Journal Home > Volume 11 , Issue 1

Innate immunity, particularly macrophages, is critical for intestinal homeostasis. Sulforaphane, a dietary isothiocyanate from cruciferous vegetables, has been reported to protect against intestinal inflammation. However, the role of macrophages in sulforaphane mediated intestinal inflammation and the underlying molecular mechanisms have not been studied yet. In this study, sulforaphane effectively attenuated dextran sodium sulphate (DSS) induced intestinal inflammation in murine model. Of note, sulforaphane skewed the switching from classically (M1) to alternatively (M2) activated phenotype both in intestinal and bone marrow-derived macrophages (BMDMs). The expression levels of M1 associated maker genes induced by DSS or lipopolysaccharide (LPS) plus interferon gamma-γ (IFN-γ) were suppressed by sulforaphane while M2 marker gene expression levels were improved. This resulted in alteration of inflammatory mediators, particularly interleukin-10 (IL-10), both in colon tissues and culture medium of BMDMs. Subsequently, IL-10 was found to mediate the sulforaphane induced M2 phenotype switching of BMDMs through the activation of STAT3 signaling. This was confirmed by immunofluorescence analysis with increased number of p-STAT3-positive cells in the colon sections. Moreover, anti-IL-10 neutralizing antibody significantly interfered M2 phenotyping of BMDMs induced by sulforaphane with reduced STAT3 phosphorylation. Findings here introduced a potential utilization of sulforaphane for intestinal inflammation treatment with macrophages as the therapeutic targets.


menu
Abstract
Full text
Outline
About this article

Sulforaphane attenuates dextran sodium sulphate induced intestinal inflammation via IL-10/STAT3 signaling mediated macrophage phenotype switching

Show Author's information Yuyang SunaJiqing TangaCui LiaJun Liua,b( )Haijie Liua( )
College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
Beijing Advanced Innovation Center for Food Nutrition and Human Health, Beijing Technology and Business University, Beijing 100048, China

Peer review under responsibility of KeAi Communications Co., Ltd.

Abstract

Innate immunity, particularly macrophages, is critical for intestinal homeostasis. Sulforaphane, a dietary isothiocyanate from cruciferous vegetables, has been reported to protect against intestinal inflammation. However, the role of macrophages in sulforaphane mediated intestinal inflammation and the underlying molecular mechanisms have not been studied yet. In this study, sulforaphane effectively attenuated dextran sodium sulphate (DSS) induced intestinal inflammation in murine model. Of note, sulforaphane skewed the switching from classically (M1) to alternatively (M2) activated phenotype both in intestinal and bone marrow-derived macrophages (BMDMs). The expression levels of M1 associated maker genes induced by DSS or lipopolysaccharide (LPS) plus interferon gamma-γ (IFN-γ) were suppressed by sulforaphane while M2 marker gene expression levels were improved. This resulted in alteration of inflammatory mediators, particularly interleukin-10 (IL-10), both in colon tissues and culture medium of BMDMs. Subsequently, IL-10 was found to mediate the sulforaphane induced M2 phenotype switching of BMDMs through the activation of STAT3 signaling. This was confirmed by immunofluorescence analysis with increased number of p-STAT3-positive cells in the colon sections. Moreover, anti-IL-10 neutralizing antibody significantly interfered M2 phenotyping of BMDMs induced by sulforaphane with reduced STAT3 phosphorylation. Findings here introduced a potential utilization of sulforaphane for intestinal inflammation treatment with macrophages as the therapeutic targets.

Keywords: STAT3, Intestinal inflammation, BMDMs, IL-10, Macrophage phenotype, Sulforaphane

References(51)

[1]

B.A. Hendrickson, R. Gokhale, J.H. Cho, Clinical aspects and pathophysiology of inflammatory bowel disease, Clin. Microbiol. Rev. 15 (2002) 79-94. http://doi.org/10.1128/CMR.15.1.79-94.2002.

[2]

B. Khor, A. Gardet, R.J. Xavier, Genetics and pathogenesis of inflammatory bowel disease, Nature 474 (2011) 307-317. https://doi.org/10.1038/nature10209.

[3]

T. Imam, S. Park, M.H. Kaplan, et al., Effector T helper cell subsets in inflammatory bowel diseases, Front Immunol. 9 (2018) 1212. http://doi.org/10.3389/fimmu.2018.01212.

[4]

A. Geremia, P. Biancheri, P. Allan, et al., Innate and adaptive immunity in inflammatory bowel disease, Autoimmun. Rev. 13 (2014) 3-10. https://doi.org/10.1016/j.autrev.2013.06.004.

[5]

P. Ruytinx, P. Proost, J. Van Damme, et al., Chemokine-induced macrophage polarization in inflammatory conditions, Front Immunol. 9 (2018) 1930. https://doi.org/10.3389/fimmu.2018.01930.

[6]

P.J. Murray, Macrophage polarization, Annu. Rev. Physiol. 79 (2017) 541-566. http://doi.org/10.1146/annurev-physiol-022516-034339.

[7]

Y.R. Na, M. Stakenborg, S.H. Seok, et al., Macrophages in intestinal inflammation and resolution: a potential therapeutic target in IBD, Nat. Rev. Gastro. Hepat. 16 (2019) 531-543. http://doi.org/10.1038/s41575-019-0172-4.

[8]

W. Zhu, J. Yu, Y. Nie, et al., Disequilibrium of M1 and M2 macrophages correlates with the development of experimental inflammatory bowel diseases, Immunol. Invest. 43 (2014) 638-652. http://doi.org/10.3109/08820139.2014.909456.

[9]

N. Eissa, H. Hussein, L. Kermarrec, et al., Chromofungin ameliorates the progression of colitis by regulating alternatively activated macrophages, Front Immunol. 8 (2017) 1131. https://doi.org/10.3389/fimmu.2017.01131.

[10]

G.R. Lichtenstein, M.T. Abreu, R. Cohen, et al., American gastroenterological association institute technical review on corticosteroids, immunomodulators, and infliximab in inflammatory bowel disease, Gastroenterology 130 (2006) 940-987. http://doi.org/10.1053/j.gastro.2006.01.048.

[11]

D.C. Baumgart, W.J. Sandborn, Gastroenterology 2-inflammatory bowel disease: clinical aspects and established and evolving therapies, Lancet 369 (2007) 1641-1657. http://doi.org/10.1016/s0140-6736(07)60751-x.

[12]

S.J. Somani, K.P. Modi, A.S. Majumdar, et al., Phytochemicals and their potential usefulness in inflammatory bowel disease, Phytother. Res. 29 (2015) 339-350. http://doi.org/10.1002/ptr.5271.

[13]

K. Guo, J.A. Ren, G.S. Gu, et al., Hesperidin protects against intestinal inflammation by restoring intestinal barrier function and up-regulating Treg cells, Mol. Nutr. Food Res. 63 (2019) e1800975. http://doi.org/10.1002/mnfr.201800975.

[14]

Y.F. Kang, G. Yang, S.M. Zhang, et al., Goji berry modulates gut microbiota and alleviates colitis in IL-10-deficient mice, Mol. Nutr. Food Res. 62 (2018) 1800535. http://doi.org/10.1002/mnfr.201800535.

[15]

N.V. Matusheski, J.A. Juvik, E.H. Jeffery, Heating decreases epithiospecifier protein activity and increases sulforaphane formation in broccoli, Phytochemistry 65 (2004) 1273-1281. https://doi.org/10.1016/j.phytochem.2004.04.013.

[16]

B. Abbaoui, C.R. Lucas, K.M. Riedl, et al., Cruciferous vegetables, isothiocyanates, and bladder cancer prevention, Mol. Nutr. Food Res. 62 (2018) 1800079. http://doi.org/10.1002/mnfr.201800079.

