Chapter Eight - Regulation of Regulatory T Cells: Epigenetics and Plasticity

https://doi.org/10.1016/B978-0-12-800147-9.00008-XGet rights and content

Abstract

Regulatory T (Treg) cells, as central mediators of immune suppression, play crucial roles in many aspects of immune system's physiology and pathophysiology. The transcription factor Foxp3 has been characterized as a master gene of Tregs. Yet Treg cells possess a distinct pattern of gene expression, including upregulation of immune-suppressive genes and silencing of inflammatory cytokine genes. Recent studies have revealed the molecular mechanisms that establish and maintain such gene regulation in Treg cells. This review discusses recent progress in our understanding of molecular features of Treg cells, with particular attention to Treg-cell lineage commitment and stability.

Introduction

A variety of autoimmune and allergic disease pathologies are caused by immune responses to “self” antigens, environmental nonmicrobial antigens, and infectious agents. Regulatory T (Treg) cells, which play an indispensable role in immunological tolerance, protecting the host from excessive immune responses, are characterized by expression of the Forkhead transcription factor Foxp3. Foxp3 plays an essential role in the suppressive function of Tregs, and Foxp3 deficiency causes a multiorgan autoimmune disease, which can be observed in the scurfy mouse and in patients with immunodysregulation polyendocrinopathy enteropathy X-linked syndrome (Sakaguchi et al., 2008, Wan and Flavell, 2007). Foxp3 is induced in naturally occurring Tregs (called nTregs or more recently thymic Tregs [tTregs]) during their development in the thymus under the influence of relatively high-avidity interactions of T-cell receptors (TCRs) with self-antigens (Lee, Bautista, & Hsieh, 2011). Foxp3+ Tregs can also be generated from naïve T cells by TCR stimulation in the presence of TGF-β and IL-2; the resulting cells are called induced Tregs (iTregs or peripheral Tregs [pTregs]). Although iTregs exert an in vitro suppression activity similar to that of nTregs, the Foxp3 expression of iTregs was shown to be unstable in vivo (Chen et al., 2011, Floess et al., 2007, Koenecke et al., 2009). Various transcription factors including c-Rel, Smad2/3, and Runx1 have been identified as important for nTreg and iTreg induction through their transactivation of the Foxp3 promoter and/or enhancers (Ohkura et al., 2011, Takimoto et al., 2010). In addition, we have recently shown that the Nr4a family of transcription factors, which could be direct sensors of TCR strength, are essential for nTreg development in the thymus (Sekiya et al., 2013).

Although the Treg suppression mechanism is now well characterized (Wing & Sakaguchi, 2012), the molecular mechanisms of Treg development and maintenance remain to be clarified. nTregs have been shown to convert to certain types of effector helper T cells such as Th1, Th17, and follicular helper T (Tfh) cells after losing Foxp3 expression (Komatsu et al., 2009, Tsuji et al., 2009). These ex-Foxp3 cells, so-called as they are derived from Foxp3+ Treg cells but have lost their Foxp3 expression, develop an effector-memory phenotype, produce pathogenic cytokines, and may be involved in triggering the development of autoimmunity. In contrast, a recent study by Miyao et al. (2012) clearly denied Treg reprogramming. Nevertheless, it remains an open question how the expression and stability of Foxp3 in Tregs are regulated.

Section snippets

Generation of nTregs in the Thymus

nTregs develop from progenitor CD4+CD8+ double-positive (DP) T cells as do other single-positive (SP) T cells (Fig. 8.1). As a result of TCR gene rearrangement, T cells that possess TCR but fail to receive the appropriate TCR signal go into “death by neglect,” which is also called “positive selection.” In contrast, high-affinity ligands, mostly from self-proteins presented on MHC molecules on thymic antigen-presenting cells, trigger clonal deletion by direct induction of apoptosis. This process

Factors Involved in Foxp3 Expression

The next question concerns how strong TCR signals induce Foxp3 expression. The Foxp3 promoter has been extensively studied: Mantel et al. (2006) first characterized the human Foxp3 promoter, which is located 6.5 kb upstream of the first exon, and found six NFAT and AP-1 binding sites and a TATA and CAAT box (Mantel et al., 2006). The promoter is highly conserved between humans, mice, and rats; in addition, three highly conserved noncoding DNA sequences (CNS), CNS1, CNS2, and CNS3, were

Generation of Tregs by TGF-β

The role of TGF-β in nTreg differentiation is still controversial. Mice deficient in CNS1 exhibit alterations in iTreg but not nTreg differentiation (Zheng et al., 2010). Selective impairment of iTreg differentiation due to CNS1 deletion results in spontaneous allergic Th2-type inflammation in the intestine and lungs under a B6 background (Josefowicz et al., 2012). T-cell-specific deletion of the TGF-β receptor II or Smad2 and Smad3 cause very severe fatal inflammation, but thymic nTreg

Differences Between nTregs and iTregs

Several groups have used microarray analyses to unveil the developmental and functional differences between nTreg and iTreg cells (Feuerer et al., 2010, Hill et al., 2007). iTreg and nTreg cells were shown to be genetically distinct. Among the genes differentially expressed, Ikzf2 (Helios) and Nrp1 (Neuropilin-1) expressions have often been used as markers of nTreg cells compared with iTreg cells. Semaphorin-4a (Sema4a) and the Treg-cell-expressed receptor Nrp1 interact with each other. Sema4a

Epigenetic Changes in Tregs and Their Role in Treg Stability

Unlike nTregs, iTregs have been shown to be unstable. This is a significant obstacle to the use of ex vivo-expanded iTregs for adoptive immune therapy (Koenecke et al., 2009). This unstable phenotype is associated with a strong methylation of the CNS2 region of the TSDRs within the Foxp3 locus. This idea is supported by the fact that treatment of iTregs with IL-2/anti-IL-2 complexes in the presence of an antigen stabilized Foxp3 expression while also enhancing demethylation of the TSDR (Chen et

Treg Stability and Its Implication in Immunological Diseases

The continuous presence of Tregs throughout life is necessary to avoid autoimmunity (Kim, Rasmussen, & Rudensky, 2007). Thus, the stability of Foxp3 expression influences the balance between tolerance and autoimmunity, and loss of Foxp3 in Tregs may underlie the pathogenesis of autoimmune diseases and infection (Komatsu et al., 2014, Oldenhove et al., 2009, Yang et al., 2008, Zhou et al., 2009). nTregs have been shown to convert to certain types of effector helper T cells such as Th1, Th17, and

Factors that Control Treg Stability

Most reports have suggested, however, that these unstable Foxp3+ cell subsets constitute a minor fraction under ordinary conditions and only increase under lymphopenic or inflammatory conditions. Moreover, Foxp3 has been shown to be stable in bona fide Tregs (Miyao et al., 2012, Rubtsov et al., 2010). Thus, there must be a mechanism(s) that prevents the pathogenic conversion of nTregs. As mentioned before, hypomethylation of TSDR is a key factor in the stability of Tregs. In addition, several

Conclusion

As shown here, Treg-cells possess the potential to treat a wide range of immunological diseases from autoimmune conditions to allergies to cancer. For the treatment of autoimmune disorders and allergies, and to promote the efficient acceptance of grafts after transplantation, the adoptive transfer of Treg cells that have been expanded ex vivo or induced in vitro is attempted. The ultimate goal of this approach is to control inflammation with minimum adverse effects through the administration of

References (101)

  • L.F. Lu et al.

    Function of miR-146a in controlling Treg cell-mediated regulation of Th1 responses

    Cell

    (2010)
  • L.F. Lu et al.

    Foxp3-dependent microRNA155 confers competitive fitness to regulatory T cells by targeting SOCS1 protein

    Immunity

    (2009)
  • C.T. Luo et al.

    Transcriptional control of regulatory T cell development and function

    Trends in Immunology

    (2013)
  • T.R. Malek et al.

    CD4 regulatory T cells prevent lethal autoimmunity in IL-2Rbeta-deficient mice. Implications for the nonredundant function of IL-2

    Immunity

    (2002)
  • T. Miyao et al.

    Plasticity of Foxp3(+) T cells reflects promiscuous Foxp3 expression in conventional T cells but not reprogramming of regulatory T cells

    Immunity

    (2012)
  • M. Murai et al.

    Regulatory T-cell stability and plasticity in mucosal and systemic immune systems

    Mucosal Immunology

    (2010)
  • N. Ohkura et al.

    T cell receptor stimulation-induced epigenetic changes and Foxp3 expression are independent and complementary events required for Treg cell development

    Immunity

    (2012)
  • N. Ohkura et al.

    FOXP3 + regulatory T cells: Control of FOXP3 expression by pharmacological agents

    Trends in Pharmacological Sciences

    (2011)
  • N. Ohkura et al.

    Development and maintenance of regulatory T cells

    Immunity

    (2013)
  • G. Oldenhove et al.

    Decrease of Foxp3 + Treg cell number and acquisition of effector cell phenotype during lethal infection

    Immunity

    (2009)
  • Q. Ruan et al.

    Development of Foxp3(+) regulatory t cells is driven by the c-Rel enhanceosome

    Immunity

    (2009)
  • S. Sakaguchi et al.

    Regulatory T cells and immune tolerance

    Cell

    (2008)
  • B. Salomon et al.

    B7/CD28 costimulation is essential for the homeostasis of the CD4+CD25 + immunoregulatory T cells that control autoimmune diabetes

    Immunity

    (2000)
  • H. Takaki et al.

    STAT6 Inhibits TGF-beta1-mediated Foxp3 induction through direct binding to the Foxp3 promoter, which is reverted by retinoic acid receptor

    The Journal of Biological Chemistry

    (2008)
  • J. van Loosdregt et al.

    Stabilization of the transcription factor Foxp3 by the deubiquitinase USP7 increases Treg-cell-suppressive capacity

    Immunity

    (2013)
  • L. Xu et al.

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

    Immunity

    (2010)
  • X.O. Yang et al.

    Molecular antagonism and plasticity of regulatory and inflammatory T cell programs

    Immunity

    (2008)
  • Z. Yao et al.

    Nonredundant roles for Stat5a/b in directly regulating Foxp3

    Blood

    (2007)
  • S.E. Allan et al.

    Activation-induced FOXP3 in human T effector cells does not suppress proliferation or cytokine production

    International Immunology

    (2007)
  • N. Arpaia et al.

    Metabolites produced by commensal bacteria promote peripheral regulatory T-cell generation

    Nature

    (2013)
  • K. Atarashi et al.

    Treg induction by a rationally selected mixture of Clostridia strains from the human microbiota

    Nature

    (2013)
  • K. Atarashi et al.

    Induction of colonic regulatory T cells by indigenous Clostridium species

    Science

    (2011)
  • I. Baine et al.

    Regulation of T-cell tolerance by calcium/NFAT signaling

    Immunological Reviews

    (2009)
  • T. Bopp et al.

    NFATc2 and NFATc3 transcription factors play a crucial role in suppression of CD4 + T lymphocytes by CD4 + CD25 + regulatory T cells

    The Journal of Experimental Medicine

    (2005)
  • M.A. Burchill et al.

    IL-2 receptor beta-dependent STAT5 activation is required for the development of Foxp3 + regulatory T cells

    Journal of Immunology

    (2007)
  • B.J. Calnan et al.

    A role for the orphan steroid receptor Nur77 in apoptosis accompanying antigen-induced negative selection

    Immunity

    (1995)
  • J.D. Carter et al.

    Deficiency of the Src homology region 2 domain-containing phosphatase 1 (SHP-1) causes enrichment of CD4+CD25 + regulatory T cells

    Journal of Immunology

    (2005)
  • Q. Chen et al.

    IL-2 controls the stability of Foxp3 expression in TGF-beta-induced Foxp3 + T cells in vivo

    Journal of Immunology

    (2011)
  • J.L. Coombes et al.

    A functionally specialized population of mucosal CD103 + DCs induces Foxp3 + regulatory T cells via a TGF-beta and retinoic acid-dependent mechanism

    The Journal of Experimental Medicine

    (2007)
  • E.K. Deenick et al.

    c-Rel but not NF-kappaB1 is important for T regulatory cell development

    European Journal of Immunology

    (2010)
  • G.M. Delgoffe et al.

    Stability and function of regulatory T cells is maintained by a neuropilin-1-semaphorin-4a axis

    Nature

    (2013)
  • M. Dominguez-Villar et al.

    Identification of T helper type 1-like, Foxp3 + regulatory T cells in human autoimmune disease

    Nature Medicine

    (2011)
  • M.S. Fassett et al.

    Nuclear receptor Nr4a1 modulates both regulatory T-cell (Treg) differentiation and clonal deletion

    Proceedings of the National Academy of Sciences of the United States of America

    (2012)
  • M. Feuerer et al.

    Genomic definition of multiple ex vivo regulatory T cell subphenotypes

    Proceedings of the National Academy of Sciences of the United States of America

    (2010)
  • S. Floess et al.

    Epigenetic control of the foxp3 locus in regulatory T cells

    PLoS Biology

    (2007)
  • W. Fu et al.

    A multiply redundant genetic switch ‘locks in’ the transcriptional signature of regulatory T cells

    Nature Immunology

    (2012)
  • Y. Furusawa et al.

    Commensal microbe-derived butyrate induces the differentiation of colonic regulatory T cells

    Nature

    (2013)
  • A.D. Gu et al.

    Requirements of transcription factor Smad-dependent and -independent TGF-beta signaling to control discrete T-cell functions

    Proceedings of the National Academy of Sciences of the United States of America

    (2012)
  • S. Haxhinasto et al.

    The AKT-mTOR axis regulates de novo differentiation of CD4+Foxp3 + cells

    The Journal of Experimental Medicine

    (2008)
  • P.G. Hogan et al.

    Transcriptional regulation by calcium, calcineurin, and NFAT

    Genes & Development

    (2003)
  • Cited by (25)

    • Restoring self-tolerance in autoimmune diseases by enhancing regulatory T-cells

      2019, Cellular Immunology
      Citation Excerpt :

      The transcriptional regulation of Foxp3 expression and Treg differentiation is summarized in Fig. -1. The Foxp3 gene contains a promoter region and three conserved non-coding sequences (CNS1, 2 & 3) in human, mouse and rat [26,27]. More recently, Kitagawa et al. reported CNS0, another conserved region bound by Satb1, which is a chromatin organizer that can activate super-enhancers even in closed chromatin where chromatin accessibility is low [28].

    • Basic science for the clinician: Mechanisms of sublingual and subcutaneous immunotherapy

      2016, Annals of Allergy, Asthma and Immunology
      Citation Excerpt :

      This raises the possibility that although clinical improvement may be seen as soon as 1 month, parallel immunologic changes that involve the shift from a TH2 signature and up-regulation of pTregs require longer treatment protocols (up to 12 months). Another explanation for the potential failure of immunotherapy is that the epigenetic changes noted in pTregs are unstable (ie, the FoxP3 locus is only transiently hypomethylated).33,34 During immunotherapy, allergen-specific IgE levels measured by ImmunoCAP initially increase above baseline and then gradually decrease during months to years of treatment.35,36

    • The NF-κB transcription factor RelA is required for the tolerogenic function of Foxp3<sup>+</sup> regulatory T cells

      2016, Journal of Autoimmunity
      Citation Excerpt :

      Our finding that Foxp3 expression is unstable in RelA-deficient Tregs also raises the possibility that ex-RelaΔ Tregs contribute to the autoimmune disease. Treg populations possess cells with a self-reactive TCR repertoire [59] and with studies showing Tregs that lose FOXP3 expression behave like effector T cells and express inflammatory cytokines [60,61], Treg instability in young RelaΔ Treg mice could potentially generate a population of auto-reactive T cells. In conclusion, we have established that the constitutive activation of RelA in Tregs is required to maintain Treg stability and control Treg functions involved in the competitive interplay between eTregs and conventional T cells, roles that are vital to Tregs maintaining peripheral tolerance.

    • Continuum model of T-cell avidity: Understanding autoreactive and regulatory T-cell responses in type 1 diabetes

      2015, Journal of Theoretical Biology
      Citation Excerpt :

      Foxp3 transcription factor is considered a master regulator and the most reliable marker for Treg cells (Corthay, 2009). The plasticity, or ability of Tregs to lose their regulatory function by depleting Foxp3 is an innate property of these cells (Murai et al., 2010; Okada et al., 2014). A reduced expression of Foxp3 in activated Treg population has been found to cause defects in IL-2 signalling and reduced expression of a deubiquitinase important for Foxp3 stability (Bending et al., 2014).

    View all citing articles on Scopus

    Competing Interests: The authors have declared that no competing interests exist.

    View full text