DOI QR코드

DOI QR Code

Hesperetin suppresses LPS/high glucose-induced inflammatory responses via TLR/MyD88/NF-κB signaling pathways in THP-1 cells

  • Lee, Aeri (Department of Food and Nutrition, Chonnam National University) ;
  • Gu, HyunJi (Department of Food and Nutrition, Chonnam National University) ;
  • Gwon, Min-Hee (Department of Education, Graduate School of Education, Chonnam National University) ;
  • Yun, Jung-Mi (Department of Food and Nutrition, Chonnam National University)
  • Received : 2020.12.23
  • Accepted : 2021.03.30
  • Published : 2021.10.01

Abstract

BACKGROUND/OBJECTIVES: Unregulated inflammatory responses caused by hyperglycemia may induce diabetes complications. Hesperetin, a bioflavonoid, is a glycoside in citrus fruits and is known to have antioxidant and anticarcinogenic properties. However, the effect of inflammation on the diabetic environment has not been reported to date. In this study, we investigated the effect of hesperetin on proinflammatory cytokine secretion and its underlying mechanistic regulation in THP-1 macrophages with co-treatment LPS and hyperglycemic conditions. MATERIALS/METHODS: THP-1 cells differentiated by PMA (1 µM) were cultured for 48 h in the presence or absence of hesperetin under normoglycemic (5.5 mM/L glucose) or hyperglycemic (25 mM/L glucose) conditions and then treated with LPS (100 ng/mL) for 6 h before harvesting. Inflammation-related proteins and mRNA levels were evaluated by enzyme-linked immunosorbent assay, western blot, and quantitative polymerase chain reaction analyses. RESULTS: Hesperetin (0-100 µM, 48 h) treatment did not affect cell viability. The tumor necrosis factor-α and interleukin-6 levels increased in cells co-treated with LPS under hyperglycemic conditions compared to normoglycemic conditions, and these increases were decreased by hesperetin treatment. The TLR2/4 and MyD88 activity levels increased in cells co-treated with LPS under hyperglycemic conditions compared to normoglycemic conditions; however, hesperetin treatment inhibited the TLR2/4 and MyD88 activity increases. In addition, nuclear factor-κB (NF-κB) and Acetyl-NF-κB levels increased in response to treatment with LPS under hyperglycemic conditions compared to normoglycemic conditions, but those levels were decreased when treated with hesperetin. SIRT3 and SIRT6 expressions were increased by hesperetin treatment. CONCLUSIONS: Our results suggest that hesperetin may be a potential agent for suppressing inflammation in diabetes.

Keywords

Acknowledgement

This study was financially supported by Chonnam National University (Grant number: 2018-3468).

