Planta Med 2019; 85(06): 453-464
DOI: 10.1055/a-0835-2301
Biological and Pharmacological Activity
Original Papers
Georg Thieme Verlag KG Stuttgart · New York

Metabolism of Scoparone in Experimental Animals and Humans

Risto O. Juvonen
1   School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
,
Filip Novák
2   Department of Biochemical Sciences, Faculty of Pharmacy, Charles University, Hradec Králové, Czech Republic
,
Eleni Emmanouilidou
3   School of Pharmacy, Faculty of Health Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece
,
Seppo Auriola
1   School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
,
Juri Timonen
1   School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
,
Aki T. Heikkinen
4   Admescope Ltd, Oulu, Finland
,
Jenni Küblbeck
1   School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
,
Moshe Finel
5   Division of Pharmaceutical Chemistry and Technology, Faculty of Pharmacy, University of Helsinki, University of Helsinki, Finland
,
Hannu Raunio
1   School of Pharmacy, Faculty of Health Sciences, University of Eastern Finland, Kuopio, Finland
› Author Affiliations
Further Information

Publication History

received 09 November 2018
revised 02 January 2019

accepted 16 January 2019

Publication Date:
08 February 2019 (online)

Abstract

Scoparone, a major constituent of the Chinese herbal medicine Yin Chen Hao, expresses beneficial effects in experimental models of various diseases. The intrinsic doses and effects of scoparone are dependent on its metabolism, both in humans and animals. We evaluated in detail the metabolism of scoparone in human, mouse, rat, pig, dog, and rabbit liver microsomes in vitro and in humans in vivo. Oxidation of scoparone to isoscopoletin via 6-O-demethylation was the major metabolic pathway in liver microsomes from humans, mouse, rat, pig and dog, whereas 7-O-demethylation to scopoletin was the main reaction in rabbit. The scoparone oxidation rates in liver microsomes were 0.8 – 1.2 µmol/(min*g protein) in mouse, pig, and rabbit, 0.2 – 0.4 µmol/(min*g protein) in man and dog, and less than 0.1 µmol/(min*g) in rat. In liver microsomes of all species, isoscopoletin was oxidized to 3-[4-methoxy-ρ-(3, 6)-benzoquinone]-2-propenoate and esculetin, which was formed also in the oxidation of scopoletin. Human CYP2A13 exhibited the highest rate of isoscopoletin and scopoletin oxidation, followed by CYP1A1 and CYP1A2. Glucuronidation of isoscopoletin and scopoletin was catalyzed by the human UGT1A1, UGT1A6, UGT1A7, UGT1A8, UGT1A9, UGT1A10, and UGT2B17. Dog was most similar to man in scoparone metabolism. Isoscopoletin glucuronide and sulfate conjugates were the major scoparone in vivo metabolites in humans, and they were completely excreted within 24 h in urine. Scoparone and its metabolites did not activate key nuclear receptors regulating CYP and UGT enzymes. These results outline comprehensively the metabolic pathways of scoparone in man and key preclinical animal species.

Supporting Information

 
  • References

  • 1 Hung HY, Kuo SC. Recent studies and progression of Yin Chen Hao (茵陳蒿 Yīn Chén Hāo), a long-term used traditional Chinese medicine. J Tradit Complement Med 2013; 3: 2-6
  • 2 Jang E, Kim BJ, Lee KT, Inn KS, Lee JH. A survey of therapeutic effects of Artemisia capillaris in liver diseases. Evid Based Complement Alternat Med 2015; 2015: 728137
  • 3 Li JY, Cao HY, Sun L, Sun RF, Wu C, Bian YQ, Dong S, Liu P, Sun MY. Therapeutic mechanism of Yīn-Chén-Hāo decoction in hepatic diseases. World J Gastroenterol 2017; 23: 1125-1138
  • 4 Wang Y, Wang M, Chen B, Shi J. Scoparone attenuates high glucose-induced extracellular matrix accumulation in rat mesangial cells. Eur J Pharmacol 2017; 815: 376-380
  • 5 Yamahara J, Kobayashi G, Matsuda H, Katayama T, Fujimura H. The effect of scoparone, a coumarin derivative isolated from the Chinese crude drug Artemisiae capillaris flos, on the heart. Chem Pharm Bull (Tokyo) 1989; 37: 1297-1299
  • 6 Choi BR, Kim HK, Park JK. Penile erection induced by scoparone from Artemisia capillaris through the nitric oxide-cyclic guanosine monophosphate signaling pathway. World J Mens Health 2017; 35: 196-204
  • 7 Jang SI, Kim YJ, Lee WY, Kwak KC, Baek SH, Kwak GB, Yun YG, Kwon TO, Chung HT, Chai KY. Scoparone from Artemisia capillaris inhibits the release of inflammatory mediators in RAW 264.7 cells upon stimulation cells by interferon-gamma Plus LPS. Arch Pharm Res 2005; 28: 203-208
  • 8 Kim EK, Kwon KB, Lee JH, Park BH, Park JW, Lee HK, Jhee EC, Yang JY. Inhibition of cytokine-mediated nitric oxide synthase expression in rat insulinoma cells by scoparone. Biol Pharm Bull 2007; 30: 242-246
  • 9 Kim JK, Kim JY, Kim HJ, Park KG, Harris RA, Cho WJ, Lee JT, Lee IK. Scoparone exerts anti-tumor activity against DU145 prostate cancer cells via inhibition of STAT3 activity. PLoS One 2013; 8: e80391
  • 10 Park S, Kim JK, Oh CJ, Choi SH, Jeon JH, Lee IK. Scoparone interferes with STAT3-induced proliferation of vascular smooth muscle cells. Exp Mol Med 2015; 47: e145
  • 11 Cho DY, Ko HM, Kim J, Kim BW, Yun YS, Park JI, Ganesan P, Lee JT, Choi DK. Scoparone inhibits LPS-simulated inflammatory response by suppressing IRF3 and ERK in BV-2 microglial cells. Molecules 2016; 21: E1718
  • 12 Xu M, Cai J, Wei H, Zhou M, Xu P, Huang H, Peng W, Du F, Gong A, Zhang Y. Scoparone protects against pancreatic fibrosis via TGF-β/Smad signaling in rats. Cell Physiol Biochem 2016; 40: 277-286
  • 13 Zhang A, Sun H, Wu G, Sun W, Yuan Y, Wang X. Proteomics analysis of hepatoprotective effects for scoparone using MALDI-TOF/TOF mass spectrometry with bioinformatics. OMICS 2013; 17: 224-229
  • 14 Zhang A, Qiu S, Sun H, Zhang T, Guan Y, Han Y, Yan G, Wang X. Scoparone affects lipid metabolism in primary hepatocytes using lipidomics. Sci Rep 2016; 6: 28031
  • 15 Liu X, Zhao X. Scoparone attenuates hepatic stellate cell activation through inhibiting TGF-β/Smad signaling pathway. Biomed Pharmacother 2017; 93: 57-61
  • 16 Huang W, Zhang J, Moore DD. A traditional herbal medicine enhances bilirubin clearance by activating the nuclear receptor CAR. J Clin Invest 2004; 113: 137-143
  • 17 Masuyama H, Mitsui T, Maki J, Tani K, Nakamura K, Hiramatsu Y. Dimethylesculetin ameliorates maternal glucose intolerance and fetal overgrowth in high-fat diet-fed pregnant mice via constitutive androstane receptor. Mol Cell Biochem 2016; 419: 185-192
  • 18 Mehta P, Shah R, Lohidasan S, Mahadik KR. Pharmacokinetic profile of phytoconstituent(s) isolated from medicinal plants – a comprehensive review. J Tradit Complement Med 2015; 5: 207-227
  • 19 Shi P, Lin X, Yao H. A comprehensive review of recent studies on pharmacokinetics of traditional Chinese medicines (2014–2017) and perspectives. Drug Metab Rev 2018; 50: 161-192
  • 20 Reichard JF, Maier MA, Naumann BD, Pecquet AM, Pfister T, Sandhu R, Sargent EV, Streeter AJ, Weideman PA. Toxicokinetic and toxicodynamic considerations when deriving health-based exposure limits for pharmaceuticals. Regul Toxicol Pharmacol 2016; 79 (Suppl. 01) S67-S78
