DOI QR코드

DOI QR Code

Cytokine modulation in Raw 264.7 macrophages treated with ginseng fermented by Penibacillus MBT213

  • Son, Ji Yoon (Laboratory of Milk Food Biochemistry and Biotechnology, College of Agriculture and Life Sciences, Chungnam National University) ;
  • Renchinkhand, Gereltuya (Laboratory of Milk Food Biochemistry and Biotechnology, College of Agriculture and Life Sciences, Chungnam National University) ;
  • Bae, Hyoung Churl (Laboratory of Milk Food Biochemistry and Biotechnology, College of Agriculture and Life Sciences, Chungnam National University) ;
  • Paik, Seung-Hee (Division of Food Service Industry, Yonam College) ;
  • Lee, Jo Yoon (College of Tourism & Health, Joongbu University) ;
  • Nam, Myoung Soo (Laboratory of Milk Food Biochemistry and Biotechnology, College of Agriculture and Life Sciences, Chungnam National University)
  • Received : 2018.05.30
  • Accepted : 2018.07.24
  • Published : 2018.12.31

Abstract

The fermentation of Panax ginseng yields many compounds including ginsenosides that have various biological functions. The objective of this study was to investigate the modulation of nitric oxide (NO), Interleukin (IL)-6 and tumor necrosis factor $(TNF)-{\alpha}$ in Raw 264.7 cells treated with ginseng fermented by Penibacillus MBT213. Nitric oxide production in the Raw 264.7 cells treated for 24 hours with fermented ginseng at 3, 7, and 14 days after the treatment decreased to 74, 43, and 36%, respectively, compared with the positive control. The production of IL-6 was inhibited in all the cells treated with fermented ginseng at 3, 7, and 14 days after the treatment except for the positive control. The $TNF-{\alpha}$ production in the Raw 264.7 cells treated with fermented ginseng for 6 hours at 3, 7, and 14 days after the treatment was about 40,000, 85,000 and 65,000 pg/mL, respectively. Moreover, the $TNF-{\alpha}$ production in the Raw 264.7 cells treated with fermented ginseng for 24 hours at 7 and 14 days after the treatment was about 160,000 and 180,000 pg/mL, respectively. However, $TNF-{\alpha}$ production was inhibited in the Raw 264.7 cells at 6 and 12 hours after the treatment with fermented ginseng. herefore, it was confirmed that the immunological activity of the Raw 264.7 macrophages was affected by the treatment with fermented ginseng. It was concluded that ginseng fermented by Paenibacillus MBT213 possesses a potential anti-inflammatory activity and could be used as an ingredient in functional foods and pharmaceutical products.

Keywords

CNNSA3_2018_v45n4_769_f0001.png 이미지

Fig. 1. Effect of fermented ginseng by Penibacillus MBT213 on the cell viability water soluble tetrazolium (WST)-1 in Raw 264.7 cells for 6, 12, 24 h at 37℃. Fermented ginseng by Penibacillus MBT213 were treated to Raw 264.7 cells for 3, 7, 14 days. Each bar represents the average ± SE of three independent experiments. Lipopolysaccharide (LPS) (1 μg/mL) treatment alone served as a positive control. Level of significance was identified statistically compare with control using Duncan's multiple range test (*p < 0.05).

CNNSA3_2018_v45n4_769_f0002.png 이미지

Fig. 2. The production quantity of nitric oxide (NO) in Raw 264.7 cells by fermented ginseng by Penibacillus MBT213 for 6, 12, 24 h at 37℃ of fermented ginseng by Penibacillus MBT 213 were treated to Raw 264.7 cells for 3, 7, 14 days. Each bar represents the average ± SD of three independent experiments. Lipopolysaccharide (LPS) (1 μg/mL) treatment alone served as a positive control. Level of significance was identified statistically compare with control using Duncan's multiple range test (*p < 0.05).

CNNSA3_2018_v45n4_769_f0003.png 이미지

Fig. 3. The expression of tumor necrosis factor (TNF)-α and Iinterleukin (IL)-6 in Raw 264.7 cells by fermented ginseng by Penibacillus MBT213 for 6, 12, 24 h at 37℃. Fermented ginseng by Penibacillus MBT213 were treated to Raw 264.7 cells for 3, 7, 14 days. GAPDH, glutaldehyde-3-phosphate dehydrogenase; LPS, Lipopolysaccharide.

CNNSA3_2018_v45n4_769_f0004.png 이미지

Fig. 4. The production quantity of tumor necrosis factor (TNF)-α and Iinterleukin (IL)-6 in Raw 264.7 cells by fermented ginseng by Penibacillus MBT213 for 6, 12, 24 h at 37℃. Fermented ginseng by Penibacillus MBT 213 were treated to Raw 264.7 cells for 3, 7, 14 days. Each bar represents the average ± SD of three independent experiments. Lipopolysaccharide (LPS) (1 μg/mL) treatment alone served as a positive control. Level of significance was identified statistically compare with control using Duncan's multiple range test (*p < 0.05).

References

  1. Attele as, wu JA, Yuan CS. 1999. Ginseng pharmacology: Multiple constituents and multiple actions. Biochemical Pharmacology 58:1685-1693. https://doi.org/10.1016/S0006-2952(99)00212-9
  2. Bae EA, Han MJ, Cho MK, Park SY, Kim DH. 2002. Metabolism of 20(S) and 20(R) ginsenoside Rg3 by human intestinal bacteria and its relation to in vitro biological activities. Biological and Pharmaceutical Bulletin 25:58-63. https://doi.org/10.1248/bpb.25.58
  3. Byun SH, Yang CH, Kim SC. 2005. Inhibitory effect of Scrophulariae Radix extract on TNF$\alpha$, IL-1$\beta$, IL-6 and Nitric Oxide production in lipopolysaccharide-activated Raw 264.7 cells. Korean Journal of Herbology 20:7-16. [in Korean]
  4. Cheng LQ, Kim MK, Lee JW, Lee YJ, Yang DC. 2006. Conversion of major ginsenoside Rb(1) to ginsenoside F(2) by Caulobacter leidyia. Biotechnology Letters 28:1121-1127. https://doi.org/10.1007/s10529-006-9059-x
  5. Cheng L-Q, Na JR, Bang MH, Kim MK, Yang DC. 2008. Conversion of major ginsenoside Rb1 to 20(S)-ginsenoside Rg3 by Microbacterium sp. GS514. Phytochemistry 69:218-224. https://doi.org/10.1016/j.phytochem.2007.06.035
  6. Chi H, Ji GE. 2005. Transformation of ginsenosides Rb1 and Re from panax ginseng by food microorganisms. Biotechnology Letter 27:765-771. https://doi.org/10.1007/s10529-005-5632-y
  7. Delgado AV, McManus AT, Chambers JP. 2003. Production of tumor necrosis factor-alpha, interleukin 1-beta, interleukin 2, and interleukin 6 by rat leukocyte subpopulations after exposure to substance P. Neuropeptides 37:355-361. https://doi.org/10.1016/j.npep.2003.09.005
  8. Han BH, Park MH, Han YN, Woo LK, Sankawa U, Yahara S, Tanaka O. 1982. Degradation of ginseng saponins under mild acidic conditions. Planta Medica 44:146-149. https://doi.org/10.1055/s-2007-971425
  9. Han YM, Rhew KY. 2013. Ginsenoside Rd induces protective anti-Candida albicans antibody through immunological adjuvant activity. International Immunopharmacology 17:651-657. https://doi.org/10.1016/j.intimp.2013.08.003
  10. Hierholzer C, Harbrecht B, Menezes JM, Kane J, MacMicking J, Nathan CF, Peitzman AB, Billiar TR, Tweardy DJ. 1998. Essential role of induced nitric oxide in the initiation of the inflammatory response after hemorrhagic shock. The Journal of Experimental Medicine 187:917-28. https://doi.org/10.1084/jem.187.6.917
  11. Kim KK, Lee JW, Lee KY, Yang DC. 2005. Microbial conversion of major ginsenoside Rb(1) to pharmaceutically active minor ginsenoside Rd. Journal of Microbiology 43:456-462.
  12. Kim MW, Ko SR, Choi KJ, Kim SC. 1987. Distribution of saponin in various sections of Panax ginseng root and changes of its contents according to root age. Korean Journal of Ginseng Science 11:10-16. [in Korean]
  13. Kim BJ. 2013. Involvement of melasatin type transient receptor potential 7 channels in ginsenoside Rd-induced apoptosis in gastric and breast cancer. Journal of Ginseng Research 37:201-209. https://doi.org/10.5142/jgr.2013.37.201
  14. Kitagawa I, Taniyama T, Shibuya H, Noda T, Yoshikawa M. 1987. Chemical studies on crude drug processing. V. On the constituents of ginseng radix rubra (2): Comparison of the constituents of white ginseng and red ginseng prepared from the same Panax ginseng root. Yakugaku Zasshi 107:495-505. [in Japan] https://doi.org/10.1248/yakushi1947.107.7_495
  15. Ko SR, Choi K, Uchida K, Suzuki Y. 2003. Enzymatic preparation of ginsenosides Rg2, Rh1, and F1 from proto-panaxatriol-type ginseng saponin mixture. Planta Medica 69:285-286. https://doi.org/10.1055/s-2003-38476
  16. Lin T, Liu Y, Shi M, Liu X, Li L, Liu Y, Zhao G. 2012. Promotive effect of ginsenoside Rd on proliferation on neural stem cells in vivo and in vitro. Journal of Ethnopharmacology 142:754-761. https://doi.org/10.1016/j.jep.2012.05.057
  17. McDaniel ML, Kwon G, Hill JR, Marshall CA, Corbett JA. 1996. Cytokines and nitric oxides in islet inflammation and diabetes. Proceedings of the Society for Experimental Biology and Medicine 211:24-32. https://doi.org/10.3181/00379727-211-43950D
  18. Stuehr DJ, Marletta MA. 1985 Mammalian nitrate biosynthesis: Mouse macrophages produce nitrite and nitrate in response to Escherichia coli lipopolysaccharide. Proceedings of the National Academy of Sciences of the United States of America 82:7738-42. https://doi.org/10.1073/pnas.82.22.7738
  19. Palaniyandi SA, Damodharan K, Lee KW, Yang SH, Suh JW. 2015. Enrichment of ginsenoside Rd in Panax ginseng extract with combination of enzyme treatment and high hydrostatic pressure. Biotechnology Bioprocess Engineering 20:608-613. https://doi.org/10.1007/s12257-014-0857-z
  20. Renchinkhand G, Cho SH, Urgamal M, Park Y-W, Nam JH, Bae HC, Song GY. Nam MS. 2017. Characterization of Paenibacillus sp. MBT213 isolated from raw milk and its ability to convert ginsenoside Rb1 into ginsenoside Rd from Panax ginseng. Korean Journal for Food Science of Animal Resources 37:735-742. https://doi.org/10.5851/kosfa.2017.37.5.735
  21. Renchinkhand G, Park YW, Cho SH, Song GY, Bae HC, Choi SJ, Nam MS. 2015. Identification of $\beta$-glucosidase activity of Lactobacillus plantarum CRNB22 in Kimchi and its potential to ginsenoside Rb1 from Panax ginseng. Journal of Food Biochemistry 39:155-163. https://doi.org/10.1111/jfbc.12116
  22. Renchinkhand G, Park YW, Song GY, Cho SH, Choi SJ, Urgamal M, Bae HC, Cho, J-W, Nam MS. 2016. Identification of $\beta$-glucosidase activity of Enterococcus faecalis CRNB-A3 in Airag and its potential to convert ginsenoside Rb1 from Panax ginseng. Journal of Food Biochemistry 40:120-129. https://doi.org/10.1111/jfbc.12201
  23. Senthil K, Veena V, Mahalakshmi M, Pulla R, Yang DC, Parvatham R. 2009. Microbial conversion of major ginsenoside Rb1 to minor ginsenoside Rd by Indian fermented food bacteria. African Journal of Biotechnology 8:6961-6966.
  24. Son JW, Kim HJ, Oh DK. 2008. Ginsenoside Rd production from the major ginsenoside Rb1 by $\beta$-glucosidase from Thermus caldophilus. Biotechnology Letters 30:713-716. https://doi.org/10.1007/s10529-007-9590-4
  25. Wang L, Zhang Y, Chen J, Li S, Wang Y, Hu L, Wang L, Wu Y. 2012. Immunosuppressive effects of ginsenoside Rd on skin allograft rejection in rats. Journal of Surgery Research 176:267-274. https://doi.org/10.1016/j.jss.2011.06.038
  26. Yan Q, Zhou W, Li XW, Feng MQ, Zhou P. 2008. Purification method improvement and characterization of a novel ginsenoside-hydrolyzing $\beta$-glucosidase from Paecilomyces Bainier sp. 229. Bioscience Biotechnology Biochemistry 72:352-359. https://doi.org/10.1271/bbb.70425
  27. Yang XL, Guo TK, Wang YH, Huang YH, Liu X, Wang XX, Li W, Zhao X, Wang LP, Yan S, Wu D, Wu YJ. 2012. Ginsenoside Rd attenuates the inflammatory response via modulating p38 and JNK signaling pathways in rats with TNBS-induced relapsing colitis. International Immunopharmacology 12:408-414. https://doi.org/10.1016/j.intimp.2011.12.014