throbber
P1: FPP
`July 25, 2000
`
`12:47
`
`AR102
`
`CHAP15
`
`Annu. Rev. Biochem. 2000. 69:419–45
`Copyright c(cid:13) 2000 by Annual Reviews. All rights reserved
`
`PLATELET-ACTIVATING FACTOR
`AND RELATED LIPID MEDIATORS
`
`Stephen M. Prescott1, Guy A. Zimmerman2,
`Diana M. Stafforini1, and Thomas M. McIntyre2
`The Huntsman Cancer Institute1 and the Program in Human Molecular Biology
`and Genetics2, University of Utah, Salt Lake City, Utah 84112;
`e-mail: steve.prescott@hci.utah.edu
`
`Key Words PAF, inflammation, phospholipases
`n Abstract Platelet-activating factor (PAF) is a phospholipid with potent, diverse
`physiological actions, particularly as a mediator of inflammation. The synthesis, trans-
`port, and degradation of PAF are tightly regulated, and the biochemical basis for many
`of these processes has been elucidated in recent years. Many of the actions of PAF can
`be mimicked by structurally related phospholipids that are derived from nonenzymatic
`oxidation, because such compounds can bind to the PAF receptor. This process cir-
`cumvents much of the biochemical control and presumably is regulated primarily by
`the rate of degradation, which is catalyzed by PAF acetylhydrolase. The isolation of
`cDNA clones encoding most of the key proteins involved in regulating PAF has allowed
`substantial recent progress and will facilitate studies to determine the structural basis
`for substrate specificity and the precise role of PAF in physiological events.
`
`CONTENTS
`
`INTRODUCTION : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : 419
`PHYSIOLOGIC ACTIONS OF PLATELET-ACTIVATING FACTOR : : : : : : : : : : : 420
`PAF Is a Cell-Associated Signal for Leukocyte Adhesion and Activation
`in Models of Acute Inflammation: Juxtacrine Signaling : : : : : : : : : : : : : : : : : : : 420
`Signaling Roles for PAF in Vivo : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : 422
`THE PAF RECEPTOR : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : 424
`SYNTHESIS OF PLATELET-ACTIVATING FACTOR
`AND GENERATION OF RELATED COMPOUNDS : : : : : : : : : : : : : : : : : : : : : : 428
`Platelet-Activating Factor : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : 428
`Oxidative Generation of Platelet Activating Factor-Like Lipids : : : : : : : : : : : : : : 431
`DEGRADATION OF PAF: THE ACETYLHYDROLASES : : : : : : : : : : : : : : : : : : 433
`The Intracellular Forms : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : 435
`The Plasma Form : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : 437
`CONCLUSION : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : : 441
`
`0066-4154/00/0707-0419/$14.00
`
`419
`
`Annu. Rev. Biochem. 2000.69:419-445. Downloaded from www.annualreviews.org
`
` by John Jones on 10/12/17. For personal use only.
`
`AKER EXHIBIT 2003 Page 1
`
`

