12
Formation of Prostaglandins E 2 and D 2 via the Isoprostane Pathway A MECHANISM FOR THE GENERATION OF BIOACTIVE PROSTAGLANDINS INDEPENDENT OF CYCLOOXYGENASE* Received for publication, April 16, 2003, and in revised form, May 5, 2003 Published, JBC Papers in Press, May 13, 2003, DOI 10.1074/jbc.M303984200 Ling Gao‡§, William E. Zackert§, Justin J. Hasford§, Michael E. Danekis§, Ginger L. Milne§, Catha Remmert§, Jeff Reese, Huiyong Yin, Hsin-Hsiung Tai**, Sudhansu K. Dey, Ned A. Porter, and Jason D. Morrow‡§‡‡ From the Departments of Medicine, §Pharmacology, Pediatrics, and Chemistry, Vanderbilt University, Nashville, Tennessee 37232 and the **College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536 It has heretofore been assumed that the cyclooxygen- ases (COXs) are solely responsible for peostaglandin (PG) synthesis in vivo. An important structural feature of PGH 2 formed by COX is the trans-configuration of side chains relative to the prostane ring. Previously, we reported that a series of PG-like compounds termed isoprostanes (IsoPs) are formed in vivo in humans from the free radi- cal-catalyzed peroxidation of arachidonate independent of COX. A major difference between these compounds and PGs is that IsoPs are formed from endoperoxide interme- diates, the vast majority of which contain side chains that are cis relative to the prostane ring. In addition, unlike the formation of eicosanoids from COX, IsoPs are formed as racemic mixtures because they are generated nonen- zymatically. IsoPs containing E- and D-type prostane rings (E 2 /D 2 -IsoPs) are one class of IsoPs formed, and we have reported previously that one of the major IsoPs gen- erated is 15-E 2t -IsoP (8-iso-PGE 2 ). Unlike PGE 2 , 15-E 2t - IsoP is significantly more unstable in buffered solutions in vitro and undergoes epimerization to PGE 2 . Analo- gously, the D-ring IsoP (15-D 2c -IsoP) would be predicted to rearrange to PGD 2 . We now report that compounds identical in all respects to PGE 2 and PGD 2 and their re- spective enantiomers are generated in vivo via the IsoP pathway, presumably by epimerization of racemic 15-E 2t - IsoP and 15-D 2c -IsoP, respectively. Racemic PGE 2 and PGD 2 were present esterified in phospholipids derived from liver tissue from rats exposed to oxidant stress at levels of 24 16 and 37 12 ng/g of tissue, respectively. In addition, racemic PGs, particularly PGD 2 , were present unesterified in urine from normal animals and humans and represented up to 10% of the total PG detected. Levels of racemic PGD 2 increased 35-fold after treatment of rats with carbon tetrachloride to induce oxidant stress. In this setting, PGD 2 and its enantiomer generated by the IsoP pathway represented 30% of the total PGD 2 present in urine. These findings strongly support the contention that a second pathway exists for the formation of bioac- tive PGs in vivo that is independent of COX. Cyclooxygenase (COX) 1 -1 and COX-2 catalyze the committed steps in formation of prostaglandins (PGs) by generating the unstable bicycloendoperoxide intermediate PGH 2 (1–3). PGH 2 is subsequently metabolized to the parent eicosanoids PGE 2 , PGD 2 , PGF 2 , PGI 2 , and thromboxane A 2 , which exert a pleth- ora of biological activities (1). The formation of PGH 2 is ste- reospecific in that, among other structural aspects, the side chains of PGH 2 are oriented in the trans-configuration relative to the prostane ring (Fig. 1A). This conformation is highly favored thermodynamically (4, 5). We have previously reported that a series of PG-like com- pounds termed isoprostanes (IsoPs) are formed in vivo from the free radical-catalyzed peroxidation of arachidonate independ- ent of COX (6). Analogous to PGs, we have determined that IsoPs contain E/D-, F-, and thromboxane-type prostane rings (7). Although the structures of these compounds are very sim- ilar to COX-derived PGs, an important distinction between IsoPs and PGs is that IsoP bicycloendoperoxide intermediates contain side chains that are predominantly (90%) oriented cis in relation to the prostane ring because the generation of these intermediates is favored kinetically (4, 7, 8). Indeed, we have previously reported that two IsoPs that are formed in abun- dance in vivo are 15-F 2t -IsoP (8-iso-PGF 2 ) and 15-E 2t -IsoP (8-iso-PGE 2 ), which are generated from the endoperoxide in- termediate 15-H 2t -IsoP (8-iso-PGH 2 ) (Fig. 1B) (9, 10). Although not reported, it would also be predicted that 15-H 2c -IsoP (12- iso-PGH 2 ) is formed in abundance and can rearrange to the analogous D-ring IsoP 15-D 2c -IsoP (12-iso-PGD 2 ) (Fig. 1C). In contrast to other types of prostanoids, E 2 /D 2 -IsoPs are -hydroxyketone-containing compounds that can undergo re- versible keto-enol tautomerization under both acidic and basic conditions, allowing rearrangement of the side chains that are initially cis to the more stable trans-configuration. That the trans-configuration is highly favored has been demonstrated by the finding that, when PGE 2 is subjected to conditions that induce keto-enol tautomerism, 10% of the compound rear- ranges to the cis-side chain isomer 15-E 2t -IsoP (11). In addi- tion, attempts to synthesize 15-D 2c -IsoP have been unsuccess- ful because epimerization at C-12 readily occurs during synthesis to yield PGD 2 (12). Furthermore, facile epimerization of a number of other PG-like compounds containing side chains cis to the prostane ring has been reported (13, 14). In the course of studies to characterize various E/D-ring IsoPs formed in vitro and in vivo from the peroxidation of * This work was supported in part by National Institutes of Health Grants DK48831, GM42056, CA77839, HD12304, and HL46296. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked “adver- tisement” in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. ‡‡ Recipient of a Burroughs Wellcome Fund Clinical Scientist Award in Translational Research. To whom correspondence should be ad- dressed: Vanderbilt University, 526 RRB, 23rd and Pierce Aves., Nash- ville, TN 37232-6602. Tel.: 615-343-1124; Fax: 615-322-3669; E-mail: [email protected]. 1 The abbreviations used are: COX, cyclooxygenase; PG, prosta- glandin; IsoP, isoprostane; GC, gas chromatography; MS, mass spec- trometry; ent-, enantiomeric; rac-, racemic; PFB, pentafluorobenzyl; HPLC, high pressure liquid chromatography. THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 278, No. 31, Issue of August 1, pp. 28479 –28489, 2003 © 2003 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A. This paper is available on line at http://www.jbc.org 28479 by guest on January 7, 2016 http://www.jbc.org/ Downloaded from

Formation of Prostaglandins E2 and D2 via the Isoprostane Pathway: A MECHANISM FOR THE GENERATION OF BIOACTIVE PROSTAGLANDINS INDEPENDENT OF CYCLOOXYGENASE

  • Upload
    sibs

  • View
    0

  • Download
    0

Embed Size (px)

Citation preview

Formation of Prostaglandins E2 and D2 via the Isoprostane PathwayA MECHANISM FOR THE GENERATION OF BIOACTIVE PROSTAGLANDINS INDEPENDENTOF CYCLOOXYGENASE*

Received for publication, April 16, 2003, and in revised form, May 5, 2003Published, JBC Papers in Press, May 13, 2003, DOI 10.1074/jbc.M303984200

Ling Gao‡§, William E. Zackert§, Justin J. Hasford§, Michael E. Danekis§, Ginger L. Milne§,Catha Remmert§, Jeff Reese¶, Huiyong Yin�, Hsin-Hsiung Tai**, Sudhansu K. Dey¶,Ned A. Porter�, and Jason D. Morrow‡§‡‡

From the Departments of ‡Medicine, §Pharmacology, ¶Pediatrics, and Chemistry, �Vanderbilt University,Nashville, Tennessee 37232 and the **College of Pharmacy, University of Kentucky, Lexington, Kentucky 40536

It has heretofore been assumed that the cyclooxygen-ases (COXs) are solely responsible for peostaglandin (PG)synthesis in vivo. An important structural feature of PGH2formed by COX is the trans-configuration of side chainsrelative to the prostane ring. Previously, we reported thata series of PG-like compounds termed isoprostanes(IsoPs) are formed in vivo in humans from the free radi-cal-catalyzed peroxidation of arachidonate independentof COX. A major difference between these compounds andPGs is that IsoPs are formed from endoperoxide interme-diates, the vast majority of which contain side chains thatare cis relative to the prostane ring. In addition, unlikethe formation of eicosanoids from COX, IsoPs are formedas racemic mixtures because they are generated nonen-zymatically. IsoPs containing E- and D-type prostanerings (E2/D2-IsoPs) are one class of IsoPs formed, and wehave reported previously that one of the major IsoPs gen-erated is 15-E2t-IsoP (8-iso-PGE2). Unlike PGE2, 15-E2t-IsoP is significantly more unstable in buffered solutionsin vitro and undergoes epimerization to PGE2. Analo-gously, the D-ring IsoP (15-D2c-IsoP) would be predictedto rearrange to PGD2. We now report that compoundsidentical in all respects to PGE2 and PGD2 and their re-spective enantiomers are generated in vivo via the IsoPpathway, presumably by epimerization of racemic 15-E2t-IsoP and 15-D2c-IsoP, respectively. Racemic PGE2 andPGD2 were present esterified in phospholipids derivedfrom liver tissue from rats exposed to oxidant stress atlevels of 24 � 16 and 37 � 12 ng/g of tissue, respectively. Inaddition, racemic PGs, particularly PGD2, were presentunesterified in urine from normal animals and humansand represented up to 10% of the total PG detected. Levelsof racemic PGD2 increased 35-fold after treatment of ratswith carbon tetrachloride to induce oxidant stress. In thissetting, PGD2 and its enantiomer generated by the IsoPpathway represented �30% of the total PGD2 present inurine. These findings strongly support the contentionthat a second pathway exists for the formation of bioac-tive PGs in vivo that is independent of COX.

Cyclooxygenase (COX)1-1 and COX-2 catalyze the committedsteps in formation of prostaglandins (PGs) by generating theunstable bicycloendoperoxide intermediate PGH2 (1–3). PGH2

is subsequently metabolized to the parent eicosanoids PGE2,PGD2, PGF2�, PGI2, and thromboxane A2, which exert a pleth-ora of biological activities (1). The formation of PGH2 is ste-reospecific in that, among other structural aspects, the sidechains of PGH2 are oriented in the trans-configuration relativeto the prostane ring (Fig. 1A). This conformation is highlyfavored thermodynamically (4, 5).

We have previously reported that a series of PG-like com-pounds termed isoprostanes (IsoPs) are formed in vivo from thefree radical-catalyzed peroxidation of arachidonate independ-ent of COX (6). Analogous to PGs, we have determined thatIsoPs contain E/D-, F-, and thromboxane-type prostane rings(7). Although the structures of these compounds are very sim-ilar to COX-derived PGs, an important distinction betweenIsoPs and PGs is that IsoP bicycloendoperoxide intermediatescontain side chains that are predominantly (�90%) oriented cisin relation to the prostane ring because the generation of theseintermediates is favored kinetically (4, 7, 8). Indeed, we havepreviously reported that two IsoPs that are formed in abun-dance in vivo are 15-F2t-IsoP (8-iso-PGF2�) and 15-E2t-IsoP(8-iso-PGE2), which are generated from the endoperoxide in-termediate 15-H2t-IsoP (8-iso-PGH2) (Fig. 1B) (9, 10). Althoughnot reported, it would also be predicted that 15-H2c-IsoP (12-iso-PGH2) is formed in abundance and can rearrange to theanalogous D-ring IsoP 15-D2c-IsoP (12-iso-PGD2) (Fig. 1C).

In contrast to other types of prostanoids, E2/D2-IsoPs are�-hydroxyketone-containing compounds that can undergo re-versible keto-enol tautomerization under both acidic and basicconditions, allowing rearrangement of the side chains that areinitially cis to the more stable trans-configuration. That thetrans-configuration is highly favored has been demonstrated bythe finding that, when PGE2 is subjected to conditions thatinduce keto-enol tautomerism, �10% of the compound rear-ranges to the cis-side chain isomer 15-E2t-IsoP (11). In addi-tion, attempts to synthesize 15-D2c-IsoP have been unsuccess-ful because epimerization at C-12 readily occurs duringsynthesis to yield PGD2 (12). Furthermore, facile epimerizationof a number of other PG-like compounds containing side chainscis to the prostane ring has been reported (13, 14).

In the course of studies to characterize various E/D-ringIsoPs formed in vitro and in vivo from the peroxidation of

* This work was supported in part by National Institutes of HealthGrants DK48831, GM42056, CA77839, HD12304, and HL46296. Thecosts of publication of this article were defrayed in part by the paymentof page charges. This article must therefore be hereby marked “adver-tisement” in accordance with 18 U.S.C. Section 1734 solely to indicatethis fact.

‡‡ Recipient of a Burroughs Wellcome Fund Clinical Scientist Awardin Translational Research. To whom correspondence should be ad-dressed: Vanderbilt University, 526 RRB, 23rd and Pierce Aves., Nash-ville, TN 37232-6602. Tel.: 615-343-1124; Fax: 615-322-3669; E-mail:[email protected].

1 The abbreviations used are: COX, cyclooxygenase; PG, prosta-glandin; IsoP, isoprostane; GC, gas chromatography; MS, mass spec-trometry; ent-, enantiomeric; rac-, racemic; PFB, pentafluorobenzyl;HPLC, high pressure liquid chromatography.

THE JOURNAL OF BIOLOGICAL CHEMISTRY Vol. 278, No. 31, Issue of August 1, pp. 28479–28489, 2003© 2003 by The American Society for Biochemistry and Molecular Biology, Inc. Printed in U.S.A.

This paper is available on line at http://www.jbc.org 28479

by guest on January 7, 2016http://w

ww

.jbc.org/D

ownloaded from

arachidonic acid, analysis of oxidation products by gas chroma-tography (GC)/mass spectrometry (MS) disclosed the genera-tion of significant amounts of compounds that had retentiontimes and molecular weights identical to those of PGE2 andPGD2. Because IsoPs are formed nonenzymatically, compoundsgenerated by this pathway would be predicted to be racemic (6,7). Using a variety of chromatographic and mass spectrometricapproaches, we present evidence that compounds identical inall respects to COX-derived PGE2 and PGD2 and their respec-tive enantiomers are formed in vitro and in vivo via the IsoPpathway. A proposed mechanism by which the formation ofPGE2 and PGD2 occurs from 15-E2t-IsoP and 15-D2c-IsoP, re-spectively, via base-catalyzed isomerization is shown in Fig. 2(A and B). Generation of PGE2 and PGD2 from 15-E2t-IsoP and15-D2c-IsoP, respectively, would also be predicted to occur viaacid catalysis (11).

These findings strongly support the contention that a secondpathway exists for the formation of bioactive PGs in vivo that isindependent of COX. This finding is of potential physiologicaland pharmacological importance because it would be predictedthat the generation of PGs via this mechanism would not beinhibited by aspirin or other COX inhibitors. For purposes ofdiscussion hereafter, PGs possessing a structure identical tothose generated by COX are referred to PGE2 and PGD2. Com-pounds that are enantiomeric to COX-derived PGs are referredto as ent-PGE2 and ent-PGD2. The racemic mixtures are termedrac-PGE2 and rac-PGD2 (Fig. 3).

EXPERIMENTAL PROCEDURES

Materials—Arachidonic acid, dimethylformamide, CCl4, and unde-cane were purchased from Aldrich. Pentafluorobenzyl (PFB) bromide,methoxyamine HCl, diisopropylethylamine, and Apis mellifera venomphospholipase A2 were from Sigma. [2H3]Methoxyamine HCl was fromCambridge Isotope Laboratories, Inc. (Andover, MA). N,O-Bis(trimeth-ylsilyl)trifluoroacetamide was from Supelco Inc. (Bellefonte, PA). N,O-[2H9]Bis(trimethylsilyl)acetamide was from CDN Isotopes (Pointe-Claire, Quebec, Canada). Organic solvents were from EM Science(Darmstadt, Germany). C18 and silica Sep-Pak cartridges were fromWaters Associates (Milford, MA). 60ALK6D TLC plates were fromWhatman (Maidstone, UK). [2H4]PGE2, [2H4]PGD2, and [2H4]PGB2

were from Cayman Chemical Co., Inc. (Ann Arbor, MI). [3H7]PGE2 and[3H7]PGD2 (�180–200 Ci/mmol) were obtained from Amersham Bio-sciences. 15-E2t-[

3H6]IsoP (160 Ci/mmol) was commercially preparedfrom [3H7]PGE2 by Amersham Biosciences as described (10). The enan-tiomer of PGE2 (ent-PGE2) was synthesized by Dr. Doug Taber (Uni-versity of Delaware) (15).

Epimerization of 15-E2t-IsoP in Phosphate Buffer—15-E2t-IsoP (1 �M)was incubated in 50 mM KPO4 buffer, pH 7.4, for 0–24 h. Subsequently,fractions were analyzed for 15-E2t-IsoP, PGE2, 15-A2t-IsoP, and PGA2

by GC/MS and NMR as described (10, 16, 17).Oxidation of Arachidonic Acid—Arachidonic acid was oxidized in

vitro using an iron/ADP/ascorbate mixture as previously described (16,17).

Isolation of E/D-ring IsoPs and PGs from Rodent and Human Tissueand Urine—A mixture of E/D-ring IsoPs and PGs was isolated from thelivers of Sprague-Dawley rats 2 h after intragastric administration ofCCl4 (2 mg/kg) in corn oil (16, 17). The animals were anesthetized withpentobarbital (60 mg/kg) intraperitoneally and killed, and the liverswere removed. Depending on the experiment, 1–4 g of tissue wasimmediately extracted to obtain a crude phospholipid extract contain-ing IsoPs and PGs esterified in phospholipids. The lipid extract was

then subjected to hydrolysis (30 min) in boronate buffer with A. mellif-era venom containing phospholipase A2 as described (16, 17). In controlexperiments, complete hydrolysis of 2-[3H7]arachidonylphosphatidylc-holine was effected during this incubation. In addition, these hydrolysisconditions resulted in �5% epimerization of 15-E2t-IsoP to PGE2.Subsequently, free IsoPs and PGs were extracted, partially purifiedusing C18 and silica Sep-Pak cartridges, and subjected to HPLC. Forselected experiments, liver tissue was also obtained from day 19 COX-1�/�/COX-2�/� mouse pups harvested in utero as described (18).

In some experiments, 24-h urine samples were collected from ratstreated with CCl4 or from normal humans. Unesterified IsoPs and PGswere extracted using Sep-Pak columns as described (6).

HPLC Separation of Racemic E/D-ring PGs and IsoPs—Dependingon the experiment, incubations of oxidized arachidonic acid, partiallypurified tissue extracts, or urine samples were analyzed for rac-PGE2,rac-PGD2, or rac-15-E2t-IsoP (9, 10). To the biological sample was added�0.5–3 �Ci of [3H7]PGE2, [3H7]PGD2, or 15-E2t-[

3H6]IsoP. The mixturewas then subjected to four successive HPLC purification steps. To

FIG. 2. Base-catalyzed mechanism of epimerization of 15-E2t-IsoP to PGE2 (A) and 15-D2c-IsoP to PGD2 (B) involving keto-enoltautomerization. Acid-catalyzed epimerization can also occur. R �either hydrogen or phospholipids depending on whether the IsoPsepimerize as free acids or esterified in phospholipids.

FIG. 1. Structure of the bicycloendoperoxide intermediate PGH2 (A) derived from COX and structures of the IsoP endoperoxides15-H2t-IsoP (B) and 15-H2c-IsoP (C).

Prostaglandin Formation via the Isoprostane Pathway28480

by guest on January 7, 2016http://w

ww

.jbc.org/D

ownloaded from

maximize purification and resolution of each compound, we used HPLCprocedures that yielded relatively long retention volumes for each com-pound (�15–40 ml), and each solvent was run isocratically. In pilotexperiments, it was also shown that the three compounds readily sep-arated from one another under the HPLC conditions utilized. In addi-tion, radiolabeled PGE2, PGD2, and 15-E2t-IsoP separated to a signifi-cant extent (1–2.5 ml) from unlabeled compounds due to the fact thatthe radiolabeled compounds contained either six or seven tritium at-oms. Thus, for each HPLC step, fractions corresponding to those con-taining both labeled and unlabeled PGs or IsoPs were collected andpooled for further purification. For rac-PGE2 and rac-15-E2t-IsoP, thefirst HPLC step was normal-phase using a Econosil SI column (25 cm �4.6 mm, 5-�m particles; Alltech Associates Inc., Deerfield, IL). Thesolvent system was 88:12:0.1 (v/v/v) hexane/isopropyl alcohol/acetic acidat a flow rate of 1 ml/min. The second HPLC step was reversed-phaseusing an Econosil C18 column (25 cm � 4.6 mm, 5 �m; Alltech Associ-ates Inc.). The solvent system was 30:70:0.1 (v/v/v) acetonitrile/water/acetic acid at a flow rate of 1 ml/min. For the third and fourth HPLCsteps, IsoPs or PGs were converted to PFB esters and rechromato-graphed on normal- and reversed-phase HPLC columns. A solventsystem of 92:8 (v/v) hexane/isopropyl alcohol was used for the thirdHPLC step, and 51:49 (v/v) acetonitrile/water was used for the fourthHPLC step, both at a flow rate of 1 ml/min. For the purification ofrac-PGD2, the same columns were utilized, but the solvent systemsvaried. For the first HPLC step, the solvent system was 93:7:0.1 (v/v/v)hexane/isopropyl alcohol/acetic acid; the second HPLC solvent systemwas 33:67:0.1 (v/v/v) acetonitrile/water/acetic acid; the third HPLC sol-vent system was 95:5 (v/v) hexane/isopropyl alcohol; and the fourthHPLC solvent system was 58:42 (v/v) acetonitrile/water.

Chiral HPLC Separation of rac-PGE2 and rac-PGD2—Racemic PGspurified by the methods described above were subsequently subjected tochiral HPLC to separate enantiomers using a Chiralpak AD column (25cm � 4.6 mm, 5 �m; Chiral Technologies, Exton, PA). To separaterac-PGE2, a solvent system of 93:7 (v/v) hexane/isopropyl alcohol wasutilized; and for rac-PGD2, a solvent system of 95:5 (v/v) hexane/isopro-pyl alcohol was employed.

Analysis of PGs and IsoPs by GC/MS—Quantification of E/D-ringPGs and IsoPs in partially purified biological extracts and throughoutsubsequent HPLC purification procedures was performed by analyzingaliquots by selected ion monitoring GC/negative ion chemical ionizationMS using either [2H4]PGE2 or [2H4]PGD2 as an internal standard.Compounds were quantified as O-methyloxime, PFB ester, trimethyl-silyl ether derivatives by monitoring the M-PFB (M � 181) ions at m/z524 for endogenous compounds and at m/z 528 for the deuteratedstandards (10, 19).

RESULTS

Epimerization of 15-E2t-IsoP in Phosphate Buffer—We ini-tially determined the extent to which 15-E2t-IsoP undergoesepimerization to PGE2 in a buffered solution at physiologicalpH (50 mM KPO4, pH 7.4). Products that were quantified in-cluded the starting material 15-E2t-IsoP and PGE2 and theirrespective dehydration products, 15-A2t-IsoP and PGA2.Amounts are expressed as percent of total PG at a particulartime point. The identification of PGE2 was also confirmed byNMR comparison with a chemically pure PGE2 standard. The

results are shown in Fig. 4. As shown, 15-E2t-IsoP epimerizedin a time-dependent manner to PGE2. The half-life for thisconversion under the conditions noted was �2 h. In addition,small amounts of 15-A2t-IsoP and PGA2 were formed, presum-ably as a result of dehydration of 15-E2t-IsoP and PGE2, re-spectively. These findings support the hypothesis that E2/D2-IsoPs can readily rearrange to E/D-ring PGs in aqueousenvironments.

