Nutr Res Pract.  2016 Dec;10(6):623-628. 10.4162/nrp.2016.10.6.623.

Induction of heme oxygenase-1 with dietary quercetin reduces obesity-induced hepatic inflammation through macrophage phenotype switching

Affiliations
  • 1Department of Food Science and Nutrition, University of Ulsan, 93 Daehak-ro, Nam-ku, Ulsan 44610, Korea. rinayu@ulsan.ac.kr
  • 2Department of Biological Science, University of Ulsan, Ulsan 44610, Korea.

Abstract

BACKGROUND/OBJECTIVES
Obesity-induced steatohepatitis accompanied by activated hepatic macrophages/Kupffer cells facilitates the progression of hepatic fibrinogenesis and exacerbates metabolic derangements such as insulin resistance. Heme oxyganase-1 (HO-1) modulates tissue macrophage phenotypes and thus is implicated in protection against inflammatory diseases. Here, we show that the flavonoid quercetin reduces obesity-induced hepatic inflammation by inducing HO-1, which promotes hepatic macrophage polarization in favor of the M2 phenotype.
MATERIALS/METHODS
Male C57BL/6 mice were fed a regular diet (RD), high-fat diet (HFD), or HFD supplemented with quercetin (HF+Que, 0.5g/kg diet) for nine weeks. Inflammatory cytokines and macrophage markers were measured by ELISA and RT-PCR, respectively. HO-1 protein was measured by Western blotting.
RESULTS
Quercetin supplementation decreased levels of inflammatory cytokines (TNFα, IL-6) and increased that of the anti-inflammatory cytokine (IL-10) in the livers of HFD-fed mice. This was accompanied by upregulation of M2 macrophage marker genes (Arg-1, Mrc1) and downregulation of M1 macrophage marker genes (TNFα, NOS2). In co-cultures of lipid-laden hepatocytes and macrophages, treatment with quercetin induced HO-1 in the macrophages, markedly suppressed expression of M1 macrophage marker genes, and reduced release of MCP-1. Moreover, these effects of quercetin were blunted by an HO-1 inhibitor and deficiency of nuclear factor E2-related factor 2 (Nrf2) in macrophages.
CONCLUSIONS
Quercetin reduces obesity-induced hepatic inflammation by promoting macrophage phenotype switching. The beneficial effect of quercetin is associated with Nrf2-mediated HO-1 induction. Quercetin may be a useful dietary factor for protecting against obesity-induced steatohepatitis.

Keyword

Obesity; quercetin; inflammation; macrophage

MeSH Terms

Animals
Blotting, Western
Coculture Techniques
Cytokines
Diet
Diet, High-Fat
Down-Regulation
Enzyme-Linked Immunosorbent Assay
Fatty Liver
Heme Oxygenase-1*
Heme*
Hepatocytes
Humans
Inflammation*
Insulin Resistance
Liver
Macrophages*
Male
Mice
NF-E2-Related Factor 2
Obesity
Phenotype*
Quercetin*
Up-Regulation
Cytokines
Heme
Heme Oxygenase-1
NF-E2-Related Factor 2
Quercetin

Figure

  • Fig. 1 Effects of quercetin on hepatic inflammatory responses and macrophage phenotypes in HFD-fed obese mice. (A) Inflammatory cytokines (TNFα, IL-6, and MCP-1) and (B) an anti-inflammatory cytokine (IL-10) in liver were determined as described in Methods. Results are means ± SEM of six mice per group. * P < 0.05, ** P < 0.01, *** P < 0.001 compared with obese mice fed an HFD. (C) F4/80 as a macrophage marker and (D-E) M1 and M2 macrophages mRNAs in livers were determined by real-time PCR. The intensity of the bands was densitometrically measured and normalized to GAPDH transcripts. Results are means ± SEM of six mice per group. * P < 0.05, ** P < 0.01 compared with obese mice fed an HFD.

  • Fig. 2 Effect of HO-1 inhibition on quercetin action in co-cultures of lipid-laden hepatocytes with macrophages. Primary hepatocytes were treated with palmitic acid (BSA: palmitic acid ratio 1:5) for 12h. Raw 264.7 macrophages were then co-cultured with the lipid-laden hepatocytes for 12h in the presence or absence of quercetin or ZnPP. L-Hepa; lipid-laden hepatocytes, Co-culture; lipid-laden hepatocytes and macrophages, Que; quercetin. (A) HO-1 protein was measured by western blotting in macrophages. Results are means ± SEM of two experiments with duplicate determinations. * P < 0.05, *** P < 0.001 compared with no treatment. (B) MCP-1 production detected by ELISA in co-cultured lipid-laden hepatocytes and macrophages. Results are means ± SEM of triplicate samples. * P < 0.05, *** P < 0.001 compared with quercetin treatment. (C) M1 macrophage mRNAs in co-cultured lipid-laden hepatocytes and macrophages determined by real-time PCR. Results are means ± SEM of quadruplicate samples. * P < 0.05, ** P < 0.01, *** P < 0.001 compared with quercetin treatment.

  • Fig. 3 Effect of Nrf2 deficiency on quercetin action in LPS-stimulated macrophages. (A) HO-1 protein was measured by western blotting in peritoneal macrophages of Nrf2 KO and WT mice. Results are means ± SEM of three independent experiments. ** P < 0.01 compared with LPS treatment. (B-C) M1/M2 marker mRNAs were determined by real-time PCR. The intensity of the bands was densitometrically measured and normalized to GAPDH transcripts. Results are means ± SEM of two experiments with duplicate determinations. * P < 0.05, ** P < 0.01, *** P < 0.001 compared with LPS treatment.


Reference

1. Bilzer M, Roggel F, Gerbes AL. Role of Kupffer cells in host defense and liver disease. Liver Int. 2006; 26:1175–1186.
Article
2. Li Z, Diehl AM. Innate immunity in the liver. Curr Opin Gastroenterol. 2003; 19:565–571.
Article
3. Lanthier N, Molendi-Coste O, Horsmans Y, van Rooijen N, Cani PD, Leclercq IA. Kupffer cell activation is a causal factor for hepatic insulin resistance. Am J Physiol Gastrointest Liver Physiol. 2010; 298:G107–G116.
Article
4. Mantovani A, Sica A, Sozzani S, Allavena P, Vecchi A, Locati M. The chemokine system in diverse forms of macrophage activation and polarization. Trends Immunol. 2004; 25:677–686.
Article
5. Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest. 2007; 117:175–184.
Article
6. Dixon LJ, Barnes M, Tang H, Pritchard MT, Nagy LE. Kupffer cells in the liver. Compr Physiol. 2013; 3:785–797.
Article
7. Odegaard JI, Ricardo-Gonzalez RR, Red Eagle A, Vats D, Morel CR, Goforth MH, Subramanian V, Mukundan L, Ferrante AW, Chawla A. Alternative M2 activation of Kupffer cells by PPARdelta ameliorates obesity-induced insulin resistance. Cell Metab. 2008; 7:496–507.
Article
8. Wan J, Benkdane M, Teixeira-Clerc F, Bonnafous S, Louvet A, Lafdil F, Pecker F, Tran A, Gual P, Mallat A, Lotersztajn S, Pavoine C. M2 Kupffer cells promote M1 Kupffer cell apoptosis: a protective mechanism against alcoholic and nonalcoholic fatty liver disease. Hepatology. 2014; 59:130–142.
Article
9. Alam J, Stewart D, Touchard C, Boinapally S, Choi AM, Cook JL. Nrf2, a Cap'n'Collar transcription factor, regulates induction of the heme oxygenase-1 gene. J Biol Chem. 1999; 274:26071–26078.
Article
10. Lee TS, Chau LY. Heme oxygenase-1 mediates the anti-inflammatory effect of interleukin-10 in mice. Nat Med. 2002; 8:240–246.
Article
11. Ndisang JF. Role of heme oxygenase in inflammation, insulin-signalling, diabetes and obesity. Mediators Inflamm. 2010; 2010:359732.
Article
12. Kim DH, Burgess AP, Li M, Tsenovoy PL, Addabbo F, McClung JA, Puri N, Abraham NG. Heme oxygenase-mediated increases in adiponectin decrease fat content and inflammatory cytokines tumor necrosis factor-alpha and interleukin-6 in Zucker rats and reduce adipogenesis in human mesenchymal stem cells. J Pharmacol Exp Ther. 2008; 325:833–840.
Article
13. L'Abbate A, Neglia D, Vecoli C, Novelli M, Ottaviano V, Baldi S, Barsacchi R, Paolicchi A, Masiello P, Drummond GS, McClung JA, Abraham NG. Beneficial effect of heme oxygenase-1 expression on myocardial ischemia-reperfusion involves an increase in adiponectin in mildly diabetic rats. Am J Physiol Heart Circ Physiol. 2007; 293:H3532–H3541.
14. Alcaraz MJ, Fernández P, Guillén MI. Anti-inflammatory actions of the heme oxygenase-1 pathway. Curr Pharm Des. 2003; 9:2541–2551.
Article
15. Tu TH, Joe Y, Choi HS, Chung HT, Yu R. Induction of heme oxygenase-1 with hemin reduces obesity-induced adipose tissue inflammation via adipose macrophage phenotype switching. Mediators Inflamm. 2014; 2014:290708.
Article
16. Yao P, Hao L, Nussler N, Lehmann A, Song F, Zhao J, Neuhaus P, Liu L, Nussler A. The protective role of HO-1 and its generated products (CO, bilirubin, and Fe) in ethanol-induced human hepatocyte damage. Am J Physiol Gastrointest Liver Physiol. 2009; 296:G1318–G1323.
Article
17. Son Y, Lee JH, Chung HT, Pae HO. Therapeutic roles of heme oxygenase-1 in metabolic diseases: curcumin and resveratrol analogues as possible inducers of heme oxygenase-1. Oxid Med Cell Longev. 2013; 2013:639541.
Article
18. Anjaneyulu M, Chopra K. Quercetin, an anti-oxidant bioflavonoid, attenuates diabetic nephropathy in rats. Clin Exp Pharmacol Physiol. 2004; 31:244–248.
Article
19. Overman A, Chuang CC, McIntosh M. Quercetin attenuates inflammation in human macrophages and adipocytes exposed to macrophage-conditioned media. Int J Obes (Lond). 2011; 35:1165–1172.
Article
20. Panchal SK, Poudyal H, Brown L. Quercetin ameliorates cardiovascular, hepatic, and metabolic changes in diet-induced metabolic syndrome in rats. J Nutr. 2012; 142:1026–1032.
Article
21. Noh HJ, Kim CS, Kang JH, Park JY, Choe SY, Hong SM, Yoo H, Park T, Yu R. Quercetin suppresses MIP-1alpha-induced adipose inflammation by downregulating its receptors CCR1/CCR5 and inhibiting inflammatory signaling. J Med Food. 2014; 17:550–557.
Article
22. Le NH, Kim CS, Park T, Park JH, Sung MK, Lee DG, Hong SM, Choe SY, Goto T, Kawada T, Yu R. Quercetin protects against obesity-induced skeletal muscle inflammation and atrophy. Mediators Inflamm. 2014; 2014:834294.
Article
23. Ying HZ, Liu YH, Yu B, Wang ZY, Zang JN, Yu CH. Dietary quercetin ameliorates nonalcoholic steatohepatitis induced by a high-fat diet in gerbils. Food Chem Toxicol. 2013; 52:53–60.
Article
24. Li X, Wang R, Zhou N, Wang X, Liu Q, Bai Y, Bai Y, Liu Z, Yang H, Zou J, Wang H, Shi T. Quercetin improves insulin resistance and hepatic lipid accumulation in vitro in a NAFLD cell model. Biomed Rep. 2013; 1:71–76.
Article
25. Marcolin E, San-Miguel B, Vallejo D, Tieppo J, Marroni N, González-Gallego J, Tuñón MJ. Quercetin treatment ameliorates inflammation and fibrosis in mice with nonalcoholic steatohepatitis. J Nutr. 2012; 142:1821–1828.
Article
26. Kim CS, Yu R. The inhibitory effect of quercetin on adipose tissue inflammation in mice fed on a high-fat diet. Korean J Obes. 2014; 23:170–178.
Article
27. Lin HY, Juan SH, Shen SC, Hsu FL, Chen YC. Inhibition of lipopolysaccharide-induced nitric oxide production by flavonoids in RAW264.7 macrophages involves heme oxygenase-1. Biochem Pharmacol. 2003; 66:1821–1832.
Article
28. Marra F, Lotersztajn S. Pathophysiology of NASH: perspectives for a targeted treatment. Curr Pharm Des. 2013; 19:5250–5269.
Article
29. Tosello-Trampont AC, Landes SG, Nguyen V, Novobrantseva TI, Hahn YS. Kuppfer cells trigger nonalcoholic steatohepatitis development in diet-induced mouse model through tumor necrosis factor-alpha production. J Biol Chem. 2012; 287:40161–40172.
Article
30. Dong J, Zhang X, Zhang L, Bian HX, Xu N, Bao B, Liu J. Quercetin reduces obesity-associated ATM infiltration and inflammation in mice: a mechanism including AMPKalpha1/SIRT1. J Lipid Res. 2014; 55:363–374.
Article
31. Matsuzawa N, Takamura T, Kurita S, Misu H, Ota T, Ando H, Yokoyama M, Honda M, Zen Y, Nakanuma Y, Miyamoto K, Kaneko S. Lipid-induced oxidative stress causes steatohepatitis in mice fed an atherogenic diet. Hepatology. 2007; 46:1392–1403.
Article
32. Kim CS, Kwon Y, Choe SY, Hong SM, Yoo H, Goto T, Kawada T, Choi HS, Joe Y, Chung HT, Yu R. Quercetin reduces obesity-induced hepatosteatosis by enhancing mitochondrial oxidative metabolism via heme oxygenase-1. Nutr Metab (Lond). 2015; 12:33.
Article
33. Endale M, Park SC, Kim S, Kim SH, Yang Y, Cho JY, Rhee MH. Quercetin disrupts tyrosine-phosphorylated phosphatidylinositol 3-kinase and myeloid differentiation factor-88 association, and inhibits MAPK/AP-1 and IKK/NF-kappaB-induced inflammatory mediators production in RAW 264.7 cells. Immunobiology. 2013; 218:1452–1467.
Article
Full Text Links
  • NRP
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