long enough to be very flexible even in the presence of the disulfide linkage (Fig. 206). To test this hypothesis, we prepared and stably transfected huCOX-2 mutants bearing various modifications of an 8-residue sequence (UPSTRM8) immediately upstream of Asn-594. The mutation V591P T592G huCOX—2 introduces a proline and a glycine into UPSTRM8 to disrupt the helix that is presumably formed by this region of huCOX-Z. This mutation was also designed to mimic the corresponding region of ovCOX-l (Fig. 21a). A second mutant was made in which the whole UPSTRM8 region of huCOX—2 was replaced with the corresponding region of ovCOX-l (Fig. 21a). This mutant is designated ovCOX-l UPSTRM8 huCOX-2. The initial rates of degradation of V591P T592G huCOX-2 (ti/2 ~4-5 h) and ovCOX-l UPSTRM8 huCOX-2 (ti/2 ~3 h) were similar to that of the native enzyme (Figs. 21b and c). However, overall, both mutants appeared to be degraded to a greater extent than native huCOX-2 (Fig. 21c). For instance, essentially all of V591P T592G huCOX-2 or ovCOX-l UPSTRM8 huCOX-2 was degraded by the 24 h time point, while ~15% of native huCOX-2 remained (Fig. 21c). Also, these mutations resulted in the detection of the 74 kDa glycosylated form even without KIF treatment (Figs. 21b, 22a, and 22b). This interesting observation led us to hypothesize that disrupting helix A facilitated Asn-594 glycosylation, and consequently, enhanced the overall degradation of the protein. If this is true, then replacing the UPSTRM8 region of huCOX-2 with that of muCOX-2 should not change the glycosylation pattern at Asn-594. As shown in Fig. 21a substantial differences exist between the UPSTRM8 regions of muCOX-2 and huCOX—2. Nonetheless, we expected that helix A of muCOX-2 would be conserved in huCOX-2. The initial degradation rate 95 of the mutant muCOX-2 UPSTRM8 huCOX-2 (ti/2 ~4-5 h) was similar to that of native huCOX-2 and the ovCOX—l UPSTRM8 mutants (Figs 21b and c). Moreover, the extent of the overall degradation of this mutant appeared to be intermediate between that of the native huCOX—2 and the ovCOX-l UPSTRM8 mutants (Fig. 21c). The Asn-594 glycosylation pattern of muCOX-2 UPSTRM8 huCOX-2 was similar to that of the native huCOX-2 in that unlike the ovCOX-l UPSTRM8 mutants, very little of the Asn-594 glycosylated form of this mutant could be detected in the absence of KIF or M6132 treatment (Figs. 22a and b). The modest detection of Asn-594-glycosylated muCOX-2 UPSTRM8 huCOX-2 is consistent with the observation that the extent of its overall degradation is slightly greater than that of native huCOX-2. Nonetheless, these results are consistent with our expectation that the UPSTRM8 regions of muCOX-Z and huCOX—2 have conserved helical secondary structure which appears to negatively regulate Asn—594 glycosylation. It is important to note that the putative helix A formed by the UPSTRM8 of huCOX-2 may actually be 2 residues longer since, unlike muCOX-2 UPSTRM8, this region lacks a proline residue at its N-terminus (Fig. 21a). Just like native huCOX-2, the degradation of all three UPSTRM8 huCOX-2 mutants involved entry into the ERAD system because the 26S proteasome inhibitor MG132 and the ER al,2-mannosidase I inhibitor KIF stabilized these mutants (Figs. 22a, b, and c). These findings lead us to propose that the UPSTRM8 of huCOX-2 acts in concert with the downstream l9-aa to control the timing of the entry of the protein into the ERAD system and the extent of protein degradation by regulating glycosylation of Asn-594. 96 20A. helix loop * helix 5638‘ IQSLICMGCPFTSFNVBDPQPEKTATINABS96 mouse SIQSLICNNVKGCPFTSFSVPDPELIKTVTINAS human Helix B n-570 -569 Ser 6 Helix A 97 Pro-583 helix + -------- > 56‘ LKKLVCLRTKTCPYVSFHVPDPRQEDRPGV ——————————————————— ERPPTELGUO ovcox-l 55‘ IQSLICNNVKGCPFTSFSVPDPELIKTVTINASSSRSGLDDINPTVLLKERSTEL“8 hucox—z E. helix loop -k helix 5“ IQSLICNNVKGCPFTSl-“SVPDPELIKTVTImisssrtsammIiu=t'1'v1.i:.RERS'I‘ELM native hucox—z 9“ LKKLVCLNTKTCPWSFHVPDPRQEDRPGVNAa "M... . . rent” insS94—612 ovCOX—l V helix loop Figure 20. Structure of the C-terminal region of COX-1 and COX-2. A, Amino acid sequences of the C-terminal regions of muCOX-2 and huCOX-Z that are located upstream of the Asn-594 glycosylation site (underlined). Residues that are colored blue are conserved between the two species. Segments that form helices in the muCOX-2 X- ray structure are highlighted. The region marked with an asterisk is not resolved in the muCOX-2 X-ray structure and is very likely to be a continuation of the resolved loop. The disulfide linkage between Cys-569 and Cys-575 is also shown. B, Ribbon diagrams depicting the structure of the C-terminal region of muCOX-2 whose sequence is shown in A. The last resolved residue in the muCOX-2 X-ray structure is Ser-596, the final amino acid of the Asn-594 glycosylation site. Asn-594 is situated in a helix (Helix A) that is positioned right under an adjacent helix (Helix B). The amide chain of Asn-594 appears to be pointed upward in the direction of helix B. The position of the disulfide linkage relative to Asn-594 is shown. C, A ribbon diagram showing the distances between Asn- 594 and Asn-570 of helix B (4.5 A), and Asn-594 and the disulfide linkage between the loop and helix B (4.7 A). D, Top, Structure of the C-terminal region of ovCOX-l. The last resolved residue in the X-ray structure of ovCOX-l is Pro-583. The conserved 98 disulfide linkage between Cys-569 and Cys-575 is shown. Bottom, Amino acid sequences of the C-termini of ovCOX-l and huCOX-2. The sequence in ovCOX-l that forms a helix is shown. The sequence highlighted with a dashed double arrow is the region of ovCOX-l that is not resolved in the X-ray crystal structure whose corresponding region is resolved in the X-ray structure of muCOX-2. Conserved residues between ovCOX-l and huCOX-2 are highlighted in red. The conserved cysteines that form the disulfide bridge in either isoform are colored blue. The last resolved residue, Pro-583, is also colored blue. E, Comparison of the C-termini of native huCOX-2 and insS94-612 ovCOX-l. Segments expected to form a helix are highlighted with black double arrows. Segments expected to form a loop are highlighted with blue double arrows. The area of native huCOX-2 marked with an asterisk is likely to be a continuation of the helix. The conserved Cys-569 and Cys-5 75 are colored red. 21A. 53°FSVPDPELIKTVTIESSRSGLDDINPTVLLKERSTELe18 native hucox-2 SBOFSVPDPELIKTPGIESSRSGLDDIN‘PTVLLKERSTEL‘S18 V591P T592G hucox-z 58°FSVPDPRQEDRPGVEA—SSSRSGLDDINPTVLLKERSTEL518 ovCOX-l UPSTRM8 huCOX—2 530FSVPDPQPTKTATIESSRSGLDDINPTVLLKERSTEL518 mucox—z UPSTRM8 huCOX-Z B. V591? T592G huCOX—Z OVCOX-l UPSTRMS huCOX—Z 0 2 4 12 24 (ii) “-9. ”2., ....‘3_ -12. 24 0') Actin can... Actin “C-.. tug” 3 h tm ~ 4-5 h muCOX-Z UPSTRM8 huCOX—Z 0 2 4 12 24 (h) Actin ..“~ t1]; ~ 4-5 h 99 21C. °/o Protein remaining O————O A- ----- A 100 “I. Protein remaining 80- 60‘ 40- 20- ovCOX-l UPSTRM8 huCOX-2 100 on O O) O 40 20 \ i ' — ~ _ * 0 I I I I—¢ 0 5 10 15 20 25 30 Time (h) native mutant V591P T592G huCOX—Z \ 0 5 10 15 20 25 Time (h) .—9 native l- ----- -I mutant lOO 21C. (cont’d) muCOX-2 UPSTRM8 huCOX-2 100 on O 60‘ 40- % Protein remaining 20- Figure 21. Degradation of the UPSTRM8 huCOX-2 mutants. A, Amino acid sequences of the C-termini of UPSTRM8 huCOX-2 mutants that were stably transfected into HEK293 cells. The underlined sequence is the Asn-594 glycosylation site. B, After stably transfecting the UPSTRM8 huCOX-2 constructs shown in A, protein expression was induced as described in the figure legend to Fig. 16. Thereafter, cells were treated with 50 M puromycin for the indicated times and remaining levels of COX-2 and actin were analyzed by Western blotting. The Western blotting results are based representative of three separate experiments from which protein half-life measurements were made. Arrows indicate differences in molecular mass due to N—glycosylation. C, Densitometry analysis of the degradation of the UPSTRM8 huCOX-2 mutants relative to native huCOX-2. Densitometry is based on three separate experiments. Error bars denote +/— SE of the mean. Red asterisk indicates that the difference in % protein levels between the mutant and native protein at a given time point is statistically significant based on a Student’s paired t-Test (p value < 0.01). Black asterisks indicate that the overall degradation of the mutant relative to the native protein is statistically significant based on a Student’s paired t-Test (p value < 0.05). 101 22A. (1) Endo H cleavage 123456123456 “kDa—v 74 kDa : 72 kDa ”W . . , MV 1. native huCOX-Z 2. native huCOX-2 + KIF 3. V591P T5926 huCOX—Z 4. V 591P T592G huCOX—Z + KIF 5. muCOX-2 UPSTRM8 huCOX-Z 6. muCOX-Z UPSTRM8 huCOX-Z +KIF Endo H cleavage l. native huCOX-Z 2. native huCOX-Z + KIF 3. ovCOX-l UPSTRM8 huCOX-Z 4. ovCOX—l UPSTRM8 huCOX-2 + KIF 102 22B. (1) V591P r5920 huCOX-Z 20 “M MG132 0 2 4 12 24 2 4 12 24 (h) Actin "wanton nun-d B. (11) ovCOX-l UPSTRMS huCOX-Z 20 1.1M MGI32 o 4 12 24 4 12 24 (h) :z I! n in «a. -., .— B. (III) muCOX—Z UPSTRMs huCOX-Z 20 MI MG132 0241224 24122401) :2: UI-iudir- i"§i‘g‘“‘ Actin . “mi! Wilt. W “8' 103 22B. (IV) native huCOX-2 20 pM MGl32 0 2 4 8 24 2 4 12 24 (h) -..__-.____ _. --.—a—.-— _.-__.._..-....,.._.._. ..._..._- ._:_..... (. .. _ . “J. _ . _, " ~.. , . ‘3‘}? ,i,. r “155': .'-1-_. :53 .‘fl: 3'; r151; 11' 'i- L,- 1l- - .- ‘ -.‘ 1' ,. --'-'-'a¥!-'. ’.- ' M9 “:I-JKC. - ‘af'? .. hi”- ' ' ‘ W" {‘2‘}; 3m 5 ‘t 3.} r ' J 'a‘ . ‘. 1’ 1‘1. ."L'. ‘_ ""rs'c ' _ “If ‘ is; "w ‘\.‘ T 7' 21".. 1 it . . f H i . ‘r. u“ - Actin ~ II ' ~ ~ |~ avw -u- v-r-I— In.“ ‘ 1:- a.-. rv v-uu --——' .. v-awr—n- - u-nu——-— r——-—-—fi - ”—— _————-. ., 9 C. V591 P T5926 huCOX-2 % Protein remaining Time (h) Q. _____ .9 No protease inhibitor I 1 20 W M6132 104 22C. (cont’d) ovCOX1 UPSTRM8 huCOX2 % Protein remaining Time (h) muCOX—2 UPSTRM8 huCOX—2 1 20 1 00 80 60 4O % Protein remaining 20 O 5 1 0 15 20 25 Time (h) 0. _____ 4 No protease inhibitor I— 1 20 11M MG132 105 22C. (cont’d) Native huCOX—2 E E 3 5: Time (h) .1. ————— 4 No protease inhibitor I—-—I 20 11M MG132 Figure 22. Asn-594 glycosylation of the UPSTRM8 huCOX-2 mutants and their degradation by the ERAD pathway. A (I) and (II), The expression of native huCOX-2 or the UPSTRM8 huCOX-2 mutants was induced with 10 ug/ml tetracycline for 24 h. After protein induction cells were incubated for an additional 12 h in normal culture medium with or without 25 M KIF. Thereafter, cell lysates were prepared and boiled in SDS sample loading buffer for 10 min and then treated with or without Endo H (0.4 mU/ul) for at least 12 h. Proteins were resolved by SDS—PAGE and subjected to Western blotting as in 18A. Arrows indicate differences in molecular mass due to N—glycosylation. B (I), (II), (III), and (IV), HEK293 cells stably expressing the UPSTRM8 huCOX-2 mutants were grown to ~80% confluency, subjected to serum-starvation for 24 h, and then treated with 10 ug/ml tetracycline for 24 h. Puromycin (50 M) was then added to block translation with or without additional treatment with 20 “M MG132. Thereafter, COX-2 and actin protein levels were analyzed at different times by Western blotting. Arrows indicate differences in molecular mass due to N—glycosylation. C, Quantitative analysis of the effect of 20 M MG132 on the protein stability of native huCOX-2 and the UPSTRM8 huCOX-2 mutants. 106 Protein Extent of Asn-594 Protein half-llfe Extent of protein glycosylation clearance Full glycosylation Full clearance ins594-612 ovCOX-l ~ 3 h ovCOX-l UPSTRM8 huCOX-Z ~ 3 h V591? T5926 huCOX-z ~ 4—5 h muCOX-z UPSTRM8 huCOX-Z ~ 4-5 h Native huCOX-Z -— 5 h V609A huCOX-Z ~ 8 h 31115597612 huCOX-2 No glycosylation >24 h (MSW-612 huCOX-z No clearance del602-612 huCOX-Z > 24 h 412507-612 huCOX-Z Table 2. Relationship between the extent of Asn-594 glycosylation and overall protein degradation. The extents of Asn-594 glycosylation and protein clearance are arbitrary and are based on Western blotting and densitometry data. Protein clearance is a relative measure of how much protein is remaining at the end of the 24 h experiment. 107 Discussion Inhibiting the N—glycosylation of COX-2 during its synthesis and maturation in the ER severely compromises its catalytic activity (151). Therefore, co-translational N- glycosylation of COX-2 appears to be required for the proper folding of the enzyme. It is well known that there are ER-resident chaperones, namely calnexin and calreticulin, that associate with folding or misfolded glycoprotein substrates mainly by binding to their exposed N—linked oligosaccharide groups ( 166). Asn-594, the last N-glycosylation site of COX-2, is usually variably glycosylated (148,151,258), and is not required for the enzyme to achieve a catalytically competent conformation (151). Previously, we have shown that the Asn-594 glycosylation site is essential for the ER-associated degradation (ERAD) of COX-2 (257). In the present study, we have addressed the question of whether glycosylation of Asn-594 is essential for enabling COX-2 ERAD. We provide evidence that Asn-594 glycosylation is regulated by interplay of an upstream segment, approximately 8 residues long and a downstream l6-residue sequence that forms part of the 19-aa cassette. Furthermore, we observed a positive correlation between the extent of Asn-594 glycosylation and the overall degradation of the protein (Table 2). This is based on the following findings: a) l9-aa deletion mutants of huCOX-2 that have an intact Asn- 594 glycosylation site but lack various portions of the l9-aa appear not to be glycosylated at Asn-594 and are completely stable; b) scrambling the l6-amino acid sequence downstream of the Asn-594 glycosylation site prevents glycosylation of Asn-594 and yields a mutant that is degraded much slower than native COX-2; c) inserting the huCOX-2 19-aa near the C-terminus of ovCOX-l results in the complete glycosylation of Asn-594, and the COX-1 mutant has a shorter tl/z (~3 h) compared to native huCOX-2 108 (~5 h)(257); and d) replacing the 8-residue sequence immediately upstream of Asn-594 in huCOX-2 with the corresponding region of ovCOX-l facilitates Asn-594 glycosylation concomitant with a shorter 11/2 (~ 3 h) and more effective clearance of the protein. Both the 72 and 74 kDa alternatively glycosylated forms of COX-2 can be readily detected by immunoblotting in various cell types, including NIH/3T3 fibroblasts and RAW264.7 macrophages. However, heterologous expression of recombinant huCOX-2 in HEK293 cells yields predominantly the 72 kDa form. The 74 kDa Asn-594- glycosylated huCOX-2 form can be detected in these cells after treatment with kifunensine (KIF), an inhibitor of ER a1,2 mannosidase I, which we have shown to stabilize COX-2 (257). ER a1,2 mannosidase I catalyzes the committed step in the ERAD pathway (259,260). Its activity releases irreversibly misfolded glycoproteins from the calnexin/calreticulin folding cycle by cleaving a single mannose residue from an N- linked MangGlcNAcz (198-200). A putative Mans-binding lectin Yos9p or the ER- degradation mannosidase I-like protein (EDEM) will bind MangGlcNAcz and facilitate the delivery of protein having a processed N—glycan to a membrane retrotranslocon for export out of the ER and into the cytosol for proteasomal degradation (201- 204,206,207,248). The identity of this membrane retrotranslocon has not been determined. KIF inhibition of ER a1,2 mannosidase I prevents the retrotranslocation and proteasomal