Journal Home > Volume 12 , Issue 2

Low-molecular-weight polysaccharides (RLPs) extracted from Rosa laevigata fruits showed multiple biofunctions in Asia. This study aimed to investigate its anti-inflammatory obesity effect in high-fat diet-fed rats and further elucidated the underlying molecular mechanism by multi-omics methods. The results showed that RLPs administration had significantly restored immune organ indexes and reduced body weight gain. RNA-seq revealed that the effect of RLPs was partially attributed to its regulation on PPARs signaling by increasing the expressions of Scd, Acox3 and Hmgcs2, and on other redox-related pathways by decreasing the expressions of Cyp2e1, Il1-r1 and Lbp. Moreover, 16S rRNA sequencing coupled with metagenome sequencing showed that RLPs had significantly reduced the ratio of Firmicutes/Bacteroidetes from 8.01 to 2.37, and significantly increased the relative abundances of Alistipes, Prevotella, and Akkermansia from 0.36%, 1.10% and 2.61% to 0.65%, 2.37% and 4.42%, respectively. Spearman correlation analysis result indicated that the abundances of Lachnospiraceae, Prevotella and Bacteroidales were significantly negatively correlated with obesity phenotype, liver function and inflammatory factors. These results revealed that RLPs exerted significant anti-inflammatory obesity property partially via regulation on gut microbiota interface and the redox balance. Therefore, RLPs could be a promising functional food resource with the potential for redox imbalance-related diseases chemoprevention.


menu
Abstract
Full text
Outline
About this article

Polysaccharide extract from Rosa laevigata fruit attenuates inflammatory obesity by targeting redox balance and gut interface in high-fat diet-fed rats

Show Author's information Xuejiao Zhanga,bChenzhong JincHan LiucYihong HucYunyun ZhoucWeiguo Wua( )Si Qina( )
Lab of Food Function and Nutrigenomics, College of Food Science and Technology, Hunan Agricultural University, Changsha 410128, China
College of Food and Chemical Engineering, Shaoyang University, Shaoyang, 422000, China
Collaborative Innovation Center for Field Weeds Control, Hunan University of Humanities, Science and Technology, Loudi 417000, China

Abstract

Low-molecular-weight polysaccharides (RLPs) extracted from Rosa laevigata fruits showed multiple biofunctions in Asia. This study aimed to investigate its anti-inflammatory obesity effect in high-fat diet-fed rats and further elucidated the underlying molecular mechanism by multi-omics methods. The results showed that RLPs administration had significantly restored immune organ indexes and reduced body weight gain. RNA-seq revealed that the effect of RLPs was partially attributed to its regulation on PPARs signaling by increasing the expressions of Scd, Acox3 and Hmgcs2, and on other redox-related pathways by decreasing the expressions of Cyp2e1, Il1-r1 and Lbp. Moreover, 16S rRNA sequencing coupled with metagenome sequencing showed that RLPs had significantly reduced the ratio of Firmicutes/Bacteroidetes from 8.01 to 2.37, and significantly increased the relative abundances of Alistipes, Prevotella, and Akkermansia from 0.36%, 1.10% and 2.61% to 0.65%, 2.37% and 4.42%, respectively. Spearman correlation analysis result indicated that the abundances of Lachnospiraceae, Prevotella and Bacteroidales were significantly negatively correlated with obesity phenotype, liver function and inflammatory factors. These results revealed that RLPs exerted significant anti-inflammatory obesity property partially via regulation on gut microbiota interface and the redox balance. Therefore, RLPs could be a promising functional food resource with the potential for redox imbalance-related diseases chemoprevention.

Keywords: Gut microbiota, Redox, Multi-omics, Polysaccharides, Rosa laevigata

References(68)

[1]

P.W. Franks, M.I. Mccarthy, Exposing the exposures responsible for type 2 diabetes and obesity, Science 354 (2016) 69-73. https://doi.org/10.1126/science.aaf5094.

[2]

W. Liu, S.Q. Zhao, J.Q. Wang, et al., Grape seed proanthocyanin extract ameliorates inflammation and adiposity by modulating gut microbiota in high-fat diet mice, Mol. Nutr. Food Res. 61 (2017) 1-33. https://doi.org/10.1002/mnfr.201601082.

[3]

J.O. Hill, H.R. Wyatt, G.W. Reed, et al., Obesity and the environment: where do we go from here, Science 299 (2016) 853-855. https://doi.org/10.1126/science.1079857.

[4]

Q.D. Xiang, Q. Yu, H. Wang, et al., Immunomodulatory activity of Ganoderma atrum polysaccharide on purified T Lymphocytes through Ca2+/CaN and mitogen-activated protein kinase pathway based on RNA sequencing, J. Agric. Food Chem. 65 (2017) 5306-5315. https://doi.org/10.1021/acs.jafc.7b01763.

[5]

X. Zhang, N.F. Zhang, J. Kan, et al., Anti-inflammatory activity of alkali-soluble polysaccharides from Arctium lappa L. and its effect on gut microbiota of mice with inflammation, Int. J. Biol. Macromol. 154 (2020) 773-787. https://doi.org/10.1016/j.ijbiomac.2020.03.111.

[6]

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.

[7]

X. Zhang, M. Zhang, C.T. Ho, et al., Metagenomics analysis of gut microbiota modulatory effect of green tea polyphenols by high fat diet-induced obesity mice model, J. Funct. Foods. 46 (2018) 268-277. https://doi.org/10.1016/j.jff.2018.05.003.

[8]

A.J. Cox, N.P. West, A.W. Cripps, Obesity, inflammation, and the gut microbiota, Lancet Diabetes endo. 3 (2015) 207-215. https://doi.org/10.1016/S2213-8587(14)70134-2.

[9]

Y.T. Tian, Y.T. Zhao, H.L. Zeng, et al., Structural characterization of a novel neutral polysaccharide from Lentinus giganteus and its antitumor activity through inducing apoptosis, Carbohydr. Polym. 154 (2016) 231-240. https://doi.org/10.1016/j.carbpol.2016.08.059.

[10]

L.L. Chu, L.C. Yang, L.Z. Lin, et al., Chemical composition, antioxidant activities of polysaccharide from Pine needle (Pinus massoniana) and hypolipidemic effect in high-fat diet-induced mice, Int. J. Biol. Macromol. 125 (2019) 445-452. https://doi.org/10.1016/j.ijbiomac.2018.12.082.

[11]

C. Li, Q. Huang, X. Fu, et al., Characterization, antioxidant and immunomodulatory activities of polysaccharides from Prunella vulgaris Linn, Int. J. Biol. Macromol. 75 (2015) 298-305. https://doi.org/10.1016/j.ijbiomac.2015.01.010.

[12]

Y.C. Wang, M. Guan, X. Zhao, et al., Effects of garlic polysaccharide on alcoholic liver fibrosis and intestinal microflora in mice, Pharm. Biol. 56 (2018) 325-332. https://doi.org/10.1080/13880209.2018.1479868.

[13]

M.Y. Chen, D. Xiao, W. Liu, et al., Intake of Ganoderma lucidum polysaccharides reverses the disturbed gut microbiota and metabolism in type 2 diabetic rats, Int. J. Biol. Macromol. 155 (2020) 890-902. https://doi.org/10.1016/j.ijbiomac.2019.11.047.

[14]

Y.M. Jin, Y.X. Wang, X.B. Wang, et al., Structure characterization of a polysaccharide extracted from noni (Morinda citrifolia L.) and its protective effect against DSS-induced bowel disease in mice, Food hydrocolloid. 90 (2019) 189-197, https://doi.org/10.1016/j.foodhyd.2018.11.049.

[15]

L. Zhao, M.Y. Li, K.C. Sun, et al., Hippophae rhamnoides polysaccharides protect IPEC-J2 cells from LPS-induced inflammation, apoptosis and barrier dysfunction in vitro via inhibiting TLR4/NF-kappa B signaling pathway, Int. J. Biol. Macromol. 155 (2020) 1202-1215. https://doi.org/10.1016/j.ijbiomac.2019.11.088.

[16]

L.L. Shi, Y. Li, Y. Wang, et al., MDG-1, an Ophiopogon polysaccharide, regulate gut microbiota in high-fat diet-induced obese C57BL/6 mice, Int. J. Biol. Macromol. 81(2015) 576-583. https://doi.org/10.1016/j.ijbiomac.2015.08.057.

[17]

B.L. Li, J. Yuan, J.W. Wu, A review on the phytochemical and pharmacological properties of Rosa laevigata: a medicinal and edible plant, Chem. Pharm. Bull. 69 (2021) 421-431. https://doi.org/10.1248/cpb.c20-00743.

[18]

C.H. Yu, X.Y. Dai, Q. Chen, et al., Hypolipidemic and antioxidant activities of polysaccharides from Rosae laevigatae Fructus in rats, Carbohydr. Polym. 94 (2013) 56-62. https://doi.org/10.1016/j.carbpol.2013.01.006.

[19]

X. Li, W. Cao, Y. Shen, et al., Antioxidant compounds from Rosa laevigata fruits, Food Chem. 130(3) (2012) 575-580. https://doi.org/10.1016/j.foodchem.2011.07.076.

[20]

D.S. Dong, L.H. Yin, Y. Qi, et al., Protective effect of the total saponins from Rosa laevigata Michx fruit against carbon tetrachloride-induced liver fibrosis in rats, Nutrients. 7(6) (2015) 4829-4850. https://doi.org/10.3390/nu7064829.

[21]

Q. Zhan, Q. Wang, R. Lin, et al., Structural characterization and immunomodulatory activity of a novel acid polysaccharide isolated from the pulp of Rosa laevigata Michx fruit, Int. J. Biol. Macromol. 145 (2020) 1080-1090. https://doi.org/10.1016/j.ijbiomac.2019.09.201.

[22]

S. Zhang, Y. Qi, Y.W. Xu, et al., Protective effect of flavonoid-rich extract from Rosa laevigata Michx on cerebral ischemia-reperfusion injury through suppression of apoptosis and inflammation, Neurochem. Int. 63(5) (2013) 522-532. https://doi.org/10.1016/j.neuint.2013.08.008.

[23]

X.G. Liu, Y.X. Gao, D.Q. Li, et al., The neuroprotective and antioxidant profiles of selenium-containing polysaccharides from the fruit of Rosa laevigata, Food Funct., 9(2018)1800-1808. https://doi.org/10.1039/c7fo01725a.

[24]

X.J. Zhang, Y.H. Hu, C.Z. Jin, et al., Extraction and hypolipidemic activity of low molecular weight polysaccharides isolated from Rosa laevigata fruits, Biomed Res. Int. (2020)1-20. https://doi.org/10.1155/2020/2043785.

[25]

J.J. Zhang, Z.T. Song, Y. Li, et al., Structural analysis and biological effects of a neutral polysaccharide from the fruits of Rosa laevigata, Carbohydr. Polym. 265 (2021) 1-12. https://doi.org/10.1016/j.carbpol.2021.118080.

[26]

M. Mashmoul, A. Azlan, B.N.M. Yusof, et al., Effects of saffron extract and crocin on anthropometrical, nutritional and lipid profile parameters of rats fed a high fat diet, J. Funct. Foods. 8 (2014) 180-187, https://doi.org/10.1016/j.jff.2014.03.017.

[27]

B.L. Liu, Y.P. Chen, H. Cheng, et al., The protective effects of curcumin on obesity-related glomerulopathy are associated with inhibition of Wnt/β-catenin signaling activation in podocytes, Evid-Based Compl. Alt. (2015)1-12. https://doi.org/10.1155/2015/827472.

[28]

C. Tang, J. Sun, B. Zhou, et al., Effects of polysaccharides from purple sweet potatoes on immune response and gut microbiota composition in normal and cyclophosphamide treated mice, Food Funct. 9 (2018) 937-950. https://doi.org/https://doi.org/10.1039/c7fo01302g.

[29]

S.R. Gill, M. Pop, R.T. Deboy, et al., Metagenomic analysis of the human distal gut microbiome, Science. 312 (2006) 1355-1359. https://doi.org/10.1126/science.1124234.

[30]

X.L. Song, Z.Z. Ren, C. Zhang, Antioxidant, anti-inflammatory and renoprotective effects of acidic-hydrolytic polysaccharides by spent mushroom compost (Lentinula edodes) on LPS-induced kindey injury, Int. J. Biol. Macromol. (2019)1-38. https://doi.org/10.1016/j.ijbiomac.2019.10.173.

[31]

Y. Wang, N.F. Zhang, J. Kan, et al., Structural characterization of water-soluble polysaccharide from Arctium lappa and its effects on colitis mice, Carbohydr. Polym. 213 (2019) 89-99. https://doi.org/10.1016/j.carbpol.2019.02.090.

[32]

T.R. Mosmann, R.L. Coffman, Th1-cell and Th2-cell-different patterns of lymphokine secretion lead to different functional properties, Ann. Rev. Immunol. 7 (1989) 145-173. https://doi.org/10.1146/annurev.iy.07.040189.001045.

[33]

T. Varga, Z. Czimmerer, L. Nagy, PPARs are a unique set of fatty acid regulated transcription factors controlling both lipid metabolism and inflammation, BBA- Bioenergetics. 1812 (2011) 1007-1002. https://doi.org/10.1016/j.bbadis.2011.02.014.

[34]

E. Albano, E. Mottaran, G. Occhino, et al., Review article: role of oxidative stress in the progression of non-alcoholic steatosis, Aliment. Pharmacol. Ther. 22 (Suppl.2) (2005) 71-73. https://doi.org/10.1111/j.1365-2036.2005.02601.x.

[35]

X.Q. Cao, Y.H. Han, M. Gu, et al., Foodborne titanium dioxide nanoparticles induce stronger adverse effects in obese mice than non-obese mice: gut microbiota dysbiosis, colonic inflammation, and proteome alterations, Small. 16(36) (2020), https://doi.org/10.1002/smll.2001858.

[36]

S. Shao, D.D. Wang, W. Zheng, et al., A unique polysaccharide from Hericium erinaceus mycelium ameliorates acetic acid-induced ulcerative colitis rats by modulating the composition of the gut microbiota, short chain fatty acids levels and GPR41/43 respectors, Int. Immunopharmacol. 71 (2019) 411-422. https://doi.org/10.1016/j.intimp.2019.02.038.

[37]

I.D. Caterson, Medical management of obesity and its complications, Ann. Acad. Med. Singap. 38 (2009) 22-27.

[38]

N. Mauras, C. DelGiorno, C. Kollman, et al., Obesity without established comorbidities of the metabolic syndrome is associated with a proinflammatory and prothrombotic state, even before the onset of puberty in children, J. Clin. Endocrinol. Metab. 95 (2010) 1060-1068. https://doi.org/10.1210/jc.2009-1887.

[39]

E. Ho, K. Karimi Galougahi, C.C. Liu, et al., Biological markers of oxidative stress: applications to cardiovascular research and practice, Redox Biol. 1 (2013) 483-491, https://doi.org/10.1016/j.redox.2013.07.006.

[40]

Z. Zhang, Z.G. Zhou, Y. Li, et al., Isolated exopolysaccharides from Lactobacillus rhamnosus GG alleviated adipogenesis mediated by TLR2 in mice, Sci. Rep-UK. 6 (2016) 1-14. https://doi.org/10.1038/srep36083.

[41]

E. Bhakkiyalakshmi, D. Sireesh, P. Rajaguru, et al., The emerging role of redox-sensitive Nrf2-Keap1 pathway in diabetes, Pharmacol. Res. 91 (2015) 104-114. https://doi.org/10.1016/j.phrs.2014.10.004.

[42]

Y. Wang, Y. Liu, I. Kirpich, et al., Lactobacillus rhamnosus GG reduces hepatic TNF-α production and inflammation in chronic alcohol-induced liver injury, J. Nutr. Biochem. 24 (2013) 1609-1615. https://doi.org/10.1016/j.jnutbio.2013.02.001.

[43]

R. Yuan, X.Y. Tao, S. Liang, et al., Protective effect of acidic polysaccharide from Schisandra chinensis on acute ethanol-induced liver injury through reducing CYP2E1-dependent oxidative stress, Biomed. Pharmacother. 99 (2018) 537-542. https://doi.org/10.1016/j.biopha.2018.01.079.

[44]

V. Cécile, P. Gaëlle, D. Jocelyne, et al., Postprandial endotoxemia linked with chylomicrons and lipopolysaccharides handling in obese versus lean men: a lipid dose-effect trial, J. Clin. Endocrinol. Metab. 100 (2015) 3427-3435. https://doi.org/10.1210/JC.2015-2518.

[45]

R.K. Singh, H.W. Chang, D. Yan et al., Influence of diet on the gut microbiome and implications for human health. J. Transl. Med. 15 (1) (2017) 73-90. https://doi.org/10.1186/s12967-017-1175-y.

[46]

A.W. Walker, J.D. Sanderson, C. Churcher, et al., High-throughput clone library analysis of the mucosa-associated microbiota reveals dysbiosis and differences between inflamed and non-inflamed regions of the intestine in inflammatory bowel disease, BMC Microbiol. 11 (2011) 1-12. https://doi.org/10.1186/1471-2180-11-7.

[47]

W.X. Chen, L.H. Ren, R.H. Shi., Enteric microbiota leads to new therapeutic strategies for ulcerative colitis, World J. Gastroenterol. 20 (2014) 15657-15663. https://doi.org/10.3748/wjg.v20.i42.15657.

[48]

M.H. Do, Y.S. Seo, H.Y. Park, Polysaccharides: bowel health and gut microbiota, Crit. Rev. Food Sci. 5 (2020) 1-13. https://doi.org/10.1080/10408398.2020.1755949.

[49]

K.M. Maslowski, A.T. Vieira, A. Ng, et al., Regulation of inflammatory responses by gut microbiota and chemoattractant receptor GPR43, Nature. 461 (2009) 1282-1286. https://doi.org/10.1038/nature08530.

[50]

G. den Besten, K. van Eunen, A.K. Groen, et al., The role of short-chain fatty acids in the interplay between diet, gut microbiota, and host energy metabolism, J. Lipid Res. 54 (2013) 2325-2340. https://doi.org/10.1194/jlr.r036012.

[51]

C.H. Kim, Microbiota or short-chain fatty acids. Which regulates diabetes? Cell. Mol. Immunol. 15 (2018) 88-91. https://doi.org/10.1038/cmi.2017.57.

[52]

G. den Besten, K. Lange, R. Havinga, et al., Gut-derived short-chain fatty acids are vividly assimilated into host carbohydrates and lipids, Am. J. Physiol-Gastr. L. 305 (2013) 900-910. https://doi.org/10.1152/ajpgi.00265.2013.

[53]

H. Yan, K.M. Ajuwon, Mechanism of butyrate stimulation of triglyceride storage and adipokin expression during adipogenic differentiation of porcine stromovascular cells, PLoS One. 10 (2015) e0145940. https://doi.org/10.1371/journal.pone.0145940.

[54]

Y.M. Jia, J. Hong, H.F. Li, et al., Butyrate stimulates adipose lipolysis and mitochondrial oxidative phosphorylation through histone hyperacetylation-associated beta3-adrenergic receptor activation in high-fat diet-induced obese mice, Exp. Physiol. 102(2017) 273-281. https://doi.org/10.1113/EP086114.

[55]

R.S. Wright, J.W. Anderson, S.R. Bridges, Propionate inhibits hepatocyte lipid synthesis, Exp. Biol. Med. 195 (1990) 26-29. https://doi.org/10.3181/00379727-195-43113.

[56]

P.J. Turnbaugh, R.E. Ley, M.A. Mahowald, et al., An obesity-associated gut microbiome with increased capacity for energy harvest, Nature. 444 (2006) 1027-1031. https://doi.org/10.1038/nature05414.

[57]

M.C. Abt, P.T. McKenney, E.G. Pamer, Clostridium difficile colitis: pathogenesis and host defence. Nat. Rev. Microbiol. (2016):1-12. https://doi.org/10.1038/nrmicro.2016.108.

[58]

K. Kameyama, K. Itoh, Intestinal colonization by a Lachnospiraceae bacterium contributes to the development of diabetes in obese mice, Microbes Environ. 29 (2014) 427-430. https://doi.org/10.1264/jsme2.ME14054.

[59]

I. Moreno-Indias, L. Sánchez-Alcoholado, E. García-Fuentes, et al., Insulin resistance is associated with specific gut microbiota in appendix samples from morbidly obese patients, Am. J. Transl. Res. 8 (12) (2016) 5672-5684, https://www.ajtr.org/ISSN:1943-8141/AJTR0033702.

[60]

L. Zhao, Q. Zhang, W. Ma, et al., A combination of quercetin and resveratrol reduces obesity in high-fat diet-fed rats by modulation of gut microbiota, Food Funct. 8 (2017) 4644-4656. https://doi.org/10.1039/C7FO01383C.

[61]

F.F. Anhê, D. Roy, G. Pilon, et al., A polyphenol-rich cranberry extract protects from diet-induced obesity, insulin resistance and intestinal inflammation in association with increased Akkermansia spp. population in the gut microbiota of mice, Gut. 64 (2015) 872-883. https://doi.org/10.1136/gutjnl-2014-307142.

[62]

L.K. Brahe, E.L. Chatelier, E. Prifti, et al., Specific gut microbiota features and metabolic markers in postmenopausal women with obesity, Nutr. Diabetes. 5 (2015) e159. https://doi.org/10.1038/nutd.2015.9.

[63]

C. Matziouridou, N. Marungruang, T.D. Nguyen, et al., Lingonberries reduce atherosclerosis in Apoe(-/-) mice in association with altered gut microbiota composition and improved lipid profile, Mol. Nutr. Food Res. 60 (2016) 1150-1160. https://doi.org/10.1002/mnfr.201500738.

[64]

C. Haro, M. Montes-Borrego, O.A. Rangel-Zúñiga, et al., Two healthy diets modulate gut microbial community improving insulin sensitivity in a human obese population, J. Clin. Endocr. Metab. 101(2016) 233-242. https://doi.org/10.1210/jc.2015-3351.

[65]

A. Marisol, B.D.S. Carlota, V. Koen, et al., The gut microbiota from lean and obese subjects contribute differently to the fermentation of arabinogalactan and inulin, PLoS One. 11 (2016)1-18. https://doi.org/10.1371/journal.pone.0159236.

[66]

C. Carlberg, Nutrigenomics of vitamin D, Nutrients. 11(3) (2019) 676-690. https://doi.org/10.3390/nu11030676.

[67]

A.C. Peña-Romero, D. Navas-Carrillo, F. Marín, et al., The future of nutrition: nutrigenomics and nutrigenetics in obesity and cardiovascular diseases, Crit. Rev. Food Sci. 58 (2018) 3030-3041. https://doi.org/10.1080/10408398.2017.1349731.

[68]

C. Braicu, N. Mehterov, B. Vladimirov, et al., Nutrigenomics in cancer: revisiting the effects of natural compounds, Semin. Cancer Biol. 46 (2017) 84-106. http://doi.org/10.1016/j.semcancer.2017.06.011.

Publication history
Copyright
Rights and permissions

Publication history

Received: 16 July 2021
Revised: 16 July 2021
Accepted: 22 November 2021
Published: 07 September 2022
Issue date: March 2023

Copyright

© 2023 Beijing Academy of Food Sciences. Publishing services by Elsevier B.V. on behalf of KeAi Communications Co., Ltd.

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