Immune Netw.  2010 Oct;10(5):145-152. 10.4110/in.2010.10.5.145.

Flowers of Inula japonica Attenuate Inflammatory Responses

Affiliations
  • 1Research and Development Division, Daegu Gyeongbuk Institute for Oriental Medicine Industry, Gyeongsan 712-210, Korea. eklee@ynu.ac.kr
  • 2College of Pharmacy, Yeungnam University, Gyongsan 712-749, Korea.
  • 3College of Medicine, Yeungnam University, Daegu 705-717, Korea.

Abstract

BACKGROUND
The flowers of Inula japonica (Inulae Flos) have long been used in traditional medicine for the treatment of inflammatory diseases. In the present study, we investigated the anti-inflammatory properties of Inulae Flos Extract (IFE).
METHODS
The anti-inflammatory effects of IFE against nitric oxide (NO), PGE2, TNF-alpha, and IL-6 release, as well as NF-kappa B and MAP kinase activation were evaluated in RAW 264.7 cells.
RESULTS
IFE inhibited the production of NO and the expression of inducible nitric oxide synthase (iNOS) in LPS-stimulated RAW264.7 cells. In addition, IFE reduced the release of pro-inflammatory cytokines, such as TNF-alpha and IL-6. Furthermore, IFE inhibited the NF-kappa B activation induced by LPS, which was associated with the abrogation of I kappa B-alpha degradation and subsequent decreases in nuclear p65 and p50 levels. Moreover, the phosphorylation of ERK, JNK, and p38 MAP kinases in LPS-stimulated RAW 264.7 cells was suppressed by IFE in a dose-dependent manner.
CONCLUSION
These results suggest that the anti-inflammation activities of IFE might be attributed to the inhibition of NO, iNOS and cytokine expression through the down-regulation of NF-kappa B activation via suppression of I kappa B alpha and MAP kinase phosphorylation in macrophages.

Keyword

Inula japonica; Inulae Flos Extract (IFE); Nitric oxide (NO); iNOS; Cytokine; NF-kappa B; MAP kinase

MeSH Terms

Cytokines
Dinoprostone
Down-Regulation
Flowers
I-kappa B Proteins
Interleukin-6
Inula
Macrophages
Medicine, Traditional
NF-kappa B
Nitric Oxide
Nitric Oxide Synthase Type II
Phosphorylation
Phosphotransferases
Tumor Necrosis Factor-alpha
Cytokines
Dinoprostone
I-kappa B Proteins
Interleukin-6
NF-kappa B
Nitric Oxide
Nitric Oxide Synthase Type II
Phosphotransferases
Tumor Necrosis Factor-alpha

Figure

  • Figure 1 Effect of IFE on LPS-induced NO production and iNOS expression in RAW 264.7 cells. Cells were pretreated with different concentrations of IFE for 1 h and then stimulated with LPS (200 ng/ml) for 24 h. The levels of nitrite were measured in the culture media by Griess reagents (A). Cells were harvested and the cell lysates were prepared as described in the experimental procedure. The protein levels of iNOS were measured by Western blot analysis using antibody against iNOS. Quantification of band intensities from three independent results was determined by densitometric analysis. The values were expressed as a percentage of maximal band intensity in the LPS-treated cells, which was set to 100% (B). Values are expressed as means±S.D. of three different samples. *p<0.01, †p<0.001, compared with the control value.

  • Figure 2 Effect of IFE on PGE2 generation and COX-2 expression in LPS-treated RAW 264.7 cells. Cells were pretreated with different concentrations of IFE for 1 h and then stimulated with LPS (200 ng/ml) for 24 h. After incubation, the PGE2 levels in the cultured media were measured using an ELISA kit (A). Cells were harvested and equal amounts of cell extracts were subjected to Western blot analysis using antibody against COX-2. Quantification of band intensities from three independent results was determined by densitometric analysis. The values were expressed as a percentage of maximal band intensity in the LPS-treated cells, which was set to 100% (B). Values are expressed as the means±S.D. of three different samples. *p<0.01, †p<0.001, when compared with the control value.

  • Figure 3 Effects of IFE on cytokine release in LPS-treated RAW 264.7 cells. Cells were pretreated with different concentrations of IFE for 1 h and then treated with LPS (200 ng/ml) for 24 h. The cytokine levels in the culture media were measured using an ELISA kit as described in the experimental procedures. Values are expressed as the means±S.D. of three independent experiments. *p<0.05, †p<0.001, when compared with the control value.