degradation of aberrant glycoproteins by causing them to be retained in the ER (199,200,248-251). Since KIF inhibits COX-2 degradation and leads to accumulation of the Asn-594 glycosylated form of the enzyme in HEK293, NIH/3T3, and RAW264.7 cells, we reason that COX-2 ERAD requires Asn-594 glycosylation and subsequent processing of the attached N-glycan group by ER a1,2 mannosidase I. 109 Even though ERAD is considered to be an ER quality control mechanism for the elimination of aberrantly folded glycoproteins, COX-2 appears to be degraded from a properly folded conformation. Sifers and co-workers have proposed that glycoprotein ERAD (GERAD) requires at least two signal determinants: a) an N—glycan component, and b) non-native protein structure (259). Their proposal is based on the observation that inhibiting the glycosylation of misfolded glycoproteins prevents their degradation (261). However, it is still unclear how GERAD quality control is able to selectively eliminate terminally defective glycoproteins while sparing folding glycoproteins that have yet to achieve their native protein structure. It is possible that the intimate physical interaction of folding glycoproteins with ER resident molecular chaperones protects them from being degraded. Taking into consideration both the criteria for GERAD and our overall findings, we provide a model for the initiation of COX-2 degradation by Asn-594 glycosylation (Fi gs 23, 24, and 25). In this model, COX-2 synthesis and maturation in the ER involves co-translational glycosylation of its first three N-glycosylation sites, but not Asn-594. Glycosylation of these sites as the COX-2 nascent protein grows from the Sec61 translocon would serve the purpose of recruiting the chaperones calnexin and/or calreticulin to assist in protein folding. COX-2 maturation also entails formation of disulfide bonds, membrane insertion, heme incorporation, and assembly into a homodimer. This yields a catalytically competent COX-2 having an Asn-594 site that may not be easily accessible for glycosylation because it is situated in an ordered helix (helix A) such that its amide group is protected by an adjacent helix (helix B) ~4.5 A away. A disulfide bridge between Cys-569 of helix B and Cys-575 of a 15-residue loop connects helices A and B. This covalent modification may further contribute to 110 constraining helix A into a conformation which prevents glycosylation of Asn-594. However, this disulfide bridge is also present in COX-1 and we have found that the COX-1 insertion mutant ins594-612 ovCOX-l appears to be fully glycosylated at Asn- 594. Therefore, we propose that the degradation of COX-2 is initiated by a local conformational change in the helix A-loop-helix B region that enables the post- translational glycosylation of the protein at Asn-594. This conformational change probably involves the last 16 amino acids of the l9-aa cassette since this region appears to be required for Asn-594 to become glycosylated. The mechanism by which the C- terminal 16 amino acid segment of l9—aa promotes glycosylation of Asn-594 is yet to be determined. It is possible that after this putative loop region may form hydrophobic and electrostatic interactions with helix A, thereby, causing helix A to change conformation so that it is no longer stacked against helix B. Alternatively, the C-terminal 16 amino acids of 19-aa may serve as a docking site for a protein complex that will remodel the helix A-loop-helix B region so that the side chain of Asn-594 is exposed for glycosylation. Based on our experimental results, it is unlikely that cleavage of the disulfide bridge between the intervening loop and helix B is required for Asn-594 to become glycosylated. Otherwise, one of the proteins in this complex may be a protein disulfide isomerase that would catalyze the breakage of the disulfide bond between the loop and helix B. Oligosaccharyltransferase (0ST) could also be part of this complex. Since OST is a membrane-bound complex that is associated with the protein translocon 111 23A. Helix B native huCOX-2 Helix A l l \v - ERSTELCOOH B. i :3” 3?: ‘ in3594-612 ovCOX-1 \ N5“ \ < \ \ X ‘ _ _ ‘ , .ERPPTEL°°°” Figure 23. Schemes illustrating how the C-terminus of COX-2 may regulate Asn- 594 glycosylation. A, In native huCOX-2, Asn-594 glycosylation may be hindered by the ordered helical conformation of the region immediately upstream of Asn-594. The C- terrninal 16 amino acid segment of l9-aa appears to promote glycosylation of Asn-594. B, Since insS94-612 ovCOX-l lacks secondary structure in the vicinity of Asn-594, this residue is easily accessible to glycosylation. Asn-594 glycosylation of ins594-612 ovCOX-l may not require reduction of the disulfide bond. The green continuous line represents the loop that is resolved in the X-ray structures of muCOX—Z and ovCOX-l. The green dashed line is the putative loop region upstream of Asn—594 that is not resolved in either isoform. The black dashed line represents the C-terminal l6-amino acid segment of 19-aa. 112 24. Possibility A Helix B 5.5 . 321 <1. <1 .e .‘e l i. Helix A “ Ma. \\ z - ERSTELCOOH _ \ \ I ‘ ERSTELC°°H Possibility B Helix B -' S—S ‘ N’“ (:5- - — ' l Helix A 1 3’0... | | .‘ \w - ERSTEL°°°“ ' : M». ‘w - ERSTELCOOH Figure 24. Two possibilities of how Asn-594 of native huCOX-Z may become accessible to glycosylation. The loop region from Cys-575 to helix A could be long and flexible enough to enable movement of helix A to permit Asn-594 glycosylation (Possibility A). Alternatively, reduction of the disulfide bond may also be needed to sufficiently expose Asn—594 for glycosylation (Possibility B). In either case, the C- terminal 16 amino acid segment of 19-aa would be required to mediate Asn-594 glycosylation. 113 25. HelixB y GI Gil S "i. ' m '--' ERp57 .fe , 3:. Disassembly, unfolding ‘ 5“. '. e-‘ and refolding PV (GlcaMangGlcNAC21‘5 (Glc1MangGlcNAc2) Helix ‘i, ".l 611 I | |‘.. - ERSTEL°°°" \.- ERSTEL°°°" UGGT (ManeGlcNAcz) f l ' ( o ding sensor) I ‘0'." I '.. ' 0 w '\ I ‘. ER Man I ‘m ERSTEL°°°N O {3" (MansGlcNAcz) / %: KIF We. _—’ ,— w - ERSTEL°°°" Figure 25. A model of how Asn-594 glycosylation mediates the entry of COX-2 into the ERAD pathway. An N—glycan group consisting of GIC3Man9GlcNAcz is transferred en bloc from ER membrane-bound dolichol pyrophosphate to Asn-594. This occurs after the helix A-loop-helix B region has been remodeled to permit glycosylation. The remodeling process could be mediated by a complex of proteins that dock onto the 16- amino acid segment of 19-AA. Glucosidases I and 11 (G1 and GII) will cleave off the two terminal glucose residues leaving a monoglucosylated N-glycan that is specifically recognized by calnexin, a membrane-bound lectin binding chaperone. The heterodimer complex of calnexin and ERp57 will participate in the disassembly, unfolding, and refolding of the protein in an attempt to correct the structural modification to the native enzyme. The protein will be maintained in the calnexin cycle by the actions of G11 and the folding sensor and glucosyltransferase UGGT. Since COX-2 is irreversibly glycosylated at Asn-594, the protein will eventually be released from the calnexin cycle by ER (21,2 mannosidase 1 (ER Man I) which cleaves off the a1,2-1inked mannose to form MangGlcNAcz. This will initiate ERAD of Asn-594 glycosylated COX-2. Processing of the MangGlcNAcz oligosaccharide group by ER Man I is the committed step of ERAD and is inhibited by KIF. 114 (262,263), the C terminal region of mature COX-2 has to be in the vicinity of the membrane for Asn-594 to become post-translationally glycosylated. Introduction of the local conformational change in the helix A-loop-helix B region and the subsequent glycosylation of Asn-594 may constitute the signals needed to initiate COX-2 GERAD. The above model for COX-2 degradation could be tested in several ways. We have preliminary evidence that COX-2 undergoes post-translational glycosylation at Asn- 594. This is based on the observation that in addition to preventing COX-2 ERAD in HEK293 cells, KIF treatment causes the accumulation of what appears to be the Asn-594 glycosylated form, even in the absence of protein synthesis; this COX-2 glycosylated form is barely detectable in untreated HEK293 cells. Accumulation of the 74 kDa Asn- 594 glycosylated form of COX-2 in KIF-treated cells may inhibit further glycosylation of Asn-594. This would explain why KIF also stabilizes the 72 kDa form of the native enzyme. To the best of our knowledge, the only published report of post-translational N- glycosylation is that of the secretory glycoprotein human coagulation factor VII (FVII). FVH has two consensus glycosylation sites, and, like COX-2, exists as two alternatively glycosylated forms of 54 and 56 kDa (264). Using 35S pulse chase analysis, Steenstrup and co—workers elegantly demonstrated that the 54 kDa form is a precursor for the 56 kDa form, and by Endo H treatment were able to show that the difference between the two forms was due to N-glycosylation. Similarly, it will be important to perform 358 pulse chase analysis to confirm that the 72 kDa form of COX-2 is indeed the precursor for the 74 kDa form in KIF-treated cells. It would also be interesting to examine the effect of mutagenesis of Cys-575 on the extent of Asn-594 glycosylation and the overall degradation of COX-2. 115 In conclusion, we have identified a potential mechanism for the initiation of COX-2 ERAD that involves Asn-594 glycosylation, its negative regulation by an 8- residue segment that is immediately upstream, and its positive regulation by the last 16 amino acids of the l9-aa cassette. The upstream 8-residue sequence may also play a role in the processing of the N—glycan group at Asn-594 that preceeds translocation of COX-2 to the cytoplasm for proteasomal degradation. We reason that both the 19-aa cassette and the upstream 8-residue sequence form a 27 amino acid sequence that constitutes the instability element responsible for controlling the intracellular stability of COX-2. Acknowledgments We thank Dr. Randal Kaufman and Dr. Billy Tsai for helpful suggestions relevant to this study. 116 CHAPTER IV SUBSTRATE DEPENDENT COX-2 PROTEIN DEGRADATION Summary Cyclooxygenases (COX-1 and COX-2) catalyze the committed step in prostanoid synthesis. Previously, we showed that the unique C-terminal 19 amino acid cassette of COX-2 enables degradation of this COX isoform in a proteasome-dependent manner. Here, we demonstrate that COX-2 degradation in NIH/3T3 and HEK293 cells is enhanced by treatment with exogenous arachidonic acid (AA) at concentrations as low as 5-20 11M. At similar concentrations AA did not cause significant COX-l degradation in either cell type. Endogenous COX-2 degradation in serum-treated NIH/3T3 cells was enhanced by treatment with 10 11M calcium ionophore (A23187) and was almost completely blocked by cyclooxygenase (COX) inhibitors. AA—induced COX-2 degradation was retarded by COX inhibitors. The 26S proteasome inhibitor M0132, which blocks basal COX-2 protein turnover in HEK293 cells, did not affect the AA- dependent degradation of the protein. Moreover, AA-dependent degradation of COX-2 occurred independently of its C-terminal 19 amino acid cassette. Only fatty acid substrates of COX-2 were able to enhance degradation. Also, 3 6533A COX-inactive point mutant of COX-2 was resistant to AA-induced degradation even though it underwent substrate—independent degradation at the same rate as native COX-2. Loss of COX specific activity and prostaglandin product formation was also observed in COX-2- expressing cells treated with AA. We propose that substrate-dependent inactivation of 117 COX-2 causes structural damage to the enzyme that subsequently leads to its degradation. Introduction Cyclooxygenases-1 and -2 (COX—1 and COX-2) are membrane-bound, ER- resident hemoproteins that catalyze the committed step in prostanoid synthesis (l,3,7,17,150). Although encoded by different genes, COX-1 and COX-2 are structural isoforms that share ~60% identity in primary structure. COX-1 is a stable protein that is constitutively expressed in resting cells of many tissues, most notably in platelets and vesicular gland (1,257,265,266). In contrast, COX-2 is a stimulus-inducible protein that is quite unstable and whose expression is observed to be short-lived in epithelial, endothelial, smooth muscle, and fibroblast cells (20,21,23,24,257). The short half-life of COX-2 protein mimics that of the COX-2 mRNA. Both the COX-2 mRNA and protein have been found to possess instability elements that target them for rapid degradation. At the 3’ untranslated region (UTR) of COX-2 mRNA are multiple AUUUA elements that are known to target the message for rapid exonuclease cleavage (20,129). COX-1 mRNA lacks these 3’ UTR AU-rich elements which would explain the prolonged expression of COX-1 mRNA in megakaryocytes and vascular endothelial cells (117,118,267). We have recently reported that a unique C-terminal l9-amino acid insertion near the C-terminus of COX-2 confers the enzyme with a short half-life by rapidly and selectively initiating its proteasomal degradation (257). This cassette is part of a larger 27 amino acid sequence that appears to be essential for directing ER-associated degradation of the enzyme. It could therefore be postulated that the evolution of COX-2 must have entailed the 118 development of post-transcriptional and post-translational mechanisms that contribute to the sophisticated tight regulation of the synthesis of COX-2-derived prostanoids. The principal endogenous fatty acid substrate of COX-1 and —2 is arachidonic acid (20:4A5'8'11’”) which is mobilized from the sn-2 position of membrane phospholipids upon the activation of cytosolic phospholiase A201 (cPLAzoc) with bradykinin, thrombin, growth factors, calcium ionophore, or cytokines (l,3,8,36-41,43). The cyclooxygenase isoforms have two catalytic activities that occur at two spatially distinct active sites. At the cyclooxygenase (COX) active site, arachidonic acid (AA) is oxygenated to form the hydroperoxide and endoperoxide prostaglandin G; (PGGZ) (1,3,7). This prostaglandin intermediate is then moved to the peroxidase (POX) site where its hydroperoxy group undergoes a two—electron reduction to form the alcohol prostaglandin H2 (PGH2) (l,3,7,156). PGH2 is a common substrate for downstream terminal prostanoid synthases that act upon it differently to form various prostanoids. The POX and COX activities of COX-1 and -2 undergo irreversible suicide inactivation in vitro during catalysis. POX and COX self inactivation is mechanism-based because it proceeds from heme and tyrosyl radical intermediates that are formed during the POX and COX reactions, respectively (3,156). The specific structural changes in the holoenzyme that lead to suicide inactivation remain unknown. Mevkh et al. noted that COX-l self inactivation is accompanied by dramatic changes in protein structure as is manifested by the increased susceptibility of the inactive enzyme to trypsin cleavage and the increased number of exposed histidine residues subject to chemical covalent modification (162). It has not been established whether the COXs can undergo suicide inactivation in vivo. If so, this could serve as an additional regulatory mechanism for 119 prostanoid synthesis. In this regard it would be interesting to investigate whether the inactive forms of COX-1 and -2 are more susceptible to degradation compared to the native forms. It is well established that misfolded or structurally damaged proteins that are present in the ER can be targeted for degradation by a process known as ER- associated degradation (ERAD) (l64-166,182-186). It is possible that COX-2 might selectively undergo suicide inactivation at endogenous hydroperoxide or AA levels that would be insufficient to inactivate COX-1. If indeed the inactive protein gets degraded, this could partly explain the short-lived nature of COX-2 protein. The present study examined the impact of COX catalysis in vivo on the protein stabilities of COX-1 and COX-2. We demonstrate that COX-2 protein degradation that is mediated by the C- terrninal 19 amino acid insertion can be substantially enhanced by COX substrates in NIH/3T3 and HEK293 cells in a proteasome-independent manner. We also provide evidence that substrate-dependent degradation of COX-2 requires a functional COX active site and proceeds from substrate-induced inactive forms of the enzyme. 