We use cookies to improve your experience. By continuing to browse this site, you accept our cookie policy.×

Engineering ‘Enzymelink’ for screening lead compounds to inhibit mPGES-1 while maintaining prostacyclin synthase activity

    Diana T Ruan

    Columbia University Vagelos College of Physicians & Surgeons, Columbia University Irving Medical Center, New York, NY, USA

    ,
    Nanhong Tang

    The Center for Experimental Therapeutics & Pharmacoinformatics, Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA

    Visiting Scholar from Department of Hepatobiliary Surgery & Fujian Institute of Hepatobiliary Surgery, Cancer Center of Fujian Medical University, Fujian Medical University Union Hospital, Fuzhou, China

    ,
    Hironari Akasaka

    The Center for Experimental Therapeutics & Pharmacoinformatics, Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA

    ,
    Renzhong Lu

    The Center for Experimental Therapeutics & Pharmacoinformatics, Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA

    &
    Ke-He Ruan

    *Author for correspondence: Tel.: +1 713 743 1771;

    E-mail Address: khruan@uh.edu

    The Center for Experimental Therapeutics & Pharmacoinformatics, Department of Pharmacological & Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX, USA

    Published Online:https://doi.org/10.4155/fmc-2021-0056

    Aim: This study investigated our Enzymelinks, COX-2-10aa-mPGES-1 and COX-2-10aa-PGIS, as cellular cross-screening targets for quick identification of lead compounds to inhibit inflammatory PGE2 biosynthesis while maintaining prostacyclin synthesis. Methods: We integrated virtual and wet cross-screening using Enzymelinks to rapidly identify lead compounds from a large compound library. Results: From 380,000 compounds virtually cross-screened with the Enzymelinks, 1576 compounds were identified and used for wet cross-screening using HEK293 cells that overexpressed individual Enzymelinks as targets. The top 15 lead compounds that inhibited mPGES-1 activity were identified. The top compound that specifically inhibited inflammatory PGE2 biosynthesis alone without affecting COX-2 coupled to PGI2 synthase (PGIS) for PGI2 biosynthesis was obtained. Conclusion: Enzymelink technology could advance cyclooxygenase pathway-targeted drug discovery to a significant degree.

    References

    • 1. Vane JR. Back to an aspirin a day? Science 296(5567), 474–475 (2002).
    • 2. Funk CD. Prostaglandins and leukotrienes: advances in eicosanoid biology. Science 294(5548), 1871–1875 (2001).
    • 3. Murakami M, Naraba H, Tanioka T et al. Regulation of prostaglandin E2 biosynthesis by inducible membrane-associated prostaglandin E2 synthase that acts in concert with cyclooxygenase-2. J. Biol. Chem. 275(42), 32783–32792 (2000).
    • 4. Makoto M, Nakatani Y, Tanioka T, Kudo I. Prostaglandin E synthase. Prostaglandins Other Lipid Mediat. 68-69, 383–399 (2002).
    • 5. Ruan KH. Advance in understanding the biosynthesis of prostacyclin and thromboxane A2 in the endoplasmic reticulum membrane via the cyclooxygenase pathway. Mini. Rev. Med. Chem. 4(6), 639–647 (2004).
    • 6. Dogné JM, de Leval X, Hanson J et al. New developments on thromboxane and prostacyclin modulators part I: thromboxane modulators. Curr. Med. Chem. 11(10), 1223–1241 (2004).
    • 7. Murakami PM, Nakatani Y, Tanioka T, Kudo I. Prostaglandin E synthase. Prostaglandins Other Lipid Mediat. 68-69, 383–399 (2002).
    • 8. Yamada T, Komoto J, Watanabe K et al. Crystal structure and possible catalytic mechanism of microsomal prostaglandin E synthase type 2 (mPGES-2). J. Mol. Biol. 348(5), 1163–1176 (2005).
    • 9. Hironari A, So SP, Ruan KH. Relationship of the topological distances and activities between mPGES-1 and COX-2 versus COX-1: implications of the different post-translational endoplasmic reticulum organizations of COX-1 and COX-2. Biochemistry 54(23), 3707–3715 (2015).
    • 10. Zhu L, Xu C, Huo X et al. The cyclooxygenase-1/mPGES-1/endothelial prostaglandin EP4 receptor pathway constrains myocardial ischemia-reperfusion injury. Nat. Commun. 10, 1888 (2019).
    • 11. Ruan KH, Cervantes V, So SP. Engineering of a novel hybrid enzyme: an anti-inflammatory drug target with triple catalytic activities directly converting arachidonic acid into the inflammatory prostaglandin E2. Protein Eng. Des. Sel. 22(12), 733–740 (2009).
    • 12. Ruan KH, Deng H, So SP. Engineering of a protein with cyclooxygenase and prostacyclin synthase activities that converts arachidonic acid to prostacyclin. Biochemistry 45(47), 14003–14011 (2006).
    • 13. Ruan K-H, So S-P, Cervantes V et al. An active triple-catalytic hybrid enzyme engineered by linking cyclo-oxygenase isoform-1 to prostacyclin synthase that can constantly biosynthesize prostacyclin, the vascular protector. FEBS J. 275(23), 5820–5829 (2008).
    • 14. Vane JR, Bakhle YS, Botting RM. Cyclooxygenases 1 and 2. Ann. Rev. Pharmacol. Toxicol. 38(January), 97–120; (1998).
    • 15. Ding K, Zhou Z, Hou S et al. Structure-based discovery of mPGES-1 inhibitors suitable for preclinical testing in wild-type mice as a new generation of anti-inflammatory drugs. Sci. Rep. 8, 5205 (2018).
    • 16. Lauro G, Cantone V, Potenza M et al. Discovery of 3-hydroxy-3-pyrrolin-2-one-based mPGES-1 inhibitors using a multi-step virtual screening protocol. Medchemcomm. 9(12), 2028–2036 (2018).
    • 17. Zhou S, Zhou Z, Ding K et al. DREAM-in-CDM approach and identification of a new generation of anti-inflammatory drugs targeting mPGES-1. Sci. Rep. 10, 10187 (2020).
    • 18. Ozen G, Gomez I, Daci A et al. Inhibition of microsomal PGE synthase-1 reduces human vascular tone by increasing PGI2: a safer alternative to COX-2 inhibition. Br. J. Pharmacol. 174(22), 4087–4098 (2017).
    • 19. Wang C, Chen Y, Wang Y et al. Inhibition of COX-2, mPGES-1 and CYP4A by isoliquiritigenin blocks the angiogenic Akt signaling in glioma through ceRNA effect of miR-194-5p and lncRNA NEAT1. J. Exp. Clin. Cancer Res. 38, 371 (2019).
    • 20. Kats A, Båge T, Georgsson P, Jönsson J et al. Inhibition of microsomal prostaglandin E synthase-1 by aminothiazoles decreases prostaglandin E2 synthesis in vitro and ameliorates experimental periodontitis in vivo. FASEB J. 27(6), 2328–2341 (2013).
    • 21. Francesco LD, Bruno A, Ricciotti E et al. Pharmacological characterization of the microsomal prostaglandin E2 synthase-1 inhibitor AF3485 in vitro and in vivo. Front Pharmacol. 11, 374 (2020).
    • 22. Terracciano S, Lauro G, Strocchia M et al. Structural insights for the optimization of dihydropyrimidin-2(1H)-one based mPGES-1 inhibitors. ACS Med. Chem. Lett. 6(2), 187–191 (2015).
    • 23. Ding K, Zhou Z, Zhou S et al. Design, synthesis, and discovery of 5-((1,3-diphenyl-1H-pyrazol-4-yl) methylene) pyrimidine-2,4,6(1H,3H,5H)-triones and related derivatives as novel inhibitors of mPGES-1. Bioorg. Med. Chem. Lett. 28(5), 858–862 (2018).
    • 24. Waltenberger B, Wiechmann KW, Bauer J et al. Pharmacophore modeling and virtual screening for novel acidic inhibitors of microsomal prostaglandin E2 synthase-1 (mPGES-1). J. Med. Chem. 54(9), 3163–3174 (2011).
    • 25. Frédéric M, Guiral S, Fortin L-J et al. Mark mPGES-1 inhibitor screening: an automated multistep high-throughput screening assay for the identification of lead inhibitors of the inducible enzyme mPGES-1. J. Biomolec. Screen. 10(6), 599–605 (2005).
    • 26. Orlando BJ, Lucido MJ, Malkowski MG. The structure of ibuprofen bound to cyclooxygenase-2. J. Struct. Biol. 189, 62–66 (2015).
    • 27. Li Y-C, Chiang C-W, Yeh H-C et al. Structures of prostacyclin synthase and its complexes with substrate analog and inhibitor reveal a ligand-specific heme conformation change. J. Biol. Chem. 283, 2917–2926 (2008).
    • 28. Luz JG, Antonysamy S, Kuklish SL et al. Crystal structures of mPGES-1 inhibitor complexes form a basis for the rational design of potent analgesic and anti-inflammatory therapeutics. J. Med. Chem. 58, 4727–4737 (2015).
    • 29. Joel S. Fluorine – a vital element in the medicine chest. Pharmaceuticals 26–30 (2005).
    • 30. Liggett JL, Zhang X, Eling TE, Baek SJ. Anti-tumor activity of non-steroidal anti-inflammatory drugs: cyclooxygenase-independent targets. Cancer Lett. 346(2), 217–224 (2014).
    • 31. Masako N, Rosenberg DW. Multifaceted roles of PGE2 in inflammation and cancer. Sem. Immunopathol. 35(2), 123–137 (2013).
    • 32. Masako N, Montrose DC, Clark P, et al. Genetic deletion of mPGES-1 suppresses intestinal tumorigenesis. Cancer Res. 68(9), 3251–3259 (2008).
    • 33. Masako N, Gokhale V, Meuillet EJ, Rosenberg DW. MPGES-1 as a target for cancer suppression. a comprehensive invited review ‘phospholipase A2 and lipid mediators’. Biochimie. 92, 660–664 (2010).
    • 34. Daisuke K, Murakami M, Nakatani Y et al. Potential role of microsomal prostaglandin E synthase-1 in tumorigenesis. J. Biol. Chem. 278(21), 19396–19405 (2003).