Browse > Article
http://dx.doi.org/10.1016/j.jgr.2018.06.005

Mediation of antiinflammatory effects of Rg3-enriched red ginseng extract from Korean Red Ginseng via retinoid X receptor α-peroxisome-proliferating receptor γ nuclear receptors  

Saba, Evelyn (Department of Physiology and Cell Signaling, College of Veterinary Medicine, Kyungpook National University)
Irfan, Muhammad (Department of Physiology and Cell Signaling, College of Veterinary Medicine, Kyungpook National University)
Jeong, Dahye (Department of Physiology and Cell Signaling, College of Veterinary Medicine, Kyungpook National University)
Ameer, Kashif (Division of Food Technology, Biotechnology and Agrochemistry, College of Agriculture & Life Science, Chonnam National University)
Lee, Yuan Yee (Department of Physiology and Cell Signaling, College of Veterinary Medicine, Kyungpook National University)
Park, Chae-Kyu (Laboratory of Cosmetic Research, R&D Headquarters, Korean Ginseng Cooperation)
Hong, Seung-Bok (Department of Clinical Laboratory Science, Chungbuk Health & Science University)
Rhee, Man Hee (Department of Physiology and Cell Signaling, College of Veterinary Medicine, Kyungpook National University)
Publication Information
Journal of Ginseng Research / v.43, no.3, 2019 , pp. 442-451 More about this Journal
Abstract
Background: Ginseng has a wide range of beneficial effects on health, such as the mitigation of minor and major inflammatory diseases, cancer, and cardiovascular diseases. There are abundant data regarding the health-enhancing properties of whole ginseng extracts and single ginsenosides; however, no study to date has determined the receptors that mediate the effects of ginseng extracts. In this study, for the first time, we explored whether the antiinflammatory effects of Rg3-enriched red ginseng extract (Rg3-RGE) are mediated by retinoid X receptor ${\alpha}$-peroxisome-proliferating receptor ${\gamma}$ ($RXR{\alpha}-PPAR{\gamma}$) heterodimer nuclear receptors. Methods: Nitric oxide assay, 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazoliumbromide assay, quantitative reverse transcription polymerase chain reaction, nuclear hormone receptor-binding assay, and molecular docking analyses were used for this study. Results: Rg3-RGE exerted antiinflammatory effects via nuclear receptor heterodimers between $RXR{\alpha}$ and $PPAR{\gamma}$ agonists and antagonists. Conclusion: These findings indicate that Rg3-RGE can be considered a potent antiinflammatory agent, and these effects are likely mediated by the nuclear receptor $RXR{\alpha}-PPAR{\gamma}$ heterodimer.
Keywords
Antiinflammation; Ginseng; Peroxisome-proliferating receptor ${\gamma}$; Retinoid X receptor ${\alpha}$; Rg3-enriched red ginseng extract;
Citations & Related Records
Times Cited By KSCI : 1  (Citation Analysis)
연도 인용수 순위
1 Zhang YH, Li HD, Li B, Jiang SD, Jiang LS. Ginsenoside Rg3 induces DNA damage in human osteosarcoma cells and reduces MNNG-induced DNA damage and apoptosis in normal human cells. Oncol Rep 2014;31:919-25.   DOI
2 Venteclef N, Jakobsson T, Steffensen KR, Treuter E. Metabolic nuclear receptor signaling and the inflammatory acute phase response. Trends Endocrinol Metab 2011;22:333-43.   DOI
3 Fuentes E, Guzman-Jofre L, Moore-Carrasco R, Palomo I. Role of PPARs in inflammatory processes associated with metabolic syndrome (Review). Mol Med Rep 2013;8:1611-6.   DOI
4 Inoue M, Ohtake T, Motomura W, Takahashi N, Hosoki Y, Miyoshi S, Suzuki Y, Saito H, Kohgo Y, Okumura T. Increased expression of PPARgamma in high fat diet-induced liver steatosis in mice. Biochem Biophys Res Commun 2005;336:215-22.   DOI
5 Monsalve FA, Pyarasani RD, Delgado-Lopez F, Moore-Carrasco R. Peroxisome proliferator-activated receptor targets for the treatment of metabolic diseases. Mediators Inflamm 2013;2013:549627.
6 Na SY, Kim HJ, Lee SK, Choi HS, Na DS, Lee MO, Chung M, Moore DD, Lee JW. IkappaBbeta interacts with the retinoid X receptor and inhibits retinoiddependent transactivation in lipopolysaccharide-treated cells. J Biol Chem 1998;273:3212-5.   DOI
7 Xu J, Storer PD, Chavis JA, Racke MK, Drew PD. Agonists for the peroxisome proliferator-activated receptor-alpha and the retinoid X receptor inhibit inflammatory responses of microglia. J Neurosci Res 2005;81:403-11.   DOI
8 Depoix C, Delmotte MH, Formstecher P, Lefebvre P. Control of retinoic acid receptor heterodimerization by ligand-induced structural transitions. A novel mechanism of action for retinoid antagonists. J Biol Chem 2001;276:9452-9.   DOI
9 Lefebvre P, Benomar Y, Staels B. Retinoid X receptors: common heterodimerization partners with distinct functions. Trends Endocrinol Metab 2010;21:676-83.   DOI
10 Meng XY, Zhang HX, Mezei M, Cui M. Molecular docking: a powerful approach for structure-based drug discovery. Curr Comput Aided Drug Des 2011;7:146-57.   DOI
11 Morris GM, Lim-Wilby M. Molecular docking. Methods Mol Biol 2008;443:365-82.   DOI
12 Kotani H, Tanabe H, Mizukami H, Amagaya S, Inoue M. A naturally occurring rexinoid, honokiol, can serve as a regulator of various retinoid x receptor heterodimers. Biol Pharm Bull 2012;35:1-9.   DOI
13 Cesario RM, Klausing K, Razzaghi H, Crombie D, Rungta D, Heyman RA, Lala DS. The rexinoid LG100754 is a novel RXR: PPARgamma agonist and decreases glucose levels in vivo. Mol Endocrinol 2001;15:1360-9.   DOI
14 Wang JH, Nao JF, Zhang M, He P. 20(s)-ginsenoside Rg3 promotes apoptosis in human ovarian cancer HO-8910 cells through PI3K/Akt and XIAP pathways. Tumour Biol 2014;35:11985-94.   DOI
15 Yoon SJ, Park JY, Choi S, Lee JB, Jung H, Kim TD, Yoon SR, Choi I, Shim S, Park YJ. Ginsenoside Rg3 regulates S-nitrosylation of the NLRP3 inflammasome via suppression of iNOS. Biochem Biophys Res Commun 2015;463:1184-9.   DOI
16 Luo Y, Zhang P, Zeng HQ, Lou SF, Wang DX. Ginsenoside Rg3 induces apoptosis in human multiple myeloma cells via the activation of Bcl-2-associated X protein. Mol Med Rep 2015;12:3557-62.   DOI
17 Lee YJ, Lee S, Ho JN, Byun SS, Hong SK, Lee SE, Lee E. Synergistic antitumor effect of ginsenoside Rg3 and cisplatin in cisplatinresistant bladder tumor cell line. Oncol Rep 2014;32:1803-8.   DOI
18 Nunez V, Alameda D, Rico D, Mota R, Gonzalo P, Cedenilla M, Fischer T, Bosca L, Glass CK, Arroyo AG, et al. Retinoid X receptor alpha controls innate inflammatory responses through the up-regulation of chemokine expression. Proc Natl Acad Sci U S A 2010;107:10626-31.   DOI
19 Sato Y, Ramalanjaona N, Huet T, Potier N, Osz J, Antony P, Peluso-Iltis C, Poussin- Courmontagne P, Ennifar E, Mely Y, et al. The "Phantom Effect" of the Rexinoid LG100754: structural and functional insights. PLoS One 2010;5:e15119.   DOI
20 Uchimura K, Nakamuta M, Enjoji M, Irie T, Sugimoto R, Muta T, Iwamoto H, Nawata H. Activation of retinoic X receptor and peroxisome proliferatoractivated receptor-gamma inhibits nitric oxide and tumor necrosis factoralpha production in rat Kupffer cells. Hepatology 2001;33:91-9.   DOI
21 Roszer T, Menendez-Gutierrez MP, Cedenilla M, Ricote M. Retinoid X receptors in macrophage biology. Trends Endocrinol Metab 2013;24:460-8.   DOI
22 Claudel T, Leibowitz MD, Fievet C, Tailleux A, Wagner B, Repa JJ, Torpier G, Lobaccaro JM, Paterniti JR, Mangelsdorf DJ, et al. Reduction of atherosclerosis in apolipoprotein E knockout mice by activation of the retinoid X receptor. Proc Natl Acad Sci U S A 2001;98:2610-5.   DOI
23 Ma F, Liu SY, Razani B, Arora N, Li B, Kagechika H, Tontonoz P, Nunez V, Ricote M, Cheng G. Retinoid X receptor alpha attenuates host antiviral response by suppressing type I interferon. Nat Commun 2014;5:5494.   DOI
24 Uchida H, Hasegawa Y, Takahashi H, Makishima M. 1alpha-dihydroxyvitamin D3 and retinoic acid increase nuclear vitamin D receptor expression in monocytic THP-1 cells. Anticancer Res 2016;36:6297-301.   DOI
25 Mangelsdorf DJ, Borgmeyer U, Heyman RA, Zhou JY, Ong ES, Oro AE, Kakizuka A, Evans RM. Characterization of three RXR genes that mediate the action of 9-cis retinoic acid. Genes Dev 1992;6:329-44.   DOI
26 Budhu AS, Noy N. Direct channeling of retinoic acid between cellular retinoic acid-binding protein II and retinoic acid receptor sensitizes mammary carcinoma cells to retinoic acid-induced growth arrest. Mol Cell Biol 2002;22:2632-41.   DOI
27 Wang CZ, Anderson S, Du W, He TC, Yuan CS. Red ginseng and cancer treatment. Chin J Nat Med 2016;14:7-16.   DOI
28 Serhan CN. Resolution phase of inflammation: novel endogenous antiinflammatory and proresolving lipid mediators and pathways. Annu Rev Immunol 2007;25:101-37.   DOI
29 Lee HN, Na HK, Surh YJ. Resolution of inflammation as a novel chemopreventive strategy. Semin Immunopathol 2013;35:151-61.   DOI
30 Shergis JL, Zhang AL, Zhou W, Xue CC. Panax ginseng in randomised controlled trials: a systematic review. Phytother Res 2013;27:949-65.   DOI
31 Nag SA, Qin JJ, Wang W, Wang MH, Wang H, Zhang R. Ginsenosides as anticancer agents: in vitro and in vivo activities, structure-activity relationships, and molecular mechanisms of action. Front Pharmacol 2012;3:25.   DOI
32 Im DS, Nah SY. Yin and Yang of ginseng pharmacology: ginsenosides vs gintonin. Acta Pharmacol Sin 2013;34:1367-73.   DOI
33 Jeong D, Irfan M, Kim SD, Kim S, Oh JH, Park CK, Kim HK, Rhee MH. Ginsenoside Rg3-enriched red ginseng extract inhibits platelet activation and in vivo thrombus formation. J Ginseng Res 2017;41:548-55.   DOI
34 Irwin JJ, Sterling T, Mysinger MM, Bolstad ES, Coleman RG. ZINC: a free tool to discover chemistry for biology. J Chem Inf Model 2012;52:1757-68.   DOI
35 Mangelsdorf DJ, Ong ES, Dyck JA, Evans RM. Nuclear receptor that identifies a novel retinoic acid response pathway. Nature 1990;345:224-9.   DOI
36 Perez E, Bourguet W, Gronemeyer H, de Lera AR. Modulation of RXR function through ligand design. Biochim Biophys Acta 1821;2012:57-69.
37 Perlmann T, Jansson L. A novel pathway for vitamin A signaling mediated by RXR heterodimerization with NGFI-B and NURR1. Genes Dev 1995;9:769-82.   DOI
38 Zhang-Gandhi CX, Drew PD. Liver X receptor and retinoid X receptor agonists inhibit inflammatory responses of microglia and astrocytes. J Neuroimmunol 2007;183:50-9.   DOI
39 Evans RM, Mangelsdorf DJ. Nuclear receptors, RXR, and the Big Bang. Cell 2014;157:255-66.   DOI
40 Castrillo A, Joseph SB, Marathe C, Mangelsdorf DJ, Tontonoz P. Liver X receptor-dependent repression of matrix metalloproteinase-9 expression in macrophages. J Biol Chem 2003;278:10443-9.   DOI
41 Terasaka N, Hiroshima A, Ariga A, Honzumi S, Koieyama T, Inaba T, Fujiwara T. Liver X receptor agonists inhibit tissue factor expression in macrophages. FEBS J 2005;272:1546-56.   DOI
42 Son YL, Park OG, Kim GS, Lee JW, Lee YC. RXR heterodimerization allosterically activates LXR binding to the second NR box of activating signal co-integrator-2. Biochem J 2008;410:319-30.   DOI
43 Makoukji J, Shackleford G, Meffre D, Grenier J, Liere P, Lobaccaro JM, Schumacher M, Massaad C. Interplay between LXR and $Wnt/{\beta}$eta-catenin signaling in the negative regulation of peripheral myelin genes by oxysterols. J Neurosci 2011;31:9620-9.   DOI
44 Joseph SB, Castrillo A, Laffitte BA, Mangelsdorf DJ, Tontonoz P. Reciprocal regulation of inflammation and lipid metabolism by liver X receptors. Nat Med 2003;9:213-9.   DOI
45 Joseph SB, Bradley MN, Castrillo A, Bruhn KW, Mak PA, Pei L, Hogenesch J, O'Connell RM, Cheng G, Saez E, et al. LXR-dependent gene expression is important for macrophage survival and the innate immune response. Cell 2004;119:299-309.   DOI
46 Park JY, Kawada T, Han IS, Kim BS, Goto T, Takahashi N, Fushiki T, Kurata T, Yu R. Capsaicin inhibits the production of tumor necrosis factor alpha by LPSstimulated murine macrophages, RAW 264.7: a PPARgamma ligand-like action as a novel mechanism. FEBS Lett 2004;572:266-70.   DOI
47 Yu XH, Zheng XL, Tang CK. Peroxisome proliferator-activated receptor alpha in lipid metabolism and atherosclerosis. Adv Clin Chem 2015;71:171-203.   DOI