Genoprotective Properties and Metabolites of β-Glucan-Rich Edible Mushrooms Following Their In Vitro Fermentation by Human Faecal Microbiota

Athina Boulaka, Paraschos Christodoulou, Marigoula Vlassopoulou, Georgios Koutrotsios, Georgios Bekiaris, Georgios I Zervakis, Evdokia K Mitsou, Georgia Saxami, Adamantini Kyriacou, Maria Zervou, Panagiotis Georgiadis, Vasiliki Pletsa, Athina Boulaka, Paraschos Christodoulou, Marigoula Vlassopoulou, Georgios Koutrotsios, Georgios Bekiaris, Georgios I Zervakis, Evdokia K Mitsou, Georgia Saxami, Adamantini Kyriacou, Maria Zervou, Panagiotis Georgiadis, Vasiliki Pletsa

Abstract

A variety of bioactive compounds, constituents of edible mushrooms, in particular β-glucans, i.e., a group of β-d-glucose polysaccharides abundant in the fungal cell walls, have been linked to immunomodulating, anticancer and prebiotic activities. The aim of the study was the investigation of the genoprotective effects of edible mushrooms produced by Pleurotus eryngii, Pleurotus ostreatus and Cyclocybe cylindracea (Basidiomycota). Mushrooms from selected strains of the species mentioned above were fermented in vitro using faecal inocula from healthy volunteers. The cytotoxic and anti-genotoxic properties of the fermentation supernatants (FSs) were investigated in Caco-2 human colon adenocarcinoma cells. The FSs were cytotoxic in a dose-dependent manner. Non-cytotoxic concentrations were used for the genotoxicity studies, which revealed that mushrooms' FSs have the ability to protect Caco-2 cells against tert-butyl hydroperoxide (t-BOOH), a known genotoxic agent. Their global metabolic profiling was assessed by 1H-NMR spectroscopy. A total of 37 metabolites were identified with the use of two-dimensional (2D) homo- and hetero-nuclear NMR experiments. Multivariate data analysis monitored the metabolic variability of gut microbiota and probed to biomarkers potentially associated with the health-promoting effects of edible mushrooms.

Keywords: NMR-based metabolomics; edible mushrooms; faecal microbiota; genoprotection; in vitro fermentation; β-glucans.

Conflict of interest statement

The authors declare no conflict of interest. The funders had no role in the design of the study; in the collection, analyses, or interpretation of data; in the writing of the manuscript, or in the decision to publish the results.

Figures

Figure 1
Figure 1
Box plots depicting (a) total and (b) β-glucan content (%, dry weight [d.w.]) of mushroom species cultivated in various substrates. The horizontal line in each box represents the median, the x represents the mean, the rectangle represents the second and third quartile. An outlier (green dot) is shown among Hericium erinaceus cases.
Figure 2
Figure 2
Inhibition of cell proliferation by post-fermentation supernatants (FSs): FSs with different concentrations of the prebiotic inulin (0.5% w/v and 2% w/v) and lyophilized P. ostreatus 1123 powder (0.5% w/v, 1% w/v and 2% w/v), fermented by fecal inocula from one healthy donor were incubated with Caco-2 cells at 1%, 5% and 10% v/v for 48 (a) and 72 h (b). Cells treated with medium only (MD) served as control. NC: basal medium with no additional carbohydrate source. All values are expressed as the mean ± standard deviation (SD) of at least two independent experiments. * p < 0.05 versus control/non-treated cells. Associations were not significant unless otherwise indicated.