  • Figure 4 Effect of IFE on NF-κB activation in RAW 264.7 cells. Cells were pretreated with different concentrations of IFE for 1 h and then stimulated with LPS (200 ng/ml) for 30 min. Total cellular proteins and nuclear extracts were prepared for Western blot analysis of NF-κB p65 and IκBα proteins. Quantification of band intensities from three independent results was determined by densitometric analysis. The values were expressed as a percentage of maximal band intensity in the LPS-treated cells, which was set to 100% (A, B). Cells were transiently transfected with the pNF-κB-Luc plasmid using a lipofectamine method. Cells were pretreated with different concentrations of IFE for 1 h and then further stimulated with LPS (200 ng/ml) for 6 h. The cells were then harvested and the luciferase activities were determined using a Promega luciferase assay system and a luminometer (C). Data represent the mean±S.D. of three different samples. *p<0.001, when compared with the LPS-treated group.

  • Figure 5 Effect of IFE on LPS-stimulated MAPK activation in RAW 264.7 cells. Cells were pretreated with different concentrations of IFE for 1 h and then were incubated with LPS (200 ng/ml) for 30 min. Whole cell lysates were analyzed for ERK (A), JNK (B) and p38 (C) phosphorylation by Western blot analysis. Quantification of band intensities from three independent results was determined by densitometric analysis. The values were expressed as a percentage of maximal band intensity in the LPS-treated cells, which was set to 100%. Data represent the mean±S.D. of three different samples. *p<0.05, †p<0.01, ‡p<0.001, when compared with the LPS-treated group.