120 Experimental Procedures Materials. Dulbecco’s modified Eagle’s medium (DMEM), fetal bovine serum (FBS), ponasterone A, and tetracycline were obtained from Gibco/Invitrogen. Bovine calf serum (BCS) was from Hyclone. Arachidonic acid (AA), eicosapentaenoic acid (EPA), eicosadienoic acid (EDA), oleic acid (OA), linoleic acid (LA), 2-arachidonoyl glycerol ether (2-AG ether), dihomo-y-linolenic acid (DHLA), (R/S)-flurbiprofen, (S)- flurbiprofen, (R)-flurbiprofen, and NS-398 were from Cayman Chemicals. (S)- flurbiprofen and NS-398 are time-dependent, irreversible inhibitors of cyclooxygenase (COX) activity whereas (R)-flurbiprofen is a competitive COX inhibitor. Cycloheximide, puromcyin, and glutathione (GSH) were purchased from Sigma. Calcium ionophore (A23187), bradykinin, and M0132 were purchased from Calbiochem. [l-‘4C] arachidonic acid (55 mCi/mmol) was from American Radiolabeled Chemicals. Microsomal PGES-l (mPGES-l) was kindly provided by Dr. Michael Garavito of Michigan State University. Construction of Plasmids for Transfection. Recombinant ovine (ov) COX-1 cDNA and N-terminal hexahistidine-tagged ovCOX-l cDNA were subcloned into pIND (Invitrogen). cDNAs for human (hu) COX-2, N-terminal hexahistidine-tagged huCOX-2, and N-terminal hexahistidine-tagged murine (mu) COX-2 were subcloned into pcDNAS/FRT/TO (Invitrogen). pIND is ecdysone-inducible whereas pcDNAS/FRT/TO is tetracycline-inducible. After subcloning, the Quick-ChangeTM site—directed mutagenesis kit (Stratagene) was used to create the following huCOX-2 mutants: 0533A huCOX-2, N594A huCOX-2, and delS95-612 huCOX-2. Correct cDNA orientation and mutations were confirmed by sequencing. 121 Cell Culture and Transfection. NIH/3T3 fibroblasts at early passage (<6 passages) were cultured in DMEM supplemented with 10% BCS and 100 u/ml penicillin/streptomycin. To induce COX-2 expression, the cells were first made quiescent by serum starvation for 48 h in DMEM containing 0.2% BCS and thereafter treated with DMEM supplemented with 20% FBS for 4 h. Stable HEK293 transfectants of native or mutant COX-2 constructs were generated using the Flp-In T-Rex tetracycline-inducible expression system (Invitrogen). Native ovCOX-l or His-tagged ovCOX-l were stably transfected into the ecdysone- inducible expression system (Invitrogen). All these transfections were conducted according to the manufacturer’s protocol. Stably transfected HEK293 cells were cultured in DMEM supplemented with 10% FBS, 100 u/ml penicillin/streptomycin, and the appropriate pharmacological selective reagents. Inducible expression was achieved by a 24 h serum starvation in serum-free medium followed by treatment with 10 pg/ml tetracycline or 10 uM ponasterone A (ecdysone analog) for an additional 24 h in normal culture medium. Fatty @ and Drug Treatments. Quiescent 3T3 cells were serum-stimulated for 4 h then treated with 50 uM cycloheximide (CHX) or 50 11M puromycin for different times in the presence or absence of 20 M arachidonic acid (AA), 100 M flurbiprofen (FB), 20 M NS-398, or a combination of 20 11M AA and one of the following: 100 1.1M flurbiprofen (FB), 20 11M NS-398, or 20 11M MGl32. Alternatively, after serum- stimulation the cells were treated for different times with 50 11M cycloheximide (CHX) or 50 M puromycin in the presence or absence of 10 M A23187 or 10 1.1M bradykinin with or without 100 11M flurbiprofen , 20 11M NS-398, or 20 11M MGl32. 122 HEK293 cells inducibly expressing wild-type or mutant cyclooxygenase constructs were grown to 80% confluency, serum-starved for 24 h, and then treated with 10 ug/ml tetracycline or 10 uM ponasterone A in complete culture medium for 24 h to induce expression. Afterwards, the cells were incubated for various times with 50 12M puromycin to block translation in the absence or presence of the polyunsaturated fatty acids AA, EPA, EDA, 2-A0 ether, LA, DHLA, 0A, or the COX inhibitor flurbiprofen. To examine the effect of COX, proteasome, lysosomal, and ER to Golgi trafficking inhibitors on AA-induced protein turnover, cells expressing native huCOX—2, delS94-612 huCOX—2, or N594A huCOX-2 were treated for 4 h with 50 11M puromycin with or without AA in combination with one of the following: 20 11M M0132, 100 M flurbiprofen, 50 11M (S)-flurbiprofen, 50 11M (R)-flurbiprofen, 20 11M NS-398, 50 M leupeptin, 25 11M E64, or 5 jig/ml brefeldin A. Microsome Prepa_ration 3nd Trypsin Cleajvagg. HEK293 cells stably expressing delS95-612 huCOX—2 or N594A huCOX-2 were lysed by sonication in 0.1 M Tris-Cl buffer, pH 8.0. The lysates were centrifuged at 5,000 rpm for 10 min at 4°C to precipitate unbroken cells. Thereafter, microsomes were prepared by ultraentrifugation at 55,000 rpm for l h at 4°C. The microsomes were resuspended in 0.1 M Tris-Cl buffer, pH 8.0 and 200 pig of this resuspension was incubated in a 100 ill reaction mixture containing 0.1 M Tris-Cl, pH 8.0, 10 11M hematin, and 1 mM phenol in the absence of substrate, or with 100 “M AA, or a combination of 100 11M AA and 100 1.1M PB. The incubation was performed at 37°C for 4 h. 100 pig of microsomes were treated with 5 pg trypsin before or after solubilization with 1% Tween-20. Trypsin treatment was performed at 37°C for 20 min. 123 Western Trmfer Blotting. After the appropriate treatments, NIH/3T3 and HEK293 cells were harvested by centrifugation at 2000 rpm for 3 min. Cell pellets were stored at -80°C until needed. RIPA lysis buffer (10 mM Tris, pH 7.2, 150 mM NaCl, SmM EDTA, 0.1% SDS, 1.0% Triton X-100, 1% deoxycholate) containing a protease inhibitor cocktail (Roche Applied Science) was used for cell lysis. Protein concentrations were determined using the BCA protein assay kit (Pierce). The NuPAGE system (Invitrogen) was used to resolve the proteins in the whole cell lysates on a 7% tris-acetate polyacrylamide gel. After transfer to a nitrocellulose membrane, immunoblotting was performed with the appropriate primary antibody. Horseradish peroxidase-conjugated anti-rabbit or anti-mouse IgG antibodies (Bio-Rad) were used as secondary antibodies. Immunodetection was performed using the Western Lighting Chemiluminescent kit (Amersham Biosciences) followed by exposure to X-ray film. Densitometry analysis was performed using Quantity One software (Bio-Rad). Cyclooxygenase Inactivation Assay. Cultured HEK293 cells stably expressing His-tagged ovCOX-l or His-tagged muCOX-2 were incubated for 10 min with different concentrations of AA ranging from 2.5 M to 50 12M. After incubation the cells were washed with 1X PBS (phosphate buffered saline) and harvested by centrifugation at 2000 rpm for 3 min. Cell pellets were resuspended in 0.1 M Tris, pH 8.0 buffer containing 1 mM EDTA and a protease inhibitor cocktail and lysed by sonication. Protein concentration in the lysates was determined by BCA assay (Pierce). COX activity assays were performed at 37°C by monitoring the initial rate of oxygen uptake using an oxygen electrode as described previously (268). Reactions were initiated by adding cell lysate to 124 the assay chamber containing 3 ml of 0.1 M Tris-H01, pH 8.0, 1 mM phenol, 5 11M hematin, and 100 M AA. Microsomes were prepared from the lysates of His muCOX-2-epxressing cells by ultracentrifugation at 55,000 rpm for 1 h at 4°C. The microsomes were resuspended in 0.1 M Tris-HCl, pH 8.0 containing 1 mM EDTA and 20% glycerol and a BCA assay (Pierce) was perfomed to determine protein concentration. The microsomal fractions were then used to perform COX radioactive TLC assays to examine COX product formation as previously described (269). Briefly, microsomes (0.5 mg) were incubated in a 100-ltl reaction mixture containing 0.1 M Tris-HCl, pH 8.0, 1 mM phenol, 10 11M hematin, and 10 11M [1-14C]arachidonic acid at room temperature for 1 min. To assay for PGE2 formation microsomal POES-l (mPGES-l) and 60 “M GSH were included in some of the reaction samples. The reactions were terminated by adding 300 pl of an ice-cold mixture of ethylether, methanol, and 0.2 M citric acid (30:4:1). The organic phase of the reaction mixture, containing the radioactive products, was isolated and applied directly onto a TLC silica plate at 4°C. The plate was developed in ethyl acetate, 2,2,4- trimethylpentane, acetic acid, and water (110:50:20:100) and exposed to x-ray film, and radioactive products were visualized by autoradiography. 125 LESLIE COX Inhibitors Retard COX-2 Degradation in Nil/3T3 cells. About nine years ago Smith and co-workers made the interesting finding that serum-stimulating quiescent NIH/3T3 cells in the presence of the non-selective NSAIDS flurbiprofen and aspirin, or the COX-2 selective inhibitor NS-398 dramatically enhanced inducible COX—2 protein expression without affecting COX-2 mRNA levels ((14), Arakawa and Smith, unpublished results). In contrast, constitutive COX-1 protein expression was not affected. These results suggested that COX inhibitors promote the stability of COX-2 protein in NIH/3T3 cells by preventing the degradation of the enzyme. After successfully reproducing these findings (Fig. 26a) 1 tested hypothesis that NSAIDS inhibit the rapid degradation of COX-2. In a cycloheximide chase experiment (R/S)-flurbiprofen, (R)- flurbiprofen, and (S)-flurbiprofen nearly completely retarded COX-2 degradation in NIH/3T3 cells within the time frame of the experiment (Fig. 26b). There is no evidence to date that NSAIDS directly inhibit the 26S proteasome. Since proteasome inhibitors partly inhibit COX-2 degradation in 3T3 cells (257), my findings suggest the possibility of a second non-proteasome—dependent pathway for COX-2 protein turnover. Moreover, flurbiprofen inhibits COX-2 degradation in 3T3 cells to a greater extent than the proteasome inhibitors M0132 and epoxomicin (data not shown). Therefore, it is likely that COX inhibitors prevent both the proteasomal and non-proteasomal degradation of COX-2 in 3T3 cells. Proteasomal Degradation of COX-2 in HEK293 cells is not inhibited by NSAIDS. We have previously reported that heterologously expressed COX-2 undergoes proteasomal degradation in HEK293 cells in a manner that is dependent upon the C- 126 terminal 19 amino acid cassette of the protein (257). To determine if COX inhibitors stabilize COX-2 in HEK293 cells, COX-2 protein degradation was examined in these cells in the presence or absence of treatment with 20 11M NS-398 or 100 pM flurbiprofen. We were surprised to find that, unlike 3T3 cells,ineither NS-398 nor flurbiprofen significantly stabilized COX-2 in HEK293 cells (Fig 26c). Furthermore, inducible COX- 2 expression in these cells was not enhanced by treatment with 20 M NS-398 (Fig. 26d). Serum-stimulation of quiescent 3T3 cells results in the coordinate induction of COX-2 and the mobilization of free AA due to activation of cPLA20t (77). Therefore, it is reasonable to expect that serum-treatment of quiescent 3T3 cells will stimulate COX-2 COX catalysis. During catalysis COX-2 may undergo suicide inactivation, a process that could initiate the rapid degradation of the enzyme. Resting HEK293 cells lack detectable phosholipase A2 activity so that heterologously expressed COX-2 is more likely to be in its latent form (33,58). This may explain the discrepancy that is observed in the two cell lines with respect to the effect of COX inhibitors on COX-2 protein turnover. It is possible that COX-2 is degraded in serum-treated 3T3 cells by at least two distinct mechanisms: (a) COX substrate-induced protein turnover which is inhibited by COX inhibitors; and (b) a proteasome-dependent basal degradation of the resting enzyme that is directed by the C—terminal 19 amino acid cassette. Substrate-dependent Degraflttion of COX-2. To test the hypothesis that COX fatty acid substrates can induce COX-2 protein turnover, we analyzed the effect of treatment with exogenous AA on the degradation of native huCOX-2, delS95-612 127 26A. 100 M FB 0 2 4 6 10 2 ..4 7 6 7 10 '(h) COX-2 Wait first. “mm A Actin _ , V and amp fl Um. V’W" 7". a 1.6 l' \ E 1.2 4 I \ \- W g - E 9 3 3- a 0 2 4 6 8 10 12 Time after induction (h) Q—O No inhibitor |___.| molt-MFR 128 26B' 100 FB 2Q“ 0‘ 2 8 2 8 TimewithCHX (h) i. .4 -I—‘hu uni-q .r cox-2 ' a... 2. 4i 2...... Actin ..,,.- 2.,” .4... ~w-«w m 100 “M FB 0 2 8 2 8 (h) Densitometry (%) 100 58 35 102 103 50 M 500 |.l.M 50 M No inhibitor (Lu-FR (R)-FB (S)-FB 0 2 4 2 4 2 4 2 4 (h) ’L'Jk- “an" .‘ern 1!: W.» \‘L‘J ' -1124- ‘ .?;~.Wr+'” ’ . .L-mm.) :‘J‘ET‘J-I' 1' .‘ MeM‘i- - in"".‘-Lj_7'73 ow ' - - . 't .170;va rue-Te: ism-er» nu.‘ ma. iv-uv-vv M'\‘ rib-.2 - ~.--o nay-27:; mh,. — -.. Inn nun-urn... I- - I ' '- wilful .‘p .-.I .bfimqu‘ '2 .rfi'ifflL‘r ACtln 3“ _" ‘ a a“ (tori-.1 w “I W“ l . . "‘ynra ‘. - . '- .-~., "" . - u - ran—n! No so M 500 11M so itM Inhibitor (R)-FB (R)-FB (sire 0 ‘2 4| ‘2 4| ‘2 4“2 4‘(h) Densitometry (%) 100 39 36 65 83 80 69 65 48 129 100 FB 26C. 100M“ 2 24 (h) I Densitometry(%) I 100 I 96 I 48 I 46 I 18 I 100 I 62 I 37 I 17 I 100 NS-398 0 2 4 12 24 2 4 12 24 (h) huCOX-2 . “ w “P‘. W (gm, ~ .. Actin r g u I!“ in; w ‘2! '5 .3 7 W M V A— “— A20MNS-398 o 2 4 12 24 2 4 12 24 (h) [Densitomeu'y(%) I 100 I 86 I 77 I 50 I 21 87 I 77 I 47 I 30 I D. l 2 huCOX-2 tiff 4'" 1. tet-lnduced Actin -" 2. tet-induced + 20 “M PIS-938 Figure 26. Cyclooxygenase inhibitors prevent the rapid degradation of COX-2 in serum- treated NIH/3T3, but not HEK293, cells. (A) NIH/3T3 cells were made quiescent by 48 h serum-starvation and serum—induced for different times in the absence or presence of 100 11M flurbiprofen (FB). (B) Quiescent (Q) 3T3 cells were serum-induced for 4 h then treated with 50 11M cycloheximide (CI-1X) with or without 100 M FB, 50 M (R)—FB, 500 uM (R)—FB, or 50 M (S)—FB. (C) HEK293 cells stably expressing huCOX—2 were serum—starved for 24 h and treated with 10 jig/ml tetracycline in normal complete medium for an additional 24 h to induce COX-2 expression. Upon COX—2 induction, cells were treated with 50 11M puromycin to block translation with or without 100 M FB or 20 11M NS-398. (D) COX expression was induced in HEK293 cells with tetracycline (tet) in the absence (1) or presence (2) of treatment with 20 11M NS-398. After the treatments in (A )-(D) the cells were harvested, lysed, and analyzed by Western blotting for COX-2 and actin. 