Sie sind auf Seite 1von 12

Current Pharmaceutical Design, 2004, 10, 577-588 577

Cyclooxygenase Enzymes: Regulation and Function

F.A. Fitzpatrick*

Department of Oncological Sciences & Department of Medicinal Chemistry, Huntsman Cancer Institute, University of
Utah School of Medicine, Salt Lake City, Utah 84112-0555, USA

Abstract: The cyclooxygenase isoenzymes, COX-1 and COX-2, catalyze the formation of prostaglandins, thromboxane,
and levuloglandins. The prostaglandins are autocoid mediators that affect virtually all known physiological and
pathological processes via their reversible interaction with G-protein coupled membrane receptors. The levuloglandins are
a newer class of products that appear to act via irreversible, covalent attachment to numerous proteins. COX enzymes are
clinically important because they are inhibited by aspirin and numerous other non-steroidal anti-inflammatory drugs. This
inhibition of COX confers relief from inflammatory, pyretic, thrombotic, neurodegenerative and oncological maladies.
About one hundred years have elapsed since Hoffman designed and synthesized acetylsalicylic (aspirin) as an agent
intended to lessen the gastrointestinal irritation of salicylates while maintaining their efficacy. During the past forty years
systematic advances in our understanding of the structure, regulation and function of COX isoenzymes have enabled the
design and synthesis of COX-2 selective inhibitors as agents intended to lessen the gastrointestinal irritation of aspirin and
non-selective NSAIDs. This review discusses: 1) how two separate catalytic processes in COX - peroxidase and
prostaglandin synthase - act in an integrated fashion manner to generate prostaglandins; 2) why irreversible inactivation of
COX is important constitutively and pharmacologically; 3) how cells have managed to use two closely related, almost
identical enzymes in ways that discriminate their physiological versus pathological roles; 4) how investigators have used
these advances to formulate and test medically important uses for old drugs (i.e. aspirin) and create new ones that still
seek to achieve Hoffman's original goal.

INTRODUCTION of its flowers, roots, or leaves; or the place that supported its
growth. Considering the immature state of chemistry at that
The cyclooxygenase (COX) enzymes have elaborate
time, physicians had few better alternatives than the Doctrine
roles in human physiology and pathology [1], spanning the
of Signatures as a guide for the discovery of medicines.
cardiovascular [2], neuronal [3], renal [4], immune [5],
Thus, in 1763 Reverend Edward Stone reported to the Royal
gastrointestinal [6] and reproductive systems [7, 8]. Since the Society in London his use of a powder prepared from the
1960's, progressive improvements in our understanding of dried bark of the white willow tree for the effective treatment
their structure, function and regulation have enabled the
of fever in over 50 patients. The Doctrine of Signatures
introduction of many chemical classes of non-steroidal anti-
recommended the willow tree to Dr. Stone because it
inflammatory drugs (NSAIDS) into the pharmacopoeia [9-
flourishes in damp locales where fevers, aches and pains are
11]. The recent discovery and design of the 'coxib' class of
common in the population. Whether divinely ordained or
NSAIDS exemplifies the doctrine of contemporary pharma-
accidental, the Doctrine of Signatures did prompt the identi-
ceutical design - the use of rational, objective, molecular
fication of salicylic acid as the active ingredient of willow;
criteria in an urgent and highly integrated quest for a new
the "design" and synthesis of acetylsalicylic acid (aspirin) to
drug [12, 13]. Few dispute the advantages of this approach, alleviate gastrointestinal toxicity while maintaining salicyl-
especially its superiority over empirical, biological appro-
ate's analgesic efficacy, and ultimately the inauguration of
aches that can distort our comprehension of structure-activity
the modern pharmaceutical industry with formation of Bayer
relationships.
& Co in 1899. After one hundred years aspirin is still widely
Oddly, the COX story begins in a mystical doctrine used as an anti-pyretic, anti-inflammatory and analgesic
popularized by a shoemaker in 17th century Germany. The agent [14].
"Doctrine of Signatures" asserts that God annotated his The discovery of the COX enzymes and the prosta-
herbal creations with a sign (signature) indicating their uses.
glandins (PG) occurred over 60 years after the introduction
Jakob Böhme (1575-1624) based his book "Signatura of aspirin and salicylates into medical practice. When a
Rerum: The Signature of all Things" on mystical visions that
therapeutic agent has a sixty years head start and an
illuminated this ancient idea. Enthusiasts adopted this
established record of efficacy for inflammation, pain and
doctrine for medical purposes, convinced that one could
fever how does understanding its mechanism of action
determine a plant's therapeutic utility from the physical traits matter? This chapter will review our current concepts on the
regulation and function of COX enzymes. First, we will
*Address correspondence to this author at the Department of Oncological present the basic catalytic mechanisms of the COX enzymes
Sciences & Department of Medicinal Chemistry, Huntsman Cancer and their regulation. Second, we will discuss how two
Institute, University of Utah School of Medicine, Salt Lake City, Utah separate catalytic processes - peroxidase and PGH synthase -
84112-0555, USA; Tel: 801-581-6204; Fax: 801-585-0101;
E-mail: frank.fitzpatrick@hci.utah.edu act in an integrated manner. Third, we will discuss the

1381-6128/04 $45.00+.00 © 2004 Bentham Science Publishers Ltd.


578 Current Pharmaceutical Design, 2004, Vol. 10, No. 6 F.A. Fitzpatrick

irreversible inactivation of COX catalysis by constitutive, levels of non-heme iron and trace levels of heme iron;
mechanism-based, "suicide" inactivation and compare this to catalyzed the insertion of the two oxygen molecules
the irreversible inactivation by acetylation of COX. Fourth, necessary for the formation of PGG2 / PGH2 from
we will present some of the lessons learned from the arachidonic acid; and reacted covalently with [3H]
determination of the crystal structure of COX. Fifth, we will acetylsalicylic acid to form a radioactive adduct [28]. The
present a glimpse at the transcriptional and post-translational Unilever group, using a different purification scheme added
regulation of COX expression. This topic is covered in that COX was a glycoprotein with both peroxidase and PG
greater detail elsewhere in this issue of the Journal. Sixth, we synthase activity [29]. They also established that loosely
will summarize some cellular regulatory mechanisms and associated haemin protected the enzyme from inactivation by
functions of the COX enzymes and consider a few peroxide. These two reports laid the foundation for our
paradoxes. For example, do COX enzymes have functions understanding of COX catalysis and its inhibition by
that are unrelated to PG formation? Are all NSAIDS NSAIDs.
equivalent? Seventh, we will summarize how all of this
The availability of methods to purify COX permitted the
detailed information has advanced the design and use of
investigation of its reaction kinetics and its interaction with
NSAIDS in modern medicine. For instance, our detailed NSAIDs. Investigators had noted that certain reducing agents
understanding of how COX enzymes catalyze PG formation stimulated purified COX in a reversible, time-dependent
has enabled the use of low dose aspirin as a unique anti- manner with modification of its Vmax and Km [30].
thrombotic therapeutic entity. Likewise, our detailed
Scientists at Merck studied the stimulation of COX, and its
understanding of the different roles and structures of the inhibition, in an integrated way using structurally distinct
COX-1 and COX-2 isoenzymes has enabled the design of
classes of NSAIDS [31]. Their findings were among the first
coxib class NSAIDS to avoid gastrointestinal toxicity while
to establish that 'stimulators', e.g. phenols, affected both
maintaining analgesic and anti-inflammatory efficacy - the
stimulation and inhibition of COX and that NSAIDS could
identical medical rationale that inspired Hoffman's synthesis
be divided into at least two distinct groups - introducing the
of aspirin in 1897.
notion of inhibitor specificity. Fenamates inhibited COX
weakly in the absence of phenol but potently in its presence.
CATALYTIC MECHANISMS OF CYCLOOXY-
In contrast, aryl propionates (ibuprofen, flurbiprofen) and
GENASE
indomethacin inhibited COX more potently in the absence of
In the sections below the term COX refers to both phenol. The relative potency of COX stimulation by phenol
members of the enzyme family, or to the enzyme used in > norepinephrine > tryptophan > benzoquinone > anisole
investigations that pre-dated the discovery of the COX-2 corresponded with their synergistic effect on the fenamates
isoenzyme. The terms COX-1 and COX-2 refer to the and their antagonist effect on the arylpropionates and
respective isoenzymes. The reader seeking more detailed indomethacin. These data also suggested that constitutive
reviews on the catalytic mechanisms of COX enzymes phenolic anti-oxidants in cells could modulate both COX
should refer to recent publications [15, 16]. activity and its sensitivity to NSAIDS. Intrinsically, this
investigation also established that a better understanding of
COX enzymes interest scientists and physicians because
COX kinetics could provide meaningful and unanticipated
they generate biologically important autocoid lipid
information about NSAIDS.
mediators, the PG and thromboxanes (Tx). How did
investigators unravel the catalytic mechanism of these One of the most important insights into the catalytic
enzymes? Between 1965-1968 investigators at the mechanisms of COX came from studies using twenty carbon
Karolinska Institute and at Unilever Laboratories established polyunsaturated fatty acids (PUFA) with olefin
the absolute configuration of PGs [17] and identified configurations different from arachidonic acid (5,8,11,14-
molecular oxygen and arachidonic acid as co-substrates in cis-eicosatetraenoic acid). Investigators showed that purified
PG biosynthesis [18, 19]. This information, plus peculiar COX, which catalyzed the formation of the PG
kinetics of oxygen consumption during PG biosynthesis, endoperoxides from arachidonic acid (20:4) in a bis-
suggested the existence of transient intermediates in the dioxygenase reaction, also catalyzed the insertion of
biosynthetic pathway, which eventually led to the isolation molecular O 2 into the C-11 carbon of 8,11-cis-eicosadienoic
and structural elucidation of the prostaglandin acid (20:2) in a mono-dioxygenase reaction [32]. Both
endoperoxides, PGG2 and PGH2 in 1973-74 [20, 21]. A oxygenation reactions had similar pH optima, substrate Km
contemporaneous discovery that aspirin abolished PG values, a need for peroxide, and an absolute requirement for
formation [22] prompted the idea that the PG biosynthesis heme. Importantly, NSAIDS inhibited C20:2 oxygenase
enzyme, COX, was a “molecular target” of great medical activity. This investigation implied that abstraction of the 13-
significance. Within 2 years, the discovery of the vasoactive, proS hydrogen from arachidonic acid, followed by C-11
pro-thrombotic substance thromboxane A2 (TxA2) and its oxygenation is one of the rate-limiting steps of COX action.
physiological antagonist, PGI2, as products derived from the It also drew attention to useful precedents with plant
COX catalysis accentuated its importance [23, 24]. lipoxygenase (LOX) enzymes [33] and provided a basis to
understand how COX catalysed the formation of the
In the 1970's investigators at the University of Michigan peroxide bridge between the C-11 and C-9 atoms of
sought experimental conditions for optimizing PG formation arachidonic acid.
by isolated purified COX enzyme [25-28]. The COX enzyme
purified to homogeneity from sheep vesicular glands had a These separate oxygenase activities (11-lipoxygenase and
molecular weight of 70,000 daltons; contained appreciable 15-lipoxygenase) of COX have interested investigators
Cyclooxygenase Enzymes Current Pharmaceutical Design, 2004, Vol. 10, No. 6 579

sporadically and for biological, enzymological, and with the substrate. About 95% of the interactions involve
pharmacological reasons. For instance, COX isolated from hydrophobic residues. Site-directed mutational analyses have
human endothelial arteries converts arachidonic acid into its positioned the residue that abstracts the proS hydrogen from
bis-dioxygenase products PGG2 / PGH2, along with its C-13 of arachidonate (Tyr-385); the residues that orients C-
mono-dioxygenase products 11-hydroxeicosatetraenoic acid 13 of arachidonate for hydrogen abstraction (Gly-533 and
(11-HETE) and 15-HETE [34]. It is uncertain whether these Tyr-348); the residue responsible for the electrostatic
HETEs are biologically relevant. From an enzymological interaction with the carboxylate anion of arachidonate (Arg-
perspective, the formation of multiple products indicates that 120); and lastly the residues that govern the orientation of
catalytic competency depends on the spatial arrangement of arachidonate so that it is optimally arranged to yield PGG2
arachidonic acid within the COX active site [35]. Briefly, (Val-349, Trp-387, and Leu-534). When these latter residues
arachidonate can occupy the active site of COX-1 in at least do not orient arachidonate optimally COX converts it into
three different, catalytically competent arrangements that 11R-HETE, and 15-HETE as mentioned above. Fig. (1)
yield PGG2, 11R-HETE, and 15-HETE, respectively. depicts the interactions between arachidonic acid and the
Arachidonic acid assumes its spatial arrangement after its amino acid residues lining the COX-1 active site and the
entry into the active site. If its arrangement distorts the catalytic reaction. Both isoforms of COX have similar
optimal spacing between C-9 and C-11 the 11-peroxyl amino-acid sequences and catalytic mechanisms. A 3A°
radical cannot attack C-9 to generate an endoperoxide crystal structure of murine apo-COX-2 also affirms that
efficiently. Instead, 11-HpETE is the default product. The arachidonic acid can assume orientations within the active
observation that preparations of COX enzyme could still site that are catalytically incompetent. For instance, the
generate 15(R)-HETE after treatment with aspirin was one of anionic carboxylate end of the substrate can form hydrogen
the important, early clues that there existed a second, unique bonds with Tyr 385 and Ser 530, rather than the predicted
COX isoform, now termed COX-2 [36]. ion pair with Arg120. Instead, the Arg 120 residue contacts
the hydrophobic -end of the substrate [39]. In its
Structural studies indicate that the COX-1 active site is a
hydrophobic channel that protrudes from its membrane- catalytically competent state COX-2 resembles COX-1, with
binding domain into its globular domain [37, 38]. A detailed the carboxylate anion of arachidonic acid coordinated by
crystal structure of Co (3+)-heme COX-1 complexed with Arg 120 and Tyr 355 and the C13 pro (S)-hydrogen atom of
arachidonic acid predicts that 19 active site residues interact the substrate positioned near Tyr 385.

Fig. (1). Amino acid residues in COX 1 that govern substrate positioning and catalysis.
580 Current Pharmaceutical Design, 2004, Vol. 10, No. 6 F.A. Fitzpatrick

BI-FUNCTIONAL CATALYSIS IN A SINGLE attributed to oxidation of the heme group of the peroxidase,
ENZYME PROTEIN: INTEGRATED ACTIONS OF which leads to controlled oxidation of the active site tyrosine
PEROXIDASE AND CYCLOOXYGENASE of COX. An iron-containing heme prosthetic group is the
oxidant that ultimately produces a tyrosine radical. Although
Once investigators established that tyrosine and tyrosyl COX contains Fe+3 in its resting state, the oxidation of Tyr-
radicals initiated, sustained and terminated COX catalysis
385 via the corresponding reduction of Fe3+ to Fe2+ is not
[40, 41] it became possible to clarify how two separate considered favorable. Instead, it is more likely that peroxides
catalytic processes - peroxidase and PG synthase - work
convert the heme of COX into a ferryl-oxo complex, which
coordinately to manage a radical reaction within a single
is more likely to oxidize Tyr-385.
COX enzyme [42].
There have been several insights into the physical
COX has an intrinsic peroxidase activity [25, 28, 29].
chemical and enzymatic role of radicals in COX catalysis
Early experiments, using limited proteolysis of COX,
since the initial report on their occurrence [40]. For instance,
investigated whether the peroxidase and COX catalytic
purified COX-1 and COX-2 enzymes that have been
processes occurred at separate or overlapping sites in the
reconstituted with manganese protoporphyrin IX and probed
enzyme. Digestion of purified COX enzyme with trypsin with peroxides yield electron paramagentic resonance (EPR)
generated an amino terminal 33 kDa fragment and a 38 kDa spectra that reflect radical-dependent oxidation [48]. This
fragment. When purified neither the 33- nor 38-kDa study indicated that the peroxide-generated radicals in COX-
fragment had PGH synthase activity; however, treatment
2 and MnCOX-1 each form the same carbon-centered
with [3H]acetylsalicylic acid introduced [3H]acetyl substi- arachidonic acid radical that subsequently reacts with oxygen
tuents into the 38-kDa fragment, but not the 33-kDa
to the lipid hydroperoxide. The EPR data also indicate that
fragment suggesting a prominent part of PG synthase active
the metastable arachidonate radical contains a planar
site occurred within this fragment [43]. Subsequent studies
pentadienyl radical, as widely speculated.
by the same group showed that the peroxidase activity
declined rapidly, corresponding to the rate of proteolysis Given the similarities between their primary sequences,
[44]. Spectrometric analyses suggested that proteolysis their crystal structures and their catalytic mechanisms the
decreased heme binding at a site that controls peroxidase constitutive regulation of COX-1 and COX-2 catalysis is
activity. Since trypsin cleaved COX between Arg253 and similar with only a few nuances. For instance, catalysis by
Gly254 the investigators proposed that the heme responsible the COX-2 isoenzyme can proceed at hydroperoxide levels
for the peroxidase activity of COX bound to the His residue 10-fold lower than those needed to promote catalysis by
of the sequence His250-Tyr251-Pro252-Arg253. A compar- COX-1. Investigators have speculated that this may be a
able study affirmed that the two separate enzymatic pro- mechanism to generate PG preferentially by COX-2 when
cesses could be differentiated. Only the PG synthase activity both isoforms occur in the same intracellular compartment
was lost during catalysis; the peroxidase activity was [49, 50]. It is unclear why cells would find this advanta-
retained. By contrast, only the peroxidase activity was lost geous. Likewise, the structural stability of COX-2, both in
by limited proteolysis with trypsin, suggesting the presence the active site regions and in the subunits overall, is less than
of separate PG synthase and peroxidase structural domains in that of COX-1 [51]. This may relate to their different
the COX enzyme [45]. It is important to note that one preferences for fatty acid substrates and inhibitors and the
investigator has reported the opposite result [46]. Namely, larger size of the pocket in the active site of COX-2. A valine
proteolytic attack on the Arg 253 region of COX diminished residue at position 509 in COX-2 corresponds to the isoleu-
both PG synthase and peroxidase activities together. The cine residue at position 523 in COX-1. However, mutage-
reason for the discrepancy is uncertain. nesis studies in both COX-1 and COX-2 suggest that
exchange of valine and isoleucine at position 509/523 does
Improved methods for quantifying peroxidase activity
not account fully for all the differences in inhibitor
fortified the notion of a regulatory relationship between the
specificity between the isoforms [52]. Thus, differences in
peroxidase and PG synthase activities of the COX enzyme
structural stability may contribute to the ligand selectivity in
[47]. Kinetic and chemical studies established that the the respective enzyme active sites.
peroxidase in COX is a classical heme peroxidase.
Comparison of a series of phenols, aromatic amines, beta-
IRREVERSIBLE INACTIVATION OF COX: CONSTI-
dicarbonyls, naturally occurring compounds, and NSAIDS TUTIVE 'SUICIDE' INACTIVATION
indicated differences in their ability to act as reducing
substrates. Importantly, there was no structure-activity As discussed above COX enzymes use peroxides and
correlation between agents that sustain peroxidase activity peroxidase in a radical dependent mechanism to stimulate
and agents that inhibited COX. Thus, pharmacological data their initial rate of catalysis. Once these processes reach
implied that the efficient hydroperoxide-reducing activity maximal velocity how can they be blunted? “Suicide”
(peroxidase) might be responsible for the hydroperoxide inactivation, or mechanism-based inactivation, appears to be
dependent stimulation of COX during the initial phase of one of the rate-limiting processes governing COX.
catalysis, as well as protection against hydroperoxide- Investigators described this aspect of COX catalysis in some
dependent inactivation of COX during the sustained phase of of the earliest studies of the enzyme [26]. It is now known
catalysis. that many other eicosanoid biosynthetic enzymes undergo
"suicide" inactivation, e.g. Tx synthase [53], PGI synthase
Thus, the current mechanistic model places peroxidase
[54], LTA4 hydrolase [55, 56], 5-LOX [57- 59], 12-LOX
and COX catalysis at separate but electronically interrelated
active sites. The stimulating effect of peroxides is best [60] and 15- lipoxygenases [61-63]. Studies on 15-LOX may
Cyclooxygenase Enzymes Current Pharmaceutical Design, 2004, Vol. 10, No. 6 581

be particularly relevant because COX carries out this same the enzyme and LG generated from PGH2 form covalent
reaction. Suicide inactivation is a cumbersome regulatory adducts with a nearly 100% reaction yield, consistent with
process; yet COX manages to use metastable radical inter- the reactivity of these electrophiles. Importantly, the lysyl-
mediates for catalysis that generates reactive hydroperoxide LG adducts retain reactivity so the Schiff base adduct with
or endoperoxide products (PGG2 and PGH 2). Thus, it is not lysine can undergo nucleophilic attack, and the oxidation of
surprising that COX enzymes have consigned the chemical the pyrrole adducts generates sites of electrophilic reactivity.
intermediates in their Michaelis complex to two separate Thus, lysyl-LG adducts can form covalent bonds with other
fates. One fate is a normal catalytic cycle yielding product nucleophiles, leading to intra- or even intermolecular cross-
and regenerating enzyme. A second fate is a catalytic cycle linking. It is likely that some of the unidentified PGH2
leading to irreversible inactivation of the enzyme. adducts reported in earlier studies are actually LG adducts.
The site of LG attack on COX is uncertain, as is any
The first detailed studies of COX auto-inactivation, by
relationship between adduct stoichiometry and COX
the Merck group, showed that the generation of PGH2 from
inactivation. This type of reaction does occur in cells [70]
arachidonic acid released oxidizing equivalents. Phenol and
and tissue [71]. Thus, separate from its relevance to auto-
methional, which quench oxygen-centered radicals,
increased the initial rate and the extent of catalysis prior to inactivation of COX this process must now be considered as
COX inactivation. Hence, it appeared that the COX was a significant aspect of many COX mediated maladies,
irreversibly self-deactivated due to its oxidation by oxygen- especially neurodegenerative disorders and cancer.
centered radicals that formed as a result of the peroxidase- “Suicide” inactivation of COX has some implications
dependent reduction of the hydroperoxide on PGG2 [64]. that have not yet been fully appreciated, or fully integrated,
This information aligns well with our current understanding into models of eicosanoid biosynthesis. For example, efforts
of how peroxide → peroxidase → tyrosine → arachidonic to correlate the amount of cellular COX enzyme with the
acid propagate radical-dependent oxidation reactions. amount of product, e.g. PGE2, need to take into account the
However, it is doubtful that this is the only chemical process catalytic history of the enzyme. For instance, if cells
that participates in the modification and inactivation of accumulate COX-2 protein via de novo synthesis and if this
COX. For example, PGG2 and PGH2 may react covalently COX-2 propagates catalysis continuously then there should
with proteins [65 -67]. Likewise, hydroperoxy-eicosate- co-exist two “pools” of enzyme - a pool that retains catalytic
traenoic acids, like the 15-HpETE formed by COX-1 or activity and a pool with depleted catalytic activity. Right
COX-2 under certain conditions may be able to oxidize now there is very little known about the intracellular
methionine residues [68]. It is not known if this actually degradation of COX-2 protein. How is auto-inactivated COX
occurs in COX but a methionine residue at position 522 recognized and targeted for degradation? Is this different
could be an attractive candidate for such a reaction. from degradation of active COX? Does cellular
accumulation of “ suicide” inactivated enzyme account, in
Recently, investigators from Vanderbilt University have
part, for any poor correlation between protein expression and
provided a novel hypothesis relevant to the catalysis-
product formation (e.g. COX-2 protein versus PGE2)? If
dependent, irreversible inactivation of COX [69]. The
cells fail to degrade suicide-inactivated enzymes are there
endoperoxide PGH 2, can rearrange spontaneously into the γ-
keto aldehydes, levuglandin (LG) E2 and LGD2. Fig. (2) any pathological consequences? The identification of LG
depicts their structures. LG can react with the ε-amino group adducts to COX should allow investigators to address these
questions experimentally.
of lysine to form both the lysyl-levuglandin Shiff base and
the pyrrole-derived lysyl-levuglandin lactam adducts. These Analogous to its PG synthase activity, the peroxidase
lysyl-LG adducts form on COX following its oxygenation of activity of COX also has a self-limited number of catalytic
arachidonic acid. Approximately 25-20% of the arachidonic turnovers. Data suggest that the auto-inactivation process
acid provided to COX becomes covalently associated with originates with an enzyme (Intermediate II), which contains

O
COOH

O
OH
PGH2

O O

COOH COOH

O OH O OH
LGD2 LGE 2

Fig. (2). Structures of levuloglandins derived from PGH2.


582 Current Pharmaceutical Design, 2004, Vol. 10, No. 6 F.A. Fitzpatrick

an oxyferryl heme and a tyrosyl radical that is converted Functionally, inhibition of COX by aspirin is "reversed"
quickly (k = 0.5-2 s-1) to an Intermediate III, which retains an only when de novo protein synthesis replaces inactive
intact heme group that is not covalently linked with COX-1 enzyme with newly translated enzyme. This has had
This implies that the heme group in Intermediate III is not profound implications for the use of aspirin in thrombosis
properly coordinated to the histidine ligand(s) but still because platelets, which lack a nucleus, cannot regenerate
remains non-covalently associated with the protein [72]. COX [78-81]. It is now incontestable that aspirin reduces the
Irreversible heme destruction occurs during COX catalysis, incidence of vascular occlusive events in patients at risk of
but at a much slower rate than the conversion of Intermediate thrombotic complications and that its survival benefit persist
II to III. Others have substantiated this model of COX through ten years of follow-up [82, 83]. These beneficial
inactivation but provided some important insights. Under effects are due to suppression of platelet aggregation from
conditions where COX inactivation is complete the enzyme the permanent depletion of COX activity for the full lifespan
retains ∼ 60% of its peroxidase activity. The rates of of a platelet (∼ 6 days). The rational use of low-dose aspirin,
peroxidase inactivation were indistinguishable for native which spares the gastrointestinal tract while maintaining
COX-1 and an Y385F mutant which cannot form the Tyr385 anti-thrombotic efficacy, results directly from understanding
radical of COX Intermediate II suggesting that another the molecular mechanism of aspirin in detail and the
species, corresponding to Intermediate III with radical availability of quantitative methods to relate biochemical
centered on a tyrosine other than Tyr385, is the precursor for effects with clinical effects. Whether measured in economic
peroxidase inactivation [73]. or human terms the detailed knowledge of COX catalysis
Overall, COX-1 and COX-2 have adapted complex and its relationship to vascular biology has been a
remarkable medical achievement.
chemical processes for the practical purpose of regulating
their own activation and inactivation in an orderly way. In Can one design an irreversible inhibitor based on selec-
this regard they are among nature's most chemically tive acetylation of the COX-2 active site? Investigators from
sophisticated creations. Vanderbilt University have made progress toward this goal
with their design and synthesis of o-(acetoxyphenyl)hept-2-
IRREVERSIBLE INACTIVATION OF COX: PHAR- ynyl sulfide (APHS) which inhibits COX-2 selectively (100
MACOLOGICAL ACETYLATION × vs. COX-1) [84]. Structure-activity relationships (SAR)
indicate that the S-alkyl chain length is a determinant of
Irreversible inactivation of COX via acetylation is a less
potency, with a heptyl chain optimal. Insertion of a triple
complex phenomenon chemically but a more significant one
bond into the heptyl chain enhanced potency and selectivity.
pharmacologically. This reaction accounts for the unique
Sulfides were also more potent and selective COX-2
value of aspirin in cardiovascular medicine and it has
inhibitors than the corresponding sulfoxides or sulfones or
allowed investigators to unmask some notable differences
other heteroatom-containing compounds. This novel class of
between the active sites of COX-1 and COX-2. covalent modifiers may serve as potential therapeutic agents
Shortly after investigators proposed that aspirin inhibited in inflammatory and proliferative disorders without gastro-
COX [22] investigators at Washington University showed intestinal or hematologic side effects [85]. The optimization
that aspirin irreversibly acetylated COX in microsomes of of the clinical pharmacology of these compounds should
seminal vesicles and human platelets [74]. The rate and prove interesting because in pathological states cells may
stoichiometry of COX acetylation correlated precisely with induce COX-2 expression continuously. In other words, the
inhibition of COX activity. At concentrations correlated with half-life of COX-2 protein may govern the dosing schedule,
its Michaelis constant the substrate, arachidonic acid, not the half-life of the compound. Clinical assessment of
prevented COX acetylation by aspirin. A reversible COX these novel agents should be an important source of
inhibitor indomethacin also inhibited the acetylation information on how expression and degradation of COX-2
reaction. These data established that aspirin exerts its anti- change in disease.
inflammatory and anti-platelet effects by acetylating the
As mentioned above, the observation that COX enzyme
COX active site. Subsequent investigations established the could still generate 15(R)-HETE after treatment with aspirin
serine 530 residue in the active site as the one modified coincided with the discovery a second, unique COX isoform,
[75,76] and, incidentally, reinforced the fact that the PG
now termed COX-2 [36, 86]. Thus, some of the earliest
synthase activity and peroxidase activity of COX were known distinctions between COX-1 and COX-2 came from
distinct and separable, pharmacologically. Treatment of investigations using aspirin as a probe. Detailed investigation
COX with an appreciable excess of other acetylating agents, of the stereospecific conversion of arachidonic acid into
such as N-acetylimidazole, can inactivate both COX and
15(R)-HETE has provided a glimpse at the alignment of
peroxidase activity. However, COX acetylated by N-
substrate within the active site of COX-2. Remarkably, the
acetylimidazole contains acetyl adducts throughout the hydrogen abstraction leading to formation of 15R-HETE by
protein. Thus, binding of the salicylate portion of aspirin in aspirin-treated COX-2 is identical to the hydrogen
the active site of COX governs its tendency to react solely at
abstraction leading to PG formation. These data imply that
Ser530 [77]. The acetylated serine 530 residue is not readily
the respective catalytic reactions must involve significantly
reversed by hydrolysis - this is consistent with our current different stereochemistry because the 15(S) hydroxyl groups
understanding that the COX active site is a hydrophobic in PG have the opposite configuration as the 15(R) hydroxyl
pocket that evolved to accommodate a very hydrophobic
groups in 15(R)-HETE. Among three models considered to
substrate - see Fig. (1). Hydrolysis is unlikely in such
explain these data, the best one predicts that acetylation of
conditions and the aspirin effect is difficult to reverse. Ser-516 in COX-2 limits the space available to accommodate
Cyclooxygenase Enzymes Current Pharmaceutical Design, 2004, Vol. 10, No. 6 583

the ω end of the arachidonic acid carbon chain [87, 88]. With acetylating its active site Ser 530 residue the enzyme cannot
intact COX-2, hydrogen abstraction and O2 insertion form any PG or other oxygenated lipids. When one inhibits
generate hydroperoxy substituents with 11(R) - and 15(S)- COX-2 by acetylating its active site Ser 516 the enzyme
configurations. With acetylated COX-2, steric hindrance by cannot form any PG but it can form 15(R)-HETE. It is will
the acetyl group distorts the normal arrangement of be interesting to ascertain whether 15(R) HETE contributes
arachidonic acid within the active site, displacing the ω end in any way to the pharmacological actions of aspirin or
of its carbon chain. Insertion of O2 into the distorted APHS [84, 85]. Although speculative, it seems possible that
arachidonate from the conventional direction then generates one might achieve a full, irreversible inactivation COX-2
the 15(R)- configuration. The distortion may also render the with other o-(alkyloxyphenyl)hept-2-ynyl sulfide that added
C-11 carbon so inaccessible to O2 that no PG can form. This an acyl group larger than acetyl to Ser 516. For instance, o-
result departs from the so-called "antarafacial rule" typifying (2-propionyloxyphenyl)hept-2-ynyl sulfide or o-(2-methyl-
the dioxygenase reactions of LOX and COX enzymes. This butyloxyphenyl)hept-2-ynyl sulfide might be candidates to
rule specifies that the enzyme abstract hydrogen from one test this hypothesis and explore whether elimination of PG
side of a planar 1,4-cis-pentadiene and insert O2 from the formation is sufficient to neutralize COX or whether
other as shown in Fig. (3). However, this rule and elimination of PG plus 15(R)-HETE is advantageous. The
terminology only apply when the 1,4-pentadiene is in a distinctive effects of aspirin on COX-2 catalysis may be
planar conformation [87]. relevant to paradoxical reports that aspirin does not protect
against intestinal tumorigenesis in mice that harbor defective
Fig. (4) depicts the two cases outlined above, along with
alleles of the adenomatous polyposis coli (APC) tumor
an hypothetical third case. When one inhibits COX-1 by suppressor.

H H
O O
O O O OOH
O H
O -H +H
O

CO2H CO2H
CO2H

Fig. (3). Antarafacial relationship between hydrogen abstraction from C13 and O2 insertion.

COX-1 + Aspirin AA
O
No products
O-C-CH3
Ser 530

COX-2 + Aspirin or APHS


No PG
AA + O2 15R-HETE ….. Function ?
O
O-C-CH3
Ser 516

COX-2 + alkoxyPHS ? AA

No products ?
O alkyl
O-C
Ser 516
Fig. (4). Effect of acetylation on substrate access to COX-1 and COX-2. Speculative effect of acylation on COX-2.
584 Current Pharmaceutical Design, 2004, Vol. 10, No. 6 F.A. Fitzpatrick

Readers interested in the coxib class of COX-2 selective expression of Tx synthase by platelets and restricted
inhibitors and the basis for inhibitor selectivity should see expression of PGI2 synthase by endothelial cells account for
the review by Patrignani in this issue as well as the following the medical benefits of low-dose aspirin in thrombo-
[89- 92]. occlusive disorders [98] and, in part, for the functional
antagonism between platelets and endothelium in vascular
LESSONS FROM THE X-RAY CRYSTAL STRUC- biology [99-101]. Restricted expression of 5-LOX in
TURES OF COX-1 AND COX-2 granulocytes is also critical for transcellular biosynthesis of
Investigators have obtained X-ray crystal structures for leukotrienes by combinations of granulocytes, erythrocytes,
COX-1 and COX-2 at about 3 angstroms resolution [93-96]. platelets and endothelium [102-104]. Numerous investigators
One lesson derived from these crystal structures is that have contributed to our current understanding of restricted
conventional biochemical, pharmacological and molecular (i.e. inducible) expression of the COX-2 isoenzyme [105-
biological approaches had provided a very accurate and 109]. Of particular note are the series of studies by
precise picture of the COX molecules. These crystal Needleman and colleagues who first showed convincingly
structures have confirmed previous held concepts such as the that in most cells and tissues there is a constitutive COX
hydrophobic nature of the COX active site, the acetylation of which is unaffected by steroids, while in inflammatory cells
serine residues within the active site as a basis for aspirin and inflamed tissue there is an inducible COX regulated by
inhibition, the relative differences in the volumes of the glucocorticoids [110-112]. Under normal physiological
active sites of COX-1 versus COX-2, the existence of two conditions glucocorticoids restrain inducible COX
separate sites for peroxidase and PG synthase catalysis, the expression. Depletion of glucocorticoids or accumulation of
basis for salicylate antagonism of aspirin acetylation, and cytokines and interleukins causes induction of COX. The
other notions. A second lesson is that the progressive motivation to discover therapeutic substitutes for
improvement in the selectivity of inhibitors for COX-2 glucocorticoids and the eventual design of COX-2 selective
versus COX-1 could not be obtained without the molecular inhibitors owes much to these studies.
coordinates and distance data from crystal structures. A third COX-2 induction involves the orchestration of
lesson is that we are still using the doctrine of signatures - membrane-to-nucleus signaling processes such as tyrosine
we just rely on more sophisticated technology to observe the kinases, protein kinase C isoforms, peroxisome proliferator
signs. activated receptor, ras, NF-κB, and other transcription
factors [113]. Many of these converge on the mitogen
CELLULAR REGULATION OF COX PROTEIN activated protein kinase (MAPK) pathway involved in the
EXPRESSION AND FUNCTION transcriptional regulation of inflammatory genes [114]. It is
A study in 1989 [97] provided a glimpse at a regulatory now established that cells modulate COX-2 expression by
process that has assumed great importance for the COX transcriptional and post-transcriptional mechanisms that
enzyme family. Investigators at Upjohn showed that a affect mRNA stability [115, 116].
typical growth factor, platelet-derived growth factor (PDGF), Presumably, high expression of COX-2 in many tumors
caused a sustained increase in PGE2 biosynthesis, derives, in part, from somatic mutations that persistently
commencing within 10 min, reaching a maximum after 2 hrs activate signaling pathways for COX-2 expression (e.g. ras,
and enduring beyond 2 hrs in NIH 3T3 cells. Second, they various growth factor receptor tyrosine kinase pathways).
showed that cycloheximide inhibited > 90% of the PGE2 However, tumors are also susceptible to dramatic changes in
synthesis initiated by PDGF, suggesting that de novo protein the methylation of cytosine in the CpG islands of their
synthesis, presumably de novo synthesis of a COX enzyme genome. It now appears COX-2 expression may be
was responsible. However, additional experiments in which "silenced" epigenetically. Investigators have reported that
NIH 3T3 cells were exposed to exogenous arachidonic acid, methylation of CpG islands near exon 1 silenced COX-2
instead of PDGF, showed that these cells had ample basal expression in 13% of sporadic colorectal cancers and 14% of
capacity for COX catalyzed formation of PGE2. colorectal adenomas [117, 118]. This may have medical
Furthermore, the steady-state level of COX mRNA in NIH implications. Specifically, adenomas with a methylation-
3T3 cells rose, but this rise followed, rather than preceded, silenced COX-2 gene may have a diminished response to
the increase in PGE2 synthesis, as one would have expected. treatment with COX-2 specific inhibitors or a better
Finally, COX protein levels did not rise appreciably prognosis, depending on what other genetic defects are
following PDGF stimulation, even when the corresponding harbored by the tumor. Several types of neoplastic and pre-
mRNA levels were elevated. These data prompted two neoplastic tissue overexpress COX-2, compared to adjacent
questions. First, why would cells need de novo synthesis of non-involved tissue, with incidences ranging from ∼ 50% -
additional COX enzyme if they already had ample COX to 90% in studies involving a minimum of 10 subjects [119-
convert any available arachidonic acid into PGE2? Second, if 124]. Thus, the converse may also be true: the extensive
de novo synthesis of COX enzyme did account for increased hypomethylation often seen in the genome of tumors may
PGE2 formation in cells incubated with PGDF, why were the release "silenced" COX-2. Investigations with the Min/+
temporal and stoichiometric relationships between mRNA mouse model of intestinal polyposis suggest that COX-2
accumulation, protein accumulation and PGE2 formation so expression is a significant factor that causes progression
distorted? Discovery of the COX-2 isoenzyme helped clarify from a pre-neoplastic to a neoplastic condition. For instance,
these paradoxes [86]. homologous disruption of the COX-2 gene diminished polyp
Generally, restricted enzyme expression is a prominent formation in Min/+ mice by ∼ 80% [125]. Investigations
mechanism for regulating eicosanoid biosynthesis. Restricted with Min/+ mice also suggest that the COX-1 isoenzyme
Cyclooxygenase Enzymes Current Pharmaceutical Design, 2004, Vol. 10, No. 6 585

participates in tumorigenesis. Episodically, investigators [3] Graham SH, Hickey RW. Cyclooxygenases in central nervous
have reported that stromal tissue adjacent to tumor harbors system diseases: a special role for cyclooxygenase 2 in neuronal
cell death. Arch Neurol 2003; 60: 628-30.
elevated levels of the COX-1 isoenzyme, not COX-2 [126]. [4] FitzGerald GA. The choreography of cyclooxygenases in the
On the one hand, such reports reinforce the notion that kidney. J Clin Invest 2002; 110: 33-4.
disease-restricted expression of COX-2 is associated [5] Tilley SL, Coffman TM, Koller BH. Mixed messages: modulation
explicitly with the disease at the cellular level. On the other of inflammation and immune responses by prostaglandins and
thromboxanes. J Clin Invest 2001; 108:15-23.
hand, such reports imply that adaptation to disease alters [6] Peura DA. Gastrointestinal safety and tolerability of nonselective
expression of COX in proximal cells. nonsteroidal anti-inflammatory agents and cyclooxygenase-2-
selective inhibitors. Cleve Clin J Med 2002; 69: Suppl 1:SI31-9.
Cells regulate COX-1 and COX-2 by intracellular [7] Kniss DA. Cyclooxygenases in reproductive medicine and biology.
compartmentation, accessory proteins, arachidonate levels, J Soc Gynecol Investig 1999; 6: 285-92.
and availability of hydroperoxide activator as discussed [8] Lim H, Paria BC, Das SK, Dinchuk JE, Langenbach R, Trzaskos
elsewhere [127, 128]. The O2 tension in most tissues exceeds JM, et al. Multiple female reproductive failures in cyclooxygenase
the Km for COX by at least 6-fold and there are few 2-deficient mice. Cell 1997; 91:197-208.
[9] Smith WL, DeWitt DL, Garavito RM. Cyclooxygenases: structural,
circumstances where O2 levels would be insufficient to cellular, and molecular biology. Annu Rev Biochem 2000; 69:145-
sustain a high rate of prostaglandin biosynthesis [129]. 82.
However, the core of solid tumors is often hypoxic and this [10] Kurumbail RG, Kiefer JR, Marnett LJ. Cyclooxygenase enzymes:
may be one of those circumstances. The nitric oxide radical catalysis and inhibition. Curr Opin Struct Biol 2001; 11: 752-60.
[11] Thun MJ, Henley SJ, Patrono C. Nonsteroidal anti-inflammatory
(NO) often occurs together with PG. Given the role of drugs as anticancer agents: mechanistic, pharmacologic, and
radical dependent reactions and radical intermediates in clinical issues. J Natl Cancer Inst 2002; 94: 252-66.
COX catalysis it is not surprising that there are some reports [12] Isakson P, Seibert K, Masferrer J, Salvemini D, Lee L, Needleman
that NO can stimulate COX activity by a direct interaction or P. Discovery of a better aspirin. Adv Prostaglandin Thromboxane
by a mechanism that involves peroxynitrite (from O2.-+NO.) Leukot Res 1995; 23: 49-54.
[13] Prasit P, Wang Z, Brideau C, Chan CC, Charleson S, Cromlish W,
reacting directly with COX [130]. et al. The discovery of rofecoxib, [MK 966, Vioxx, 4-(4'-
methylsulfonylphenyl)-3-phenyl-2(5H)-furanone], an orally active
PARADOXES AND CONTROVERSIES cyclooxygenase-2-inhibitor. Bioorg Med Chem Lett 1999; 9:1773-
8.
FDA approved COX-2 selective NSAIDs should have no [14] Elwood PC. Aspirin: past, present and future. Clinical Medicine
greater liability than non-selective NSAIDS. However, this 2001; 1: 132-137.
has been controversial and the debate ranges from their [15] Smith WL, Song I. The enzymology of prostaglandin endoperoxide
H synthases-1 and -2. Prostaglandins Other Lipid Mediat 2002;
relative superiority as gastric sparing NSAIDs [131] to their 68:115-28.
cardiovascular and renal safety [132]. Mercifully, for those [16] Marnett LJ, Rowlinson SW, Goodwin DC, Kalgutkar AS, Lanzo
interested in medical science, this debate can be informed by CA. Arachidonic acid oxygenation by COX-1 and COX-2.
our appreciable understanding of the role of COX-1 and Mechanisms of catalysis and inhibition. J Biol Chem 1999; 274:
22903-6.
COX-2 isoforms. [17] Nugteren DH, Van Dorp DA, Bergstrom S, Hamberg M,
Other paradoxes are more topical and more interesting. Samuelsson B. Absolute configuration of the prostaglandins.
Nature 1966; 212: 38-9
Several groups have now reported that aspirin has no effect [18] Nugteren DH, van Dorp DA. The participation of molecular
on tumour number and size when administered to Min/+ oxygen in the biosynthesis of prostaglandins. Biochim Biophys
mice with established polyposis - its effect is only evident Acta 1965; 98: 654-6.
when administered during gestation or prior to weaning [19] Lands WE, Samuelsson B. Phospholipid precursors of
[133-135]. Likewise, there are reports that overexpression of prostaglandins. Biochim Biophys Acta 1968; 164: 426-9.
[20] Nugteren DH, Hazelhof E. Isolation and properties of intermediates
COX-2 induces cell cycle arrest via a prostaglandin- in prostaglandin biosynthesis. Biochim Biophys Acta 1973; 326:
independent mechanism [136]. These challenge current 448-61.
concepts about the role of COX-2 in neoplasia and the role [21] Hamberg M, Svensson J, Samuelsson B. Prostaglandin
of NSAIDS in cancer chemoprevention. Paradoxes like these endoperoxides. A new concept concerning the mode of action and
release of prostaglandins. Proc Natl Acad Sci USA 1974; 71: 3824-
assure continued research on COX enzymes. 8.
[22] Ferreira SH, Moncada S, Vane JR. Indomethacin and aspirin
ACKNOWLEDGEMENT abolish prostaglandin release from the spleen. Nature New Biol
1971; 231: 237-9.
F.A. Fitzpatrick is the Dee Glenn & Ida Smith Chair of [23] Hamberg M, Svensson J, Samuelsson B. Thromboxanes: a new
Cancer Research and an investigator of the Huntsman group of biologically active compounds derived from prostaglandin
Cancer Institute. This work was supported by NIH AI 26730. endoperoxides. Proc Natl Acad Sci USA 1975; 72: 2994-8.
[24] Moncada S, Gryglewski R, Bunting S, Vane JR. An enzyme
isolated from arteries transforms prostaglandin endoperoxides to an
REFERENCES unstable substance that inhibits platelet aggregation. Nature 1976;
263: 663-5.
References 137-139 are related articles recently published in [25] Smith WL, Lands WE. Stimulation and blockade of prostaglandin
Current Pharmaceutical Design. biosynthesis. J Biol Chem 1971; 246: 6700-2.
[26] Smith WL, Lands WE. Oxygenation of polyunsaturated fatty acids
[1] Williams CS, Mann M, DuBois RN. The role of cyclooxygenases
during prostaglandin biosynthesis by sheep vesicular gland.
in inflammation, cancer, and development. Oncogene 1999; 18:
Biochemistry 1972; 11:3276-85.
7908-16.
[27] Rome LH, Lands WE. Properties of a partially-purified preparation
[2] Lettino M, Cantu F, Mariani M. Cyclo-oxygenase-1 and cyclo-
oxygenase-2 and cardiovascular system. Dig Liver Dis 2001; 33: of the prostaglandin-forming oxygenase from sheep vesicular
gland. Prostaglandins 1975; 10: 813-24.
Suppl 2:S12-20.
586 Current Pharmaceutical Design, 2004, Vol. 10, No. 6 F.A. Fitzpatrick

[28] Hemler M, LandsWE. Purification of the cyclooxygenase that [49] Kulmacz RJ, Wang LH. Comparison of hydroperoxide initiator
forms prostaglandins. Demonstration of two forms of iron in the requirements for the cyclooxygenase activities of prostaglandin H
holoenzyme. J Biol Chem 1976; 251: 5575-9. synthase-1 and -2. J Biol Chem 1995; 270: 24019-23.
[29] Van der Ouderaa FJ, Buytenhek M, Nugteren DH, Van Dorp DA. [50] Lu G, Tsai AL, Van Wart HE, Kulmacz RJ. Comparison of the
Purification and characterisation of prostaglandin endoperoxide peroxidase reaction kinetics of prostaglandin H synthase-1 and -2. J
synthetase from sheep vesicular glands. Biochim Biophys Acta Biol Chem 1999; 274: 16162-7.
1977; 487: 315-31 [51] Xiao G, Chen W, Kulmacz RJ. Comparison of structural stabilities
[30] Marnett LJ, Wilcox CL. Stimulation of prostaglandin biosynthesis of prostaglandin H synthase-1 and -2. J Biol Chem 1998; 273:
by lipoic acid. Biochim Biophys Acta 1977; 487: 222-30. 6801-11.
[31] Egan RW, Humes JL, Kuehl FA Jr. Differential effects of [52] Gierse JK, McDonald JJ, Hauser SD, Rangwala SH, Koboldt CM,
prostaglandin synthetase stimulators on inhibition of Seibert K. A Single amino acid difference between
cyclooxygenase. Biochemistry 1978; 17: 2230-4. cyclooxygenase-1 (COX-1) and -2 (COX-2) reverses the selectivity
[32] Hemler ME, Crawford CG, Lands WE. Lipoxygenation activity of of COX-2 specific inhibitors J Biol Chem 1996; 271: 15810-15814.
purified prostaglandin-forming cyclooxygenase. Biochemistry [53] Jones DA, Fitzpatrick FA. "Suicide" inactivation of thromboxane
1978; 17:1772-9. A2 synthase. Characteristics of mechanism-based inactivation with
[33] Smith WL, Lands WE. Oxygenation of unsaturated fatty acids by isolated enzyme and intact platelets. J Biol Chem 1990; 265:
soybean lipoxygenase. J Biol Chem 1972; 247: 1038-47. 20166-71.
[34] Setty BN, Stuart MJ, Walenga RW. Formation of 11- [54] Wade ML, Voelkel NF, Fitzpatrick FA. "Suicide" inactivation of
hydroxyeicosatetraenoic acid and 15-hydroxyeicosatetraenoic acid prostaglandin I2 synthase: characterization of mechanism-based
in human umbilical arteries is catalyzed by cyclooxygenase. inactivation with isolated enzyme and endothelial cells. Arch
Biochim Biophys Acta 1985; 833: 484-94. Biochem Biophys 1995; 321: 453-8.
[35] Thuresson ED, Lakkides KM, Smith WL. Different catalytically [55] Evans JF, Nathaniel DJ, Zamboni RJ, Ford-Hutchinson AW.
competent arrangements of arachidonic acid within the Leukotriene A 3. A poor substrate but a potent inhibitor of rat and
cyclooxygenase active site of prostaglandin endoperoxide H human neutrophil leukotriene A4 hydrolase. J Biol Chem 1985;
synthase-1 lead to the formation of different oxygenated products. J 260: 10966-70.
Biol Chem 2000; 275: 8501-7. [56] Orning L, Jones DA, Fitzpatrick FA. Mechanism-based
[36] Holtzman MJ, Turk J, Shornick LP. Identification of a inactivation of leukotriene A4 hydrolase during leukotriene B4
pharmacologically distinct prostaglandin H synthase in cultured formation by human erythrocytes. J Biol Chem 1990; 265: 14911-
epithelial cells. J Biol Chem 1992; 267: 21438-45. 6.
[37] Thuresson ED, Lakkides KM, Rieke CJ, Sun Y, Wingerd BA, [57] Aharony D, Redkar-Brown DG, Hubbs SJ, Stein RL. Kinetic
Micielli R, et al. Prostaglandin endoperoxide H synthase-1: the studies on the inactivation of 5-lipoxygenase by 5(S)-
functions of cyclooxygenase active site residues in the binding, hydroperoxyeicosatetraenoic acid. Prostaglandins 1987; 33: 85-
positioning, and oxygenation of arachidonic acid. J Biol Chem 100.
2001; 276: 10347-57. [58] Lepley RA, Fitzpatrick FA. Irreversible inactivation of 5-
[38] Malkowski MG, Ginell S, Smith WL, Garavito RM. The lipoxygenase by leukotriene A4. Characterization of product
productive conformation of arachidonic acid bound to inactivation with purified enzyme and intact leukocytes. J Biol
prostaglandin synthase. Science 2000; 289: 1933-1937. Chem 1994; 269: 2627-31.
[39] Kiefer JR, Pawlitz JL, Moreland KT, Stegeman RA, Hood WF, [59] De Carolis E, Denis D, Riendeau D. Oxidative inactivation of
Gierse JK, et al. Structural insights into the stereochemistry of the human 5-lipoxygenase in phosphatidylcholine vesicles. Eur J
cyclooxygenase reaction. Nature 2000; 405: 97-101. Biochem 1996; 235: 416-23.
[40] Karthein R, Dietz R, Nastainczyk W, Ruf HH. Higher oxidation [60] Kishimoto K, Nakamura M, Suzuki H, Yoshimoto T, Yamamoto S,
states of prostaglandin H synthase. EPR study of a transient tyrosyl Takao T, et al. Suicide inactivation of porcine leukocyte 12-
radical in the enzyme during the peroxidase reaction. Eur J lipoxygenase associated with its incorporation of 15-hydroperoxy-
Biochem 1988; 171: 313-320. 5,8,11,13-eicosatetraenoic acid derivative. Biochim Biophys Acta
[41] Smith WL, Eling TE, Kulmacz RJ, Marnett LJ, Tsai A. Tyrosyl 1996; 1300: 56-62.
radicals and their role in hydroperoxide-dependent activation and [61] Smith WL, Lands WE. The self-catalyzed destruction of
inactivation of prostaglandin endoperoxide synthase. Biochemistry lipoxygenase. Biochem Biophys Res Commun 1970; 41: 846-51.
1992; 31: 3-7. [62] Kim MR, Sok DE. Irreversible inhibition of soybean lipoxygenase-
[42] Kulmacz RJ, Pendleton RB, Lands WE. Interaction between 1 by hydroperoxy acids as substrates. Arch Biochem Biophys 1991;
peroxidase and cyclooxygenase activities in prostaglandin- 288: 270-5.
endoperoxide synthase. Interpretation of reaction kinetics. J Biol [63] Wiesner R, Suzuki H, Walther M, Yamamoto S, Kuhn H. Suicidal
Chem 1994; 269: 5527-36. inactivation of the rabbit 15-lipoxygenase by 15S-HpETE is
[43] Chen YN, Bienkowski MJ, Marnett LJ. Controlled tryptic digestion paralleled by covalent modification of active site peptides. Free
of prostaglandin H synthase. Characterization of protein fragments Radic Biol Med 2003; 34: 304-15.
and enhanced rate of proteolysis of oxidatively inactivated enzyme. [64] Egan RW, Paxton J, Kuehl FA. Mechanism for irreversible self-
J Biol Chem 1987; 262: 16892-9. deactivation of prostaglandin synthetase. J Biol Chem 1976; 251:
[44] Marnett LJ, Chen YN, Maddipati KR, Ple P, Labeque R. 7329-35.
Functional differentiation of cyclooxygenase and peroxidase [65] Fitzpatrick FA, Gorman RR. Platelet rich plasma transforms
activities of prostaglandin synthase by trypsin treatment. Possible exogenous prostaglandin endoperoxide H2 into thromboxane A2.
location of a prosthetic heme binding site. J Biol Chem 1988; 263: Prostaglandins 1977; 14: 881-9.
16532-5. [66] Crutchley DJ, Hawkins HJ, Eling TE, Anderson MW. Covalent
[45] Raz A, Needleman P. Differential modification of cyclo-oxygenase binding of prostaglandins G2 and H2 to components of ram
and peroxidase activities of prostaglandin endoperoxidase synthase seminal vesicle microsomal fraction. Biochem Pharmacol 1979;
by proteolytic digestion and hydroperoxides. Biochem J 1990; 269: 28:1519-23.
603-7. [67] Lecomte M, Lecocq R, Dumont JE, Boeynaems J-M. Covalent
[46] Kulmacz RJ. Concerted loss of cyclooxygenase and peroxidase binding of arachidonic acid metabolites to human platelet proteins.
activities from prostaglandin H synthase upon proteolytic attack. Identification of prostaglandin H synthase as one of the modified
Prostaglandins 1989; 38: 277-88. substrates. J Biol Chem 1990; 265: 5178-87.
[47] Markey CM, Alward A, Weller PE, Marnett LJ. Quantitative [68] Rapoport S, Hartel B, Hausdorf G. Methionine sulfoxide
studies of hydroperoxide reduction by prostaglandin H synthase. formation: the cause of self-inactivation of reticulocyte
Reducing substrate specificity and the relationship of peroxidase to lipoxygenase. Eur J Biochem 1984; 139: 573-6.
cyclooxygenase activities. J Biol Chem 1987; 262: 6266-79. [69] Boutaud O, Brame CJ, Chaurand P, Li J, Rowlinson SW, Crews
[48] Tsai A, Palmer G, Xiao G, Swinney DC, Kulmacz RJ. Structural BC, et al. Characterization of the lysyl adducts of prostaglandin H-
characterization of arachidonyl radicals formed by prostaglandin H synthases that are derived from oxygenation of arachidonic acid.
synthase-2 and prostaglandin H synthase-1 reconstituted with Biochemistry 2001; 40: 6948-55.
mangano protoporphyrin IX. J Biol Chem 1998; 273: 3888-94.
Cyclooxygenase Enzymes Current Pharmaceutical Design, 2004, Vol. 10, No. 6 587

[70] Boutaud O, Li J, Zagol I, Shipp EA, Davies SS, Roberts LJ, et al. [92] Stichtenoth DO, Frolich JC. The second generation of COX-2
Levuglandinyl adducts of proteins are formed via a prostaglandin inhibitors: what advantages do the newest offer? Drugs 2003; 63:
H2 synthase-dependent pathway after platelet activation. J Biol 33-45.
Chem 2003; 278: 16926-8. [93] Picot D, Loll PJ, Garavito RM. The X-ray crystal structure of the
[71] Bernoud-Hubac N, Roberts LJ. Identification of oxidized membrane protein prostaglandin H2 synthase-1. Nature 1994; 367:
derivatives of neuroketals. Biochemistry 2002; 41:11466-71. 243-9.
[72] Wu G, Wei C, Kulmacz RJ, Osawa Y, Tsai AL. A mechanistic [94] Loll PJ, Picot D, Garavito RM. The structural basis of aspirin
study of self-inactivation of the peroxidase activity in prostaglandin activity inferred from the crystal structure of inactivated
H synthase-1. J Biol Chem 1999; 274: 9231-7. prostaglandin H2 synthase. Nat Struct Biol 1995; 2: 637-43.
[73] Song I, Ball TM, Smith WL. Different suicide inactivation [95] Kurumbail RG, Stevens AM, Gierse JK, McDonald JJ, Stegeman
processes for the peroxidase and cyclooxygenase activities of RA, Pak JY, et al. Structural basis for selective inhibition of
prostaglandin endoperoxide H synthase-1. Biochem Biophys Res cyclooxygenase-2 by anti-inflammatory agents. Nature 1996; 384:
Commun 2001; 289: 869-75. 644-8.
[74] Roth GJ, Stanford N, Majerus PW. Acetylation of prostaglandin [96] Luong C, Miller A, Barnett J, Chow J, Ramesha C, Browner MF.
synthase by aspirin. Proc Natl Acad Sci USA 1975; 72: 3073-6. Flexibility of the NSAID binding site in the structure of human
[75] Roth GJ, Machuga ET, Ozols J. Isolation and covalent structure of cyclooxygenase-2. Nat Struct Biol 1996; 3: 927-33.
the aspirin-modified, active-site region of prostaglandin synthetase. [97] Lin AH, Bienkowski MJ, Gorman RR. Regulation of prostaglandin
Biochemistry 1983; 22: 4672-5. H synthase mRNA levels and prostaglandin biosynthesis by
[76] Van Der Ouderaa FJ, Buytenhek M, Nugteren DH,Van Dorp DA. platelet-derived growth factor. J Biol Chem 1989; 264: 17379-83.
Acetylation of prostaglandin endoperoxide synthetase with [98] Clarke RJ, Mayo G, Price P, FitzGerald GA. Suppression of
acetylsalicylic acid. Eur J Biochem 1980; 109:1-8. thromboxane A2 but not of systemic prostacyclin by controlled-
[77] Wells I, Marnett LJ. Acetylation of prostaglandin endoperoxide release aspirin. N Engl J Med 1991; 325: 1137-41.
synthase by N-acetylimidazole: comparison to acetylation by [99] Marcus AJ, Weksler BB, Jaffe EA, Broekman MJ. Synthesis of
aspirin. Biochemistry 1992; 31: 9520-5. prostacyclin from platelet-derived endoperoxides by cultured
[78] Patrono C, Ciabattoni G, Patrignani P, Pugliese F, Filabozzi P, human endothelial cells. J Clin Invest 1980; 66: 979-86.
Catella F, et al. Clinical pharmacology of platelet cyclooxygenase [100] FitzGerald GA, Reilly IA, Pedersen AK. The biochemical
inhibition. Circulation 1985; 72:1177-84. pharmacology of thromboxane synthase inhibition in man.
[79] Patrono C. Measurement of cyclooxygenase isozyme inhibition in Circulation 1985; 72: 1194-201.
humans: exploring the clinical relevance of biochemical selectivity. [101] Nowak J, FitzGerald GA. Redirection of prostaglandin
Clin Exp Rheumatol 2001; 19: (6 Suppl 25):S45-50. endoperoxide metabolism at the platelet-vascular interface in man.
[80] Patrono C. Aspirin: new cardiovascular uses for an old drug. Am J J Clin Invest 1989; 83: 380-5.
Med 2001; 110: 62S-65S. [102] McGee JE, Fitzpatrick FA. Erythrocyte-neutrophil interactions:
[81] Harter HR, Burch JW, Majerus PW, Stanford N, Delmez JA, formation of leukotriene B 4 by transcellular biosynthesis. Proc Natl
Anderson CB, et al. Prevention of thrombosis in patients on Acad Sci USA 1986; 83: 1349-53.
hemodialysis by low-dose aspirin. N Engl J Med 1979; 301: 577-9 [103] Maclouf JA, Murphy RC. Transcellular metabolism of neutrophil-
[82] Randomised trial of intravenous streptokinase, oral aspirin, both, or derived leukotriene A4 by human platelets. A potential cellular
neither among 17,187 cases of suspected acute myocardial source of leukotriene C4. J Biol Chem 1988; 263: 174-181.
infarction: ISIS-2. ISIS-2 (Second International Study of Infarct [104] Feinmark SJ, Cannon PJ. Endothelial cell leukotriene C4 synthesis
Survival) Collaborative Group. Lancet 1988; 2: (8607), 349-60. results from intercellular transfer of leukotriene A4 synthesized by
[83] Baigent C, Collins R, Appleby P, Parish S, Sleight P, Peto R. ISIS- polymorphonuclear leukocytes. J Biol Chem 1986; 261: 16466-72.
2: 10 year survival among patients with suspected acute myocardial [105] Bailey JM, Muza B, Hla T, Salata K. Restoration of prostacyclin
infarction in randomised comparison of intravenous streptokinase, synthase in vascular smooth muscle cells after aspirin treatment:
oral aspirin, both, or neither. The ISIS-2 (Second International regulation by epidermal growth factor. J Lipid Res 1985; 26: 54-61.
Study of Infarct Survival) Collaborative Group. Brit Med J 2001; [106] Bartolini G, Orlandi M, Chiricolo M, Minghetti L, Guerrini F,
316: 1337-43. Fidan M, et al. Regulation of thromboxane A2 biosynthesis in
[84] Kalgutkar AS, Crews BC, Rowlinson SW, Garner C, Seibert K, platelet-free human monocytes and the possible role of polypeptide
Marnett LJ. Aspirin-like molecules that covalently inactivate growth factor(s) in the induction of cyclooxygenase system.
cyclooxygenase-2. Science 1998; 280:1268-70. Biochim Biophys Acta 1986; 876: 486-93.
[85] Kalgutkar AS, Kozak KR, Crews BC, Hochgesang GP, Marnett LJ. [107] O'Neill LA, Barrett ML, Lewis GP. Induction of cyclooxygenase
Covalent modification of cyclooxygenase-2 (COX-2) by 2- by interleukin-1 in rheumatoid synovial cells. FEBS Lett 1987;
acetoxyphenyl alkyl sulfides, a new class of selective COX-2 212: 35-9.
inactivators. J Med Chem 1998; 41: 4800-18. [108] Pash JM, Bailey JM. Inhibition by corticosteroids of epidermal
[86] Fletcher BS, Kujubu DA, Perrin DM, Herschman HR. Structure of growth factor-induced recovery of cyclooxygenase after aspirin
the mitogen-inducible TIS10 gene and demonstration that the inactivation. FASEB J 1988; 2: 2613-8.
TIS10-encoded protein is a functional prostaglandin G/H synthase. [109] Xie WL, Chipman JG, Robertson DL, Erikson RL, Simmons DL.
J Biol Chem 1992; 267: 4338-44. Expression of a mitogen-responsive gene encoding prostaglandin
[87] Schneider C, Brash AR. Stereospecificity of hydrogen abstraction synthase is regulated by mRNA splicing. Proc Natl Acad Sci USA
in the conversion of arachidonic acid to 15R-HETE by aspirin- 1991; 88: 2692-6.
treated cyclooxygenase-2. Implications for the alignment of [110] Fu JY, Masferrer JL, Seibert K, Raz A, Needleman P. The
substrate in the active site. J Biol Chem 2000; 275: 4743-6. induction and suppression of prostaglandin H2 synthase
[88] Lecomte M, Laneuville O, Ji C, DeWitt DL, SmithWL. Acetylation (cyclooxygenase) in human monocytes. J Biol Chem 1990; 265:
of human rostaglandin endoperoxide synthase-2 (cyclooxygenase- 16737-40.
2) by aspirin. J Biol Chem 1994; 269: 13207-15. [111] Masferrer JL, Zweifel BS, Seibert K, Needleman P. Selective
[89] Patrignani P, Panara MR, Sciulli MG, Santini G, Renda G, Patrono regulation of cellular cyclooxygenase by dexamethasone and
C. Differential inhibition of human prostaglandin endoperoxide endotoxin in mice. J Clin Invest 1990; 86: 1375-9.
synthase-1 and -2 by nonsteroidal anti-inflammatory drugs. J [112] Masferrer JL, Seibert K, Zweifel B, Needleman P. Endogenous
Physiol Pharmacol 1997; 48: 623-31. glucocorticoids regulate an inducible cyclooxygenase enzyme. Proc
[90] Meade EA, Smith WL, DeWitt DL. Differential inhibition of Natl Acad Sci USA 1992; 89: 3917-21.
prostaglandin endoperoxide synthase (cyclooxygenase) isozymes [113] Subbaramaiah K, Lin DT, Hart JC, Dannenberg AJ. Peroxisome
by aspirin and other non-steroidal anti-inflammatory drugs. J Biol proliferator-activated receptor gamma ligands suppress the
Chem 1993; 268: 6610-4. transcriptional activation of cyclooxygenase-2. Evidence for
[91] Trummlitz G, van Ryn J. Designing selective COX-2 inhibitors: involvement of activator protein-1 and CREB-binding
molecular modeling approaches. Curr Opin Drug Discov Devel protein/p300. J Biol Chem 2001; 276: 12440-8.
2002; 5: 550-61. [114] Guan Z, Baier LD, Morrison AR. p38 mitogen-activated protein
kinase down-regulates nitric oxide and up-regulates prostaglandin
588 Current Pharmaceutical Design, 2004, Vol. 10, No. 6 F.A. Fitzpatrick

E2 biosynthesis stimulated by interleukin-1beta. J Biol Chem 1997; [126] Hwang D, Scollard D, Byrne J, Levine E. Expression of
272: 8083-9. cyclooxygenase-1 and cyclooxygenase-2 in human breast cancer. J
[115] Newton R, Seybold J, Kuitert LM, Bergmann M, Barnes PJ. Natl Cancer Inst 1998; 90: 455-60.
Repression of cyclooxygenase-2 and prostaglandin E2 release by [127] Kulmacz RJ. Cellular regulation of prostaglandin H synthase
dexamethasone occurs by transcriptional and post-transcriptional catalysis. FEBS Lett 1998; 430:154-7.
mechanisms involving loss of polyadenylated mRNA. J Biol Chem [128] Marshall PJ, Kulmacz RJ, Lands WE. Constraints on prostaglandin
1998; 273: 32312-21. biosynthesis in tissues. J Biol Chem 1987; 262: 3510-7.
[116] Dixon DA, Kaplan CD, McIntyre TM, Zimmerman GA, Prescott [129] Lands WE, Sauter J, Stone GW. Oxygen requirement for
SM. Post-transcriptional control of cyclooxygenase-2 gene prostaglandin biosynthesis. Prostaglandins Med 1978; 1: 117-20.
expression. The role of the 3'-untranslated region. J Biol Chem [130] Goodwin DC, Landino LM, Marnett LJ. Effects of nitric oxide and
2000; 275: 11750-7. nitric oxide-derived species on prostaglandin endoperoxide
[117] Toyota M, Shen L, Ohe-Toyota M, Hamilton SR, Sinicrope FA, synthase and prostaglandin biosynthesis. FASEB J 1999; 13: 1121-
Issa JP. Aberrant methylation of the cyclooxygenase-2 CpG island 36.
in colorectal tumors. Cancer Res 2000; 60: 4044-8. [131] Juni P, Rutjes AW, Dieppe PA. Are selective COX 2 inhibitors
[118] Song SH, Jong HS, Choi HH, Inoue H, Tanabe T, Kim NK, et al. superior to traditional non steroidal anti-inflammatory drugs? Brit
Transcriptional silencing of cyclooxygenase-2 by hyper- Med J. 2002; 324: 1287-8.
methylation of the 5' CpG island in human gastric carcinoma cells. [132] FitzGerald GA. Cardiovascular pharmacology of nonselective
Cancer Res 2001; 61: 4628-35. nonsteroidal anti-inflammatory drugs and coxibs: clinical
[119] Kutchera W, Jones DA, Matsunami N, Groden J, McIntyre TM, considerations. Am J Cardiol 2002; 89: 26D-32D.
Zimmerman GA, et al. Prostaglandin H synthase 2 is expressed [133] Reuter BK, Zhang XJ, Miller MJ. Therapeutic utility of aspirin in
abnormally in human colon cancer: evidence for a transcriptional the ApcMin/+ murine model of colon carcinogenesis. BMC Cancer
effect. Proc Natl Acad Sci USA 1996; 93: 4816-20. 2002; 2: 19.
[120] Soslow RA, Dannenberg AJ, Rush D, Woerner BM, Khan KN, [134] Sansom OJ, Stark LA, Dunlop MG, Clarke AR. Suppression of
Masferrer J, et al. COX-2 is expressed in human pulmonary, intestinal and mammary neoplasia by lifetime administration of
colonic, and mammary tumors. Cancer 2000; 89: 2637-2645. aspirin in Apc(Min/+) and Apc(Min/+), Msh2(-/-) mice. Cancer
[121] Lim HY, Joo HJ, Choi JH, Yi JW, Yang MS, Cho DY, et al. Res 2001; 61: 7060-4.
Increased expression of cyclooxygenase-2 protein in human gastric [135] Williamson SL, Kartheuser A, Coaker J, Kooshkghazi MD, Fodde
carcinoma. Clin Cancer Res 2000; 6: 519-25. R, Burn J, et al. Intestinal tumorigenesis in the Apc1638N mouse
[122] Chan G, Boyle JO, Yang EK, Zhang F, Sacks PG, Shah JP, et al. treated with aspirin and resistant starch for up to 5 months.
Cyclooxygenase-2 expression is up-regulated in squamous cell Carcinogenesis 1999; 20: 805-10.
carcinoma of the head and neck. Cancer Res 1999; 59: 991-4. [136] Trifan OC, Smith RM, Thompson BD, Hla T. Overexpression of
[123] Tucker ON, Dannenberg AJ, Yang EK, Zhang F, Teng L, Daly JM, cyclooxygenase-2 induces cell cycle arrest. Evidence for a
et al. Cyclooxygenase-2 expression is up-regulated in human prostaglandin-independent mechanism. J Biol Chem 1999; 274:
pancreatic cancer. Cancer Res 1999; 59: 987-90. 34141-7.
[124] Fujita T, Matsui M, Takaku K, Uetake H, Ichikawa W, Taketo [137] Baraldi PG, Cacciari B, Borean PA, Varanin K, Pastorin G, Da Ros
MM, et al. Size- and invasion-dependent increase in T, et al. Pyrazolo-triazolo-pyrimidine derivatives as adenosine
cyclooxygenase 2 levels in human colorectal carcinomas. Cancer receptor antagonists: a possible template for adenosine receptor
Res 1998; 58: 4823-6. subtypes? Curr Pharm Design 2002; 8(26): 2299-332.
[125] Chulada PC, Thompson MB, Mahler JF, Doyle CM, Gaul BW, Lee [138] Oshima M, Taketo MM. COX selectivity and animal models for
C, et al. Genetic disruption of Ptgs-1, as well as Ptgs-2, reduces colon cancer. Curr Pharm Design 2002; 8(12): 1021-34.
intestinal tumorigenesis in Min mice. Cancer Res 2000; 60: 4705-8. [139] Rodriguez-Tellez M, Arguelles F, Herrerias JM Jr, Ledro D,
Esteban J, Herrerias JM. Antiinflamatory agents less dangerous for
gastrointestinal tract. Curr Pharm Design 2001; 7(10): 951-76.

Das könnte Ihnen auch gefallen