Reference

1. Ritchlin CT, Haas-Smith SA, Li P, Hicks DG, Schwarz EM. Mechanisms of TNF-alpha- and RANKL-mediated osteoclastogenesis and bone resorption in psoriatic arthritis. J Clin Invest. 2003. 111:821–831.
Article
2. Makarov SS. NF-kappaB as a therapeutic target in chronic inflammation: recent advances. Mol Med Today. 2000. 6:441–448.
3. Lappas M, Permezel M, Georgiou HM, Rice GE. Nuclear factor kappa B regulation of proinflammatory cytokines in human gestational tissues in vitro. Biol Reprod. 2002. 67:668–673.
Article
4. Surh YJ, Chun KS, Cha HH, Han SS, Keum YS, Park KK, Lee SS. Molecular mechanisms underlying chemopreventive activities of anti-inflammatory phytochemicals: down-regulation of COX-2 and iNOS through suppression of NF-kappa B activation. Mutat Res. 2001. 480-481:243–268.
Article
5. Yamamoto Y, Gaynor RB. Therapeutic potential of inhibition of the NF-kappaB pathway in the treatment of inflammation and cancer. J Clin Invest. 2001. 107:135–142.
Article
6. Geng HM, Zhang DQ, Zha JP, Qi JL. Simultaneous HPLC determination of five flavonoids in flos inulae. Chromatographia. 2007. 66:271–275.
7. Liu S, Liu H, Yan W, Zhang L, Bai N, Ho CT. Studies on 1-O-acetylbritannilactone and its derivative, (2-O-butyloxime-3-phenyl)-propionyl-1-O-acetylbritannilactone ester. Bioorg Med Chem Lett. 2004. 14:1101–1104.
Article
8. Grilli M, Chiu JJ, Lenardo MJ. NF-kappa B and Rel: participants in a multiform transcriptional regulatory system. Int Rev Cytol. 1993. 143:1–62.
9. Shan JJ, Yang M, Ren JW. Anti-diabetic and hypolipidemic effects of aqueous-extract from the flower of Inula japonica in alloxan-induced diabetic mice. Biol Pharm Bull. 2006. 29:455–459.
Article
10. Han M, Wen JK, Zheng B, Zhang DQ. Acetylbritannilatone suppresses NO and PGE2 synthesis in RAW 264.7 macrophages through the inhibition of iNOS and COX-2 gene expression. Life Sci. 2004. 75:675–684.
Article
11. Hernández V, del Carmen Recio M, Máñez S, Prieto JM, Giner RM, Ríos JL. A mechanistic approach to the in vivo anti-inflammatory activity of sesquiterpenoid compounds isolated from Inula viscosa. Planta Med. 2001. 67:726–731.
Article
12. Hernández V, Máñez S, Recio MC, Giner RM, Ríos JL. Anti-inflammatory profile of dehydrocostic acid, a novel sesquiterpene acid with a pharmacophoric conjugated diene. Eur J Pharm Sci. 2005. 26:162–169.
Article
13. Hernández V, Recio MC, Máñez S, Giner RM, Ríos JL. Effects of naturally occurring dihydroflavonols from Inula viscosa on inflammation and enzymes involved in the arachidonic acid metabolism. Life Sci. 2007. 81:480–488.
Article
14. Whan Han J, Gon Lee B, Kee Kim Y, Woo Yoon J, Kyoung Jin H, Hong S, Young Lee H, Ro Lee K, Woo Lee H. Ergolide, sesquiterpene lactone from Inula britannica, inhibits inducible nitric oxide synthase and cyclo-oxygenase-2 expression in RAW 264.7 macrophages through the inactivation of NF-kappaB. Br J Pharmacol. 2001. 133:503–512.
Article
15. Yu NJ, Zhao YM, Zhang YZ, Li YF. Japonicins A and B from the flowers of Inula japonica. J Asian Nat Prod Res. 2006. 8:385–390.
16. Bai N, Lai CS, He K, Zhou Z, Zhang L, Quan Z, Zhu N, Zheng QY, Pan MH, Ho CT. Sesquiterpene lactones from Inula britannica and their cytotoxic and apoptotic effects on human cancer cell lines. J Nat Prod. 2006. 69:531–535.
Article
17. Cheng PY, Lee YM, Wu YS, Chang TW, Jin JS, Yen MH. Protective effect of baicalein against endotoxic shock in rats in vivo and in vitro. Biochem Pharmacol. 2007. 73:793–804.
Article
18. Aggarwal BB, Natarajan K. Tumor necrosis factors: developments during the last decade. Eur Cytokine Netw. 1996. 7:93–124.
19. Thanos D, Maniatis T. Identification of the rel family members required for virus induction of the human beta interferon gene. Mol Cell Biol. 1995. 15:152–164.
Article
20. Kim HG, Shrestha B, Lim SY, Yoon DH, Chang WC, Shin DJ, Han SK, Park SM, Park JH, Park HI, Sung JM, Jang Y, Chung N, Hwang KC, Kim TW. Cordycepin inhibits lipopolysaccharide-induced inflammation by the suppression of NF-kappaB through Akt and p38 inhibition in RAW 264.7 macrophage cells. Eur J Pharmacol. 2006. 545:192–199.
Article
21. Choi MS, Lee SH, Cho HS, Kim Y, Yun YP, Jung HY, Jung JK, Lee BC, Pyo HB, Hong JT. Inhibitory effect of obovatol on nitric oxide production and activation of NF-kappaB/MAP kinases in lipopolysaccharide-treated RAW 264.7cells. Eur J Pharmacol. 2007. 556:181–189.
Article
22. Choi HJ, Eun JS, Park YR, Kim DK, Li R, Moon WS, Park JM, Kim HS, Cho NP, Cho SD, Soh Y. Ikarisoside A inhibits inducible nitric oxide synthase in lipopolysaccharide-stimulated RAW 264.7 cells via p38 kinase and nuclear factor-kappaB signaling pathways. Eur J Pharmacol. 2008. 601:171–178.
Article
23. Zingarelli B, Sheehan M, Wong HR. Nuclear factor-kappaB as a therapeutic target in critical care medicine. Crit Care Med. 2003. 31:1 Suppl. S105–S111.
24. Johnson GL, Lapadat R. Mitogen-activated protein kinase pathways mediated by ERK, JNK, and p38 protein kinases. Science. 2002. 298:1911–1912.
Article
25. Kim JH, Kim DH, Baek SH, Lee HJ, Kim MR, Kwon HJ, Lee CH. Rengyolone inhibits inducible nitric oxide synthase expression and nitric oxide production by down-regulation of NF-kappaB and p38 MAP kinase activity in LPS-stimulated RAW 264.7 cells. Biochem Pharmacol. 2006. 71:1198–1205.
Article
26. Suh SJ, Chung TW, Son MJ, Kim SH, Moon TC, Son KH, Kim HP, Chang HW, Kim CH. The naturally occurring biflavonoid, ochnaflavone, inhibits LPS-induced iNOS expression, which is mediated by ERK1/2 via NF-kappaB regulation, in RAW264.7 cells. Arch Biochem Biophys. 2006. 447:136–146.
Article
Full Text Links
  • IN
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr