Immunomodulatory Activities of a Fungal Protein Extracted from Hericium erinaceus through Regulating the Gut Microbiota

Chen Diling, Zheng Chaoqun, Yang Jian, Li Jian, Su Jiyan, Xie Yizhen, Lai Guoxiao, Chen Diling, Zheng Chaoqun, Yang Jian, Li Jian, Su Jiyan, Xie Yizhen, Lai Guoxiao

Abstract

A single-band protein (HEP3) was isolated from Hericium erinaceus using a chemical separation combined with pharmacodynamic evaluation methods. This protein exhibited immunomodulatory activity in lipopolysaccharide-activated RAW 264.7 macrophages by decreasing the overproduction of tumor necrosis factor-α, interleukin (IL)-1β, and IL-6, and downregulating the expression of inducible nitric oxide synthase and nuclear factor-κB p65. Further researches revealed that HEP3 could improve the immune system via regulating the composition and metabolism of gut microbiota to activate the proliferation and differentiation of T cells, stimulate the intestinal antigen-presenting cells in high-dose cyclophosphamide-induced immunotoxicity in mice, and play a prebiotic role in the case of excessive antibiotics in inflammatory bowel disease model mice. Aided experiments also showed that HEP3 could be used as an antitumor immune inhibitor in tumor-burdened mice. The results of the present study suggested that fungal protein from H. erinaceus could be used as a drug or functional food ingredient for immunotherapy because of its immunomodulatory activities.

Keywords: Hericium erinaceus; anti-inflammation; functional food ingredient; fungal immunomodulatory protein; gut microbiota; immunotherapy.

Figures

Figure 1
Figure 1
Effect of crude protein extracts from Hericium erinaceus on trinitrobenzenesulfonic acid solution (TNBS)-induced inflammatory bowel disease (IBD) rats. (A) The technical route of this study; (B) the fresh fruiting bodies of H. erinaceus and the protein electrophoresis; (C) the hematoxylin and eosin-staining and immunohistochemistry results; (D) the Disease Activity Index scores (calculated according to the weight loss, stool consistency, and blood in feces) and observation of colons of the TNBS-induced IBD rats. Control is the normal group without any treatments, Model is the TNBS-induced IBD rats, HEP is the crude protein extract-treated group after TNBS enema, and 5-aminosalicylic acid (5-ASA) is the positive control group treated with 100 mg/(kg ⋅ day) of 5-ASA after TNBS enema. Values were expressed as means ± SDs. #P < 0.05 vs the control group, *P < 0.05, **P < 0.01 vs the model group, indicating significant differences compared with the model group.
Figure 2
Figure 2
Effects of HEP on trinitrobenzenesulfonic acid solution (TNBS)-induced rats. Normal group; model group, induced by TNBS enema; HEP group, the crude protein extract-treated group after TNBS enema; and positive control group, treated with 100 mg/(kg ⋅ day) of 5-aminosalicylic acid after TNBS enema. After treatment for 14 days, cytokines interleukin (1L)-1α (A), 1L-2 (B), 1L-8 (C), 1L-10 (D), 1L-11 (E), IL-12 (F), tumor necrosis factor (TNF)-γ (H), TNF-α (G), vascular endothelial growth factor (VEGF) (I), MIP-α (J), macrophage colony-stimulating factor (M-CSF) (K), and myeloperoxidase (MPO) (L) were produced. The assays were carried out according to the procedures recommended in the enzyme-linked immunosorbent assay kit manual. Values were means ± SDs of three independent experiments. #P < 0.05 vs the normal group, *P < 0.05, **P < 0.01 vs the TNBS-treated group.
Figure 3
Figure 3
Effects of HEP3 on proinflammatory cytokine productions in lipopolysaccharide (LPS)-activated RAW 264.7 cells, and effects on the d-galactose-induced HIEpiC senescent cells. Cell viability was measured by quantitative colorimetric methylthiazolyl tetrazolium (MTT) after incubation with HEP3 for 48 h [(A), a]; cells were preincubated with 0.05–0.20 mg/mL HEP3 for 4 h, and then treated with 1 μg/mL LPS for 24 h. Using an ELISA kit, tumor necrosis factor-α [(A), b], interleukin (IL)-1β [(A), c], IL-6 [(A), d], nitric oxide [(A), e], and inducible nitric oxide synthase [(A), f] in the supernatant were detected. [(B), a–e] The cells were treated with 40 mg/mL of d-galactose for 72 h combined with different concentrations of HEP3, and the number of senescent cells (blue-stained cells) was detected using β-galactosidase staining. [(B), f] The cells were treated with different concentrations of HEP for 24 h, and the cytotoxicity was detected by an MTT assay. [(B), g–i] The activities of malondialdehyde, total superoxide dismutase, and glutathione peroxidase of the cells. Values were means ± SDs of three independent experiments. #P < 0.05 vs the normal group, *P < 0.05, **P < 0.01 vs the model group, indicating significant differences compared with the model group.
Figure 4
Figure 4
Effect of HEP3 on the cyclophosphamide-induced immunotoxicity mice. Body weight changes (A), thymus index (B), and spleen index (C), neutral red engulfment (D), splenocyte proliferation (E), platelet (F), and white blood cell (G), the tissue structure of the spleen (H), the CD3+(I), CD4+/CD8 (J), CD4+(L), CD8+(M), CD28+/CD8 (K), and naive T cells (N). CT is the control group treated with just vehicle, CTX is the cyclophosphamide-induced group (intraperitoneal injection of 80 mg/kg) group, HEP3-D is the group treated with 100 mg/kg HEP3 and intraperitoneal injection of 80 mg/kg cyclophosphamide, and HEP3-G is the group treated with 200 mg/kg HEP3 and intraperitoneal injection of 80 mg/kg cyclophosphamide. Values were means ± SDs of six independent experiments. #P < 0.05 vs the control group, *P < 0.05, **P < 0.01 vs the CTX, indicating significant differences.
Figure 5
Figure 5
Influence of cyclophosphamide on the cecal microbiota of mice. (A) The Venn diagram; (B) the PCA analysis of operational taxonomic units; (C) heat map of 16S rRNA gene sequencing analysis of cecal content at the genus level. Z denotes the control (normal) group just treated with vehicle, and M is the cyclophosphamide-induced (intraperitoneal injection of 80 mg/kg) group.
Figure 6
Figure 6
Effects of HEP3 on the microbiota of cecal contents in rats with cyclophosphamide-induced immunotoxicity. (A) The rarefaction curve; (B) the principal component analysis of operational taxonomic units; (C) the classification and abundance of cecal contents at the phylum level; (D) the Venn diagram of OTUs; (E) the sample species classification tree. Z denotes the control group just treated with vehicle, M is the cyclophosphamide-induced (intraperitoneal injection of 80 mg/kg) group, D is the 100 mg/kg HEP3-treated group, and G is the 200 mg/kg HEP3-treated group. Values were means ± SDs of six independent experiments. #P < 0.05 vs the control group, *P < 0.05, **P < 0.01 vs the cyclophosphamide-induced group, indicating significant differences.
Figure 6
Figure 6
Effects of HEP3 on the microbiota of cecal contents in rats with cyclophosphamide-induced immunotoxicity. (A) The rarefaction curve; (B) the principal component analysis of operational taxonomic units; (C) the classification and abundance of cecal contents at the phylum level; (D) the Venn diagram of OTUs; (E) the sample species classification tree. Z denotes the control group just treated with vehicle, M is the cyclophosphamide-induced (intraperitoneal injection of 80 mg/kg) group, D is the 100 mg/kg HEP3-treated group, and G is the 200 mg/kg HEP3-treated group. Values were means ± SDs of six independent experiments. #P < 0.05 vs the control group, *P < 0.05, **P < 0.01 vs the cyclophosphamide-induced group, indicating significant differences.
