DOI QR코드

DOI QR Code

Anti-inflammatory effect of a mixture of Astragalus membranaceus and Lithospermum erythrorhizon extracts by inhibition of MAPK and NF-κB signaling pathways in RAW264.7 cells

RAW264.7 대식세포에서 MAPK 및 NF-κB 신호전달 경로 억제를 통한 황기 및 지치 복합물의 항염증 효과

  • Choi, Doo Jin (Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA) ;
  • Kim, Geum Soog (Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA) ;
  • Choi, Bo-Ram (Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA) ;
  • Lee, Young-Seob (Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA) ;
  • Han, Kyung Sook (Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA) ;
  • Lee, Dong-Sung (College of Pharmacy, Chosun University) ;
  • Lee, Dae Young (Department of Herbal Crop Research, National Institute of Horticultural and Herbal Science, RDA)
  • Received : 2020.09.28
  • Accepted : 2020.11.13
  • Published : 2020.12.31

Abstract

This study investigated a mixture of Astragalus membranaceus (AM) and Lithospermum erythrorhizon (LE) extracts (ALM16), exerts anti-inflammatory effects in lipopolysaccharide (LPS)-induced RAW264.7 macrophage cells, and its underlying mechanism. ALM16 was prepared by mixing AM and LE extracts in a ratio of 7:3 (w/w). Cytotoxicity of ALM16 in RAW264.7 cells was not shown up to 200 ㎍/mL of ALM16. The results of this study showed that ALM16 does-dependently inhibits the production of nitric oxide, prostaglandin E2 and pro-inflammatory cytokines (interleukin-1β, interleukin-6, and tumor necrosis factor-α) in LPS-induced RAW264.7 cells. ALM16 not only markedly reduced the protein expression levels of inducible nitric oxide synthase and cyclooxygenase-2 (COX-2) in LPS-stimulated RAW264.7 cells, but also inhibited the nuclear translocation and DNA-binding activity of nuclear factor-kappa B (NF-κB). In addition, ALM16 specifically inhibited the phosphorylation of c-Jun N-terminal kinase and extracellular signal-regulated kinases in LPS-stimulated RAW264.7 cells. In conclusion, these results suggest that ALM16 may exert anti-inflammatory effect by modulating mitogen-activated protein kinase and NF-κB signaling pathways.

본 연구는 황기와 지치 복합물인 ALM16이 lipopolysaccharide 처리에 의해 자극된 RAW264.7 대식세포의 염증반응에 미치는 영향에 대하여 조사하였다. ALM16은 RAW264.7 대식세포에 대하여 최대 200 ㎍/mL의 농도까지 독성은 보이지 않았다. 항염증 활성을 검정하기 위해 nitric oxide (NO), prostaglandin E2 (PGE2) 및 pro-inflammatory cytokines 생성량을 측정한 결과, ALM16은 각각의 생성량을 농도의존적으로 감소시켰다. 또한 ALM16은 NO와 PGE2 생성에 관여하는 inducible nitric oxide synthase (iNOS)와 cyclooxygenase-2 (COX-2)의 단백질 발현을 억제하였다. 한편, 항염증 활성 조절 기전을 확인하기 위하여 NK-κB의 핵으로의 이동과 DNA-binding activity 및 MAPK 신호전달 경로에 대한 ALM16의 영향을 확인한 결과, ALM16은 NF-κB의 핵으로 이동과 DNA-binding activity를 유의적으로 억제하였으며, JNK와 ERK 특이적으로 인산화를 억제함으로써 MAPK 신호전달 경로 활성을 억제하였다. 이러한 결과를 종합하여 볼 때 ALM16이 MAPK와 NF-κB의 신호전달 경로 억제를 통한 iNOS와 COX-2의 발현을 조절하고, 이로 인하여 NO, PGE2 및 pro-inflammatory cytokines의 생성이 감소하여 염증 반응을 조절하는 능력이 있는 것으로 판단된다.

Keywords

References

  1. Medzhitov R (2010) Inflammation 2010: new adventures of an old flame. Cell 140: 771-776 https://doi.org/10.1016/j.cell.2010.03.006
  2. Zhou Y, Hong Y, Huang H (2016) Triptolide attenuates inflammatory response in membranous glomerulo-nephritis rat via downregulation of NF-κB signaling pathway. Kidney Blood Press Res 41: 901-910 https://doi.org/10.1159/000452591
  3. Arulselvan P, Fard MT, Tan WS, Gothai S, Fakurazi S, Norhaizan ME, Kumar SS (2016) Role of antioxidants and natural products in inflammation. Oxid Med Cell Longev 2016: 5276130
  4. Lee EH, Park HJ, Kim BO, Choi HW, Park KI, Kang IK, Cho YJ (2020) Anti-inflammatory effect of Malus domestica cv. Green ball apple peel extract on Raw 264.7 macrophages. J Appl Biol Chem 63: 117-123 https://doi.org/10.3839/jabc.2020.016
  5. Choi DH, Cho UM, Hwang HS (2018) Anti-inflammation effect of rebaudioside A by inhibition of the MAPK and NF-κB signal pathway in RAW264.7 macrophage. J Appl Biol Chem 61: 205-211 https://doi.org/10.3839/jabc.2018.030
  6. Takeuchi O, Akira S (2010) Pattern recognition receptors and inflammation. Cell 140: 805-820 https://doi.org/10.1016/j.cell.2010.01.022
  7. Negishi H, Fujita Y, Yanai H, Sakaguchi S, Ouyang X, Shinohara M, Takayanagi H, Ohba Y, Taniguchi T, Honda K (2006) Evidence for licensing of IFN-gamma-induced IFN regulatory factor 1 transcription factor by MyD88 in Toll-like receptor-dependent gene induction program. Proc Natl Acad Sci USA 103: 15136-15141 https://doi.org/10.1073/pnas.0607181103
  8. Sharif O, Bolshakov VN, Raines S, Newham P, Perkins ND (2007) Transcriptional profiling of the LPS induced NF-kappaB response in macrophages. BMC Immunol 8: 1 https://doi.org/10.1186/1471-2172-8-1
  9. Cagiola M, Giulio S, Miriam M, Katia F, Paola P, Macri A, Pasquali P (2006) In vitro down regulation of proinflammatory cytokines induced by LPS tolerance in pig CD14+ cells. Vet Immunol Immunopathol 112: 316-320 https://doi.org/10.1016/j.vetimm.2006.04.002
  10. Yoon YK, Woo HJ, Kim Y (2015) Orostachys japonicus inhibits expression of the TLR4, NOD2, iNOS, and COX-2 genes in LPS-stimulated human PMA-differentiated THP-1 cells by inhibiting NF-κB and MAPK activation. Evid Based Complement Alternat Med 2015: 682019
  11. Kim GS, Lee DY, Lee SE, Noh HJ, Choi JH, Park CG, Choi SI, Hong SJ, Kim SY (2013) Evaluation on extraction conditions and HPLC analysis method for bioactive compounds of Astragali radix. Korean J Medicinal Crop Sci 21: 486-492 https://doi.org/10.7783/KJMCS.2013.21.6.486
  12. Shahzad M, Shabbir A, Wojcikowski K, Wohlmuth H, Gobe GC (2016) The antioxidant effects of Radix Astragali (Astragalus membranaceus and related species) in protecting tissues from injury and disease. Curr Drug Targets 17: 1331-1340 https://doi.org/10.2174/1389450116666150907104742
  13. Zhu H, Zhang Y, Ye G, Li Z, Zhou P, Huang C (2009) In vivo and in vitro antiviral activities of calycosin-7-O-β-D-glucopyranoside against coxsackie virus B3. Biol Pharm Bull 32: 68-73 https://doi.org/10.1248/bpb.32.68
  14. Chan JY, Lam F, Leung P, Che C, Fung K (2009) Antihyperglycemic and antioxidative effects of an herbal formulation of radix astragali, radix codonopsis and cortex lycii in a mouse model of type 2 diabetes mellitus. Phytother Res 23: 658-665 https://doi.org/10.1002/ptr.2694
  15. Xiao W, Han L, Shi B (2009) Isolation and purification of flavonoid glucosides from Radix Astragali by high-speed counter-current chromatography. J Chromatogr B Analyt Technol Biomed Life Sci 877: 697-702 https://doi.org/10.1016/j.jchromb.2009.01.034
  16. Zhang Y, Shi S, Guo J, You Q, Feng D (2013) On-line surface plasmon resonance-high performance liquid chromatography-tandem mass spectrometry for analysis of human serum albumin binders from Radix Astragali. J Chromatogr A 1293: 92-99 https://doi.org/10.1016/j.chroma.2013.04.015
  17. Su X, Huang Q, Chen J, Wang M, Pan H, Wang R, Zhou H, Zhou Z, Liu J, Yang F, Li T, Liu L (2016) Calycosin suppresses expression of pro-inflammatory cytokines via the activation of p62/Nrf2-linked heme oxygenase 1 in rheumatoid arthritis synovial fibroblasts. Pharmacol Res 113: 695-704 https://doi.org/10.1016/j.phrs.2016.09.031
  18. Zhang WJ, Hufnagl P, Binder BR, Wojta J (2003) Antiinflammatory activity of astragaloside IV is mediated by inhibition of NF-kappa B activation and adhesion molecule expression. Thromb Haemost 90: 904-914 https://doi.org/10.1160/TH03-03-0136
  19. Lee DY, Noh HJ, Choi J, Lee KH, Lee MH, Lee JH, Hong Y, Lee SE, Kim SY, Kim GS (2013) Anti-Inflammatory cycloartane-type saponins of Astragalus membranaceus. Molecules. 18: 3725-3732 https://doi.org/10.3390/molecules18043725
  20. Kim GS, Kim HJ, Lee DY, Choi SM, Lee SE, Noh HJ, Choi JG, Choi SI (2013) Effects of supercritical fluid extract, shikonin and acetylshikonin from Lithospermum erythrorhizon on chondrocytes and MIA-induced osteoarthritis in rats. Korean J Medicinal Crop Sci 21: 466-473 https://doi.org/10.7783/KJMCS.2013.21.6.466
  21. Papageorgiou VP, Assimopoulou AN, Couladouros EA, Hepworth D, Nicolaou KC (1999) The chemistry and biology of alkannin, shikonin, and related naphthazarin natural products. Angew Chem Int Ed Engl 38: 270-301 https://doi.org/10.1002/(SICI)1521-3773(19990201)38:3<270::AID-ANIE270>3.0.CO;2-0
  22. Gwon SY, Ahn JY, Chung CH, Moon BK, Ha TY (2012) Lithospermum erythrorhizon suppresses high-fat diet-induced obesity, and acetylshikonin, a main compound of Lithospermum erythrorhizon, inhibits adipocyte differentiation. J Agric Food Chem 60: 9089-9096 https://doi.org/10.1021/jf3017404
  23. Kim GS, Park CG, Lee KH, Choi JH, Lee SE, Noh HJ, Lee JH, Kim SY (2011) Investigation of shikonin pigments and antioxidant activity of the roots from Lithospermum erythrorhizon according to the different growth stages and areas of cultivation. Korean J Medicinal Crop Sci 19: 435-440 https://doi.org/10.7783/KJMCS.2011.19.6.435
  24. Kim JS, Han YS, Kang MH (2006) Identification of shikonin and its derivatives from Lithospermum erythrorhizon. J Korean Soc Food Sci Nutr 35: 177-181 https://doi.org/10.3746/JKFN.2006.35.2.177
  25. Chung HS, Kang M, Cho C, Park S, Kim H, Yoon YS, Kang J, Shin MK, Hong MC, Bae H (2005) Inhibition of lipopolysaccharide and interferon-gamma-induced expression of inducible nitric oxide synthase and tumor necrosis factor-alpha by Lithospermi radix in mouse peritoneal macrophages. J Ethonophamocol 102: 412-417 https://doi.org/10.1016/j.jep.2005.06.028
  26. Yoshida LS, Kakegawa T, Yuda Y, Takano-Ohmuro H (2017) Shikonin changes the lipopolysaccharide-induced expression of inflammation-related genes in macrophages. J Nat Med 71: 723-734 https://doi.org/10.1007/s11418-017-1106-5
  27. Choi DJ, Choi BR, Lee DY, Choi SI, Lee YS, Kim GS (2019) Inhibitory effect of mixed extracts obtained from Astragali Radix and Lithospermi Radix on matrix metalloproteinases in IL-1β-induced SW1353 cells and quantitative analysis of active compounds. Korean J Medicinal Crop Sci 27: 247-258 https://doi.org/10.7783/KJMCS.2019.27.4.247
  28. Choi DJ, Choi SI, Choi BR, Lee YS, Lee DY, Kim GS (2019) Cartilage protective and anti-analgesic effects of ALM16 on monosodium iodoacetate induced osteoarthritis in rats. BMC complement Altern Med 19: 325 https://doi.org/10.1186/s12906-019-2746-7
  29. Choi DJ, Kim SC, Park GE, Choi BR, Lee DY, Lee YS, Park SB, Park YI, Kim GS (2020) Protective effect of a mixture of Astragalus membranaceus and Lithospermum erythrorhizon extract against hepatic steatosis in high fat diet-induced nonalcoholic fatty liver disease mice. Evid Based Complement Alternat Med 2020: 8370698
  30. Funk CD (2001) Prostaglandins and leukotrienes: advances in eicosanoid biology. Science 294: 1871-1875 https://doi.org/10.1126/science.294.5548.1871
  31. Nathan C (1992) Nitric oxide as a secretory product of mammalian cells. FASEB J 6: 3051-3064 https://doi.org/10.1096/fasebj.6.12.1381691
  32. Hanada T, Yoshimura A (2002) Regulation of cytokine signaling and inflammation. Cytokine Growth F R 13: 413-421 https://doi.org/10.1016/S1359-6101(02)00026-6
  33. Dinarello CA (1999) Cytokines as endogenous pyrogens. J Infect Dis 179: 294-304 https://doi.org/10.1086/513856
  34. Masters SL, Simon A, Aksentijevich I, Kastner DL (2009) Horror autoinflammaticus: The molecular pathophysiology of autoinflammatory disease. Annu Rev Immunol 27: 621-668 https://doi.org/10.1146/annurev.immunol.25.022106.141627
  35. Chung YS, Choi M, Park I, Park KY, Kim KH (2010) Effects of chitosan on the production of TNF-α, IL-1β, and IL-6 in mice. Cancer Prev Res 15: 204-210
  36. Park MJ, Park HJ, Lee EH, Jung HY, Cho YJ (2018) Anti-inflammatory effect potentials of ethanol extracts from fermentated Caryopteris incana by Lactobacillus plantarum in induced to LPS with Raw 264.7 cell. J Appl Biol Chem 61: 141-150 https://doi.org/10.3839/jabc.2018.021
  37. Moncada S, Palmer RM, Higgs EA (1991) Nitric oxide: physiology, pathophysiology, and pharmacology. Pharmacol.Rev 43: 109-142
  38. Liu SF, Malik AB (2006) NF-κB activation as a pathological mechanism of septic shock and inflammation. Am J Physiol Lung Cell Mol Physiol 290: L622-L645 https://doi.org/10.1152/ajplung.00477.2005
  39. Chan AT (2003) Aspirin, non-steroidal anti-inflammatory drugs and colorectal neoplasia: Future challenges in chemoprevention. Cancer Causes Control 14: 413-418 https://doi.org/10.1023/A:1024986220526
  40. Yang EJ, Kim MS, Kim SY, Hyun CG (2019) Anti-inflammatory activity of Euphorbia jolkini extract in lipopolysaccharide-stimulated RAW264.7 cells. KSBB J 34: 120-125 https://doi.org/10.7841/ksbbj.2019.34.2.120
  41. Surh YJ, Chun KS, Cha HH, Han SS, Keum YS, Park KK, Lee SS (2001) Molecular mechanisms underlying chemopreventive activities of anti-inflammatory phytochemicals: down-regulation of COX-2 and iNOS through suppression of NF-κB activation. Mutat Res 480: 243-268
  42. Roberts PJ, Der CJ (2007) Targeting the Raf-MEK-ERK mitogen-activated protein kinase cascade for the treatment of cancer. Oncogene 26: 3291-3310 https://doi.org/10.1038/sj.onc.1210422
  43. Johnson GL, Lapadat R (2002) Mitogen-activated protein kinase pathways mediated by ERK, JNK, and p38 protein kinases. Science 298: 1911-1912 https://doi.org/10.1126/science.1072682
  44. Kim HK (2014) Role of ERK/MAPK signalling pathway in anti-inflammatory effects of Ecklonia cava in activated human mast cell line-1 cells. Asian Pac J Trop Med 7: 703-708 https://doi.org/10.1016/S1995-7645(14)60120-6
  45. Lee SC, Sim SY, Kim YS (2016) Effects of Gardeniae Fructus on the metabolic process of antioxidant and anti-inflammation by JNK and NF-κB. J Korean Med Ophthalmol Otolaryngol Dermatol 29: 56-64 https://doi.org/10.6114/jkood.2016.29.2.056