LC-MS/MS confirms that COX-1 drives vascular prostacyclin whilst gene expression pattern reveals non-vascular sites of COX-2 expression

Nicholas S Kirkby, Anne K Zaiss, Paula Urquhart, Jing Jiao, Philip J Austin, Malak Al-Yamani, Martina H Lundberg, Louise S MacKenzie, Timothy D Warner, Anna Nicolaou, Harvey R Herschman, Jane A Mitchell, Nicholas S Kirkby, Anne K Zaiss, Paula Urquhart, Jing Jiao, Philip J Austin, Malak Al-Yamani, Martina H Lundberg, Louise S MacKenzie, Timothy D Warner, Anna Nicolaou, Harvey R Herschman, Jane A Mitchell

Abstract

There are two schools of thought regarding the cyclooxygenase (COX) isoform active in the vasculature. Using urinary prostacyclin markers some groups have proposed that vascular COX-2 drives prostacyclin release. In contrast, we and others have found that COX-1, not COX-2, is responsible for vascular prostacyclin production. Our experiments have relied on immunoassays to detect the prostacyclin breakdown product, 6-keto-PGF1α and antibodies to detect COX-2 protein. Whilst these are standard approaches, used by many laboratories, antibody-based techniques are inherently indirect and have been criticized as limiting the conclusions that can be drawn. To address this question, we measured production of prostanoids, including 6-keto-PGF1α, by isolated vessels and in the circulation in vivo using liquid chromatography tandem mass spectrometry and found values essentially identical to those obtained by immunoassay. In addition, we determined expression from the Cox2 gene using a knockin reporter mouse in which luciferase activity reflects Cox2 gene expression. Using this we confirm the aorta to be essentially devoid of Cox2 driven expression. In contrast, thymus, renal medulla, and regions of the brain and gut expressed substantial levels of luciferase activity, which correlated well with COX-2-dependent prostanoid production. These data are consistent with the conclusion that COX-1 drives vascular prostacyclin release and puts the sparse expression of Cox2 in the vasculature in the context of the rest of the body. In doing so, we have identified the thymus, gut, brain and other tissues as target organs for consideration in developing a new understanding of how COX-2 protects the cardiovascular system.

Conflict of interest statement

Competing Interests: JAM and TDW have received funds from GSK and Astra Zeneca and acted as consultants and expert witnesses for Pfizer in cases where anti-inflammatory drugs, including COX-2 inhibitors have been involved. AN has received research funds and acted as consultant for projects related to supplement and drug analysis from/for Allergan, Unilever, Amarin, Equateq, GSK. This does not alter the authors' adherence to all the PLOS ONE policies on sharing data and materials.

