Journal Home > Volume 12 , Issue 6

In order to effectively deliver lutein to the inflamed colon and better exert its pharmacological activity, this paper constructed a sodium alginate hydrogel-based delivery system loaded with lutein nanoparticles, evaluated the regulation on the expression and secretion of related inflammatory factors in mice with colitis, and its impact on intestinal microbial environment. The results showed that comparing lutein crystal and its nanoparticle, lutein hydrogel alleviated dextran sodium sulfate (DSS)-induced colitis in mice more effectively by adjusting fecal heme content, colon tissue damage, and inflammatory factor levels. Moreover, lutein hydrogel increased the expression of intestinal tight junction proteins zonula occluden-1 (ZO-1), claudin-1 and occludin to maintain the integrity of the intestinal-barrier, inhibited the nuclear factor-κB (NF-κB) pathway and reduced expression and secretion of inflammatory factors including tumour necrosis factor-α (TNF-α), inducible nitric oxide synthase (iNOS), NOD-like receptors 3 (NLRP3) and interleukin (IL)-1β. In addition, the intestinal microbial environment of mice with colitis was improved by down-regulating the relative abundance of Desulfovibrionaceae and up-regulating the relative abundance of Erysipelotrichaceae and Rikenellaceae. As a slow-release carrier to load lutein nanoparticles, sodium alginate-based hydrogel has potential application prospect.


menu
Abstract
Full text
Outline
About this article

Effect of sodium alginate-based hydrogel loaded with lutein on gut microbiota and inflammatory response in DSS-induced colitis mice

Show Author's information Pengxiang Xua,b,1Shuwei Luoa,b,1Jiangfeng Songa,b( )Zhuqing DaiaDajing LiaCai’e Wub
Institute of Agro-product Processing, Jiangsu Academy of Agricultural Sciences, Nanjing 210014, China
College of Light Industry and Food Engineering, Nanjing Forestry University, Nanjing 210037, China

1 The authors contributed equally to this article.

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

Abstract

In order to effectively deliver lutein to the inflamed colon and better exert its pharmacological activity, this paper constructed a sodium alginate hydrogel-based delivery system loaded with lutein nanoparticles, evaluated the regulation on the expression and secretion of related inflammatory factors in mice with colitis, and its impact on intestinal microbial environment. The results showed that comparing lutein crystal and its nanoparticle, lutein hydrogel alleviated dextran sodium sulfate (DSS)-induced colitis in mice more effectively by adjusting fecal heme content, colon tissue damage, and inflammatory factor levels. Moreover, lutein hydrogel increased the expression of intestinal tight junction proteins zonula occluden-1 (ZO-1), claudin-1 and occludin to maintain the integrity of the intestinal-barrier, inhibited the nuclear factor-κB (NF-κB) pathway and reduced expression and secretion of inflammatory factors including tumour necrosis factor-α (TNF-α), inducible nitric oxide synthase (iNOS), NOD-like receptors 3 (NLRP3) and interleukin (IL)-1β. In addition, the intestinal microbial environment of mice with colitis was improved by down-regulating the relative abundance of Desulfovibrionaceae and up-regulating the relative abundance of Erysipelotrichaceae and Rikenellaceae. As a slow-release carrier to load lutein nanoparticles, sodium alginate-based hydrogel has potential application prospect.

Keywords: Hydrogel, Intestinal microbiota, Colitis, Lutein, Inflammation regulation

References(41)

[1]

M.A. McGuckin, R. Eri, L.A. Simms, et al., Intestinal barrier dysfunction in inflammatory bowel diseases, Inflamm. Bowel. Dis. 15 (2009) 100-113. https://doi.org/10.1002/ibd.20539.

[2]

R.R. Mccarthy, F. O'gara, The impact of phytochemicals present in the diet on microbial signalling in the human gut, J. Funct. Foods 14 (2015) 684-691. https://doi.org/10.1016/j.jff.2015.02.032.

[3]

I.B. Jeffery, P.W. O'Toole, Ã. Lena, et al., An irritable bowel syndrome subtype defined by species-specific alterations in faecal microbiota, Gut. 61 (2012) 997-1006. https://doi.org/10.4161/gmic.21772.

[4]

M.C. Meurer, M. Mees, L.N.B. Mariano, et al., Hydroalcoholic extract of Tagetes erecta L. flowers, rich in the carotenoid lutein, attenuates inflammatory cytokine secretion and improves the oxidative stress in an animal model of ulcerative colitis, Nutr. Res. 66 (2019) 95-106. https://doi.org/10.1016/j.nutres.2019.03.005.

[5]

G. El-Akabawy, N.M. El-Sherif, Zeaxanthin exerts protective effects on acetic acid-induced colitis in rats via modulation of pro-inflammatory cytokines and oxidative stress, Biomed. Pharmacother. 111 (2019) 841-851. https://doi.org/10.1016/j.biopha.2019.01.001.

[6]

Y. Lyu, L. Wu, F. Wang, et al., Carotenoid supplementation and retinoic acid in immunoglobulin A regulation of the gut microbiota dysbiosis, Exp. Biol. Med. 243 (2018) 613-620. https://doi.org/10.1177/1535370218763760.

[7]

Y. Xu, X.Y. Ma, W. Gong, et al., Nanoparticles based on carboxymethylcellulose-modified rice protein for efficient delivery of lutein, Food Funct. 11 (2020) 2380-2394. https://doi.org/10.1039/C9FO02439E.

[8]

A.S. Awaad, R.M. El-Meligy, G.A. Soliman, Natural products in treatment of ulcerative colitis and peptic ulcer, J. Saudi Chem. Soc. 17 (2013) 101-124. https://doi.org/10.1016/j.jscs.2012.03.002.

[9]

M. Abbasi, M. Sohail, M.U. Minhas, et al., Novel biodegradable pH-sensitive hydrogels: an efficient controlled release system to manage ulcerative colitis, Int. J. Biol. Macromol. 136 (2019) 83-96. https://doi.org/10.1016/j.ijbiomac.2019.06.046.

[10]

M. George, T.E. Abraham, Polyionic hydrocolloids for the intestinal delivery of protein drugs: alginate and chitosan: a review, J. Control. Release 114 (2006) 1-14. https://doi.org/10.1016/j.jconrel.2006.04.017.

[11]

S. Uzun, H. Kim, C. Leal, et al., Ethanol-induced whey protein gels as carriers for lutein droplets, Food Hydrocolloid. 61 (2016) 426-432. https://doi.org/10.1016/j.foodhyd.2016.05.013.

[12]

J. Su, Q. Guo, Y. Chen, et al., Characterization and formation mechanism of lutein pickering emulsion gels stabilized by β-lactoglobulin-gum arabic composite colloidal nanoparticles, Food Hydrocolloid. 98 (2020) 105276. https://doi.org/10.1016/j.foodhyd.2019.105276.

[13]

C. Soukoulis, S. Cambier, L. Hoffmann, et al., Chemical stability and bioaccessibility of β-carotene encapsulated in sodium alginate o/w emulsions: impact of Ca2+ mediated gelation, Food Hydrocolloid. 57 (2016) 301-310. https://doi.org/10.1016/j.foodhyd.2016.02.001.

[14]

P. Xu, J. Song, Z. Dai, et al., Effect of Ca2+ cross-linking on the properties and structure of lutein-loaded sodium alginate hydrogels, Int. J. Biol. Macromol. 193 (2021) 53-63. https://doi.org/10.1016/j.ijbiomac.2021.10.114.

[15]

A. Jain, G. Sharma, G. Ghoshal, et al., Lycopene loaded whey protein isolate nanoparticles: an innovative endeavor for enhanced bioavailability of lycopene and anti-cancer activity, Int. J. Pharm. 546 (2018) 97-105. https://doi.org/10.1016/j.ijpharm.2018.04.061.

[16]

J. Yi, T.I. Lam, W. Yokoyama, et al., Beta-carotene encapsulated in food protein nanoparticles reduces peroxyl radical oxidation in Caco-2 cells, Food Hydrocolloid. 43 (2015) 31-40. https://doi.org/10.1016/j.foodhyd.2014.04.028.

[17]

E. Josef, M. Zilberman, H. Bianco-Peled, Composite alginate hydrogels: an innovative approach for the controlled release of hydrophobic drugs, Acta Biomater. 6 (2010) 4642-4649. https://doi.org/10.1016/j.actbio.2010.06.032.

[18]

S. Feng, M. Sui, D.Wang, et al., Pectin-zein based stigmasterol nanodispersions ameliorate dextran sulfate sodium-induced colitis in mice, Food Funct. 12 (2021) 11656-11670. https://doi.org/10.1039/D1FO02493K.

[19]

H. Zhao, N. Cheng, W. Zhou, et al., Honey polyphenols ameliorate DSS-induced ulcerative colitis via modulating gut microbiota in rats, Mol. Nutr. Food Res. 63 (2019) 1900638. https://doi.org/10.1002/mnfr.201900638.

[20]

X. Mao, X. Li, W. Zhang, et al., Development of microspheres based on thiol-modified sodium alginate for intestinal-targeted drug delivery, ACS Appl. Bio Mater. 2 (2019) 5810-5818.

[21]

A. Ya Sklyarov, N.B. Panasyuk, I.S., Fomenko, Role of nitric oxide-synthase and cyclooxygenase/lipooxygenase systems in development of experimental ulcerative colitis, J. Physiol. Pharmacol. 62 (2011) 65-73. https://doi.org/10.1152/jn.01060.2010.

[22]

X. Wang, F. Fan, Q. Cao, Modified Pulsatilla decoction attenuates oxazolone-induced colitis in mice through suppression of inflammation and epithelial barrier disruption, Mol. Med. Rep. 14 (2016) 1173-1179. https://doi.org/10.3892/mmr.2016.5358.

[23]

S. Lechuga, A.I. Ivanov, Disruption of the epithelial barrier during intestinal inflammation: quest for new molecules and mechanisms, Bba-Mol. Cell Biol. L. 1864 (2017) 1183-1194. https://doi.org/10.1016/j.bbamcr.2017.03.007.

[24]

Y. Tan, Y. Guan, Y. Sun, et al., Correlation of intestinal mucosal healing and tight junction protein expression in ulcerative colitis patients, Am. J. Med. Sci. 357 (2019) 195-204. https://doi.org/10.1016/j.amjms.2018.11.011.

[25]

W.N. D’Souza, J. Douangpanya, S. Mu, et al., Differing roles for short chain fatty acids and GPR43 agonism in the regulation of intestinal barrier function and immune responses, PLoS One 12 (2017) e0180190. https://doi.org/10.1371/journal.pone.0180190.

[26]

M. Kim, L. Friesen, J. Park, et al., Microbial metabolites, short-chain fatty acids, restrain tissue bacterial load, chronic inflammation, and associated cancer in the colon of mice, Eur. J. Immunol. 48 (2018) 1235-1247. https://doi.org/10.1002/eji.201747122.

[27]

W. Gao, C. Wang, L. Yu, et al., Chlorogenic acid attenuates dextran sodium sulfate-induced ulcerative colitis in mice through MAPK/ERK/JNK pathway, BioMed Res. Int. 2019 (2019) 1-13. https://doi.org/10.1155/2019/6769789.

[28]

A. Salminen, M. Lehtonen, T. Suuronen, et al., Terpenoids: natural inhibitors of NF-κB signaling with anti-inflammatory and anticancer potential, Cell. Mol. Life Sci. 65 (2008) 2979-2999. https://doi.org/10.1007/s00018-008-8103-5.

[29]

K. Ling, D. Mingzhao, Z. Chaopu, et al., The hypoglycemic, hypolipidemic, and anti-diabetic nephritic activities of zeaxanthin in diet-streptozotocin-induced diabetic sprague dawley rats, Appl. Biochem. Biotech. 182 (2017) 944-955. https://doi.org/10.1007/s12010-016-2372-5.

[30]

I. Soufli, R. Toumi, H. Rafa, et al., Overview of cytokines and nitric oxide involvement in immuno-pathogenesis of inflammatory bowel diseases, World J. Gastrointest. Pharmacol. Ther. 7 (2016) 353-360. https://doi.org/10.4292/wjgpt.v7.i3.353.

[31]

M.M. Rafi, Y. Shafaie, Dietary lutein modulates inducible nitric oxide synthase (iNOS) gene and protein expression in mouse macrophage cells (RAW 264.7), Mol. Nutr. Food Res. 51 (2007) 333-340. https://doi.org/10.1002/mnfr.200600170.

[32]

L. Xue, B. Lu, B. Gao, et al., NLRP3 promotes glioma cell proliferation and invasion via the interleukin-1β/NF-κB p65 signals, Oncol. Res. 27 (2019) 557-564. https://doi.org/10.3727/096504018X15264647024196.

[33]

I. Khan, N. Ullah, L. Zha, et al., Alteration of gut microbiota in inflammatory bowel disease (IBD): cause or consequence? IBD treatment targeting the gut microbiome, Pathogens. 8 (2019) 126. https://doi.org/10.3390/pathogens8030126.

[34]

T. Zuo, S.C. Ng, The gut microbiota in the pathogenesis and therapeutics of inflammatory bowel disease, Front. Microbiol. 9 (2018) 2247. https://doi.org/10.3389/fmicb.2018.02247.

[35]

W. Liu, S. Tang, Q. Zhao, et al., The α-D-glucan from marine fungus Phoma herbarum YS4108 ameliorated mice colitis by repairing mucosal barrier and maintaining intestinal homeostasis, Int. J. Biol. Macromol. 149 (2020) 1180-1188. https://doi.org/10.1016/j.ijbiomac.2020.01.303.

[36]

A. Andoh, H. Imaeda, T. Aomatsu, et al., Comparison of the fecal microbiota profiles between ulcerative colitis and Crohn’s disease using terminal restriction fragment length polymorphism analysis, J. Gastroenterol. 46 (2011) 479-486. https://doi.org/10.1007/s00535-010-0368-4.

[37]

J. Sun, H. Chen, J. Kan, et al., Anti-inflammatory properties and gut microbiota modulation of an alkali-soluble polysaccharide from purple sweet potato in DSS-induced colitis mice, Int. J. Biol. Macromol. 153 (2020) 708-722. https://doi.org/10.1016/j.ijbiomac.2020.03.053.

[38]

I.V. Kushkevych, Activity and kinetic properties of phosphotransacetylase from intestinal sulfate-reducing bacteria, Acta Biochim. Pol. 62 (2015) 103-108. https://doi.org/10.18388/abp.2014_845.

[39]

M.S. Attene-Ramos, E.D. Wagner, M.J. Plewa, et al., Evidence that hydrogen sulfide is a genotoxic agent, Mol. Cancer Res. 4 (2006) 9-14. https://doi.org/10.1158/1541-7786.MCR-05-0126.

[40]

A. Labbé, J.G. Ganopolsky, C.J. Martoni, et al., Bacterial bile metabolising gene abundance in Crohn's, ulcerative colitis and type 2 diabetes metagenomes, PLoS One 9 (2014) e115175. https://doi.org/10.1371/journal.pone.0115175.

[41]

X. Chen, Q. Zuo, Y. Hai, et al., Lactulose: an indirect antioxidant ameliorating inflammatory bowel disease by increasing hydrogen production, Med. Hypotheses 76 (2011) 325-327. https://doi.org/10.1016/j.mehy.2010.09.026.

Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 03 January 2022
Revised: 01 February 2022
Accepted: 28 March 2022
Published: 04 April 2023
Issue date: November 2023

Copyright

© 2023 Beijing Academy of Food Sciences.

Acknowledgements

Acknowledgements

This work was supported by the Independent Innovation Fund Project of Agricultural Science and Technology in Jiangsu Province (Project No. CX (20)3047).

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