DOI QR코드

DOI QR Code

LC-MS/MS analysis and anti-inflammatory effects of crude extract from Coptidis Rhizoma

황련 추출물의 LC-MS/MS 분석 및 항염증 효과

  • Min-Jung, Kim (College of Veterinary Medicine, Gyeongsang National University) ;
  • Ye-Jin, Yang (College of Veterinary Medicine, Gyeongsang National University) ;
  • Kwang-Youn, Kim (Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine) ;
  • Hun Hwan, Kim (College of Veterinary Medicine, Gyeongsang National University) ;
  • Jae Dong, Son (College of Veterinary Medicine, Gyeongsang National University) ;
  • Ju-Hye, Yang (Korean Medicine (KM) Application Center, Korea Institute of Oriental Medicine) ;
  • Dong bin, Lee (College of Veterinary Medicine, Gyeongsang National University) ;
  • Woo Hyun, Kim (College of Veterinary Medicine, Gyeongsang National University) ;
  • Hu-Jang, Lee (College of Veterinary Medicine, Gyeongsang National University) ;
  • Seon Been, Bak (School of Korean Medicine, Dongguk University) ;
  • Kwang-Il, Park (College of Veterinary Medicine, Gyeongsang National University)
  • 김민정 (경상국립대학교 수의과대학) ;
  • 양예진 (경상국립대학교 수의과대학) ;
  • 김광연 (한국한의학연구원 한의기술응용센터) ;
  • 김훈환 (경상국립대학교 수의과대학) ;
  • 손재동 (경상국립대학교 수의과대학) ;
  • 양주혜 (한국한의학연구원 한의기술응용센터) ;
  • 이동빈 (경상국립대학교 수의과대학) ;
  • 김우현 (경상국립대학교 수의과대학) ;
  • 이후장 (경상국립대학교 수의과대학) ;
  • 박선빈 (동국대학교 한의과대학 방제학교실) ;
  • 박광일 (경상국립대학교 수의과대학)
  • Received : 2023.01.09
  • Accepted : 2023.01.12
  • Published : 2023.02.28

Abstract

Objectives : The main aim of this study was to examine the LC-MS/MS used to identify phenolic compounds of CRE(Coptidis Rhizoma 70% EtOH Extract). Also, we investigated antioxidative activities and Anti-inflammatory activities. Methods : LC-MS/MS Analysis HPLC and LC-MS/MS were performed on a 1260 series HPLC system (Agilent Technologies, Inc., California, USA) and 3200 QTrap tandem mass system (Sciex LLC) operated in positive ion mode (spray voltage set at -4.5 kV). The solvent used was DW and Acetonitrile containing 0.1% formic acid, a gradient system was used at a flow rate of 0.5 mL/min for analysis, and a Prontosil C18 column (length, 250 mm; inner diameter, 4.6 mm; particle size, 5 ㎛; Phenomenex Co., Ltd., California, USA, Biochoff Chromatography) was used. The solvent conditions used in the mobile phases were 0-10 min at 10-15% B, 10-20 min at 20% B, 20-30 min at 25%, 30-40 min at 40%, 40-50 min at 70%, 50-60 min at 95%, and 60-70 min at 95%. The analysis was performed at a wavelength of 284 nm and a temperature of 35℃. The cell viability was measured using a 3-(4,5-dimethyethiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) assay and 1,1-diphenyl-2-picrylhydrazyl (DPPH) radical scavenging activity. We examined the effects of CRE on the lipopolysaccharide (LPS)-induced production of nitric oxide (NO) in a RAW 264.7 cells Results : The chemical analysis CRE by Liquid chromatography-tandem mass spectrometry (LC-MS/MS) confirmed that Rosmarinic acid, Ferrulic acid, 3-O-feruloylquinic acid, and 5-O-feruloylquinic acid as phenolic components. DPPH radical scavenging activity was the inhibitory activity of CRE showed at 200 ㎍/mL a statistically significant level. MTT assay demonstrated that the CRE did not have a cytotoxic effect in RAW 264.7 and LPS-induced RAW264.7 cells. Also, CRE reduced NO production in RAW 264.7 cells stimulated with LPS. Conclusions : Based on these findings, The chemical analysis 4 major components CRE such as Rosmarinic acid, Ferrulic acid, 3-O-feruloylquinic acid, and 5-O-feruloylquinic acid. Moreover, we confirmed that CRE has effects antioxidant and anti-inflammatory. The results demonstrate that CRE can be used as an antioxidant and a powerful chemopreventive ingredient against inflammatory diseases.