`

`P1: FPP
`July 25, 2000
`
`12:47
`
`AR102
`
`CHAP15
`
`420
`
`PRESCOTT ET AL
`
`INTRODUCTION
`
`Platelet-activating factor (PAF) is the trivial name for a phospholipid, 1-O-alkyl-2-
`acetyl-sn-glycero-3-phosphocholine that has potent, diverse physiological actions.
`Its discovery and the early studies to characterize its actions, synthesis, and degra-
`dation were described in a previous chapter in this series (1) and other reviews
`(2–5). Many lipid mediators are derived from phospholipids (6), but PAF was the
`first one shown to have autocoid, or messenger, functions as an intact phospholipid.
`This signaling results from PAF binding to a specific receptor—not from physico-
`chemical effects on the plasma membrane of the target cell. One of the most in-
`triguing aspects of PAF has been the strict structural requirement for binding to
`its receptor and for recognition as a substrate both by synthetic and degradative
`enzymes. The last point seems to be an eminently practical requirement because, if
`the degradative enzymes were not specific for this unusual structure, there would
`be continuous hydrolysis of structural phospholipids.
`
`PHYSIOLOGIC ACTIONS OF PLATELET-ACTIVATING
`FACTOR
`
`PAF’s primary role seems clearly to be to mediate intercellular interactions; when
`synthesized by cells of a variety of types, it binds to receptors on the plasma
`membranes of other cells, which activates them and changes their phenotypes. In
`addition to intercellular signaling, it has been proposed that PAF has both autocrine
`and intracrine effects, the latter occurring via one or more intracellular receptors.
`Although postulated to exist for many years based on pharmacological studies,
`intracellular receptors for PAF are yet to be rigorously characterized. We focus
`here on intercellular signaling by PAF. Many of its best-characterized signaling
`roles are in the vascular and inflammatory systems (see below), but it also trans-
`mits information between cells in the central nervous system and in endocrine,
`gastrointestinal, and other organs (see review articles above). Most of the follow-
`ing examples are drawn from studies in inflammation, but the principles likely are
`relevant in multiple systems. Several mechanisms regulate the PAF intercellular-
`signaling system. These include tightly controlled synthetic pathways, spatial reg-
`ulation of the display and biologic availability of PAF, cell-specific expression of
`the receptor for PAF, homologous and heterologous desensitization of the receptor,
`and rapid degradation of PAF by extracellular and intracellular acetylhydrolases.
`Each of these regulatory features is considered in more detail elsewhere in the
`chapter. Together, they indicate specialization of function for PAF as an intercel-
`lular signal, and the redundant mechanisms appear to have evolved to precisely
`control its biologic activities. Thus, unregulated or dysregulated signaling by PAF
`can be a mechanism of disease. Disease models in isolated cell systems (7–10) and
`experimental animals (5) and observations in human syndromes (5) also support
`this possibility.
`
`Annu. Rev. Biochem. 2000.69:419-445. Downloaded from www.annualreviews.org
`
` by John Jones on 10/12/17. For personal use only.
`
`AKER EXHIBIT 2003 Page 2
`
`

`

`P1: FPP
`July 25, 2000
`
`12:47
`
`AR102
`
`CHAP15
`
`PAF AND RELATED LIPIDS
`
`421
`
`PAF Is a Cell-Associated Signal for Leukocyte Adhesion
`and Activation in Models of Acute Inflammation:
`Juxtacrine Signaling
`In the inflammatory response, PAF has well-characterized actions, mediating cell-
`cell interactions in models of acute and chronic inflammation in virtually all or-
`gans. The cells involved include endothelial cells, leukocytes of several classes, and
`others (5). These studies have shown that PAF can mediate paracrine signaling by
`acting over short distances in solution. In some cases, PAF may also circulate and
`act in an endocrine fashion. The latter mode of action is controlled by plasma PAF
`acetylhydrolase, which limits the half-life of PAF to a few minutes in the blood
`of humans and experimental animals (see below). Experiments in models of acute
`inflammation have also shown, however, that most signaling by PAF may occur be-
`tween closely juxtaposed cells and that it can be recognized by its receptor on target
`cells while associated with the plasma membrane of the signaling cell. Thus, PAF
`can signal in a juxtacrine fashion (Figure 1). These models have also demonstrated
`that PAF acts cooperatively with an adhesion protein, P-selectin, on the endothe-
`lial cell surface and that activation responses triggered by signals delivered via the
`PAF receptor in the target cell further modify the intercellular interaction.
`
`Figure 1 Platelet-activating factor (PAF) signals the priming and activation of leukocytes
`at the surfaces of inflamed human endothelial cells (a juxtacrine system for spatial control of
`inflammation). PAF and an adhesion protein, P-selectin, are coordinately displayed on the
`plasma membranes of stimulated human endothelial cells. P-selectin tethers the leukocyte
`to the endothelial cell, which allows the PAF from the endothelial cell to bind to its receptor
`on the polymorphonuclear leukocyte. This constitutes a form of juxtacrine signaling, which
`may be a general way to spatially restrict the actions of a potent, pleiotropic mediator such
`as PAF. (cid:12)2 integrins are not shown on the polymorphonuclear leukocyte (left side of the
`figure), for convenience. This figure is reproduced from previously published work (10a),
`with permission of the publisher.
`
`Annu. Rev. Biochem. 2000.69:419-445. Downloaded from www.annualreviews.org
`
` by John Jones on 10/12/17. For personal use only.
`
`AKER EXHIBIT 2003 Page 3
`
`