[17]

K.L. Cheung, A.N. Kong, Molecular targets of dietary phenethyl isothiocyanate and sulforaphane for cancer chemoprevention, AAPS J. 12 (2010) 87-97. https://doi.org/10.1208/s12248-009-9162-8.

[18]

A. Yanaka, J.W. Fahey, A. Fukumoto, et al., Dietary sulforaphane-rich broccoli sprouts reduce colonization and attenuate gastritis in helicobacter pylori-infected mice and humans, Cancer Prev. Res. 2 (2009) 353-360. http://doi.org/10.1158/1940-6207.Capr-08-0192.

[19]

R.H. Brown, C. Reynolds, A. Brooker, et al., Sulforaphane improves the bronchoprotective response in asthmatics through Nrf2-mediated gene pathways, Resp. Res. 16 (2015) 106. http://doi.org/10.1186/s12931-015-0253-z.

[20]

E.S. Son, J.W. Park, Y.J. Kim, et al., Effects of antioxidants on oxidative stress and inflammatory responses of human bronchial epithelial cells exposed to particulate matter and cigarette smoke extract, Toxicol. In Vitro. 67 (2020) 104883. http://doi.org/10.1016/j.tiv.2020.104883.

[21]

R.T. Ruhee, K. Suzuki, The integrative role of sulforaphane in preventing inflammation, oxidative stress and fatigue: a review of a potential protective phytochemical, Antioxidants 9 (2020) 521. http://doi.org/10.3390/antiox9060521.

[22]

C.E. Guerrero-Beltran, P. Mukhopadhyay, B. Horvath, et al., Sulforaphane, a natural constituent of broccoli, prevents cell death and inflammation in nephropathy, J. Nutr. Biochem. 23 (2012) 494-500. http://doi.org/10.1016/j.jnutbio.2011.02.004.

[23]

H.R. Liu, X.Q. Yang, K. Tang, et al., Sulforaphane elicts dual therapeutic effects on renal inflammatory injury and crystal deposition in calcium oxalate nephrocalcinosis, Theranostics 10 (2020) 7319-7334. http://doi.org/10.7150/thno.44054.

[24]

V. Hernandez-Rabaza, A. Cabrera-Pastor, L. Taoro-Gonzalez, et al., Neuroinflammation increases GABAergic tone and impairs cognitive and motor function in hyperammonemia by increasing GAT-3 membrane expression. Reversal by sulforaphane by promoting M2 polarization of microglia, J. Neuroinflamm. 13 (2016) 83. http://doi.org/10.1186/s12974-016-0549-z.

[25]

S. Pal, V.B. Konkimalla, Sulforaphane regulates phenotypic and functional switching of both induced and spontaneously differentiating human monocytes, Int. Immunopharmacol. 35 (2016) 85-98. http://doi.org/10.1016/j.intimp.2016.03.008.

[26]

Y.L. Lin, X.G. Yang, W.J. Yue, et al., Chemerin aggravates DSS-induced colitis by suppressing M2 macrophage polarization, Cell. Mol. Immunol. 11 (2014) 355-366. http://doi.org/10.1038/cmi.2014.15.

[27]

P. Stopfer, F. Obermeier, N. Dunger, et al., Blocking lymphotoxin-β receptor activation diminishes inflammation via reduced mucosal addressin cell adhesion molecule-1 (MAdCAM-1) expression and leucocyte margination in chronic DSS-induced colitis, Clin. Exp. Immunol. 136 (2004) 21-29. https://doi.org/10.1111/j.1365-2249.2004.02402.x.

[28]

J. Liu, J. Tang, X. Li, et al., Curdlan (alcaligenes faecalis)(1→3)-β-D-glucan oligosaccharides drive M1 phenotype polarization in murine bone marrow-derived macrophages via activation of MAPKs and NF-κB pathways, Molecules 24 (2019) 4251. http://doi.org/10.3390/molecules24234251.

[29]

A. Vossenkamper, C. Hundsrucker, K. Page, et al., A CD3-specific antibody reduces cytokine production and alters phosphoprotein profiles in intestinal tissues from patients with inflammatory bowel disease, Gastroenterology 147 (2014) 172-183. http://doi.org/10.1053/j.gastro.2014.03.049.

[30]

J.Q. Tang, H.M. Zhen, N.N. Wang, et al., Curdlan oligosaccharides having higher immunostimulatory activity than curdlan in mice treated with cyclophosphamide, Carbohyd. Polym. 207 (2019) 131-142. http://doi.org/10.1016/j.carbpol.2018.10.120.

[31]

S. Fujisaka, I. Usui, A. Bukhari, et al., Regulatory mechanisms for adipose tissue M1 and M2 macrophages in diet-Induced obese mice, Diabetes 58 (2009) 2574-2582. http://doi.org/10.2337/db08-1475.

[32]

K. Socała, D. Nieoczym, E. Kowalczuk-Vasilev, et al., Increased seizure susceptibility and other toxicity symptoms following acute sulforaphane treatment in mice, Toxicol. Appl. Pharm. 326 (2017) 43-53. https://doi.org/10.1016/j.taap.2017.04.010.

[33]

N. Petri, C. Tannergren, B. Holst, et al., Absorption/metabolism of sulforaphane and quercetin, and regulation of phase II enzymes, in human jejunum in vivo, Drug Metab. Dispos. 31 (2003) 805-813. https://doi.org/10.1124/dmd.31.6.805.

[34]

A.E. Wagner, O. Will, C. Sturm, et al., DSS-induced acute colitis in C57BL/6 mice is mitigated by sulforaphane pre-treatment, J. Nutr. Biochem. 24 (2013) 2085-2091. http://doi.org/10.1016/j.jnutbio.2013.07.009.

[35]

Y. Zhang, L.X. Tan, C. Li, et al., Sulforaphane alter the microbiota and mitigate colitis severity on mice ulcerative colitis induced by DSS, AMB Express. 10 (2020) 119. http://doi.org/10.1186/s13568-020-01053-z.

[36]

L.Y. Wei, J.J. Wang, L. Yan, et al., Sulforaphane attenuates 5-fluorouracil induced intestinal injury in mice, J. Funct. Food. 69 (2020) 103965. http://doi.org/10.1016/j.jff.2020.103965.

[37]

M. Ali, M. Bonay, V. Vanhee, et al., Comparative effectiveness of 4 natural and chemical activators of Nrf2 on inflammation, oxidative stress, macrophage polarization, and bactericidal activity in an in vitro macrophage infection model, PLoS One 15 (2020) e0234484. http://doi.org/10.1371/journal.pone.0234484.

[38]

R.T. Ruhee, S.H. Ma, K. Suzuki, Sulforaphane protects cells against lipopolysaccharide-stimulated inflammation in murine macrophages, Antioxidants 8 (2019) 577. http://doi.org/10.3390/antiox8120577.

[39]

J.W. Zhao, B.X. Li, S.Y. Sun, et al., Sulforaphane alleviates dextran sodium sulfate (DSS)-induced inflammation via switch mir-155/AHR mediated macrophage polarization, Gastroenterology 156 (2019) S491. https://doi.org/10.1016/S0016-5085(19)38090-4.

[40]

W. Zhu, Z.S. Jin, J.B. Yu, et al., Baicalin ameliorates experimental inflammatory bowel disease through polarization of macrophages to an M2 phenotype, Int. Immunopharmacol. 35 (2016) 119-126. http://doi.org/10.1016/j.intimp.2016.03.030.

[41]

Y.S. Zhu, X.Q. Li, J.Q. Chen, et al., The pentacyclic triterpene Lupeol switches M1 macrophages to M2 and ameliorates experimental inflammatory bowel disease, Int. Immunopharmacol. 30 (2016) 74-84. http://doi.org/10.1016/j.intimp.2015.11.031.

[42]

H.X. Wei, B.L. Wang, B.F. Li, IL-10 and IL-22 in mucosal immunity: driving protection and pathology, Front Immunol. 11 (2020) 1315. http://doi.org/10.3389/fimmu.2020.01315.

[43]

P. Xiao, H.L. Zhang, Y. Zhang, et al., Phosphatase Shp2 exacerbates intestinal inflammation by disrupting macrophage responsiveness to interleukin-10, J. Exp. Med. 216 (2019) 337-349. http://doi.org/10.1084/jem.20181198.

[44]

D.S. Shouval, J. Ouahed, A. Biswas, et al., Interleukin 10 receptor signaling: master regulator of intestinal mucosal homeostasis in mice and humans, Adv. Immunol. 122 (2014) 177-210. http://doi.org/10.1016/B978-0-12-800267-4.00005-5.

[45]

C. Niemand, A. Nimmesgern, S. Haan, et al., Activation of STAT3 by IL-6 and IL-10 in primary human macrophages is differentially modulated by suppressor of cytokine signaling 3, J. Immunol. 170 (2003) 3263-3272. http://doi.org/10.4049/jimmunol.170.6.3263.

[46]

R. Kuhn, J. Lohler, D. Rennick, et al., Interleukin-10-deficient mice develop chronic enterocolitis, Cell 75 (1993) 263-274. http://doi.org/10.1016/0092-8674(93)80068-p.

[47]

L. Steidler, W. Hans, L. Schotte, et al., Treatment of murine colitis by Lactococcus lactis secreting interleukin-10, Science 289 (2000) 1352-1355. http://doi.org/10.1126/science.289.5483.1352.

[48]

A.C. Cevey, F.N. Penas, C.D.A. Soto, et al., IL-10/STAT3/SOCS3 axis is involved in the anti-inflammatory effect of benznidazole, Front Immunol. 10 (2019) 1267. http://doi.org/10.3389/fimmu.2019.01267.

[49]

P. Hutchins, D. Diez, D. Miranda-Saavedra, The IL-10/STAT3-mediated anti-inflammatory response: recent developments and future challenges, Brief Funct. Genomics. 12 (2013) 489-498. http://doi.org/10.1093/bfgp/elt028.

[50]

S.W. Wang, Y.F. Bai, Y.Y. Weng, et al., Cinobufacini ameliorates dextran sulfate sodium-induced colitis in mice through inhibiting M1 macrophage polarization, J. Pharmacol. Exp. Ther. 368 (2019) 391-400. http://doi.org/10.1124/jpet.118.254516.

[51]

X. Zhang, F. Xu, L. Liu, et al., (+)-Borneol improves the efficacy of edaravone against DSS-induced colitis by promoting M2 macrophages polarization via JAK2-STAT3 signaling pathway, Int. Immunopharmacol. 53 (2017) 1-10. http://doi.org/10.1016/j.intimp.2017.10.002.

Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 16 September 2020
Revised: 29 October 2020
Accepted: 01 November 2020
Published: 11 September 2021
Issue date: January 2022

Copyright

© 2021 Beijing Academy of Food Sciences. Publishing services by Elsevier B.V. on behalf of KeAi Communications Co., Ltd.

Acknowledgements

Acknowledgements

We gratefully acknowledge the financial support provided by "Modern Food Processing, Food Storage, Transportation Technology, and Equipment" State Key Research and Development Plan (2017YFD0400204) and the National Science Foundation of China (31972091).

Rights and permissions

This is an open access article under the CC BY-NC-ND license (http://creativecommons.org/licenses/by-nc-nd/4.0/).

Return