Figure 3
Figure 3
Genoprotective effect of fermentation supernatants (FSs): Fermentations were performed at increasing concentrations 0.5%, 1% and 2% w/v inulin or lyophilized P. ostreatus 1123 and with no additional carbohydrate source (NC) as a control. Caco-2 cells were incubated with NC, inulin (INU) and P. ostreatus (PO) FS at 1%, 5% and 10% v/v of culture medium for 48 h. The genotoxic agent tert-butyl hydroperoxide (BOOH) (500 μΜ) was added one hour prior to harvesting. Cells in medium (MD) and cells treated only with BOOH were used as negative and positive controls, respectively. All values are expressed as the mean ± SD of two independent experiments. * p < 0.05 versus NC. NC: basal medium with no additional carbohydrate source. Associations were not significant unless otherwise indicated.
Figure 4
Figure 4
Inhibition of cell proliferation by pre- and post-fermentation supernatants (FSs): Caco-2 cells were treated with 1%, 2.5% and 5% v/v of (a) pre-fermentation (t = 0 h) and (b) post-fermentation (t = 24 h) supernatants of selected mushrooms for 48 h. Cells treated with medium only served as control (MD). NC: basal medium with no additional carbohydrate source, INU: inulin, PO: P. ostreatus strain 1123, POL: P. ostreatus strain LGM 22, PE: P. eryngii strain LGAM 216, CC2: C. cylindracea strain CC2, CC505: C. cylindracea strain 505. Faecal inocula derived from 8 donors and the fermentation supernatants were pooled for this process. All values are expressed as the mean ± SD of at least two independent experiments. * p < 0.05 versus control/non-treated cells. Associations were not significant unless otherwise indicated.
Figure 5
Figure 5
Genotoxic and genoprotective effects of pre- and post-fermentation supernatants. Caco-2 cells were treated with 1% v/v pre- and post-fermentation supernatants of NC, INU, PO, POL, PE, CC2 and CC505 for 24 h. The genotoxic agent BOOH (500 μM) was added for one hour. (a) genotoxic effect of BOOH, NC 0 and NC 24 (% of BOOH), (b) genotoxic effect of pre-fermentation supernatants (% NC t = 0 h), (c) genoprotective effect of post-fermentation supernatants (% NC t = 24 h). NC: basal medium with no additional carbohydrate source, INU: inulin, PO: P. ostreatus strain 1123, POL: P. ostreatus strain LGM 22, PE: P. eryngii strain LGAM 216, CC2: C. cylindracea strain CC2, CC505: C. cylindracea strain CC505. All values are expressed as the mean ± 95% CI (Confidence Interval). * p < 0.05 versus control (paired t-test). Associations were not significant unless otherwise indicated.
Figure 6
Figure 6
Direct genotoxic effects of pre- and post-fermentation samples: Caco-2 cells were treated with 1% v/v pre- and post-fermentation samples of NC, INU, PO, POL, PE, CC2 and CC505. Cells treated with culture medium only served as control (MD). NC: basal medium with no additional carbohydrate source, INU: inulin, PO: P. ostreatus strain 1123, POL: P. ostreatus strain LGM 22, PE: P. eryngii strain LGAM 216, CC2: C. cylindracea strain CC2, CC505: C. cylindracea strain CC505. All values are expressed as the mean ± SD of two independent experiments. * p < 0.05 versus medium. Pooled samples from 8 donors were used. Associations were not significant unless otherwise indicated.
Figure 7
Figure 7
Representative 1H-NMR spectra of the pre- (t = 0 h) and post-fermentation (t = 24 h) supernatants from one volunteer, for both NC (basal medium with no additional carbohydrate source) and C. cylindracea CC505 (a) 0.8–4.4 ppm, and (b) 4.5–9.0 ppm. NC: basal medium with no additional carbohydrate source, CC505: C. cylindracea strain 505.
Figure 8
Figure 8
Illustration of multivariate statistical analysis of the 1H-NMR data for the pre- and post-fermentation samples from 3 randomly selected volunteers. (a) Orthogonal projections to latent structures with discriminant analysis (OPLS-DA) scores map. (R2X(cum) = 0.68, (R2X(cum) = 0.92, Q2(cum) = 0.90, Pareto scaling, Hotelling T2 = 95%). (b) Validation of the OPLS-DA model by permutation analysis indicating that the extracted model is significantly different from a model built on random data. The permutation tests were carried out with 999 random permutations, thus providing significance of the model at the 0.05 level. PO: P. ostreatus strain 1123, POL: P. ostreatus strain LGM 22, PE: P. eryngii strain LGAM 216, CC2: C. cylindracea strain CC2, CC505: C. cylindracea strain CC505.
Figure 9
Figure 9
S-line plot based on the OPLS-DA model in order to visualise the most discriminative metabolites. The colour coding corresponds to the significance of their contribution with the red colour depicting the metabolites that most influence the separation of the groups. Resonance lines with positive values correspond to the most characteristic metabolites for the post-fermentation supernatants, and those with negative values constitute the most discriminative for the pre-fermented supernatants. PO: P. ostreatus strain 1123, POL: P. ostreatus strain LGM 22, PE: P. eryngii strain LGAM 216, CC2: C. cylindracea strain CC2, CC505: C. cylindracea strain CC505.

References

    1. Gargano M.L., van Griensven L.J., Isikhuemhen O.S., Lindequist U., Venturella G., Wasser S.P., Zervakis G.I. Medicinal mushrooms: Valuable biological resources of high exploitation potential. Plant Biosyst. 2017;151:548–565. doi: 10.1080/11263504.2017.1301590.
    1. Xie J.H., Jin M.L., Morris G.A., Zha X.Q., Chen H.Q., Yi Y., Li J.E., Wang Z.J., Gao J., Nie S.P., et al. Advances on Bioactive Polysaccharides from Medicinal Plants. Crit. Rev. Food Sci. Nutr. 2016;56(Suppl. 1):S60–S84. doi: 10.1080/10408398.2015.1069255.
    1. Giavasis I. Bioactive fungal polysaccharides as potential functional ingredients in food and nutraceuticals. Curr. Opin. Biotechnol. 2014;26:162–173. doi: 10.1016/j.copbio.2014.01.010.
    1. Ooi V.E., Liu F. Immunomodulation and anticancer activity of polysaccharide-protein complexes. Curr. Med. Chem. 2000;7:715–729. doi: 10.2174/0929867003374705.
    1. Wasser S.P., Weis A.L. Therapeutic effects of substances occurring in higher Basidiomycetes mushrooms: A modern perspective. Crit. Rev. Immunol. 1999;19:65–96.
    1. Laroche C., Michaud P. New developments and prospective applications for beta (1,3) glucans. Recent Pat. Biotechnol. 2007;1:59–73. doi: 10.2174/187220807779813938.
    1. Du B., Meenu M., Liu H., Xu B. A Concise Review on the Molecular Structure and Function Relationship of β-Glucan. Int. J. Mol. Sci. 2019;20:4032. doi: 10.3390/ijms20164032.
    1. Lazaridou A., Biliaderis C.G. Molecular aspects of cereal b-glucan functionality: Physical properties, technological applications and physiological effects. J. Cereal Sci. 2007;46:101–118. doi: 10.1016/j.jcs.2007.05.003.
    1. FDA Food labeling: Health claims; oats and coronary heart disease. Fed. Regist. 1997;62:3584–3601.
    1. EFSA Scientific Opinion on the substantiation of health claims related to beta-glucans and maintenance of normal blood cholesterol concentrations (ID 754, 755, 757, 801, 1465, 2934) and maintenance or achievement of a normal body weight (ID 820, 823) pursuant to Article 13(1) of Regulation (EC) No. 1924/2006. EFSA J. 2009;7:1254.
    1. Kim J., Lee S.M., Bae I.Y., Park H.G., Lee H.G., Lee S. (1-3)(1-6)-β-Glucan-enriched materials from Lentinus edodes mushroom as a high-fibre and low-calorie flour substitute for baked foods. J. Sci. Food Agric. 2011;91:1915–1919. doi: 10.1002/jsfa.4409.
    1. Nakashima A., Yamada K., Iwata O., Sugimoto R., Atsuji K., Ogawa T., Ishibashi-Ohgo N., Suzuki K. β-Glucan in Foods and Its Physiological Functions. J. Nutr. Sci. Vitaminol. 2018;64:8–17. doi: 10.3177/jnsv.64.8.
    1. Koutrotsios G., Kalogeropoulos N., Kaliora A., Zervakis G.I. Toward an increased functionality in oyster (Pleurotus) mushrooms produced on grape marc or olive mill wastes serving as sources of bioactive compounds. J. Agric. Food Chem. 2018;66:5971–5983. doi: 10.1021/acs.jafc.8b01532.
    1. Mitsou E.K., Panopoulou N., Turunen K., Spiliotis V., Kyriacou A. Prebiotic potential of barley-derived β-glucan at low intake levels: A randomized, double-blinded, placebo-controlled clinical study. Food Res. Int. 2010;43:1086–1092. doi: 10.1016/j.foodres.2010.01.020.
    1. Human Microbiome Project Consortium Structure, function and diversity of the healthy human microbiome. Nature. 2012;486:207–214. doi: 10.1038/nature11234.
    1. Jayachandran M., Xiao J., Xu B.A. Critical Review on Health Promoting Benefits of Edible Mushrooms through Gut Microbiota. Int. J. Mol. Sci. 2017;18:1934. doi: 10.3390/ijms18091934.
    1. Zimmermann C.E., Cruz I.B., Cadoná F.C., Machado A.K., Assmann C., Schlemmer K.B., Zanette R.A., Leal D.B.R., Santurio J.M. Cytoprotective and genoprotective effects of β-glucans against aflatoxin B₁-induced DNA damage in broiler chicken lymphocytes. Toxicol. In Vitro. 2015;29:538–543. doi: 10.1016/j.tiv.2015.01.005.
    1. Kerche-Silva L.E., Cólus I.M., Malini M., Mori M.P., Dekker R.F., Barbosa-Dekker A.M. In vitro protective effects of botryosphaeran, a (1→3;1→6)-β-d-glucan, against mutagens in normal and tumor rodent cells. Mutat. Res. 2017;814:29–36. doi: 10.1016/j.mrgentox.2016.12.003.
    1. Madrigal-Bujaidar E., Morales-González J.A., Sánchez-Gutiérrez M., Izquierdo-Vega J.A., Reyes-Arellano A., Álvarez-González I., Pérez-Pasten R., Madrigal-Santillán E. Prevention of Aflatoxin B₁-Induced DNA Breaks by β-d-Glucan. Toxins. 2015;7:2145–2158. doi: 10.3390/toxins7062145.
    1. Tohamy A.A., El-Ghor A.A., El-Nahas S.M., Noshy M.M. Beta-glucan inhibits the genotoxicity of cyclophosphamide, adriamycin and cisplatin. Mutat. Res. 2003;541:45–53. doi: 10.1016/S1383-5718(03)00184-0.
    1. Silva-Sena G.G., Malini M., Delarmelina J.M., Dutra J.C.V., Gervásio S.V., Leal M.A.S., Costa Pereira T.M., Barbosa-Dekker A.M., Dekker R.F.H., de Paula F., et al. In vivo antimutagenic and antiatherogenic effects of the (1 → 3)(1 → 6)-β-d-glucan botryosphaeran. Mutat. Res. Genet. Toxicol. Environ. Mutagen. 2018;826:6–14. doi: 10.1016/j.mrgentox.2017.12.008.
    1. De Souza Silva P.M., de Sousa R.V., Simão A.A., Cesar P.H.S., Trento M.V.C., Marcussi S. Protective effect of β-d-glucan and glutamine on the genomic instability induced by Cytarabine/Ara-C in BALB/c mice. Int. J. Biol. Macromol. 2018;117:559–564. doi: 10.1016/j.ijbiomac.2018.05.206.
    1. Turunen K.T., Pletsa V., Georgiadis P., Triantafillidis J.K., Karamanolis D., Kyriacou A. Impact of β-glucan on the Fecal Water Genotoxicity of Polypectomized Patients. Nutr. Cancer. 2016;68:560–567. doi: 10.1080/01635581.2016.1156713.
    1. Nowak A., Śliżewska K., Otlewska A. Antigenotoxic activity of lactic acid bacteria, prebiotics, and products of their fermentation against selected mutagens. Regul. Toxicol. Pharmacol. 2015;73:938–946. doi: 10.1016/j.yrtph.2015.09.021.
    1. Christophersen C.T., Petersen A., Licht T.R., Conlon M.A. Xylo-oligosaccharides and inulin affect genotoxicity and bacterial populations differently in a human colonic simulator challenged with soy protein. Nutrients. 2013;5:3740–3756. doi: 10.3390/nu5093740.
    1. Allshop P., Possemiers S., Campbell D., Oyarzábal I.S., Gill C., Rowland I. An exploratory study into the putative prebiotic activity of fructans isolated from Agave angustifolia and the associated anticancer activity. Anaerobe. 2013;22:38–44. doi: 10.1016/j.anaerobe.2013.05.006.
    1. Burns A.J., Rowland I.R. Antigenotoxicity of probiotics and prebiotics on faecal water-induced DNA damage in human colon adenocarcinoma cells. Mutat. Res. 2004;551:233–243. doi: 10.1016/j.mrfmmm.2004.03.010.
    1. Munjal U., Scharlau D., Glei M. Gut fermentation products of inulin-type fructans modulate the expression of xenobiotic-metabolizing enzymes in human colonic tumour cells. Anticancer Res. 2012;32:5379–5386.
    1. Markovina N., Banjari I., Bucevic Popovic V., Jelicic Kadic A., Puljak L. Efficacy and safety of oral and inhalation commercial beta-glucan products: Systematic review of randomized controlled trials. Clin. Nutr. 2020;39:40–48. doi: 10.1016/j.clnu.2019.01.003.
    1. Polemis E., Zervakis G.I. Mushrooms in Greece. Present status and threats; Proceedings of the COST Action FP1203 European Non-Wood Forest Products (NWFPs) Network: Workshop on Mushroom (Including Truffles) Regulating Policies; Ioannina, Greece. 20 April 2016.
    1. Ntougias S., Baldrian P., Ehaliotis C., Nerud F., Merhautová V., Zervakis G.I. Olive mill wastewater biodegradation potential of white-rot fungi-Mode of action of fungal culture extracts and effects of ligninolytic enzymes. Bioresour. Technol. 2015;189:121–130. doi: 10.1016/j.biortech.2015.03.149.
    1. Koutrotsios G., Patsou M., Mitsou E.K., Bekiaris G., Kotsou M., Tarantilis P.A., Pletsa V., Kyriacou A., Zervakis G.I. Valorization of Olive By-Products as Substrates for the Cultivation of Ganoderma lucidum and Pleurotus ostreatus Mushrooms with Enhanced Functional and Prebiotic Properties. Catalysts. 2019;9:537. doi: 10.3390/catal9060537.
    1. Koutrotsios G., Kalogeropoulos N., Stathopoulos P., Kaliora A., Zervakis G.I. Bioactive compounds and antioxidant activity exhibit high intraspecific variability in Pleurotus ostreatus mushrooms and correlate well with cultivation performance parameters. World J. Microbiol. Biotechnol. 2017;33:98. doi: 10.1007/s11274-017-2262-1.
    1. Koutrotsios G., Larou E., Mountzouris K., Zervakis G.I. Detoxification of olive mill wastewater and bioconversion of olive crop residues into high-value added biomass by the choice edible mushroom Hericium erinaceus. Appl. Biochem. Biotechnol. 2016;180:195–209. doi: 10.1007/s12010-016-2093-9.
    1. Mitsou E.K., Saxami G., Stamoulou E., Kerezoudi E., Terzi E., Koutrotsios G., Bekiaris G., Zervakis G.I., Mountzouris K.C., Pletsa V., et al. Effects of Rich in B-Glucans Edible Mushrooms on Aging Gut Microbiota Characteristics: An In Vitro Study. Molecules. 2020;25:2806. doi: 10.3390/molecules25122806.
    1. An J., Liu J., Liang Y., Ma Y., Chen C., Cheng Y., Peng P., Zhou N., Zhang R., Addy M., et al. Characterization, bioavailability and protective effects of phenolic-rich extracts from almond hulls against pro-oxidant induced toxicity in Caco-2 cells. Food Chem. 2020;322:126742. doi: 10.1016/j.foodchem.2020.126742.
    1. Sauer J., Richter K.K., Pool-Zobel B.L. Products formed during fermentation of the prebiotic inulin with human gut flora enhance expression of biotransformation genes in human primary colon cells. Br. J. Nutr. 2007;97:928–937. doi: 10.1017/S0007114507666422.
    1. Klinder A., Forster A., Caderni G., Femia A.P., Pool-Zobel B. Fecal water genotoxicity is predictive of tumor-preventive activities by inulin-like oligofructoses, probiotics (Lactobacillus rhamnosus and Bifidobacterium lactis), and their symbiotic combination. Nutr. Cancer. 2004;49:144–155. doi: 10.1207/s15327914nc4902_5.
    1. Pool-Zobel B., Cherbut C. Diets enriched with cereal brans or inulin modulate protein kinase C activity and isozyme expression in rat colonic mucosa. Br. J. Nutr. 2003;89:283–284. doi: 10.1079/BJN2002744.
    1. Roberton A.M. Roles of endogenous substances and bacteria in colorectal cancer. Mutat. Res. 1993;290:71–78. doi: 10.1016/0027-5107(93)90034-D.
    1. Vogrinčič M., Kreft I., Filipič M., Zegura B. Antigenotoxic effect of Tartary (Fagopyrum tataricum) and common (Fagopyrum esculentum) buckwheat flour. J. Med. Food. 2013;16:944–952. doi: 10.1089/jmf.2012.0266.
    1. Abrahamse S.L., Pool-Zobel B.L., Rechkemmer G. Potential of short chain fatty acids to modulate the induction of DNA damage and changes in the intracellular calcium concentration by oxidative stress in isolated rat distal colon cells. Carcinogenesis. 1999;20:629–634. doi: 10.1093/carcin/20.4.629.
    1. Dhawan A., Anderson D. The Comet Assay in Toxicology. 2nd ed. Royal Society of Chemistry; Cambridge, UK: 2016.
    1. Yang J., Martínez I., Jens W., Ali K., Devin J.R. In vitro Characterization of the Impact of Selected Dietary Fibers on Fecal Microbiota Composition and Short Chain Fatty Acid Production. Anaerobe. 2013;23:74–81. doi: 10.1016/j.anaerobe.2013.06.012.
    1. Marchese S., Polo A., Ariano A., Velotto S., Costantini S., Severino L. Aflatoxin B1 and M1: Biological Properties and Their Involvement in Cancer Development. Toxins. 2018;10:214. doi: 10.3390/toxins10060214.
    1. De Vadder F., Kovatcheva-Datchary P., Goncalves D., Vinera J., Zitoun C., Duchampt A., Bäckhed F., Mithieux G. Microbiota-generated metabolites promote metabolic benefits via gut-brain neural circuits. Cell. 2014;156:84–96. doi: 10.1016/j.cell.2013.12.016.
    1. Sawangwan T., Wansanit W., Pattani L., Noysang C. Study of Prebiotic Properties from Edible Mushroom Extraction. Agric. Nat. Resour. 2018;52:519–524. doi: 10.1016/j.anres.2018.11.020.
    1. Rowland I., Gibson G., Heinken A., Scott K., Swann J., Thiele I., Tuohy K. Gut microbiota functions: Metabolism of nutrients and other food components. Eur. J. Nutr. 2018;57:1–24. doi: 10.1007/s00394-017-1445-8.
    1. Zhu F., Du B., Bian Z., Xu B. β-Glucans from Edible and Medicinal Mushrooms: Characteristics, Physicochemical and Biological Activities. J. Food Compost. Anal. 2015;41:165–173. doi: 10.1016/j.jfca.2015.01.019.
    1. Zeisel S.H., Mar M.-H., Howe J.C., Holden J.M. Concentrations of Choline-Containing Compounds and Betaine in Common Foods. J. Nutr. 2003;133:1302–1307. doi: 10.1093/jn/133.5.1302.
    1. Canyelles M., Tondo M., Cedó L., Farràs M., Escolà-Gil J.C., Blanco-Vaca F. Trimethylamine N-oxide: A link among diet, gut microbiota, gene regulation of liver and intestine cholesterol homeostasis and HDL function. Int. J. Mol. Sci. 2018;19:3228. doi: 10.3390/ijms19103228.
    1. Fennema D., Phillips I.R., Shephard E.A. Trimethylamine and trimethylamine N-oxide, a Flavin-Containing Monooxygenase 3 (FMO3)-mediated host-microbiome metabolic axis implicated in health and disease. Drug Metab. Dispos. 2016;44:1839–1850. doi: 10.1124/dmd.116.070615.
    1. Mazzoli R., Pessione E. The neuro-endocrinological role of microbial glutamate and GABA signaling. Front. Microbiol. 2016;7:1934. doi: 10.3389/fmicb.2016.01934.
    1. Strandwitz P., Kim K.H., Terekhova D., Liu J.K., Sharma A., Levering J., McDonald D., Dietrich D., Ramadhar T.R., Lekbua A., et al. GABA-modulating bacteria of the human gut microbiota. Nat. Microbiol. 2019;4:396–403. doi: 10.1038/s41564-018-0307-3.
    1. Jewett B.E., Sharma S. StatPearls. StatPearls Publishing; Treasure Island, FL, USA: 2020. Physiology, GABA.
    1. Uerlings J., Schroyen M., Willems E., Tanghe S., Bruggeman G., Bindelle J., Everaert N. Differential effects of inulin or its fermentation metabolites on gut barrier and immune function of porcine intestinal epithelial cells. J. Funct. Foods. 2020;67:103855. doi: 10.1016/j.jff.2020.103855.
    1. Philippoussis A., Zervakis G., Diamantopoulou P. Bioconversion of agricultural lignocellulosic wastes through the cultivation of the edible mushrooms Agrocybe aegerita, Volvariella volvacea and Pleurotus spp. World J. Microbiol. Biotechnol. 2001;17:191–200. doi: 10.1023/A:1016685530312.
    1. Xia J., Bjorndahl T.C., Tang P., Wishart D.S. MetaboMiner-Semi-automated identification of metabolites from 2D NMR spectra of complex biofluids. BMC Bioinform. 2008;9:507. doi: 10.1186/1471-2105-9-507.
    1. Bjerrum J.T., Wang Y., Hao F., Coskun M., Ludwig C., Günther U., Nielsen O.H. Metabonomics of human fecal extracts characterize ulcerative colitis, Crohn’s disease and healthy individuals. Metabolomics. 2014;11:122–133. doi: 10.1007/s11306-014-0677-3.
    1. Eriksson L., Johansson E., Kettaneh-Wold N., Trygg J., Wikström C., Wold S. Multi- and Megavariate Data Analysis. 2nd ed. Umetrics Academy; Umeå, Sweden: 2006.

Source: PubMed

3
Abonner