Journal Home > Volume 12 , Issue 2

In this study, the in vitro digestion and fermentation of two intra-polysaccharide fractions (IPS1 and IPS2) from Paecilomyces cicadae TJJ1213 were investigated. The constituent monosaccharides of IPS1 and IPS2 were not changed after simulated saliva, gastric and small intestinal digestion. However, they can be hydrolyzed and utilized by gut microbiota, and short-chain fatty acids (SCFAs) level were increased after IPS1and IPS2 treatments. Furthermore, 16S rRNA sequencing analysis of fermentation samples were performed. Alpha-diversity, beta-diversity and taxonomic composition differences analysis revealed that IPS1 and IPS2 promoted the proliferation of beneficial bacteria and modulated the overall structure of gut microbiota. Taxonomic comparison analysis found that IPS1 increased the relative abundances of beneficial bacteria including Megamonas, Bifidobacterium and Lactobacillus, while IPS2 could increase the abundance of Bacteroides, Parabacteroides and Phascolarctobacterium. In addition, they can also decrease the levels of pathogenic bacteria containing Escherichia-Shigella, Klebsiella and Fusobacterium. These results indicated that IPS from Paecilomyces cicadae TJJ1213 could be used as potential candidates for new functional foods.


menu
Abstract
Full text
Outline
About this article

Artificial simulated saliva, gastric and intestinal digestion and fermentation in vitro by human gut microbiota of intrapolysaccharide from Paecilomyces cicadae TJJ1213

Show Author's information Juanjuan TianXiaomeng WangXueliang ZhangXiaohong ChenMingsheng DongXin RuiQiuqin ZhangMei JiangWei Li( )
College of Food Science and Technology, Nanjing Agricultural University, Nanjing 210095, China

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

Abstract

In this study, the in vitro digestion and fermentation of two intra-polysaccharide fractions (IPS1 and IPS2) from Paecilomyces cicadae TJJ1213 were investigated. The constituent monosaccharides of IPS1 and IPS2 were not changed after simulated saliva, gastric and small intestinal digestion. However, they can be hydrolyzed and utilized by gut microbiota, and short-chain fatty acids (SCFAs) level were increased after IPS1and IPS2 treatments. Furthermore, 16S rRNA sequencing analysis of fermentation samples were performed. Alpha-diversity, beta-diversity and taxonomic composition differences analysis revealed that IPS1 and IPS2 promoted the proliferation of beneficial bacteria and modulated the overall structure of gut microbiota. Taxonomic comparison analysis found that IPS1 increased the relative abundances of beneficial bacteria including Megamonas, Bifidobacterium and Lactobacillus, while IPS2 could increase the abundance of Bacteroides, Parabacteroides and Phascolarctobacterium. In addition, they can also decrease the levels of pathogenic bacteria containing Escherichia-Shigella, Klebsiella and Fusobacterium. These results indicated that IPS from Paecilomyces cicadae TJJ1213 could be used as potential candidates for new functional foods.

Keywords: Gut microbiota, Polysaccharide, Fermentation, Paecilomyces cicadae TJJ1213

References(61)

[1]

O. Paliy, H. Kenche, F. Abernathy, et al., High-throughput quantitative analysis of the human intestinal microbiota with a phylogenetic microarray, Appl. Environ. Microb. 75 (2009) 3572-3579. https://doi.org/10.1128/AEM.02764-08.

[2]

J. Qin, R. Li, J. Raes, et al., A human gut microbial gene catalogue established by metagenomic sequencing, Nat. 464 (2010) 59-65. https://doi.org/10.1038/nature08821.

[3]

P.B. Eckburg, E.M. Bik, C.N. Bernstein, et al., Diversity of the human intestinal microbial flora, Sci. 308 (2005) 1635-1638. https://doi.org/ 10.1126/science.1110591.

[4]

D. Li, P. Wang, P.P. Wang, et al., The gut microbiota: a treasure for human health, Biotechnol. Adv. 34 (2016) 1210-1224. https://doi.org/10.1016/j.biotechadv.2016.08.003.

[5]

A. Koh, F. de Vadder, P. Kovatcheva-Datchary, et al., From dietary fiber to host physiology: short-chain fatty acids as key bacterial metabolites, Cell, 165 (2016) 1332-1345. https://doi.org/10.1016/j.cell.2016.05.041.

[6]

R.E. Ley, P. Turnbaugh, S. Klein, et al., Human gut microbes associated with obesity. Nat. 444 (2006) 10220-11023. https://doi.org/10.1038/4441022a.

[7]

W.A. Walters, Z. Xu, R. Knight, Meta-analyses of human gut microbes associated with obesity and IBD. Febs Lett. 588 (2014) 4223-4233. https://doi.org/10.1016/j.febslet.2014.09.039.

[8]

E. Sherwin, T.G. Dinan, J.F. Cryan, Recent developments in understanding the role of the gut microbiota in brain health and disease, Ann. N. Y. Acad. 1420 (2018) 1-21. https://doi.org/10.1111/nyas.13416.

[9]

C.S. Oriach, R.C. Robertson, C. Stanton, et al., Food for thought: The role of nutrition in the microbiota-gut–brain axis, Clin. Nutr. Experiment. 6 (2016) 25-38. https://doi.org/10.1016/j.yclnex.2016.01.003.

[10]

C. Zhang, M. Zhang, S. Wang, et al., Interactions between gut microbiota, host genetics and diet relevant to development of metabolic syndromes in mice, ISME J. 4 (2010) 312-313. https://doi.org/10.1038/ismej.2009.112.

[11]

A.W. Walker, J. Ince, S.H. Duncan, et al., Dominant and diet-responsive groups of bacteria within the human colonic microbiota, ISME J. 5 (2011) 220-230. https://doi.org/10.1038/ismej.2010.118.

[12]

A.E. Kaoutari, F. Armougom, J.I. Gordon, et al., The abundance and variety of carbohydrate-active enzymes in the human gut microbiota, Nat. Rev. Microbiol. 11 (2013) 497-504. https://doi.org/10.1038/nrmicro3050.

[13]

W.Z. Tang, J.Z. Zhou, Q.D. Xu, et al., In vitro digestion and fermentation of released exopolysaccharides (r-EPS) from Lactobacillus delbrueckii ssp. bulgaricus SRFM-1, Carbohydr. Polym. 230 (2020) 115593. https://doi.org/10.1016/j.carbpol.2019.115593.

[14]

Y. Ding, Y. Yan, Y.J. Peng, et al., In vitro digestion under simulated saliva, gastric and small intestinal conditions and fermentation by human gut microbiota of polysaccharides from the fruits of Lycium barbarum, Int. J. Biol. Macromol. 125 (2019) 751-760. https://doi.org/10.1016/j.ijbiomac.2018.12.081.

[15]

Y.H. Mao, A.X. Song, L.Q. Li, et al., Effects of exopolysaccharide fractions with different molecular weights and compositions on fecal microflora during in vitro fermentation, Int. J. Biol. Macromol. 144 (2020) 76-84, https://doi.org/10.1016/j.ijbiomac.2019.12.072.

[16]

Y. Sun, E. Kmonickona, R. Han, et al., Comprehensive evaluation of wild Cordyceps cicadae from different geographical origins by TOPSIS method based on the macroscopic infrared spectroscopy (IR) fingerprint, Spectrochim. Acta. A, 214 (2019) 252-260. https://doi.org/10.1016/j.saa.2019.02.031.

[17]

Y.F. Sun, M. Wink, P. Wang, et al., Biological characteristics, bioactive components and antineoplastic properties of sporoderm-broken spores from wild Cordyceps cicadae, Phytomedicine, 36 (2017) 217-228. https://doi.org/10.1016/j.phymed.2017.10.004.

[18]

L.F. He, W.J. Shi, X.C. Liu, et al., Anticancer action and mechanism of ergosterol peroxide from Paecilomyces cicadae fermentation broth, Int. J. Mol. Sci. 19 (2018) 3935. https://doi.org/10.3390/ijms19123935.

[19]

L. Li, T. Zhang, C.R. Li, et al., Potential therapeutic effects of Cordyceps cicadae and Paecilomyces cicadae on adenine-induced chronic renal failure in rats and their phytochemical analysis, Drug Des. Devel. Ther. 13 (2019) 103-117. https://doi.org/10.2147/DDDT.S180543.

[20]

J.Z. Yang, J. Zhuo, B.K. Chen, et al., Regulating effects of Paecilomyces cicadae polysaccharides on immunity of aged rats, China J. Chinese Materia Med. 33 (2008) 292-295.

[21]

J.J. Tian, C.P. Zhang, X.M. Wang, et al., Structural characterization and immunomodulatory activity of intracellular polysaccharide from the mycelium of Paecilomyces cicadae TJJ1213, Food Res. Int. (2021) 110515. https://doi.org/10.1016/j.foodres.2021.110515.

[22]

H. Liu, F. Gong, F. Wei, et al., Artificial simulation of salivary and gastrointestinal digestion, and fermentation by human fecal microbiota, of polysaccharides from Dendrobium aphyllum, RSC Adv. 8 (2018) 13954-13963. https://doi.org/10.1039/C8RA01179F.

[23]

G. Chen, M. Xie, P. Wan, et al., Digestion under saliva, simulated gastric and small intestinal conditions and fermentation in vitro by human intestinal microbiota of polysaccharides from Fuzhuan brick tea, Food Chem. 244 (2018) 331-339. https://doi.org/10.1016/j.foodchem.2017.10.074.

[24]

F. Huang, R.Y. Hong, Y. Yi, et al., In vitro digestion and human gut microbiota fermentation of longan pulp polysaccharides as affected by Lactobacillus fermentum fermentation, Int. J. Biol. Macro. 147 (2020) 363-368. https://doi.org/10.1016/j.ijbiomac.2020.01.059.

[25]

W. Li, K.Q. Wang, Y. Sun, et al., Influences of structures of galactooligosaccharides and fructooligosaccharides on the fermentation in vitro by human intestinal microbiota, J. Funct. Foods 13 (2015) 158-168. https://doi.org/10.1016/j.jff.2014.12.044.

[26]

Y. Su, L. Li, Structural characterization and antioxidant activity of polysaccharide from four auriculariales, Carbohydr. Polym. 229 (2020) 115407. https://doi.org/10.1016/j.carbpol.2019.115407.

[27]

Q.X. Yuan, Y.F. Xie, W. Wang, et al., Jabbar, Extraction optimization, characterization and antioxidant activity in vitro of polysaccharides from mulberry (Morus alba L.) leaves, Carbohydr. Polym. 128 (2015) 52-62. https://doi.org/10.1016/j.carbpol.2015.04.028.

[28]

S.J. Hur, B.O. Lim, E.A. Decker, et al., Mcclements, in vitro human digestion models for food applications, Food Chem. 125 (2011) 1-12. https://doi.org/10.1016/j.foodchem.2010.08.036.

[29]

J.L. Hu, S.P. Nie, C. Li, et al., In vitro fermentation of polysaccharide from the seeds of Plantago asiatica L. by human fecal microbiota, Food Hydrocoll. 33 (2013) 384-392. https://doi.org/10.1016/j.foodhyd.2013.04.006.

[30]

J.L. Hu, S.P. Nie, F.F. Min, et al, Artificial simulated saliva, gastric and intestinal digestion of polysaccharide from the seeds of Plantago asiatica L., Carbohydr. Polym. 92 (2013) 1143-1150, https://doi.org/10.1016/j.carbpol.2012.10.072.

[31]

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) 11619. https://doi.org/10.1016/j.carbpol.2020.116194.

[32]

Y.Q. Yuan, C. Li, Q.W. Zheng, et al., Effect of simulated gastrointestinal digestion in vitro on the antioxidant activity, molecular weight and microstructure of polysaccharides from a tropical sea cucumber (Holothuria leucospilota), Food Hydrocoll. 89 (2019) 735-741. https://doi.org/10.1016/j.foodhyd.2018.11.040.

[33]

L. Wang, C. Li, Q. Huang, et al., In vitro digestibility and prebiotic potential of a novel polysaccharide from Rosa roxburghii Tratt fruit, J. Funct. Foods 52 (2019) 408-417. https://doi.org/10.1016/j.jff.2018.11.021.

[34]

L. Payling, K. Fraser, S.M. Loveday, et al., The effects of carbohydrate structure on the composition and functionality of the human gut microbiota, Trends Food Sci. Tech. 97 (2020) 233-248. https://doi.org/10.1016/j.tifs.2020.01.009.

[35]

S.H. Duncan, P. Louis, J.M. Thomson, et al., The role of pH in determining the species composition of the human colonic microbiota, Environ. Microbiol. 11 (2009) 2112-2122. https://doi.org/10.1111/j.1462-2920.2009.01931.

[36]

S.L. Malhotra, Faecal urobilinogen levels and pH of stools in population groups with different incidence of cancer of the colon, and their possible role in its aetiology, J. R. Soc. Med. 75 (1982) 709-714.

[37]

G.T. Macfarlane, G.R. Gibson, J.H. Cummings, Comparison of fermentation reactions in different regions of the human colon, J. Appl. Bacterial. 72 (1992) 57-64. https://doi.org/10.1111/j.1365-2672.1992.tb04882.

[38]

J.K. Nicholson, E. Holmes, J. Kinross, et al., Host-gut microbiota metabolic interactions, Sci. 336 (2012) 1262-1267. https://doi.org/10.1126/science.1223813.

[39]

V.T. Pham, M.H. Mohajeri, The application of in vitro human intestinal models on the screening and development of pre-and probiotics, Benef. Microbes, 9 (2018) 725-742. https://doi.org/10.3920/BM2017.0164.

[40]

L.G. Chen, W. Xu, D. Chen, et al., Digestibility of sulfated polysaccharide from the brown seaweed Ascophyllum nodosum and its effect on the human gut microbiota in vitro, Int. J. Biol. Macro. 112 (2018) 1055-1061. https://doi.org/10.1016/j.ijbiomac.2018.01.183.

[41]

W.T. Zhou, Y.M. Yan, J. Mi, et al., Simulated digestion and fermentation in vitro by human gut microbiota of polysaccharides from bee collected pollen of Chinese wolfberry, J. Agric. Food Chem. 66 (2018) 898-907. https://doi.org/10.1021/acs.jafc.7b05546.

[42]

N.R. Shin, T.W. Whon, J.W. Bae, Proteobacteria: microbial signature of dysbiosis in gut microbiota, Trends Biotechnol. 33 (2015) 496-503. https://doi.org/10.1016/j.tibtech.2015.06.011.

[43]

H.M. Wexler, Bacteroides: the good, the bad, and the nitty-gritty, Clin. Microbiol. Rev. 20 (4) 593-621. https://doi.org/10.1128/CMR.00008-07.

[44]

G.D. Wu, J. Chen, C. Hoffmann, et al., Linking long-term dietary patterns with gut microbial enterotypes, Sci. 334 (2011) 105-108. https://doi.org/10.1126/science.1208344.

[45]

L. Li, Q. Su, B. Xie, et al., Gut microbes in correlation with mood: case study in a closed experimental human life support system, Neurogastroenterol. Moti. 28 (2016) 1233-1240. https://doi.org/10.1111/nmo.12822.

[46]

T.R. Wu, C.S. Lin, C.J. Chang, et al., Gut commensal parabacteroides goldsteinii plays a predominant role in the anti-obesity effects of polysaccharides isolated from Hirsutella sinensi, Gut, 68 (2019) 248-262, http://dx.doi.org/10.1136/gutjnl-2017-315458.

[47]

F.F. Wu, X.F. Guo, J.C. Zhang, et al., Phascolarctobacterium faecium abundant colonization in Human gastrointestinal tract, Experi. Ther. Med. 14 (2017) 3122-3126. https://doi.org/10.3892/etm.2017.4878.

[48]

J.P. Zackular, M.A. Rogers, M.T. Ruffin, et al., The human gut microbiome as a screening tool for colorectal cancer, Cancer Prev. Res. 7 (2014) 1112-1121. https://doi.org/10.1158/1940-6207.CAPR-14-0129.

[49]

B. Wang, Y.K. Mao, C. Diorio, et al., Bienenstock, Luminal administration ex vivo of a live Lactobacillus species moderates mouse jejunal motility within minutes, FASEB J. 24 (2010) 4078-4088. https://doi.org/10.1096/fj.09-153841.

[50]

S. Yamazaki, H. Kamimura, H. Momose, Protective effect of Bifidobacterium monoassociation against lethal activity of E. coli, Bifidobacteria Microflora, 1 (1982) 55-59. https://doi.org/10.12938/bifidus1982.1.1_55.

[51]

Y. Kohwi, K. Imai, Z. Tamura, et al., Antitumor effect of Bifidobacterium infantis in mice, Gan, 69 (1978) 613-618. https://doi.org/10.20772/cancersci1959.69.5_613.

[52]

S. Brisse, F. Grimont, P.A.D. Grimont, The genus Klebsiella, Prokaryotes, (2006) 159-196. https://doi.org/10.1007/0-387-30746-X_8.

[53]

J.D. Bronzato, R.A. Bomfim, D.H. Edwards, et al., Detection of Fusobacterium in oral and head and neck cancer samples: A systematic review and meta-analysis, Arch Oral Biol, 112 (2020) 104669. https://doi.org/10.1016/j.archoralbio.2020.104669.

[54]

M. Maaloum, B.A. Diop, A.S. Ndongo, et al., Draft genome sequence of Megamonas Funiformis strain marseille-p3344, isolated from a human fecal microbiota. Genome Announcem, 6 (2018) e01459-17. https://doi.org/10.1128/genomeA.01459-17.

[55]

M.X. Ying, Q. Yu, B. Zheng, Cultured Cordyceps Sinensis polysaccharides modulate intestinal mucosal immunity and gut microbiota in cyclophosphamide-treated mice. Carbohydr. Polym. 235 (2020) 115957. https://doi.org/10.1016/j.carbpol.2020.115957.

[56]

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.

[57]

M.C. Jonathan, J.J.G.C. Borne, P. Wiechen, et al., In vitro fermentation of 12 dietary fibres by faecal from pigs and humans, Food Chem. 133 (2012) 889-897, https://doi.org/10.1016/j.foodchem.2012.01.110.

[58]

B. Gullón, P. Gullón, F. Tavaria, et al., Structural features and assessment of prebiotic activity of refined arabinoxylooligosaccharides from wheat bran, J. Funct. Foods. 6 (2014) 438-449. https://doi.org/10.1016/j.jff.2013.11.010.

[59]

J. Yang, I. Martínez, J. Walter, et al., In vitro characterization of the impact of selected dietary fibers on fecal microbiota composition and short chain fatty acid production, Anaerobe, 23 (2013) 74-81. https://doi.org/10.1016/j.anaerobe.2013.06.012.

[60]

Y. Zhou, Y.H. Cui, X.J. Qu, Exopolysaccharides of lactic acid bacteria: structure, bioactivity and associations: a review, Carbohydr. Polym. 207 (2019) 317-332. https://doi.org/10.1016/j.carbpol.2018.11.093.

[61]

I. Henningsson, I. Bjiirck, M. Nyman, Short-chain fatty acid formation at fermentation of indigestible carbohydrates, Food Nutr. Res. 45 (2001) 165-168, https://doi.org/10.3402/fnr.v45i0.1801.

Publication history
Copyright
Acknowledgements
Rights and permissions

Publication history

Received: 29 June 2021
Revised: 20 July 2021
Accepted: 19 August 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.

Acknowledgements

Acknowledgements

This work was co-financed by National Natural Science Foundation of China (U1903108, 31871771 and 31571818), Natural Science Foundation of Jiangsu Province (BK20201320), Jiangsu Agriculture Science and Technology Innovation Fund (CX (20)3043), Postgraduate Research & Practice innovation Program of Jiangsu Province (KYCX19_0589), Qing Lan Project of Jiangsu Province and Priority Academic Program Development of Jiangsu Higher Education Institutions (PAPD).

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