Keywords

Acknowledgement

This work was supported by the National Research Foundation (NRF) grant funded by the Korean government (No. 2022R1I1A3053818), the "Leaders in Industry-university Cooperation 3.0" Project, and supported by the Ministry of Education and National Research Foundation of Korea and by grant (No. 20190055) funded by the Ministry of Oceans and Fisheries, Republic of Korea.

References

  1. Hsu YY, et al. Berberine activates Nrf2 nuclear translocation and protects against oxidative damage via a phosphatidylinositol 3-kinase/Akt-dependent mechanism in NSC34 motor neuron-like cells. Eur J Pharm Sci. 2012 ; 46(5) : 415-25.  https://doi.org/10.1016/j.ejps.2012.03.004
  2. Yan B, et al. Palmatine inhibits TRIF-dependent NF-kappaB pathway against inflammation induced by LPS in goat endometrial epithelial cells. Int Immunopharmacol. 2017 ; 45 : 194-200.  https://doi.org/10.1016/j.intimp.2017.02.004
  3. Zou ZY, et al. Coptisine attenuates obesity-related inflammation through LPS/TLR-4-mediated signaling pathway in Syrian golden hamsters. Fitoterapia. 2015 ; 105 : 139-46.  https://doi.org/10.1016/j.fitote.2015.06.005
  4. Gao MY, et al. Berberine inhibits LPS-induced TF procoagulant activity and expression through NF-kappaB/p65, Akt and MAPK pathway in THP-1 cells. Pharmacol Rep. 2014; 66(3) : 480-4.  https://doi.org/10.1016/j.pharep.2013.12.004
  5. Zhao H, et al. Berberine suppresses geroconversion from cell cycle arrest to senescence. Aging. 2013 ; 5(8) :623-36.  https://doi.org/10.18632/aging.100593
  6. Yokozawa T, et al. Protective role of Coptidis Rhizoma alkaloids against peroxynitrite-induced damage to renal tubular epithelial cells. J Pharm Pharmacol. 2005 ; 57(3) : 367-74.  https://doi.org/10.1211/0022357055470
  7. Zhang Y, Liang Y, He C. Anticancer activities and mechanisms of heat-clearing and detoxicating traditional Chinese herbal medicine. Chin Med. 2017 ; 12(1) : 20. 
  8. Feng M, et al. Comparative effect of berberine and its derivative 8-cetylberberine on attenuating atherosclerosis in ApoE(-/-) mice. Int Immunopharmacol. 2017 ; 43:195-202.  https://doi.org/10.1016/j.intimp.2016.12.001
  9. Zhang DS, et al. Effect of berberine on the insulin resistance and TLR4/IKKbeta/NF-kappaB signaling pathways in skeletal muscle of obese rats with insulin resistance. J Sichuan Univ Med Sci Ed. 2015 ; 46(6) : 827-31. 
  10. Shin HS, You HK, Song KB. Effects of Rhizorna Coptidis on Cellular Activity and IL-6 Production of LPS-treated Periodontal Ligarnent Cells. Journal of Wonkwang dental Research institute. 1997 ; 7(1) : 177-91. 
  11. Tang J, Feng Y, Tsao S, Wang N, Curtain R, Wang Y. Berberine and Coptidis rhizoma as nov- el antineoplastic agents: a review of traditional use and biomedical investigations. J Ethnopharmacol. 2009 ; 126(1) : 5-17  https://doi.org/10.1016/j.jep.2009.08.009
  12. Liu F, Ng T. Antioxidative and free radical scav- enging activities of selected medicinal herbs. Life Sci. 2000 ; 66(8) : 725-35.  https://doi.org/10.1016/S0024-3205(99)00643-8
  13. Hou C, Chen L, Yang L, Ji X. An insight into anti-inflammatory effects of natural polysaccharides. Int J Biol Macromol. 2020 ; 153 : 248-255.  https://doi.org/10.1016/j.ijbiomac.2020.02.315
