AI Chat Paper
Note: Please note that the following content is generated by AMiner AI. SciOpen does not take any responsibility related to this content.
{{lang === 'zh_CN' ? '文章概述' : 'Summary'}}
{{lang === 'en_US' ? '中' : 'Eng'}}
Chat more with AI
PDF (4.7 MB)
Collect
Submit Manuscript AI Chat Paper
Show Outline
Outline
Show full outline
Hide outline
Outline
Show full outline
Hide outline
Research Article | Open Access

Adsorption, in vitro digestion and human gut microbiota regulation characteristics of three Poria cocos polysaccharides

Fangming Zhanga,1Hui Zhenga,1Tao Zhenga,Pan XuaYao XuaYuxin CaoaFan JiaaYiqiong ZengaYubing FanaKai Heb( )Xinwen DaicFengfei HoucYong Yanga( )
College of Pharmacy, Hunan University of Chinese Medicine, Changsha 410208, China
School of Pharmaceutical Science, Hunan University of Medicine, Huaihua 418000, China
Hunan Fuling Engineering Technology Research Center, Huaihua 418100, China

1 Co-first authors of the article.

Peer review under responsibility of Tsinghua University Press.

Show Author Information

Abstract

Poria cocos (PC) is a famous traditional Chinese medicine (TCM) and a widely used healthcare ingredient, which has antiobesity, enhancing immunity and improving sleep effects. Traditionally, only water-soluble poria polysaccharide (WSP) is extracted and applied for clinical application, while insoluble polysaccharide (alkali-soluble poria polysaccharide, ASP) is discarded as herb residue. However, the whole PC has also been historically utilized as functional herbal food. Considering the beneficial role of dietary fiber and the traditional use of PC, ASP may also contribute substantially to the therapy function of PC. Compared to WSP, little attention has been paid to ASP and ASP modified product carboxymethyl poria polysaccharide (CMP) which has been used as an antitumor adjuvant drug. In this study, the oil, cholesterol, metal ions and polyphenols adsorption ability, in vitro simulated digestive and the gut microbiota fermentation characteristics of WSP, ASP and CMP were studied to evaluate the functional values of three P. cocos polysaccharides (PCPs). The results showed that all three PCPs had good adsorption capacity on cholesterol, polyphenols and metal ions (Cd2+/Zn2+/Mg2+), among which ASP showed the highest capacity than WSP and CMP. The adsorption capacity of all three PCPs on heavy metal ions (Cd2+/Zn2+) was stronger than that of non-heavy metal ions (Mg2+); The in vitro digestibility of all three PCPs was very low, but WSP was slightly higher than ASP and CMP; Moreover, the indigestible residue of all three PCPs could improve the richness and diversity of gut microbiota, among which ASP had the greatest influence. In general, ASP and CMP could significantly promote the proliferation of some probiotics and inhibit the growth of some harmful bacteria. The gut microbiota diversity of CMP was reduced, but the richness of probiotics, especially Parabacteroides distasonis was significantly enhanced compared with the ASP group, and the growth of harmful bacteria Klebsiella pneumoniae was inhibited after CMP treatment. The short-chain fatty acids (SCFAs) analysis results showed that all three PCPs could significantly promote the production of acetic acid, propionic acid and the total acid content compared with blank control group, and SCFAs producing activity was positively correlated with the proliferative capacity of probiotics. Taken together, the good adsorption characteristics and gut microbiota regulatory activity of ASP may lay foundation for its lipid-lowering and immune-improving function. Additionally, the probiotic effect of CMP and ASP indicated that except for only use the water extract of PC in clinic, CMP and ASP also can be used in healthcare to take full advantage of this valuable medicine.

References

[1]
National Pharmacopoeia Commission (Ed.), Pharmacopoeia of the People’s Republic of China I, Beijing, China Medical Science and Technology Press, 2020, pp. 251-252.
[2]

J.J. Liu, W.Y. Hong, M. Li, et al., Transcriptome analysis reveals immune and metabolic regulation effects of Poria cocos polysaccharides on Bombyx mori larvae, Front. Immunol. 13 (2022) 1014985. https://doi.org/10.3389/fimmu.2022.1014985.

[3]

A. Singh, K. Zhao, Treatment of insomnia with traditional Chinese herbal medicine, Int. Rev. Neurobiol. 135 (2017) 97-115. https://doi.org/10.1016/bs.irn.2017.02.006.

[4]

L. Zhu, C. Ye, B.F. Hu, et al., Regulation of gut microbiota and intestinal metabolites by Poria cocos oligosaccharides improves glycolipid metabolism disturbance in high-fat diet-fed mice, J. Nutr. Biochem. 107 (2022) 109019. https://doi.org/10.1016/j.jnutbio.2022.109019.

[5]

M.H. Zhao, Z.Y. Guan, N. Tang, et al., The differences between the water- and alkaline-soluble Poria cocos polysaccharide: a review, Int. J. Biol. Macromol. 235 (2023) 123925. https://doi.org/10.1016/j.ijbiomac.2023.123925.

[6]

Y.T. Zou, J. Zhou, C.Y. Wu, et al., Protective effects of Poria cocos and its components against cisplatin-induced intestinal injury, J. Ethnopharmacol. 269 (2021) 113722. https://doi.org/10.1016/j.jep.2020.113722.

[7]

H.C. Xu, S.Q. Wang, Y.W. Jiang, et al., Poria cocos polysaccharide ameliorated antibiotic-associated diarrhea in mice via regulating the homeostasis of the gut microbiota and intestinal mucosal barrier, Int. J. Mol. Sci. 24 (2023) 1423. https://doi.org/10.3390/ijms24021423.

[8]

Y. Zhang, Extraction and purification of Poria cocos polysaccharides and its regulation on immune function and intestinal flora in rats with insufficiency of the spleen, Anhui University of Chinese Medicine (2020). https://doi.org/10.26922/d.cnki.ganzc.2020.000308.

[9]

S.S. Sun, K. Wang, K. Ma, et al., An insoluble polysaccharide from the sclerotium of Poria cocos improves hyperglycemia, hyperlipidemia and hepatic steatosis in ob/ob mice via modulation of gut microbiota, Chin. J. Nat. Med. 17 (2019) 3-14. https://doi.org/10.1016/S1875-5364(19)30003-2.

[10]

F. Liu, L.J. Zhang, X. Feng, et al., Immunomodulatory activity of carboxymethyl pachymaran on immunosuppressed mice induced by cyclophosphamide, Molecules 26 (2021) 5733. https://doi.org/10.3390/molecules26195733.

[11]

Y.H. Jiang, L. Wang, W.D. Chen, et al., Poria cocos polysaccharide prevents alcohol-induced hepatic injury and inflammation by repressing oxidative stress and gut leakiness, Front. Nutr. 9 (2022) 963598. https://doi.org/10.3389/fnut.2022.963598.

[12]
C.X. Chen, High substitution carboxymethyl poria polysaccharide and its preparation and application, Hunan Province: CN100509857C, 2009-07-08.
[13]

X.W. Liu, Study on the preparation and structure-activity relationships of carboxymethyl-pachymaran, South China University of Technology (2021). https://doi.org/10.27151/d.cnki.ghnlu.2021.001578.

[14]

J.Y. Si, C.R. Yang, W.J. Ma, et al., Screen of high efficiency cellulose degrading strains and effects on tea residues dietary fiber modification:structural properties and adsorption capacities, Int. J. Biol. Macromol. 220 (2022) 337-347. https://doi.org/10.1016/j.ijbiomac.2022.08.092.

[15]

L. Xin, K.K. Suo, P. Wang, et al., Modification of wheat bran insoluble and soluble dietary fibers with snail enzyme, Food Sci. Human Well. 10 (2021) 356-361. https://doi.org/10.1016/j.fshw.2021.02.027

[16]

J.J. Wu, Y, Ni, Y. Li, et al., Adsorption behavior and kinetics of different kinds of polyphenols by ginkgo husk cellulose, Food Ferment. Ind. 48 (2022) 121-128. https://doi.org/10.13995/j.cnki.11-1802/ts.028304.

[17]

Y.J. Huang, J.R. Wu, L. Zhu, et al., Adsorption of heavy metal by polysaccharide of Cyanobacteria from Taihu Lake, Chinese J. Bioprocess Eng. 17 (2019) 214-219. https://doi.org/CNKI:SUN:SWJG.0.2019-02-017.

[18]

A. Brodkorb, L. Egger, M. Alminger, et al., INFOGEST static in vitro simulation of gastrointestinal food digestion, Nat Protoc. 14 (2019) 991-1014. https://doi.org/10.1038/s41596-018-0119-1.

[19]

J.J. Tian, X.M. Wang, X.L. Zhang, et al., Artificial simulated saliva, gastric and intestinal digestion and fermentation in vitro by human gut microbiota of intrapolysaccharide from Paecilomyces cicadae TJJ1213, Food Sci. Human Well. 12 (2023) 622-633. https://doi.org/10.1016/j.fshw.2022.07.065.

[20]

C. Amorim, S.C. Silvério, B.B. Cardoso, et al., In vitro fermentation of raffinose to unravel its potential as prebiotic ingredient, LWT-Food Sci. Technol. 126 (2020) 109322. https://doi.org/10.1016/j.lwt.2020.109322.

[21]

Y.F. Wang, M. Zhang, D. Ruan, et al., Chemical components and molecular mass of six polysaccharides isolated from the sclerotium of Poria cocos, Carbohydr. Res. 339 (2004) 327-334. https://doi.org/10.1016/j.carres.2003.10.006.

[22]

D.D. Wang, C.G. Huang, Y. Zhao, et al., Comparative studies on polysaccharides, triterpenoids, and essential oil from fermented mycelia and cultivated sclerotium of a medicinal and edible mushroom, Poria cocos, Molecules 25 (2020) 1269. https://doi.org/10.3390/molecules25061269

[23]

M. Bie, B.J. Xie, Z.D. Sun, et al., Preparation, structural characterization and in vitro antibacterial activity of water-soluble carboxymethyl pachymaran with different degrees of substitution, Food Sci. 41 (2020) 67-76. https://doi.org/10.7506/spkx1002-6630-20190603-015.

[24]

X.Q. Yang, L.H. Li, B. Qi, Adsorption of Cd2+, Pb2+ and Hg2+ by dietary fibers from four seaweeds, Zhongguo Shui Chan Ke Xue 14 (2007) 132-138. https://doi.org/10.3321/j.issn:1005-8737.2007.01.019.

[25]

L.H. Li, S.L. Yang, B Qi, Effects of Eucheuma dietary fiber on adsorption of calcium, zinc and iron, Zhongguo Shui Chan Ke Xue 15 (2008) 323-329. https://doi.org/10.3321/j.issn:1005-8737.2008.02.017.

[26]

L.P. Zhang, B. Meng, L.Z. Li, et al., Boletus aereus protects against acute alcohol-induced liver damage in the C57BL/6 mouse via regulating the oxidative stress-mediated NF-κB pathway, Pharm. Biol. 58 (2020) 905-914. https://doi.org/10.1080/13880209.2020.1812672.

[27]

X.J. Li, R. Guo, X.J. Wu, et al., Dynamic digestion of tamarind seed polysaccharide: indigestibility in gastrointestinal simulations and gut microbiota changes in vitro, Carbohydr. Polym. 239 (2020) 116194. https://doi.org/10.1016/j.carbpol.2020.116194.

[28]

B.J. Callahan, P.J. McMurdie, M.J. Rosen, et al., DADA2: High-resolution sample inference from Illumina amplicon data, Nat. Methods 13 (2016) 581-583. https://doi.org/10.1038/nmeth.3869.

[29]

X. Yan, L.Z. Ai, Y.J. Xia, et al., Research progress on safety, probiotic potential and functional properties of Weisella confusa, Ind. Microbiol. 52 (2022) 37-48.

[30]

Y. Gao, M. Li, Y Sun, et al., Development and prospects of Lactococcus lactis expression system, Acta Microbiologica Sinica 62 (2022) 895-905. https://doi.org/10.13343/j.cnki.wsxb.20210341.

[31]

L.P. Zhang, K.N. Wang, Bacteroides thetaiotaomicron and its utilization of nutrients in host, Chinese J. Anim. Sci. 45 (2009) 57-61. https://doi.org/CNKI:SUN:ZGXM.0.2009-09-018.

[32]

W.J. Wu, J. Hu, H.Y. Gao, et al., The potential cholesterol-lowering and prebiotic effects of bamboo shoot dietary fibers and their structural characteristics, Food Chem. 332 (2020) 127372. https://doi.org/10.1016/j.foodchem.2020.127372.

[33]

C.H. Chen. A preliminary study on the mechanism of red chicory leaf dietary fiber alleviating colitis via the “intestinal barrier-gut microbiota” pathway, Nanchang University (2022). https://doi.org/10.27232/d.cnki.gnchu.2022.004153.

[34]

Y. Liu, X.L.J. Fu, H. Zhou, et al., Different types of non-starch polysaccharides alter the growth, intestinal flora and serum metabolite profile of grass carp, Ctenopharyngodon idella, Metabolites 12 (2022) 1003. https://doi.org/doi: 10.3390/metabo12101003.

[35]

E.C. Martens, N.M. Koropatkin, T.J. Smith, et al., Complex glycan catabolism by the human gut microbiota: the Bacteroidetes Sus-like paradigm, J. Biol. Chem. 284 (2009) 24673-24677. https://doi.org/10.1074/jbc.R109.022848.

[36]

G.D. Besten, K.V. 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.

[37]

E. Angelakis, F. Armougom, M. Million, et al., The relationship between gut microbiota and weight gain in humans, Future Microbiol. 7 (2012) 91-109. https://doi.org/10.2217/fmb.11.142.

[38]

X.N. Sun, M.M. Duan, Y.L. Liu, et al., The beneficial effects of Gracilaria lemaneiformis polysaccharides on obesity and the gut microbiota in high fat diet-fed mice, J. Funct. Foods 46 (2018) 48-56. https://doi.org/10.1016/j.jff.2018.04.041.

[39]

S.K. Mazmanian, J.L. Round, D.L. Kasper, A microbial symbiosis factor prevents intestinal inflammatory disease, Nature 453 (2008) 620-625. https://doi.org/10.1038/nature07008.

[40]

J. Li, F.Q. Zhao, Y.D. Wang, et al., Gut microbiota dysbiosis contributes to the development of hypertension, Microbiome 5 (2017) 14. https://doi.org/10.1186/s40168-016-0222-x.

[41]

R. Pittayanon, J.T. Lau, Y.H. Yuan, et al., Gut microbiota in patients with irritable bowel syndrome-a systematic review, Gastroenterology 157 (2019) 97-108. https://doi.org/10.1053/j.gastro.2019.03.049.

[42]

D.T. Wu, Y. Fu, H. Guo, et al., In vitro simulated digestion and fecal fermentation of polysaccharides from loquat leaves: Dynamic changes in physicochemical properties and impacts on human gut microbiota, Int. J. Biol. Macromol. 168 (2021) 733-742. https://doi.org/10.1016/j.ijbiomac.2020.11.130.

[43]

K. Wang, M.F. Liao, N. Zhou, et al., Parabacteroides distasonis Alleviates obesity and metabolic dysfunctions via production of succinate and secondary bile acids, Cell Rep. 26 (2019) 222-235.e5. https://doi.org/10.1016/j.celrep.2018.12.028.

[44]

P. Sun, J.Q. Wang, Y.M. Jiang, Effects of Enterococcus faecium (SF68) on immune function in mice, Food Chem. 123 (2010) 63-68. https://doi.org/10.1016/j.foodchem.2010.03.128.

[45]

K. Xia, Z.Q. Chen, L. Wang, Feeding effects of Enterococcus faeciumis and its mechanism of action, Cereal Feed Ind. 5 (2016) 44-47. https://doi.org/10.7633/j.issn.1003-6202.2016.05.011.

[46]

D.T. Wu, X.R. Nie, R.Y. Gan, et al., In vitro digestion and fecal fermentation behaviors of a pectic polysaccharide from okra (Abelmoschus esculentus) and its impacts on human gut microbiota, Food Hydrocoll. 114 (2021) 106577. https://doi.org/10.1016/j.foodhyd.2020.106577.

[47]

M. Snelson, N.J. Kellow, M.T. Coughlan, Modulation of the gut microbiota by resistant starch as a treatment of chronic kidney diseases: evidence of efficacy and mechanistic insights, Adv. Nutr. 10 (2019) 303-320. https://doi.org/10.1093/advances/nmy068.

[48]

F. Mahmoodpoor, Y.R. Saadat, A. Barzegari, et al., The impact of gut microbiota on kidney function and pathogenesis, Biomed. Pharmacother. 93 (2017) 412-419. https://doi.org/10.1016/j.biopha.2017.06.066.

[49]

J. Fernández, S. Redondo-Blanco, I. Gutiérrez-del-Río, et al., Colon microbiota fermentation of dietary prebiotics towards short-chain fatty acids and their roles as anti-inflammatory and antitumour agents: a review, J. Funct. Foods 25 (2016) 511-522. https://doi.org/10.1016/j.jff.2016.06.032.

[50]

L. Liang, Q. Bi, J.D. Wei, et al., Analytical progress of short-chain fatty acids in biological samples, Life Science Instruments 15 (2017) 9-14. https://doi.org/CNKI:SUN:SBKY.0.2017-05-002.

[51]

Y.Q. Xu, Y. Zhu, X.T. Li, et al., Dynamic balancing of intestinal short-chain fatty acids: the crucial role of bacterial metabolism, Trends Food Sci. Technol. 100 (2020) 118-130. https://doi.org/10.1016/j.tifs.2020.02.026.

[52]

G.Z. Mao, S. Li, C. Orfila, et al., Depolymerized RG-I-enriched pectin from citrus segment membranes modulates gut microbiota, increases SCFA production, and promotes the growth of Bifidobacterium spp., Lactobacillus spp. and Faecalibaculum spp., Food Funct. 10 (2019) 7828-7843. https://doi.org/10.1039/c9fo01534e.

Food Science and Human Wellness
Pages 1685-1697
Cite this article:
Zhang F, Zheng H, Zheng T, et al. Adsorption, in vitro digestion and human gut microbiota regulation characteristics of three Poria cocos polysaccharides. Food Science and Human Wellness, 2024, 13(3): 1685-1697. https://doi.org/10.26599/FSHW.2022.9250195

1111

Views

206

Downloads

1

Crossref

0

Web of Science

1

Scopus

0

CSCD

Altmetrics

Received: 11 May 2023
Revised: 05 June 2023
Accepted: 20 June 2023
Published: 08 February 2024
© 2024 Beijing Academy of Food Sciences. Publishing services by Tsinghua University Press.

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

Return