`

`P1: FPP
`July 25, 2000
`
`12:47
`
`AR102
`
`CHAP15
`
`422
`
`PRESCOTT ET AL
`
`PAF had been shown in early studies to be a fluid-phase mediator that could
`activate platelets and various leukocytes in suspension (1). Subsequently, it was
`demonstrated that PAF is synthesized in regulated fashion by endothelial cells
`stimulated with thrombin or other inflammatory mediators and that this PAF ac-
`tivated polymorphonuclear leukocytes (PMNs) (11–13), which are key effector
`cells in the acute inflammatory response. This was the first observation of synthe-
`sis of a signaling factor for PMNs by inflamed endothelial cells, and it established
`a molecular mechanism for local activation of leukocytes at the endothelial sur-
`face rather than requiring the diffusion of chemotactic factors into the blood. This
`local mechanism of leukocyte signaling had been predicted by classic in vivo
`studies but never proven. The adhesion of PMNs in response to PAF is mediated
`by activation-dependent alterations in the affinity and avidity of (cid:12)2 integrins on
`their surface (14). PAF also induces other responses of PMNs that are critical in
`acute inflammation, including cell polarization, enhanced motility, priming for en-
`hanced granular enzyme release, and redistribution of surface ligands (Figure 1).
`In addition, PAF activates other leukocytes involved in inflammation, such as
`monocytes, where PAF signals NF-(cid:20)B translocation to the nucleus and alterations
`in gene expression (15) and other functional responses.
`A key additional observation was that PAF synthesized by stimulated endothe-
`lial cells is not released into solution but that almost all of it remains cell-associated
`even in the presence of albumin, an acceptor molecule to which it binds avidly
`(13, 16, 17). Furthermore, a significant fraction of PAF that is synthesized by stim-
`ulated endothelial cells is translocated to the outer surface of the plasma membrane,
`where it is available for binding to its receptor on target PMNs (18). The specific
`mechanism of translocation to the endothelial surface and whether PAF is local-
`ized in hydrophobic patches or is noncovalently linked to a presenting protein are
`currently unknown. A variety of experimental strategies were used to demonstrate
`that PAF that is endogenously synthesized by endothelial cells and is retained at
`their surfaces ligates the PAF receptor on target PMNs. These strategies included
`receptor blockade, receptor desensitization, and degradation of PAF in the endothe-
`lial plasma membrane with extracellular preparations of PAF acetylhydrolase (18).
`These experiments demonstrated that PAF displayed by inflamed endothelial cells
`has the requisite features of a juxtacrine signaling molecule (19). Subsequent ex-
`periments have confirmed that PAF can operate in this fashion when displayed by
`stimulated endothelial cells both in static systems and when the target PMNs are
`subjected to flow to model in vivo conditions (20–23; reviewed in 24). Recently,
`strategies with exogenous recombinant PAF acetylhydrolase (25) or PAF receptor
`antagonists (26) demonstrated that PAF is a juxtacrine signaling molecule at the
`surfaces of activated human platelets, which—like endothelial cells—are critical
`in cell-cell interactions in inflammatory and thrombotic responses (27, 28). These
`findings suggest that this mode of action, presentation of PAF on the cell surface
`by the cell originating the signal with transfer to an adjacent cell that has a recep-
`tor, may be a general mechanism. If so, it has profound implications for how this
`mediator and other small signaling molecules function in many tissues. In essence,
`
`Annu. Rev. Biochem. 2000.69:419-445. Downloaded from www.annualreviews.org
`
` by John Jones on 10/12/17. For personal use only.
`
`AKER EXHIBIT 2003 Page 4
`
`

`

`P1: FPP
`July 25, 2000
`
`12:47
`
`AR102
`
`CHAP15
`
`PAF AND RELATED LIPIDS
`
`423
`
`it would make the concentration, as determined in biological fluids such as blood,
`irrelevant because the crucial determinant would be its relative abundance in a
`pseudo-two-dimensional space. This could have attractive implications for signal-
`ing events such as occur in the neuronal synapse and other environments where
`spatial limitation of the signal would be important.
`
`Signaling Roles for PAF in Vivo
`Many studies involving exogenous administration of PAF or its endogenous gener-
`ation in experimental animals indicate that it has signaling roles in vivo (reviewed
`in 5). Again, many of these involve models of inflammation or inflammatory injury.
`Recent studies with genetically manipulated mice (29, 30) further document signal-
`ing roles of PAF in vivo. In the first, the guinea pig PAF receptor was overexpressed
`in mice by Shimizu and colleagues (29), who originally cloned the guinea pig re-
`ceptor (31). The transgenic animals had increased mortality when challenged with
`endotoxin, developed melanocytic tumors of the skin, and had increased bronchial
`hyperreactivity in response to inhaled methacholine. Increased bronchial reactivity
`and susceptibility to endotoxin were consistent with previous studies in wild-type
`animals, where there is considerable evidence that dysregulated signaling by PAF
`or PAF-like lipids occurs in endotoxemia and models of asthma (5). Melanocytic
`tumors were, however, unexpected although there is evidence that PAF has neo-
`plastic effects, and it is known to mediate skin inflammation. In a converse strategy,
`the same group created mice with targeted deletions in the PAF receptor (30). These
`PAF receptor knockout animals were developmentally normal and reproduced ef-
`fectively, indicating that some of the actions in which PAF had been implicated pre-
`viously are not essential actions. Future studies with these mice are important be-
`cause more sensitive assays might detect subtle alterations in reproductive capacity,
`neurological function, and other normal physiological events in which PAF might
`play a role. In the reported studies, the knockout animals were tested in a model
`of inflammation and had much milder anaphylactic responses to exogenous anti-
`gen challenge than did wild-type animals, including less cardiovascular instability,
`airway constriction, and alveolar edema. These results were again consistent with
`findings in earlier animal models, involving receptor blockade or other manipula-
`tions of the PAF system, and were also consistent with the respiratory physiology
`of the transgenic mice overexpressing the PAF receptor. In contrast, the knockout
`animals remained susceptible to endotoxin, with vascular and cytokine responses
`equivalent to those in the paired animals with intact PAF receptors. This suggested
`that PAF is not required for endotoxic shock, although it is a modulating signal
`(30). A potentially important variable is that the genetic background of mice used in
`this study was different from that of the transgenic mice that overexpressed to PAF
`receptor.
`In a third study, Shimizu and colleagues examined inflammatory acute lung
`injury caused by acid aspiration in knockout mice and in transgenic mice overex-
`pressing the PAF receptor. The injury was reduced in animals deficient in the PAF
`
`Annu. Rev. Biochem. 2000.69:419-445. Downloaded from www.annualreviews.org
`
` by John Jones on 10/12/17. For personal use only.
`
`AKER EXHIBIT 2003 Page 5
`
`

`

`P1: FPP
`July 25, 2000
`
`12:47
`
`AR102
`
`CHAP15
`
`424
`
`PRESCOTT ET AL
`
`receptor and increased in the overexpressing mice when assessed by histologic and
`physiologic indices and mortality (10). An unexpected finding was that local accu-
`mulation or sequestration of leukocytes in pulmonary vessels of acid-injured mice
`was not different from that of the wild type in either the knockout or transgenic
`animals. However, this is consistent with evidence that there are mechanisms of
`adhesion and accumulation of leukocytes that are independent of activation of (cid:12)2
`integrins in the mouse pulmonary system. The fact that injury was reduced sug-
`gests that signaling by the PAF receptor to priming pathways that enhance granular
`enzyme release or oxygen radical generation (5), both of which are known to cause
`lung injury, is critical in this model.
`Studies in humans also support signaling roles for PAF in vivo. Many reports of
`individual patients and small groups of subjects indicate that dysregulated signal-
`ing by PAF may be a mechanism of disease (5, 32). As examples, developmental
`deficiency of PAF acetylhydrolase may lead to unregulated proinflammatory sig-
`naling by PAF in infants with neonatal necrotizing enterocolitis and in laboratory
`animals with an experimental form of this condition (32–34), and dysregulated sig-
`naling by PAF contributes to the severity of asthma in patients genetically deficient
`in PAF acetylhydrolase (35).
`
`THE PAF RECEPTOR
`
`The intercellular actions of PAF are mediated through a G-protein–linked receptor
`that is expressed on the surface of a variety of cell types (31, 36–38). The human
`gene codes for 342 amino acids, and the sequence is >80% identical to the guinea
`pig protein. After the isolation of the receptor cDNA, the signaling pathway was
`defined in transfection experiments, which complemented previous pharmacolog-
`ical studies (39). Ali et al (40) showed that the signal from the PAF receptor caused
`phosphatidylinositol turnover and raised the intracellular calcium. Both responses
`were inhibited by GTP analogs, but had different responses to pertussis toxin. This
`suggested that there were two different G proteins downstream of the PAF receptor.
`Honda et al (41) used stably transfected cells to assess the role of mitogen-activated
`protein (MAP) kinases and described activation of the 42- and 44-kDa forms of
`ERK by tyrosine phosphorylation when the cells were exposed to PAF. They, too,
`observed increased phosphatidylinositol turnover and found G-protein pathways
`that were sensitive and insensitive to pertussis toxin. They were unable to detect
`activation of ras, which indicated that MAP kinase activation occurred through a
`different pathway (41).
`The structure-function relationships of the PAF receptor have been defined by
`site-directed mutagenesis and transfection (Figure 2). One goal was to identify
`the PAF-binding site, and, based on mutagenesis studies, Ishii et al proposed that
`histidine residues 188, 248, and 249 form a binding pocket (42) and systematically
`mutated all of the polar amino acids in the transmembrane domains of the receptor
`
`Annu. Rev. Biochem. 2000.69:419-445. Downloaded from www.annualreviews.org
`
` by John Jones on 10/12/17. For personal use only.
`
`AKER EXHIBIT 2003 Page 6
`
`

`

`P1: FPP
`July 25, 2000
`
`12:47
`
`AR102
`
`CHAP15
`
`PAF AND RELATED LIPIDS
`
`425
`
`permissionofthepublisher.
`ligandbinding,intracellularsignaling,anddesensitization.Thisfigureisreproducedfrompreviouslypublishedwork(5),with
`cDNAsencodingwild-typeandmutantreceptorsintotargetcellshavedefinedtheregionsofthereceptorresponsiblefor
`Figure2Thestructure-activityrelationshipsoftheplatelet-activatingfactor(PAF)receptor.Studiesusingtransfectionof
`
`Annu. Rev. Biochem. 2000.69:419-445. Downloaded from www.annualreviews.org
`
` by John Jones on 10/12/17. For personal use only.
`
`AKER EXHIBIT 2003 Page 7
`
`

`

`P1: FPP
`July 25, 2000
`
`12:47
`
`AR102
`
`CHAP15
`
`426
`
`PRESCOTT ET AL
`
`(Figure 2) and found that mutations in domains II, III, and VII resulted in higher
`affinities for PAF than the wild-type receptor. Conversely, mutants in transmem-
`brane segments V, VI, and VII had lower affinities (42). The binding affinities
`correlated well with functional responses. A mutation in the third transmembrane
`domain was constitutively active and had lost some of the substrate specificity; the
`mutant receptor responded well to lyso-PAF, which gives little or no response with
`the wild-type receptor. Parent et al (43) examined the transmembrane domains
`and found two adjacent phenylalanines (Figure 2) to be important for structural
`stability of the receptor but, surprisingly, they had no substantial effect on lig-
`and binding (43). They also explored the role of several cysteine residues and
`showed that there likely is a disulfide bond between the conserved residues at
`positions 90 and 163 (44). The cysteine at position 95 was required for optimal
`function, and they concluded that it, too, might be bound to another cysteine.
`In addition to the mutation (N100A) described by Ishii et al, which results in
`constitutive activation of the PAF receptor, Parent et al showed that mutation of
`alanine 230 to glutamic acid also yielded constitutive activity and an increased
`affinity for binding of PAF (45). Mutation of the adjacent residue led to a marked
`decrease in activity. Thus, the central portion of the receptor appears to func-
`tion in ligand binding. This type of binding site is consistent with findings for
`other G-protein-coupled receptors, in which the ligand-binding site is defined in
`three dimensions by amino acids from several of the transmembrane-spanning
`regions.
`Another series of studies defined the portions of the receptor responsible for
`transducing PAF binding into intracellular signals. The third intracellular loop has
`been implicated by studies from multiple laboratories. Carlson et al (46) showed
`that constructs with the sequence from this region could block the response from
`other receptors (this essentially was a “dominant negative” strategy to saturate the
`downstream signaling pathways). They also showed that chimeric receptors that
`included or lacked this region could shift the signaling response. From these ex-
`periments, it seems clear that the third intracellular loop of the receptor is critically
`important for initiating phosphatidylinositol turnover.
`As with other G-protein–coupled receptors, the PAF receptor is desensitized
`after its initial activation. Ali et al (40) found that desensitization of the PAF recep-
`tor was accompanied by phosphorylation, which was partially blocked by protein
`kinase C (PKC) inhibitors. PKC is activated by PAF, so their finding suggested
`a negative feedback mechanism to desensitize the receptor (40). This was ex-
`plored by Takano et al (47), who created a PAF receptor that lacked the C-terminal
`intracellular region and another in which several serine residues and one threo-
`nine residue were replaced with alanine. Both the tail region and these putative
`phosphorylation sites were required for desensitization. Unexpectedly, however,
`pretreatment either with phorbol esters or PKC inhibitors had no effect, which
`indicated that PKC was not the responsible kinase. They also found that a peptide
`representing the carboxyl-terminal 18 amino acids of the receptor was an excellent
`
`Annu. Rev. Biochem. 2000.69:419-445. Downloaded from www.annualreviews.org
`
` by John Jones on 10/12/17. For personal use only.
`
`AKER EXHIBIT 2003 Page 8
`
`

`

`P1: FPP
`July 25, 2000
`
`12:47
`
`AR102
`
`CHAP15
`
`PAF AND RELATED LIPIDS
`
`427
`
`substrate for the G-protein–coupled receptor kinase-1 (47). Taken together, these
`experiments showed that the carboxyl terminus of the PAF receptor is the target for
`phosphorylation and that this is required to desensitize the receptor. The respon-
`sible kinase is likely to be a member of the G-protein–coupled receptor kinase-1
`family.
`At a cellular level there may be additional mechanisms for desensitization of
`responses to PAF—a phenomenon known as heterologous desensitization. In the
`down-regulation described above (homologous desensitization), receptors are de-
`sensitized only by their own ligand (or family members). In contrast, in heterolo-
`gous desensitization, receptors of one type can be down-regulated after activation
`of a different type of receptor. From previous work, it was known that the PAF
`receptor undergoes heterologous desensitization when some peptide receptors are
`activated, and other work showed that this results from PKC-mediated phosphory-
`lation at the carboxyl terminus (48). The converse can also be true—signals from
`the PAF receptor can down-regulate peptide receptors by changes in downstream
`enzymes (49). In summary, the PAF receptor can be desensitized in multiple way;
`homologous desensitization of PAF uses two mechanisms—phosphorylation of
`the receptor (probably a G-protein–coupled receptor kinase-1) and inactivation
`of the key downstream effector PLC-(cid:12)3 by PKC. Heterologous desensitization
`of the PAF receptor uses a third mechanism—phosphorylation of the receptor by
`PKC.
`The structure of the human gene encoding the PAF receptor has been deter-
`mined, and it does not contain introns (50, 51). The gene is on chromosome
`1p35-p34.4 (50, 51). In cells that normally express the PAF receptor, this gene
`seems to be present constitutively, but it can be transcriptionally regulated by
`inflammatory agonists including PAF itself (52–55). There are two different tran-
`scripts, which are found in different tissues. The first transcript described is found
`only in peripheral leukocytes and is the one that increases in response to PAF.
`It has consensus sequences for NF-(cid:20)B, SP-1, and INR, and the response to PAF
`and phorbol esters requires the NF-(cid:20)B consensus binding sites (53). Transcrip-
`tion of the second transcript is induced by phorbol esters, but not PAF, and this
`response requires a functional AP-1 site (53). The second transcript also is reg-
`ulated by retinoic acid and thyroid hormone (56). The cis-acting elements re-
`sponsible for this effect were mapped to three hexamer repeats located between
`−67 to −44 of the transcription start site. Thus, the regulation of expression of
`0
`sequence of transcript 1 re-
`the two transcripts seems to be quite specific—the 5
`sponds to inflammatory stimuli, whereas transcript 2 is regulated by differentiation
`signals.
`It seems likely that the major control of expression of the PAF receptor is at
`the transcriptional level, but one group observed that the PAF receptor mRNA
`levels in human monocytes fell in response to PKC and that the t1=2 for the mRNA
`was markedly decreased (52). They concluded that the stability of the mRNA was
`another potential regulatory mechanism, and this warrants further study.
`
`Annu. Rev. Biochem. 2000.69:419-445. Downloaded from www.annualreviews.org
`
` by John Jones on 10/12/17. For personal use only.
`
`AKER EXHIBIT 2003 Page 9
`
`