Figures

Figure 1. 6-keto-PGF 1α production in isolated…
Figure 1. 6-keto-PGF1α production in isolated mouse aorta; measurement by enzyme immunoassay, radio immunoassay, and liquid chromatography tandem mass spectrometry (LC-MS/MS).
Prostacyclin release by isolated rings of mouse aorta stimulated with Ca2+ ionophore A23187 (50µM), measured as the stable breakdown product 6-keto-PGF1α, was not altered byCox2 gene deletion, but was reduced >10-fold byCox1 gene deletion. The pattern and level of 6-keto-PGF1α accumulation was similar whether measured by (a) enzyme immunoassay, (b) radio immunoassay or (c) LC-MS/MS. Representative LC-MS/MS chromatograms show the presence or absence of 6-keto PGF1α in all sample types (retention time 2.81 min; transition ion m/z 369>163). n=4-7. *, p<0.05 by 1-way ANOVA with Bonferonni’s post-test.
Figure 2. Bradykinin-stimulated prostanoid accumulation in the…
Figure 2. Bradykinin-stimulated prostanoid accumulation in the circulation in vivo in wild-type, Cox1 -/-, and Cox2 -/- mice.
Accumulation of the stable prostacyclin breakdown product, 6-keto-PGF1α in plasma after bradykinin administration (100nmol/kg i.v.) is dependent on COX-1 but not COX-2 when measured by LC-MS/MS (a). Representative LC-MS/MS chromatograms show the presence or absence of 6-keto PGF1α in all sample types (retention time 2.81 min; transition ion m/z369>163). Similar data were obtained for plasma levels of PGE2 (b), 13,14-dihydro-15-keto-PGE2 (c), PGD2 (d), TXB2 (e) and (f) PGF2α. Plasma 6-keto-PGF1α levels in all genotypes compare well with those previously published using enzyme immunoassay measurements. n=6. *, p<0.05 by 1-way ANOVA with Bonferonni’s post-hoc test.
Figure 3. Distribution of luciferin-dependent bioluminescence in…
Figure 3. Distribution of luciferin-dependent bioluminescence in cardiovascular tissue from Cox2 fLuc/+ mice.
(a) Quantification of basal expression from the aortic tree, vena cava, chambers of the heart and, for comparison, brain fromCox2fLuc/+ mice and (b) and representative images of bioluminescence. Arteries, veins and chambers of the heart were essentially devoid of expression from theCox2 gene, in comparison with the brain as a reference tissue. The only exception to this was weak, but detectable, expression in the region of the aortic arch. n=3.
Figure 4. Distribution of luciferin-dependent bioluminescence in…
Figure 4. Distribution of luciferin-dependent bioluminescence in tissues from Cox2 fLuc/+ mice.
(a) Basal expression from organs of theCox2fLuc/+ mice was visualized by bioluminescent imaging of tissues dissected fromCox2fLuc/+ reporter mice after injection of D-luciferin in vivo (125mg/kg i.p.). (b) Imaging data are expressed as maximum luminescent emission from each tissue. Basal Cox2 gene driven luciferase expression was present in many tissues including the vas deferens, brain, intestine, and thymus but was notably low to absent in the aorta (highlighted with red circles). Sub-division of the (c) brain, (d) intestine, (e) kidney and (f) stomach revealed regional expression patterns within each tissue. n=5.
Figure 5. COX-2-dependent prostanoid production by aorta…
Figure 5. COX-2-dependent prostanoid production by aorta versus other mouse tissues in Cox1 -/- mice.
(a) PGE2 formation, normalized to tissue mass, was measured by immunoassay in supernatants of Ca2+ ionophore A23187 (50µM)-stimulated tissue segments fromCox1-/- mice. Cox1-/- tissues released a variable amount of PGE2 with low levels in the aorta (highlighted in red), and substantially higher levels in the thymus, intestines, renal medulla, brain and vas deferens. This distribution correlates well with luciferase expression in organs of theCox2fLuc/+ mouse, as described in Figures 3 and 4. n=6.

References

    1. Grosser T, Fries S, FitzGerald GA (2006) Biological basis for the cardiovascular consequences of COX-2 inhibition: therapeutic challenges and opportunities. J Clin Invest 116: 4-15. PubMed: .
    1. Kujubu DA, Fletcher BS, Varnum BC, Lim RW, Herschman HR (1991) TIS10, a phorbol ester tumor promoter-inducible mRNA from Swiss 3T3 cells, encodes a novel prostaglandin synthase/cyclooxygenase homologue. J Biol Chem 266: 12866-12872. PubMed: .
    1. Xie WL, Chipman JG, Robertson DL, Erikson RL, Simmons DL (1991) Expression of a mitogen-responsive gene encoding prostaglandin synthase is regulated by mRNA splicing. Proc Natl Acad Sci U S A 88: 2692-2696. doi:. PubMed: .
    1. Rosen GD, Birkenmeier TM, Raz A, Holtzman MJ (1989) Identification of a cyclooxygenase-related gene and its potential role in prostaglandin formation. Biochem Biophys Res Commun 164: 1358-1365. doi:. PubMed: .
    1. Kirkby NS, Lundberg MH, Harrington LS, Leadbeater PD, Milne GL et al. (2012) Cyclooxygenase-1, not cyclooxygenase-2, is responsible for physiological production of prostacyclin in the cardiovascular system. Proc Natl Acad Sci U S A 109: 17597-17602. doi:. PubMed: .
    1. Langenbach R, Morham SG, Tiano HF, Loftin CD, Ghanayem BI et al. (1997) Disruption of the mouse cyclooxygenase 1 gene. Characteristics of the mutant and areas of future study. Adv Exp Med Biol 407: 87-92. PubMed: .
    1. Mitchell JA, Warner TD (2006) COX isoforms in the cardiovascular system: understanding the activities of non-steroidal anti-inflammatory drugs. Nat Rev Drug Discov 5: 75-86. doi:. PubMed: .
    1. Herschman HR, Reddy ST, Xie W (1997) Function and regulation of prostaglandin synthase-2. Adv Exp Med Biol 407: 61-66. PubMed: .
    1. Wallace JL, Vong L (2008) NSAID-induced gastrointestinal damage and the design of GI-sparing NSAIDs. Curr Opin Investig Drugs 9: 1151-1156. PubMed: .
    1. Bombardier C, Laine L, Reicin A, Shapiro D, Burgos-Vargas R et al. (2000) Comparison of upper gastrointestinal toxicity of rofecoxib and naproxen in patients with rheumatoid arthritis. VIGOR Study Group. N Engl J Med 343: 1520-1528, 1522 p following 1528. doi:10.1056/NEJM200011233432103. PubMed; : 11087881
    1. Rodriguez LAG, Gonzalez-Perez A, Bueno H, Hwa J (2011) NSAID Use Selectively Increases the Risk of Non-Fatal Myocardial Infarction: A Systematic Review of Randomised Trials and Observational Studies. PLOS ONE 6.
    1. Cheng Y, Wang M, Yu Y, Lawson J, Funk CD et al. (2006) Cyclooxygenases, microsomal prostaglandin E synthase-1, and cardiovascular function. J Clin Invest 116: 1391-1399. doi:. PubMed: .
    1. Fitzgerald DJ, Fitzgerald GA (2013) Historical Lessons in Translational Medicine: Cyclooxygenase Inhibition and P2Y12 Antagonism. Circ Res 112: 174-194. doi:. PubMed: .
    1. Yu Y, Ricciotti E, Scalia R, Tang SY, Grant G et al. (2012) Vascular COX-2 modulates blood pressure and thrombosis in mice. Sci Transl Med 4: 132ra154 PubMed: .
    1. Barbieri SS, Amadio P, Gianellini S, Tarantino E, Zacchi E et al. (2012) Cyclooxygenase-2-derived prostacyclin regulates arterial thrombus formation by suppressing tissue factor in a sirtuin-1-dependent-manner. Circulation 126: 1373-1384. doi:. PubMed: .
    1. Ricciotti E, Yu Y, Grosser T, Fitzgerald GA (2013) COX-2, the dominant source of prostacyclin. Proc Natl Acad Sci U S A 110: E183. doi:. PubMed: .
    1. Catella-Lawson F, McAdam B, Morrison BW, Kapoor S, Kujubu D et al. (1999) Effects of specific inhibition of cyclooxygenase-2 on sodium balance, hemodynamics, and vasoactive eicosanoids. J Pharmacol Exp Ther 289: 735-741. PubMed: .
    1. Egan KM, Lawson JA, Fries S, Koller B, Rader DJ et al. (2004) COX-2-derived prostacyclin confers atheroprotection on female mice. Science 306: 1954-1957. doi:. PubMed: .
    1. Cathcart MC, Tamosiuniene R, Chen G, Neilan TG, Bradford A et al. (2008) Cyclooxygenase-2-linked attenuation of hypoxia-induced pulmonary hypertension and intravascular thrombosis. J Pharmacol Exp Ther 326: 51-58. doi:. PubMed: .
    1. Adegboyega PA, Ololade O (2004) Immunohistochemical expression of cyclooxygenase-2 in normal kidneys. Applied immunohistochemistry & molecular morphology : AIMM / official publication of the Society for Applied Immunohistochemistry 12: 71-74
    1. Liu B, Luo W, Zhang Y, Li H, Zhu N et al. (2012) Involvement of cyclo-oxygenase-1-mediated prostacyclin synthesis in the vasoconstrictor activity evoked by ACh in mouse arteries. Exp Physiol 97: 277-289. PubMed: .
    1. Fu JY, Masferrer JL, Seibert K, Raz A, Needleman P (1990) The induction and suppression of prostaglandin H2 synthase (cyclooxygenase) in human monocytes. J Biol Chem 265: 16737-16740. PubMed: .
    1. Patrignani P, Panara MR, Greco A, Fusco O, Natoli C et al. (1994) Biochemical and pharmacological characterization of the cyclooxygenase activity of human blood prostaglandin endoperoxide synthases. J Pharmacol Exp Ther 271: 1705-1712. PubMed: .
    1. Mitchell JA, Akarasereenont P, Thiemermann C, Flower RJ, Vane JR (1993) Selectivity of nonsteroidal antiinflammatory drugs as inhibitors of constitutive and inducible cyclooxygenase. Proc Natl Acad Sci U S A 90: 11693-11697. doi:. PubMed: .
    1. Chan CC, Boyce S, Brideau C, Charleson S, Cromlish W et al. (1999) Rofecoxib [Vioxx, MK-0966; 4-(4'-methylsulfonylphenyl)-3-phenyl-2-(5H)-furanone]: a potent and orally active cyclooxygenase-2 inhibitor. Pharmacological and biochemical profiles. J Pharmacol Exp Ther 290: 551-560. PubMed: .
    1. Warner TD, Giuliano F, Vojnovic I, Bukasa A, Mitchell JA et al. (1999) Nonsteroid drug selectivities for cyclo-oxygenase-1 rather than cyclo-oxygenase-2 are associated with human gastrointestinal toxicity: a full in vitro analysis. Proc Natl Acad Sci U S A 96: 7563-7568. doi:. PubMed: .
    1. Ishikawa TO, Jain NK, Taketo MM, Herschman HR (2006) Imaging cyclooxygenase-2 (Cox-2) gene expression in living animals with a luciferase knock-in reporter gene. Mol Imaging Biol MIB Off Publ Academy Of Molecular Imaging 8: 171-187. doi:. PubMed: .
    1. Langenbach R, Morham SG, Tiano HF, Loftin CD, Ghanayem BI et al. (1995) Prostaglandin synthase 1 gene disruption in mice reduces arachidonic acid-induced inflammation and indomethacin-induced gastric ulceration. Cell 83: 483-492. doi:. PubMed: .
    1. Masoodi M, Nicolaou A (2006) Lipidomic analysis of twenty-seven prostanoids and isoprostanes by liquid chromatography/electrospray tandem mass spectrometry. Rapid Commun Mass Spectrom RCM 20: 3023-3029. doi:. PubMed: .
    1. Salmon JA (1978) A radioimmunoassay for 6-keto-prostaglandin F1alpha. Prostaglandins 15: 383-397. PubMed: .
    1. Cottee F, Flower RJ, Moncada S, Salmon JA, Vane JR (1977) Synthesis of 6-keto-PGF1alpha by ram seminal vesicle microsomes. Prostaglandins 14: 413-423. PubMed: .
    1. Kirkby NS (2012) Deletion of COX-2 augments atherosclerosis and vascular inflammation in ApoE-/-mice, independently of local prostacyclin production. British journal of pharmacology, PA2online Winter BPS meeting
    1. Yu Z, Crichton I, Tang SY, Hui Y, Ricciotti E et al. (2012) Disruption of the 5-lipoxygenase pathway attenuates atherogenesis consequent to COX-2 deletion in mice. Proc Natl Acad Sci U S A 109: 6727-6732. doi:. PubMed: .
    1. McGettigan P, Henry D (2011) Cardiovascular risk with non-steroidal anti-inflammatory drugs: systematic review of population-based controlled observational studies. PLOS Med 8: e1001098 PubMed: .
    1. Ishikawa TO, Jain N, Herschman HR (2009) Feedback regulation of cyclooxygenase-2 transcription ex vivo and in vivo. Biochem Biophys Res Commun 378: 534-538. doi:. PubMed: .
    1. Ishikawa TO, Jain NK, Herschman HR (2010) Cox-2 gene expression in chemically induced skin papillomas cannot predict subsequent tumor fate. Mol Oncol 4: 347-356. doi:. PubMed: .
    1. Cuhlmann S, Van der Heiden K, Saliba D, Tremoleda JL, Khalil M et al. (2011) Disturbed blood flow induces RelA expression via c-Jun N-terminal kinase 1: A novel mode of NF-κB regulation that promotes arterial inflammation. Circ Res 108: 950-959. doi:. PubMed: .
    1. Jongstra-Bilen J, Haidari M, Zhu SN, Chen M, Guha D et al. (2006) Low-grade chronic inflammation in regions of the normal mouse arterial intima predisposed to atherosclerosis. J Exp Med 203: 2073-2083. doi:. PubMed: .
    1. Wang D, Patel VV, Ricciotti E, Zhou R, Levin MD et al. (2009) Cardiomyocyte cyclooxygenase-2 influences cardiac rhythm and function. Proc Natl Acad Sci U S A 106: 7548-7552. doi:. PubMed: .
    1. Papanicolaou KN, Streicher JM, Ishikawa TO, Herschman H, Wang Y et al. (2010) Preserved heart function and maintained response to cardiac stresses in a genetic model of cardiomyocyte-targeted deficiency of cyclooxygenase-2. J Mol Cell Cardiol 49: 196-209. doi:. PubMed: .

Source: PubMed

Подписаться