Elsevier

Cellular Immunology

Volume 359, January 2021, 104251
Cellular Immunology

Review article
Role of orally induced regulatory T cells in immunotherapy and tolerance

https://doi.org/10.1016/j.cellimm.2020.104251Get rights and content

Highlights

  • Oral antigen-based oral immunotherapies (OITs) alleviate allergies and autoimmune diseases.

  • OIT induces expansion of several subtypes of regulatory T cells (Treg) such as FoxP3+ and LAP+ Treg and Tr1 cells.

  • Orally induced Treg exhibit overlapping function and cooperation between each other.

  • Tolerogenic dendritic cells are drivers of Treg cells expansion and differentiation in the small intestine.

  • TGF-β, RA, IL-27 and IDO determine differentiation of orally induced Treg.

Abstract

Oral antigen administration to induce regulatory T cells (Treg) takes advantage of regulatory mechanisms that the gastrointestinal tract utilizes to promote unresponsiveness against food antigens or commensal microorganisms. Recently, antigen-based oral immunotherapies (OITs) have shown efficacy as treatment for food allergy and autoimmune diseases. Similarly, OITs appear to prevent anti-drug antibody responses in replacement therapy for genetic diseases. Intestinal epithelial cells and microbiota possibly condition dendritic cells (DC) toward a tolerogenic phenotype that induces Treg via expression of several mediators, e.g. IL-10, transforming growth factor-β, retinoic acid. Several factors, such as metabolites derived from microbiota or diet, impact the stability and expansion of these induced Treg, which include, but are not limited to, FoxP3+ Treg, LAP+ Treg, and/or Tr1 cells. Here, we review various orally induced Treg, their plasticity and cooperation between the Treg subsets, as well as underlying mechanisms controlling their induction and role in oral tolerance.

Introduction

The small and large intestine are continuously exposed to a large variety of foreign antigens derived from food as well as commensal bacteria. Nevertheless, the intestinal immune system does not necessarily mount cellular or humoral immune responses to non-self-antigens due to regulatory mechanisms. One such mechanism is termed “oral tolerance”, referring to the natural development of induced tolerance to orally ingested stimuli in the gut-associated lymphoid tissue (GALT) [1]. Failure to establish oral tolerance leads to food allergy or the development of intestinal inflammatory diseases [2], [3]. While the colon is thought to respond to the massive saturation stimulation by bacterial or bacterially induced antigens, the immune system of the small intestine is distinct; it processes orally delivered antigens as well as systemic antigens, contains specialized lymphoid structures such as Peyer’s patches, has a distinct microbiome, distinct mechanisms of immune regulation, and uniquely interacts with the mesenteric lymph node (MLN) [4], [5], [6], [7], [8], [9], [10]. Hence oral tolerance mechanisms, including oral induction of regulatory T cells (Treg), take place in the small intestine.

Recently, the term oral immunotherapy (OIT) was coined to define the oral delivery of antigen with the objective to suppress immune responses. Delivering a specific target antigen or antibody that affects T cell function have been the two main methods used to achieve OIT. Alternatives routes of OIT include nasal, sublingual, subcutaneous and epicutaneous administration [11], [12], [13], [14], [15].

Oral tolerance was first demonstrated by Wells and Osbourne more than a century ago [16]. These investigators found that guinea pigs fed with corn-containing diet, but not corn-free diet, failed to show anaphylactic reactions against zein, a major protein of corn. In 1946, another study reported that prior feeding of certain allergenic compounds to non-sensitive subjects induced a state of immunological tolerance against subsequent experimental dermal sensitization with the same compounds [17]. Subsequent studies confirmed the existence of an oral tolerance mechanism. For instance, rats developed tolerance to horse serum or pollen extract when fed with these antigens prior to non-oral exposure [18]. This was also demonstrated in other animal models that were fed bovine serum albumin [19] or sheep red blood cells [20]. However, induction of oral tolerance in humans was only demonstrated in the early 1990s, when adults fed with keyhole limpet hemocyanin followed by subcutaneous immunization with the same antigen were prevented from developing a subsequent delayed type hypersensitivity response [21]. Currently, OIT has been applied toward tolerance induction in autoimmunity and inflammatory diseases such as peanut allergy [22], [23], allergic asthma [24], [25], pollen allergy [26], hepatitis C infection [27], nonalcoholic steatohepatitis (NASH) [28], Pompe disease [29], rheumatoid arthritis [30], [31], type I diabetes [32], [33], hemophilia A and B [34], [35], [36], [37] in clinical as well as preclinical studies.

The development of oral tolerance is thought to take place in both the small and large gastrointestinal (GI) tract, where the GALT plays a key role in regulating responses to ingested antigens [38]. Antigen can be acquired directly by phagocytes or can be delivered through goblet cell associated passages prior to capture by dendritic cells (DCs) in lamina propria (LP) [39]. Antigen uptake by a subset of regulatory DCs expressing CD103, which migrate from the gut mucosa to the MLN, concomitant secretory IgA production [40], forkhead box protein P3 (FoxP3) expressing Treg that produce transforming growth factor β (TGF-β) and IL-10, and expression of indoleamine 2,3-dioxygenase (IDO) or the vitamin A metabolite retinoic acid (RA) [41] are all implicated in this process.

For the last three decades, the role of T cells in oral tolerance induction has been studied in increasing detail. It was found that depletion of CD4+ T cells abolished oral tolerance development [42], and that oral tolerance could be transferred from one animal to another by adoptive CD4+ T cell transfer [43]. Moreover, a population of TGF-β secreting CD4+ T cells termed Th3 were found to play a key role in oral tolerance [44], supporting the role of Treg in the induction of oral tolerance. Another conceptual advance in the oral tolerance field was the discovery of CD4+ T cells expressing a membrane-bound form of TGF-β that contains latency-associated peptide (LAP) [1], [45], [46]. LAP+CD4+ T cells were found to have important immune regulatory functions in oral tolerance. Interestingly, TGF-β is required for the induction of both FoxP3+ Treg and LAP+ Treg [47], [48].

Here we review current understanding of oral tolerance mechanisms, applications of OIT in allergy, autoimmune disease and in inducing tolerance to protein replacement therapies for monogenic disorders. We highlight key regulatory cells that are induced by orally delivered antigen, circumstances leading to their induction and the suppressive mechanisms exerted by them. Understanding these mechanisms are critical to identify new strategies for modulating tolerance.

Section snippets

Subsets of orally induced regulatory T cells

In the intestine, crosstalk among several cells occurs in order to induce a naturally tolerogenic environment. Unlike thymus derived (t)Treg in other organs, which have a self-antigen TCR repertoire, intestinal Treg display a peripheral (p) Treg TCR repertoire responsive to resident and non-resident microbiota and dietary antigens, thus playing a crucial role in controlling pro-inflammatory responses [49], [50], [51], [52]. Both tTreg and pTreg are located in the intestine, but it appears that

Regulatory CD8+ T cells

The majority of cells involved in oral tolerance are thought to be CD4+ T cells, but these may not be the only immune regulatory cells involved in OIT. For example, it has been reported that CD8+ T cells with regulatory activity may be induced upon interaction with intestinal epithelial cells [126]. Regulatory CD8+ T cells express lower levels of FoxP3 compared to CD4+ Treg in mice, rats and humans [127]. In mice, surface markers such as CD122(+) or CD28(−) have been used to identify regulatory

RORγt-expressing Treg

The abundant microbial community found in the intestine plays a prominent role in the regulation of oral tolerance. Retinoic acid-related orphan receptor gamma t (RORγt)-expressing Treg (RORγt+ Treg) represent a substantial number among Treg in the intestine and are dependent on the microbiota for maintenance and thus might be sensitive to microbiota shifts [3], [53], [158], [159]. Clostridia and Helicobacter species are examples of bacteria that may induce RORγt+ Treg differentiation in the

Mechanisms of oral Treg induction

Orally induced Treg cells have a crucial role in maintaining tolerance to microbiota, diet and other harmless antigens [50]. Here, we describe mechanisms that mediate Treg differentiation during OIT.

Mechanisms of suppression

Treg preserve homeostasis in the intestinal tract through multiple suppressive mechanisms: production of inhibitory cytokines IL-10, IL-35 and TGF-β; apoptosis or anergy of effector T cells, perforin and granzyme-dependent cytolysis of target cells; suppression of DC maturation and function through expression of PD-1, CTLA4, LAG3; and apoptosis of effector T cells by deprivation of IL-2. Of these, inhibitory cytokines have been studied the most in intestinal Treg.

Examples of OIT-induced Treg in treatment of disease

OIT can be used to alleviate inflammatory disease in an antigen-nonspecific fashion by non-specific enhancement of Treg [1], [97], [216]. This can be accomplished by supply of IL-2, which is critical for Treg development, expansion and maintenance of suppressive function [49], [217], [218]. Several studies have shown that low dose subcutaneous injections of IL-2 leads to an increase in number and function of Treg, thereby ameliorating autoimmune disease in patients [12], [15]. Bonnet and

Conclusions and future perspectives

In the past decade, there has been an increase in knowledge regarding OIT, as well as mechanisms that mediate the development of this process. Three distinct Treg subsets have been shown to majorly contribute to OIT: FoxP3+ Treg, LAP+ Treg and Tr1 cells. These cells share several key characteristics and functions, such as the ability to produce IL-10 and TGF-β, which are critical mediators of immune suppression in OIT. TGF-β is also a key cytokine for FoxP3+ and LAP+ Treg differentiation. CD103+

Acknowledgments

This work was supported by NIH grants R01 HL133191 and R01 HL107904 to HD and RWH; R01 HL131093 to RWH and CT; and U54 HL142012 to RWH. Figures were generated using BioRender.

References (228)

  • M.A. Curotto de Lafaille

    Adaptive Foxp3+ regulatory T cell-dependent and -independent control of allergic inflammation

    Immunity

    (2008)
  • H.L. Weiner

    Oral tolerance: immune mechanisms and the generation of Th3-type TGF-beta-secreting regulatory cells

    Microbes Infect.

    (2001)
  • X. Wang

    Plant-based oral tolerance to hemophilia therapy employs a complex immune regulatory response including LAP+CD4+ T cells

    Blood

    (2015)
  • L. Xu

    Positive and negative transcriptional regulation of the Foxp3 gene is mediated by access and binding of the Smad3 protein to enhancer I

    Immunity

    (2010)
  • M.B. Geuking

    Intestinal bacterial colonization induces mutualistic regulatory T cell responses

    Immunity

    (2011)
  • U. Hadis

    Intestinal tolerance requires gut homing and expansion of FoxP3+ regulatory T cells in the lamina propria

    Immunity

    (2011)
  • J.A. Hill

    Foxp3 transcription-factor-dependent and -independent regulation of the regulatory T cell transcriptional signature

    Immunity

    (2007)
  • C. Campbell

    Extrathymically generated regulatory T cells establish a niche for intestinal border-dwelling bacteria and affect physiologic metabolite balance

    Immunity

    (2018)
  • X.O. Yang

    Molecular antagonism and plasticity of regulatory and inflammatory T cell programs

    Immunity

    (2008)
  • M. Kleinewietfeld et al.

    The plasticity of human Treg and Th17 cells and its role in autoimmunity

    Semin. Immunol.

    (2013)
  • H. Ochi

    New immunosuppressive approaches: oral administration of CD3-specific antibody to treat autoimmunity

    J. Neurol. Sci.

    (2008)
  • K. Forster

    An oral CD3-specific antibody suppresses T-cell-induced colitis and alters cytokine responses to T-cell activation in mice

    Gastroenterology

    (2012)
  • H.L. Weiner

    Oral tolerance

    Immunol. Rev.

    (2011)
  • C. Ohnmacht

    The microbiota regulates type 2 immunity through RORγt+ T cells

    Science

    (2015)
  • S.Y. Chang

    Circulatory antigen processing by mucosal dendritic cells controls CD8(+) T cell activation

    Immunity

    (2013)
  • S.R.P. Kumar

    Role of small intestine and gut microbiome in plant-based oral tolerance for hemophilia

    Front. Immunol.

    (2020)
  • M.S. O'Keeffe

    SLAMF4 is a negative regulator of expansion of cytotoxic intraepithelial CD8+ T cells that maintains homeostasis in the small intestine

    Gastroenterology

    (2015)
  • C. Kuhn et al.

    Immunology. How does the immune system tolerate food?

    Sci. (New York, N.Y.)

    (2016)
  • X. Wang

    Mechanism of oral tolerance induction to therapeutic proteins

    Adv. Drug Deliv. Rev.

    (2013)
  • Y. Zhong

    CD4+LAP + and CD4 +CD25 +Foxp3 + regulatory T cells induced by nasal oxidized low-density lipoprotein suppress effector T cells response and attenuate atherosclerosis in ApoE-/- mice

    J. Clin. Immunol.

    (2012)
  • E. Castela

    Effects of low-dose recombinant interleukin 2 to promote T-regulatory cells in alopecia areata

    JAMA Dermatol.

    (2014)
  • E.H. Kim

    Sublingual immunotherapy for peanut allergy: clinical and immunologic evidence of desensitization

    J. Allergy Clin. Immunol.

    (2011)
  • J.Y. Humrich

    Rapid induction of clinical remission by low-dose interleukin-2 in a patient with refractory SLE

    Ann. Rheum. Dis.

    (2015)
  • H.G. Wells et al.

    The biological reactions of the vegetable proteins I. Anaphylaxis

    J. Infect. Dis.

    (3 January 1911)
  • M.W. Chase

    Inhibition of experimental drug allergy by prior feeding of the sensitizing agent

    Proc. Soc. Exp. Biol. Med.

    (1946)
  • H.C. Thomas et al.

    The induction of tolerance to a soluble protein antigen by oral administration

    Immunology

    (1974)
  • C. André

    A mechanism for the induction of immunological tolerance by antigen feeding: antigen-antibody complexes

    J. Exp. Med.

    (1975)
  • S. Husby

    Oral tolerance in humans. T cell but not B cell tolerance after antigen feeding

    J. Immunol.

    (1994)
  • K. Suzuki

    Prevention of allergic asthma by vaccination with transgenic rice seed expressing mite allergen: induction of allergen-specific oral tolerance without bystander suppression

    Plant Biotechnol. J.

    (2011)
  • C.C. Lee

    Construction of a Der p2-transgenic plant for the alleviation of airway inflammation

    Cell. Mol. Immunol.

    (2011)
  • Y. Wakasa

    Oral immunotherapy with transgenic rice seed containing destructed Japanese cedar pollen allergens, Cry j 1 and Cry j 2, against Japanese cedar pollinosis

    Plant Biotechnol. J.

    (2013)
  • W. Halota

    Oral anti-CD3 immunotherapy for HCV-nonresponders is safe, promotes regulatory T cells and decreases viral load and liver enzyme levels: results of a phase-2a placebo-controlled trial

    J. Viral Hepat.

    (2015)
  • G. Lalazar

    Oral Administration of OKT3 MAb to patients with NASH, promotes regulatory T-cell induction, and alleviates insulin resistance: results of a phase IIa blinded placebo-controlled trial

    J. Clin. Immunol.

    (2015)
  • J. Su

    Oral delivery of acid alpha glucosidase epitopes expressed in plant chloroplasts suppresses antibody formation in treatment of Pompe mice

    Plant Biotechnol. J.

    (2015)
  • M. Iizuka

    Suppression of collagen-induced arthritis by oral administration of transgenic rice seeds expressing altered peptide ligands of type II collagen

    Plant Biotechnol. J.

    (2014)
  • C. Hansson

    Feeding transgenic plants that express a tolerogenic fusion protein effectively protects against arthritis

    Plant Biotechnol. J.

    (2016)
  • Y. Chen

    Targeted delivery of antigen to intestinal dendritic cells induces oral tolerance and prevents autoimmune diabetes in NOD mice

    Diabetologia

    (2018)
  • A.L. Posgai

    Plant-based vaccines for oral delivery of type 1 diabetes-related autoantigens: evaluating oral tolerance mechanisms and disease prevention in NOD mice

    Sci. Rep.

    (2017)
  • K.C. Kwon

    Expression and assembly of largest foreign protein in chloroplasts: oral delivery of human FVIII made in lettuce chloroplasts robustly suppresses inhibitor formation in haemophilia A mice

    Plant Biotechnol. J.

    (2018)
  • P. Satitsuksanoa

    Regulatory immune mechanisms in tolerance to food allergy

    Front. Immunol.

    (2018)
  • Cited by (45)

    View all citing articles on Scopus
    View full text