Korean J Physiol Pharmacol.  2013 Jun;17(3):217-222. 10.4196/kjpp.2013.17.3.217.

2-(4-Hydroxyphenyl)-5-(3-Hydroxypropenyl)-7-Methoxybenzofuran, a Novel Ailanthoidol Derivative, Exerts Anti-Inflammatory Effect through Downregulation of Mitogen-Activated Protein Kinase in Lipopolysaccharide-Treated RAW 264.7 Cells

Affiliations
  • 1Department of Biomedical Science, College of Natural Science, Catholic University of Daegu, Gyeongsan 700-712, Korea. toto0818@cu.ac.kr
  • 2Department of Chemistry and Institute of Natural Medicine, Hallym University, Chuncheon 200-702, Korea.

Abstract

We reported that ailanthoidol, a neolignan from Zanthoxylum ailanthoides and Salvia miltiorrhiza Bunge, inhibited inflammatory reactions by macrophages and protected mice from endotoxin shock. We examined the anti-inflammatory activity of six synthetic ailanthoidol derivatives (compounds 1-6). Among them, compound 4, 2-(4-hydroxyphenyl)-5-(3-hydroxypropenyl)-7-methoxybenzofuran, had the lowest IC50 value concerning nitric oxide (NO) release from lipopolysaccharide (LPS)-stimulated RAW264.7 cells. Compound 4 suppressed the generation of prostaglandin (PG) E2 and the expression of inducible NO synthase and cyclooxygenase (COX)-2 induced by LPS, and inhibited the release of LPS-induced pro-inflammatory cytokines from RAW264.7 cells. The underlying mechanism of compound 4 on anti-inflammatory action was correlated with the down-regulation of mitogen-activated protein kinase and activator protein-1 activation. Compound 4 is potentially an effective functional chemical candidate for the prevention of inflammatory diseases.

Keyword

Ailanthoidol derivatives; AP-1; Cytokines; Inflammation; Macrophage

MeSH Terms

Animals
Benzofurans
Cytokines
Down-Regulation
Inflammation
Inhibitory Concentration 50
Macrophages
Mice
Nitric Oxide
Nitric Oxide Synthase
Prostaglandin-Endoperoxide Synthases
Protein Kinases
Salvia miltiorrhiza
Shock
Transcription Factor AP-1
Zanthoxylum
Benzofurans
Cytokines
Nitric Oxide
Nitric Oxide Synthase
Prostaglandin-Endoperoxide Synthases
Protein Kinases
Transcription Factor AP-1

Figure

  • Fig. 1 Chemical structure of ailanthoidol and its derivatives, compounds 1-6.

  • Fig. 2 Effects of compound 4 on murine macrophage viability. RAW264.7 cells were treated with indicated concentrations of compound 4 for 24 h, and proliferation was determined. The results are reported as mean±SEM of three independent experiments in triplicate.

  • Fig. 3 Effects of compound 4 on LPS-induced NO and PGE2 release. RAW264.7 cells were treated with 0~10 µM of compound in the presence of 100 ng/ml of LPS or with LPS alone for 24 h, and (A) NO and (B) PGE2 release was determined. The results are reported as mean±SEM of three independent experiments in triplicate. Statistical significance is based on the difference when compared with LPS-stimulated cells (**p<0.01, ***p<0.001). Thirty micrograms of protein obtained from each cell lysate was resolved by 10% SDS-PAGE for (C) iNOS and (D) COX-2 determination. β-actin expression is shown as a loading control. The bands were quantified using image analysis software and their relative intensity was expressed as fold against the image of the LPS-stimulated RAW264.7 cells.

  • Fig. 4 Effects of compound 4 on LPS-induced inflammatory cytokine production in murine macrophages. RAW264.7 cells were treated with 0~10 µM of compound 4 in the presence of 100 ng/ml LPS or with LPS alone for 24 h. The cell culture media were then collected, and the amount of (A) IL-1β and (B) IL-6 released was measured. The results are reported as mean±SEM of three independent experiments in triplicate. Statistical significance is based on the difference when compared with LPS-stimulated cells (**p<0.01, ***p<0.001).

  • Fig. 5 Effect of compound 4 on LPS-induced MAPK and AP-1 activation. RAW264.7 cells were plated in 100 mm-diameter dishes. After 12 h of seeding, cells were treated with different doses of compound 4 for 1 h, followed by stimulation with 500 ng/ml of LPS for 30 min. Cell lysates (30 µg protein) were prepared and subjected to Western blot analysis by using antibodies specific for (A) IκB-α, phosphorylated forms of ERK1/2, JNK, p38 MAPK and (B) phosphorylated forms of c-Jun. Equivalent loading of cell lysates was determined by reprobing the blots with anti-β-actin, total ERK1/2, JNK or p38 MAPK antibody. (C) Nuclear protein (30 µg protein) was prepared and subjected to Western blot analysis by using antibodies specific for c-Jun and equivalent loading of nuclear protein was determined by reprobing the blots with anti-PARP. The bands were quantified using image analysis software and their relative intensity was expressed as fold against the image of the LPS-stimulated RAW264.7 cells.


Reference

1. Aller MA, Arias N, Fuentes-Julian S, Blazquez-Martinez A, Argudo S, Miguel MP, Arias JL, Arias J. Coupling inflammation with evo-devo. Med Hypotheses. 2012; 78:721–731. PMID: 22405850.
Article
2. Biasi F, Astegiano M, Maina M, Leonarduzzi G, Poli G. Polyphenol supplementation as a complementary medicinal approach to treating inflammatory bowel disease. Curr Med Chem. 2011; 18:4851–4865. PMID: 21919842.
Article
3. Kim JK, Jun JG. Ailanthoidol suppresses lipopolysaccharidestimulated inflammatory reactions in RAW264.7 cells and endotoxin shock in mice. J Cell Biochem. 2011; 112:3816–3823. PMID: 21826708.
Article
4. Lee YJ, Kao ES, Chu CY, Lin WL, Chiou YH, Tseng TH. Inhibitory effect of ailanthoidol on 12-O-tetradecanoyl-phorbol-13-acetate-induced tumor promotion in mouse skin. Oncol Rep. 2006; 16:921–927. PMID: 16969515.
Article
5. Lee WS, Baek YI, Kim JR, Cho KH, Sok DE, Jeong TS. Antioxidant activities of a new lignan and a neolignan from Saururus chinensis. Bioorg Med Chem Lett. 2004; 14:5623–5628. PMID: 15482936.
Article
6. Abdel-Wahab BF, Abdel-Aziz HA, Ahmed EM. Synthesis and antimicrobial evaluation of 1-(benzofuran-2-yl)-4-nitro-3-arylbutan-1-ones and 3-(benzofuran-2-yl)-4,5-dihydro-5-aryl-1-[4-(aryl)-1,3-thiazol-2-yl]-1H-pyrazoles. Eur J Med Chem. 2009; 44:2632–2635. PMID: 18995932.
Article
7. Lin SY, Chen CL, Lee YJ. Total synthesis of ailanthoidol and precursor XH14 by Stille coupling. J Org Chem. 2003; 68:2968–2971. PMID: 12662079.
Article
8. Rao MLN, Awasthi DK, Banerjee D. An expeditious and convergent synthesis of ailanthoidol. Tetrahedron Lett. 2010; 51:1979–1981.
Article
9. Chen GY, Nuñez G. Sterile inflammation: sensing and reacting to damage. Nat Rev Immunol. 2010; 10:826–837. PMID: 21088683.
Article
10. Martinez FO, Helming L, Gordon S. Alternative activation of macrophages: an immunologic functional perspective. Annu Rev Immunol. 2009; 27:451–483. PMID: 19105661.
Article
11. Natoli G, Ghisletti S, Barozzi I. The genomic landscapes of inflammation. Genes Dev. 2011; 25:101–106. PMID: 21245163.
Article
12. Lee LR, Lee JJ, Kim , Jun JG. Ailanthoidol derivatives and their anti-inflammatory effects. Bull Korean Chem Soc. 2012; 33:1907–1912.