Figure 7
Figure 7
Effect of HEP3 on the microbiota classification and abundance detected by 16S rRNA gene sequencing analysis of cecal contents from the mice with cyclophosphamide-induced immunotoxicity at the genus level (A,B). Effect of HEP3 on KEGG pathways of gut microbiota in mice with cyclophosphamide-induced immunotoxicity (C,D). Z denotes the control group just treated with vehicle, M is the cyclophosphamide-induced (intraperitoneal injection of 80 mg/kg) group, D is the 100 mg/kg HEP3-treated group, and G is the 200 mg/kg HEP3-treated group.
Figure 7
Figure 7
Effect of HEP3 on the microbiota classification and abundance detected by 16S rRNA gene sequencing analysis of cecal contents from the mice with cyclophosphamide-induced immunotoxicity at the genus level (A,B). Effect of HEP3 on KEGG pathways of gut microbiota in mice with cyclophosphamide-induced immunotoxicity (C,D). Z denotes the control group just treated with vehicle, M is the cyclophosphamide-induced (intraperitoneal injection of 80 mg/kg) group, D is the 100 mg/kg HEP3-treated group, and G is the 200 mg/kg HEP3-treated group.
Figure 8
Figure 8
HEP3 extracted from Hericium erinaceus improved the pathological parameters of the trinitrobenzenesulfonic acid solution (TNBS)-induced mice. (A) The body weight changes; (B) the levels of lipopolysaccharide in serum; (C) the levels of cytokines GM-CSF, tumor necrosis factor (TNF)-γ, 1L-10, interleukin (IL)-12, 1L-17α, 1L-4, TNF-α, and vascular endothelial growth factor in serum; (D) the histopathological changes in colon; and (E) the histopathological changes in spleen. Control is the normal group; model is the TNBS-induced group; model and high-dose antibiotics; HEP3 [100 mg/(kg ⋅ day)], Bifidobacterium, HEP3 and high-dose antibiotics, HEP3 and Bifidobacterium, Bifidobacterium and high-dose antibiotics, HEP3 and Bifidobacterium and high-dose antibiotics.
Figure 9
Figure 9
Immunohistochemical staining of tumor necrosis factor-α (A), NF-κB p65 (B), interleukin-17 (C), and Foxp3 (D) in the colons of different experimental groups in inflammatory bowel disease mice after treatment with HEP3. Control is the normal group; model is the trinitrobenzenesulfonic acid solution-induced group; model and high-dose antibiotics; HEP3 [100 mg/(kg ⋅ day)], Bifidobacterium, HEP3 and high-dose antibiotics, HEP3 and Bifidobacterium, Bifidobacterium and high-dose antibiotics, HEP3 and Bifidobacterium and high-dose antibiotics.
Figure 10
Figure 10
HEP3 showed good prebiotic effects in trinitrobenzenesulfonic acid solution (TNBS)-induced mice. (A,B) The relative abundance at the phylum level; (C–F) the relative abundance at the family level. Control is the normal group; model is the TNBS-induced group; model and high-dose antibiotics; HEP3 [100 mg/(kg ⋅ day)], Bifidobacterium, HEP3 and high-dose antibiotics, HEP3 and Bifidobacterium, Bifidobacterium and high-dose antibiotics, HEP3 and Bifidobacterium and high-dose antibiotics. Values were means of six independent experiments.
Figure 11
Figure 11
Effects of HEP on the average tumor weight and the content of various cytokines in the CC531 cell tumor xenograft model mice. The tumor tissue of HL, HH, and model groups (A); the tumor inhibition rate (B); the contents of various cytokines (C) were detected using an enzyme-linked immunosorbent assay kit: tumor necrosis factor-α (a), PSA (b), interferon-γ (c), macrophage colony-stimulating factor (d), transforming growth factor (e), and vascular endothelial growth factor (f). Values were means ± SDs. #P < 0.05, ##P < 0.01 vs the normal group; *P < 0.05, **P < 0.01 vs the model group, indicating significant differences.
Figure 12
Figure 12
Dominant species interaction-associated network analysis results. Nodes represent the dominant species under the identity of different colors: the red line shows a positive correlation and the green line shows a negative correlation. More connections by nodes indicated more association with others. (A) Antibiotics rapidly declined the diversity and destroyed the stability of the whole ecological system; (B) HEP3 can main the diversity and stability of the whole ecological system in IBD model mice.
Figure 12
Figure 12
Dominant species interaction-associated network analysis results. Nodes represent the dominant species under the identity of different colors: the red line shows a positive correlation and the green line shows a negative correlation. More connections by nodes indicated more association with others. (A) Antibiotics rapidly declined the diversity and destroyed the stability of the whole ecological system; (B) HEP3 can main the diversity and stability of the whole ecological system in IBD model mice.

References

    1. Ko JL, Hsu CI, Lin RH, Kao CL, Lin JY. A new fungal immunomodulatory protein, FIP-fve isolated from the edible mushroom, Flammulina velutipes and its complete amino acid sequence. Eur J Biochem (1995) 228:244–9.10.1111/j.1432-1033.1995.tb20256.x
    1. Kawagishi H, Mori H. Chemical modification and NMR studies on a mushroom lectin Ischnoderma resinosum agglutinin (IRA). Biochim Biophys Acta (1991) 1076:179–86.10.1016/0167-4838(91)90263-Y
    1. Kawagishi H, Takagi JI, Taira T, Murata T, Usui T. Purification and characterization of a lectin from the mushroom Mycoleptodonoides aitchisonii. Phytochemistry (2001) 56:53–8.10.1016/S0031-9422(00)00351-4
    1. Li QZ, Wang XF, Zhou XW. Recent status and prospects of the fungal immunomodulatory protein family. Crit Rev Biotechnol (2011) 31:365–75.10.3109/07388551.2010.543967
    1. Hsin IL, Ou CC, Wu TC, Jan MS, Wu MF, Chiu LY, et al. GMI, an immunomodulatory protein from Ganoderma microsporum, induces autophagy in non-small cell lung cancer cells. Autophagy (2011) 7:873–82.10.4161/auto.7.8.15698
    1. Wang PH, Hsu CI, Tang SC, Huang YL, Lin JY, Ko JL. Fungal immunomodulatory protein from Flammulina velutipes induces interferon-gamma production through p38 mitogen-activated protein kinase signaling pathway. J Agric Food Chem (2004) 52:2721–5.10.1021/jf034556s
    1. Chang YC, Chow YH, Sun HL, Liu YF, Lee YT, Lue KH, et al. Alleviation of respiratory syncytial virus replication and inflammation by fungal immunomodulatory protein FIP-fve from Flammulina velutipes. Antiviral Res (2014) 110:124–31.10.1016/j.antiviral.2014.08.006
    1. Hsieh CW, Lan JL, Meng Q, Cheng YW, Huang HM, Tsai JJ. Eosinophil apoptosis induced by fungal immunomodulatory peptide-fve via reducing IL-5 α receptor. J Formos Med Assoc (2007) 106:36–43.10.1016/S0929-6646(09)60214-X
    1. Kong XH, Zhang JC, Han X, Zhang PQ, Dai XD, Liu JN, et al. High-yield production in Escherichia coli of fungal immunomodulatory protein isolated from Flammulina velutipes and its bioactivity assay in vivo. Int J Mol Sci (2013) 14:2230–41.10.3390/ijms14022230
    1. Singh SS, Wang H, Chan YS, Pan W, Dan X, Yin CM, et al. Lectins from edible mushrooms. Molecules (2014) 20(1):446–69.10.3390/molecules20010446
    1. Isikhuemhen OS, Mikiashvilli NA. Lignocellulolytic enzyme activity, substrate utilization, and mushroom yield by Pleurotus ostreatus cultivated on substrate containing anaerobic digester solids. J Ind Microbiol Biotechnol (2009) 36(11):1353–62.10.1007/s10295-009-0620-1
    1. Adav SS, Ravindran A, Sze SK. Quantitative proteomic analysis of lignocellulolytic enzymes by Phanerochaete chrysosporium on different lignocellulosic biomass. J Proteomics (2012) 75(5):1493–504.10.1016/j.jprot.2011.11.020
    1. Montoya S, Orrego CE, Levin L. Growth, fruiting and lignocellulolytic enzyme production by the edible mushroom Grifola frondosa (maitake). World J Microbiol Biotechnol (2012) 28(4):1533–41.10.1007/s11274-011-0957-2
    1. Lim SH, Lee YH, Kang HW. Efficient recovery of lignocellulolytic enzymes of spent mushroom compost from Oyster mushrooms, Pleurotus spp., and potential use in dye decolorization. Mycobiology (2013) 41(4):214–20.10.5941/MYCO.2013.41.4.214
    1. Choi BS, Sapkota K, Choi JH, Shin CH, Kim S, Kim SJ. Herinase: a novel bi-functional fibrinolytic protease from the monkey head mushroom, Hericium erinaceum. Appl Biochem Biotechnol (2013) 170(3):609–22.10.1007/s12010-013-0206-2
    1. Ali PP, Sapna K, Mol KR, Bhat SG, Chandrasekaran M, Elyas KK. Trypsin inhibitor from edible mushroom Pleurotus floridanus active against proteases of microbial origin. Appl Biochem Biotechnol (2014) 173(1):167–78.10.1007/s12010-014-0826-1
    1. Akanbi MH, Post E, van Putten SM, de Vries L, Smisterova J, Meter-Arkema AH, et al. The antitumor activity of hydrophobin SC3, a fungal protein. Appl Microbiol Biotechnol (2013) 97(10):4385–92.10.1007/s00253-012-4311-x
    1. Erjavec J, Kos J, Ravnikar M, Dreo T, Sabotič J. Proteins of higher fungi – from forest to application. Trends Biotechnol (2012) 30(5):259–73.10.1016/j.tibtech.2012.01.004
    1. De Stefano L, Rea I, De Tommasi E, Rendina I, Rotiroti L, Giocondo M, et al. Bioactive modification of silicon surface using self-assembled hydrophobins from Pleurotus ostreatus. Eur Phys J E Soft Matter (2009) 30(2):181–5.10.1140/epje/i2009-10481-y
    1. Mori K, Inatomi S, Ouchi K, Azumi Y, Tuchida T. Improving effects of the mushroom Yamabushitake (Hericium erinaceus) on mild cognitive impairment: a double-blind placebo-controlled clinical trial. Phytother Res (2009) 23:367–72.10.1002/ptr.2634
    1. Mori K, Obara Y, Hirota M, Azumi Y, Kinugasa S, Inatomi S, et al. Nerve growth factor-inducing activity of Hericium erinaceus in 1321N1 human astrocytoma cells. Biol Pharm Bull (2008) 31:1727–32.10.1248/bpb.31.1727
    1. Wong KH, Vikineswary S, Abdullah N, Naidu M, Keynes R. Activity of aqueous extracts of lion’s mane mushroom Hericium erinaceus (Bull.: Fr.) Pers. (Aphyllophoromycetideae) on the neural cell line NG108-15. Int J Med Mushrooms (2007) 9:57–65.10.1615/IntJMedMushr.v9.i1.70
    1. Jinn CW, Shu HH, Jih TW, Ker SC, Yi CC. Hypoglycemic effect of extract of Hericium erinaceus. J Sci Food Agric (2004) 85:641–6.10.1002/jsfa.1928
    1. Li JJ, Hu XQ, Zhang XF, Liu JJ, Cao LS. Study on variation of main ingredients from spores and fruiting bodies of Ganoderma lucidum. Zhongguo Zhong Yao Za Zhi (2014) 39(21):4246–51.
    1. Kim SP, Nam SH, Friedman M. Correction to Hericium erinaceus (Lion’s Mane) mushroom extracts inhibit metastasis of cancer cells to the lung in CT-26 colon cancer-transplanted mice. J Agric Food Chem (2014) 62:528–528.10.1021/jf405673a
    1. Gong M, An J, Lu HZ, Wu CF, Li YJ, Cheng JQ, et al. Acid modification on fluorescence spectrum and hemagglutinating activity of Hericium erinaceum lectin. Acta Biochim Biophys Sin (2004) 36:343–50.10.1093/abbs/36.5.343
    1. Laemmli UK, Favre M. Maturation of the head of bacteriophage T4. I. DNA packaging events. J Mol Biol (1973) 80(4):575–99.
    1. Ling ZX, Xia L, Jia XY, Cheng YW, Luo YQ, Li Y, et al. Impacts of infection with different toxigenic Clostridium difficile strains on faecal microbiota in children. Sci Rep (2014) 4:7485.10.1038/srep07485
    1. Asnicar F, Weingart G, Tickle TL, Huttenhower C, Segata N. Compact graphical representation of phylogenetic data and metadata with GraPhlAn. Peer J (2015) 3:e1029.10.7717/peerj.1029
    1. Fangkrathok N, Junlatat J, Umehara K, Noguchi H, Sripanidkulchai B. Cytotoxic and immunomodulatory effects of polyhydroxyoctane isolated from Lentinus polychrous mycelia. J Nat Med (2014) 68(2):302–9.10.1007/s11418-013-0797-5
    1. Lin CH, Hsiao YM, Ou CC, Lin YW, Chiu YL, Lue KH, et al. GMI, a Ganoderma immunomodulatory protein, down-regulates tumor necrosis factor α-induced expression of matrix metalloproteinase 9 via NF-κB pathway in human alveolar epithelial A549 cells. J Agric Food Chem (2010) 58(22):12014–21.10.1021/jf103068w
    1. Xu X, Yan H, Chen J, Zhang X. Bioactive proteins from mushrooms. Biotechnol Adv (2011) 29(6):667–74.10.1016/j.biotechadv.2011.05.003
    1. Sun X, Huang W, Xiao S, Liang C, Zhang S, Liu Z, et al. Extracellular expression and efficient purification of a functional recombinant Volvariella volvacea immunomodulatory protein (FIP-vvo) using Pichia pastoris system. Protein Expr Purif (2014) 94:95–100.10.1016/j.pep.2013.10.022
    1. Wong JH, Ng TB, Cheung RC, Ye XJ, Wang HX, Lam SK, et al. Proteins with antifungal properties and other medicinal applications from plants and mushrooms. Appl Microbiol Biotechnol (2010) 87(4):1221–35.10.1007/s00253-010-2690-4
    1. Huang Y, Chen Z. Inflammatory bowel disease related innate immunity and adaptive immunity. Am J Transl Res (2016) 8(6):2490–7.
    1. Corridoni D, Arseneau KO, Cominelli F. Inflammatory bowel disease. Immunol Lett (2014) 161(2):231–5.10.1016/j.imlet.2014.04.004
    1. De Mattos BR, Garcia MP, Nogueira JB, Paiatto LN, Albuquerque CG, Souza CL, et al. Inflammatory bowel disease: an overview of immune mechanisms and biological treatments. Mediators Inflamm (2015) 2015:493012.10.1155/2015/493012
    1. Miyoshi J, Chang EB. The gut microbiota and inflammatory bowel diseases. Transl Res (2017) 179:38–48.10.1016/j.trsl.2016.06.002
    1. Xavier RJ. Microbiota as therapeutic targets. Dig Dis (2016) 34(5):558–65.10.1159/000445263
    1. Vindigni SM, Zisman TL, Suskind DL, Damman CJ. The intestinal microbiome, barrier function, and immune system in inflammatory bowel disease: a tripartite pathophysiological circuit with implications for new therapeutic directions. Therap Adv Gastroenterol (2016) 9(4):606–25.10.1177/1756283X16644242
    1. Geuking MB, Köller Y, Rupp S, McCoy KD. The interplay between the gut microbiota and the immune system. Gut Microbes (2014) 5(3):411–8.10.4161/gmic.29330
    1. Magrone T, Jirillo E. The interplay between the gut immune system and microbiota in health and disease: nutraceutical intervention for restoring intestinal homeostasis. Curr Pharm Des (2013) 19(7):1329–42.10.2174/1381612811319070017
    1. Daillère R, Vétizou M, Waldschmitt N, Yamazaki T, Isnard C, Poirier-Colame V, et al. Enterococcus hirae and Barnesiella intestinihominis facilitate cyclophosphamide-induced therapeutic immunomodulatory effects. Immunity (2016) 45(4):931–43.10.1016/j.immuni.2016.09.009
    1. Xu X, Zhang X. Effects of cyclophosphamide on immune system and gut microbiotain mice. Microbiol Res (2015) 171:97–106.10.1016/j.micres.2014.11.002
    1. Xie G, Wang X, Liu P, Wei R, Chen W, Rajani C, et al. Distinctly altered gut microbiota in the progression of liver disease. Oncotarget (2016) 7(15):19355–66.10.18632/oncotarget.8466
    1. Xie JH, Fan ST, Nie SP, Yu Q, Xiong T, Gong D, et al. Lactobacillus plantarum NCU116 attenuates cyclophosphamide-induced intestinal mucosal injury, metabolism and intestinal microbiota disorders in mice. Food Funct (2016) 7(3):1584–92.10.1039/c5fo01516b
    1. Zhernakova A, Kurilshikov A, Bonder MJ, Tigchelaar EF, Schirmer M, Vatanen T, et al. Population-based metagenomics analysis reveals markers for gut microbiome composition and diversity. Science (2016) 352(6285):565–9.10.1126/science.aad3369
    1. Liu JP, Zou WL, Chen SJ, Wei HY, Yin YN, Zou YY, et al. Effects of different diets on intestinal microbiota and nonalcoholic fatty liver disease development. World J Gastroenterol (2016) 22(32):7353–64.10.3748/wjg.v22.i32.7353
    1. Song X, Bao MM, Li DD, Li YM. Advanced glycation in d-galactose induced mouse aging model. Mech Ageing Dev (1999) 108:239–51.10.1016/S0047-6374(99)00022-6
    1. Haider S, Liaquat L, Shahzad S, Sadir S, Madiha S, Batool Z, et al. A high dose of short term exogenous d-galactose administration in young male rats produces symptoms simulating the natural aging process. Life Sci (2015) 124:110–9.10.1016/j.lfs.2015.01.016
    1. Liu YY, Nagpure BV, Wong PT, Bian JS. Hydrogen sulfide protects SH-SY5Y neuronal cells against d-galactose induced cell injury by suppression of advanced glycation end products formation and oxidative stress. Neurochem Int (2013) 62:603–9.10.1016/j.neuint.2012.12.010
    1. Blagosklonny MV. Rejuvenating immunity: “anti-aging drug today” eight years later. Oncotarget (2015) 6(23):19405–12.10.18632/oncotarget.3740
    1. Hassan MA, Rouf R, Tiralongo E, May TW, Tiralongo J. Mushroom lectins: specificity, structure and bioactivity relevant to human disease. Int J Mol Sci (2015) 16(4):7802–38.10.3390/ijms16047802
    1. Kobayashi Y, Kawagishi H. Fungal lectins: a growing family. Methods Mol Biol (2014) 1200:15–38.10.1007/978-1-4939-1292-6_2

Source: PubMed

3
구독하다