130 huCOX-2, and N594A huCOX-2 in HEK293 cells. The basal degradation of native huCOX-2 was dramatically enhanced by treatment with 20 M AA (Fig. 27a, panel I). An AA dose response of COX-2 degradation revealed that protein turnover could be substantially enhanced by fatty acid concentrations as low as 5 11M (Fig. 27b, panel I). The mutants delS95-612 huCOX-2 and N594A huCOX-2, which are usually stable under basal conditions, also rapidly degraded upon treatment with 10 M AA (Fig. 27a, panels 11 and III, and Fig. 27b, panel II). Therefore, substrate-induced degradation of COX-2 is not mediated by the C-terminal l9-amino acid cassette. Since removal of the cassette does not affect COX-2 catalytic activity, these results imply that substrate—induced degradation could be a consequence of substrate turnover at the COX active site. To test this hypothesis, we examined the effect of COX inhibitors on substrate-induced COX-2 degradation in HEK293 cells. We also analyzed a panel of polyunsaturated fatty acids only some of which are COX-2 substrates. Flurbiprofen, (R)-flurbiprofen, (S)- flurbiprofen, and NS-398 significantly blocked the AA-induced degradation of huCOX-Z in 293 cells (Figs. 27c and d). Moreover, these NSAIDS selectively inhibited COX-2 degradation that was substrate-dependent without affecting the basal degradation of the enzyme (Figs. 27c and d). In contrast, the degradation of delS95-612 huCOX-2 and N594A huCOX—2, which was only observed upon exogenous AA treatment, was nearly completely blocked by flurbiprofen and (S)-flurbiprofen (Fig. 27e). In order to be utilized as substrate by COX-2, exogenous AA would have to penetrate the cell, move to the ER membrane by diffusion or carrier-mediated transport, and be delivered to COX-2 on the luminal side of the membrane. This transport and delivery process should be very rapid since prostaglandin product formation is 131 known to reach a plateau within minutes after treatment with exogenous AA in cultured cells expressing COX activity (270). Therefore, a brief challenge of cells with exogenous AA should be sufficient to induce COX-2 degradation. HEK293 cells stably expressing huCOX—2 were exposed to 20 11M AA for 5 min. or 4 h. After aspirating the AA- containing medium, cells that were treated for 5 min with AA were washed with 1X PBS then treated with the protein translation inhibitor puromycin with or without flurbiprofen for an additional 4 h. It was observed that regardless of the duration of AA treatment (5 min or 4 h), COX-2 degradation was enhanced to a similar extent (Fig. 27f). Flurbiprofen added after the 5 min treatment with AA did not protect the enzyme from degradation as well as when the inhibitor was added together with substrate. These observations indicate that a short-term incubation of cells with AA, during which prostaglandin products are expected to be formed, is sufficient to further destabilize COX-2. Like native huCOX-2, the N-terminal hexahistidine (His6)-tagged huCOX-2 was susceptible to AA-induced degradation that could be inhibited by NSAIDS (Figs. 28a and b). The IC50 values for the inhibition of huCOX—2 COX activity by flurbiprofen and NS- 398 are 0.51 M and 1.77 11M, respectively (271). I found that 10 M PB and 500 nM NS-398 significantly inhibited the degradation of Hisn-tagged huCOX-2 induced by 15 11M AA (Fig. 28b). Just like AA, the COX-2 polyunsaturated fatty acid (PUFA) substrates EPA and DHLA enhanced COX-2 degradation albeit at higher concentrations than AA (Figs. 28c and d, panel 11). In contrast, 2-A0 ether, a derivative of the COX-2 substrate 2-arachidonyl glycerol, and OA did not affect COX-2 protein turnover (Fig.28d, panels II and III). Similarly, LA and EDA which are known to be poor COX-2 substrates did not induce COX-2 degradation even at concentrations as high as 50-100 M 132 M 27A.(1) 0 2 4 12 24 2 4 12 24 (h) huCOX-2 , Actin (11) 20mm 0 2 4 12 2 4 12 (11) 1-3.1:. 7 «In; ,. ,._ 6‘ :2“.— mm: .v‘fiau? 'qu 4‘ «1595-612 i .t .,,5 ,3 huCOX-2 i; ”'u‘ “‘ . . I " mu: 9- ~_ ii". '11-'— main-um un- rub- tuna—Ind say i.-. m racy. aerrnxxri-Ir,.n.u.vv‘";’,q u. it: ‘- l‘ M 1‘ Actin i '1'” m .1: <5 #5:.rn'4 :2: ‘ .4 ’ c.41- -. Av (In) 10 111! AA 024122424122401) «a. _ "7 it '~ ‘ ‘ . I" I N594A huCOX-Z {mfi ” gird-d : . . T- ~ “le ‘ 2&1 _° , . 1' .y‘ °:"- .ru: » ‘3' 7 ' .,-:~— 4.1.1: ”3" "~52 Actin 133 27B. (1) 1 2 3 4 5 1. 24hinduction huCOX-Z «fl- 4.. 2. “ + 4hpuromycin 3. “ + “ +5pMAA 4. “ + “ +10pMAA Actin "fl““* 5, “ + “ +20IJMAA 120 2" E 100 :E': 80 h 60 = E 40 O 3: 20 e\° 0 (11M) B. (II) -1_.2....3 4 5. delS95-612 , 7 .,,. . ,_ 1 24hinduction huCOX-2 . T‘Vf‘fi" " ”' ° ' 2. “ + 4hpuromycin 2.. .. —.__.'.'.' _' v . -.- 1.: 3. u + ‘6 +5 "M AA ‘ ‘ " ' 4 “ + “ +10uMAA Actin 5 “ + “ +20llMAA 0 5 10 20 (11M) 134 27C. ' huCOX-2 . vl 1 '23.. 1‘ . I ‘, 4 , .' hi. . Actin 1 2 3 4 5 6 7 Densitometry (%)I 100 J 51.2 Il7.6 I 26.4 I 39.4 I 45.6 I 44.3 I 1. 24 h induction 2. “ + 4 h puromycin 3. “ + “ + 10 pM AA 4. “ + “ + “ + 20 nM M0132 5. “ + “ + “ + 50 11M R-FB 6. “ + “ + “ + 50 11M S-FB 7. “ + “ + “ + 100 M FB D. 1 2 3 4 5 6 ram. .... .. _. 2......“ . 5+4 ‘1. 'i ,,_ F I“. e- 4 an. on Iraq if 1". P_-rr....'.::_.!!.-.l 1. 24 h induction 2. “ + 4 h puromycin 3. “ + “ + 20 M AA 4. “ + “ + “ + 20 11M NS-398 5. “ + “ + “ + 100 “M FB 6. “ + “ + “ + 20 M M0132 135 27E. 1234567 delS95-612 ..._, .2 M . .... huCOX-2 l. 24hinduction . 2. “ + 4 h puromycin ACtIn ~~~hflflww 3. 66 + u + ZOILMAA ' 4. “ + “ + “ + 20uMMGl32 .. ..5. “ + “ + “ + 10011MFB N594A " 6. “ + “ + “ + 50 R-FB huCOX-z, ~-- ~- --«---~ ,7. « . .. + « + some. Aetin -"' 1"” *‘V W H W w. F. 1 2 3 4 5 6 hUCOX-Z ‘5‘“ race: ' q .4 ~_. Actin ilr“ E- V -:_'_. ‘.'. . i' 1.1322“ ’1 j. . l 2 3 4 5 6 Demimmetl'y (%) 100 78.3 50.7 77.0 36.3 37.4 1. 24h induction 2 “ + 4 h puromycin 3. “ + “ + 2011MAA 4. “ + “ + “ + 10011MFB 5 “ + 5minpuromycin+20pMAA; 1X PBS wash;4hpuromycin 6 6‘ + 66 + 66 66 o 66 + lmuMFB Figure 27. COX-2 degradation in HEK293 cells is enhanced by exogenous AA treatment. (A) After serum—starvation and challenge with tetracycline to induce expression HEK293 cells stably expressing native huCOX-2 (I), delS95-612 huCOX-2 (H), or N594A huCOX—2 (III) were treated with 50 p.M puromycin for the indicated times with or without the addition of 10 11M or 20 11M AA. Alternatively, the cells were treated for 4 h with puromycin, a combination of puromycin and different concentrations of AA (5 M, 10 11M, or 20 12M) (B), or a combination of puromycin, AA, and the indicated inhibitors (C, D, and E). (F) After a 24 h COX-2 induction with 10 jig/ml tetracycline cells were treated with 50 12M puromycin and 20 p.M AA for 5 min or 4 h in the presence or absence of 100 12M FB. Following the 5 min treatment with AA, cells were washed with PBS and treated for an additional 4 h with puromycin in the presence or absence of 100 “M PB. After the treatments in (A)-(F) protein levels were analyzed by Western blotting and densitometry analysis. The results in HO) and B(II) are representative of three independent experiments and the graph error bars denote 1 SE of the mean. The result in F is representative of two different experiments. 136 (Fig. 28d, panels I, II, and 111). Thus, COX-2 degradation in 293 cells is selectively induced by COX PUFA substrates. Collectively, these results suggest that substrate binding to the COX active site is a prerequisite for the substrate-dependent degradation of COX-2. 6533A huCOX-g is Refractory to Substrate-dependent Degrafltjgl. In a standard in vitro oxygen electrode assay, GS33A huCOX-2 has less than 5% of the COX specific activity of native enzyme with AA as substrate (17,272). We stably expressed 6533A huCOX-2 in HEK293 cells and analyzed its degradation profile after treatment with exogenous AA. Under basal conditions GS33A huCOX-2 degraded with a half-life that was similar to that of the native huCOX-2 (Fig. 29a). However, this mutant was resistant to degradation that was induced by COX substrate (Figs. 29b and c). Our findings suggest that a functional COX active site is required for substrate-induced degradation of COX-2. Substrate-dependent COX-2 Degragrtion in NIH/3T3 cells. We attempted to reproduce the enhancement of COX-2 degradation by fatty acid substrate in a different cell type. Serum-stimulation of quiescent 3T3 cells induced endogenous COX-2 degradation which was enhanced by treatment with 20 MM exogenous AA (Fig. 30a, panels I and II). Flurbiprofen significantly blocked both the endogenous degradation of COX-2 and AA-induced degradation of the protein (Fig. 30b). We also conducted experiments to determine if further stimulating endogenous AA release with bradykinin or A23187 in addition to serum-induction would also cause an enhancement of COX-2 degradation. In one experiment, treatment with 10 M A23187 modestly enhanced COX- 2 degradation albeit to a similar extent as treatment with 20 M AA (Fig. 30c). The enhancement of COX-2 degradation by both A23187 and AA was inhibited by NS-398. 137 28A. MA... AA. _AA+FB_ AA+NS-398 024 24 24,214 a.) HisshuCOX-Z a...” . “A w B. l 2 3 4 5 6 7 3 His; huCOX-2 ~n « ‘- 3 , fl.“ 1. tet-induced 2 “ + 4 h puromycin 3 “ + “ + 20 “M AA 4. “ + “ + “ + 10 pM FB 5. u + ‘6 + “ + 30 ”M FB 6 u + u + u + 100 [M FB 7 “ + “ + “ + 500 nM NS-398 s “ + n + “ + 20 M NS-398 C. AGE)— EPA q 2 __ 10 100 7, , 2 ‘10 50. 100 His, huCOX-Z.“ I" ,_ . _fi, _. ...., ..,_ L‘._‘:.-.,;J_-_.... 2 - _ __ _1.. Actin ‘. EPA 9‘ Rundnlng o 2 10 100(uM) o 2 10 50100 (m) 138 28D (1) AA LA 0 3.75 7.5 15 100 3.75 7.5 15 100 (11M) o 3.75 7.5 15 100 3.75 75 15 10? (pM) FDensitometry(%) I 100 I 75.2 I 39.7 I 43.7 I 31.6 I105.3 I100.0I123.7I105.7I D(II) AA DHLA EDA 2-AG ether ,0 5 20 2_ .10 so 100 10 5010010 50 (11M) HisshuCOX-Zhwg . ‘1.“ 1‘2?" rfl‘fl‘ an a a .1“ AA I DHLA EDA IL? 2-AG ethe'l' 0 {s 20‘6 2 10 so 100\ [10 so 100\ ’10 501 (11M) IDensitometry(%)I 100I 48.2 I 17.3 I 68.5 I795 50.0 I58.0I 67.1 I 74.1 I 63.2 I 76.2 I 84.7 I III 2-AG ether EDA 0A ( ) 0 2 5 10 20. 2 10 50 100 2 10 50 100 (11M) ';’_‘1 . . .1 . . .7, .4 . . _...“_. ___._.. __. r . fa» 2-AG ether EDA 0A o 6 s 10 20\ f o 10? G 1 50 R Densitometry(%)I 100I 126.l I 132.3I 111.7 I115.6I94.9 I89'9t‘7'3 163.7 I 146.2 I 145.0 I 110.3I12:7“IM Figure 28. COX-2 degradation is selectively induced by PUFA COX substrates for the enzyme. N-terminal Hiss-tagged huCOX-Z-expressing HEK293 cells were treated as in 27(A) to induce protein expression. (A) Cells were treated for the indicated times with 50 11M puromycin with or without 20 11M AA, or a combination of puromcyin, AA, and the indicated concentrations of F8 or NS398. (B) After induction of His huCOX-2 (1), cells were treated for 4 h with 50 [AM puromycin only (2), or in combination with 15 11M AA (3) and 10 M PB (4), 30 11M FB (5), 100 [AM FB (6), 500 nM NS-398 (7), or 20 M NS-398 (8). In (C) and (D) puromycin treatment was followed by treatment with different concentrations of AA, linoleic acid (LA), eicosapentaenoic acid (EPA), 2-arachidonyl glycerol ether (2-AG ether), dihomo-Y-linolenic acid (DHLA), eicosadienoic acid (EDA), and oleic acid (0A). COX-2 protein levels were analyzed by Western blotting and densitometry. The result in C is representative of at least three independent experiments and the graph error bars denote 1 SE of the mean. 139 29A. 20 AA 0 2 4 12 24 2 4 12724 (b) 1' ..:r _. a. - an n a. .1 . —~ 1:: a. 1,3- ,. u- u. G533A huCOX-2 "u .1. i— q... w I . U" U .11 a“- B- 1 2 3 4 5 f 3‘" i " i ' "9‘3 1. 24hinduction G533A huCOX-2 I P “h w w . i 2. “ + 4 h puromycin r... ‘11" ‘ , #7:)": am; m1 3, “ + “ + 5 “1M AA "11‘ *—‘ 1 ‘ 4. “ + “ + lOuMAA Actin flflkg‘flal 5. “ + “ +20uMAA C. % Protein remaining @ Native COX-2 l G533A cox-2 "' 0 5 10 20 (11M AA) Figure 29. The G533A cyclooxygenase-inactive COX-2 mutant is not susceptible to substrate-dependent degradation. (A) Degradation of GS33A huCOX—2 was examined with or without addition of 20 11M AA. (B) Cells expressing G533A huCOX-2 were treated for 4 h with 50 pLM puromycin plus different concentrations of AA (5 11M, 10 11M, or 20 1.1M). Protein levels were analyzed by Western blotting. (C) Densitometry was performed on the levels of native huCOX-2 or G533A huCOX-2 and normalized to those of actin. Densitometry analysis for each protein is based on at least three independent experiments. The error bars denote 1 SE of the mean. In contrast, bradykinin at 10 MM did not facilitate COX-2 protein turnover. It is possible that A23187 may cause more release of endogenous AA than bradykinin and that the amount of bradykinin-induced free AA may not be sufficient to facilitate COX-2 degradation in serum-stimulated 3T3 cells. SubstLate-dependent Degra_dz_1tion of COX-1. COX-l is known to be a very stable protein (257). This is consistent with the observation that NIH/3T3 cells that have been made quiescent by serum deprivation for 48 h continue to express the enzyme. Furthermore, stimulating quiescent 3T3 cells with serum will induce COX-1 mRNA (22). However, serum-stimulation of 3T3 cells in the presence of flurbiprofen does not lead to an accumulation of COX-1 protein. Collectively, these results imply that the constitutive expression of this COX isoform observed in many tissues is likely to be mainly due to the stability of the enzyme rather than a continuous balance of protein synthesis and degradation. Experiments were performed to determine if COX-l protein turnover could be induced by COX fatty acid substrate. In NIH/3T3 cells, treatment with 20 11M AA did not affect the stability of endogenous COX-1 protein (Fig. 31a). This was a surprising result because treatment of quiescent 3T3 cells with 10 p.M AA results in prostanoid product formation due to COX-1 activity. Therefore, at the AA concentrations that were used in our experiments COX-1 is catalyzing oxygenation. In HEK293 cells, treatment with _>_20 11M of AA resulted in a modest loss of COX-1 protein (Figs. 31b and c). If indeed COX-2 degradation due to AA challenge is a consequence of suicide inactivation during COX catalysis, it is possible that COX-l may be more resistant to COX substrate- induced inactivation in vivo compared to COX-2. Alternatively, the inactive form(s) of COX-1 formed during COX catalysis may be less susceptible to degradation. 141 30A. (1) 20”,“, 0 0.5 l 2 0.5 l 2 Time with puromycin (h) COX-2 Actin $6 Protein rornalnlng I F 0 0.5 1 1.5 2 2.5 Tlm 0 after purom yeln (h) ._. No substrate I-——-I zopMAA 142 30A. (11) MA 0 1 2 1 2 Time with CHX (h) COX-2 W Irv-‘1 g,» Actin we: , ,, “a, .1. 1..» 120 100 O .E .E 80. C E .0. E ‘0 s 40‘ \ a2 20. i— _ _ _l 0 . fi _ fl 0 0.5 1 1.5 2 2.5 Time after CHX (h) P * No Substrate F—_—. 201LMAA 143 30B. _h N O _A O o 804 60- % Protein remainlng 40- 20- 0 0:5 1 1:5 2 2.5 Tlme after puromycin (h) H No substrate H 20 11M AA l———-I ZOuMAA+100uMFB A---i 100uMFB M1 '2 3 4 5 6 7 8 9 30C. .3. ..___ 3................ .._3 COX-2 Ihufi. «it a“. Actin 1- w r 2 3 4 5 6 7 s 9 Densitometry(°/ o) 100 73 80 87 94 62 76 59 80 Figure 30. The enhancement of COX-2 degradation in NIH/3T3 cells by exogenous and endogenous AA is inhibited by NSAIDS. (A and B) Serum-starved, quiescent 3T3 cells were serum-stimulated for 4 h to induce COX-2 expression. Thereafter, cells were treated with 50 11M puromycin or 50 pM CHX for the indicated times in the presence or absence of 20 MM AA, 100 M FB, or a combination of both AA and PB. After Western analysis, densitometry was performed on the levels of COX-2 and normalized to those of actin. (C) After inducing COX-2 expression (1) as in A and B, cells were treated with 50 12M CHX (2) in the presence or absence of the following: 20 M NS-398 (3), 10 11M bradykinin (4) , 10 M bradykinin + 20 [AM NS398 (5), 10 11M A23187 (6), 10 |.l.M A23187 + 20 uM NS-398 (7), 20 11M AA (8), or 20 11M AA + 20 [AM NS-398 (9). Densitometry was performed on COX-2 protein levels and normalized to those of actin. 