Article
13. Chen CY, Peng WH, Tsai KD, Hsu SL. Luteolin suppresses inflammation-associated gene expression by blocking NF-kappaB and AP-1 activation pathway in mouse alveolar macrophages. Life Sci. 2007; 81:1602–1614. PMID: 17977562.
14. Shaulian E. AP-1--The Jun proteins: Oncogenes or tumor suppressors in disguise? Cell Signal. 2010; 22:894–899. PMID: 20060892.
15. Kawada N, Moriyama T, Kitamura H, Yamamoto R, Furumatsu Y, Matsui I, Takabatake Y, Nagasawa Y, Imai E, Wilcox CS, Rakugi H, Isaka Y. Towards developing new strategies to reduce the adverse side-effects of nonsteroidal anti-inflammatory drugs. Clin Exp Nephrol. 2012; 16:25–29. PMID: 22038259.
Article
16. Li C, Lin G, Zuo Z. Pharmacological effects and pharmacokinetics properties of Radix Scutellariae and its bioactive flavones. Biopharm Drug Dispos. 2011; 32:427–445. PMID: 21928297.
Article
17. Pontiki E, Hadjipavlou-Litina D, Litinas K, Nicolotti O, Carotti A. Design, synthesis and pharmacobiological evaluation of novel acrylicacid derivatives acting as lipoxygenase and cyclooxygenase-1 inhibitors with antioxidant and anti-inflammatory activities. Eur J Med Chem. 2011; 46:191–200. PMID: 21106277.
18. Altavilla D, Squadrito F, Bitto A, Polito F, Burnett BP, Di Stefano V, Minutoli L. Flavocoxid, a dual inhibitor of cyclooxygenase and 5-lipoxygenase, blunts pro-inflammatory phenotype activation in endotoxin-stimulated macrophages. Br J Pharmacol. 2009; 157:1410–1418. PMID: 19681869.
Article
19. Sharma N, Mohanakrishnan D, Shard A, Sharma A, Saima , Sinha AK, Sahal D. Stilbene-chalcone hybrids: design, synthesis, and evaluation as a new class of antimalarial scaffolds that trigger cell death through stage specific apoptosis. J Med Chem. 2012; 55:297–311. PMID: 22098429.
Article
20. Karapetyan G, Chakrabarty K, Hein M, Langer P. Synthesis and bioactivity of carbohydrate derivatives of indigo, its isomers and heteroanalogues. Chem Med Chem. 2011; 6:25–37. PMID: 21108279.
Article
21. Silvia V, Baldisserotto A, Scalambra E, Malisardi G, Durini E, Manfredini S. Novel molecular combination deriving from natural aminoacids and polyphenols: Design, synthesis and free-radical scavenging activities. Eur J Med Chem. 2012; 50:383–392. PMID: 22385675.
Article
22. Fokialakis N, Magiatis P, Mitaku S, Pratsinis H, Tillequin F. Estrogenic activity of phenylpropanoids from Sarcomelicope megistophylla and structure determination of a new norneolignan. Planta Med. 2003; 69:566–568. PMID: 12865982.
23. Murray PJ, Wynn TA. Protective and pathogenic functions of macrophage subsets. Nat Rev Immunol. 2011; 11:723–737. PMID: 21997792.
Article
24. Vesely PW, Staber PB, Hoefler G, Kenner L. Translational regulation mechanisms of AP-1 proteins. Mutat Res. 2009; 682:7–12. PMID: 19167516.
Article
25. Pasparakis M. Regulation of tissue homeostasis by NF-kappaB signalling: implications for inflammatory diseases. Nat Rev Immunol. 2009; 9:778–788. PMID: 19855404.
26. Schonthaler HB, Guinea-Viniegra J, Wagner EF. Targeting inflammation by modulating the Jun/AP-1 pathway. Ann Rheum Dis. 2011; 70:i109–i112. PMID: 21339212.
Article
Full Text Links
  • KJPP
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