Elsevier

Current Opinion in Immunology

Volume 43, December 2016, Pages 16-23
Current Opinion in Immunology

The role of FOXP3 in autoimmunity

https://doi.org/10.1016/j.coi.2016.07.004Get rights and content

Highlights

  • Dysregulation of FOXP3 expression can occur at multiple levels in autoimmunity.

  • Autoimmune SNPs diminish IL-2 sensitivity and FOXP3, resulting in impaired Tregs.

  • Splicing, post-translational modification, and subcellular localization regulate FOXP3.

  • Many new Treg-targeted therapies are being tested in autoimmunity.

FOXP3 controls the development and function of T regulatory cells (Tregs). Autoimmunity is linked to changes in FOXP3 activity that can occur at multiple levels and lead to Treg dysfunction. For example, changes in IL-2 signaling, FOXP3 transcription and/or post-translational modifications can all contribute to loss of self-tolerance. As additional pathways of FOXP3 regulation are elucidated, new therapeutic approaches to increase Treg activity either by cell therapy or pharmacological intervention are being tested. Early success from pioneering studies of Treg-based therapy in transplantation has promoted the undertaking of similar studies in autoimmunity, with emerging evidence for the effectiveness of these approaches, particularly in the context of type 1 diabetes.

Introduction

Forkhead box protein 3 (FOXP3) is the master transcription factor for CD4+ regulatory T cells (Tregs) [1], a cell type that plays a critical role in immune regulation. The essential role of FOXP3 and Tregs in autoimmunity was discovered through studies of humans with immune dysregulation, polyendocrinopathy, enteropathy, X-linked (IPEX) syndrome and the scurfy mouse model [2]. IPEX patients and scurfy mice have monogenic mutations in FOXP3 causing absent or poorly functional FOXP3 protein, a lack of normal Tregs, and the consequent development of multi-system autoimmunity [2].

Following these seminal studies in primary immunodeficiencies, many groups investigated whether changes in FOXP3 and associated changes in Treg numbers or function might also underlie the common polygenic forms of autoimmunity. Indeed there is now ample evidence that FOXP3 can be dysregulated in many ways, leading to altered Tregs that initiate and/or perpetuate autoimmunity. Here we review advances made in our understanding of how FOXP3 regulates autoimmunity, focusing on research in humans in the past 2 years addressing two main questions: firstly, how is FOXP3 dysregulated in autoimmunity? and secondly, how can FOXP3 be therapeutically targeted to treat autoimmunity?

Section snippets

How is FOXP3 dysregulated in autoimmunity?

Autoimmunity is clearly associated with changes in the proportion and/or function of FOXP3-expressing Tregs [3], but there is no dominant mechanism driving these changes. Rather, factors affecting Treg function range from the effects of genetics, to changes in FOXP3-promoting signaling pathways, FOXP3 mRNA expression, or protein modification, summarized in Figure 1. Autoimmunity may be driven by one or more of these mechanisms, ultimately resulting in disrupted balance between Tregs and

How can FOXP3 be targeted in autoimmunity?

With more than a decade of evidence that poor FOXP3 expression and Treg function causes or perpetuates autoimmunity, a variety of approaches to reverse these phenomena have been explored. The approaches are broadly classified as cellular or non-cellular treatments, with a combined approach likely being the most effective.

Conclusions and perspective

How autoimmunity is affected by changes in FOXP3 as it specifically relates to CD4+ Tregs has been extensively studied, but it is important to note that FOXP3 also has regulatory roles in other immune cells. Activated CD4+ T cells express FOXP3, leading to restraint of cytokine production and proliferation [64], and there are also reports of FOXP3 expression in CD8+ and invariant NKT cells. Whether or not autoimmunity is linked to changes in FOXP3 in non-CD4+ Tregs is an underexplored area of

References and recommended reading

Papers of particular interest, published within the period of review, have been highlighted as:

  • • of special interest

  • •• of outstanding interest

Acknowledgements

The authors’ own work on FOXP3 and autoimmunity is supported by grants from the Canadian Institutes for Health Research, the Canucks for Kids Foundation, and JDRF. AMP holds fellowships from the CIHR-STIR Training program in Transplantation, the 4 What Matters foundation fellowship and the JDRF postdoctoral fellowship. LC holds a fellowship from the JDRF Canadian Clinical Trial Network. MKL receives a Scientist Salary Award from the BC Children's Hospital Research Institute. We thank Dr. Paul

References (69)

  • R.B. Elliott et al.

    A population based strategy to prevent insulin-dependent diabetes using nicotinamide

    J Pediatr Endocrinol Metab

    (1996)
  • Z. Li et al.

    PIM1 kinase phosphorylates the human transcription factor FOXP3 at serine 422 to negatively regulate its activity under inflammation

    J Biol Chem

    (2014)
  • H. Nie et al.

    Phosphorylation of FOXP3 controls regulatory T cell function and is inhibited by TNF-alpha in rheumatoid arthritis

    Nat Med

    (2013)
  • J.A. Bluestone et al.

    Type 1 diabetes immunotherapy using polyclonal regulatory T cells

    Sci Transl Med

    (2015)
  • C.G. Brunstein et al.

    Umbilical cord blood-derived T regulatory cells to prevent GVHD: kinetics, toxicity profile, and clinical effect

    Blood

    (2016)
  • X. Song et al.

    The roles and functional mechanisms of interleukin-17 family cytokines in mucosal immunity

    Cell Mol Immunol

    (2016)
  • Y.C. Kim et al.

    Engineered antigen-specific human regulatory T cells: immunosuppression of FVIII-specific T- and B-cell responses

    Blood

    (2015)
  • C. von Spee-Mayer et al.

    Low-dose interleukin-2 selectively corrects regulatory T cell defects in patients with systemic lupus erythematosus

    Ann Rheum Dis

    (2015)
  • J.B. Spangler et al.

    Antibodies to interleukin-2 elicit selective T cell subset potentiation through distinct conformational mechanisms

    Immunity

    (2015)
  • S. Mitra et al.

    Interleukin-2 activity can be fine tuned with engineered receptor signaling clamps

    Immunity

    (2015)
  • T. Magg et al.

    Subcellular localization of FOXP3 in human regulatory and nonregulatory T cells

    Eur J Immunol

    (2012)
  • C.M. Huijts et al.

    mTOR inhibition per se induces nuclear localization of FOXP3 and conversion of invariant NKT (iNKT) cells into immunosuppressive regulatory iNKT Cells

    J Immunol

    (2015)
  • K. Matsuoka et al.

    Low-dose interleukin-2 therapy restores regulatory T cell homeostasis in patients with chronic graft-versus-host disease

    Sci Transl Med

    (2013)
  • A.M. Pesenacker et al.

    A Treg gene signature is a specific and sensitive biomarker to identify children with new onset type 1 diabetes

    Diabetes

    (2016)
  • R. Bacchetta et al.

    From IPEX syndrome to FOXP3 mutation: a lesson on immune dysregulation

    Ann N Y Acad Sci

    (2016)
  • A.M. Pesenacker et al.

    Control of tissue-localized immune responses by human regulatory T cells

    Eur J Immunol

    (2015)
  • H. Morikawa et al.

    Genetic and epigenetic basis of Treg cell development and function: from a FoxP3-centered view to an epigenome-defined view of natural Treg cells

    Immunol Rev

    (2014)
  • H. Wu et al.

    Critical link between epigenetics and transcription factors in the induction of autoimmunity: a comprehensive review

    Clin Rev Allergy Immunol

    (2016)
  • K.K. Farh et al.

    Genetic and epigenetic fine mapping of causal autoimmune disease variants

    Nature

    (2015)
  • J.H. Yang et al.

    Natural variation in IL-2 sensitivity influences regulatory T cell frequency and function in individuals with long-standing type 1 diabetes

    Diabetes

    (2015)
  • Y. Kochi

    Genetics of autoimmune diseases: perspectives from genome-wide association studies

    Int Immunol

    (2016)
  • M. Sebode et al.

    Reduced FOXP3(+) regulatory T cells in patients with primary sclerosing cholangitis are associated with IL2RA gene polymorphisms

    J Hepatol

    (2014)
  • S.J. Patterson et al.

    T regulatory cell chemokine production mediates pathogenic T cell attraction and suppression

    J Clin Invest

    (2016)
  • N. Bottini et al.

    Tyrosine phosphatase PTPN22: multifunctional regulator of immune signaling, development, and disease

    Annu Rev Immunol

    (2014)
  • Cited by (26)

    • Building a CAR-Treg: Going from the basic to the luxury model

      2020, Cellular Immunology
      Citation Excerpt :

      Regulatory T cells (Tregs) control immune homeostasis and dampen exacerbated immune responses by suppressing the activation, proliferation and/or effector function of a range of immune cells [1–3]. Their impaired function is associated with a variety of disorders including Immunodysregulation Polyendocrinopathy Enteropathy X-linked (IPEX) syndrome [4,5], and many common polygenic autoimmune diseases [6,7]. This ability of Treg therapy to promote and re-shape immune tolerance makes them an attractive candidate for a cellular therapy-based approach to minimize autoimmunity and/or promote the development of transplantation tolerance [8–10].

    • General features of autoimmune disease

      2019, The Autoimmune Diseases
    • Epigenetic and gene expression alterations of FOXP3 in the T cells of EAE mouse model of multiple sclerosis

      2017, Journal of the Neurological Sciences
      Citation Excerpt :

      It has been shown that Treg cells are the main cellular constituents of the tolerance system to suppress autoimmunity [4]. Meanwhile, the main transcription factor predominantly expressed in the Treg cells is forkhead box P (FOXP3) acting as the main regulator to polarize naïve T cells into the Treg lineage [5,6]. Thus, FOXP3 expression was found to be critical for Treg cell development and its suppressive function.

    View all citing articles on Scopus
    3

    These authors contributed equally to the work.

    View full text