Analysis of E2/D2-IsoPs from the Oxidation of ArachidonicAcid in Vitro and in Vivo—Fig. 5A shows the selected ioncurrent chromatograms for E2/D2-IsoPs obtained from theanalysis of arachidonic acid with iron/ADP/ascorbate for 2 h.Compounds were analyzed as O-methyloxime, PFB ester, tri-methylsilyl ether derivatives. In the lower m/z 528 chromato-gram are two peaks representing the syn- and anti-O-methyl-oxime isomers of the [2H4]PGE2 internal standard. In theupper m/z 524 chromatogram are a series of peaks representingvarious E2/D2-IsoPs. The peaks indicated by asterisks repre-sent compounds that co-chromatographed upon GC with theO-methyloxime isomers of chemically synthesized PGE2. Inaddition, the peaks denoted by plus signs co-chromatographedwith the O-methyloxime isomers of chemically pure PGD2. Thetotal E2/D2-IsoPs present were �1500 ng/g of arachidonic acid.The materials designated by the peaks denoted by asterisksand plus signs each represent �20% of the total E2/D2-IsoPs inthe mixture.

In addition to the analysis of oxidized arachidonic acid invitro, Fig. 5B shows the selected ion current chromatogramsobtained from the hydrolysis of rat liver phospholipids afteradministration of CCl4 to animals to induce oxidant stress. Asshown, a similar pattern of peaks was present as shown in Fig.5A. The analyses in Fig. 5 (A and B) were performed on sepa-rate days, accounting for differences in GC retention time.

FIG. 3. Structures of PGE2 and PGD2 and their respective enantiomers.

FIG. 4. Time course of epimerization of 15-E2t-IsoP in KPO4buffer, pH 7.4. Data are expressed as means � S.D.

Prostaglandin Formation via the Isoprostane Pathway 28481

by guest on January 7, 2016http://w

ww

.jbc.org/D

ownloaded from

Again, the peaks indicated by asterisks represent compoundsthat co-chromatographed upon GC with chemically synthesizedPGE2, whereas those denoted by plus signs co-chromato-

graphed with PGD2. The total E2/D2-IsoPs present in this sam-ple were �400 ng/g of liver tissue. The materials designated bythe peaks denoted by the asterisks and plus signs each repre-

FIG. 5. A, analysis of arachidonic acid oxidized in vitro for E2/D2-IsoPs by GC/negative ion chemical ionization MS; B, analysis ofhydrolyzed lipid extracts from livers of rats treated with CCl4 for E2/D2-IsoPs by GC/negative ion chemical ionization MS. The twopeaks in the m/z 528 ion current chromatograms represent syn- and anti-O-methyloxime isomers of the [2H4]PGE2 internal standard. A series ofpeaks representing E2/D2-IsoPs are in the m/z 524 chromatograms. Compounds denoted by asterisks co-chromatographed with the O-methyloximeisomers of PGE2, whereas those denoted by the plus signs co-chromatographed with the O-methyloxime isomers of PGD2. Analyses were performedon separate days, accounting for differences in GC retention time.

FIG. 6. HPLC analysis of a mixture of E2/D2-IsoPs from hydrolyzed rat liver phospholipids for rac-PGE2 in Fig. 5B. Details regardingsolvent systems used are described under “Experimental Procedures.” Tritiated PGE2 was added to the mixture at the beginning, and aliquots ofeluted fractions were assayed for radioactivity (●). Aliquots were also assayed and quantified by GC/MS for the presence of E 2/D2-IsoP peaks withthe same retention time as authentic PGE2 (Œ). All HPLC purifications were carried out isocratically. A, normal-phase HPLC as free acids of theinitial mixture of E2/D2-IsoPs as shown in Fig. 5B. The plus sign denotes fractions in which chemically pure unlabeled PGE2 eluted using thissolvent system. B, reversed-phase HPLC as free acids of the material that eluted at the retention volume between 14.5 and 18.5 ml in A. C,normal-phase HPLC as PFB esters of the material that eluted at the retention volume between 28.5 and 33 ml in B. D, reversed-phase HPLC asPFB esters of the material that eluted at the retention volume between 27.5 and 34 ml in C.

Prostaglandin Formation via the Isoprostane Pathway28482

by guest on January 7, 2016http://w

ww

.jbc.org/D

ownloaded from

sent �15–20% of the total E2/D2-IsoPs in the mixture. A verysimilar pattern of peaks representing E2/D2-IsoPs was ob-tained from the livers of both COX-1�/�/COX-2�/� and controlfetal mice without induction of oxidant stress, although totalE2/D2-IsoP levels were �5–10 ng/g of liver tissue (data notshown). In addition, a pattern of peaks virtually identical tothose shown in the chromatograms in Fig. 5 was obtained afterhydrolysis of rat liver phospholipids that had been treated withmethyloxime HCl prior to hydrolysis. These latter findingssuggest that epimerization of E2/D2-IsoPs occurs while com-pounds are esterified in phospholipids. Taken together, thesedata support the contention that significant amounts of com-pounds that coelute upon GC with PGE2 and PGD2 are gener-ated from the peroxidation of arachidonic acid in vitro and invivo.

Purification of Putative rac-PGE2 from Rat Liver Hydroly-sates by HPLC—We subsequently sought to determine whetherthe compounds from rat liver hydrolysates that coeluted uponGC with PGE2 and PGD2 were, in fact, structurally identical toPGE2 and PGD2 and their respective enantiomers. If PGE2 andPGD2 are formed via the IsoP pathway, it would be predictedthat they would be racemic mixtures because they would beformed from the epimerization of rac-15-E2t-IsoP and rac-15-D2c-IsoP, respectively (6, 7). Of note, enantiomers of PGE2 andPGD2 would not be expected to separate using standard non-chiral HPLC methods.

The formation of rac-PGE2 was assessed initially. For thesestudies, �2000 ng of E2/D2-IsoPs from rat liver containing 3�Ci of [3H7]PGE2 was subjected to four successive HPLC puri-fication steps. The first HPLC step was normal-phase using asolvent system of 88:12:0.1 (v/v/v) hexane/isopropyl alcohol/acetic acid. Aliquots of fractions that eluted from the HPLCcolumn were then analyzed for E2/D2-IsoPs by GC/MS and forradioactivity (Fig. 6A). Radiolabeled PGE2 eluted in this sys-tem between 16 and 18.5 min. Compounds representing endog-enous E2/D2-IsoPs were present that had the same retentiontime upon GC as PGE2, but that eluted with different retentionvolumes compared with PGE2 upon HPLC (10–12, 18–21, and22.5–25 ml). Radiolabeled PGE2 eluted at a volume of �1.0–1.5ml after unlabeled PGE2 using this HPLC solvent system.Significantly, as shown in Fig. 6A, an endogenous E2/D2-IsoPpeak (indicated by the plus sign) was detected that coelutedwith unlabeled PGE2, suggesting that this compound is endo-genously derived rac-PGE2.

The material that eluted from the HPLC column between14.5 and 18.5 ml in Fig. 6A was subsequently subjected toreversed-phase HPLC using an isocratic solvent system of 30:70:0.1 (v/v/v) acetonitrile/water/acetic acid. Aliquots of frac-tions collected were again analyzed for endogenous E2/D2-IsoPsby GC/MS and for radioactivity (Fig. 6B). Radiolabeled PGE2

eluted from the HPLC column with a retention volume of28.5–32.5 ml. Analysis of aliquots of the eluted fractions byGC/MS showed that almost all of the unlabeled E2/D2-IsoPmaterial detected in the chromatogram eluted at the retentionvolume of unlabeled PGE2 (29.5–33 ml), except for a smallamount of additional material that eluted at 37–39 ml.

Altering the polarity of a compound by derivatization andrechromatography of the compound can provide a powerfulapproach for purification and separation of biomolecules (9).Thus, the material that eluted from the HPLC column between28.5 and 33 ml in Fig. 6B was converted to a PFB ester andrechromatographed on a normal-phase HPLC column using asolvent system of 92:8 (v/v) hexane/isopropyl alcohol. Fig. 6Cshows the result of this HPLC step. Radiolabeled PGE2 elutedbetween 29.5 and 33.5 ml. A large peak representing endoge-

nous E2/D2-IsoPs that coeluted with the PFB ester of unlabeledPGE2 was detected (27.5–31.5 ml).

Compounds that eluted from the HPLC column between 27.5and 34 ml in Fig. 6C were then pooled. This material wassubjected to further purification by reversed-phase HPLC us-ing a solvent system of 51:49 (v/v) acetonitrile/water. The re-sults of the analyses for radioactivity and endogenous E2/D2-IsoPs in the eluted fractions are shown in Fig. 6D. A singleE2/D2-IsoP peak presumably representing rac-PGE2 was pres-ent that coeluted exactly with the PFB ester of unlabeled PGE2

(42–44.5 ml). Radiolabeled PGE2 eluted slightly before theendogenous E2/D2-IsoP compound. Virtually identical resultswere obtained when putative rac-PGE2 generated from arachi-donic acid oxidized in vitro was analyzed by the HPLC proto-cols described above.

Analysis of Endogenous Putative rac-PGE2 by GC/MS—Thematerial that eluted between 40.5 and 44.5 ml upon the fourthHPLC step was then analyzed by GC/MS. As shown in Fig. 7,two E2/D2-IsoP peaks were present in the m/z 524 chromato-gram, representing the syn- and anti-O-methyloxime isomersof putative rac-PGE2. The amount of putative rac-PGE2 pres-ent in this rat liver hydrolysate was �35 ng/1000 ng of totalE2/D2-IsoP based upon losses of [3H7]PGE2 that occurred withthe four HPLC purification steps. When the material indicatedby peaks in the m/z 524 chromatogram in Fig. 7 was mixed withan equivalent amount of derivatized synthetic PGE2, the twocompounds co-chromatographed perfectly upon capillary GCwithout any suggestion of a shoulder on the GC peaks (data notshown). Additional experiments were subsequently under-taken to confirm the identification of the compound in Fig. 7 asPGE2. First, analysis of the material as a deuterated O-meth-yloxime derivative disclosed the presence of one carbonyl

FIG. 7. Selected ion current chromatogram obtained from theGC/MS analysis of the material that eluted at a retention vol-ume between 40.5 and 44.5 ml in Fig. 6D. Only a single set of m/z524 peaks representing the syn- and anti-O-methyloxime isomers ofendogenous putative rac-PGE2 remained after the four HPLC purifica-tion procedures shown in Fig. 6. The peaks in the m/z 528 chromato-gram represent the syn- and anti-O-methyloxime isomers of the deu-terated PGE2 internal standard. The amount of putative rac-PGE2 inthe fraction analyzed was �35 ng/1000 ng of total E2/D2-IsoP.

Prostaglandin Formation via the Isoprostane Pathway 28483

by guest on January 7, 2016http://w

ww

.jbc.org/D

ownloaded from

group. Second, analysis as a deuterated trimethylsilyl etherderivative revealed that the compound had two hydroxylgroups. Third, catalytic hydrogenation showed two doublebonds (16). Finally, treatment of putative rac-PGE2 with 15%methanolic KOH for 30 min converted it to a compound with amolecular weight and retention time identical to those of PGB2

when analyzed by GC/MS (Fig. 8) (20). Taken together, thesefindings strongly support the contention that the material rep-resented in the m/z 524 chromatogram in Fig. 7 is rac-PGE2.

Analysis of Putative rac-PGE2 by Chiral HPLC—As noted, itis predicted that PGE2 generated by the IsoP pathway shouldbe racemic. The HPLC steps utilized above to purify putative

FIG. 8. Selected ion current chromatogram from the GC/MS analysis of putative rac-PGE2 in Fig. 7 after treatment withmethanolic KOH. In the lower m/z 438 chromatogram is a single peak representing the [2H4]PGB2 internal standard. In the upper m/z 434chromatogram is a single peak that co-chromatographed perfectly with unlabeled PGB2 and that represents endogenous rac-PGB2.

Prostaglandin Formation via the Isoprostane Pathway28484

by guest on January 7, 2016http://w

ww

.jbc.org/D

ownloaded from

rac-PGE2 will not separate enantiomers. Thus, the compoundsrepresented in the chromatogram in Fig. 7 were subjected tochiral column chromatography, and fractions that eluted fromthe HPLC column were analyzed by GC/MS. Fig. 9 shows theresults of the analysis. The peak indicated by the asteriskco-chromatographed under these HPLC conditions with chem-ically synthesized PGE2, whereas the peak denoted by the plussign co-chromatographed with chemically synthesized ent-PGE2. The material from each peak co-chromatographed per-fectly with both PGE2 and ent-PGE2 upon GC and was indis-tinguishable upon MS analysis. Approximately equal amountsof the compounds were present, as would be expected. Further-more, the ratio of methyloxime isomers of ent-PGE2 was essen-tially identical to that of PGE2. Taken together, these studiesprovide compelling evidence that PGE2 and ent-PGE2 are gen-erated in vivo in significant quantities from the IsoP pathway.Essentially identical results were obtained from the analysis ofputative rac-PGE2 formed from the peroxidation of arachido-nate in vitro.

Purification of Putative rac-PGD2 from Rat Liver Hydroly-sates by HPLC—As noted in Fig. 5 (A and B), chromatographicpeaks were present that coeluted not only with PGE2, but alsowith PGD2. We thus employed similar approaches as thoseused to obtain evidence for the formation of rac-PGE2 in vitroand in vivo to determine whether rac-PGD2 is also generated.Table I shows the HPLC conditions utilized to purify putativerac-PGD2 and the retention time of the compound at each step.Fig. 10 illustrates the results from GC/MS analysis of thematerial that eluted between 21 and 24 ml, where PGD2 eluted,upon the fourth HPLC step. As shown, two E2/D2-IsoP peakswere present in the m/z 524 chromatogram, representing thesyn- and anti-O-methyloxime isomers of putative rac-PGD2.The amount of putative rac-PGD2 present in the rat liverhydrolysate from this analysis was �55 ng/1000 ng of totalE2/D2-IsoP, based upon losses of [3H7]PGD2 that occurred withthe four HPLC purification steps. When the material denotedby the peaks in the m/z 524 chromatogram in Fig. 10 was mixedwith an equivalent amount of derivatized synthetic PGD2, thetwo compounds co-chromatographed perfectly upon capillaryGC without any suggestion of a shoulder on the GC peaks (datanot shown). Additional experiments confirmed the identifica-tion of the compound in Fig. 10 as PGD2. First, analysis of the

material as a deuterated O-methyloxime derivative disclosedthe presence of one carbonyl group. Second, analysis as a deu-terated trimethylsilyl ether derivative revealed that the com-pound had two hydroxyl groups. Third, catalytic hydrogenationshowed two double bonds.

Analysis of Putative rac-PGD2 by Chiral HPLC—As withPGE2 generated by the IsoP pathway, it is predicted that PGD2

should be racemic. Thus, the compounds represented in the m/z524 chromatogram in Fig. 10 were subjected to chiral columnchromatography, and fractions that eluted from the HPLCcolumn were analyzed by GC/MS (Table I). Fig. 11 shows theresults of the analysis. The peak indicated by the asteriskco-chromatographed under these HPLC conditions with chem-

FIG. 9. Chiral HPLC separation of rac-PGE2. The putative rac-PGE2 purified as described in the legend to Fig. 6 was subjected tochiral column chromatography using the solvent system 93:7 (v/v) hex-ane/isopropyl alcohol. Aliquots of fractions that eluted from the HPLCcolumn were then analyzed for PGE2 by GC/MS. The peak indicated bythe asterisk co-chromatographed in the HPLC solvent system withchemically pure PGE2, whereas the peak denoted by the plus signcoeluted with ent-PGE2.

TABLE IHPLC retention times for purification of putatitive rac-PGD2 and

chiral separation of enantiomersSee “Experimental Procedures” for details regarding the columns

used and solvent conditions employed for each HPLC step.

HPLC step Conditions Retention volume ofputative rac-PGD2

a

ml

1 Straight-phase (as free acid) 22–242 Reversed-phase (as free acid) 22–243 Straight-phase (as PFB ester) 29–344 Reversed-phase (as PFB ester) 21–24Chiral HPLC Straight-phase (as PFB ester)

PGD2 28–33ent-PGD2 36–41a The flow rate those of for all HPLC solvents was 1 ml/min. The

retention times of putative rac-PGD2 were identical to chemically purePGD2 in HPLC Steps 1–4.

FIG. 10. Selected ion current chromatogram obtained from theGC/MS analysis of the material that eluted at a retention vol-ume of 21–24 ml after the fourth HPLC step to purify rac-PGD2as noted in Table I. Only a single set of m/z 524 peaks representingthe syn- and anti-O-methyloxime isomers of endogenous putative rac-PGD2 remained after the four HPLC purification procedures shown inTable I. The peaks in the m/z 528 chromatogram represent the syn- andanti-O-methyloxime isomers of the deuterated PGD2 internal standard.The amount of putative rac-PGE2 in the fraction analyzed was �55ng/1000 ng of total E2/D2-IsoP.

Prostaglandin Formation via the Isoprostane Pathway 28485

by guest on January 7, 2016http://w

ww

.jbc.org/D

ownloaded from

ically synthesized PGD2, whereas the peak denoted by the plussign represents ent-PGD2. The material from each peak co-chromatographed perfectly with PGD2 upon GC and was indis-tinguishable upon MS analysis. Approximately equal amountsof compounds were present, as would be expected; and therelative amounts of methyloxime isomers of PGD2 and ent-PGD2 were very similar. Taken together, these studies providestrong evidence that PGD2 and ent-PGD2, in addition to PGE2

and ent-PGE2, are generated in vivo in significant quantitiesfrom the IsoP pathway. Again, essentially identical resultswere obtained from the analysis of putative rac-PGD2 formedfrom the peroxidation of arachidonate in vitro.

Quantitative Analysis of rac-PGE2 and rac-PGD2 Generatedin Vitro and in Rat Liver—The above studies provide substan-tial support for the hypothesis that PGE2 and PGD2 and theirrespective enantiomers can be generated via the IsoP pathway.We subsequently undertook experiments to determine the totalamounts of rac-PGD2 and rac-PGE2 generated from the oxida-tion of arachidonate in vitro and in vivo in comparison withother E2/D2-IsoPs. These determinations are highly importantbecause, based on previous reports (4), the vast majority(�90%) of endoperoxide intermediates generated by the autox-idation of polyunsaturated fatty acids have side chains that arecis in relation to the prostane ring. Indeed, we have recentlyconfirmed that endoperoxides with cis-side chains predominateover trans-side chain compounds when arachidonate is oxi-dized in vitro (5). Thus, it would be predicted that the IsoPendoperoxide with a structure identical to PGH2 generatedfrom the peroxidation of arachidonate in vitro and in vivowould compose a trivial fraction of the total endoperoxides thatare formed. Therefore, the amounts of rac-PGE2 and rac-PGD2

that are subsequently generated from this endoperoxide inter-mediate would be present at no more than a few nanograms/1000 ng of total E2/D2-IsoPs (4). Employing the HPLC protocolsutilized for the studies described above, we quantified rac-PGE2, rac-PGD2, and rac-15-E2t-IsoP in vitro and in vivo andalso assessed the relative formation of each enantiomer in theracemic mixture. Losses of endogenous material during thechromatographic procedures were accounted for by determin-ing the percent loss of the respective radiolabeled PG added tothe samples prior to purification. As noted in Table II, theamounts of rac-PGE2 and rac-PGD2 far exceeded those pre-dicted based upon the observations of O’Connor et al. (4), and

the quantities of rac-PGE2 were at least as great as, if notgreater than, those of rac-15-E2t-IsoP both in vitro and in vivo.In summary, these quantitative data provide support thatPGE2 and PGD2 and their respective enantiomers are gener-ated in significant amounts via the IsoP pathway.

Excretion of Unesterified rac-PGE2 and rac-PGD2 in Rat andHuman Urine in Vivo—In addition to detecting the in vivoformation of rac-PGE2 and rac-PGD2 esterified in rat livertissue, we also sought to determine whether these compoundsare present unesterified in human and rodent urine at base lineand whether they increase in association with oxidant stress.Table III shows the results of studies performed to determinethe relative amounts of these eicosanoids under these condi-tions. As shown, at baseline, the relative levels of both ent-PGE2 and ent-PGD2 in humans and rats were low and com-posed no more than 10% of the total rac-PGE2 and rac-PGD2

generated. In addition, in several of the urine samples obtainedfrom normal humans and rats at baseline, the levels of rac-PGE2 were below the limits of assay detection. On the otherhand, after treatment of rats with CCl4, the levels of bothent-PGE2 and ent-PGD2 increased significantly. This was par-ticularly the case for ent-PGD2. If one assumes that an amountof PGD2 equivalent to that of ent-PGD2 is generated via theIsoP pathway after administration of CCl4, then �15% of PGD2

present in rat urine under these conditions is formed by amechanism independent of COX. Analogously, �30% of rac-PGD2 (total of PGD2 and ent-PGD2) would thus be predicted tobe generated by this mechanism. Fig. 12 (A and B) shows theresults from chiral analysis of rat urine for PGD2 and ent-PGD2

at base line and after treatment with CCl4. As shown, at baseline, the chromatographic peak comprising PGD2 (*) in Fig.12A greatly exceeded the enantiomer (�), suggesting that COXcontributes to the vast majority of PGD2 production at baseline. After CCl4 administration (Fig. 12B), the levels of ent-PGD2 (�) increased dramatically in relation to PGD2 (*), sup-porting the contention that CCl4 has induced PG formation viathe IsoP pathway.

To provide further evidence that PGs can be generated viathe IsoP pathway, we pretreated rats with indomethacin (10mg/kg intraperitoneally for 24, 12, and 1 h) prior to CCl4administration and collected urine for 24 h after the oxidantwas given (6, 21). Fig. 12C shows the results from the chiralanalysis of rac-PGD2. As shown, the chromatographic peaksrepresenting PGD2 and ent-PGD2 are very similar. The levelsof PGD2 and ent-PGD2 were �30% of those present in CCl4-treated rats not given indomethacin (Table III) and support thecontention that significant amounts of unesterified PGD2 (andto a lesser extent, PGE2) can be formed by a mechanism inde-pendent of COX in settings of oxidant stress.

FIG. 11. Chiral HPLC separation of rac-PGD2. The putative rac-PGD2 purified as noted in Table I was subjected to chiral columnchromatography using the solvent system 95:5 (v/v) hexane/isopropylalcohol. Aliquots of fractions that eluted from the HPLC column werethen analyzed for PGD2 by GC/MS. The peak indicated by the asteriskco-chromatographed in the HPLC solvent system with chemically purePGD2, whereas the peak denoted by the plus sign represents ent-PGD2.

TABLE IILevels of rac-PGE2, rac-PGD2, and rac-15-F2t-IsoP generated from the

peroxidation of arachidonic acid in vitro and in liver hydrolysatesfrom rats after treatment with CCl4

Data are expressed as means � S.D.

ng/mg arachidonate PG/ent-PG ratio

In vitro incubationsa

rac-PGE2 56 � 21 1.06 � 0.18rac-PGD2 78 � 19 0.93 � 0.19rac-15-E2t-IsoP 39 � 16 1.13 � 0.22

ng/g liver PG/ent-PG ratio

Rat liver hydrolysatesb

rac-PGE2 24 � 16 0.93 � 0.21rac-PGD2 37 � 12 1.01 � 0.14rac-15-E2t-IsoP 21 � 18 0.99 � 0.18

a Incubation time was 2 h. Total E2/D2-IsoPs generated in experi-ments were 1240 � 450 ng/mg of arachidonate (n � 8).

b Total E2/D2-IsoPs were 561 � 232 ng/g of liver tissue (n � 6).

Prostaglandin Formation via the Isoprostane Pathway28486

by guest on January 7, 2016http://w

ww

.jbc.org/D

ownloaded from

DISCUSSION

This study describes the formation of PGE2 and PGD2 inde-pendent of COX and involving the free radical-catalyzed per-oxidation of arachidonate. We have reported that significantamounts of rac-PGE2 and rac-PGD2 are generated in vitro andin vivo in settings of oxidant stress. Unlike PGs formed viaCOX, generation of eicosanoids by this mechanism results inthe formation of compounds as racemic mixtures because theoxygenation of arachidonic acid does not occur stereospecifi-cally (2, 3, 6). Our initial interest in determining whether PGsare generated via the IsoP pathway emerged from the obser-vation that compounds with the same molecular weights andGC retention times as PGE2 and PGD2 are present when ana-lyzed by GC/MS in mixtures of arachidonate oxidized in vitroand in rat liver hydrolysates. Utilizing a variety of high resolv-ing chromatographic, chemical, and mass spectrometric ap-proaches, we have found that substantial quantities of theseracemic PGs can be generated. Analysis of putative rac-PGE2

and rac-PGD2 by chiral HPLC revealed that each compound iscomposed of two enantiomers generated in equal amounts invitro and in liver tissue from rats exposed to oxidant stress.rac-PGE2 and rac-PGD2 were also present in the unesterifiedform in significant amounts in urine from rats treated withCCl4, and their formation was unaffected by COX inhibition.That COX is not involved in the formation of these compoundsis also supported by the findings that these PGs could begenerated in vitro without COX and were present in vivo es-terified in phospholipids. COX is not active on arachidonateesterified in phospholipids (1). Finally, compounds with reten-tion times and molecular weights identical to those of PGE2

and PGD2 were present when liver tissue from COX-1�/�/COX-2�/� mice was analyzed for E2/D2-IsoPs.

We propose that the formation of PGs independent of COXinvolves the generation of two IsoP endoperoxide intermediates(rac-15-H2t-IsoP and rac-15-H2c-IsoP) that isomerize to rac-15-E2t-IsoP and rac-15-D2c-IsoP, respectively. These eicosanoidssubsequently undergo rapid epimerization to compounds iden-tical in all respects to racemic PGE2 and PGD2, respectively(Fig. 2). A number of lines of evidence that we and others haveobtained support this proposed mechanism of formation. Asnoted, we have previously shown that IsoPs contain E/D-, F-,and thromboxane-type prostane rings (7). However, an impor-tant distinction between IsoPs and PGs is that IsoP bicycloen-doperoxide intermediates contain side chains that are predom-inantly (�90%) oriented cis in relation to the prostane ring (4).Indeed, we have recently confirmed that endoperoxides withcis-side chains predominate over trans-side chain compoundswhen arachidonate is oxidized (5). One IsoP that is formed inabundance in vivo is 15-E2t-IsoP, which is generated from theendoperoxide intermediate 15-H2t-IsoP (10). It would also bepredicted that the endoperoxide 15-H2c-IsoP can rearrange toform the analogous D-ring IsoP 15-D2c-IsoP. In contrast toother types of prostanoids, E2/D2-IsoPs are �-hydroxyketone-containing compounds that can undergo reversible keto-enoltautomerization under both acidic and basic conditions, allow-ing rearrangement of the side chains that are initially cis to the

more stable trans-configuration. That the trans-configurationis highly favored has been demonstrated by the finding that,when PGE2 is subjected to conditions that induce keto-enoltautomerism, �10% of the compound rearranges to the cis-sidechain isomer 15-E2t-IsoP (11). Also, attempts to synthesize15-D2c-IsoP have been unsuccessful because epimerization atC-12 readily occurs during synthesis to yield PGD2 (12).

In this study, we have shown that chemically synthesized15-E2t-IsoP is unstable and rapidly epimerizes nonenzymati-cally to PGE2 in phosphate buffer at physiological pH. It islikely that the isomerization is further enhanced in the pres-ence of protein-containing biological solutions, which have beenshown to facilitate epimerization and dehydration of other ei-cosanoids (22). Whether the isomerization can be catalyzedenzymatically is unknown. That epimerization of IsoP endoper-oxides occurred in the in vitro and in vivo studies reportedherein is strongly supported by the fact that comparableamounts of rac-PGE2 and rac-15-E2t-IsoP were generated fromthe peroxidation of arachidonate. In addition, the abundance ofrac-PGD2 lends credence to the hypothesis that epimerizationoccurs readily. As noted by our findings, the formation of rac-PGD2 predominates over that of rac-PGE2 both at base line andafter oxidant stress, perhaps because this compound would bepredicted to form more readily from the epimerization of rac-15-D2c-IsoP compared with PGE2 from 15-E2t-IsoP. In thisstudy, the lack of a chemically synthesized 15-D2c-IsoP stand-ard precludes our detection of this compound in vitro and invivo, although it would be predicted that it would not be pres-ent in significant amounts.

Our results also suggest that epimerization of 15-E2t-IsoPand 15-D2c-IsoP to PGE2 and PGD2, respectively, occurs to asignificant extent while these compounds are esterified inphospholipids based on two lines of evidence. First, a pattern ofpeaks virtually identical to that shown in the chromatogramsin Fig. 5 was obtained after hydrolysis of rat liver phospholip-ids that had been treated with methyloxime HCl prior to hy-drolysis. Second, in control experiments, the conversion of ex-ogenously added 15-E2t-IsoP to PGE2 occurred to a negligibleextent during sample workup.

A number of important physiological and pharmacologicalissues emerge from the this study. The first relates to the factthat formation of bioactive PGs occurs in vivo to a significantextent via the IsoP pathway in settings of oxidative stress andpotentially in other inflammatory situations. Although levels ofPGs derived via this mechanism are low at base line in normalhumans and animals, they represent up to 15% of PGD2 pres-ent in the urine of rats treated with CCl4, and these PGs areformed independent of COX inhibition. IsoPs have been impli-cated as mediators of oxidant stress (23–25). Thus, it will beimportant to investigate the extent to which not only IsoPs, butPGs, contribute to adverse sequelae of oxidative injury.

Although the biological properties of PGE2 and PGD2 havebeen well characterized (1), our studies suggest that equalamounts of the enantiomers of these PGs are also produced. Itwill thus be of interest to explore the bioactivity of ent-PGE2

and ent-PGD2. In this respect, the former compound was syn-

TABLE IIILevels of PGE2, ent-PGE2, PGD2, and ent-PGD2 in urine from rats and humans

Data are expressed as means � S.D. (n � 8).

PGE2 ent-PGE2 PGD2 ent-PGD2

pg/mg creatinineRat urine

Base line 247 � 104 7 � 2 260 � 76 10 � 8�CCl4 1790 � 820 52 � 14 2690 � 640 421 � 123�Indomethacin � CCl4 191 � 110 48 � 21 533 � 220 410 � 78

Human urine (baseline) 127 � 69 4 � 4 195 � 97 12 � 2

Prostaglandin Formation via the Isoprostane Pathway 28487

by guest on January 7, 2016http://w

ww

.jbc.org/D

ownloaded from

thesized for the studies reported herein, and experiments todetermine its biological relevance will likely yield importantinsights into its role in oxidative injury.

The metabolism of PGE2 and PGD2 has been extensivelystudied in animals and humans (1). The metabolism of parentPGs via the formation of C-13,14-dihydro-15-keto derivativesand subsequent �- or �-oxidation generally renders them inac-tive. However, this is not the case for the one IsoP whosemetabolism has been studied in detail, 15-F2t-IsoP. The majormetabolite of this compound is 2,3-dinor-5,6-dihydro-15-F2t-IsoP, which results from one step of �-oxidation and an unusualC-5–C-6 double bond reduction (26). Interestingly, this metab-olite displays bioactivity as a vasoconstrictor similar to that of15-F2t-IsoP (27). Thus, studying the metabolism of ent-PGs, inaddition to their biological activities, may provide importantinsights into their role as mediators of oxidant stress. In thisregard, we have recently found that, unlike PGE2, ent-PGE2 isa poor substrate for 15-hydroxyprostaglandin dehydrogenase,suggesting that the metabolism of this eicosanoid is signifi-cantly different from that PGE2.2

The studies reported herein are highly relevant with re-gard to human pharmacology in that they suggest that asecond pathway operates in vivo to generate PGs and isindependent of COX. That this pathway contributes to theformation of PGs in settings of oxidant stress has been dis-cussed above. On the other hand, the extent to which itcontributes to PG production in other disease states or atbase line has not been elucidated. Administration of nonste-roidal anti-inflammatory agents to humans has been shownto significantly decrease production of PGs and PG metabo-lites, although the degree of suppression varies depending onthe eicosanoid measured. For example, administration ofhigh doses of nonsteroids (e.g. 1.5 g of aspirin or more or theequivalent) to normal human volunteers is associated with a90% reduction in thromboxane formation and a �80% reduc-tion in PGI2 (28–30). In contrast, the same doses of theseagents have been reported to be associated with no greaterthan a 60% decrease in PGE2 excretion (28). In this regard,we have made similar observations (31). The reasons for thisdiscrepancy are unknown; but in light of our findings thatPGs, particularly PGE2, are formed via a non-COX mecha-nism, it is intriguing to postulate that part of the reason thataspirin-like drugs fail to inhibit PGE2 production comparedwith other PGs in certain settings is that the former com-pound can be produced from IsoP intermediates.

In summary, we report that a second pathway exists for theformation of bioactive PGs in vivo that is independent of COX.This finding is likely of physiological and pharmacological im-portance because it would be predicted that the generation ofPGs via this mechanism would not be inhibited by aspirin orother COX inhibitors. The extent to which formation of PGsindependent of COX contributes to human physiology andpathophysiology remains to be elucidated.

Acknowledgments—We thank Drs. Jack Roberts and Doug Taber forhelpful discussions.

REFERENCES

1. Hardman, J. G., and Limbird L. E. (2001) Goodman and Gilman’s The Phar-macological Basis of Therapeutics, 10th Ed., McGraw-Hill Book Co., NewYork

2. Marnett, L. J. (2000) Curr. Opin. Chem. Biol. 4, 545–5523. Marnett, L. J., Rowlinson, S. W., Goodwin, D. C., Kalgutkar, A. S., and Lanzo,

C. A. (1999) J. Biol. Chem. 274, 22903–229064. O’Connor, D. E., Mihelich, E. D., and Coleman, M. C. (1984) J. Am. Chem. Soc.

106, 3577–3584

2 L. Gao, W. E. Zackert, J. J. Hasford, M. E. Danekis, G. L. Milne, C.Remmert, J. Reese, H. Yin, H.-H. Tai, S. K. Dey, N. A. Porter, and J. D.Morrow, unpublished data.

FIG. 12. Chiral analysis of rat urine for rac-PGD2. The puta-tive rac-PGD2 in each case was purified as noted in Table I and under“Results” and subjected to chiral column chromatography using thesolvent system 95:5 (v/v) hexane/isopropyl alcohol. Aliquots of frac-tions that eluted from the HPLC column were then analyzed for PGD2by GC/MS. The peaks indicated by the asterisks co-chromatographedin the HPLC solvent system with chemically pure PGD2, whereas thepeaks denoted by the plus signs represent ent-PGD2. A, chiral anal-ysis of rac-PGD2 in urine from a normal rat. The predominant ste-reoisomer is PGD2. B, chiral analysis of rac-PGD2 in urine from a rattreated with CCl4 (2 ml/kg) to induce oxidant stress. As shown, therelative height of the chromatographic peak representing ent-PGD2compared with PGD2 is significantly higher than that in A. C, chiralanalysis of rac-PGD2 in urine from a rat pretreated with indometha-cin prior to receiving CCl4. Although the relative height of the peakrepresenting PGD2 decreased significantly compared with that in B,the height of the peak representing ent-PGD2 was largely unchanged.See Table III for details.

Prostaglandin Formation via the Isoprostane Pathway28488

by guest on January 7, 2016http://w

ww

.jbc.org/D

ownloaded from

5. Yin, H., Havrilla, C. M., Morrow, J. D., and Porter, N. A. (2002) J. Am. Chem.Soc. 124, 7745–7754

6. Morrow, J. D., Hill, K. E., Burk, R. F., Nammour, T. M., Badr, K. F., andRoberts, L. J. (1990) Proc. Natl. Acad. Sci. U. S. A. 87, 9383–9387

7. Morrow, J. D., Chen, Y., Brame, C. J., Yang, J., Sanchez, S. C., Xu, J., Zackert,W. E., Awad, J. A., and Roberts, L. J. (1999) Drug Metab. Rev. 31, 117–139

8. Morrow, J. D., Harris, T. M., and Roberts, L. J. (1990) Anal. Biochem. 184,1–10

9. Morrow, J. D., Minton, T. A., Badr, K. F., and Roberts, L. J. (1994) Biochim.Biophys. Acta 1210, 244–248

10. Morrow, J. D., Scruggs, J., Chen, Y., Zackert, W. E., and Roberts, L. J. (1998)J. Lipid Res. 39, 1589–1593

11. Daniels, E. G., Krueger, W. C., Kupiecki, F. P., Pike, J. E., and Schneider, W. P.(1968) J. Am. Chem. Soc. 90, 5894–5895

12. Taber, D. F., and Kanai, K. (1998) J. Org. Chem. 63, 6607–660913. Marino, J. P., Fernandez de la Pradilla, R., and Laborde, E. (1987) J. Org.

Chem. 52, 4898–491314. Roth, G. J., Kirschbaum, S., and Bestmann, H. J. (1997) Synlett 618–62015. Taber, D. F., and Jiang, Q. (2000) Tetrahedron 56, 5991–599416. Morrow, J. D., Minton, T. A., Mukundan, C. R., Campbell, M. D., Zackert,

W. E., Daniel, V. C., Badr, K. F., Blair, I. A., and Roberts, L. J. (1994)J. Biol. Chem. 269, 4317–4326

17. Chen, Y., Morrow, J. D., and Roberts, L. J. (1999) J. Biol. Chem. 274,10863–10868

18. Reese, J., Paria, B. C., Brown, N., Zhao, X., Morrow, J. D., and Dey, S. K. (2000)Proc. Natl. Acad. Sci. U. S. A. 97, 9759–9764

19. DuBois, R. N., Awad, J., Morrow, J., Roberts, L. J., and Bishop, P. R. (1994)J. Clin. Invest. 93, 493–498

20. Schneider, C., Boeglin, W. E., and Brash, A. R. (2000) Anal. Biochem. 287,186–189

21. Morrow, J. D., Awad, J. A., Boss, H. J., Blair, I. A., and Roberts, L. J. (1992)Proc. Natl. Acad. Sci. U. S. A. 89, 10721–10725

22. Fitzpatrick, F. A., and Wynalda, M. A. (1983) J. Biol. Chem. 258, 11713–1171823. Longmire, A. W., Roberts, L. J., and Morrow, J. D. (1998) Prostaglandins 48,

247–25624. Morrow, J. D., Minton, T. A., and Roberts, L. J. (1992) Prostaglandins 44,

155–16325. Janssen, L. J. (2001) Am. J. Physiol. 280, L1067–L108226. Roberts, L. J., Moore, K. P., Zackert, W. E., Oates, J. A., and Morrow, J. D.

(1996) J. Biol. Chem. 271, 20617–2062027. Hou, X., Roberts, L. J., Taber, D. F., Morrow, J. D., Kanai, K., Gobeil, F.,

Beauchamp, M. H., Bernier, S. G., Lepage, G., Varma, D. R., and Chemtob,S. (2001) Am. J. Physiol. 281, R391–R400

28. Bippi, H., and Frolich, J. C. (1990) Br. J. Clin. Pharmacol. 29, 305–31029. Rane, A., Oelz, O., Frolich, J. C., Seyberth, H. W., Sweetman, B. J., Watson,

J. T., Wilkinson, G. R., and Oates, J. A. (1978) Clin. Pharmacol. Exp. Ther.23, 658–668

30. FitzGerald, G. A., Oates, J. A., Hawiger, J., Maas, R. L., Roberts, L. J., Lawson,J. A., and Brash, A. R. (1983) J. Clin. Invest. 71, 676–688

31. Daniel, V. C., Minton, T. A., Brown, N. J., Nadeau, J. H., and Morrow, J. D.(1993) J. Chromatogr. 653, 117–122

Prostaglandin Formation via the Isoprostane Pathway 28489

by guest on January 7, 2016http://w

ww

.jbc.org/D

ownloaded from

A. Porter and Jason D. MorrowYin, Hsin-Hsiung Tai, Sudhansu K. Dey, Ned Milne, Catha Remmert, Jeff Reese, HuiyongHasford, Michael E. Danekis, Ginger L. Ling Gao, William E. Zackert, Justin J.  CYCLOOXYGENASEINDEPENDENT OF BIOACTIVE PROSTAGLANDINSFOR THE GENERATION OF

MECHANISMthe Isoprostane Pathway: A via2 and D2Formation of Prostaglandins E

Lipids and Lipoproteins:

doi: 10.1074/jbc.M303984200 originally published online May 13, 20032003, 278:28479-28489.J. Biol. Chem. 

  10.1074/jbc.M303984200Access the most updated version of this article at doi:

  .JBC Affinity SitesFind articles, minireviews, Reflections and Classics on similar topics on the

 Alerts:

  When a correction for this article is posted• 

When this article is cited• 

to choose from all of JBC's e-mail alertsClick here

  http://www.jbc.org/content/278/31/28479.full.html#ref-list-1

This article cites 28 references, 9 of which can be accessed free at

by guest on January 7, 2016http://w

ww

.jbc.org/D

ownloaded from