31A. 20 AA 0 l 2 l 2 Time with puromycin (h) cox-1 m5} “'3 :4 3‘32. A 20 EM AA 0 1 2 1 2 Timewith CHX (h) COX'I . , ,3, ‘ _..- “'3‘ “*0:- Av we 4‘! ~- '9. 9..-.“ *M“ 33.... Actrn .. ,3. 3.... ,3. ...., .3... 145 31B. 0 2 10 25 50 (uMAA) ovCOX—l ACtin 9""?! $31-an ~3: ~13 . -e~. $3.3: 0 2 10 25 50 Densitometry (%) 100 81 78 74 69 C3 120 100 L; % .‘s‘ T ‘ ~ 5 80- \ ‘ 0 ‘ Em i 3' 0 4° ‘ 32 20 i 0 . 0 2 4 6 Time (h) .__. No substrate '- — " '" i 20 [1M AA (LAM AA) Figure 31. COX-1 stability is not significantly affected by exogenous AA. (A) Quiescent 3T3 cells were serum-stimulated for 4 h then treated with 50 MM puromycin or 50 M CHX for the indicated times with or without the addition of 20 [1M AA. (B) 293 cells stably expressing ovCOX-l were treated with 50 11M puromycin for 4 h with or without the addition of different concentrations of AA. (C) Densitometry of the effect of 20 mM AA on ovCOX—l stability in 293 cells. Quantitative analysis is based on six independent experiments. The graph error bars denote 1 SE of the mean. 146 Substrate-dependent Degradation of COX-2 is not Proteasome- or Lvsosome— dexndent. We tested an inhibitor of the 26S proteasome and selective inhibitors of lysosomal degradation in an attempt to identify the pathway responsible for substrate- dependent COX-2 protein turnover. The 268 proteasome inhibitor M0132 failed to inhibit substrate-induced COX-2 degradation in both 3T3 and 293 cells (Figs. 27c, 27d, 27e, and 32a). Similarly, the lysosomal degradation inhibitors E64 and leupeptin, and brefeldin A, an inhibitor of ER to Golgi trafficking, failed to prevent AA-induced COX-2 degradation in 293 cells (Figs. 32a and b). Therefore, substrate-dependent degradation of COX-2 does not appear to require the proteolytic activity of the proteasome or lysosome. Experiments were carried out to determine if the protease(s) responsible for cleaving COX-2 in a substrate-dependent manner was ER-associated. Microsomes prepared from 293 cells expressing delS95-612 huCOX—Z or N594A huCOX-2 were incubated with or without 100 “M AA in a reaction buffer containing 10 uM hematin and 1 mM phenol. After 4 h at 37°C the samples were analyzed by Western blotting to determine whether there was loss of COX-2 protein as a consequence of substrate treatment. There was no observable change in the levels of delS95-612 huCOX-2 or N594A huCOX-Z in microsomes that were exposed to AA (Fig. 32c). It is possible that the microsomes that we prepared were not sufficiently intact to retain any ER luminal protease(s) responsible for COX-2 proteolytic cleavage. To test the integrity of the microsomes, I treated solubilized or non-solubilized microsomes with trypsin. Limited digestion of COX-2 with trypsin yields a single cleavage at its C-terminus that produces a ~2-3 kDa C-terminal fragment. The C-terminal trypsin cleavage site is within the epitope for the primary antibody used to detect COX-2. Since COX-2 is an ER luminal protein it 147 should not be cleaved by trypsin if the microsomes are intact. However, both delS95-612 huCOX-Z and N594A huCOX-2 were proteolytically cleaved by trypsin in non- solubilized microsomes (Fig. 32d). These results indicated that the prepared microsomes were permeable and/or wrong-side-out. Therefore, we cannot eliminate the possibility that there could be an ER-associated protease involved in substrate-dependent COX-2 degradation. The protease is likely to be a luminal enzyme or it may be ER membrane- associated but requires luminal co-factors for its catalytic activity. COX Self-inactivation Preceeds Substgte-depenggnt COX-2 Degradation. Both COX-1 and COX-2 are known to undergo irreversible, mechanism-based COX inactivation during in vitro catalysis (3,156). It is possible that the self-inactivation of COX-2 in HEK293 cells and NIH/3T3 cells treated with AA would accompany the formation of cyclooxygenase oxygenation products. If indeed the enzyme self-inactivates in vivo during catalysis, it would reflect damage at the COX active site. Structurally damaged COX-2 would likely be more susceptible to degradation than the native enzyme, which could explain why AA treatment facilitates COX-2 protein turnover in HEK293 and NIH/3T3 cells. I decided to examine whether COX-1 and COX-2 undergo self- inactivation in vivo during COX catalysis. For this purpose, I utilized HEK293 cells stably expressing N-terminal His-tagged versions of ovCOX-l (His ovCOX-l) or murine COX-2 (His muCOX-2). These cells were treated for 10 min with varying concentrations of AA (0, 2.5, 10, 25, and 50 MM). Thereafter, the cells were washed with PBS, pH 7.4 and centrifuged at 2000 rpm for 3 min. Cell pellets were resuspended in 0.1 M Tris, pH 7.4 and sonicated. The COX activity of the whole cell lysates was then assayed using an 148 32A. delS95-6l2 . . huCOX-Z tet-mductlon “ + 4 h puromycin Actin “ + “ +20pMAA “ + “ + “ +50 Leu N594A “ + “ + “ +25 ”um E64p huCOX-2 “ + “ + “ +20 uMMGl32 “ + “ + “ +20 pMNS-398 Actin F3 B. huCOX-2 Actin qr “WWT‘CF ._. “ delS95-612 £533; 33. ' ”‘1. .' f3 huCOX-2 If. ‘ "5 J _ “i h ' 1" aygffiéézsfi .1 A . 5;?1'riaj-‘fifnf‘rfi-‘fi‘g Actin fififlt-flg 3 ,3. .... 3-..“, ,3 4 ‘_l l. tet-induction 2. “ + 4 h puromycin 3. “ + “ + 20 [1M AA 4. “ + “ + “ + 5 ug/ml BFA 5. “ + “ + “ + 5 ug/ml BFA 6. “ + “ + “ +20uMNS-398 149 32C. 123456 N594A ""” ' -‘ 71 huCOX-2 Eh," I TV, “ ' o r ' 1 :ng93X6-1223 "HHHH— 3 1,4. No substrate "7 2,5.100 [AM AA 3,6.100uMAA-l-100uMFB 32D. 1 2 3 4 3......3 _3 .3__... “.3. l. unsolubilized N594A huCOX-2 ~ .. r 2. unsolubilized + trypsin tmt. . 7 1 ' . 3. solubilized 4. solubilized + trypsin tmt. Figure 32. Inhibitors of proteasomal and lysosomal degradation do not prevent substrate-dependent COX-2 protein turnover. (A) HEK293 cells stably expressing delS95-612 huCOX-2 and N594A huCOX-2 were treated as in 27A to induce protein expression (1). Thereafter, cells were treated for 4 h with 50 pM puromycin (2) or in combination with 20 pM AA (3), 20 pM AA + 50 pM leupeptin (4), 20 pM AA + 25 pM E64 (5), 20 pM AA + 20 pM MGl32 (6), or 20 pM AA + 20 pM NS398 (7). (B) Cells stably expressing native huCOX—2 and delS95-612 huCOX-2 were treated as in 27A to induce protein expression (1). This was followed by treatment with 50 pM puromycin only (2) or in combination with 20 pM AA (3), 20 pM AA + 5 pg/ml brefeldin A (4), 20 pM AA + 5 pg/ml brefeldin A (5), or 20 pM AA + 20 pM NSB98 (6). (C) Microsomes were prepared from 293 cells stably expressing delS95-612 huCOX-2 or N594A huCOX- 2 as described in the text. Thereafter, the microsomes were resuspended in 0.1 M Tris-Cl buffer, pH 8.0 and 200 pg of this resuspension was incubated in a 100 pl reaction mixture containing 0.1 M Tris-Cl, pH 8.0, 10 pM hematin, and 1 mM phenol with no substrate (1,4) or with 100 pM AA (2,5) and 100 pM FB (3,6). The incubation was performed at 37°C for 4 h. (D) After resuspension, 100 pg of microsomes were treated with 5 pg trypsin before or after solubilization with 1% Tween-20. Trypsin treatment was performed at 37°C for 20 min. After the treatments in A-D protein levels were analyzed by Western blotting. 150 oxygen electrode. Microsomes were also prepared from the whole cell lystates and assayed for prostaglandin product formation by radioactive TLC. There was a dramatic concentration-dependent decrease in COX activity of His6-tagged muCOX-Z-expressing whole cell lysates (Fig. 33a), which could not be a result of a decline in His6-tagged muCOX—2 protein levels during the 10 min duration of AA pretreatment (data not shown). In contrast, the decline in COX activity observed with Hisé—tagged ovCOX-l- expressing HEK293 cells was less dramatic (Fig. 33a). For instance, His6-tagged muCOX-2-expressing lysates pretreated with 10 pM had a ~40% decrease in specific activity compared to only ~20% for Hing-tagged ovCOX-l-expressing lysates pretreated with the same concentration of AA. The observations made with the oxygen electrode assays are consistent with results from radioactive COX TLC assays which demonstrated that prostaglandin product formation of Hisa-tagged muCOX-Z-expressing microsomes prepared from cells that were treated with 10 pM or 25 pM AA was significantly reduced (Fig. 33b). Overall, these findings indicate that, at least in HEK293 cells, both COX-1 and COX-2 undergo substrate-induced catalytic inactivation with COX-2 appearing to be the more susceptible isoform. 151 33A. His ovCOX-l His mucox-z spadflc athy (nmnlIn-Inlrm lynx.) apodflc adlvlty (mrnllninlflu In“) mPGES—l + GSH B. AA.» HETE‘ _ W . HHI"~ *‘I-“ww PGE2” < 5 5 5 < < I ‘ 5 5 < a a. Pretreatment -5 g 1 I: O o '0 ~ 1 2 3 4 5 6 Figure 33. COX activity and prostaglandin product formation of His ovCOX-l and His muCOX—2 cells pretreated with exogenous AA. Upon induction of His ovCOX-l or His muCOX-Z, cultured 293 cells stably expressing these enzymes were incubated with the indicated concentrations of AA for 10 min. Afterwards, the cells were washed with 1X PBS, harvested and centrifuged at 2000 rpm for 3 min. Cell pellets were resuspended in 0.1 M Tris-Cl, pH 8.0 and lysed by sonication. (A) The COX activity of the whole cell lysates was measured by an oxygen electrode assay in a standard reaction mixture containing 100 pM AA, 1 mM phenol, and 5 pM hematin dissolved in 0.1 M Tris-Cl, pH 8.0. (B) Microsomes were isolated from the whole cell lysates pretreated with 0 pM (1, 4), 10 pM (2, 5) or 25 pM AA (3, 6) and prepared for radioactive TLC as described in the text. Equivalent amounts of microsomes (0.5 mg) were incubated in a IOO-pl reaction mixture containing 0.1 M Tris-HCI, pH 8.0, 1 mM phenol, 10 pM hematin, and 10 pM [1-“C]arachidonic acid at room temperature for 1 min. To assay for PGE2 formation microsomal PGES-l (mPGES-l) and 60 pM GSH were included in reaction samples 4, 5 and 6. Afier the reactions were terminated as described in the text the radioactive products were isolated, resolved by TLC, and analyzed by autoradiography. HETE—hydroxyeicosatetraenoic acid; HHT — hydroxyheptadecatrienoic acid. 152 Discussion The secondary and tertiary structures of COX-2 are more susceptible to guadinium hydrochloride chemical denaturation compared with COX-1, demonstrating a difference in the inherent structural stabilities of the COX isoforms (150). This difference in protein stability parallels that observed in viva where COX-l is very stable under conditions in which COX-2 is rapidly degraded (257). We have previously attributed the short protein half-life of COX-2 to a unique C-terminal l9-amino acid cassette (l9-aa) that is required for ER-associated degradation of the enzyme in HEK293 cells. In the present study we have shown that COX-2 undergoes a second distinct form of degradation that is induced by COX fatty acid substrate and is independent of the l9-aa. We also demonstrate that the COX substrate-induced degradation of COX-2 requires a functional COX active site because it can be blocked by: (a) both competitive and time- dependent, irreversible COX inhibitors, and (b) a G533A point mutation that inactivates COX catalysis. That the substrate-induced degradation of COX-2 is dependent on substrate binding and turnover at the COX active site is suggested by the finding that only known COX-2 COX fatty acid substrates are able to facilitate COX-2 protein turnover. Furthermore, a 5 min brief challenge of COX-Z-expressing cells with AA, during which prostaglandin products are expected to be formed, is sufficient to significantly enhance COX-2 degradation. Overall, these findings enable us to postulate that COX substrate-dependent degradation of COX-2 is initiated at the COX active site by the binding of fatty acid substrate. It is not clear whether the inactivity observed with GS33A COX-2 is due to a defect in substrate binding or substrate turnover. This point mutant has some residual COX activity with AA as substrate (~5% relative to the native 153 enzyme) (17,272) suggesting that it may bind AA but inefficiently bis-oxygenate the substrate to form PGGz. Proteasome activity appears to be dispensable for the COX substrate-dependent degradation of COX-2. We have previously shown that COX-2 degradation in NIH/3T3 cells is partly proteasome-independent in NIH/3T3 cells, but largely proteasome- dependent in HEK293 cells (257). Also noteworthy is the finding that COX inhibitors dramatically prevented the basal degradation of COX-2 in serum-induced NIH/3T3 cells but did not affect the proteasome-dependent degradation of the enzyme in HEK293 cells. Collectively, these findings suggest that COX inhibitors may not inhibit the proteasome- dependent degradation of COX-2 that is initiated by the C-terminal 27 amino acid instability element. Therefore, there appears to be a second distinct mechanism for COX- 2 protein turnover present in NIH/3T3 cells that is triggered by the endogenous release of AA. HEK293 cells lack detectable phospholipase A2 activity whereas NIH/3T3 cells can activate cPLAmI in response to serum treatment (33,58,77). This could explain why COX inhibitors do not retard the basal, proteasome-dependent degradation of COX-2 in HEK293 cells. In NIH/3T3 cells, endogenously released AA will be utilized as COX substrate by COX-2, thereby, initiating the substrate-dependent degradation of the enzyme. Addition of exogenous free AA or A23187, but not bradykinin, further enhanced the NSAID-inhibitable degradation of COX-2 in serum-treated 3T3 cells. In this regard, it is possible that a copious amount of endogenous AA was released upon serum- stimulation that was sufficient to induce substantial degradation of COX-2, so that adding an additional stimulus like bradykinin did not dramatically affect COX-2 protein levels. It would be interesting to determine if the effect of bradykinin and A23187 on COX-2 154 degradation can be amplified in 3T3 cells that have been induced to express COX-2 with phorbol ester instead of serum. Since phorbol ester is not a major activator of cPLAza in fibroblast cells it should induce a relatively smaller release of AA compared to serum resulting in a proportionate attenuation of the endogenous COX-2 degradation. If indeed endogenous COX-2 degradation is partly induced by cPLAz-mobilized AA in serum- treated NIH/3T3 cells, it would also be reasonable to test the effect of cPLAz inhibition on the degradation of the protein. COX-1 and COX-2 undergo irreversible suicide inactivation during COX catalysis under cell-free conditions (3,156,162,163). Whether or not these enzymes undergo suicide inactivation in intact cells has not yet been clarified. Riese et al. have provided evidence to suggest that COX-2 may inactivate in RAW264.7 macrophage cells that have been challenged with a combination of LPS and IFN-y (273). In their study, they observed that while COX-2 expression levels remained at maximal levels 24—48 h after induction with LPS and IFN-y , the specific activity of the enzyme was transient during this time period. In vitro COX inactivation is accompanied by significant changes in the structural conformation of the enzyme, suggesting that this process is a culmination of the destruction of the native structure of heme-protein complex (162). The concept of inactivation-coupled structural damage due to COX catalysis becomes important in the context of COX protein stability since it is well known that the ER has a sophisticated quality control system that recognizes and selectively eliminates structurally-damaged ER-associated proteins (164-166,182-186). In the present study, evidence has been provided to show that both COX-1 and -2 can inactivate in intact HEK293 cells upon exposure to exogenous AA. From this finding we propose that the inactivation-coupled 155 structural damage to COX-2 as a consequence of substrate turnover at the COX active site precedes the substrate-dependent degradation of the enzyme (Fig. 34). We are yet to determine whether this model is also applicable to COX-1. It is interesting to observe that although the COX substrate-dependent degradation of COX-1 in HEK293 is relatively modest compared to COX-2, it appears to be proportionate to its COX substrate-induced inactivation. HE K293 NIH/3T3 FTPII'RCFCIIH? challengej [ Serum stimulation 1 2 Induction of (0&2 LCOX-Z induction 1 ELA2 activation 1 FCPLA, lnhlbltors?? heterologous expression 1 A Mobilization of free . . f g endogenous AA , Basal ( OX-Z 1 “F I ERAD ‘ x Proteolysis by l \‘n v 1 "‘ - 1 ; -65 proteasome .