Health Benefits of Mushroom Dietary Fiber: Comparison
Please note this is a comparison between Version 2 by Catherine Yang and Version 1 by Yan Zhao.

Mushroom dietary fiber is a type of bioactive macromolecule derived from the mycelia, fruiting bodies, or sclerotia of edible or medicinal fungi. The use of mushroom dietary fiber as a prebiotic has recently gained significant attention for providing health benefits to the host by promoting the growth of beneficial microorganisms; therefore, mushroom dietary fiber has promising prospects for application in the functional food industry and in drug development. The gut microbiota, as the core microecological system in the human intestinal tract, helps maintain the normal physiological function of the human body by preventing the invasion of various viral antigens. Dietary fibers (DFs) are fermented by intestinal microorganisms to yield SCFAsshort-chain fatty acids (SCFAs), which can improve host health and have many beneficial effects in the human body.

  • dietary fiber
  • mushroom
  • gut microbiota
  • beneficial effects

1. Improving Metabolic Syndromes

Metabolic syndrome (MS) is a dysbiosis of physiological metabolism caused by insulin resistance. MS manifests as a pathological state of metabolic disorders related to nutrition, including hyperglycemia, dyslipidemia, central obesity, and hypertension. An increasing amount of evidence indicates that the etiology of MS is associated with dysbiosis of the gut microbiota [147,148,149][1][2][3]. The HFD-induced dysbiosis of the gut microbiota may disrupt intestinal barrier function and increase endotoxin levels in the circulatory system. This leads to metabolic endotoxemia and induces MSs, such as insulin resistance, obesity, and even diabetes [150][4]. By generating enzymes such as CAZymes and proteases, the gut microbiota promotes the digestion of carbohydrates, such as DFs, and produces metabolites such as SCFAs that can be absorbed and used by the body. Studies have shown that changes in the gut microbiota and SCFAs are associated with the development of metabolic diseases [151][5]. According to experimental evidence, an increase in Firmicutes and a decrease in Bacteroides have been observed in obese individuals and mouse models [152][6]. Indeed, DFs from mushrooms can effectively improve diet-induced MSs in mice and rats (Table 31). For example, P. eryngii DFs reduced LDL cholesterol levels and body weight in HFD-fed mice by altering the abundance of SCFA-producing gut microbiota [116][7]. G. lucidum DFs reverse the HFD-induced dysbiosis of gut microbiota by reducing the proportion of Firmicutes/Bacteroides and the Proteobacteria level. They also maintain the integrity of the intestinal barrier and reduce metabolic endotoxemia [120][8]. F. velutipes DFs can alleviate lipid metabolism dysbiosis in obese mice by regulating the intestinal-flora-mediated AMPK signaling pathway [119][9]. In addition, other types of DFs from mushrooms can play a positive role in MSs by regulating the gut microbiota composition (Table 31). Thus, mushroom DFs appear to play a positive role in regulating dysbiosis in the intestine that is induced by metabolic disturbances and maintaining the integrity of the intestinal barrier, further highlighting the potential value of mushroom DFs as foods or drugs.
Table 31.
Effects of mushroom DF on the gut microbiota and SCFAs.
DF Source Model Gut Microbiota Regulation SCFA Generation Effect on Host Reference
Pleurotus eryngii HFD-induced obese rat The relative abundances of Roseburia and

Lactobacillus ↓, the relative abundances of Anaerostipes,

Clostridium and Lactococcus ↑.
Increased the

concentrations of total

SCFAs.
Reduced BW gain, adipose tissue

weight, FBG level; the expression

of FASN and ACC.
[116][7]
Pleurotus eryngii HFD-fed mice The relative abundances of Methylobacterium and Lactobacillus ↑, the relative abundances of unidentified_Lachnospiraceae and Helicobacter ↓. Increased the

content of SCFAs, including acetic acid, propionic acid, and butyric acid.
Decreased the weight, promoted the proliferation of beneficial bacteria, reduced the risks of many chronic diseases. [53][10]
Agaricus blazei Murrill Hyperlipidemia rats The ratio of

Firmicutes/Bacteroidetes ↓; the abundance of Peptostreptococcaceae, Erysipelaceae, and Clostridium ↑.
Nm Regulated dyslipidemia in rats with hyperlipidemia possibly by regulating imbalance in the intestinal microflora. [117][11]
Hericium caput-medusae One-day-old Arbor Acres male broilers The count of Lactobacilli and Bifidobacteria ↑, the count of acecum Escherichia coli ↓. Increased the concentration of propionic acid. Decreased cholesterol content in broiler chickens. [118][12]
Flammulina velutipes Male C57BL/6 J mice The relative abundance of some beneficial bacteria ↑, such as Akkermansia and Prevotellaceae UCG-001; the relative abundance of some harmful bacteria ↓, such as Lachnospiraceae

NK4A136 group and Desulfovibrio.
Nm Reduced the weight gain, triglycerides and total

cholesterol, low-density lipoprotein cholesterol; increased the activity of enzymes related to

scavenging ability of oxygen free radicals.
[119][9]
Flammulina velutipes Mice The relative abundance of Firmicutes ↓, the relative abundance of Bacteroidetes ↑; the ratio of

Firmicutes/Bacteroidetes ↓.
Increased the

concentrations of total

SCFAs, acetic acid, propionic acid, and n-butyric acid.
Suppressed obesity and immune regulation. [101][13]
Ganoderma lucidum C57BL/6NCrlBltw genetic lineage mice The ratio of

Firmicutes/Bacteroidetes, Proteobacteria ↓.
Nm Reduced body weight gain, chronic inflammation, and insulin resistance in obese individuals. [120][8]
Poria cocos C57BL/6J mice The relative abundance of Lachnospiracea, Clostridium ↑. Increased butyrate levels. Activated the intestinal PPAR-γ pathway, modulated gut microbiota to improve hyperglycemia and hyperlipidemia. [121][14]
Agaricus bisporus Human The relative abundance of Firmicutes ↑, the relative abundance of Bacteroidetes ↓. Increased the concentrations of acetic acid and propionic acid. Increased the relative abundance of beneficial bacteria, exhibited an effective prebiotic regulation function on human gut microbiota. [105][15]
Cordyceps militaris Liver and kidney injury induced by lead acetate in mice The relative abundance of Clostridium and

Bacteroidetes ↑, the relative abundance of Firmicutes ↓.
Nm Reduced the Pb2+ content and organ index of liver and kidney in mice, had a protective effect on organs against damage in mice. [122][16]
Pleurotus eryngii C57BL/6 male mice The relative abundances of

Firmicutes ↓, Bacteroidetes
Increased the

concentrations of Acetate and Propionate.
Regulated the host immune function effectively. [123][17]
Ganoderma lucidum Chronic pancreatitis mice The relative abundance of Bacteroidetes ↓ and that of Firmicutes ↑;

at the genus level, the

relative abundance of beneficial bacteria

such as Lactobacillus, Roseburia, and

Lachnospira ↑.
Nm Indicated beneficial effects on pancreas fibrosis, and impeded an inflammatory response. [124][18]
Dictyophora indusiata Antibiotic-induced intestinal microflora disorder in mice Beneficial bacteria ↑, including

Lactobacilli and Ruminococcaceae; harmful bacteria ↓, such as

Enterococcus, Bacteroides, and

Proteobacteria.
Nm Enhanced the restoration of gut microbiota and gut barrier integrity, reduce the inflammation and endotoxin levels in mice. [125][19]
Coprinus comatus Human The relative abundances of

Bacteroides and Bifidobacterium ↑, the ratio of Firmicutes/Bacteroidetes ↓.
Increased the production of propionic acid and butyric acid. Demonstrated potential prebiotic effects. [50][20]
Ganoderma lucidum C57BL/6J mice The relative abundances of Actinobacteria at the family level, and Leuconostoc, Lactobacillus spp. ↑. Nm Improved low-grade chronic inflammation, ectopic lipid accumulation, and systemic insulin sensitivity. [126][21]
Hericium erinaceus Mice The relative abundance of Lachnospiraceae and Akkermansiaceae ↑, the relative abundance of Rikenellaceae and Bacteroidaceae ↓. Nm Promoted the production of NO, IL-6, IL-10, INF-γ, and TNF-α. [127][22]
Auricularia auricular ICR mice The ratio of Firmicutes/Bacteroidetes ↓, the relative abundance of Porphyromoadaceae and Bacteroidaceae ↑. Increased the concentration of total SCFAs and propanoic acid. Increased microbial community diversity, and increased the immunoglobulin levels in mouse serum. [128][23]
Ganoderma lucidum DSS-induced colitis male

Wistar rats
The relative abundance of Firmicutes, Paraprevotella,

etc. ↑, the relative abundance of Proteobacteria, Escherichia, etc. ↓.
Increased total SCFAs,

acetic acid, propionic acid,

and butyric acid.
Enhanced the immunity and reduced inflammatory response and colonic cancer risk. [129][24]
Ganoderma lucidum BALB/C mice The ratio of Firmicutes/Bacteroidetes ↓, the relative abundance of Alistipes ↑. Nm Demonstrated tumor-suppressing activity in mice. [130][25]
Ganoderma lucidum BALB/c mice The relative abundance of Oscillospira and unknown genus of Desulfovibrionaceae ↓. Nm Prevented colon from shortening and reduced mortality by 30% of mortality in CRC mice. [131][26]
Note: HFD: high-fat diet; BW: body weight; FBG: fasting blood glucose; FASN: fatty acid synthase; ACC: acetyl-CoA carboxylase; CRC: colorectal cancer; ↑: increase; ↓: decrease.

2. Immunomodulatory Effects

The intestinal tract is the largest immune organ of the human body, and it is involved in immune and inflammatory reactions [153][27]. Although DFs cannot be completely digested in the intestine, they are decomposed into various metabolites through enzymes produced by intestinal microorganisms. Some microbial metabolites, such as tryptophan metabolites and SCFAs, interact with host cells through the intestinal barrier, thereby affecting the immune response [154][28]. An increasing number of studies have shown that SCFAs can inhibit the expression of inflammatory factors or alleviate inflammation by promoting histone acetylation or activating GPRs [155][29], activating peroxisome-proliferator-activated receptors [156][30], inhibiting the NF-κB signaling pathway [157][31], facilitating T-cell apoptosis [158][32], increasing antimicrobial peptide production [159][33], and downregulating the expression of signal transduction and activating transcription factor-3 [160][34]. Relevant studies have also reported that DFs from mushrooms can promote SCFA production and the growth of intestinal microorganisms to stimulate the host immune response and regulate the differentiation, maturation, and function of immune cells (Table 31). Vlassopoulou et al. [161][35] found that supplementation with mushroom β-(1→3, 1→6)-d-glucan is well tolerated and promotes health through the potentiation of the immune system. In addition, F. velutipes DF may affect immune function regulation by mediating the gut microbiota [101][13]. H. erinaceus DFs can regulate the gut microbiota composition and immune activity through the NF-κB, MAPK, and PI3K/Akt pathways [127][22]. G. lucidum DFs change the diversity of the gut microbiota and significantly alleviate pancreatitis symptoms in mice by reducing the levels of lipase, interferon-γ (IFN-γ), and TNF-α and increasing SOD levels and total antioxidant activity [124][18]. A. auricular DFs may affect intestinal nutritional metabolism and immune regulation by changing the composition of the gut microbiota [128][23]. G. lucidum DFs not only regulate the gut microbiota composition and SCFA production but also participate in the regulation of gene expression in KEGG pathways related to different types of inflammation [129][24]. Thus, these studies indicate that DFs from different mushrooms may be associated with different immune regulatory pathways. This immune regulation can be attributed to the diversity of the gut microbiota and SCFA production, which may act as signaling molecules for mediating and maintaining the host’s immune system.

3. Antitumor Effects

The gut microbiota affects the metabolism and endocrine and immune systems of the host. The gut microbiota is associated with the occurrence of many diseases, including inflammatory bowel disease, nonalcoholic fatty liver disease, type 2 diabetes, and neurodegenerative diseases [162,163,164][36][37][38]. Importantly, increasing evidence shows that the gut microbiota can affect tumor occurrence, tumor progression, and the response to treatment [19][39]. For example, Helicobacter pylori induces gastritis and canceration by producing toxic factors such as cytotoxin-associated gene A and vacuolating cytotoxin A [165][40]. A study investigating the gut microbiota of patients with early lung cancer reported that Akkermansia muciniphila may cause lung cancer [166][41]. The decrease in the abundance of Lactobacillidae and Bifidobacteriaceae in colorectal cancer (CRC) patients is related to colon and rectal tumors, respectively [167][42]. In patients with multiple polypoid adenoma and intramucosal carcinoma, significant changes in the microbiome and metabolome have been observed. The relative abundance of Fusobacterium nucleatum spp. increased during the progression of intramucosal carcinoma to the more advanced stage, while the abundance of Atobobium parvulum and Actinomyces odontolyticus significantly increased in patients with multiple polypoid adenoma and/or intramucosal carcinoma [168][43]. Moreover, the gut microbiota is closely correlated with the effect of chemotherapy and immunotherapy [169,170][44][45]. For example, the gut microbiota can alleviate chemotherapy-induced adverse reactions in CRC patients [171][46]. Butyric acid, a metabolite of the gut microbiota, can directly improve the antitumor cytotoxic effect of CD8+ T cells in vitro and in vivo in an ID2-dependent manner by promoting the IL-12 signaling pathway. Butyric acid can also promote the antitumor efficacy of oxaliplatin [172][47]. L. rhamnosus GG-induced cGAS/STING-dependent type I interferon can enhance the response to immune checkpoint inhibitors [173][48].
DFs from mushrooms play an anticancer/antitumor role by regulating the gut microbiota composition and diversity, and this role has attracted increasing attention (Table 31). For example, G. lucidum polysaccharide can reverse the proportion of Firmicutes/Bacteroides and increase the levels of Alistipes, resulting in the production of SCFAs, and Helicobacter and Riskenella, which are related to immunosuppression and carcinogenesis [130][25]. Moreover, G. lucidum DFs can alleviate CRC by altering special intestinal microorganisms [131][26]. Thus, these studies indicate that mushroom DFs inhibit tumor growth or metastasis by regulating the gut microbiota composition and diversity; furthermore, immunomonitoring mediated by gut microbiota-produced metabolites such as SCFAs may be beneficial. However, the exact anticancer/antitumor mechanism of mushroom DFs remains unclear.

4. Other Beneficial Effects

Because of their strong water absorption and swelling capacities, DFs from mushrooms, especially IDF, can promote intestinal peristalsis, increase stool volume, increase the frequency of bowel movements, and avert constipation, thereby preventing and treating gastrointestinal diseases. Feeding constipated rats with A. polytricha DFs increased the wet weight of their stool and intestinal propulsion rate, thereby indicating high constipation-relieving activity [73][49]. Furthermore, DFs from mushrooms have antioxidant capacity and can eliminate free radicals from the body. For example, DFs extracted from Boletus edulis have significant reducing power and chelating activity and strong antioxidant activity [174][50].

References

  1. Ley, R.E.; Turnbaugh, P.J.; Klein, S.; Gordon, J.I. Human gut microbes associated with obesity. Nature 2006, 444, 1022–1023.
  2. Larsen, N.; Vogensen, F.K.; Van Den Berg, F.W.; Nielsen, D.S.; Andreasen, A.S.; Pedersen, B.K.; AI-Soud, W.A.; Sorensen, S.J.; Hansen, L.H.; Jakobsen, M. Gut microbiota in human adults with type 2 diabetes differs from non-diabetic adults. PLoS ONE 2010, 5, e9085.
  3. Cani, P.D.; Bibiloni, R.; Knauf, C.; Waget, A.; Neyrinck, A.M.; Delzenne, N.M.; Burcelin, R. Changes in gut microbiota control metabolic endotoxmia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes 2008, 57, 1470–1481.
  4. Cani, P.D.; Amar, J.; Iglesias, M.A.; Poggi, M.; Knauf, C.; Bastelica, D.; Neyrinck, A.M.; Fava, F.; Tuohy, K.M.; Chabo, C.; et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007, 56, 1761–1772.
  5. Backhed, F.; Manchester, J.K.; Semenkovich, C.F.; Gordon, J.I. Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proc. Nat. Acad. Sci. USA 2007, 104, 979–984.
  6. Turnbaugh, P.J.; Ley, R.E.; Mahowald, M.A.; Magrini, V.M.; Mardis, E.R.; Gordon, J.I. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 2006, 444, 1027–1031.
  7. Nakahara, D.; Nan, C.; Mori, K.; Hanayama, M.; Kikuchi, H.; Hirai, S.; Egashira, Y. Effect of mushroom polysaccharides from Pleurotus eryngii on obesity and gut microbiota in mice fed a high-fat diet. Eur. J. Nutr. 2020, 59, 3231–3244.
  8. Chang, C.J.; Lin, C.S.; Lu, C.C.; Martel, J.; Ko, Y.F.; Ojcius, D.M.; Tseng, S.F.; Wu, T.R.; Chen, Y.Y.M.; Yong, J.D.; et al. Ganoderma lucidum reduces obesity in mice by modulating the composition of the gut microbiota. Nat. Commun. 2015, 6, 7489.
  9. Wang, W.; Yang, S.; Song, S.; Zhang, J.; Jia, F. Flammulina velutipes mycorrhizae dietary fiber improves lipid metabolism disorders in obese mice through activating AMPK signaling pathway mediated by gut microbiota. Food Biosci. 2021, 43, 101246.
  10. Han, X.; Yang, D.; Zhang, S.; Liu, X.; Zhao, Y.; Song, C.; Sun, Q. Characterization of insoluble dietary fiber from Pleurotus eryngii and evaluation of its effects on obesity-preventing or relieving effects via modulation of gut microbiota. J. Future Foods 2023, 3, 55–66.
  11. Li, Y.; Lu, X.; Li, X.; Guo, X.; Sheng, Y.; Li, Y.; Xu, G.; Han, X.; An, L.; Du, P. Effects of Agaricus blazei Murrill polysaccharides on hyperlipidemic rats by regulation of intestinal microflora. Food Sci. Nutr. 2020, 8, 2758–2772.
  12. Shang, H.M.; Song, H.; Wang, L.N.; Wu, B.; Ding, G.D.; Jiang, Y.Y.; Yao, X.; Shen, S.J. Effects of dietary polysaccharides from the submerged fermentation concentrate of Hericium caput-medusae (Bull.:Fr.) Pers. On performance, gut microflora, and cholesterol metabolism in broiler chickens. Livest. Sci. 2014, 167, 276–285.
  13. Zhao, R.; Hu, Q.; Ma, G.; Su, A.; Xie, M.; Li, X.; Chen, G.; Zhao, L. Effects of Flammulina velutipes polysaccharide on immune response and intestinal microbiota in mice. J. Funct. Foods 2019, 56, 255–264.
  14. Sun, S.S.; Wang, K.; Ma, K.; Bao, L.; Liu, H.W. 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. 2019, 17, 3–14.
  15. Xiang, Q.R.; Li, W.Y.; Feng, T. Regulating effects of dietary fiber and powder of Agaricus bisporus based on in vitro fermentation on human gut microbiota. Sci. Technol. Food Ind. 2022, 44, 130–137. (In Chinese)
  16. Song, Q.; Zhu, Z. Using Cordyceps militaris extracellular polysaccharides to prevent Pb2+-induced liver and kidney toxicity by activating Nrf2 signals and modulating gut microbiota. Food Funct. 2020, 11, 9226–9239.
  17. Ma, G.; Kimatu, B.M.; Zhao, L.; Yang, W.; Pei, F.; Hu, Q. In vivo fermentation of a Pleurotus eryngii polysaccharide and its effects on fecal microbiota composition and immune response. Food Funct. 2017, 8, 1810–1821.
  18. Li, K.; Zhuo, C.; Teng, C.; Yu, S.; Wang, X.; Hu, Y.; Ren, G.; Yu, M.; Qu, J. Effects of Ganoderma lucidum polysaccharides on chronic pancreatitis and intestinal microbiota in mice. Int. J. Biol. Macromol. 2016, 93, 904–912.
  19. Kanwal, S.; Joseph, T.P.; Owusu, L.; Ren, X.; Li, M.; Xin, Y. A polysaccharide isolated from Dictyophora indusiata promotes recovery from antibiotic-driven intestinal dysbiosis and improves gut epithelial barrier function in a mouse model. Nutrients 2018, 10, 1003.
  20. Zhang, Z.; Zhao, L.; Qu, H.; Zhou, H.; Yang, H.; Chen, H. Physicochemical characterization, adsorption function and prebiotic effect of chitin-glucan complex from mushroom Coprinus comatus. Int. J. Biol. Macromol. 2022, 206, 255–263.
  21. Xu, S.; Dou, Y.; Ye, B.; Wu, Q.; Wang, Y.; Hu, M.; Ma, F.; Rong, X.; Guo, J. Ganoderma lucidum polysaccharide insulin sensitivity by regulating inflammatory cytokines and gut microbiota composition in mice. J. Funct. Foods 2017, 38, 545–552.
  22. Yang, Y.; Ye, H.; Zhao, C.; Ren, L.; Wang, C.; Georgiev, M.I.; Xiao, J.; Zhang, T. Value added immunoregulatory polysaccharides of Hericium erinaceus and their effect on the gut microbiota. Carbohyd. Polym. 2021, 262, 117668.
  23. Zhao, R.; Cheng, N.; Nakata, P.A.; Zhao, L.; Hu, Q. Consumption of polysaccharides from Auricularia auricular modulates the intestinal microbiota in mice. Food Res. Int. 2019, 123, 383–392.
  24. Xie, J.; Liu, Y.; Chen, B.; Zhang, G.; Ou, S.; Luo, J.; Peng, X. Ganoderma lucidum polysaccharide improves rat DSS-induced colitis by altering cecal microbiota and gene expression of colonic epithelial cells. Food Nutr. Res. 2019, 63, 1559.
  25. Li, L.F.; Liu, H.B.; Zhang, Q.W.; Li, Z.P.; Wong, T.L.; Fung, H.Y.; Zhang, J.X.; Bai, S.P.; Lu, A.P.; Han, Q.B. Comprehensive comparison of polysaccharides from Ganoderma lucidum and G. sinense: Chemical, antitumor, immunomodulating and gut-microbiota modulatory properties. Sci. Rep. 2018, 8, 6172.
  26. Luo, J.; Zhang, C.; Liu, R.; Gao, L.; Ou, S.; Liu, L.; Peng, X. Ganoderma lucidum polysaccharide alleviating colorectal cancer by alteration of special gut bacteria and regulation of gene expression of colonic epithelial cells. J. Funct. Foods 2018, 47, 127–135.
  27. Hachimura, S.; Totsuka, M.; Hosono, A. Immunomodulation by food: Impact on gut immunity and immune cell function. Biosci. Biotechnol. Biochem. 2018, 82, 584–599.
  28. Koh, A.; De Vadder, F.; Kovatcheva-Datchary, P.; Backhed, F. From Dietary fiber to host physiology: Short-chain fatty acids as key bacterial metabolites. Cell 2016, 165, 1332–1345.
  29. Luu, M.; Pautz, S.; Kohl, V.; Singh, R.; Romero, R.; Lucas, S.; Hofmann, J.; Raifer, H.; Vachharajani, N.; Carrascosa, L.C.; et al. The short-chain fatty acid pentanoate suppresses autoimmunity by modulating the metabolic-epigenetic crosstalk in lymphocytes. Nat. Commun. 2019, 10, 760.
  30. Kinoshita, M.; Suzuki, Y.; Saito, Y. Butyrate reduces colonic paracellular permeability by enhancing PPARγ activation. Biochem. Biophys. Res. Commun. 2002, 293, 827–831.
  31. Lee, C.; Kim, B.G.; Kim, J.H.; Chun, J.; Im, J.P.; Kim, J.S. Sodium butyrate inhibits the NF-kappa B signaling pathway and histone deacetylation, and attenuates experimental colitis in an IL- 10 independent manner. Int. Immunopharmacol. 2017, 51, 47–56.
  32. Ogawa, K.; Yasumura, S.; Atarashi, Y.; Minemura, M.; Miyazaki, T.; Lwamoto, M.; Higuchi, K.; Watanabe, A. Sodium butyrate enhances Fas-mediated apoptosis of human hepatoma cells. J. Hepatol. 2004, 40, 278–284.
  33. Xiong, H.; Guo, B.; Gan, Z.; Song, D.; Lu, Z.; Yi, H.; Wu, Y.; Wang, Y.; Du, H. Butyrate upregulates endogenous host defense peptides to enhance disease resistance in piglets via histone deacetylase inhibition. Sci. Rep. 2016, 6, 27070.
  34. Kanauchi, O.; Serizawa, I.; Araki, Y.; Suzuki, A.; Andoh, A.; Fujitama, Y.; Mitsuyama, K.; Takaki, K.; Toyonaga, A.; Sata, M.; et al. Germinated barley foodstuff, a prebiotic product, ameliorates inflammation of colitis through modulation of the enteric environment. J. Gastroenterol. 2003, 38, 134–141.
  35. Vlassopoulou, M.; Yannakoulia, M.; Pletsa, V.; Zervakis, G.I.; Kyriacou, A. Effects of fungal beta-glucans on health—A systematic review of randomized controlled trials. Food Funct. 2021, 12, 3366.
  36. Castellarin, M.; Warren, R.L.; Freeman, J.D.; Dreolini, L.; Krzywinski, M.; Stauss, J.; Barnes, R.; Watson, P.; Allen-Vercoe, E.; Moore, R.A.; et al. Fusobacterium nucleatum infection is prevalent in human colorectal carcinoma. Genome Res. 2012, 22, 299–306.
  37. Song, H.; Yoo, Y.; Hwang, J.; Na, Y.; Kim, H.S. Faecalibacterium prausnitzii subspecies-level dysbiosis in the human gut microbiome underlying atopic dermatitis. J. Allergy Clin. Immun. 2016, 137, 852–860.
  38. Amar, J.; Chabo, C.; Waget, A.; Klopp, P.; Vachoux, C.; Bermudez-Humaran, L.G.; Smirnova, N.; Berge, M.; Sulpice, T.; Lahtinen, S.; et al. Intestinal mucosal adherence and translocation of commensal bacteria at the early onset of type 2 diabetes: Molecular mechanisms and probiotic treatment. EMBO Mol. Med. 2011, 3, 559–572.
  39. Zitvogel, L.; Galluzzi, L.; Viaud, S.; Vetizou, M.; Daillere, R.; Merad, M.; Kroemer, G. Cancer and the gut microbiota: An unexpected link. Sci. Transl. Med. 2015, 7, 271ps1.
  40. Wang, F.; Meng, W.; Wang, B.; Qiao, L. Helicobacter pylori-induced gastric inflammation and gastric cancer. Cancer Lett. 2014, 345, 196–202.
  41. Zheng, Y.; Fang, Z.; Xue, Y.; Zhang, J.; Zhu, J.; Gao, R. Specific gut microbiome signature predicts the early-stage lung cancer. Gut Microbes 2020, 11, 1030–1042.
  42. Youssef, O.; Lahti, L.; Kokkola, A.; Karla, T.; Tikkanen, M.; Ehsan, H.; Carpelan-Holmstrom, M.; Koskensalo, S.; Bohling, T.; Rautelin, H.; et al. Stool microbiota composition differs in patients with stomach, colon, and rectal neoplasms. Digest. Dis. Sci. 2018, 63, 2950–2958.
  43. Yachida, S.; Mizutani, S.; Shiroma, H.; Shiba, S.; Nakajima, T.; Sakamoto, T.; Watanabe, H.; Masuda, K.; Nishimoto, Y.; Kubo, M.; et al. Metagenomic and metabolomic analyses reveal distinct stage-specific phenotypes of the gut microbiota in colorectal cancer. Nat. Med. 2019, 25, 968–976.
  44. Viaud, S.; Saccheri, F.; Mignot, G.; Yamazaki, T.; Daillere, R.; Hannani, D.; Enot, D.P.; Pfirschke, C.; Engblom, C.; Pittet, M.J.; et al. The intestinal microbiota modulates the anticancer immune effects of cyclophosphamide. Science 2013, 342, 971–976.
  45. Sivan, A.; Corrales, L.; Hubert, N.; Williams, J.B.; Aquino-Michaels, K.; Earley, Z.M.; Benyamin, F.W.; Lei, Y.M.; Jabri, B.; Alegre, M.; et al. Commensal Bifidobacterium promotes antitumor immunity and facilitates anti-PD-L1 efficacy. Science 2015, 350, 1084–1089.
  46. Mego, M.; Chovanec, J.; Vochyanova-Andrezalova, I.; Konkolovsky, P.; Mikulova, M.; Reckova, M.; Miskovska, V.; Bystricky, B.; Beniak, J.; Medvecova, L.; et al. Prevention of irinotecan induced diarrhea by probiotics: A randomized double blind, placebo controlled pilot study. Complement. Ther. Med. 2015, 23, 356–362.
  47. He, Y.; Fu, L.; Li, Y.; Wang, W.; Gong, M.; Zhang, J.; Fu, Y.X.; Chen, Y.; Guo, X. Gut microbial metabolites facilitate anticancer therapy efficacy by modulating cytotoxic CD8+ T cell immunity. Cell Metab. 2021, 33, 988–1000.
  48. Si, W.; Liang, H.; Bugno, J.; Xu, Q.; Ding, X.; Yang, K.; Fu, Y.; Weichselbaum, R.R.; Zhao, X.; Wang, L. Lactobacillus rhamnosus GG induces cGAS/STING-dependent type I interferon and improves response to immune checkpoint blockade. Gut 2022, 71, 521–533.
  49. Gan, J.; Xie, L.; Peng, G.; Xie, J.; Chen, Y.; Yu, Q. Systematic review on modification methods of dietary fiber. Food Hydrocoll. 2021, 119, 106872.
  50. Zhang, A.; Xiao, N.; He, P.; Sun, P. Chemical analysis and antioxidant activity in vitro of dietary fibers extracted from Boletus edulis. Int. J. Biol. Macromol. 2011, 49, 1092–1095.
More
ScholarVision Creations