  • 21 Yin Q, Sun H, Zhang A, Wang X. Pharmacokinetics and tissue distribution study of scoparone in rats by ultraperformance liquid-chromatography with tandem high-definition mass spectrometry. Fitoterapia 2012; 83: 795-800
  • 22 Yang D, Yang J, Shi D, Deng R, Yan B. Scoparone potentiates transactivation of the bile salt export pump gene and this effect is enhanced by cytochrome P450 metabolism but abolished by a PKC inhibitor. Br J Pharmacol 2011; 164: 1547-1557
  • 23 Mennes WC, Van Holsteijn CW, Timmerman A, Noordhoek J, Blaauboer BJ. Biotransformation of scoparone used to monitor changes in cytochrome P450 activities in primary hepatocyte cultures derived from rats, hamsters and monkeys. Biochem Pharmacol 1991; 41: 1203-1208
  • 24 Horsmans Y, Desager JP, Harvengt C. Scoparone O-demethylase assay is not useful to differentiate the effects of model inducers of cytochrome P-450 in rabbit and guinea pig liver. Life Sci 1993; 53: PL421-PL426
  • 25 Witkamp RF, Nijmeijer SM, Mennes WC, Rozema AW, Noordhoek J, Van Miert AS. Regioselective O-demethylation of scoparone (6,7-dimethoxycoumarin) to assess cytochrome P450 activities in vitro in rat. Effects of gonadal steroids and the involvement of constitutive P450 enzymes. Xenobiotica 1993; 23: 401-410
  • 26 Wang X, Lv H, Sun H, Liu L, Sun W, Cao H. Development of a rapid and validated method for investigating the metabolism of scoparone in rat using ultra-performance liquid chromatography/electrospray ionization quadruple time-of-flight mass spectrometry. Rapid Commun Mass Spectrom 2007; 21: 3883-3890
  • 27 Meyer RP, Hagemeyer CE, Knoth R, Kurz G, Volk B. Oxidative hydrolysis of scoparone by cytochrome P450 CYP2C29 reveals a novel metabolite. Biochem Biophys Res Commun 2001; 285: 32-39
  • 28 Fayyaz A, Makwinja S, Auriola S, Raunio H, Juvonen RO. Comparison of in vitro hepatic scoparone 7-O-demethylation between humans and experimental animals. Planta Med 2018; 84: 320-328
  • 29 Luukkanen L, Taskinen J, Kurkela M, Kostiainen R, Hirvonen J, Finel M. Kinetic characterization of the 1A subfamily of recombinant human UDP-glucuronosyltransferases. Drug Metab Dispos 2005; 33: 1017-1026
  • 30 Rautio A, Kraul H, Kojo A, Salmela E, Pelkonen O. Interindividual variability of coumarin 7-hydroxylaton in healthy volunteers. Pharmacogenetics 1992; 2: 227-233
  • 31 Kaipainen P, Koivusaaari U, Lang M. Catalytic and immunologic comparison of coumarin 7-hydroxylation in different species. Comp Biochem Physiol 1985; 81C: 293-296
  • 32 Raunio H, Rahnasto-Rilla M. CYP2A6: genetics, structure, regulation, and function. Drug Metabol Drug Interact 2012; 27: 73-88
  • 33 Heikkinen AT, Friedlein A, Matondo M, Hatley OJ, Petsalo A, Juvonen R, Galetin A, Rostami-Hodjegan A, Aebersold R, Lamerz J, Dunkley T, Cutler P, Parrott N. Quantitative ADME proteomics – CYP and UGT enzymes in the Beagle dog liver and intestine. Pharm Res 2015; 32: 74-90
  • 34 Kurkela M, Patana AS, Mackenzie PI, Court MH, Tate CG, Hirvonen J, Goldman A, Finel M. Interactions with other human UDP-glucuronosyltransferases attenuate the consequences of the Y485D mutation on the activity and substrate affinity of UGT1A6. Pharmacogenet Genomics 2007; 17: 115-126
  • 35 Kuuranne T, Aitio O, Vahermo M, Elovaara E, Kostiainen R. Enzyme-assisted synthesis and structure characterization of glucuronide conjugates of methyltestosterone (17 alpha-methylandrost-4-en-17 beta-ol-3-one) and nandrolone (estr-4-en-17 beta-ol-3-one) metabolites. Bioconjug Chem 2002; 13: 194-199
  • 36 Sneitz N, Court MH, Zhang X, Laajanen K, Yee KK, Dalton P, Ding X, Finel M. Human UDP-glucuronosyltransferase UGT2A2: cDNA construction, expression, and functional characterization in comparison with UGT2A1 and UGT2A3. Pharmacogenet Genomics 2009; 19: 923-934
  • 37 Kurkela M, Siiskonen A, Finel M, Tammela P, Taskinen J, Vuorela P. Microplate screening assay to identify inhibitors of human catechol-O-methyltransferase. Anal Biochem 2004; 331: 198-200
  • 38 Küblbeck J, Laitinen T, Jyrkkärinne J, Rousu T, Tolonen A, Abel T, Kortelainen T, Uusitalo J, Korjamo T, Honkakoski P, Molnár F. Use of comprehensive screening methods to detect selective human CAR activators. Biochem Pharmacol 2011; 82: 1994-2007