‘ [COX 2 (‘OX ”my,“ I |__ NSAIDS stem—4 Inactivation induced structural damage to cyclooxygenase site Proteasome independent degradation Figure 34. Model for the basal and COX substrate-initiated degradation of COX-2 in HEK293 (red) and serum-treated NIH/3T3 cells (blue). Proteasome-dependent glycoprotein ERAD is the predominant pathway for COX-2 degradation in HEK293 cells which essentially lack phospholipase A2 (PLA2) activity. Addition of exogenous AA to COX-Z-expressing HEK293 cells will activate a second distinct proteasome-independent, NSAID-inhibitable degradation pathway that involves COX catalysis. The degradation of endogenous inducible COX—2 in serum-treated NIH/3T3 cells may occur via both pathways. If COX-2 degradation in these cells is further enhanced by endogenous AA release, then phospholipase A2 inhibitors should stabilize the protein to the same extent as NSAIDS. 156 It seems unlikely that the COX substrate-dependent loss of COX-2 protein as determined by immunoblotting is due to the formation of COX reactive oxygenated side products that covalently modify a region(s) of the COX-2 protein that serves as an epitope for antibody detection. In examining the effect of COX substrate on the stability of COX-2 we utilized two different antibodies for immunoblotting. In NIH/3T3 cells a peptide-directed antibody against the epitope SS98-K612 of muCOX-2 was used to detect serum-induced COX-2. In HEK293 cells an antibody against Q583-N594 of huCOX-2 was used to detect tetracycline-inducible stably expressed COX-2. Both antibodies yielded similar results even though they are directed against different epitopes on the protein. Therefore, we believe that the substrate-dependent decrease in COX-2 immunoreactivity in both cell lines reflects the degradation of the enzyme. We were unable to identify the degradation pathway or the protease(s) responsible for the substrate-induced proteolytic cleavage of COX-2. We have found that the proteolytic activities of the 26S proteasome and the lysosome, and trafficking between the ER and Golgi appear to be dispensable for this process. It is likely that the protease(s) involved in substrate-induced cleavage of COX-2 may be resident in the ER. It may not have been possible to identify the ER- or microsome-associated proteolytic activity because the microsomes that were used in our studies were not intact; trypsin cleavage analysis suggested that the microsomes were likely to be wrong-side-out. Very few enzymes have been hitherto identified as ER-resident proteases. Signal peptidase, which is known to be physically associated with the Sec6l translocon, cleaves N-terminal ER targeting sequences of proteins as they are imported into the ER (274- 276). Signal peptide peptidase (SPP) is an ER membrane-associated aspartyl protease that 157 cleaves the signal peptide fragments generated by signal peptidase (277). Recently, SPP has been shown to be essential for the retrotranslocation and subsequent proteasomal degradation of major histocompatibility complex (MHC) Class I heavy chains that is induced by the US2 protein of cytomegalovirus (278). However, this study did not clarify whether the protease activity of SPP per se was required for this process. ER aminopeptidases-l and -2 (ERAP-l and -2) are ER luminal, interferon-y—inducible metallopeptidases that are believed to be involved in the final proteolytic processing of short proteasome-generated peptide fragments that will be loaded onto MHC Class I molecules for antigen presentation on the surface of an antigen presenting cell (279-282). There are no reports thus far implicating ERAP-l and -2 in the degradation of whole proteins. The membrane remodeling enzyme A9 steroyl-CoA desaturase is an ER integral membrane protein that is rapidly degraded (tug ~3-4 h) under basal conditions in a proteasome-independent manner (283,284). Heinemann et al have purified and characterized a 90 kDa plasminogen-related protease with Arg/Lys specificity that it is responsible for the rapid degradation of steroyl-CoA desaturase (284,285). This protease is unlikely to act in the ER lumen because the sequence determinant of steroyl-CoA desaturase that directs its rapid turnover is found on the cytosolic N-terminal portion of the enzyme (286). Although the mechanism for steroyl-CoA proteolysis is still unclear, there is evidence to show that the protein gets cleaved within a putative membrane binding region, most likely by the ER plasminogen-related protease (283). It has been suggested that the N-terminal instability sequence of steroyl-CoA desaturase may help to bring the membrane protease to close vicinity with the enzyme (286). Recently, Donoso et al. have shown that the degradation of a misfolded mutant form of BiP is initiated in 158 the ER lumen by a serine protease that is yet to be identified (287). In this regard, it is likely that a proteolytic system exists in the ER that may serve as an alternative or a complement to the ERAD-proteasome pathway in the quality control of structurally defective ER-associated proteins. Therefore, we speculate that the catalytically inactivated form(s) of COX-2 is degraded by this putative ER protease system. In conclusion, we have found that AA, the principal PUFA COX-2 substrate, induces both the COX catalytic inactivation and the rapid degradation of the enzyme. We have also observed that inhibition of COX-2 COX catalysis using site-directed mutagenesis and pharmacological approaches prevent AA-dependent degradation. Based on these observations we have proposed that the AA-dependent degradation of COX-2 is a consequence of structural damage to the COX active site. In the future, it will be important to measure and compare the kinetic constants for the catalytic inactivation of the COX isoforms in intact cells. The proposition that COX-2 degradation in NIH/3T3 cells is enhanced by serum-induced mobilization of free AA will need to be tested further using inhibitors of cPLAz. A protocol for preparing intact microsomes will also be designed in an effort to identify the ER-associated protease(s) that degrades catalytically inactive COX-2. Indeed, the unconventional COX substrate-dependent quality control of COX-2 may serve as an additional physiological mechanism for the metabolic regulation of prostanoid synthesis. 159 CONCLUSION The structural basis for the selection of normal short-lived proteins for rapid proteolysis in an intracellular environment where they coexist with many stable proteins is not clearly defined. We have identified a 27 amino acid instability element close to the C-terminal end of COX-2 that regulates the proteasomal degradation of this ER-resident membrane-bound protein by mediating its entry into the ERAD system. We believe that COX-2 is the only known ER luminal integral membrane protein that is degraded via ERAD from its native or properly folded form. The ERAD pathway is an important protein quality control pathway that is not well characterized in mammalian cells. ERAD components in the ER lumen that participate in the initial selective recognition of an ERAD substrate and the mechanism by which they do so are not clearly defined. The identity of the ER membrane retrotranslocon is also yet to be determined. Therefore, since COX-2 is one of the very few mammalian proteins that are known to undergo proteasomal degradation from the ER it could serve as a suitable reporter substrate for studying ERAD in a mammalian system. We have also identified a second distinct mechanism for COX-2 degradation that is preceded by the COX substrate (AA)-induced suicide inactivation of the enzyme. Intracellular COX-1 appears to be less susceptible to substrate-induced suicide inactivation and degradation. COX-l is a stable protein that is constitutively expressed in many cells. It is also able to utilize endogenous AA at higher concentrations than COX-2. Therefore, based on our experimental data we reason that the housekeeping function of COX-1 might extend to include regulating the intracellular levels of free AA. The proteolysis of COX-2 in response to two distinct signals, namely a structural and a 160 metabolic signal, reflects the tight and elaborate control of COX-2 expression at all three levels of gene regulation. Since the differential expression profiles of COX-1 and COX-2 are thought to contribute to their specialized biological functions, our observations may provide a clue to the unsolved mystery of why mammals have two COX isoforms. 161 APPENDIX Table 3. C-terminal mutants designed by QuickChangeTM site-directed mutagenesis Mutation Primers Template G533 A Forward: 5 ’CT CCTT GAAAGCACTT A huCQX-2 hUCOX-2 TGGGTAATG 3’ Reverse: 5’CATTACCCATAAGTGCTTT CAAGGAG 3’ N594 A Forward: 5’ GTCACCATCGCAGCAAGTT huCOX-2 huCOX-2 CTTCCCGC 3’ Reverse: 5’ GCGGGAAGAACTTGCTG CGATGGTGAC 3’ P607 A T608 A Forward: 5’ CTA GAT GAT ATC AAT GCA huCOX_2 huCOX-2 GCA GTA CT A CTA AAA GAA 3’ Reverse: 5’ 'I'I’C 'I'I'I‘ TAG TAG TAC TGC TGC ATT GAT ATC ATC TAG 3’ V609 A Forward: 5’ GAT GAT ATC AAT CCC ACA huCQX-2 huCOX-2 GCA CT A CTA AAA GAA CGT 3’ Reverse: 5’ ACG TTC T'I'I‘ TAG TAG TGC TGT GGG A'I'I’ GAT ATC ATC 3’ L610 A Forward: 5’ GAT ATC AAT CCC ACA GTA huCOX-2 hUCOX-2 GCA CT A AAA GAA CGT TCG 3’ Reverse: 5’ CGA ACG TTC 'I'I'I‘ TAG TGC TAC TGT GGG ATT GAT ATC 3’ 162 Table 3 (cont’d) Reverse: 5"I‘TCAGTCGAACGTTCA TTGATATCATC 3’ L61 1A Forward: 5’ ATC AAT CCC ACA GTA CT A huCOX-2 hUCOX-2 GCA AAA GAA CGT TCG ACT 3’ Reverse: 5’ AGT CGA ACG TTC TTT TGC TAG TAC TGT GGG ATT GAT 3’ . K6 1 2A Forward: 5 ’ CCCACAGTACTACT A huCOX-2 hUCOX-2 GCAGAACGTTCGACT 3' Reverse: 5’ AGTCGAACGTTCTGCTAGT AGTACTGTGGG 3’ dd 5 9 5-6 1 2 Forward: 5 ’ AC AGTCACCATCAAT huCQX-2 huCOX-2 GAACGTTCGAC 3’ Reverse: 5’ CT ACAG'I'I’CAGTC GAACGTTCATTG 3’ (131597-61 2 Forward: 5 ’ ACCATCAATGCA huCOX-2 huCOX-2 AGTGAACGTTCGACT 3’ Reverse: 5’ AGTCGAACGTTCACTTGCATT GATGGTGAC 3’ de1602-61 2 Forward: 5 ’CTTCCCGCTCCGGAG huCQX-2 hUCOX-2 AACGTTCGACTGAA 3’ Reverse: 5’ TTCAGTCGAACG'I'I’CT C CGGAGCGGGAAG 3’ (131607 -6 1 2 Forward: 5 ’GATGATATCAAT huCOX_2 hUCOX-2 GAACGTTCGACTGAA 3’ 163 Table 3 (cont’d) MuCOX-2 Forward: 5’ AGT GT'I’ CCA GAT CCA CAG huCQX-2 UPSTRM8 CCT ACC AAA ACA GCC ACC ATC AAT huCOX-2 GCA AGT 3’ Reverse: 5’ ACT TGC ATT GAT GGT GGC TGT TTT GGT AGG CTG TGG ATC TGG AAC ACT 3’ ovCOX- 1 Forward: 5' AGT GTT CCA GAT CCA CGT huCOX-2 UPSTRM8 CAG GAG GAC AGG CCT GGG GTG AAT h COX 2 GCA AGT TCT TCC 3’ u ' Reverse: 5’ GGA AGA ACT TGC ATT CAC CCC AGG CCT GTC CT C CT G ACG TGG ATC TGG AAC ACT 3’ V591P T592G Forward: 5’ GAG CTC ATT AAA ACA CCC huCOX-2 huCOX-2 GGC ATC AAT GCA AGT TCT 3’ Reverse: 5’ AGA ACT TGC ATT GAT GCC GGG TGT TTT AAT GAG CTC 3’ insS94-596 Forward: 5’ GACAGGCCTGGGGTGGAGA ovCOX-l ovCOX-l ATGCAAGTCGGCCACCCACA 3' Reverse: 5’ TGTGGGTGGCCGACTTGCA TTCTCCACCCCAGGCCTGTC 3’ {113594-601 Forward: 5’ TCCCGCTCCGGAGAGCGG in3594-612 ovCOX-l CCACCC 3’ ovCOX-l Reverse: 5’ GGGTGGCCGCTCTCCGGAGC GGGA 3’ in5594-612 Forward: 5 ’ GACAGGCCTGGGGTGGCAGC in5594-612 (N594A) AAGTTCTTCCCGC 3’ OVCOX-l ovCOX-l Reverse: 5’ GCGGGAAGAACTTGCTGC CACCCCAGGCCTGTC 3’ 164 Table 4. C-terminal mutants desimed by overlap-extension PCR mutagenesis Mutation Primers Template ins594-612 Mutation Primers ovCOX-l OVCOX-l Forward: 5 ’ GACAGGCCTGGGGTGAATGC AAG'I'I’CTTCCCGCTCCGGACTAGATGAT ATCAATCCC ACAGTACT ACT AAAAGAG CGGCCACCCACA 3’ Reverse: 5’ TGTGGGTGGCCGCT CTTTT AG TAGTACTGTGGGATTGATATCATCI’AGT CCGGAGCGGGAAGAACTTGCA’I‘TCACC CC AGGCCTGTC 3 ’ Common Primers Forward: 5’ GCG'ITTAAAC’I'I‘AAGCTT 3’ Reverse: 5’ GAGCTCGGTACCAAGCTT 3’ Sin5594-612 Mutation primers huCOX-2 huCOX-2 Forward: 5’ ACAGTCACCATCGCT GCT A GTATTAGAGGTCCTTT AACT AAACTT AG TGATAATAGTCTTG’I‘TAGTGATGAACGT TCGACTGAACT G 3’ Reverse: 5’ CAGTTCAGTCGAACGTTCATC ACTAACAAGACTATTATCACTAAGTTI’A GTTAAAGGACCTCTAATACTAGCAGCG ATGGTGACT GT 3’ Common primers Forward: 5’ GC'I‘TGGTACCGAGCTCGGA TCC 3’ Reverse: 5’ CGGGCCCTCTAGACTCGAG CGGCC 3’ 165 Table 4 (cont’d) Sins597-6 12 huCOX-2 Mutation primers Forward: 5’ GTCACCATCAA'I‘GCAAGTAT TAGAGGTCCTTI‘AACTAAACTTAGTGAT AATAGTC’I'FGTTAGTGATGAACGTTCGA CTGAACTG 3’ Reverse: 5’ CAGTTCAGTCGAACGTTCAT C ACTAAC AAGACTATTATC ACT AAGT'I'I’ AGT'I’AAAGGACCTCTAATACTTGCATTG ATGGTGAC 3’ Common primers Forward: 5’ GCTTGGTACCGAGCTCGGA TCC 3’ Reverse: 5’ CGGGCCCTCTAGACTCGAG CGGCC 3’ huCOX-2 A. Conditions for QuickChangeTM site-directed mutagenesis Cycles Temperature (°C) Time (min) 1 94 3 20 94 l 52 l 68 20 1 72 7 166 B. Site-directed mutagenesis by overlap-extension PCR 1) Mutation step a) PCR reaction set-up: 50 ng DNA template 5 11.1 10X Vent DNA polymerase reaction buffer - 25 picomoles each of forward common primer* and reverse mutation primer (Reaction A), or 25 picomoles each of reverse common primer* and forward mutation primer (Reaction B) - 1.25 m of40deNTPmix - 1 ul Vent DNA polymerase (2.5 U/ul) Add ddeO to final volume of 50 pd * The common primers carry the restriction sites b) PCR conditions: Cycles Temperature (°C) Time (min) 1 94 3 3O 94 l 52 l 72 5 l 72 5 167 - After the PCR reaction is completed, treat with Reactions A and B with l u] DpnI for at least 37°C for at least 4 h - Purify the PCR fragments by gel extraction and measure DNA concentration 2) Extension step a) Reaction set-up: - Equal amounts of the purified PCR fragments from Reactions A and B - 5 pl Vent DNA 10X polymerase reaction buffer -1.25 11.140 mM dNTP mix - 1 11.1 Vent DNA polymerase - Bring total volume to 50 111 with ddeO b) PCR conditions Cycles Temperature (°C) Time (min) 1 94 3 6 94 I 52 1 72 5 168 3) Amplification step a) Reaction set-up: To the reaction obtained from the extension step, add: - 25 picomoles of each of the common primers - 1 ul Vent DNA polymerase b) PCR conditions Cycles Temperature (°C) Time (min) 1 94 3 30 94 1 52 l 72 5 l 72 5 - Purify the PCR product using a PCR purification kit - Perform a restriction digestion with the appropriate enzymes and li gate onto vector of interest. 169 ' 10. ll. 12. 13. BIBLIOGRAPHY Smith, W. L., DeWitt, D. L., and Garavito, R. M. (2000) Annu Rev Biochem 69, 145—182 Narumiya, S., and FitzGerald, G. A. (2001) J Clin Invest 108(1), 25-30 Rouzer, C. A., and Mamett, L. J. (2003) Chem Rev 103(6), 2239-2304 Wang, D., Mann, J. R., and DuBois, R. N. (2005) Gastroenterology 128(5), 1445- 1461 Grosser, T., Fries, S., and FitzGerald, G. A. (2006) J Clin Invest 116(1), 4-15 Honda, T., Segi-Nishida, E., Miyachi, Y., and Narumiya, S. (2006) J Exp Med 203(2), 325-335 van der Donk, W. A., Tsai, A. L., and Kulmacz, R. J. (2002) Biochemistry 41(52), 15451-15458 Leslie, C. C. (2004) Biochem Cell Biol 82(1), l-l7 Murakami, M., Kambe-Ohkura, T., and Kudo, I. (2002) Adv Exp Med Biol 507, 15-19 Kudo, I., and Murakami, M. (2002) Prostaglandins Other Lipid Mediat 68-69, 3- 58 Picot, D., Loll, P. J ., and Garavito, R. M. (1994) Nature 367(6460), 243-249 Kurumbail, R. G., Stevens, A. M., Gierse, J. K., McDonald, J. J., Stegeman, R. A., Pak, J. Y., Gildehaus, D., Miyashiro, J. M., Penning, T. D., Seibert, K., Isakson, P. C., and Stallings, W. C. (1996) Nature 384(6610), 644-648 Otto, J. C., and Smith, W. L. (1994) J Biol Chem 269(31), 19868—19875 170 14. 15. 16. 17. 18. 19. 20. 21. 22. 23. 24. 25. 26. 27. Spencer, A. G., Woods, J. W., Arakawa, T., Singer, II, and Smith, W. L. (1998) J Biol Chem 273(16), 9886-9893 MirAfzali, Z., Leipprandt, J. R., McCracken, J. L., and DeWitt, D. L. (2006) J Biol Chem 281(38), 28354-28364 Spencer, A. G., Thuresson, E., Otto, J. C., Song, 1., Smith, T., DeWitt, D. L., Garavito, R. M., and Smith, W. L. (1999) J Biol Chem 274(46), 32936-32942 Yuan, C., Rieke, C. J ., Rimon, G., Wingerd, B. A., and Smith, W. L. (2006) Proc Natl Acad Sci U S A 103(16), 6142-6147 Munro, 8., and Pelham, H. R. (1987) Cell 48(5), 899-907 Song, I., and Smith, W. L. (1996) Arch Biochem Biophys 334(1), 67-72 Tanabe, T., and Tohnai, N. (2002) Prostaglandins Other Lipid Mediat 68-69, 95- l 14 Perkins, D. J ., and Kniss, D. A. (1997) Biochem J 321 ( Pt 3), 677-681 DeWitt, D. L., and Meade, E. A. (1993) Arch Biochem Biophys 306(1), 94-102 Rich, G., Yoder, E. J ., and Moore, S. A. (1998) J Cell Physiol 176(3), 495-505 Zaric, J ., and Ruegg, C. (2005) J Biol Chem 280(2), 1077-1085 Funk, C. D. (2001) Science 294(5548), 1871-1875 Zhang, Y., Shaffer, A., Portanova, J ., Seibert, K., and Isakson, P. C. (1997) J Pharmacol Exp Ther 283(3), 1069-1075 Samad, T. A., Moore, K. A., Sapirstein, A., Billet, S., Allchome, A., Poole, S., Bonventre, J. V., and Woolf, C. J. (2001) Nature 410(6827), 471-475 171 28. 29. 30. 31. 32. 33. 34. 35. 36. 37. 38. 39. 40. Cao, C., Matsumura, K., Yamagata, K., and Watanabe, Y. (1998) Brain Res Mol Brain Res 56(1-2), 45-56 Leahy, K. M., Omberg, R. L., Wang, Y., Zweifel, B. S., Koki, A. T., and Masferrer, J. L. (2002) Cancer Res 62(3), 625-631 Sinicrope, F. A. (2006) Mol Carcinog 45(6), 447-454 Yedgar, S., Cohen, Y., and Shoseyov, D. (2006) Biochim Biophys Acta 1761(11), 1373-1382 Ghosh, M., Tucker, D. E., Burchett, S. A., and Leslie, C. C. (2006) Prog Lipid Res 45(6), 487-510 Mounier, C. M., Ghomashchi, F., Lindsay, M. R., James, 8., Singer, A. G., Parton, R. G., and Gelb, M. H. (2004) J Biol Chem 279(24), 25024-25038 Ni, Z., Okeley, N. M., Smart, B. P., and Gelb, M. H. (2006) J Biol Chem 281(24), 16245-16255 Leslie, C. C. (1997) J Biol Chem 272(27), 16709-16712 Lin, L. L., Lin, A. Y., and DeWitt, D. L. (1992) J Biol Chem 267(33), 23451- 23454 Hoeck, W. G., Ramesha, C. 8., Chang, D. J ., Fan, N., and Heller, R. A. (1993) Proc Natl Acad Sci U S A 90(10), 4475-4479 Maxwell, A. P., Goldberg, H. J ., Tay, A. H., Li, Z. G., Arbus, G. S., and Skorecki, K. L. (1993) Biochem J 295 ( Pt 3), 763-766 Wu, T., Levine, S. J ., Lawrence, M. G., Logun, C., Angus, C. W., and Shelhamer, J. H. (1994) J Clin Invest 93(2), 571-577 Murakami, M., Matsumoto, R., Urade, Y., Austen, K. F., and Ann, J. P. (1995) J Biol Chem 270(7), 3239-3246 172 41. 42. 43. 45. 46. 47. 48. 49. 50. 51. 52. 53. Miyakawa, T., Kojima, M., and Ui, M. (1998) Biochem J 329 (Pt 1), 107-114 Clapham, D. E. (1995) Nature 375(6533), 634-635 Bootman, M. D., and Berridge, M. J. (1995) Cell 83(5), 675-678 Reynolds, L. J ., Hughes, L. L., Louis, A. I., Kramer, R. M., and Dennis, E. A. (1993) Biochim Biophys Acta 1167(3), 272-280 Loo, R. W., Conde-Frieboes, K., Reynolds, L. J ., and Dennis, E. A. (1997) J Biol Chem 272(31), 19214-19219 Clark, J. D., Lin, L. L., Kriz, R. W., Ramesha, C. S., Sultzman, L. A., Lin, A. Y., Milona, N ., and Knopf, J. L. (1991) Cell 65(6), 1043-1051 Das, S., Rafter, J. D., Kim, K. P., Gygi, S. P., and Cho, W. (2003) J Biol Chem 278(42), 41431-41442 Bonventre, J. V., Huang, Z., Taheri, M. R., O'Leary, E., Li, B, Moskowitz, M. A., and Sapirstein, A. (1997) Nature 390(6660), 622-625 Uozumi, N., Kume, K., Nagase, T ., Nakatani, N., Ishii, S., Tashiro, F., Komagata, Y., Maki, K., Ikuta, K., Ouchi, Y., Miyazaki, J ., and Shimizu, T. (1997) Nature 390(6660), 618-622 Hirabayashi, T., Murayama, T., and Shimizu, T. (2004) Biol Pharm Bull 27(8), 1 168-1 173 Hegen, M., Sun, L., Uozumi, N., Kume, K., Goad, M. E., Nickerson-Nutter, C. L., Shimizu, T., and Clark, J. D. (2003) J Exp Med 197(10), 1297-1302 Takaku, K., Sonoshita, M., Sasaki, N., Uozumi, N., Doi, Y., Shimizu, T., and Taketo, M. M. (2000) J Biol Chem 275(44), 34013-34016 Hong, K. H., Bonventre, J. C., O'Leary, E., Bonventre, J. V., and Lander, E. S. (2001) Proc Natl Acad Sci U S A 98(7), 3935-3939 173 54. 55. 56. 57. 58. 59. 60. 61. 62. 63. 65. 66. Nagase, T., Uozumi, N., Ishii, S., Kita, Y., Yamamoto, H., Ohga, E., Ouchi, Y., and Shimizu, T. (2002) Nat Med 8(5), 480—484 Nagase, T., Uozumi, N., Ishii, S., Kume, K., Izumi, T., Ouchi, Y., and Shimizu, T. (2000) Nat Immunol 1(1), 42-46 Kalyvas, A., and David, S. (2004) Neuron 41(3), 323-335 Oikawa, Y., Yamato, E., Tashiro, F., Yamamoto, M., Uozumi, N., Shimada, A., Shimizu, T., and Miyazaki, J. (2005) FEBS Lett 579(18), 3975-3978 Murakami, M., Shimbara, S., Kambe, T., Kuwata, H., Winstead, M. V., Tischfield, J. A., and Kudo, I. (1998) J Biol Chem 273(23), 14411-14423 Murakami, M., Koduri, R. S., Enomoto, A., Shimbara, S., Seki, M., Yoshihara, K., Singer, A., Valentin, E., Ghomashchi, F., Lambeau, G., Gelb, M. H., and Kudo, I. (2001) J Biol Chem 276(13), 10083-10096 Satake, Y., Diaz, B. L., Balestrieri, B., Lam, B. K., Kanaoka, Y., Grusby, M. J ., and Arm, J. P. (2004) J Biol Chem 279(16), 16488-16494 Murakami, M., and Kudo, I. (2004) Biol Pharm Bull 27(8), 1158-1164 Alaoui-El-Azher, M., Wu, Y., Havet, N ., Israel, A., Lilienbaum, A., and Touqui, L. (2002) Mol Pharmacol 61(4), 786-794 Nakano, T., and Arita, H. (1990) FEBS Lett 273(1-2), 23-26 Crowl, R. M., Stoller, T. J ., Conroy, R. R., and Stoner, C. R. (1991) J Biol Chem 266(4), 2647-2651 Pfeilschifter, J ., Schalkwijk, C., Briner, V. A., and van den Bosch, H. (1993) J Clin Invest 92(5), 2516-2523 Kuwata, H., Sawada, H., Murakami, M., and Kudo, I. (1999) Adv Exp Med Biol 469, 183-188 174 67. 68. 69. 70. 71. 72. 73. 74. 75. 76. 77. 78. 79. Pruzanski, W., Vadas, P., Stefanski, E., and Urowitz, M. B. (1985) J Rheumatol 12(2), 211-216 Pruzanski, W., Keystone, E. C., Stemby, B., Bombardier, C., Snow, K. M., and Vadas, P. (1988) J Rheumatol 15(9), 1351-1355 Vadas, P. (1984) J Lab Clin Med 104(6), 873-881 Green, J. A., Smith, G. M., Buchta, R., Lee, R., Ho, K. Y., Rajkovic, I. A., and Scott, K. F. (1991) Inflammation 15(5), 355-367 Vadas, P., and Pruzanski, W. (1993) Circ Shock 39(2), 160-167 Bidgood, M. J ., Jamal, O. S., Cunningham, A. M., Brooks, P. M., and Scott, K. F. (2000) J Immunol 165(5), 2790-2797 Gijon, M. A., Spencer, D. M., Siddiqi, A. R., Bonventre, J. V., and Leslie, C. C. (2000) J Biol Chem 275(26), 20146-20156 Balsinde, J ., Balboa, M. A., and Dennis, E. A. (1998) Proc Natl Acad Sci U S A 95(14), 7951-7956 Reddy, S. T., and Herschman, H. R. (1997) J Biol Chem 272(6), 3231-3237 Murakami, M., Kambe, T., Shimbara, S., and Kudo, I. (1999) J Biol Chem 274(5), 3103-31 15 Reddy, S. T., and Herschman, H. R. (1994) J Biol Chem 269(22), 15473-15480 Murakami, M., Kuwata, H., Amakasu, Y., Shimbara, S., Nakatani, Y., Atsumi, G., and Kudo, I. (1997) J Biol Chem 272(32), 19891-19897 Roshak, A., Sathe, G., and Marshall, L. A. (1994) J Biol Chem 269(42), 25999- 26005 175 80. 81. 82. 83. 84. 85. 86. 87. 88. 89. 90. Langenbach, R., Morham, S. G., Tiano, H. F., Loftin, C. D., Ghanayem, B. I., Chulada, P. C., Mahler, J. F., Lee, C. A., Goulding, E. H., Kluckman, K. D., Kim, H. S., and Smithies, O. (1995) Cell 83(3), 483-492 Kobayashi, T., and Narumiya, S. (2002) Prostaglandins Other Lipid Mediat 68- 69, 557-573 Naraba, H., Murakami, M., Matsumoto, H., Shimbara, S., Ueno, A., Kudo, I., and Oh-ishi, S. (1998) J Immunol 160(6), 2974-2982 Moncada, S., and Vane, J. R. (1979) Fed Proc 38(1), 66-71 Liou, J. Y., Shyue, S. K., T sai, M. J ., Chung, C. L., Chu, K. Y., and Wu, K. K. (2000) J Biol Chem 275(20), 15314-15320 Bustos, M., Coffman, T. M., Saadi, S., and Platt, J. L. (1997) J Clin Invest 100(5), 1150-1158 Okahara, K., Sun, B., and Kambayashi, J. (1998) Arterioscler Thromb Vasc Biol 18(12), 1922-1926 Caughey, G. E., Cleland, L. G., Penglis, P. S., Gamble, J. R. and James, M. J. (2001) J Immunol 167(5), 2831-2838 Solomon, S. D., McMurray, J. J ., Pfeffer, M. A., Wittes, J ., Fowler, R., Finn, R, Anderson, W. F., Zauber, A., Hawk, E., and Bertagnolli, M. (2005) N Engl J Med 352(11), 1071-1080 Bresalier, R. S., Sandler, R. S., Quan, H., Bolognese, J. A., Oxenius, B., Horgan, K., Lines, C., Riddell, R., Morton, D., Lanas, A., Konstam, M. A., and Baron, J. A. (2005) N Engl J Med 352(11), 1092-1102 DeWitt, D. L., Day, J. S., Sonnenburg, W. K., and Smith, W. L. (1983) J Clin Invest 72(6), 1882-1888 Schildknecht, S., Bachschmid, M., Baumann, A., and Ullrich, V. (2004) FASEB J 18(6), 757-759 176 92. 93. 94. 95. 96. 97. 98. 99. 100. 101. 102. 103. 1 ()4. Brock, T. G., McNish, R. W., and Peters-Golden, M. (1999) J Biol Chem 274(17), 1 1660-1 1666 Urade, Y., and Hayaishi, O. (1999) Biochim Biophys Acta 1436(3), 606-615 Eguchi, N., Minarni, T., Shirafuji, N., Kanaoka, Y., Tanaka, T., Nagata, A., Yoshida, N., Urade, Y., Ito, S., and Hayaishi, O. (1999) Proc Natl Acad Sci U S A 96(2), 726-730 Heavey, D. J., Ernst, P. B., Stevens, R. L., Befus, A. D., Bienenstock, J ., and Austen, K. F. (1988) J Immunol 140(6), 1953-1957 Diaz, B. L., Satake, Y., Kikawada, E., Balestrieri, B., and Ann, J. P. (2006) Biochim Biophys Acta 1761(12), 1489-1497 Kabashima, K., and Narumiya, S. (2003) Prostaglandins Leukot Essent Fatty Acids 69(2-3), 187-194 Lehmann, J. M., Moore, L. B., Smith-Oliver, T. A., Wilkison, W. 0., Willson, T. M., and Kliewer, S. A. (1995) J Biol Chem 270(22), 12953-12956 Forman, B. M., Tontonoz, P., Chen, J ., Brun, R. P., Spiegelman, B. M., and Evans, R. M. (1995) Cell 83(5), 803-812 Scher, J. U., and Pillinger, M. H. (2005) Clin Immunol 114(2), 100-109 Ide, T., Egan, K., Bell-Parikh, L. C., and FitzGerald, G. A. (2003) Thromb Res 110(5-6), 311-315 Chinetti, G., Griglio, S., Antonucci, M., Torra, I. P., Delerive, P., Majd, Z., Fruchart, J. C., Chapman, J ., Najib, J ., and Staels, B. (1998) J Biol Chem 273(40), 25573-25580 Azuma, Y., Shinohara, M., Wang, P. L., and Ohura, K. (2001) Biochem Biophys Res Commun 283(2), 344-346 Urade, Y., and Eguchi, N. (2002) Prostaglandins Other Lipid Mediat 68-69, 375- 382 177 105. 106. 107. 108. 109. 110. 111. 112. 113. 114. 115. Grill, M., Peskar, B. A., Schuligoi, R., and Amann, R. (2006) Neuropharmacology 50(2), 165-173 Murakami, M., Matsumoto, R., Austen, K. F., and Ann, J. P. (1994) J Biol Chem 269(35), 22269-22275 Harada, Y., Hatanaka, K., Kawamura, M., Saito, M., Ogino, M., Majima, M., Ohno, T., Ogino, K., Yamamoto, K., Taketani, Y., Yamamoto, S., and Katori, M. (1996) Prostaglandins 51(1), 19-33 Li, S., Wang, Y., Matsumura, K., Ballou, L. R., Morham, S. G., and Blatteis, C. M. (1999) Brain Res 825(1-2), 86-94 Ueno, N., Takegoshi, Y., Kamei, D., Kudo, I., and Murakami, M. (2005) Biochem Biophys Res Commun 338(1), 70-76 Tanioka, T., Nakatani, Y., Semmyo, N ., Murakami, M., and Kudo, I. (2000) J Biol Chem 275(42), 32775-32782 Murakami, M., and Kudo, I. (2004) Prog Lipid Res 43(1), 3-35 Kamei, D., Yamakawa, K., Takegoshi, Y., Mikami-Nakanishi, M., Nakatani, Y., Oh-Ishi, S., Yasui, H., Azuma, Y., I-Iirasawa, N., Ohuchi, K., Kawaguchi, H., Ishikawa, Y., Ishii, T., Uematsu, S., Akira, S., Murakami, M., and Kudo, I. (2004) J Biol Chem 279(32), 33684-33695 Myers, L. K., Kang, A. H., Postlethwaite, A. E., Rosloniec, E. F., Morham, S. G., Shlopov, B. V., Goorha, S., and Ballou, L. R. (2000) Arthritis Rheum 43(12), 2687-2693 Trebino, C. E., Stock, J. L., Gibbons, C. P., Naiman, B. M., Wachtrnann, T. S., Umland, J. P., Pandher, K., Lapointe, J. M., Saha, S., Roach, M. L., Carter, D., Thomas, N. A., Durtschi, B. A., McNeish, J. D., Hambor, J. E., Jakobsson, P. J ., Carty, T. J ., Perez, J. R., and Audoly, L. P. (2003) Proc Natl Acad Sci U S A 100(15), 9044-9049 Engblom, D., Saha, S., Engstrom, L., Westman, M., Audoly, L. P., J akobsson, P. J ., and Blomqvist, A. (2003) Nat Neurosci 6(11), 1137-1138 178 116. 117. 118. 119. 120. 121. 122. 123. 124. 125. 126. 127. 128. Wang, L. H., Hajibeigi, A., Xu, X. M., Loose-Mitchell, D., and Wu, K. K. (1993) Biochem Biophys Res Commun 190(2), 406-411 Xu, X. M., Tang, J. L., Chen, X., Wang, L. H., and Wu, K. K. (1997) J Biol Chem 272(11), 6943-6950 Duquette, M., and Laneuville, O. (2002) J Biol Chem 277(47), 44631-44637 DeLong, C. J ., and Smith, W. L. (2005) Biochem Biophys Res Commun 338(1), 53-61 Topper, J. N ., Cai, J ., Falb, D., and Gimbrone, M. A., Jr. (1996) Proc Natl Acad Sci U SA 93(19), 10417-10422 Xie, W. L., Chipman, J. G., Robertson, D. L., Erikson, R. L., and Simmons, D. L. (1991) Proc Natl Acad Sci U S A 88(7), 2692-2696 Kujubu, D. A., Reddy, S. T., Fletcher, B. S., and Herschman, H. R. (1993) J Biol Chem 268(8), 5425-5430 Fletcher, B. S., Kujubu, D. A., Penin, D. M., and Herschman, H. R. (1992) J Biol Chem 267(7), 4338-4344 Herschman, H. R., Fletcher, B. S., and Kujubu, D. A. (1993) J Lipid Mediat 6(1- 3), 89-99 Asano, K., Lilly, C. M., and Drazen, J. M. (1996) Am J Physiol 271(1 Pt 1), L126-131 Kaufmann, W. E., Worley, P. F., Taylor, C. V., Bremer, M., and Isakson, P. C. (1997) Brain Dev 19(1), 25-34 Harris, R. C., and Breyer, M. D. (2001) Am J Physiol Renal Physiol 281(1), Fl-ll Bagga, D., Wang, L., Farias-Eisner, R., Glaspy, J. A., and Reddy, S. T. (2003) Proc Natl Acad Sci U S A 100(4), 1751-1756 179 129. 130. 131. 132. 133. 134. 135. 136. 137. 138. 139. 140. 141. Chen, C. Y., and Shyu, A. B. (1994) Mol Cell Biol 14(12), 8471-8482 Kang, Y. J ., Wingerd, B. A., Arakawa, T., and Smith, W. L. (2006) J Immunol 177(11), 8111-8122 Kang, Y. J ., Mbonye, U. R., Delong, C. J., Wada, M., and Smith, W. L. (2007) Prog Lipid Res Hwang, D., Jang, B. C., Yu, G., and Boudreau, M. ( 1997) Biochem Pharmacol 54(1), 87-96 Mestre, J. R., Mackrell, P. J ., Rivadeneira, D. E., Stapleton, P. P., Tanabe, T., and Daly, J. M. (2001) J Biol Chem 276(6), 3977-3982 Scheinman, R. I., Cogswell, P. C., Lofquist, A. K., and Baldwin, A. S., Jr. (1995) Science 270(5234), 283-286 Auphan, N., DiDonato, J. A., Rosette, C., Helmberg, A., and Karin, M. (1995) Science 270(5234), 286-290 Farrajota, K., Cheng, S., Martel-Pelletier, J ., Afif, H., Pelletier, J. R, Li, X., Ranger, P., and Fahmi, H. (2005) Arthritis Rheum 52(1), 94-104 Kapoor, M., Kojima, F., Qian, M., Yang, L., and Crofford, L. J. (2006) J Biol Chem Subbaramaiah, K., Lin, D. T., Hart, J. C., and Dannenberg, A. J. (2001) J Biol Chem 276(15), 12440-12448 Chen, C. Y., and Shyu, A. B. (1995) Trends Biochem Sci 20(11), 465-470 Chen, C. Y., Gherzi, R., Ong, S. E., Chan, E. L., Raijmakers, R., Pruijn, G. J ., Stoecklin, G., Moroni, C., Mann, M., and Karin, M. (2001) Cell 107(4), 451-464 Mukherjee, D., Gao, M., O'Connor, J. P., Raijmakers, R., Pruijn, G., Lutz, C. S., and Wilusz, J. (2002) EMBO J 21(1-2), 165-174 180 142. 143. 144. 145. 146. 147. 148. 149. 150. 151. 152. 153. 154. Stoecklin, G., Mayo, T., and Anderson, P. (2006) EMBO Rep 7(1), 72-77 Lasa, M., Mahtani, K. R., Finch, A., Brewer, G., Saklatvala, J ., and Clark, A. R. (2000) Mol Cell Biol 20(12), 4265-4274 Lasa, M., Brook, M., Saklatvala, J ., and Clark, A. R. (2001) Mol Cell Biol 21(3), 771-780 Dixon, D. A., Tolley, N. D., King, P. H., Nabors, L. B., McIntyre, T. M., Zimmerman, G. A., and Prescott, S. M. (2001) J Clin Invest 108(11), 1657-1665 Murmu, N ., Jung, J ., Mukhopadhyay, D., Houchen, C. W., Riehl, T. E., Stenson, W. F., Morrison, A. R., Arumugam, T., Dieckgraefe, B. K., and Anant, S. (2004) Proc Natl Acad Sci U S A 101(38), 13873-13878 Mukhopadhyay, D., Houchen, C. W., Kennedy, 8., Dieckgraefe, B. K., and Anant, S. (2003) Mol Cell 11(1), 113-126 Rockwell, P., Yuan, H., Magnusson, R., and Figueiredo-Pereira, M. E. (2000) Arch Biochem Biophys 374(2), 325-333 Figueiredo-Pereira, M. R, Li, Z., Jansen, M., and Rockwell, P. (2002) J Biol Chem 277(28), 25283-25289 Xiao, G., Chen, W., and Kulmacz, R. J. (1998) J Biol Chem 273(12), 6801-6811 Otto, J. C., DeWitt, D. L., and Smith, W. L. (1993) J Biol Chem 268(24), 18234- 18242 Garavito, R. M., Malkowski, M. G., and DeWitt, D. L. (2002) Prostaglandins Other Lipid Mediat 68-69, 129-152 Malkowski, M. G., Ginell, S. L., Smith, W. L., and Garavito, R. M. (2000) Science 289(5486), 1933-1937 Rieke, C. J ., Mulichak, A. M., Garavito, R. M., and Smith, W. L. ( 1999) J Biol Chem 274(24), 17109-17114 181 155. 156. 157. 158. 159. 160. 161. 162. 163. 164. 165. 166. 167. Kulmacz, R. J ., and Wang, L. H. (1995) J Biol Chem 270(41), 24019-24023 Smith, W. L., and Song, I. (2002) Prostaglandins Other Lipid Mediat 68-69, 115- 128 Kiefer, J. R., Pawlitz, J. L., Moreland, K. T., Stegeman, R. A., Hood, W. F., Gierse, J. K., Stevens, A. M., Goodwin, D. C., Rowlinson, S. W., Mamett, L. J ., Stallings, W. C., and Kurumbail, R. G. (2000) Nature 405(6782), 97-101 Dietz, R., Nastainczyk, W., and Ruf, H. H. (1988) Eur J Biochem 171(1-2), 321- 328 Lu, G., Tsai, A. L., Van Wart, H. E., and Kulmacz, R. J. (1999) J Biol Chem 274(23), 16162-16167 Landino, L. M., Crews, B. C., Gierse, J. K., Hauser, S. D., and Mamett, L. J. (1997) J Biol Chem 272(34), 21565-21574 Laneuville, O., Breuer, D. K., Xu, N., Huang, Z. H., Gage, D. A., Watson, J. T., Lagarde, M., DeWitt, D. L., and Smith, W. L. (1995) J Biol Chem 270(33), 19330-19336 Mevkh, A. T., Miroshnikov, K. A., Igumnova, N. D., and Varfolomeev, S. D. (1993) FEBS Lett 321(2-3), 205-208 Mevkh, A. T., Miroshnikov, K. A., Igumnova, N. D., and Varfolomeev, S. D. (1993) Biokhimiia 58(10), 1573-1579 Meusser, B., Hirsch, C., J arosch, E., and Sommer, T. (2005) Nat Cell Biol 7(8), 766-772 McCracken, A. A., and Brodsky, J. L. (2003) Bioessays 25(9), 868-877 Spiro, R. G. (2004) Cell Mol Life Sci 61(9), 1025-1041 Stoka, V., Turk, B., and Turk, V. (2005) IUBMB Life 57(4-5), 347-353 182 168. Rubinsztein, D. C. (2006) Nature 443(7113), 780-786 169. Ciechanover, A. (2005) Cell Death Differ 12(9), 1178-1190 170. Finn, P. F., and Dice, J. F. (2006) Nutrition 22(7-8), 830-844 171. Zwickl, P., Voges, D., and Baumeister, W. (1999) Philos Trans R Soc Lond B Biol Sci 354(1389), 1501-1511 172. Peters, J. M., Cejka, 2., Harris, J. R., Kleinschmidt, J. A., and Baumeister, W. (1993) J Mol Biol 234(4), 932-937 173. Adams, J. (2003) Cancer Treat Rev 29 Suppl 1, 3-9 174. Wang, J ., and Maldonado, M. A. (2006) Cell Mol Immunol 3(4), 255-261 175. Miller, J ., and Gordon, C. (2005) FEBS Lett 579(15), 3224-3230 176. Yao, T., and Cohen, R. E. (2002) Nature 419(6905), 403-407 177. Braun, B. C., Glickman, M., Kraft, R., Dahlmann, B., Kloetzel, P. M., Finley, D., and Schmidt, M. (1999) Nat Cell Biol 1(4), 221-226 178. Coffino, P. (2001) Nat Rev Mol Cell Biol 2(3), 188-194 179. Newman, R. M., Mobascher, A., Mangold, U., Koike, C., Diah, S., Schmidt, M., Finley, D., and letter, B. R. (2004) J Biol Chem 279(40), 41504-41511 180. Zhang, M., Pickart, C. M., and Coffino, P. (2003) EMBO J 22(7), 1488-1496 181. Kahana, C., Asher, G., and Shaul, Y. (2005) Cell Cycle 4(11), 1461-1464 182. Hampton, R. Y. (2002) Curr Opin Cell Biol 14(4), 476-482 183 183. 184. 185. 186. 187. 188. 189. 190. 191. 192. 193. 194. 195. 196. 197. McCracken, A. A., and Brodsky, J. L. (2005) Curr Top Microbiol Immunol 300, 17—40 Pety de Thozee, C., and Ghislain, M. (2006) ScientificWorldJoumal 6, 967-983 Hirsch, C., Jarosch, E., Sommer, T., and Wolf, D. H. (2004) Biochim Biophys Acta 1695(1-3), 215-223 Trombetta, E. S., and Parodi, A. J. (2003) Annu Rev Cell Dev Biol 19, 649-676 Guo, W., Shea, J. E., and Berry, R. S. (2005) Ann N Y Acad Sci 1066, 34-53 Carvalho, P., Goder, V., and Rapoport, T. A. (2006) Cell 126(2), 361-373 Ismail, N., and N g, D. T. (2006) Cell 126(2), 237-239 Ahner, A., and Brodsky, J. L. (2004) Trends Cell Biol 14(9), 474-478 Mueller, B., Lilley, B. N., and Ploegh, H. L. (2006) J Cell Biol 175(2), 261-270 Nishikawa, S. I., Fewell, S. W., Kato, Y., Brodsky, J. L., and Endo, T. (2001) J Cell Biol 153(5), 1061-1070 Brodsky, J. L., Werner, E. D., Dubas, M. E., Goeckeler, J. L., Kruse, K. B., and McCracken, A. A. (1999) J Biol Chem 274(6), 3453-3460 McCracken, A. A., and Brodsky, J. L. (1996) J Cell Biol 132(3), 291-298 Pollock, S., Kozlov, G., Pelletier, M. F., Trempe, J. F., Jansen, G., Sitnikov, D., Bergeron, J. J ., Gehring, K., Ekiel, I., and Thomas, D. Y. (2004) Embo J 23(5), 1020-1029 Caramelo, J. J ., Castro, 0. A., Alonso, L. G., De Prat-Gay, G., and Parodi, A. J. (2003) Proc Natl Acad Sci U S A 100(1), 86-91 Trombetta, E. S., and Helenius, A. (2000) J Cell Biol 148(6), 1123-1129 184 198. 199. 200. 201 . 202. 203. 204. 205. 206. 207. 208. 209. 210. Jakob, C. A., Burda, P., Roth, J ., and Aebi, M. (1998) J Cell Biol 142(5), 1223- 1233 Tokunaga, F., Brostrom, C., Koide, T., and Arvan, P. (2000) J Biol Chem 275(52), 40757-40764 Hosokawa, N., Tremblay, L. 0., You, 2., Herscovics, A., Wada, I., and Nagata, K. (2003) J Biol Chem 278(28), 26287-26294 Denic, V., Quan, E. M., and Weissman, J. S. (2006) Cell 126(2), 349-359 Kim, W., Spear, E. D., and Ng, D. T. (2005) Mol Cell 19(6), 753-764 Gnann, A., Riordan, J. R., and Wolf, D. H. (2004) Mol Biol Cell 15(9), 4125-4135 Oda, Y., Hosokawa, N., Wada, I., and Nagata, K. (2003) Science 299(5611), 1394-1397 Molinari, M., Calanca, V., Galli, C., Lucca, P., and Paganetti, P. (2003) Science 299(5611), 1397-1400 Plemper, R. K., Bohmler, S., Bordallo, J., Sommer, T., and Wolf, D. H. (1997) Nature 388(6645), 891-895 Kabani, M., Kelley, S. S., Morrow, M. W., Montgomery, D. L., Sivendran, R., Rose, M. D., Gierasch, L. M., and Brodsky, J. L. (2003) Mol Biol Cell 14(8), 3437-3448 Fagioli, C., Mezghrani, A., and Sitia, R. (2001) J Biol Chem 276(44), 40962- 40967 Tortorella, D., Story, C. M., Huppa, J. B., Wiertz, E. J ., Jones, T. R., Bacik, I., Bennink, J. R., Yewdell, J. W., and Ploegh, H. L. (1998) J Cell Biol 142(2), 365- 376 Kalies, K. U., Allan, S., Sergeyenko, T., Kroger, H., and Romisch, K. (2005) EMBO J 24(13), 2284-2293 185 211. 212. 213. 214. 215. 216. 217. 218. 219. 220. 221. 222. 223. Wiertz, E. J., Tortorella, D., Bogyo, M., Yu, J ., Mothes, W., Jones, T. R., Rapoport, T. A., and Ploegh, H. L. (1996) Nature 384(6608), 432-438 Pariyarath, R., Wang, H., Aitchison, J. D., Ginsberg, H. N., Welch, W. J ., Johnson, A. E., and Fisher, E. A. (2001) J Biol Chem 276(1), 541-550 Pilon, M., Schekman, R., and Romisch, K. (1997) EMBO J 16(15), 4540-4548 Van den Berg, B., Clemons, W. M., Jr., Collinson, I., Modis, Y., Hartmann, E., Harrison, S. C., and Rapoport, T. A. (2004) Nature 427(6969), 36-44 Clemons, W. M., Jr., Menetret, J. F., Akey, C. W., and Rapoport, T. A. (2004) Curr Opin Struct Biol 14(4), 390-396 Wirth, A., Jung, M., Bies, C., Frien, M., Tyedmers, J ., Zimmermann, R., and Wagner, R. (2003) Mol Cell 12(1), 261-268 Hamman, B. D., Chen, J. C., Johnson, E. E., and Johnson, A. E. (1997) Cell 89(4), 535-544 Lilley, B. N., and Ploegh, H. L. (2004) Nature 429(6994), 834-840 Ye, Y., Shibata, Y., Yun, C., Ron, D., and Rapoport, T. A. (2004) Nature 429(6994), 841-847 Jarosch, E., Taxis, C., Volkwein, C., Bordallo, J ., Finley, D., Wolf, D. H., and Sommer, T. (2002) Nat Cell Biol 4(2), 134-139 J arosch, E., Geiss-Friedlander, R., Meusser, B., Walter, J ., and Sommer, T. (2002) Traffic 3(8), 530-536 Oda, Y., Okada, T., Yoshida, H., Kaufman, R. J ., Nagata, K., and Mori, K. (2006) J Cell Biol 172(3), 383-393 Pye, V. E., Beuron, F., Keetch, C. A., McKeown, C., Robinson, C. V., Meyer, H. H., Zhang, X., and Freemont, P. S. (2007) Proc Natl Acad Sci U S A 104(2), 467- 472 186 224. 225. 226. 227. 228. 229. 230. 231. 232. 233. 234. 235. 236. 237. Chen, B., Mariano, J ., Tsai, Y. C., Chan, A. H., Cohen, M., and Weissman, A. M. (2006) Proc Natl Acad Sci U S A 103(2), 341-346 Fang, S., Ferrone, M., Yang, C., Jensen, J. P., Tiwari, S., and Weissman, A. M. (2001) Proc Natl Acad Sci U S A 98(25), 14422-14427 Schulze, A., Standera, S., Buerger, E., Kikkert, M., van Voorden, S., Wiertz, E., Koning, F., Kloetzel, P. M., and Seeger, M. (2005) J Mol Biol 354(5), 1021-1027 Zhong, X., Shen, Y., Ballar, P., Apostolou, A., Agami, R., and Fang, S. (2004) J Biol Chem 279(44), 45676-45684 Schuberth, C., and Buchberger, A. (2005) Nat Cell Biol 7(10), 999-1006 Bar-Nun, S. (2005) Curr Top Microbiol Immunol 300, 95-125 Katiyar, S., Joshi, S., and Lennarz, W. J. (2005) Mol Biol Cell 16(10), 4584-4594 Li, G., Zhao, G., Zhou, X., Schindelin, H., and Lennarz, W. J. (2006) Proc Natl Acad Sci U S A 103(22), 8348-8353 Ravid, T., Doolman, R., Avner, R., Harats, D., and Roitelman, J. (2000) J Biol Chem 275(46), 35840-35847 Murray, B. P., and Correia, M. A. (2001) Arch Biochem Biophys 393(1), 106-116 Webster, J. M., Tiwari, S., Weissman, A. M., and Wojcikiewicz, R. J. (2003) J Biol Chem 278(40), 38238-38246 Alzayady, K. J ., Panning, M. M., Kelley, G. G., and Wojcikiewicz, R. J. (2005) J Biol Chem 280(41), 34530-34537 Sever, N., Song, B. L., Yabe, D., Goldstein, J. L., Brown, M. S., and DeBose- Boyd, R. A. (2003) J Biol Chem 278(52), 52479-52490 Sever, N., Yang, T., Brown, M. S., Goldstein, J. L., and DeBose-Boyd, R. A. (2003) Mol Cell 11(1), 25-33 187 238. 239. 240. 241. 242. 243. 244. 245. 246. 247. 248. 249. 250. 251. Song, B. L., Javitt, N. B., and DeBose-Boyd, R. A. (2005) Cell Metab 1(3), 179- 189 Song, B. L., Sever, N., and DeBose-Boyd, R. A. (2005) Mol Cell 19(6), 829-840 Evett, G. E., Xie, W., Chipman, J. G., Robertson, D. L., and Simmons, D. L. (1993) Arch Biochem Biophys 306(1), 169-177 Herschman, H. R. (1996) Biochim Biophys Acta 1299(1), 125-140 Shao, J., Sheng, H., Inoue, H., Morrow, J. D., and DuBois, R. N. (2000) J Biol Chem 275(43), 33951-33956 Tsai, B., Ye, Y., and Rapoport, T. A. (2002) Nat Rev Mol Cell Biol 3(4), 246-255 Ellgaard, L., and Helenius, A. (2001) Curr Opin Cell Biol 13(4), 431-437 Robertson, R. P. (1998) Diabetes 47(9), 1379-1383 Ristimaki, A., Honkanen, N., Jankala, H., Sipponen, P., and Harkonen, M. (1997) Cancer Res 57(7), 1276-1280 Nemeth, J. F., Hochgesang, G. P., Jr., Mamett, L. J ., and Caprioli, R. M. (2001) Biochemistry (Mose) 40(10), 3109-31 16 Fagioli, C., and Sitia, R. (2001) J Biol Chem 276(16), 12885-12892 Hosokawa, N ., Wada, I., Hasegawa, K., Yorihuzi, T., Tremblay, L. O., Herscovics, A., and Nagata, K. (2001) EMBO Rep 2(5), 415-422 Kitzmuller, C., Caprini, A., Moore, S. E., Frenoy, J. P., Schwaiger, E., Kellermann, O., Ivessa, N. E., and Ermonval, M. (2003) Biochem J 376(Pt 3), 687-696 Tsuda, H., Tokunaga, F., Nagarnitsu, H., and Koide, T. (2006) Thromb Res 117(3), 323-331 188 252. 253. 254. 255. 256. 257. 258. 259. 260. 261. 262. 263. 264. 265. Yu, Y., Fan, J ., Hui, Y., Rouzer, C. A., Mamett, L. J ., Klein-Szanto, A. J ., FitzGerald, G. A., and Funk, C. D. (2007) J Biol Chem 282(2), 1498-1506 Cok, S. J., and Morrison, A. R. (2001) J Biol Chem 276(25), 23179-23185 Dixon, D. A., Kaplan, C. D., McIntyre, T. M., Zimmerman, G. A., and Prescott, S. M. (2000) J Biol Chem 275(16), 11750-11757 Kostova, Z., and Wolf, D. H. (2003) EMBO J 22(10), 2309-2317 Spear, E. D., and Ng, D. T. (2005) J Cell Biol 169(1), 73-82 Mbonye, U. R., Wada, M., Rieke, C. J., Tang, H. Y., Dewitt, D. L., and Smith, W. L. (2006) J Biol Chem 281(47), 35770-35778 Sevigny, M. B., Li, C. F., Alas, M., and Hughes-Fulford, M. (2006) FEBS Lett 580(28-29), 6533-6536 Wu, Y., Swulius, M. T., Moremen, K. W., and Sifers, R. N. (2003) Proc Natl Acad Sci U S A 100(14), 8229-8234 Wu, Y., Terrnine, D. J ., Swulius, M. T., Moremen, K. W., and Sifers, R. N. (2007) J Biol Chem 282(7), 4841-4849 Cabral, C. M., Liu, Y., and Sifers, R. N. (2001) Trends Biochem Sci 26(10), 619- 624 Kelleher, D. J ., Kreibich, G., and Gilmore, R. (1992) Cell 69(1), 55-65 Nilsson, I. M., and von Heijne, G. ( 1993) J Biol Chem 268(8), 5798-5801 Bolt, G., Kristensen, C., and Steenstrup, T. D. (2005) Glycobiology 15(5), 541- 547 Tanaka, N., Sato, T., Fujita, H., and Morita, I. (2004) Arterioscler Thromb Vasc Biol 24(3), 607-612 189 266. 267. 268. 269. 270. 271. 272. 273. 274. 275. 276. 277. 278. Miyamoto, T., Ogino, N., Yamamoto, S., and Hayaishi, O. (1976) J Biol Chem 251(9), 2629-2636 Xu, X. M., Tang, J. L., Hajibeigi, A., Loose-Mitchell, D. S., and Wu, K. K. (1996) Am J Physiol 270(1 Pt 1), C259-264 } Song, 1., Ball, T. M., and Smith, W. L. (2001) Biochem Biophys Res Commun 289(4), 869-875 Wada, M., Yokoyama, C., Hatae, T., Shimonishi, M., Nakamura, M., Irnai, Y., Ullrich, V., and Tanabe, T. (2004) J Biochem (Tokyo) 135(4), 455-463 Habenicht, A. J ., Goeri g, M., Grulich, J ., Rothe, D., Gronwald, R., Loth, U., Schettler, G., Kommerell, B., and Ross, R. (1985) J Clin Invest 75(4), 1381-1387 Barnett, J ., Chow, J ., Ives, D., Chiou, M., Mackenzie, R., Osen, E., Nguyen, B., Tsing, S., Bach, C., Freire, J ., and et al. (1994) Biochim Biophys Acta 1209(1), 130-139 Rowlinson, S. W., Crews, B. C., Lanzo, C. A., and Mamett, L. J. (1999) J Biol Chem 274(33), 23305-23310 Riese, J ., Hoff, T., Nordhoff, A., DeWitt, D. L., Resch, K., and Kaever, V. (1994) J Leukoc Biol 55(4), 476-482 Martoglio, B., and Dobberstein, B. (1998) Trends Cell Biol 8(10), 410-415 Sanders, S. L., and Schekman, R. (1992) J Biol Chem 267(20), 13791-13794 Schmitz, A., and Herzog, V. (2004) Eur] Cell Biol 83(10), 501-509 Sato, T ., Nyborg, A. C., Iwata, N., Diehl, T. S., Saido, T. C., Golde, T. E., and Wolfe, M. S. (2006) Biochemistry (Mose) 45(28), 8649-8656 Loureiro, J ., Lilley, B. N., Spooner, E., Noriega, V., Tortorella, D., and Ploegh, H. L. (2006) Nature 441(7095), 894-897 190 279. 280. 281. 282. 283. 284. 285. 286. 287. Serwold, T., Gonzalez, F., Kim, J ., Jacob, R., and Shastri, N. (2002) Nature 419(6906), 480-483 Saric, T., Chang, S. C., Hattori, A., York, I. A., Markant, 8., Rock, K. L., Tsujimoto, M., and Goldberg, A. L. (2002) Nat Immunol 3(12), 1169-1176 York, I. A., Chang, S. C., Saric, T., Keys, J. A., Favreau, J. M., Goldberg, A. L., and Rock, K. L. (2002) Nat Immunol 3(12), 1177-1184 Saveanu, L., Carroll, 0., Lindo, V., Del Val, M., Lopez, D., Lepelletier, Y., Greer, F., Schomburg, L., Fruci, D., Niedermann, G., and van Endert, P. M. (2005) Nat Immunol 6(7), 689-697 Heinemann, F. S., and Ozols, J. (1998) Mol Biol Cell 9(12), 3445-3453 Heinemann, F. S., Mziaut, H., Korza, G., and Ozols, J. (2003) Biochemistry (Mose) 42(23), 6929-6937 Heinemann, F. S., Korza, G., and Ozols, J. (2003) J Biol Chem 278(44), 42966- 42975 Mziaut, H., Korza, G., and Ozols, J. (2000) Proc Natl Acad Sci U S A 97(16), 8883-8888 Donoso, G., Herzog, V., and Schmitz, A. (2005) Biochem J 387(Pt 3), 897-903 191 111111111111111111