References

  1. Capes SE, Hunt D, Malmberg K, Gerstein HC. Stress hyperglycaemia and increased risk of death after myocardial infarction in patients with and without diabetes: a systematic overview. Lancet 2000;355:773-8. https://doi.org/10.1016/S0140-6736(99)08415-9
  2. Urakami T, Kubota S, Nitadori Y, Harada K, Owada M, Kitagawa T. Annual incidence and clinical characteristics of type 2 diabetes in children as detected by urine glucose screening in the Tokyo metropolitan area. Diabetes Care 2005;28:1876-81. https://doi.org/10.2337/diacare.28.8.1876
  3. Saeedi P, Petersohn I, Salpea P, Malanda B, Karuranga S, Unwin N, Colagiuri S, Guariguata L, Motala AA, Ogurtsova K, Shaw JE, Bright D, Williams RIDF Diabetes Atlas Committee. Global and regional diabetes prevalence estimates for 2019 and projections for 2030 and 2045: results from the International Diabetes Federation Diabetes Atlas, 9th edition. Diabetes Res Clin Pract 2019;157:107843. https://doi.org/10.1016/j.diabres.2019.107843
  4. Lumeng CN, Saltiel AR. Inflammatory links between obesity and metabolic disease. J Clin Invest 2011;121:2111-7. https://doi.org/10.1172/JCI57132
  5. Calles-Escandon J, Cipolla M. Diabetes and endothelial dysfunction: a clinical perspective. Endocr Rev 2001;22:36-52. https://doi.org/10.1210/edrv.22.1.0417
  6. Shanmugam N, Kim YS, Lanting L, Natarajan R. Regulation of cyclooxygenase-2 expression in monocytes by ligation of the receptor for advanced glycation end products. J Biol Chem 2003;278:34834-44. https://doi.org/10.1074/jbc.M302828200
  7. de Winther MP, Kanters E, Kraal G, Hofker MH. Nuclear factor kappaB signaling in atherogenesis. Arterioscler Thromb Vasc Biol 2005;25:904-14. https://doi.org/10.1161/01.ATV.0000160340.72641.87
  8. Arkan MC, Hevener AL, Greten FR, Maeda S, Li ZW, Long JM, Wynshaw-Boris A, Poli G, Olefsky J, Karin M. IKK-beta links inflammation to obesity-induced insulin resistance. Nat Med 2005;11:191-8. https://doi.org/10.1038/nm1185
  9. Yang SM, Kim SY, Lee KY, Kim YS, Nam MS, Park IB. Inflammatory markers are associated with microvascular complications in type 2 diabetes. J Korean Diabetes Assoc 2007;31:472-9. https://doi.org/10.4093/jkda.2007.31.6.472
  10. Dasu MR, Ramirez S, Isseroff RR. Toll-like receptors and diabetes: a therapeutic perspective. Clin Sci (Lond) 2012;122:203-14. https://doi.org/10.1042/CS20110357
  11. Rhee SH, Hwang D. Murine TOLL-like receptor 4 confers lipopolysaccharide responsiveness as determined by activation of NF kappa B and expression of the inducible cyclooxygenase. J Biol Chem 2000;275:34035-40. https://doi.org/10.1074/jbc.M007386200
  12. Liang H, Hussey SE, Sanchez-Avila A, Tantiwong P, Musi N. Effect of lipopolysaccharide on inflammation and insulin action in human muscle. PLoS One 2013;8:e63983. https://doi.org/10.1371/journal.pone.0063983
  13. Park BS, Lee JO. Recognition of lipopolysaccharide pattern by TLR4 complexes. Exp Mol Med 2013;45:e66-66. https://doi.org/10.1038/emm.2013.97
  14. Kany S, Vollrath JT, Relja B. Cytokines in inflammatory disease. Int J Mol Sci 2019;20:20. https://doi.org/10.3390/ijms20010020
  15. Haigis MC, Sinclair DA. Mammalian sirtuins: biological insights and disease relevance. Annu Rev Pathol 2010;5:253-95. https://doi.org/10.1146/annurev.pathol.4.110807.092250
  16. Bosch-Presegue L, Vaquero A. The dual role of sirtuins in cancer. Genes Cancer 2011;2:648-62. https://doi.org/10.1177/1947601911417862
  17. Ma Y, Chen H, He X, Nie H, Hong Y, Sheng C, Wang Q, Xia W, Ying W. NAD+ metabolism and NAD(+)-dependent enzymes: promising therapeutic targets for neurological diseases. Curr Drug Targets 2012;13:222-9. https://doi.org/10.2174/138945012799201711
  18. Roth M, Chen WY. Sorting out functions of sirtuins in cancer. Oncogene 2014;33:1609-20. https://doi.org/10.1038/onc.2013.120
  19. Frye RA. Characterization of five human cDNAs with homology to the yeast SIR2 gene: Sir2-like proteins (sirtuins) metabolize NAD and may have protein ADP-ribosyltransferase activity. Biochem Biophys Res Commun 1999;260:273-9. https://doi.org/10.1006/bbrc.1999.0897
  20. Michishita E, Park JY, Burneskis JM, Barrett JC, Horikawa I. Evolutionarily conserved and nonconserved cellular localizations and functions of human SIRT proteins. Mol Biol Cell 2005;16:4623-35. https://doi.org/10.1091/mbc.e05-01-0033
  21. Wang CH, Wei YH. Roles of mitochondrial sirtuins in mitochondrial function, redox homeostasis, insulin resistance and type 2 diabetes. Int J Mol Sci 2020;21:5266. https://doi.org/10.3390/ijms21155266
  22. Kawahara TL, Michishita E, Adler AS, Damian M, Berber E, Lin M, McCord RA, Ongaigui KC, Boxer LD, Chang HY, Chua KF. SIRT6 links histone H3 lysine 9 deacetylation to NF-kappaB-dependent gene expression and organismal life span. Cell 2009;136:62-74. https://doi.org/10.1016/j.cell.2008.10.052
  23. Parhiz H, Roohbakhsh A, Soltani F, Rezaee R, Iranshahi M. Antioxidant and anti-inflammatory properties of the citrus flavonoids hesperidin and hesperetin: an updated review of their molecular mechanisms and experimental models. Phytother Res 2015;29:323-31. https://doi.org/10.1002/ptr.5256
  24. Aranganathan S, Nalini N. Efficacy of the potential chemopreventive agent, hesperetin (citrus flavanone), on 1,2-dimethylhydrazine induced colon carcinogenesis. Food Chem Toxicol 2009;47:2594-600. https://doi.org/10.1016/j.fct.2009.07.019
  25. Morin B, Nichols LA, Zalasky KM, Davis JW, Manthey JA, Holland LJ. The citrus flavonoids hesperetin and nobiletin differentially regulate low density lipoprotein receptor gene transcription in HepG2 liver cells. J Nutr 2008;138:1274-81. https://doi.org/10.1093/jn/138.7.1274
  26. Kumar B, Gupta SK, Srinivasan BP, Nag TC, Srivastava S, Saxena R, Jha KA. Hesperetin rescues retinal oxidative stress, neuroinflammation and apoptosis in diabetic rats. Microvasc Res 2013;87:65-74. https://doi.org/10.1016/j.mvr.2013.01.002
  27. Jialal I, Kaur H. The role of toll-like receptors in diabetes-induced inflammation: implications for vascular complications. Curr Diab Rep 2012;12:172-9. https://doi.org/10.1007/s11892-012-0258-7
  28. Amaral S, Oliveira PJ, Ramalho-Santos J. Diabetes and the impairment of reproductive function: possible role of mitochondria and reactive oxygen species. Curr Diabetes Rev 2008;4:46-54. https://doi.org/10.2174/157339908783502398
  29. Nishikawa T, Araki E. Investigation of a novel mechanism of diabetic complications: impacts of mitochondrial reactive oxygen species. Rinsho Byori 2008;56:712-9.
  30. Jain SK, Kannan K, Lim G, Matthews-Greer J, McVie R, Bocchini JA Jr. Elevated blood interleukin-6 levels in hyperketonemic type 1 diabetic patients and secretion by acetoacetate-treated cultured U937 monocytes. Diabetes Care 2003;26:2139-43. https://doi.org/10.2337/diacare.26.7.2139
  31. Jain SK, Kannan K, Lim G, McVie R, Bocchini JA Jr. Hyperketonemia increases tumor necrosis factor-alpha secretion in cultured U937 monocytes and Type 1 diabetic patients and is apparently mediated by oxidative stress and cAMP deficiency. Diabetes 2002;51:2287-93. https://doi.org/10.2337/diabetes.51.7.2287
  32. Kern PA, Ranganathan S, Li C, Wood L, Ranganathan G. Adipose tissue tumor necrosis factor and interleukin-6 expression in human obesity and insulin resistance. Am J Physiol Endocrinol Metab 2001;280:E745-51. https://doi.org/10.1152/ajpendo.2001.280.5.E745
  33. Yun JM, Jialal I, Devaraj S. Epigenetic regulation of high glucose-induced proinflammatory cytokine production in monocytes by curcumin. J Nutr Biochem 2011;22:450-8. https://doi.org/10.1016/j.jnutbio.2010.03.014
  34. Matulewicz N, Karczewska-Kupczewska M. Insulin resistance and chronic inflammation. Postepy Hig Med Dosw 2016;70:1245-58.
  35. Wu H, Ballantyne CM. Metabolic inflammation and insulin resistance in obesity. Circ Res 2020;126:1549-64. https://doi.org/10.1161/CIRCRESAHA.119.315896
  36. Hossain M, Faruque MO, Kabir G, Hassan N, Sikdar D, Nahar Q, Ali L. Association of serum TNF-α and IL-6 with insulin secretion and insulin resistance in IFG and IGT subjects in a Bangladeshi population. Int J Diabetes Mellit 2010;2:165-8. https://doi.org/10.1016/j.ijdm.2010.08.004
  37. Schmidt MI, Duncan BB, Sharrett AR, Lindberg G, Savage PJ, Offenbacher S, Azambuja MI, Tracy RP, Heiss G. Markers of inflammation and prediction of diabetes mellitus in adults (Atherosclerosis Risk in Communities study): a cohort study. Lancet 1999;353:1649-52. https://doi.org/10.1016/S0140-6736(99)01046-6
  38. Xie C, Kang J, Ferguson ME, Nagarajan S, Badger TM, Wu X. Blueberries reduce pro-inflammatory cytokine TNF-α and IL-6 production in mouse macrophages by inhibiting NF-κB activation and the MAPK pathway. Mol Nutr Food Res 2011;55:1587-91. https://doi.org/10.1002/mnfr.201100344
  39. Guha M, Bai W, Nadler JL, Natarajan R. Molecular mechanisms of tumor necrosis factor alpha gene expression in monocytic cells via hyperglycemia-induced oxidant stress-dependent and -independent pathways. J Biol Chem 2000;275:17728-39. https://doi.org/10.1074/jbc.275.23.17728
  40. Choi RY, Ham JR, Lee MK. Esculetin prevents non-alcoholic fatty liver in diabetic mice fed high-fat diet. Chem Biol Interact 2016;260:13-21. https://doi.org/10.1016/j.cbi.2016.10.013
  41. Lee H, Yang SJ. In vitro and in vivo effects of piceatannol and resveratrol on glucose control and TLR4-NF-κB pathway. J Korean Soc Food Sci Nutr 2017;46:267-72. https://doi.org/10.3746/jkfn.2017.46.2.267
  42. Youn HS, Lee JY, Fitzgerald KA, Young HA, Akira S, Hwang DH. Specific inhibition of MyD88-independent signaling pathways of TLR3 and TLR4 by resveratrol: molecular targets are TBK1 and RIP1 in TRIF complex. J Immunol 2005;175:3339-46. https://doi.org/10.4049/jimmunol.175.5.3339
  43. Ghanim H, Sia CL, Upadhyay M, Korzeniewski K, Viswanathan P, Abuaysheh S, Mohanty P, Dandona P. Orange juice neutralizes the proinflammatory effect of a high-fat, high-carbohydrate meal and prevents endotoxin increase and Toll-like receptor expression. Am J Clin Nutr 2010;91:940-9. https://doi.org/10.3945/ajcn.2009.28584
  44. Hua KF, Wang SH, Dong WC, Lin CY, Ho CL, Wu TH. High glucose increases nitric oxide generation in lipopolysaccharide-activated macrophages by enhancing activity of protein kinase C-α/δ and NF-κB. Inflamm Res 2012;61:1107-16. https://doi.org/10.1007/s00011-012-0503-1
  45. Yang F, de Villiers WJ, McClain CJ, Varilek GW. Green tea polyphenols block endotoxin-induced tumor necrosis factor-production and lethality in a murine model. J Nutr 1998;128:2334-40. https://doi.org/10.1093/jn/128.12.2334
  46. Tsai SH, Lin-Shiau SY, Lin JK. Suppression of nitric oxide synthase and the down-regulation of the activation of NFkappaB in macrophages by resveratrol. Br J Pharmacol 1999;126:673-80. https://doi.org/10.1038/sj.bjp.0702357
  47. Chan MM, Mattiacci JA, Hwang HS, Shah A, Fong D. Synergy between ethanol and grape polyphenols, quercetin, and resveratrol, in the inhibition of the inducible nitric oxide synthase pathway. Biochem Pharmacol 2000;60:1539-48. https://doi.org/10.1016/S0006-2952(00)00471-8
  48. Huynh FK, Hershberger KA, Hirschey MD. Targeting sirtuins for the treatment of diabetes. Diabetes Manag (Lond) 2013;3:245-57. https://doi.org/10.2217/dmt.13.6
  49. Elliott P, Walpole S, Morelli L, Lambert P, Lunsmann W, Westphal C, Lavu S. Resveratrol/SRT-501. Drugs Fut 2009;34:291-5. https://doi.org/10.1358/dof.2009.034.04.1360696