5,301
Views
75
CrossRef citations to date
0
Altmetric
Special Issue: Functional Food Science (REVIEW)

Immunomodulation by food: impact on gut immunity and immune cell function

, &
Pages 584-599 | Received 15 Nov 2017, Accepted 23 Jan 2018, Published online: 16 Feb 2018

References

  • Tangye SG, Ma CS, Brink R, et al. The good, the bad and the ugly - TFH cells in human health and disease. Nat Rev Immunol. 2013;13:412–426.
  • Qi H. T follicular helper cells in space-time. Nat Rev Immunol. 2016;16:612–625.
  • Walker JA, Barlow JL, McKenzie AN. Innate lymphoid cells–how did we miss them? Nat Rev Immunol. 2013;13:75–87.
  • Eberl G, Colonna M, Di Santo JP, et al. Innate lymphoid cells: a new paradigm in immunology. Science. 2015;348:aaa6566.
  • Suzuki K, Kawamoto S, Maruya M, et al. GALT: organization and dynamics leading to IgA synthesis. Adv Immunol. 2010;107:153–185.
  • Rezende RM, Weiner HL. History and mechanisms of oral tolerance. Semin Immunol. 2017;30:3–11.
  • Ise W, Nakamura K, Shimizu N, et al. Orally tolerized T cells can form conjugates with APCs but are defective in immunological synapse formation. J Immunol. 2005;175:829–838.
  • Mucida D, Park Y, Kim G, et al. Reciprocal TH17 and regulatory T cell differentiation mediated by retinoic acid. Science. 2007;317:256–260.
  • Shiokawa A, Kotaki R, Takano T, et al. Mesenteric lymph node CD11b- CD103+ PD-L1High dendritic cells highly induce regulatory T cells. Immunology. 2017;152:52–64.
  • Sato A, Hashiguchi M, Toda E, et al. CD11b+ Generation of gut-homing IgA-secreting B cells by intestinal dendritic cells. Peyer’s patch dendritic cells secrete IL-6 and induce IgA secretion from naive B cells. J Immunol. 2003;171:3684–3690.
  • Mora JR, Iwata M, Eksteen B, et al. Generation of gut-homing IgA-secreting B cells by intestinal dendritic cells. Science. 2006;314:1157–1160.
  • Tezuka H, Abe Y, Iwata M, et al. Regulation of IgA production by naturally occurring TNF/iNOS-producing dendritic cells. Nature. 2007;448:929–933.
  • Goodrich ME, McGee DW. Preferential enhancement of B cell IgA secretion by intestinal epithelial cell-derived cytokines and interleukin-2. Immunol Invest. 1999;28:67–75.
  • Atarashi K, Umesaki Y, Honda K. Microbiotal influence on T cell subset development. Semin Immunol. 2011;23:146–153.
  • Hattori M, Taylor TD. The human intestinal microbiome: a new frontier of human biology. DNA Res. 2009;16:1–12.
  • Umesaki Y, Okada Y, Matsumoto S, et al. Segmented filamentous bacteria are indigenous intestinal bacteria that activate intraepithelial lymphocytes and induce MHC class II molecules and fucosyl asialo GM1 glycolipids on the small intestinal epithelial cells in the ex-germ-free mouse. Microbiol Immunol. 1995;39:555–562.
  • Yanagibashi T, Hosono A, Oyama A, et al. IgA production in the large intestine is modulated by a different mechanism than in the small intestine: Bacteroides acidifaciens promotes IgA production in the large intestine by inducing germinal center formation and increasing the number of IgA+ B cells. Immunobiology. 2013;218:645–651.
  • Sudo N, Sawamura S, Tanaka K, et al. The requirement of intestinal bacterial flora for the development of an IgE production system fully susceptible to oral tolerance induction. J Immunol. 1997;159:1739–1745.
  • Agache I, Sugita K, Morita H, et al. The complex type 2 endotype in allergy and asthma: from laboratory to bedside. Curr Allergy Asthma Rep. 2015;15:29.
  • Takahashi T, Nakagawa E, Nara T, et al. Effects of orally ingested Bifidobacterium longum on the mucosal IgA response of mice to dietary antigens. Biosci Biotechnol Biochem. 1998;62:10–15.
  • Tominari K, Sakai F, Yasui H. Prevention of rotavirus-induced diarrhea by preferential secretion of IgA in breast milk via maternal administration of Lactobacillus gasseri SBT2055. J Pediatr Gastroenterol Nutr. 2012;55:66–71.
  • Kikuchi Y, Kunitoh-Asari A, Hayakawa K, et al. Oral administration of Lactobacillus plantarum strain AYA enhances IgA secretion and provides survival protection against influenza virus infection in mice. PLoS One. 2014;9:e86416.
  • Sakai F, Hosoya T, Ono-Ohmachi A, et al. Lactobacillus gasseri SBT2055 induces TGF-β expression in dendritic cells and activates TLR2 signal to produce IgA in the small intestine. PLoS One. 2014;9:e105370.
  • Kotani Y, Kunisawa J, Suzuki Y, et al. Role of Lactobacillus pentosus Strain b240 and the Toll-like receptor 2 axis in Peyer’s patch dendritic cell-mediated immunoglobulin A enhancement. PLoS One. 2014;9:e91857.
  • Nakamura Y, Terahara M, Iwamoto T, et al. Upregulation of Polymeric Immunoglobulin Receptor Expression by the Heat-Inactivated Potential Probiotic Bifidobacterium bifidum OLB6378 in a Mouse Intestinal Explant Model. Scand J Immunol. 2012;75:176–183.
  • Takeda K, Suzuki T, Shimada SI, et al. Interleukin-12 is involved in the enhancement of human natural killer cell activity by Lactobacillus casei Shirota. Clin Exp Immunol. 2006;146:109–115.
  • Makino S, Sato A, Goto A, et al. Enhanced natural killer cell activation by exopolysaccharides derived from yogurt fermented with Lactobacillus delbrueckii ssp. bulgaricus OLL1073R-1. J Dairy Sci. 2016;99:915–923.
  • Ishizuka T, Kanmani P, Kobayashi H, et al. Immunobiotic Bifidobacteria Strains Modulate Rotavirus Immune Response in Porcine Intestinal Epitheliocytes via Pattern Recognition Receptor Signaling. PLoS One. 2016;11:e0152416.
  • Hori T, Kiyoshima J, Shida K, et al. Augmentation of cellular immunity and reduction of influenza virus titer in aged mice fed Lactobacillus casei strain Shirota. Clin Diagn Lab Immunol. 2002;9:105–108.
  • Hosono A, Ozawa A, Kato R, et al. Dietary fructooligosaccharides induce immunoregulation of intestinal IgA secretion by murine Peyer’s patch cells. Biosci Biotechnol Biochem. 2003;67:758–764.
  • Nakanishi Y, Murashima K, Ohara H, et al. Increase in terminal restriction fragments of Bacteroidetes-derived 16S rRNA genes after administration of short-chain fructooligosaccharides. Appl Environ Microbiol. 2006;72:6271–6276.
  • Bakker-Zierikzee AM, Tol EA, Kroes H, et al. Faecal SIgA secretion in infants fed on pre- or probiotic infant formula. Pediatr Allergy Immunol. 2006;17:134–140.
  • Fukuda S, Toh H, Hase K, et al. Bifidobacteria can protect from enteropathogenic infection through production of acetate. Nature. 2011;469:543–547.
  • Kim MH, Kang SG, Park JH, et al. Short-chain fatty acids activate GPR41 and GPR43 on intestinal epithelial cells to promote inflammatory responses in mice. Gastroenterology. 2013;145:396–406.e1-10.
  • Dalonso N, Goldman GH, Gern RMM. β-(1→3), (1→6)-Glucans: medicinal activities, characterization, biosynthesis and new horizons. Appl Microbiol Biotechnol. 2015;99:7893–7906.
  • Brown GD, Gordon S. Immune recognition of fungal β-glucan. Cell Microbiol. 2005;7:471–479.
  • Zekovic DB, Kwiatkowski S, Vrvic MM, et al. Natural and modified (1→3)-β-D-glucan in health promotion and disease alleviation. Crit Rev Biotechnol. 2005;25:205–230.
  • Shi SH, Yang WT, Huang KY, et al. β-glucan from Coriolus versicolor protect mice against S. typhimurium challenge by activation of macrophages. Int J Biol Macromol. 2016;86:352–361.
  • Davis JM, Murphy EA, Brown AS, et al. Effects of moderate exercise and oat beta-glucan on innate immune function and susceptibility to respiratory infection. Am J Physiol Regul Integr Comp Physiol. 2004;286:R366–R372.
  • Yun CH, Estrada A, Van Kessel A, et al. β-Glucan, extracted from oat, enhances disease resistance against bacterial and parasitic infections. FEMS Immunol Med Microbiol. 2003;35:67–75.
  • Dambuza IM, Brown GD. C-type lectins in immunity: recent developments. Current Opin Immunol. 2015;32:21–27.
  • Kakutani R, Adachi Y, Kajiura H, et al. Relationship between structure and immunostimulating activity of enzymatically synthesized glycogen. Carbohydr Res. 2007;342:2371–2379.
  • Kakutani R, Adachi Y, Takata H, et al. Essential role of Toll-like receptor 2 in macrophage activation by glycogen. Glycobiology. 2012;22:146–159.
  • Kakutani R, Adachi Y, Kajiura H, et al. The effect of orally administered glycogen on anti-tumor activity and natural killer cell activity in mice. Int Immunopharmacol. 2012;12:80–87.
  • Maruyama H, Tamauchi H, Iizuka M, et al. The Role of NK cells in antitumor activity of dietary fucoidan from Undaria pinnatifida sporophylls (mekabu). Planta Med. 2006;72:1415–1417.
  • Hayashi K, Nakano T, Hashimoto M, et al. Defensive effects of a fucoidan from brown alga Undaria pinnatifida against herpes simplex virus infection. Int Immunopharmacol. 2008;8:109–116.
  • Hayashi K, Lee J-B, Nakano T, et al. Anti-influenza A virus characteristics of a fucoidan from sporophyll of Undaria pinnatifida in mice with normal and compromised immunity. Microb Infection. 2013;15:302–309.
  • Zou P, Yang X, Wang J, et al. Advances in characterisation and biological activities of chitosan and chitosan oligosaccharides. Food Chem. 2016;190:1174–1181.
  • Lundahl MLE, Scanlan EM, Lavelle EC. Therapeutic potential of carbohydrates as regulators of macrophage activation. Biochem Pharmacol. 2017;146:23–41.
  • Ma J-X, Qian L, Zhou Y. Stimulation effect of chitosan on the immunity of radiotherapy patients suffered from lung cancer. Int J Biol Macromol. 2015;72:195–198.
  • Iwata M, Hirakiyama A, Eshima Y, et al. Retinoic acid imprints gut-homing specificity on T cells. Immunity. 2004;21:527–538.
  • Kim MH, Taparowsky EJ, Kim CH. Retinoic acid differentially regulates the migration of innate lymphoid cell subsets to the gut. Immunity. 2015;43:107–119.
  • Gebremichael A, Levy EM, Corwin LM. Adherent cell requirement for the effect of vitamin E on in vitro antibody synthesis. J Nutr. 1984;114:1297–1305.
  • Kowdley KV, Mason JB, Meydani SN, et al. Vitamin E deficiency and impaired cellular immunity related to intestinal fat malabsorption. Gastroenterology. 1992;102:2139–2142.
  • Pae M, Wu D. Nutritional modulation of age-related changes in the immune system and risk of infection. Nutr Res. 2017;41:14–35.
  • Jubri Z, Latif AA, Top AG, et al. Perturbation of cellular immune functions in cigarette smokers and protection by palm oil vitamin E supplementation. Nutr J. 2013;12:2–13.
  • Pearce EL, Pearce EJ. Metabolic pathways in immune cell activation and quiescence. Immunity. 2013;38:633–643.
  • Kunisawa J, Sugiura Y, Wake T, et al. Mode of bioenergetic metabolism during B cell differentiation in the intestine determines the distinct requirement for vitamin B1. Cell Rep. 2015;13:122–131.
  • Huskisson E, Maggini S, Ruf M. The role of vitamins and minerals in energy metabolism and well-being. J Int Med Res. 2007;35:277–289.
  • Corbett AJ, Eckle SB, Birkinshaw RW, et al. T-cell activation by transitory neo-antigens derived from distinct microbial pathways. Nature. 2014;509:361–365.
  • Cowley SC. MAIT cells and pathogen defense. Cell Mol Life Sci. 2014;71:4831–4840.
  • Gammoh NZ, Rink L. Zinc in infection and inflammation. Nutrients. 2017;9:624.
  • Zinc Investigators’ Collaborative Group. Prevention of diarrhea and pneumonia by zinc supplementation in children in developing countries: pooled analysis of randomized controlled trials. J Pediatr. 1999;135:689–697.
  • Haase H, Rink L. Functional significance of zinc-related signaling pathways in immune cells. Annu Rev Nutr. 2009;29:133–152.
  • McKenzie RC, Rafferty TS, Beckett GJ. Selenium: an essential element for immune function. Immunol Today. 1998;19:342–345.
  • Connelly-Frost A, Poole C, Satia JA, et al. Selenium, folate, and colon cancer. Nutr Cancer. 2009;61:165–178.
  • Galli C, Calder PC. Effects of fat and fatty acid intake on inflammatory and immune responses: a critical review. Ann Nutr Metab. 2009;55:123–139.
  • de Jong AJ, Kloppenburg M, Toes REM, et al. Fatty acids, lipid mediators, and T-cell function. Front Immunol. 2014;5:483.
  • Kunisawa J, Hashimoto E, Inoue A, et al. Regulation of intestinal IgA responses by dietary palmitic acid and its metabolism. J Immunol. 2014;193:1666–1671.
  • Lamas B, Vergnaud-Gauduchon J, Goncalves-Mendes N, et al. Altered functions of natural killer cells in response to L-arginine availability. Cell Immunol. 2012;280:182–190.
  • Rodriguez PC, Quiceno DG, Ochoa AC. L-arginine availability regulates T-lymphocyte cell-cycle progression. Blood. 2007;109:1568–1573.
  • Rodríguez PC, Ochoa AC. Arginine regulation by myeloid derived suppressor cells and tolerance in cancer: mechanisms and therapeutic perspectives. Immunol Rev. 2008;222:180–191.
  • Norian LA, Rodriguez PC, O’Mara LA, et al. Tumor-infiltrating regulatory dendritic cells inhibit CD8+ T cell function via L-arginine metabolism. Cancer Res. 2009;69:3086–3094.
  • Oberlies J, Watzl C, Giese T, et al. Regulation of NK cell function by human granulocyte arginase. J Immunol. 2009;182:5259–5267.
  • Dufour C, Dandrifosse G, Forget P, et al. Spermine and spermidine induce intestinal maturation in the rat. Gastroenterology. 1988;95:112–116.
  • Deloyer P, Peulen O, Dandrifosse G. Dietary polyamines and non-neoplastic growth and disease. Eur J Gastroenterol Hepatol. 2001;13:1027–1032.
  • van der Hulst RR, von Meyenfeldt MF, Soeters PB. Glutamine: an essential amino acid for the gut. Nutrition. 1996;12:S78–S81.
  • Calder PC, Yaqoob P. Glutamine and the immune system. Amino Acids. 1999;17:227–241.
  • Santos RG, Quirino IE, Viana ML, et al. Effects of nitric oxide synthase inhibition on glutamine action in a bacterial translocation model. Br J Nutr. 2014;111:93–100.
  • Klysz D, Tai X, Robert PA, et al. Glutamine- dependent -ketoglutarate production regulates the balance between T helper 1 cell and regulatory T cell generation. Sci Signal. 2015;8:ra97–ra97.
  • Fallarino F, Grohmann U, You S, et al. Tryptophan catabolism generates autoimmune-preventive regulatory T cells. Transpl Immunol. 2006;17:58–60.
  • Yamamoto S, Hayaishi O. Tryptophan pyrrolase of rabbit intestine d-and l-tryptophan-cleaving enzyme or enzymes. J Biol Chem. 1967;242:5260–5266.
  • Munn DH, Sharma MD, Baban B, et al. GCN2 kinase in T cells mediates proliferative ar- rest and anergy induction in response to indoleamine 2, 3-dioxygenase. Immunity. 2005;22:633–642.
  • Manlapat AK, Kahler DJ, Chandler PR, et al. Cell-autonomous control of interferon type I expression by indoleamine 2, 3-dioxygenase in regulatory CD19+ dendritic cells. Eur J Immunol. 2007;37:1064–1071.
  • Cuevas A, Saavedra N, Salazar LA, et al. Modulation of immune function by polyphenols: possible contribution of epigenetic factors. Nutrients. 2013;5:2314–2332.
  • Burkard M, Leischner C, Lauer UM, et al. Dietary flavonoids and modulation of natural killer cells: implications in malignant and viral diseases. J Nutr Biochem. 2017;46:1–12.
  • Romero-Pérez GA, Egashira M, Harada Y, et al. Orally administered Salacia reticulata extract reduces h1n1 in uenza clinical symptoms in Murine lung Tissues Putatively Due to enhanced natural Killer cell activity. Front Immunol. 2016;7:115.
  • Segawa S, Fujiya M, Konishi H, et al. Probiotic-derived polyphosphate enhances the epithelial barrier function and maintains intestinal homeostasis through integrin-p38 MAPK pathway. PLoS One. 2011;6:e23278.
  • Carver JD. Dietary nucleotides: effects on the immune and gastrointestinal systems. Acta Pediatr Suppl. 1999;430:83–88.
  • Grimble GK, Westwood OM. Nucleotides as immunomodulators in clinical nutrition. Curr Opin Clin Nutr Metab Care. 2001;4:57–64.
  • Nagafuchi S, Hachimura S, Totsuka M, et al. Dietary nucleotides can up-regulate antigen-specific Th1 immune responses and suppress antigen-specific IgE responses in mice. Int Arch Allergy Immunol. 2000;122:33–41.
  • Nagafuchi S, Totsuka M, Hachimura S, et al. Dietary nucleotides increase the proportion of a TCRγδ+ subset of intraepithelial lymphocytes (IEL) and IL-7 production by intestinal epithelial cells (IEC); Implications for modification of cellular and molecular cross-talk between IEL and IEC by dietary nucleotides. Biosci Biotechnol Biochem. 2000;64:1459–1465.
  • Nagafuchi S, Totsuka M, Hachimura S, et al. Dietary nucleotides increase the mucosal IgA response and the secretion of transforming growth factor β from intestinal epithelial cells in mice. Cytotechnology. 2002;40:49–58.
  • Watanabe M, Watanabe J, Sonoyama K, et al. Novel method for producing hypoallergenic wheat flour by enzymatic fragmentation of the constituent allergens and its application to food processing. Biosci Biotechnol Biochem. 2000;64:2663–2667.
  • Du Toit G, Roberts G, Sayre PH, et al. Randomized trial of peanut consumption in infants at risk for peanut allergy. N Engl J Med. 2015;372:803–813.
  • Fukuda K, Ishida W, Harada Y, et al. Efficacy of oral immunotherapy with a rice-based edible vaccine containing hypoallergenic Japanese cedar pollen allergens for treatment of established allergic conjunctivitis in mice. Allergol Int. 2017;S1323-8930(17):30070-9.
  • Björkstén B, Sepp E, Julge K, et al. Allergy development and the intestinal microflora during the first year of life. J Allergy Clin Immunol. 2001;108:516–520.
  • Kalliomäki M, Kirjavainen P, Eerola E, et al. Distinct patterns of neonatal gut microflora in infants in whom atopy was and was not developing. J Allergy Clin Immunol. 2001;107:129–134.
  • Kalliomäki M, Salminen S, Arvilommi H, et al. Probiotics in primary prevention of atopic disease: a randomised placebo-controlled trial. Lancet. 2001;357:1076–1079.
  • Ishida Y, Nakamura F, Kanzato H, et al. Effect of milk fermented with Lactobacillus acidophilus strain L-92 on symptoms of Japanese cedar pollen allergy: a randomized placebo-controlled trial. Biosci Biotechnol Biochem. 2005;69:1652–1660.
  • Xiao JZ, Kondo S, Yanagisawa N, et al. Probiotics in the treatment of Japanese cedar pollinosis: a double-blind placebo-controlled trial. Clin Exp Allergy. 2006;36:1425–1435.
  • Torii S, Torii A, Itoh K, et al. Effects of oral administration of Lactobacillus acidophilus L-92 on the symptoms and serum markers of atopic dermatitis in children. Int Arch Allergy Immunol. 2011;154:236–245.
  • Ishida Y, Nakamura F, Kanzato H, et al. Clinical effects of Lactobacillus acidophilus strain L-92 on perennial allergic rhinitis: a double-blind, placebo-controlled study. J Dairy Sci. 2005;88:527–533.
  • Lee J, Seto D, Bielory L. Meta-analysis of clinical trials of probiotics for prevention and treatment of pediatric atopic dermatitis. J Allergy Clin Immunol. 2008;121:116–121.
  • Elazab N, Mendy A, Gasana J, et al. Probiotic administration in early life, atopy, and asthma: a meta-analysis of clinical trials. Pediatrics. 2013;132:e666–76.
  • Shida K, Makino K, Morishita A, et al. Lactobacillus casei inhibits antigen-induced IgE secretion through regulation of cytokine production in murine splenocyte cultures. Int Arch Allergy Immunol. 1998;115:278–287.
  • Murosaki S, Yamamoto Y, Ito K, et al. Heat-killed Lactobacillus plantarum L-137 suppresses naturally fed antigen-specific IgE production by stimulation of IL-12 production in mice. J Allergy Clin Immunol. 1998;102:57–64.
  • Fujiwara D, Inoue S, Wakabayashi H, et al. The anti-allergic effects of lactic acid bacteria are strain dependent and mediated by effects on both Th1/Th2 cytokine expression and balance. Int Arch Allergy Immunol. 2004;135:205–215.
  • Kanzato H, Fujiwara S, Ise W, et al. Lactobacillus acidophilus strain L-92 induces apoptosis of antigen-stimulated T cells by modulating dendritic cell function. Immunobiology. 2008;213:399–408.
  • Yoshida T, Fujiwara W, Enomoto M, et al. An increased number of CD4+CD25+ cells induced by an oral administration of Lactobacillus plantarum NRIC0380 are involved in antiallergic activity. Int Arch Allergy Immunol. 2013;162:283–289.
  • Noguchi S, Hattori M, Sugiyama H, et al. Lactobacillus plantarum NRIC1832 enhances IL-10 production from CD4⁺ T cells in vitro. Biosci Biotechnol Biochem. 2012;76:1925–1931.
  • Yoshida T, Enomoto M, Nakayama S, et al. Induction of ALDH activity in intestinal dendritic cells by Lactobacillus plantarum NRIC0380. Biosci Biotechnol Biochem. 2013;77:1826–1831.
  • Iwabuchi N, Takahashi N, Xiao JZ, et al. Suppressive effects of Bifidobacterium longum on the production of Th2-attracting chemokines induced with T cell-antigen-presenting cell interactions. FEMS Immunol Med Microbiol. 2009;55:324–334.
  • Aoki-Yoshida A, Yamada K, Hachimura S, et al. Enhancement of oral tolerance induction in DO11.10 mice by Lactobacillus gasseri OLL2809 via Increase of Effector Regulatory T Cells. PLoS One. 2016;11:e0158643.
  • Miyauchi E, Ogita T, Miyamoto J, et al. Bifidobacterium longum alleviates dextran sulfate sodium-induced colitis by suppressing IL-17A response: involvement of intestinal epithelial costimulatory molecules. PLoS One. 2013;8:e79735.
  • Shimazu T, Villena J, Tohno M, et al. Immunobiotic Lactobacillus jensenii elicits anti-inflammatory activity in porcine intestinal epithelial cells by modulating negative regulators of the Toll-like receptor signaling pathway. Infect Immun. 2012;80:276–288.
  • Kaji R, Kiyoshima-Shibata J, et al. Bacterial teichoic acids reverse predominant IL-12 production induced by certain Lactobacillus strains into predominant IL-10 production via TLR2-dependent ERK activation in macrophages. J Immunol. 2010;184:3505–3513.
  • Shida K, Kiyoshima-Shibata J, et al. Induction of interleukin-12 by Lactobacillus strains having a rigid cell wall resistant to intracellular digestion. J Dairy Sci. 2006;89:3306–3317.
  • Ichikawa S, Fujii R, Fujiwara D, et al. MyD88 but not TLR2, 4 or 9 is essential for IL-12 induction by lactic acid bacteria. Biosci Biotechnol Biochem. 2007;71:3026–3032.
  • Kawashima T, Kosaka A, Yan H, et al. Double-stranded RNA of intestinal commensal but not pathogenic bacteria triggers production of protective interferon-β. Immunity. 2013;38:1187–1197.
  • Yanagihara S, Kanaya T, Fukuda S, et al. Uromodulin-SlpA binding dictates Lactobacillus acidophilus uptake by intestinal epithelial M cells. Int. Immunol. 2017;29:357–363.
  • Nagura T, Hachimura S, Hashiguchi M, Ueda Y, Kanno T, Kikuchi H, Sayama K, Kaminogawa S. Suppressive effect of dietary raffinose on T-helper 2 cell-mediated immunity. Br J Nutr. 2002;88:421–426.
  • Tsuda M, Arakawa H, Ishii N, et al. Dietary fructooligosaccharides attenuate early activation of CD4+ T cells which produce both Th1 and Th2 cytokines in the intestinal lymphoid tissues of a murine food allergy model. Int Arch Allergy Immunol. 2017;174:121–132.
  • Yasuda A, Inoue KI, Sanbongi C, et al. Dietary supplementation with fructooligosaccharides attenuates airway inflammation related to house dust mite allergen in mice. Int J Immunopathol Pharmacol. 2010;23:727–735.
  • Furusawa Y, Obata Y, Fukuda S, et al. Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells. Nature. 2013;504:446–450.
  • Kimura Y, Sumiyoshi M, Suzuki T, et al. Inhibitory effects of water-soluble low-molecular-weight β-(1,3–1,6) d-glucan purified from Aureobasidium pullulans GM-NH-1A1 strain on food allergic reactions in mice. Int Immunopharmacol. 2007;7:963–972.
  • Yamada J, Hamuro J, Hatanaka H, et al. Alleviation of seasonal allergic symptoms with superfine beta-1,3-glucan: a randomized study. J Allergy Clin Immunol. 2007;119:1119–1126.
  • Maruyama H, Tamauchi H, Hashimoto M, et al. Suppression of Th2 immune responses by mekabu fucoidan from undaria pinnatifida sporophylls. Int Arch Allergy Immunol. 2005;137:289–294.
  • Oomizu S, Yanase Y, Suzuki H, et al. Fucoidan prevents C epsilon germline transcription and NF-κB p52 translocation for IgE production in B cells. Biochem Biophys Res Commun. 2006;350:501–507.
  • Liu B, Lin Q, Yang T, et al. Oat β-glucan ameliorates dextran sulfate sodium (DSS)-induced ulcerative colitis in mice. Food Funct. 2015;6:3454–3463.
  • Lean QY, Eri RD, Fitton JH, et al. Fucoidan extracts ameliorate acute colitis. PLoS ONE. 2015;10:e0128453.
  • Suchecka D, Harasym JP, Wilczak J, et al. Antioxidative and anti-inflammatory effects of high β-glucan concentration purified aqueous extract from oat in experimental model of LPS-induced chronic enteritis. J Func Foods. 2015;14:244–254.
  • Miyata J, Arita M. Role of omega-3 fatty acids and their metabolites in asthma and allergic diseases. Allergol Intern. 2015;64:27–34.
  • Calder PC. Omega-3 fatty acids and inflammatory processes: from molecules to man. Biochem Soc Trans. 2017;45:1105–1115.
  • Wendell SG, Baffi C, Holguin F. Fatty acids, inflammation, and asthma. J Allergy Clin Immunol. 2014;133:1255–1264.
  • Shimanaka Y, Kono N, Taketomi Y, et al. Omega-3 fatty acid epoxides are autocrine mediators that control the magnitude of IgE-mediated mast cell activation. Nat Med. 2017;23(11):nm.4417.
  • Prietl B, Treiber G, Pieber TR, et al. Vitamin D and immune function. Nutrients. 2013;5:2502–2521.
  • Wei R, Christakos S. Mechanisms underlying the regulation of innate and adaptive immunity by Vitamin D. Nutrients. 2015;7:8251–8260.
  • Yepes-Nuñez JJ, Brozek JL, Fiocchi A, et al. Vitamin D supplementation in primary allergy prevention: systematic review of randomized and non-randomized studies. Allergy. 2018;73:37–39.
  • Kunisawa J, Hashimoto E, Ishikawa I, et al. A pivotal role of vitamin B9 in the maintenance of regulatory T cells in vitro and in vivo. PLoS One. 2012;7:e32094.
  • Yamaguchi T, Hirota K, Nagahama K, et al. Control of immune responses by antigen-specific regulatory T cells expressing the folate receptor. Immunity. 2007;27:145–159.
  • Kinoshita M, Kayama H, Kusu T, et al. Dietary folic acid promotes survival of Foxp3+ regulatory T cells in the colon. J Immunol. 2012;189:2869–2878.
  • Kudva AK, Shay AE, Prabhu KS. Selenium and inflammatory bowel disease. Am J Physiol Gastrointest Liver Physiol. 2015;309:G71–G77.
  • Kaushal N, Kudva AK, Patterson AD, et al. Crucial role of macrophage selenoproteins in experimental colitis. J Immunol. 2014;193:3683–3692.
  • Son DO, Satsu H, Kiso Y, et al. Inhibitory effect of carnosine on interleukin-8 production in intestinal epithelial cells through translational regulation. Cytokine. 2008;42:265–276.
  • Hisatsune T, Kaneko J, Kurashige H, et al. Effect of anserine/carnosine supplementation on verbal episodic memory in elderly people. J Alzheimer’s Dis. 2016;50:149–159.
  • Ono-Ohmachi A, Nakajima-Adachi H, Morita Y, Kato K, Hachimura S. Milk basic protein supplementation exerts an anti-inflammatory effect in a food-allergic enteropathy model mouse. J Dairy Sci. In press.
  • Soda K, Kano Y, Nakamura T, et al. Spermine, a natural polyamine, suppresses LFA-1 expression on human lymphocyte. J Immunol. 2005;175:237–245.
  • Kano Y, Soda K, Konishi F. Suppression of LFA-1 expression by spermine is associated with enhanced methylation of ITGAL, the LFA-1 promoter area. PLoS One. 2013;8:e56056.
  • Wang J, Pae M, Meydani SN, et al. Green tea epigallocatechin-3-gallate modulates differentiation of naive CD4+ T cells into specific lineage effector cells. J Mol Med. 2013;91:485–495.
  • Yang J, Yang X, Chu Y, et al. Identification of baicalin as an immunoregulatory compound by controlling t(h)17 cell differentiation. PLoS One. 2011;6:e17164.
  • Wong CP, Nguyen LP, Noh SK, et al. Induction of regulatory T cells by green tea polyphenol EGCG. Immunol Lett. 2011;139:7–13.
  • Yang J, Yang X, Li M. Baicalin, a natural compound, promotes regulatory T cell differentiation. BMC Complement Altern Med. 2012;12:64.
  • Wang HK, Yeh CH, Iwamoto T, et al. Dietary flavonoid naringenin induces regulatory T cells via an aryl hydrocarbon receptor mediated pathway. J Agric Food Chem. 2012;60:2171–2178.
  • Tachibana H, Sunada Y, Miyase T, et al. Identification of a methylated tea catechin as an inhibitor of degranulation in human basophilic KU812 cells. Biosci Biotechnol Biochem. 2000;64:452–454.
  • Maeda-Yamamoto M. Human clinical studies of tea polyphenols in allergy or life style-related diseases. Curr Pharm Des. 2013;19:6148–6155.
  • Yamamoto T, Yoshimura M, Yamaguchi F, et al. Anti-allergic activity of naringenin chalcone from a tomato skin extract. Biosci Biotechnol Biochem. 2004;68:1706–1711.
  • Li Y, Yao J, Han C, et al. Quercetin, inflammation and immunity. Nutrients. 2016;8:167.
  • Shin HS, Bae MJ, Zhao Z, et al. Anti-inflammatory effect of chlorogenic acid on the IL-8 production in Caco-2 cells and the dextran sulfate sodium-induced colitis symptoms in C57BL/6 mice. Food Chem. 2015;168:167–175.
  • Peluso I, Miglio C, Morabito G, et al. Flavonoids and immune function in human: A systematic review. Crit Rev Food Sci Nutr. 2015;55:383–395.

Reprints and Corporate Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

To request a reprint or corporate permissions for this article, please click on the relevant link below:

Academic Permissions

Please note: Selecting permissions does not provide access to the full text of the article, please see our help page How do I view content?

Obtain permissions instantly via Rightslink by clicking on the button below:

If you are unable to obtain permissions via Rightslink, please complete and submit this Permissions form. For more information, please visit our Permissions help page.