`

`P1: FPP
`July 25, 2000
`
`12:47
`
`AR102
`
`CHAP15
`
`428
`
`PRESCOTT ET AL
`
`SYNTHESIS OF PLATELET-ACTIVATING FACTOR
`AND GENERATION OF RELATED COMPOUNDS
`
`Platelet-Activating Factor
`PAF may be synthesized through either of two enzymatic pathways, one defined as
`the remodeling pathway that substitutes an acetyl residue for the long-chain fatty
`acyl residue (Figure 3) of cellular phospholipids. A second de novo pathway to
`form PAF parallels phospholipid synthesis, in which a phosphocholine function is
`transferred to alkyl acetyl glycerol (5). Recent evidence supports the former path-
`way as contributing the bulk of the PAF synthesized in an inflammatory setting.
`PAF, like the eicosanoids, is not stored in a preformed state, but rather is rapidly
`synthesized by inflammatory cells in response to cell-specific stimuli. Stimulated
`PAF synthesis through the remodeling pathway begins with the activation of a
`phospholipase that hydrolyzes phosphatidylcholines to radyl (an inclusive term
`that does not distinguish what type of bond is at the sn-1 position of phospho-
`lipids) lysophosphatidylcholines. This enzyme, cPLA2, is regulated by Ca2C
`and
`by phosphorylation by MAP kinases (57). The stimulation by Ca2C
`occurs at sub-
`micromolar concentrations, and so its activity will be modulated by the intracellular
`Ca2C
`changes found in stimulated cells. This enzyme is unusual in that it displays
`a marked specificity for an sn-2 arachidonoyl residue in contrast to the broadly
`permissive nature of most of the members of the phospholipase A2 super family.
`This enzyme is responsible for nearly all of the arachidonate release necessary for
`eicosanoid synthesis, but it also is an essential first step in the synthesis of PAF.
`Both points were suggested by antisense inhibition of the enzyme in two types
`of cell lines (58, 59) and then firmly established by the nearly complete loss of
`prostanoid (60) and PAF synthesis by the elicited peritoneal macrophages of two
`independently established cPLA2 knockout animals (61, 62). This enzyme, then,
`is essential for arachidonate and radyl glycerophosphocholine release in inflam-
`matory settings. It does not distinguish what structure is present at the sn-1 posi-
`tion (63) and therefore attacks alkyl phosphatidylcholines in addition to the more
`abundant diacyl species. This implies that the initial lysophospholipid products re-
`flect the sn-1 alkyl and acyl arachidonoyl phosphatidylcholine composition of the
`cell.
`The cellular contents of 1-O-alkyl-arachidonoyl-sn-glycero-3-phosphocholine
`and 1-fatty acyl-arachidonoyl-l-sn-glycero-3-phosphocholine vary over a wide
`range. Alkyl phospholipids are trace components of the phosphatidylcholine pool
`of most cells, but, in cells that synthesize PAF such as endothelial cells (64) and
`neutrophils (65), the alkyl phosphatidylcholine content ranges from 10% to 40%,
`respectively, of the total cellular choline phosphoglyceride pool. In leukocytes and
`monocytes, the alkyl phosphatidylcholines are enriched for arachidonate (66, 67),
`where (cid:24)60% of the cellular arachidonate is found in this species. Accordingly,
`this subclass of phosphatidylcholine accounts for 60% of the arachidonate released
`from stimulated cells (66, 67). As would be predicted from the role of cPLA2 in
`
`Annu. Rev. Biochem. 2000.69:419-445. Downloaded from www.annualreviews.org
`
` by John Jones on 10/12/17. For personal use only.
`
`AKER EXHIBIT 2003 Page 10
`
`

`

`P1: FPP
`July 25, 2000
`
`12:47
`
`AR102
`
`CHAP15
`
`PAF AND RELATED LIPIDS
`
`429
`
`andsoarepotentligandsandagonistsofthePAFreceptor.
`fragmentsandmayoxidizethesn-2residue.SomeofthemanyphospholipidoxidationproductscontainthesamefeaturesofPAF
`ashortsn-2residue,andaphosphocholineheadgroup.Uncontrolledoxidation(right)ofalkylpolyunsaturatedphosphatidylcholines
`arachidonoylresidue.ThePAFreceptorrecognizesthreefeaturesofthisphospholipidhormone(shadedarea):ansn-1etherbond,
`synthesisofPAF(left)isacarefullycontrolledprocessinwhichanacetylresidueissubstitutedforapolyunsaturatedsn-2
`Figure3Enzymaticsynthesisofplatelet-activatingfactor(PAF)andnonenzymaticgenerationofPAF-likephospholipids.The
`
`Annu. Rev. Biochem. 2000.69:419-445. Downloaded from www.annualreviews.org
`
` by John Jones on 10/12/17. For personal use only.
`
`AKER EXHIBIT 2003 Page 11
`
`

`

`P1: FPP
`July 25, 2000
`
`12:47
`
`AR102
`
`CHAP15
`
`430
`
`PRESCOTT ET AL
`
`the release of esterified arachidonate in stimulated cells, leukocytes depleted of
`arachidonate by culture in arachidonate-deficient medium showed an 85% de-
`crease in PAF synthesis. All of this shows a strong correlation between PAF and
`eicosanoid synthesis as cells specialized for PAF synthesis store the precursors
`for these two types of lipid messengers together. It also suggests that one way
`to control the formation of PAF, compared with its much less active diacyl ho-
`molog, is by maintaining a high ratio of arachidonate in the sn-2 position of alkyl
`phosphatidylcholines.
`The second and final step in the synthesis of PAF through the remodeling path-
`way is performed by the acetyl-CoA-lysoPAF acetyltransferase. Little is known
`about this activity because this unstable enzyme remains unpurified and uncharac-
`terized, although it is stimulated in a Ca2C
`and phosphorylation-dependent fashion
`(68–70). In leukocytes, the relevant kinase appears to be the p38 MAP kinase (71).
`This acetyltransferase activity possesses little specificity for the sn-1 bond, and
`it will form either PAF from 1-alkyl-sn-glycero-3-phosphocholine (lysoPAF) or
`lysophosphocholine in in vitro assays (72). This lack of substrate selection for the
`sn-1 bond or the presence of two activities is reflected by the relative abundance
`of the acetylated products made by activated cells. For example, the abundance of
`alkyl phosphatidylcholine in endothelial cells is 10% (64), and the relative amount
`of PAF and acylPAF made by these cells is also arou

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket