Elsevier

Journal of Autoimmunity

Volume 63, September 2015, Pages 23-30
Journal of Autoimmunity

Foxp3 lacking exons 2 and 7 is unable to confer suppressive ability to regulatory T cells in vivo

https://doi.org/10.1016/j.jaut.2015.06.009Get rights and content

Highlights

  • The human FOXP3Δ2Δ7 isoform cannot cover suppressive ability to Tregs in vitro.

  • We generated mice exclusively expressing Foxp3 lacking exon 2 and 7 (Foxp3δ2δ7/Y).

  • Foxp3δ2δ7/Y mice succumb after 3–6 weeks in a lymphoproliferative disease.

  • Foxp3δ2δ7/Y mice display T cell activation and activation of antigen-presenting cells.

  • The mouse counterpart of FOXP3Δ2Δ7 cannot confer suppressive ability to Tregs in vivo.

Abstract

The forkhead/winged-helix transcription factor FOXP3 confers suppressive ability to CD4+FOXP3+ regulatory T (Treg) cells. Human Treg cells express several different isoforms of FOXP3 that differ in function. However, the regulation and functional consequences of FOXP3 isoform expression remains poorly understood. In order to study the function of the FOXP3Δ2Δ7 isoform in vivo we generated mice that exclusively expressed a Foxp3 isoform lacking exon 2 and 7. These mice exhibited multi-organ inflammation, increased cytokine production, global T cell activation, activation of antigen-presenting cells and B cell developmental defects, all features that are shared with mice completely deficient in FOXP3. Our results demonstrate that the mouse counterpart of human FOXP3Δ2Δ7 is unable to confer suppressive ability to Treg cells.

Introduction

The immune system is delicately regulated to allow responses against foreign- but not self-antigens. Several distinct mechanisms uphold such immunological tolerance, including deletion of highly self-reactive lymphocytes during their development, lymphocyte hypo-responsiveness when antigens are encountered in the absence of co-stimulatory signals, and suppression of immune responses by cells with regulatory capacity. Among these tolerance mechanisms is the CD4+FOXP3+ regulatory T (Treg) cell population, which comprises a subset of T cells that suppresses immune activation in a dominant manner [1]. Abolished Treg cell function is the direct cause of the lymphoproliferative disease seen in immunodysregulation polyendocrinopathy enteropathy X-linked syndrome (IPEX) patients and scurfy mice [2], [3], [4]. In addition, a large number of studies have suggested that deficiencies in Treg cell function are an underlying cause for disease conditions ranging from infections to chronic inflammatory disorders [5].

Treg cells are characterized by expression of the forkhead/winged-helix transcription factor FOXP3 [6], [7], [8]. FOXP3 contains an N-terminal repressor domain, centrally located C2H2 zinc finger and leucine zipper domains and a forkhead domain at the C-terminus [9], [10], [11], [12]. The N-terminal repressor domain, partially encoded by exon 2, allows FOXP3 to interact with transcriptional activators and repressors to modulate gene expression [13]. The function of FOXP3's C2H2 zinc finger domain remains unknown and no IPEX causative mutations have been found in this domain. The leucine zipper, encoded by exon 7, domain is required for formation of FOXP3 multimers as well as for FOXP3 to be able to confer suppressor ability [9], [11], [14], [15]. The C-terminal forkhead domain mediates DNA-binding by FOXP3 and it is a hotspot mutation region in IPEX [12], [16].

The expression and function of FOXP3 is normally regulated at a transcriptional, post-transcriptional, and post-translational level. At the transcriptional level, alternative splicing appears to play a key role in FOXP3 regulation. The two most abundant FOXP3 isoforms, full-length FOXP3 (FOXP3fl) and FOXP3 lacking exon 2 (FOXP3Δ2), confer a suppressive ability to Treg cells [17], [18]. In contrast, FOXP3 lacking exons 2 and 7 (FOXP3Δ2Δ7) has been suggested to inhibit other FOXP3 isoforms in a dominant negative manner [19]. A recent study has also demonstrated that exon 7 of FOXP3 is required for proper Treg cell function, as two different point mutations, located near the intron 7 splice donor site, were found to result in exon 7 excision and IPEX syndrome [20]. However, despite the importance of FOXP3 in Treg cells, the regulation and functional consequences of FOXP3 isoform expression remains poorly understood.

Up to date all experiments addressing the function of FOXP3 isoforms have been performed in vitro as mice do not utilize alternative splicing of coding exons for FOXP3 transcripts [21]. However, FOXP3 is highly conserved and can function over species barriers [19], [22]. Here, we characterized the in vivo function of the mouse counterpart to FOXP3Δ2Δ7 by generating mice exclusively expressing Foxp3 lacking exon 2 and 7 (Foxp3δ2δ7). We show that mice exclusively expressing Foxp3δ2δ7 in essence phenocopy scurfy mice and that Foxp3δ2δ7 is unable to confer a suppressive ability to mouse Treg cells.

Section snippets

Mice

A targeting vector was constructed such that a Foxp3 cDNA lacking exons 2 and 7 was inserted at the translation initiation ATG codon of Foxp3 and the TGA stop of Foxp3 was replaced by a 2A peptide-GFP cassette followed by a double polyadenylation signal (pA) and an FRT flanked Neo cassette (Fig. 1a and Supplementary Fig. 1). The Foxp3δ2δ7-2A-GFP cassette was synthesized and the sequence was verified using DNA sequencing prior to subcloning it into the targeting vector. The

Mice exclusively expressing Foxp3 lacking exons 2 and 7 succumb to lymphoproliferative disease early in life

In vitro suppression assays have been helpful to study the function of mouse and human Treg cells, yet it still remains uncertain as to what extent these assays mimic suppression in vivo. Mice do not express different Foxp3 isoforms [21], which makes it challenging to study the functional consequences of alternative splicing of FOXP3 in vivo. However, FOXP3 is highly conserved and both FOXP3 and Treg cell-mediated suppression can function over species barriers [19], [22]. To define the function

Discussion

The regulation and functional consequences of FOXP3 isoform expression remains poorly understood. In this study we demonstrate that mice exclusively expressing an artificial FOXP3 isoform FOXP3δ2δ7, which correspond to human FOXP3Δ2Δ7, succumbs early in life to lymphoproliferative disease. Thus our in vivo findings corroborate previous in vitro studies that have suggested that FOXP3Δ2Δ7 is unable to confer a suppressive ability to Treg cells.

In 2005 Allen et al. described the existence of the

Conclusions

Mice that exclusively expressing an artificial FOXP3 isoform FOXP3δ2δ7, which correspond to human FOXP3Δ2Δ7, succumbs early in life to lymphoproliferative disease characterized by multi-organ inflammation, activation of T cells and APCs, increased cytokine expression, autoantibody production and severely impaired B cell development. In fact, these Foxp3δ2δ7/Y mice appear to phenocopy scurfy mice suggesting that the Foxp3δ2δ7 isoform is completely unable to confer a suppressive phenotype to

Funding

This work was supported by the Swedish Research Council (projects: 2008-3055 and 2012-1851), the Swedish Cancer Foundation, the Swedish Heart-Lung Foundation, the European Union (FP7 Marie Curie IRG program), the Swedish Society of Medicine, Sigurd and Elsa Goljes minne and Ruth och Richard Julin's foundation.

Authorship contributions

A.-L.J., S.L., R.M. and J.A. designed the research; A.-L.J., S.L., C.D. and R.M. performed experiments; A.-L.J., S.L., L.W and J.A. wrote the manuscript and all authors discussed the results and their implications and commented on the manuscript.

Acknowledgments

We thank Ellen Chen at Ingenious targeting Laboratories, Meimei Shan and Cindy Gutzeit at the Icahn School of Medicine at Mount Sinai for their help as well as Professor Robert A. Harris at Karolinska Institutet for linguistic advice.

References (39)

  • C.L. Bennett et al.

    The immune dysregulation, polyendocrinopathy, enteropathy, X-linked syndrome (IPEX) is caused by mutations of FOXP3

    Nat. Genet.

    (2001)
  • M.E. Brunkow et al.

    Disruption of a new forkhead/winged-helix protein, scurfin, results in the fatal lymphoproliferative disorder of the scurfy mouse

    Nat. Genet.

    (2001)
  • R.S. Wildin et al.

    X-linked neonatal diabetes mellitus, enteropathy and endocrinopathy syndrome is the human equivalent of mouse scurfy

    Nat. Genet.

    (2001)
  • L.S. Taams et al.

    Regulatory T cells in human disease and their potential for therapeutic manipulation

    Immunology

    (2006)
  • J.D. Fontenot et al.

    Foxp3 programs the development and function of CD4+CD25+ regulatory T cells

    Nat. Immunol.

    (2003)
  • S. Hori et al.

    Control of regulatory T cell development by the transcription factor Foxp3

    Science

    (2003)
  • R. Khattri et al.

    An essential role for scurfin in CD4+CD25+ T regulatory cells

    Nat. Immunol.

    (2003)
  • W.J. Chae et al.

    The mutant leucine-zipper domain impairs both dimerization and suppressive function of Foxp3 in T cells

    Proc. Natl. Acad. Sci. U. S. A.

    (2006)
  • E. Bettelli et al.

    Foxp3 interacts with nuclear factor of activated T cells and NF-kappa B to repress cytokine gene expression and effector functions of T helper cells

    Proc. Natl. Acad. Sci. U. S. A.

    (2005)
  • Cited by (0)

    1

    These authors contributed equally to this work.

    View full text