  14. Kim KB, Lee HT, Ku KH, Hong JW, Cho SI. Review of Pharmacological Effects of Coptidis Rhizoma and its Bioactive Compounds. Journal of Korean Traditional Medicine. 2012 ; 33(3) : 160-183. 
  15. Han S, Gao H, Chen S, Wang Q, Li X, Du LJ, Li J, Luo YY, Li JX, Zhao LC, Feng J, Yang S. Procyanidin A1 Alleviates Inflammatory Response induced by LPS through NF-κB, MAPK, and Nrf2/HO-1 Pathways in RAW264.7 cells. Sci Rep. 2019 ; 9(1) : 15087. 
  16. Jain S, Dash P, Minz AP, Satpathi S, Samal AG, Behera PK, Satpathi PS, Senapati S. Lipopolysaccharide (LPS) enhances prostate cancer metastasis potentially through NF-κB activation and recurrent dexamethasone administration fails to suppress it in vivo. Prostate. 2019 ; 79(2) : 168-182.  https://doi.org/10.1002/pros.23722
  17. Lee MH, Chao CH, Hsu YC, Lu MK. Production, characterization, and functions of sulfated polysaccharides from zinc sulfate enriched cultivation of Antrodia cinnamomea. Int J Biol Macromol. 2020 ; 159 : 1013-1021.  https://doi.org/10.1016/j.ijbiomac.2020.05.068
  18. Arulselvan P, Fard MT, Tan WS, et al. Role of Antioxidants and Natural Products in Inflammation. Oxid Med Cell Longev. 2016 ; 2016 : 5276130. 
  19. Reuter S, Gupta SC, Chaturvedi MM, Aggarwal BB. Oxidative stress, inflammation, and cancer: how are they linked?. Free Radic Biol Med. 2010 ; 49(11) : 1603-1616.  https://doi.org/10.1016/j.freeradbiomed.2010.09.006
  20. Singh N, Baby D, Rajguru JP, Patil PB, Thakkannavar SS, Pujari VB. Inflammation and cancer. Ann Afr Med. 2019 ; 18(3) :121-126.  https://doi.org/10.4103/aam.aam_56_18
  21. L. M. Coussens and Z. Werb, Inflammation and cancer. Nature, 2022 ; 420(6917) : 860-867.  https://doi.org/10.1038/nature01322
  22. S. P. Hussain, L. J. Hofseth, and C. C. Harris, "Radical causes of cancer," Nature Reviews Cancer. 2003 ; 3(4) : 276-285.  https://doi.org/10.1038/nrc1046
  23. Harmey JH, Bucana CD, Lu W, Byrne AM, McDonnell S, Lynch C, Bouchier-Hayes D, Dong Z. Lipopolysaccharide-induced metastatic growth is associated with increased angiogenesis, vascular permeability and tumor cell invasion. Int J Cancer. 2002 ; 101(5) : 415-22.  https://doi.org/10.1002/ijc.10632
  24. Lim JP, Song YC, Kim JW, Ku CH, Eun JS, Leem KH, Kim DK. Free radical scavengers from the heartwood of Juniperus chinensis. Arch Pharm Res 2022; 25: 449-452.  https://doi.org/10.1007/BF02976600
  25. Desta KT, Kim GS, Kim, YH, Lee WS, Lee SJ, Jin JS, Shin SC, The polyphenolic profiles and antioxidant effects of Agastache rugose Kuntze (Banga) flower, leaf, stem and root. Biomedical Chromatography. 2015 ; 30(2), 225-231.  https://doi.org/10.1002/bmc.3539
  26. Li W, Sun Y, Liang W, Fitzloff JF, van Breemen, R. B. Identification of caffeic acid derivatives inActea racemosa (Cimicifuga racemosa, black cohosh) by liquid chromatography/tandem mass spectrometry. Rapid Communications in Mass Spectrometry, 2003 ; 17(9) : 978-982.  https://doi.org/10.1002/rcm.1008
  27. Kose, Y. B., Iscan, G., Goger, F., Akalin, G., Demirci, B., & Baser, K. H. C. Chemical Composition and Biological Activity ofCentaurea baseri: New Species from Turkey. Chemistry & Biodiversity, 2016 ; 13(10) ; 1369-1379.  https://doi.org/10.1002/cbdv.201600070
  28. Rice-Evans C, Miller N, Paganga G. Antioxidant properties of phenolic compounds. Trends in Plant Science, 2(4), 152-159.  https://doi.org/10.1016/S1360-1385(97)01018-2
  29. Crozier, A., Jaganath, I. B., & Clifford, M. N. (2009). Dietary phenolics: chemistry, bioavailability and effects on health. Natural Product Reports, 1997 ; 26(8) : 1001-1043.  https://doi.org/10.1039/b802662a
  30. Ma JS, Kim WJ, Kim JJ, Kim TJ, Ye SK, Song MD, Kang H, Kim DW, Moon WK, Lee KH. Gold nanoparticles attenuate LPS-induced NO production through the inhibition of NF-jBand IFN-b/STAT1 pathways in RAW 264.7 cells. Nitric Oxide 2010 ; 23(3) : 241-219. 
  31. Thomas, M.J. The role of free radicals and antioxidants. Nutrition. 2000 ; 16(7-8) : 716-718.  https://doi.org/10.1016/S0899-9007(00)00343-9
  32. Mattila P, Hellstrom J. Phenolic acids in potatoes, vegetables, and some of their products. Journal of Food Composition and Analysis, 2007 ; 20(3-4) : 152-160.  https://doi.org/10.1016/j.jfca.2006.05.007
  33. Heleno SA, Martins A, Queiroz MJR, Ferreira IC. Bioactivity of phenolic acids: Metabolites versus parent compounds: A review. Food Chemistry. 2015 ; 173 : 501-513.  https://doi.org/10.1016/j.foodchem.2014.10.057
  34. Clifford MN, Jaganath IB, Ludwig IA, Crozier A. Chlorogenic acids and the acyl-quinic acids: discovery, biosynthesis, bioavailability and bioactivity. Natural Product Reports, 2017 ; 34(12) : 1391-1421.  https://doi.org/10.1039/c7np00030h
  35. K. Azuma M. Nakayama M. Koshika K. Lppoushi Y, Yamaguchi K, Kohata Y, Yamaguchi H Ito , Higashio H. Phenolic antioxidants from the leaves of Corchorus olitorius L. J Agric Food Chem, 1999 ; 47 : 3963-3966.  https://doi.org/10.1021/jf990347p
  36. Jeong JB, Hong SC, Jeong HJ, Koo JS. Anti-inflammatory effects of ethyl acetate fraction from cnidium officinale makino on LPS-stimulated RAW 264.7 and THP-1 cells. Korean J.Plant Res, 2012 ; 25 : 299-307.  https://doi.org/10.7732/kjpr.2012.25.3.299
  37. Lee CB. Anti-inflammation activity of water extracts from Hericium erinacium among medicinal mushrooms. Culinary science and hospitality research. 2012 ; 18(4) : 233-242  https://doi.org/10.20878/cshr.2012.18.5.016016016
  38. LEE DH., SOHN DS., CHO DY., KIM, BJ, LIM YY, KIM YH. Anti-inflammatory and antioxidant effects of Sophora flavescens root extraction in lipopolysaccharide-activated Raw 264.7 cells. Korean Journal of Medical Mycology, 2010 ; 15(2) : 39-50. 
  39. Yun HY, Dawson VL, Dawson TM. Neurobiology of nitric oxide. Crit Rev Neurobiol. 1996 ; 10(3-4) : 291-316.  https://doi.org/10.1615/critrevneurobiol.v10.i3-4.20
  40. Weisz A, Cicatiello L, Esumi H. Regulation of the mouse inducible-type nitric oxide synthase gene promoter by interferon-gamma, bacterial lipopoly- saccharide and NG-monomethyl-L-arginine. Biochem J. 1996 ; 316 : 209-215.  https://doi.org/10.1042/bj3160209
  41. Kubes P. Inducible nitric oxide synthase: a little bit of good in all of us. 2000 ; 47(1) : 6-9. https://doi.org/10.1136/gut.47.1.6