Allergy Asthma Immunol Res. 2012 May;4(3):113-115. English.
Published online Mar 22, 2012.
Copyright © 2012 The Korean Academy of Asthma, Allergy and Clinical Immunology • The Korean Academy of Pediatric Allergy and Respiratory Disease
Editorial

Immune Tolerance by Induced Regulatory T Cells in Asthma

Inseon S. Choi
    • Department of Allergy, Chonnam National University Medical School, Gwangju, Korea.
Received January 29, 2012; Accepted February 01, 2012.

This is an Open Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/3.0/) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Dr. Ralph M. Steinman, who discovered the dendritic cells (DCs) that play a key role in the adaptive immune response, was awarded the 2011 Nobel Prize in Physiology or Medicine 3 days after his untimely death.1 Dr. Steinman used autologous DCs, cultured with tumor-specific antigens, to treat his own pancreatic cancer; the efficacy of this so-called "DC-based tumor vaccination" has recently been proven, and many researchers are now actively investigating this field. In contrast to the strategy of treating diseases by inducing active immunity, a strategy that involves suppressing unnecessary active immunity is required to avoid the development of autoimmune or allergic diseases, as these ailments occur due to failure to achieve immune tolerance to self or innocuous foreign materials. An article by Jang et al.,2 which is published in this issue, demonstrates that Lactobacillus rhamnosus prevents the development of asthmatic reactions by inducing immune tolerance via regulatory T (Treg) cells in a mouse asthma model. This study highlights the possibility of preventing or treating asthma using cutting-edge immunological techniques.

Mechanisms of immune tolerance include the deletion of self-reactive T cells in the thymus (central tolerance) and the deletion, ignorance, anergy, inhibition, suppression, or deviation of the cells in peripheral organs (peripheral tolerance); Treg cells mediate immune suppression.3 Since Sakaguchi et al.4 first described in 1995 that T cells expressing interleukin (IL)-2 receptor α chain (CD25) are self-tolerant, it has been shown that the transcription factor forkhead box P3 (Foxp3) is a prerequisite for the development and function of immune suppression of such Treg cells. In fact, immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome, a disease that arises due to a mutation in the Foxp3 gene, results in various severe autoimmune and allergic diseases. As both innate and adaptive immunity work in response to bacterial infection, Treg cells come in two varieties: natural Treg (nTreg) cells, which are produced in the thymus, and adaptive or induced Treg (iTreg) cells, which are induced by antigen and transforming growth factor (TGF)-β, mainly in mucosal tissue.5, 6 nTreg cells make up 5%-10% of peripheral CD4+ T cells and express CD4, CD25, and Foxp3 on their surface, but their IL-7 receptor (CD127) expression is low, and they develop early and play a primary role in self-tolerance. There are three types of iTreg cells: those that transform from peripheral naïve (CD4+CD25-Foxp3-CD127high) T cells by acquiring CD25 and Foxp3; Th3 cells (CD4+CD25-Foxp3- latency-associated peptide+), which were discovered in 1994 by a group researching the induction of oral tolerance and which secrete TGF-β;7 and type 1 regulatory T cells (Tr1; CD4+CD25-Foxp3-), which were discovered in 2001 and which secrete IL-10 and TGF-β.8

Immune tolerance is not simply the absence of an immune reaction to an antigen, but an active immune process that prevents inappropriate immune responses. In healthy humans, Tr1 cells are generated in response to inhaled allergens in the presence of partially matured DCs or plasmacytoid DCs, and, thus, Tr1 cells are the dominant T cell subset in healthy persons, while Th2 cells dominate in allergic individuals.9, 10 The generation of Tr1 cells depends on DC-derived IL-10, IL-27, and TGF-β along with inducible costimulatory molecule (ICOS) signaling by DCs;11 however, IL-10 expression is diminished in allergic airways.12 Inhaled allergens induce indoleamine 2,3-dioxygenase (IDO) in airway DCs, which catalyze tryptophan metabolism to produce TGF-β, suppress IL-6, and promote the generation of Foxp3+ iTreg cells.11 Because the function, but not the number, of CD4+CD25+Foxp3+ Treg cells in atopy and allergic rhinitis,13 the number of Tr1 cells in allergic rhinitis,14 and both the function and number of CD4+CD25+Foxp3+ Treg cells all decrease in asthma,15 and because the lung is normally a Th2-biased compartment,10 atopic individuals develop allergic diseases via Th2 responses, instead of developing tolerance to innocuous allergens. A recent study reported Foxp3 gene polymorphisms in allergic rhinitis.16 IL-6 suppresses Foxp3 expression and iTreg cell generation to release the suppression of active immunity by Treg cells and induces Th17 cell differentiation, resulting in neutrophilic asthma.6, 10

Allergen-specific immunotherapy (SIT) is a well-known strategy to develop immune tolerance to innocuous allergens that is naturally present in healthy persons and, thus, to modify the natural course of asthma. This treatment induces Tr1 cells17 and Foxp3+ iTreg cells18 and thus suppresses pathogenic immune responses through secretion of IL-10 and TGF-β. In addition, sublingual immunotherapy (SLIT) may induce Th3 cells, as can be seen in oral tolerance.19 The vitamin A metabolite retinoic acid, another agent that promotes the differentiation of Foxp3+ iTreg cells, suppresses IL-6 production by DC and CD44hi effector memory T cells that suppress TGF-β. Vitamin D (calcitriol) and glucocorticoid also induce Foxp3+ iTreg cells by promoting TGF-β production by DC.11 In addition, both calcitriol and glucocorticoid convert CD4+ T cells into Tr1 cells, and the effect by calcitriol, but not glucocorticoid, is associated with programmed death ligand (PDL)-1 expression in DCs.20 CpG induces Foxp3+ iTreg cells by binding to Toll-like receptor (TLR) 9 and activating IDO,21 and bacille Calmette-Guérin vaccination induces those cells in association with ICOS signaling.22

It has been thought that immune tolerance to inhaled allergens in healthy persons, or when acquired after SIT in allergy patients, depends on Tr1 cells, not nTreg cells or Th3 cells, and that the transient expression of CD25 and Foxp3 in some iTreg cells may simply be a feature of activated effector T cells.23 However, because allergy is severe in patients with IPEX syndrome, a disease due to a mutation in the Foxp3 gene, it is apparent that Foxp3 plays an important role in immune tolerance to allergens. In addition, CD25+ T cells that are activated nonspecifically without allergen suppress Th2 cells,24 and helminth infestation is associated with an increased number of Foxp3+ Treg cells and a low prevalence of allergic diseases.25 Therefore, nTreg cells may prevent allergic diseases in an allergen-nonspecific manner, but iTreg cells that develop after encountering allergens in the periphery mediate allergen-specific immune tolerance.25 However, among self-reactive T cells, only cells expressing Foxp3 can survive when they receive T cell receptor (TCR) signals in the thymus,26 and nTreg cells proliferate when their TCRs recognize a specific antigen to become antigen-specific Treg cells.27 More importantly, Foxp3+ iTreg cells can be induced by the abovementioned agents, including SIT;11, 18, 21, 22 consistent with this, Lactobacillus rhamnosus initiates immune tolerance to ovalbumin-induced allergic airway reactions by inducing Foxp3+ iTreg cells, as presented later in this issue.2

Intestinal commensal bacteria have an immunomodulatory effect, and reduction in gut probiotics, such as Lactobacillus or Bifidobacterium species, is associated with atopy.28 In 1997, Majamaa and Isolauri29 first introduced probiotics for treating allergies by demonstrating that Lactobacillus GG ingestion for 1 month significantly improved atopic dermatitis in infants. In 2007, Feleszko et al.30 demonstrated that oral administration of Lactobacillus rhamnosus GG or Bifidobacterium lactis for 8 weeks to newborn mice suppressed asthmatic airway reactions in association with increased Foxp3+ T cells in peribronchial lymph nodes. In addition, Forsythe et al.31 showed that Lactobacillus reuteri, but not Lactobacillus salivarius, attenuated asthmatic reactions, which was dependent on TLR9 and associated with increased IDO activity. The beneficial effects of probiotics seem to be strain-specific, and treatment may be most effective during the neonatal period. Orally administered probiotics induce IL-10 production and IDO activation systemically, and thus suppress allergic reactions in the airways.28 Treg cells induced by probiotics cause "bystander suppression" in an antigen-nonspecific manner, and create a regulatory milieu that promotes antigen-specific Treg cell production. Moreover, combined treatment with Bifidobacterium bifidum and SLIT using birch pollen extract has been shown to be superior to SLIT alone in inducing Foxp3+ iTreg cells and suppressing asthmatic reactions.32 Therefore, probiotic therapy for inducing immune tolerance to innocuous allergens is promising, but appropriate selection of the strain(s), administration timing, selection of subjects, and allergen-specificity, among other things, should be carefully considered.

As treatments using autologous iTreg cells expanded in vitro are actively pursued in the field of transplantation and autoimmune diseases, further investigations of such treatments for asthma and allergies are warranted. nTreg cells show complete demethylation in CpG motif methylation of the Foxp3 locus, conserved non-coding DNA sequence (CNS) 2 (formerly called the Treg cell-specific demethylated region [TSDR]),33 and thus their Foxp3 expression is stable.34 In contrast, iTreg cells show partial demethylation with TGF-β dependency, and thus their Foxp3 expression is unstable and progressively decreases over time. Therefore, to maintain the efficacy of Foxp3+ iTreg cells, repeated induction of the cells should be done, or methods to stabilize their Foxp3 expression should be investigated. It has been reported that when Foxp3+ iTreg cells are expanded in vitro, their function is occasionally lost or they are converted into effector T cells, resulting in the opposite of immune tolerance,35 a problem that needs to be resolved. Because CD127 expression is high in effector T cells, but low in Treg cells, it has been suggested that the combination of CD127 with other markers, such as CD25, may be useful for distinguishing between these T cell subsets.36 However, there is still no apparent marker that can distinguish between Foxp3+ iTreg cells and nTreg cells, even though the TCR repertoire and induction signal for development of the cells (i.e., CD28 and CTLA-4) differ between nTreg and Foxp3+ iTreg cells.26, 34, 35

Notes

There are no financial or other issues that might lead to conflict of interest.

References

    1. Ciechanover AJ, Sznajder JI. Innate and adaptive immunity: the 2011 Nobel Prize in physiology or medicine. Am J Respir Crit Care Med 2011;184:i–ii.
    1. Jang SO, Kim HJ, Kim YJ, Kang MJ, Kwon JW, Seo JH, Kim HY, Kim BJ, Yu J, Hong SJ. Asthma prevention by Lactobacillus rhamnosus in a mouse model is associated with CD4+CD25+Foxp3+ T Cells. Allergy Asthma Immunol Res 2012;4:150–156.
    1. Kamradt T, Mitchison NA. Tolerance and autoimmunity. N Engl J Med 2001;344:655–664.
    1. Sakaguchi S, Sakaguchi N, Asano M, Itoh M, Toda M. Immunologic self-tolerance maintained by activated T cells expressing IL-2 receptor alpha-chains (CD25). Breakdown of a single mechanism of self-tolerance causes various autoimmune diseases. J Immunol 1995;155:1151–1164.
    1. Chatila TA. Molecular mechanisms of regulatory T-cell development. Chem Immunol Allergy 2008;94:16–28.
    1. Venuprasad K, Kong YC, Farrar MA. Control of Th2-mediated inflammation by regulatory T cells. Am J Pathol 2010;177:525–531.
    1. Weiner HL, da Cunha AP, Quintana F, Wu H. Oral tolerance. Immunol Rev 2011;241:241–259.
    1. Roncarolo MG, Bacchetta R, Bordignon C, Narula S, Levings MK. Type 1 T regulatory cells. Immunol Rev 2001;182:68–79.
    1. 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.
    1. de Heer HJ, Hammad H, Kool M, Lambrecht BN. Dendritic cell subsets and immune regulation in the lung. Semin Immunol 2005;17:295–303.
    1. Kushwah R, Hu J. Role of dendritic cells in the induction of regulatory T cells. Cell Biosci 2011;1:20.
    1. Borish L, Aarons A, Rumbyrt J, Cvietusa P, Negri J, Wenzel S. Interleukin-10 regulation in normal subjects and patients with asthma. J Allergy Clin Immunol 1996;97:1288–1296.
    1. Ling EM, Smith T, Nguyen XD, Pridgeon C, Dallman M, Arbery J, Carr VA, Robinson DS. Relation of CD4+CD25+ regulatory T-cell suppression of allergen-driven T-cell activation to atopic status and expression of allergic disease. Lancet 2004;363:608–615.
    1. Han D, Wang C, Lou W, Gu Y, Wang Y, Zhang L. Allergen-specific IL-10-secreting type I T regulatory cells, but not CD4+CD25+Foxp3+ T cells, are decreased in peripheral blood of patients with persistent allergic rhinitis. Clin Immunol 2010;136:292–301.
    1. Mamessier E, Nieves A, Lorec AM, Dupuy P, Pinot D, Pinet C, Vervloet D, Magnan A. T-cell activation during exacerbations: a longitudinal study in refractory asthma. Allergy 2008;63:1202–1210.
    1. Hassannia H, Abediankenari S, Ghaffari J. FOXP3 and TGF-β gene polymorphisms in allergic rhinitis. Iran J Immunol 2011;8:218–225.
    1. Möbs C, Slotosch C, Löffler H, Jakob T, Hertl M, Pfützner W. Birch pollen immunotherapy leads to differential induction of regulatory T cells and delayed helper T cell immune deviation. J Immunol 2010;184:2194–2203.
    1. Radulovic S, Jacobson MR, Durham SR, Nouri-Aria KT. Grass pollen immunotherapy induces Foxp3-expressing CD4+CD25+ cells in the nasal mucosa. J Allergy Clin Immunol 2008;121:1467–1472. 1472.e1.
    1. Moingeon P, Batard T, Fadel R, Frati F, Sieber J, Van Overtvelt L. Immune mechanisms of allergen-specific sublingual immunotherapy. Allergy 2006;61:151–165.
    1. Unger WW, Laban S, Kleijwegt FS, van der Slik AR, Roep BO. Induction of Treg by monocyte-derived DC modulated by vitamin D3 or dexamethasone: differential role for PD-L1. Eur J Immunol 2009;39:3147–3159.
    1. Baban B, Chandler PR, Sharma MD, Pihkala J, Koni PA, Munn DH, Mellor AL. IDO activates regulatory T cells and blocks their conversion into Th17-like T cells. J Immunol 2009;183:2475–2483.
    1. Gao X, Bai H, Cheng J, Fan Y, Wang S, Jiao L, Xiu N, Yang X. CD8α+ and CD8α- DC subsets from BCG-infected mice inhibit allergic Th2-cell responses by enhancing Th1-cell and Treg-cell activity respectively. Eur J Immunol 2012;42:165–175.
    1. Commins SP, Borish L, Steinke JW. Immunologic messenger molecules: cytokines, interferons, and chemokines. J Allergy Clin Immunol 2010;125:S53–S72.
    1. Stassen M, Jonuleit H, Müller C, Klein M, Richter C, Bopp T, Schmitt S, Schmitt E. Differential regulatory capacity of CD25+ T regulatory cells and preactivated CD25+ T regulatory cells on development, functional activation, and proliferation of Th2 cells. J Immunol 2004;173:267–274.
    1. Umetsu DT, DeKruyff RH. The regulation of allergy and asthma. Immunol Rev 2006;212:238–255.
    1. Josefowicz SZ, Lu LF, Rudensky AY. Regulatory T Cells: Mechanisms of differentiation and function. Annu Rev Immunol 2012;30:531–564.
    1. Hall BM, Verma ND, Tran GT, Hodgkinson SJ. Distinct regulatory CD4+T cell subsets; differences between naïve and antigen specific T regulatory cells. Curr Opin Immunol 2011;23:641–647.
    1. Forsythe P. Probiotics and lung diseases. Chest 2011;139:901–908.
    1. Majamaa H, Isolauri E. Probiotics: a novel approach in the management of food allergy. J Allergy Clin Immunol 1997;99:179–185.
    1. Feleszko W, Jaworska J, Rha RD, Steinhausen S, Avagyan A, Jaudszus A, Ahrens B, Groneberg DA, Wahn U, Hamelmann E. Probiotic-induced suppression of allergic sensitization and airway inflammation is associated with an increase of T regulatory-dependent mechanisms in a murine model of asthma. Clin Exp Allergy 2007;37:498–505.
    1. Forsythe P, Inman MD, Bienenstock J. Oral treatment with live Lactobacillus reuteri inhibits the allergic airway response in mice. Am J Respir Crit Care Med 2007;175:561–569.
    1. Moussu H, Van Overtvelt L, Horiot S, Tourdot S, Airouche S, Zuercher A, Holvoet S, Prioult G, Nutten S, Mercenier A, Mascarell L, Moingeon P. Bifidobacterium bifidum NCC 453 promotes tolerance induction in murine models of sublingual immunotherapy. Int Arch Allergy Immunol 2011;158:35–42.
    1. Zheng Y, Josefowicz S, Chaudhry A, Peng XP, Forbush K, Rudensky AY. Role of conserved non-coding DNA elements in the Foxp3 gene in regulatory T-cell fate. Nature 2010;463:808–812.
    1. Curotto de Lafaille MA, Lafaille JJ. Natural and adaptive foxp3+ regulatory T cells: more of the same or a division of labor? Immunity 2009;30:626–635.
    1. Leguern C. Regulatory T cells for tolerance therapy: revisiting the concept. Crit Rev Immunol 2011;31:189–207.
    1. Liu W, Putnam AL, Xu-Yu Z, Szot GL, Lee MR, Zhu S, Gottlieb PA, Kapranov P, Gingeras TR, Fazekas de St Groth B, Clayberger C, Soper DM, Ziegler SF, Bluestone JA. CD127 expression inversely correlates with FoxP3 and suppressive function of human CD4+ T reg cells. J Exp Med 2006;203:1701–1711.

Metrics
Share
PERMALINK