DOI QR코드

DOI QR Code

Anti-inflammatory effect of sulforaphane on LPS-stimulated RAW 264.7 cells and ob/ob mice

  • Ranaweera, Sachithra S. (College of Veterinary Medicine, Jeju National University) ;
  • Dissanayake, Chanuri Y. (College of Veterinary Medicine, Jeju National University) ;
  • Natraj, Premkumar (College of Veterinary Medicine, Jeju National University) ;
  • Lee, Young Jae (College of Veterinary Medicine, Jeju National University) ;
  • Han, Chang-Hoon (College of Veterinary Medicine, Jeju National University)
  • Received : 2020.09.18
  • Accepted : 2020.10.28
  • Published : 2020.11.30

Abstract

Background: Sulforaphane (SFN) is an isothiocyanate compound present in cruciferous vegetables. Although the anti-inflammatory effects of SFN have been reported, the precise mechanism related to the inflammatory genes is poorly understood. Objectives: This study examined the relationship between the anti-inflammatory effects of SFN and the differential gene expression pattern in SFN treated ob/ob mice. Methods: Nitric oxide (NO) level was measured using a Griess assay. The inducible nitric oxide synthase (iNOS) and cyclooxygenase-2 (COX-2) expression levels were analyzed by Western blot analysis. Pro-inflammatory cytokines (tumor necrosis factor [TNF]-α, interleukin [IL]-1β, and IL-6) were measured by enzyme-linked immunosorbent assay (ELISA). RNA sequencing analysis was performed to evaluate the differential gene expression in the liver of ob/ob mice. Results: The SFN treatment significantly attenuated the iNOS and COX-2 expression levels and inhibited NO, TNF-α, IL-1β, and IL-6 production in lipopolysaccharide (LPS)-stimulated RAW 264.7 cells. RNA sequencing analysis showed that the expression levels of 28 genes related to inflammation were up-regulated (> 2-fold), and six genes were down-regulated (< 0.6-fold) in the control ob/ob mice compared to normal mice. In contrast, the gene expression levels were restored to the normal level by SFN. The protein-protein interaction (PPI) network showed that chemokine ligand (Cxcl14, Ccl1, Ccl3, Ccl4, Ccl17) and chemokine receptor (Ccr3, Cxcr1, Ccr10) were located in close proximity and formed a "functional cluster" in the middle of the network. Conclusions: The overall results suggest that SFN has a potent anti-inflammatory effect by normalizing the expression levels of the genes related to inflammation that were perturbed in ob/ob mice.

Keywords

Acknowledgement

This study was supported by the program of supporting Promising Small and Medium Industry (Grant No: C0518751) funded by the Korea Small and Medium Business Administration in 2017. The study was also supported by the 2019 scientific promotion program funded by Jeju National University.

References

  1. Harrison DG, Guzik TJ, Lob HE, Madhur MS, Marvar PJ, Thabet SR, et al. Inflammation, immunity, and hypertension. Hypertension. 2011;57(2):132-140. https://doi.org/10.1161/hypertensionaha.110.163576
  2. Shapiro H, Lutaty A, Ariel A. Macrophages, meta-inflammation, and immuno-metabolism. Sci World J. 2011;11:2509-2529. https://doi.org/10.1100/2011/397971
  3. Biswas SK, Lewis CE. NF-κB as a central regulator of macrophage function in tumors. J Leukoc Biol. 2010;88(5):877-884. https://doi.org/10.1189/jlb.0310153
  4. Li Q, Verma IM. NF-κB regulation in the immune system. Nat Rev Immunol. 2002;2(10):725-734. https://doi.org/10.1038/nri910
  5. Ye J, McGuinness OP. Inflammation during obesity is not all bad: evidence from animal and human studies. Am J Physiol Endocrinol Metab. 2013;304(5):E466-E477. https://doi.org/10.1152/ajpendo.00266.2012
  6. Ellulu MS, Patimah I, Khaza'ai H, Rahmat A, Abed Y. Obesity and inflammation: the linking mechanism and the complications. Arch Med Sci. 2017;13(4):851-863.
  7. Hotamisligil GS. Inflammation and metabolic disorders. Nature. 2006;444(7121):860-867. https://doi.org/10.1038/nature05485
  8. Sutter AG, Palanisamy AP, Lench JH, Jessmore AP, Chavin KD. Development of steatohepatitis in ob/ob mice is dependent on Toll-like receptor 4. Ann Hepatol. 2015;14(5):735-743. https://doi.org/10.1016/S1665-2681(19)30769-0
  9. Perfield JW 2nd, Ortinau LC, Pickering RT, Ruebel ML, Meers GM, Rector RS. Altered hepatic lipid metabolism contributes to nonalcoholic fatty liver disease in leptin-deficient ob/ob mice. J Obes. 2013;2013:296537.
  10. Lauterbach MA, Wunderlich FT. Macrophage function in obesity-induced inflammation and insulin resistance. Pflugers Arch. 2017;469(3-4):385-396. https://doi.org/10.1007/s00424-017-1955-5
  11. Santin-Marquez R, Alarcon-Aguilar A, Lopez-Diazguerrero NE, Chondrogianni N, Konigsberg M. Sulforaphane - role in aging and neurodegeneration. Geroscience. 2019;41(5):655-670. https://doi.org/10.1007/s11357-019-00061-7
  12. Angelino D, Jeffery E. Glucosinolate hydrolysis and bioavailability of resulting isothiocyanates: focus on glucoraphanin. J Funct Foods. 2014;7:67-76. https://doi.org/10.1016/j.jff.2013.09.029
  13. Xu L, Nagata N, Ota T. Glucoraphanin: a broccoli sprout extract that ameliorates obesity-induced inflammation and insulin resistance. Adipocyte. 2018;7(3):218-225. https://doi.org/10.1080/21623945.2018.1474669
  14. Galuppo M, Giacoppo S, De Nicola GR, Iori R, Mazzon E, Bramanti P. RS-Glucoraphanin bioactivated with myrosinase treatment counteracts proinflammatory cascade and apoptosis associated to spinal cord injury in an experimental mouse model. J Neurol Sci. 2013;334(1-2):88-96. https://doi.org/10.1016/j.jns.2013.07.2514
  15. Sotokawauchi A, Ishibashi Y, Matsui T, Yamagishi SI. Aqueous extract of glucoraphanin-rich broccoli sprouts inhibits formation of advanced glycation end products and attenuates inflammatory reactions in endothelial cells. Evid Based Complement Alternat Med. 2018;2018:9823141.
  16. Vo QV, Nam P, Dinh T, Mechler A, Tran T. Anti-inflammatory activity of synthetic and natural glucoraphanin. J Serbian Chem Soc. 2019;84(5):445-453. https://doi.org/10.2298/jsc180518108v
  17. Dinkova-Kostova AT, Fahey JW, Kostov RV, Kensler TW. KEAP1 and done? Targeting the NRF2 pathway with sulforaphane. Trends Food Sci Technol. 2017;69(Pt B):257-269. https://doi.org/10.1016/j.tifs.2017.02.002
  18. Li B, Cui W, Liu J, Li R, Liu Q, Xie XH, et al. Sulforaphane ameliorates the development of experimental autoimmune encephalomyelitis by antagonizing oxidative stress and Th17-related inflammation in mice. Exp Neurol. 2013;250:239-249. https://doi.org/10.1016/j.expneurol.2013.10.002
  19. Yang G, Lee HE, Lee JY. A pharmacological inhibitor of NLRP3 inflammasome prevents non-alcoholic fatty liver disease in a mouse model induced by high fat diet. Sci Rep. 2016;6(1):1. https://doi.org/10.1038/s41598-016-0001-8
  20. Sanchez-Moreno C, Larrauri JA, Saura-Calixto F. A procedure to measure the antiradical efficiency of polyphenols. J Sci Food Agric. 1998;76(2):270-276. https://doi.org/10.1002/(SICI)1097-0010(199802)76:2<270::AID-JSFA945>3.0.CO;2-9
  21. Vuong LD, Nguyen QN, Truong VL. Anti-inflammatory and anti-oxidant effects of combination between sulforaphane and acetaminophen in LPS-stimulated RAW 264.7 macrophage cells. Immunopharmacol Immunotoxicol. 2019;41(3):413-419. https://doi.org/10.1080/08923973.2019.1569049
  22. Coleman DL. Obese and diabetes: two mutant genes causing diabetes-obesity syndromes in mice. Diabetologia. 1978;14(3):141-148. https://doi.org/10.1007/BF00429772
  23. Bai Y, Wang X, Zhao S, Ma C, Cui J, Zheng Y. Sulforaphane protects against cardiovascular disease via Nrf2 activation. Oxid Med Cell Longev. 2015;2015:407580. https://doi.org/10.1155/2015/407580
  24. Yuan H, Yao S, You Y, Xiao G, You Q. Antioxidant activity of isothiocyanate extracts from broccoli. Chin J Chem Eng. 2010;18(2):312-321. https://doi.org/10.1016/S1004-9541(08)60358-4
  25. Salzano S, Checconi P, Hanschmann EM, Lillig CH, Bowler LD, Chan P, et al. Linkage of inflammation and oxidative stress via release of glutathionylated peroxiredoxin-2, which acts as a danger signal. Proc Natl Acad Sci U S A. 2014;111(33):12157-12162. https://doi.org/10.1073/pnas.1401712111
  26. MacMicking J, Xie QW, Nathan C. Nitric oxide and macrophage function. Annu Rev Immunol. 1997;15(1):323-350. https://doi.org/10.1146/annurev.immunol.15.1.323
  27. Tripathi P, Tripathi P, Kashyap L, Singh V. The role of nitric oxide in inflammatory reactions. FEMS Immunol Med Microbiol. 2007;51(3):443-452. https://doi.org/10.1111/j.1574-695X.2007.00329.x
  28. Mitchell JA, Larkin S, Williams TJ. Cyclooxygenase-2: regulation and relevance in inflammation. Biochem Pharmacol. 1995;50(10):1535-1542. https://doi.org/10.1016/0006-2952(95)00212-X
  29. Hwang JH, Lim SB. Bin. Antioxidant and anti-inflammatory activities of Broccoli florets in LPS-stimulated RAW 264.7 Cells. Prev Nutr Food Sci. 2014;19(2):89-97. https://doi.org/10.3746/PNF.2014.19.2.089
  30. Choi WJ, Kim SK, Park HK, Sohn UD, Kim W. Anti-Inflammatory and Anti-Superbacterial Properties of Sulforaphane from Shepherd's Purse. Korean J Physiol Pharmacol. 2014;18(1):33-39. https://doi.org/10.4196/kjpp.2014.18.1.33
  31. An YW, Jhang KA, Woo SY, Kang JL, Chong YH. Sulforaphane exerts its anti-inflammatory effect against amyloid-β peptide via STAT-1 dephosphorylation and activation of Nrf2/HO-1 cascade in human THP-1 macrophages. Neurobiol Aging. 2016;38:1-10. https://doi.org/10.1016/j.neurobiolaging.2015.10.016
  32. Witte A, Chatterjee M, Lang F, Gawaz M. Platelets as a novel source of pro-inflammatory chemokine CXCL14. Cell Physiol Biochem. 2017;41(4):1684-1696. https://doi.org/10.1159/000471821
  33. Nara N, Nakayama Y, Okamoto S, Tamura H, Kiyono M, Muraoka M, et al. Disruption of CXC motif chemokine ligand-14 in mice ameliorates obesity-induced insulin resistance. J Biol Chem. 2007;282(42):30794-30803. https://doi.org/10.1074/jbc.M700412200
  34. Roos RS, Loetscher M, Legler DF, Clark-Lewis I, Baggiolini M, Moser B. Identification of CCR8, the receptor for the human CC chemokine I-309. J Biol Chem. 1997;272(28):17251-17254. https://doi.org/10.1074/jbc.272.28.17251
  35. Heymann F, Hammerich L, Storch D, Bartneck M, Huss S, Russeler V, et al. Hepatic macrophage migration and differentiation critical for liver fibrosis is mediated by the chemokine receptor C-C motif chemokine receptor 8 in mice. Hepatology. 2012;55(3):898-909. https://doi.org/10.1002/hep.24764
  36. Ajuebor MN, Hogaboam CM, Le T, Proudfoot AE, Swain MG. CCL3/MIP-1α is pro-inflammatory in murine T cell-mediated hepatitis by recruiting CCR1-expressing CD4+ T cells to the liver. Eur J Immunol. 2004;34(10):2907-2918. https://doi.org/10.1002/eji.200425071
  37. Gilet J, Chang Y, Chenivesse C, Legendre B, Vorng H, Duez C, et al. Role of CCL17 in the generation of cutaneous inflammatory reactions in Hu-PBMC-SCID mice grafted with human skin. J Invest Dermatol. 2009;129(4):879-890. https://doi.org/10.1038/jid.2008.333
  38. Murdoch C, Finn A. Chemokine receptors and their role in inflammation and infectious diseases. Blood. 2000;95(10):3032-3043. https://doi.org/10.1182/blood.v95.10.3032.010k17_3032_3043
  39. Crispe IN. The liver as a lymphoid organ. Annu Rev Immunol. 2009;27(1):147-163. https://doi.org/10.1146/annurev.immunol.021908.132629
  40. Watanabe Y, Suzuki O, Haruyama T, Akaike T. Interferon-gamma induces reactive oxygen species and endoplasmic reticulum stress at the hepatic apoptosis. J Cell Biochem. 2003;89(2):244-253. https://doi.org/10.1002/jcb.10501
  41. Gjelstrup LC, Boesen T, Kragstrup TW, Jorgensen A, Klein NJ, Thiel S, et al. Shedding of large functionally active CD11/CD18 Integrin complexes from leukocyte membranes during synovial inflammation distinguishes three types of arthritis through differential epitope exposure. J Immunol. 2010;185(7):4154-4168. https://doi.org/10.4049/jimmunol.1000952
  42. Fruman DA, Chiu H, Hopkins BD, Bagrodia S, Cantley LC, Abraham RT. The PI3K pathway in human disease. Cell. 2017;170(4):605-635. https://doi.org/10.1016/j.cell.2017.07.029