Allergy Asthma Immunol Res.  2017 Mar;9(2):107-115. 10.4168/aair.2017.9.2.107.

Role of Regulatory Cells in Oral Tolerance

Affiliations
  • 1Swiss Institute of Allergy and Asthma Research (SIAF), University of Zurich, Davos, Switzerland. akdism@siaf.uzh.ch

Abstract

The immune system is continuously exposed to great amounts of different antigens from both food and intestinal microbes. Immune tolerance to these antigens is very important for intestinal and systemic immune homeostasis. Oral tolerance is a specific type of peripheral tolerance induced by exposure to antigen via the oral route. Investigations on the role of intestinal immune system in preventing hypersensitivity reactions to innocuous dietary and microbial antigens have been intensively performed during the last 2 decades. In this review article, we discuss how food allergens are recognized by the intestinal immune system and draw attention to the role of regulatory T (Treg) and B (Breg) cells in the establishment of oral tolerance and tolerogenic features of intestinal dendritic cells. We also emphasize the potential role of tonsils in oral tolerance induction because of their anatomical location, cellular composition, and possible usage to develop novel ways of specific immunotherapy for the treatment of allergic diseases.

Keyword

Food allergy; oral tolerance; regulatory cells; dendritic cells; Tregs

MeSH Terms

Allergens
Dendritic Cells
Food Hypersensitivity
Homeostasis
Hypersensitivity
Immune System
Immune Tolerance
Immunotherapy
Palatine Tonsil
Peripheral Tolerance
Allergens

Figure

  • Figure Role of regulatory cells in the induction of oral tolerance. Food antigens are acquired by the immune system via several different mechanisms. First of all, dendritic cells (DC) expressing CD11b and CX3CR1 extend dendritic processes between epithelial cells and sample antigens from lumen (1). As CX3CR1+CD11b+ DC do not express CCR7 or migrate to mesenteric lymph node, they mainly contribute to local, antigen-specific expansion of regulatory cells. Additionally, M cells localized in Payer's patches (2), intestinal cells via paracellular (3), or transcellular route (4), and goblet cells (5) might contribute to antigen sampling. Antigens are uptaken by CD11C+CD103+DC. DC expressing CD11c and CD103 co-express CCR7 and migrate to MLN under homeostatic conditions. In MLN produced by of TGF-beta, IL-10, RALDH, and IDO contribute to active promotion and development of Treg and Breg cells. Treg cells differentiated in MLN by CD11C+CD103+ DC up-regulate α4β7 integrin and CCR9 receptor that are responsible for T-cell homing from MLN to the lamina propria. In lamina propria, CD25-Foxp3--, IL-10-and TGF-β-producing Th3/Tr1 cells, CD25+Foxp3+ α4β7+CCR9+ iTreg cells, and IL-10+IgG4+ Breg cells suppress Th2-dependent allergic inflammation by decreasing mast cell activation, IL-4/IL-5/IL-13 production from ILC2 and Th2 cells, and IgE production from plasma cells.


Cited by  3 articles

Decreased CRTH2 Expression and Response to Allergen Re-stimulation on Innate Lymphoid Cells in Patients With Allergen-Specific Immunotherapy
Wat Mitthamsiri, Panitan Pradubpongsa, Atik Sangasapaviliya, Tadech Boonpiyathad
Allergy Asthma Immunol Res. 2018;10(6):662-674.    doi: 10.4168/aair.2018.10.6.662.

Short-term Haze Exposure Predisposes Healthy Volunteers to Nasal Inflammation
Mu Xian, Kuiji Wang, Hongfei Lou, Yang Wang, Luo Zhang, Chengshuo Wang
Allergy Asthma Immunol Res. 2019;11(5):632-643.    doi: 10.4168/aair.2019.11.5.632.

Allergen-Specific Immunotherapies for Food Allergy
Elizabeth Feuille, Anna Nowak-Wegrzyn
Allergy Asthma Immunol Res. 2018;10(3):189-206.    doi: 10.4168/aair.2018.10.3.189.


Reference

1. Muraro A, Werfel T, Hoffmann-Sommergruber K, Roberts G, Beyer K, Bindslev-Jensen C, et al. EAACI food allergy and anaphylaxis guidelines: diagnosis and management of food allergy. Allergy. 2014; 69:1008–1025.
2. Sicherer SH, Sampson HA. Food allergy: Epidemiology, pathogenesis, diagnosis, and treatment. J Allergy Clin Immunol. 2014; 133:291–307.
3. Zuidmeer L, Goldhahn K, Rona RJ, Gislason D, Madsen C, Summers C, et al. The prevalence of plant food allergies: a systematic review. J Allergy Clin Immunol. 2008; 121:1210–1218.e4.
4. Savage JH, Matsui EC, Skripak JM, Wood RA. The natural history of egg allergy. J Allergy Clin Immunol. 2007; 120:1413–1417.
5. Ahn K. The usefulness of component-resolved diagnostics in food allergy. Allergy Asthma Immunol Res. 2014; 6:103–104.
6. Song TW. Diagnostic Decision Points of Specific IgE Titers in Patients With Food Allergy: Are They Appropriate in All Clinical Settings? Allergy Asthma Immunol Res. 2015; 07. 7:309–311.
7. Cavkaytar O, Akdis CA, Akdis M. Modulation of immune responses by immunotherapy in allergic diseases. Curr Opin Pharmacol. 2014; 17:30–37.
8. Akdis CA. Therapies for allergic inflammation: refining strategies to induce tolerance. Nat Med. 2012; 18:736–749.
9. Chang HC, Sehra S, Goswami R, Yao W, Yu Q, Stritesky GL, et al. The transcription factor PU.1 is required for the development of IL-9-producing T cells and allergic inflammation. Nat Immunol. 2010; 11:527–534.
10. Jones CP, Gregory LG, Causton B, Campbell GA, Lloyd CM. Activin A and TGF-beta promote T(H)9 cell-mediated pulmonary allergic pathology. J Allergy Clin Immunol. 2012; 129:1000–1010.e3.
11. Annunziato F, Cosmi L, Liotta F, Maggi E, Romagnani S. Defining the human T helper 17 cell phenotype. Trends Immunol. 2012; 33:505–512.
12. Wolk K, Kunz S, Witte E, Friedrich M, Asadullah K, Sabat R. IL-22 increases the innate immunity of tissues. Immunity. 2004; 21:241–254.
13. Sugimoto K, Ogawa A, Mizoguchi E, Shimomura Y, Andoh A, Bhan AK, et al. IL-22 ameliorates intestinal inflammation in a mouse model of ulcerative colitis. J Clin Invest. 2008; 118:534–544.
14. Farfariello V, Amantini C, Nabissi M, Morelli MB, Aperio C, Caprodossi S, et al. IL-22 mRNA in peripheral blood mononuclear cells from allergic rhinitic and asthmatic pediatric patients. Pediatr Allergy Immunol. 2011; 22:419–423.
15. Chu DK, Llop-Guevara A, Walker TD, Flader K, Goncharova S, Boudreau JE, et al. IL-33, but not thymic stromal lymphopoietin or IL-25, is central to mite and peanut allergic sensitization. J Allergy Clin Immunol. 2013; 131:187–200.e1-8.
16. Turnquist HR, Thomson AW. IL-33 broadens its repertoire to affect DC. Eur J Immunol. 2009; 39:3292–3295.
17. Annunziato F, Romagnani C, Romagnani S. The 3 major types of innate and adaptive cell-mediated effector immunity. J Allergy Clin Immunol. 2015; 135:626–635.
18. Mowat AM. Anatomical basis of tolerance and immunity to intestinal antigens. Nat Rev Immunol. 2003; 3:331–341.
19. Spahn TW, Weiner HL, Rennert PD, Lügering N, Fontana A, Domschke W, et al. Mesenteric lymph nodes are critical for the induction of high-dose oral tolerance in the absence of Peyer's patches. Eur J Immunol. 2002; 32:1109–1113.
20. McDole JR, Wheeler LW, McDonald KG, Wang B, Konjufca V, Knoop KA, et al. Goblet cells deliver luminal antigen to CD103+ dendritic cells in the small intestine. Nature. 2012; 483:345–349.
21. Tordesillas L, Gómez-Casado C, Garrido-Arandia M, Murua-García A, Palacín A, Varela J, et al. Transport of Pru p 3 across gastrointestinal epithelium - an essential step towards the induction of food allergy? Clin Exp Allergy. 2013; 43:1374–1383.
22. Akdis CA, Akdis M. Mechanisms of immune tolerance to allergens: role of IL-10 and Tregs. J Clin Invest. 2014; 124:4678–4680.
23. Jutel M, Kosowska A, Smolinska S. Allergen Immunotherapy: Past, Present, and Future. Allergy Asthma Immunol Res. 2016; 8:191–197.
24. Wells HG, Osborne TB. The biological reactions of the vegetable proteins. J Infect Dis. 1911; 8:66–124.
25. Chehade M, Mayer L. Oral tolerance and its relation to food hypersensitivities. J Allergy Clin Immunol. 2005; 115:3–12.
26. Chen YH, Weiner HL. Dose-dependent activation and deletion of antigen-specific T cells following oral tolerance. Ann N Y Acad Sci. 1996; 778:111–121.
27. Du Toit G, Roberts G, Sayre PH, Bahnson HT, Radulovic S, Santos AF, et al. Randomized trial of peanut consumption in infants at risk for peanut allergy. N Engl J Med. 2015; 372:803–813.
28. Burks AW, Laubach S, Jones SM. Oral tolerance, food allergy, and immunotherapy: implications for future treatment. J Allergy Clin Immunol. 2008; 121:1344–1350.
29. Schiavi E, Smolinska S, O'Mahony L. Intestinal dendritic cells. Curr Opin Gastroenterol. 2015; 31:98–103.
30. Berin MC, Shreffler WG. Mechanisms underlying induction of tolerance to foods. Immunol Allergy Clin North Am. 2016; 36:87–102.
31. Schulz O, Jaensson E, Persson EK, Liu X, Worbs T, Agace WW, et al. Intestinal CD103+, but not CX3CR1+, antigen sampling cells migrate in lymph and serve classical dendritic cell functions. J Exp Med. 2009; 206:3101–3114.
32. Denning TL, Wang YC, Patel SR, Williams IR, Pulendran B. Lamina propria macrophages and dendritic cells differentially induce regulatory and interleukin 17-producing T cell responses. Nat Immunol. 2007; 8:1086–1094.
33. Bogunovic M, Ginhoux F, Helft J, Shang L, Hashimoto D, Greter M, et al. Origin of the lamina propria dendritic cell network. Immunity. 2009; 31:513–525.
34. Matteoli G, Mazzini E, Iliev ID, Mileti E, Fallarino F, Puccetti P, et al. Gut CD103+ dendritic cells express indoleamine 2,3-dioxygenase which influences T regulatory/T effector cell balance and oral tolerance induction. Gut. 2010; 59:595–604.
35. Mora JR, Iwata M, Eksteen B, Song SY, Junt T, Senman B, et al. Generation of gut-homing IgA-secreting B cells by intestinal dendritic cells. Science. 2006; 314:1157–1160.
36. Hadis U, Wahl B, Schulz O, Hardtke-Wolenski M, Schippers A, Wagner N, et al. Intestinal tolerance requires gut homing and expansion of FoxP3+ regulatory T cells in the lamina propria. Immunity. 2011; 34:237–246.
37. Abbas AK, Benoist C, Bluestone JA, Campbell DJ, Ghosh S, Hori S, et al. Regulatory T cells: recommendations to simplify the nomenclature. Nat Immunol. 2013; 14:307–308.
38. Carrier Y, Yuan J, Kuchroo VK, Weiner HL. Th3 cells in peripheral tolerance. I. Induction of Foxp3-positive regulatory T cells by Th3 cells derived from TGF-beta T cell-transgenic mice. J Immunol. 2007; 178:179–185.
39. Ng TH, Britton GJ, Hill EV, Verhagen J, Burton BR, Wraith DC. Regulation of adaptive immunity; the role of interleukin-10. Front Immunol. 2013; 4:129.
40. Palomares O, Yaman G, Azkur AK, Akkoc T, Akdis M, Akdis CA. Role of Treg in immune regulation of allergic diseases. Eur J Immunol. 2010; 40:1232–1240.
41. Meiler F, Klunker S, Zimmermann M, Akdis CA, Akdis M. Distinct regulation of IgE, IgG4 and IgA by T regulatory cells and toll-like receptors. Allergy. 2008; 63:1455–1463.
42. Torgerson TR, Linane A, Moes N, Anover S, Mateo V, Rieux-Laucat F, et al. Severe food allergy as a variant of IPEX syndrome caused by a deletion in a noncoding region of the FOXP3 gene. Gastroenterology. 2007; 132:1705–1717.
43. Curotto de Lafaille MA, Kutchukhidze N, Shen S, Ding Y, Yee H, Lafaille JJ. Adaptive Foxp3+ regulatory T cell-dependent and -independent control of allergic inflammation. Immunity. 2008; 29:114–126.
44. Josefowicz SZ, Niec RE, Kim HY, Treuting P, Chinen T, Zheng Y, et al. Extrathymically generated regulatory T cells control mucosal TH2 inflammation. Nature. 2012; 482:395–399.
45. Shreffler WG, Wanich N, Moloney M, Nowak-Wegrzyn A, Sampson HA. Association of allergen-specific regulatory T cells with the onset of clinical tolerance to milk protein. J Allergy Clin Immunol. 2009; 123:43–52.e7.
46. Brimnes J, Allez M, Dotan I, Shao L, Nakazawa A, Mayer L. Defects in CD8+ regulatory T cells in the lamina propria of patients with inflammatory bowel disease. J Immunol. 2005; 174:5814–5822.
47. Arnaboldi PM, Roth-Walter F, Mayer L. Suppression of Th1 and Th17, but not Th2, responses in a CD8(+) T cell-mediated model of oral tolerance. Mucosal Immunol. 2009; 2:427–438.
48. Rosser EC, Mauri C. Regulatory B cells: origin, phenotype, and function. Immunity. 2015; 42:607–612.
49. van de Veen W, Stanic B, Yaman G, Wawrzyniak M, Söllner S, Akdis DG, et al. IgG4 production is confined to human IL-10-producing regulatory B cells that suppress antigen-specific immune responses. J Allergy Clin Immunol. 2013; 131:1204–1212.
50. Stanic B, van de Veen W, Wirz OF, Rückert B, Morita H, Söllner S, et al. IL-10-overexpressing B cells regulate innate and adaptive immune responses. J Allergy Clin Immunol. 2015; 135:771–780.e8.
51. Kim AR, Kim HS, Kim DK, Nam ST, Kim HW, Park YH, et al. Mesenteric IL-10-producing CD5(+) regulatory B cells suppress cow's milk casein-induced allergic responses in mice. Sci Rep. 2016; 6:19685.
52. Kim EH, Bird JA, Kulis M, Laubach S, Pons L, Shreffler W, et al. Sublingual immunotherapy for peanut allergy: clinical and immunologic evidence of desensitization. J Allergy Clin Immunol. 2011; 127:640–646.e1.
53. Jones SM, Pons L, Roberts JL, Scurlock AM, Perry TT, Kulis M, et al. Clinical efficacy and immune regulation with peanut oral immunotherapy. J Allergy Clin Immunol. 2009; 124:292–300. 300.e1–300.e97.
54. Rachid R, Umetsu DT. Immunological mechanisms for desensitization and tolerance in food allergy. Semin Immunopathol. 2012; 34:689–702.
55. Karlsson MR, Johansen FE, Kahu H, Macpherson A, Brandtzaeg P. Hypersensitivity and oral tolerance in the absence of a secretory immune system. Allergy. 2010; 65:561–570.
56. Kim BJ, Lee SY, Kim HB, Lee E, Hong SJ. Environmental changes, microbiota, and allergic diseases. Allergy Asthma Immunol Res. 2014; 6:389–400.
57. Hooper LV, Littman DR, Macpherson AJ. Interactions between the microbiota and the immune system. Science. 2012; 336:1268–1273.
58. Bashir ME, Louie S, Shi HN, Nagler-Anderson C. Toll-like receptor 4 signaling by intestinal microbes influences susceptibility to food allergy. J Immunol. 2004; 172:6978–6987.
59. Hill DA, Siracusa MC, Abt MC, Kim BS, Kobuley D, Kubo M, et al. Commensal bacteria-derived signals regulate basophil hematopoiesis and allergic inflammation. Nat Med. 2012; 18:538–546.
60. Lyons A, O'Mahony D, O'Brien F, MacSharry J, Sheil B, Ceddia M, et al. Bacterial strain-specific induction of Foxp3+ T regulatory cells is protective in murine allergy models. Clin Exp Allergy. 2010; 40:811–819.
61. Konieczna P, Ferstl R, Ziegler M, Frei R, Nehrbass D, Lauener RP, et al. Immunomodulation by Bifidobacterium infantis 35624 in the murine lamina propria requires retinoic acid-dependent and independent mechanisms. PLoS One. 2013; 8:e62617.
62. Konieczna P, Schiavi E, Ziegler M, Groeger D, Healy S, Grant R, et al. Human dendritic cell DC-SIGN and TLR-2 mediate complementary immune regulatory activities in response to Lactobacillus rhamnosus JB-1. PLoS One. 2015; 10:e0120261.
63. Rodriguez B, Prioult G, Bibiloni R, Nicolis I, Mercenier A, Butel MJ, et al. Germ-free status and altered caecal subdominant microbiota are associated with a high susceptibility to cow's milk allergy in mice. FEMS Microbiol Ecol. 2011; 76:133–144.
64. Noval Rivas M, Burton OT, Wise P, Zhang YQ, Hobson SA, Garcia Lloret M, et al. A microbiota signature associated with experimental food allergy promotes allergic sensitization and anaphylaxis. J Allergy Clin Immunol. 2013; 131:201–212.
65. Rodriguez B, Prioult G, Hacini-Rachinel F, Moine D, Bruttin A, Ngom-Bru C, et al. Infant gut microbiota is protective against cow's milk allergy in mice despite immature ileal T-cell response. FEMS Microbiol Ecol. 2012; 79:192–202.
66. Noval Rivas M, Burton OT, Wise P, Charbonnier LM, Georgiev P, Oettgen HC, et al. Regulatory T cell reprogramming toward a Th2-cell-like lineage impairs oral tolerance and promotes food allergy. Immunity. 2015; 42:512–523.
67. Ohnmacht C, Park JH, Cording S, Wing JB, Atarashi K, Obata Y, et al. MUCOSAL IMMUNOLOGY. The microbiota regulates type 2 immunity through RORgammat(+) T cells. Science. 2015; 349:989–993.
68. Frei R, Lauener RP, Crameri R, O'Mahony L. Microbiota and dietary interactions: an update to the hygiene hypothesis? Allergy. 2012; 67:451–461.
69. Tan J, McKenzie C, Potamitis M, Thorburn AN, Mackay CR, Macia L. The role of short-chain fatty acids in health and disease. Adv Immunol. 2014; 121:91–119.
70. Smolinska S, Jutel M, Crameri R, O'Mahony L. Histamine and gut mucosal immune regulation. Allergy. 2014; 69:273–281.
71. Frei R, Ferstl R, Konieczna P, Ziegler M, Simon T, Rugeles TM, et al. Histamine receptor 2 modifies dendritic cell responses to microbial ligands. J Allergy Clin Immunol. 2013; 132:194–204.
72. McClory S, Hughes T, Freud AG, Briercheck EL, Martin C, Trimboli AJ, et al. Evidence for a stepwise program of extrathymic T cell development within the human tonsil. J Clin Invest. 2012; 122:1403–1415.
73. Palomares O, Rückert B, Jartti T, Kücüksezer UC, Puhakka T, Gomez E, et al. Induction and maintenance of allergen-specific FOXP3+ Treg cells in human tonsils as potential first-line organs of oral tolerance. J Allergy Clin Immunol. 2012; 129:510–520.e1-9.
74. Kücüksezer UC, Palomares O, Rückert B, Jartti T, Puhakka T, Nandy A, et al. Triggering of specific Toll-like receptors and proinflammatory cytokines breaks allergen-specific T-cell tolerance in human tonsils and peripheral blood. J Allergy Clin Immunol. 2013; 131:875–885.
75. Wawrzyniak P, Wawrzyniak M, Wanke K, Sokolowska M, Bendelja K, Rückert B, et al. Regulation of bronchial epithelial barrier integrity by type 2 cytokines and histone deacetylases in asthmatic patients. J Allergy Clin Immunol. 2016.
76. Akdis M, Aab A, Altunbulakli C, Azkur K, Costa RA, Crameri R, et al. Interleukins (from IL-1 to IL-38), interferons, transforming growth factor β, and TNF-α: Receptors, functions, and roles in diseases. J Allergy Clin Immunol. 2016.
77. Wawrzyniak P, Akdis CA, Finkelman FD, Rothenberg ME. Advances and highlights in mechanisms of allergic disease in 2015. J Allergy Clin Immunol. 2016; 137:1681–1696.
Full Text Links
  • AAIR
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr