GET THE APP

Approaches to Discriminate Naturally Induced Foxp3+ Treg cells of
Journal of Clinical and Cellular Immunology

Journal of Clinical and Cellular Immunology
Open Access

ISSN: 2155-9899

+44 1223 790975

Review Article - (2018) Volume 9, Issue 1

Approaches to Discriminate Naturally Induced Foxp3+ Treg cells of Intra- and Extrathymic Origin: Helios, Neuropilin-1, and Foxp3RFP/GFP

Sebastian Dohnke1,2, Maria Schreiber2,3, Sonja Schallenberg2, Mario Simonetti2,4,5, Luise Fischer1,2, Annette I. Garbe1, Antonios Chatzigeorgiou6 and Karsten Kretschmer2,4,5*
1Osteoimmunology, DFG-Center for Regenerative Therapies Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Fetscherstr. 105, 01307, Dresden, Germany
2Molecular and Cellular Immunology/Immune Regulation, DFG-Center for Regenerative Therapies Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Fetscherstr. 105, 01307 Dresden, Germany
3Regenerative Therapies for Diabetes, DFG-Center for Regenerative Therapies Dresden (CRTD), Center for Molecular and Cellula r Bioengineering (CMCB), Technische Universität Dresden, Fetscherstr. 105, 01307 Dresden, Germany
4Paul Langerhans Institute Dresden (PLID) of the Helmholtz Zentrum München at the University Hospital and Medical Faculty Carl Gustav Carus of TU Dresden, Fetscherstrasse 74, 01307 Dresden, Germany
5German Center for Diabetes Research (DZD e.V.), Ingolstädter Landstraße 1, 85764 Neuherberg, Germany
6Institute for Clinical Chemistry and Laboratory Medicine, Medical Faculty, TU Dresden, Fetscherstrasse 74, 01307, Dresden, Germany
*Corresponding Author: Karsten Kretschmer, Molecular and Cellular Immunology/Immune Regulation, DFG-Center for Regenerative Therapies Dresden (CRTD), Center for Molecular and Cellular Bioengineering (CMCB), Technische Universität Dresden, Fetscherstr. 105, 01307 Dresden, Germany Email:

Abstract

CD4+ regulatory T (Treg) cells expressing the transcription factor fork head box P3 (Foxp3) prevent catastrophic autoimmunity and maintain immune homeostasis throughout life, and are being increasingly implicated in non-immune functions, such as the control of metabolic and regenerative processes in mice and humans. Early studies have attributed a major role of the thymus and intrathymic Foxp3+ Treg lineage commitment (referred to as ‘tTreg’ cells) in the establishment and maintenance of the mature Foxp3+ Treg cell pool residing in peripheral lymphoid tissue. In addition, numerous experimental modalities have been shown to instruct Foxp3+ Treg cell commitment in peripheral, initially naïve CD4+Foxp3- T cells, which includes the induction of a Foxp3+ Treg cell phenotype and suppressor function by TGF-β in vitro (‘iTreg’ cells) and by sub-immunogenic T cell receptor stimulation in vivo (‘pTreg’ cells). This led to the hypothesis that, under physiological conditions, the induction of pTreg cells may also contribute to the peripheral Foxp3+ Treg cell compartment in the steady state of nonmanipulated, immunocompetent mice. However, until recently, studies on developmental Foxp3+ Treg cell heterogeneity have been hampered by the lack of suitable markers to discriminate naturally induced tTreg and pTreg cells. Here, we provide an overview of recently proposed approaches to track such developmental sub-lineages, with a particular emphasis on Helios, Neuropilin-1, and Foxp3RFP/GFP mice, in which Foxp3RFP+ tTreg and pTreg cells are stably marked by differential GFP expression.

Keywords: T cell development; tTreg; pTreg; iTreg; Foxp3

Introduction

Apart from the artificial induction of a Foxp3+ regulatory T (Treg) cell phenotype in post-thymic, initially naïve CD4+Foxp3 T cells [1], there is accumulating evidence that, under physiological conditions, Foxp3+ Treg cells are generated both within the thymus (referred to as ‘tTreg’ cells) and extrathymically at peripheral sites (‘pTreg’ cells). Reminiscent of Foxp3+ tTreg cell development mediated by developmental progression of CD4+CD8 single-positive (CD4SP) thymocyte precursors with a CD25+Foxp3 phenotype [2], mature Foxp3+ pTreg cells continuously differentiate from precommitted CD4+Foxp3CD25+ pTreg cell precursors in peripheral lymphoid tissues of nonmanipulated mice [1,3] and act in concert with tTreg cells to enforce immune tolerance [4-7]. The identification of immediate CD4+Foxp3CD25+ pTreg cell precursors provided direct evidence that extrathymic developmental pathways feed into the overall pool of mature Foxp3+ Treg cells in the steady state [3]. However, the lack of unambiguous markers to faithfully discriminate naturally induced Foxp3+ tTreg and pTreg cells remained a considerable limitation of experimental efforts to directly address the processes that generate such developmental sub-lineages and govern their incorporation into the mature Treg cell pool, which exhibits remarkable phenotypic heterogeneity [8]. A universal marker to discriminate murine Foxp3+ tTreg and pTreg cells should preferably fulfill several criteria: i) exclusive expression either by tTreg or pTreg cells; ii) its expression status should be highly stable and preserved under pro-inflammatory conditions; and iii) it should enable the isolation and sub-fractionation of viable cells for downstream applications, such as gene expression surveys, functional studies, and Foxp3+ Treg cell-based therapies. Moreover, a murine marker with evolutionary conservation across species may facilitate translation to human CD4+FOXP3+ Treg cells. While a marker combining all above characteristics remains to be identified, several approaches to discriminate tTreg and pTreg cells have recently been proposed: the Ikaros transcription factor Helios (Ikzf2) [9], the surface marker Neuropilin-1 (Nrp-1) [10,11], and differential GFP expression in Foxp3RFP+ Treg cells of Foxp3RFP/GFP mice [7].

Endogenous markers for tTreg/pTreg cell discrimination: Helios and Nrp-1

Helios: Initial studies by Shevach and colleagues [9] suggested that Helios might be selectively expressed in Foxp3+ tTreg cells. This conclusion was based on three key findings: i) In the steady state, Helios was found to be co-expressed in the vast majority (≥ 99%) of developing Foxp3+ CD4SP thymocytes, whereas its expression was restricted to ~70% of peripheral CD4+Foxp3+ Treg cells; ii) Foxp3+ iTreg cells generated from naïve CD4+Foxp3 T cells in vitro by TCR stimulation in the presence of added TGF-β maintained a Helios phenotype; and iii) Foxp3+ pTreg cells generated from TCR transgenic CD4+Foxp3 T cells in vivo after adoptive transfer into immunocompetent recipients and subsequent oral antigen administration also failed to up-regulate Helios expression. Overall, these data are consistent with the interpretation that the Foxp3+Helios phenotype of ~30% of peripheral Treg cells is indicative of their extrathymic developmental origin.

Subsequent studies [12-15] further corroborated the initial observation [9] that TGF-β-driven Foxp3+ iTreg cell generation fails to promote Helios co-expression, when TCR stimulation was mediated by plate-bound anti-CD3 monoclonal antibodies (mAbs) ± co-stimulatory anti-CD28 mAbs. However, TCR stimulation through peptide antigen-loaded splenocytes [12,13] or anti-CD3/CD28 mAb- coated beads (i.e. in the absence of antigen-presenting cells; APCs) [15] resulted in robust Helios expression in ~50% of newly generated CD4+Foxp3+ iTreg cells, irrespective of whether or not IL-2R signaling was abrogated by neutralizing anti-IL-2 mAbs (Figure 1A). In vivo, and in contrast to oral antigen feeding [9], Foxp3+ pTreg cell induction by intravenous antigen administration [12] or recombinant anti-DEC-205 Ab-mediated delivery of antigen to DEC-205+ DCs (Figure 1B) [16-18] resulted in Helios co-expression in the majority of de novo induced Foxp3+ pTreg cells, although continued TCR stimulation is required to maintain high levels of Helios expression [12].

clinical-cellular-immunology-Helios-expression

Figure 1: Helios expression in extrathymically induced Foxp3+ Treg cells. Flow cytometry of Foxp3 and Helios protein was performed at day (A, C) three and (B) 14 using the fluorochrome-conjugated mAbs FJK-16s and 22F6, respectively. (A) TGF-β-mediated Foxp3+ iTreg cell induction. Naïve CD4+ T cells (CD62LhighCD25 Foxp3GFP–) were FACS purified from peripheral lymphoid organs of Foxp3GFP mice and TCR stimulated in the presence of added IL-2 (100 U/ml) and TGF-β (5 ng/ml). TCR stimulation, top panels: plate-bound anti-CD3 mAbs (5 μg/ml) ± soluble anti-CD28 mAbs (1 μg/ml); bottom panels: anti-CD3/CD28 mAb-coated beads (2 beads/ cell) ± neutralizing anti-IL-2 mAbs (20 μg/ml). (B) DEC-205+ DC-targeted Foxp3+ pTreg cell induction in vivo. Immunocompetent Thy1.1+ recipients of initially naïve Thy1.2+CD4+Foxp3Helios T cells with transgenic TCR-HA107-119 expression were injected with anti-DEC-205/HA107-119 fusion antibodies (50 ng), and induced Foxp3 and Helios expression was assessed among gated Thy1.2+CD4+ T cells. Lines with arrowheads illustrate the gating scheme. (C) Precommitted Foxp3GFP– Treg cell precursors were FACS isolated from thymus (CD4+CD8CD25+Foxp3GFP−) and lymph nodes (CD4+CD25+Foxp3GFP−) of Foxp3GFP mice and cultured with IL-2 (1000 U/ ml) in the absence of deliberate TCR and TGF-βR stimulation. Numbers in histograms show percentages of Helios co-expression among differentiated Foxp3+ tTreg (left) and pTreg (right) cells. Adapted from Schallenberg et al. [3,15].

We also tracked Helios expression in the progeny of physiological CD4+(CD8)CD25+Foxp3 Treg cell precursors from thymus [2] and peripheral lymphoid tissues [3] of nonmanipulated Foxp3GFP mice. In the absence of deliberate TCR and TGF-βR stimulation, developmental progression of thymic CD25+Foxp3 CD4SP tTreg cell precursors in IL- 2-supplemented cultures were accompanied by Helios expression in the majority of differentiated Foxp3+ tTreg cells (Figure 1C, left panel). In contrast, only ~50% of lymph node-derived CD4+CD25+Foxp3 pTreg cell precursors acquired Helios expression during in vitro differentiation into Foxp3+ pTreg cells (Figure 1C, right panel). When we track naturally induced Foxp3+ Treg cells of intrathymic (RFP+GFP+ tTreg) and peripheral (RFP+GFP pTreg) origin in peripheral lymphoid tissues of nonmanipulated Foxp3RFP/GFP mice (see below), it became clear that >90% of mature RFP+GFP+ tTreg cells expressed Helios, whereas ~50% of their RFP+GFP pTreg cell counterpart were Helios [7].

In summary, induction of Helios expression during Foxp3+ Treg cell development appears more context-dependent than initially anticipated and may not be exclusive to tTreg cells. Nevertheless, lack of Helios expression identifies a significant proportion of Foxp3+ pTreg cells in lymphoid tissues of steady-state mice. Clearly, some modalities of Foxp3+ iTreg and pTreg cell generation promote Helios expression, which might be transient, unless continuous TCR stimulation is provided [12,19]. The mechanisms underlying differential Helios expression in developmental Foxp3+ Treg cell sub-lineages are likely to include differences in co-stimulatory and/or cytokine receptor signaling during the induction phase of Foxp3 expression. In any case, transgenic mice with Helios-dependent fluorochrome reporter expression [20] will pave the way to fully establish the relationship between Helios+ and Helios Treg cells. Another unresolved question is whether Helios expression can be dynamically regulated during the lifetime of mature Foxp3+ Treg cell populations.

Nrp-1: Two studies [10,11] proposed that differential Nrp-1 expression can be exploited to distinguish Foxp3+ tTreg and pTreg cells under non-inflammatory conditions: the majority of developing Foxp3+ CD4SP thymocytes and ~70% of peripheral CD4+Foxp3+ Treg cells were Nrp-1high, whereas low Nrp-1 expression levels on Foxp3+ Treg cells correlated with their extrathymic developmental origin in certain experimental settings of pTreg cell induction; this included homeostatic proliferation in lymphopenic recipient mice, free antigen administration (oral, intravenous, or via subcutaneously implanted osmotic mini- pumps), and spontaneous pTreg cell differentiation in Rag−/− mice with transgenic expression of a myelin basic protein (MBP)-reactive TCR. Consistent with the important role of the microbiota in intestinal pTreg cell homeostasis, Foxp3+Nrp-1low Treg cells were severely reduced in the lamina propria of germ-free mice, whereas this intestinal subset was enriched in mice housed under specific pathogen-free conditions, as compared to other secondary lymphoid tissues [10]. Also, in peripheral lymphoid tissues of mice with partial pTreg cell deficiency due to targeted deletion of an enhancer element (CNS1) within the Foxp3 gene locus that mediates TGF-β responsiveness during iTreg and pTreg cell development [5,21], the proportion of Nrp-1low cells among CD4+Foxp3+ Treg cells was ~2-fold reduced, as compared to CNS1- proficient mice [10].

Nonetheless, the correlation between extrathymic Treg cell development and their Foxp3+Nrp-1low phenotype may not be absolute, both in vitro and in vivo: While CD4+Foxp3 TCR-MBP+ T cells failed to up-regulate Nrp-1 expression upon TGF-β-mediated Foxp3+ iTreg cell generation, irrespective of whether TCR stimulation was mediated by anti-CD3/CD28 mAbs or peptide antigen-loaded APCs [11], polyclonal CD4+Foxp3 T cells significantly up-regulated Nrp- 1 expression during TGF-β-driven Foxp3 induction upon anti-CD3/ CD28 mAb-mediated TCR stimulation [10]. Along the same lines, oral antigen feeding of lymphopenic recipients of ovalbumin-reactive CD4+Foxp3DO11.10+ T cells promoted induction of Foxp+3 pTreg cells with a predominantly Nrp-1low phenotype [10], but oral antigen feeding of Rag1−/− × DO11.10+ mice promoted Nrp-1high expression on up to 70% of de novo induced Foxp3+DO11.10+ pTreg cells [7]. Some of the observed discrepancies in Nrp-1 up-regulation during iTreg/pTreg cell generation can potentially be attributed to differences in TCR/co- stimulatory signaling and the kinetics of transient Nrp-1 expression [7,10]. Still, a Nrp-1low/− phenotype is suitable to track Foxp3+ pTreg cells, at least under non-inflammatory/sub-immunogenic conditions and in the intestinal lamina propria.

Correlating Helios and Nrp-1 expression: In adult mice, Helios and Nrp-1 expression by peripheral Foxp3+ Treg cells closely correlate in that the majority of Helios+ cells co-express Nrp-1 [10,11,20], but a significant fraction of Foxp3+Helios Treg cells is Nrp-1high, resulting in <40% of Foxp3+Helios Treg cells with a Nrp-1low phenotype [20]. One plausible explanation for discordant Helios and Nrp-1 expression is that the Nrp-1low phenotype may not represent an immutable characteristic of Foxp3+ pTreg cells. In fact, while TCR-MBP+ Treg cells remained Nrp-1low, when stimulated in vitro with peptide antigen-loaded APCs [11], TCR stimulation using anti-CD3/CD28 mAbs [10] or anti- CD3/CD28 mAb-coated beads [7] up-regulated Nrp-1 expression on polyclonal Foxp3+Nrp-1low Treg cells. Upon adoptive transfer into otherwise nonmanipulated immunocompetent recipient mice, FACS-purified Nrp-1+ and Nrp-1low Treg cell populations largely maintained their respective Nrp-1 expression status [10,11]; in contrast, adoptively transferred Foxp3+Nrp-1low Treg cells increased Nrp-1 expression levels in lymphopenic recipients [11] and at site of acute inflammation, such as the central nervous system of mice with acute experimental autoimmune encephalomyelitis [11], indicating that Nrp-1 expression can indeed be up-regulated under pro-inflammatory conditions.

In steady-state Foxp3RFP/GFP mice (see below) [7], >45% of lymph node-derived RFP+GFP pTreg cells expressed Nrp-1 ex vivo, at levels comparable to that of RFP+GFP+ tTreg cells. In vitro, RFP+GFP pTreg cells that initially lacked Nrp-1 expression readily acquired Nrp-1 expression in IL-2-supplemented TCR stimulation cultures. These observations further support the notion that the Nrp-1 expression status of naturally induced tTreg and pTreg cells can be dynamically regulated under T cell stimulatory conditions. Consistently, at day 16 after injection of NOD.Rag1−/− mice with CD4+Foxp3BDC2.5+ T cells exhibiting a naïve CD62LhighCD44low phenotype, ~70% of homeostatically converted Foxp3+ pTreg cells expressed high levels of Nrp-1 [7].

Differential BAC.Foxp3GFP reporter expression in Foxp3RFP+ Treg cells as a transgenic approach for tTreg/pTreg cell discrimination

Our studies in the Foxp3RFP/GFP model were initiated by the serendipitous observation that peripheral lymphoid tissues of mice with Foxp3 BAC-driven expression of a Cre recombinase-GFP (Cre- GFP) fusion protein (BAC-Foxp3Cre-GFP) [22] harbor a sizable CD4+ Treg cell population that is GFP but expresses Foxp3 protein [7,23], as judged by intracellular staining using FACS-purified CD4+CD25+GFP T cells and fluorochrome-conjugated anti-Foxp3 mAbs (Figure 2). To assess whether such peripheral Foxp3+GFP Treg cells resulted from down-regulated BAC-Foxp3Cre-GFP reporter expression, we performed genetic fate mapping studies employing BAC-Foxp3Cre-GFP × R26Y mice, in which cells are marked by ROSA26-driven expression of yellow fluorescent protein (YFP) after Cre recombinase-mediated, irreversible excision of a loxP-flanked STOP cassette [24]. However, rather than transgene silencing in initially Foxp3+GFP+ Treg cells (e.g. by epigenetic mechanisms), these studies established that the BAC-Foxp3Cre-GFP reporter was transcriptionally inactive throughout the entire development and lifespan of the Foxp3+GFP Treg cell population [7]. Consistently, in BAC-Foxp3Cre-GFP mice additionally expressing RFP from an IRES downstream of the Foxp3 coding region (Foxp3IRES-RFP) [25], essentially all peripheral Foxp3+GFP Treg cells co-expressed Foxp3IRES-RFP (Figure 3A) [7]. Conversely, a considerable fraction of peripheral Foxp3IRES-RFP+ Treg cells consistently lacked any BAC-Foxp3Cre-GFP expression, and this RFP+GFP population increased from ~20% in adult to up to 45% in aged mice (Figure 3A).

clinical-cellular-immunology-lymphoid-tissues

Figure 2: Tracking CD4+Foxp3+ Treg cells that lack GFP expression in peripheral lymphoid tissues of BAC-Foxp3Cre-GFP mice. The dot plot depicts presort analysis of CD25 and GFP expression among gated CD4+ T cells from lymph nodes of BAC-Foxp3Cre-GFP mice. Histograms show postsort analysis of Foxp3-driven GFP expression (left) and Foxp3 expression, as revealed by intracellular staining (ICS) using anti-Foxp3 mAbs (right), among indicated postsort populations. Adapted from Schallenberg et al. [23].

clinical-cellular-immunology-Discrimination-mature

Figure 3: Discrimination of mature Foxp3RFP+ tTreg and pTreg cells in peripheral tissues of Foxp3RFP/GFP mice based on differential GFP expression. (A) Flow cytometry of RFP/GFP expression among CD4-gated cells in spleen of 2-month-old (top) and 18-month-old (bottom) Foxp3RFP/GFP mice. Numbers in dot plots indicate percentages of GFP pTreg cells and GFP+ tTreg cells among total RFP+ Treg cells in the respective quadrant. (B) In the thymus, tTreg-cell development is mediated by CD4+CD8 single-positive (SP) thymocytes that exhibit a CD25+Foxp3 phenotype (Foxp3 mRNA+, Foxp3 protein) and are precommitted to up-regulate Foxp3 expression upon IL-2 receptor signaling. BAC-Foxp3Cre-GFP expression is induced in such Treg cell lineage committed CD4SP precursors and maintained in their peripheral tTreg cell progeny after thymic export. In contrast, outside the thymus, IL-2 promotes the differentiation of precommitted CD25+Foxp3 pTreg precursors (Foxp3 mRNA+, Foxp3 protein) into Foxp3+ pTreg cells that fail to up-regulate BAC-Foxp3Cre-GFP expression, resulting in mature Foxp3+GFP pTreg cells. Foxp3IRES-RFP expression marks all peripheral Foxp3+ Treg cells, irrespective of their GFP expression status. In the steady state, Helios Treg cells are enriched among RFP+GFP pTreg cells, although this correlation is not absolute. Adapted from Petzold et al. [7].

Selective BAC-Foxp3Cre-GFP reporter activity in Foxp3+ tTreg cells but not iTreg/pTreg cells

To determine the origin of peripheral Foxp3+ Treg cells with a RFP+GFP phenotype, we next analyzed the activity of the BAC- Foxp3Cre-GFP reporter in thymic and peripheral pathways of Foxp3+ Treg lineage commitment.

Intrathymic BAC-Foxp3Cre-GFP reporter activity: In the thymus of Foxp3RFP/GFP mice, the vast majority of GFP-expressing CD4+CD8+ double-positive (DP) and CD4SP cells co-expressed RFP, and intrathymically injected RFP−GFPCD25+ CD4SP tTreg cell precursors efficiently up-regulated both RFP and GFP during developmental progression to Foxp3+ CD4SP thymocytes; consistently, in peripheral lymphoid tissues of recipient mice, the progeny of intrathymically injected GFPCD25+ CD4SP tTreg cell precursors exhibited a RFP+GFP- tTreg cell phenotype [7]. Furthermore, in a well-characterized double- transgenic model (TCR-HA × Pgk-HA) of intrathymic Foxp3+ Treg cell induction [26], recognition of a thymic epithelium-derived model antigen (influenza hemagglutinin, HA) by developing TCR-HA+Foxp3 CD4SP cells promoted intrathymic induction and peripheral accumulation of TCR-HA+Foxp3+ tTreg cells with a RFP+GFP+ phenotype [7]. These data indicate that the BAC-Foxp3Cre-GFP reporter is efficiently upregulated during tTreg cell lineage commitment in vivo. Consequently, Foxp3-driven RFP and GFP co-expression in peripheral CD4+Foxp3+ Treg cells closely correlates with their thymic developmental origin (Figures 3A and 3B).

Extrathymic BAC-Foxp3Cre-GFP reporter activity: In striking contrast to intrathymic tTreg lineage commitment, the BAC-Foxp3Cre-GFP reporter was refractory to GFP up-regulation in post-thymic, peripheral CD4+Foxp3 T cells in various settings of Foxp3 induction:

i. During Foxp3+ iTreg cell generation in TGF-β-supplemented TCR stimulation cultures, initially naïve CD4+Foxp3 T cells failed to up-regulate BAC-Foxp3Cre-GFP expression, despite efficient induction of Foxp3 protein and Foxp3IRES-RFP expression; this striking defect was already established at the transcriptional level and maintained under conditions of enhanced Foxp3 induction, e.g. by the addition of excessive amounts of TGF-β or the vitamin A metabolite retinoic acid. The selective BAC- Foxp3Cre-GFP reporter inactivity in vitro could also not be over come by pharmacological inhibition of DNA methyltransfe - rases and CpG methylation by 5-Aza-2-deoxycytidine (5-aza- -dC) [18,27], not withstanding efficient 5-aza-dC-mediated Foxp3+ iTreg cell generation in the absence of added TGF-β.

ii. The differentiation of lymph node-derived physiological CD4+CD25+Foxp3 pTreg cell precursors into Foxp3+ pTreg cells, either in vitro in IL-2-supplemented cultures (in the absence of deliberate TCR or TGF-βR stimulation) or upon adoptive transfer into IL-2-proficient recipient mice, coincided with the up-regulation of Foxp3IRES-RFP expression, whereas expression of the BAC-Foxp3Cre-GFP reporter remained below the detection limit. Additionally, several well-established experimental modalities of Foxp3+ pTreg cell induction in vivo failed to promote BAC-Foxp3Cre-GFP reporter expression, including lymphopenia-driven proliferation of CD4+CD25Foxp3 T cells expressing a transgenic BDC2.5 TCR after adoptive transfer into Rag1−/− recipients, adoptive transfer of CD4+CD25Foxp3 BDC2.5+ T cells into fully immunocompetent recipients and subsequent DEC-205+ DC-targeted delivery of minute amounts of a mimotope peptide to steady-state DCs, as well as oral ovalbumin feeding of Rag2-deficient Foxp3RFP/GFP mice with transgenic DO11.10 TCR expression.

Harnessing Foxp3RFP/GFP mice for studies on developmental Treg cell heterogeneity

The above outlined experiments make a strong case for a scenario in which the activity of the BAC-Foxp3Cre-GFP reporter is exclusively confined to tTreg lineage commitment due to a previously unrealized defect in post-thymic CD4+ T cells. This finding provides a unique opportunity to track viable populations of naturally occurring RFP+GFP+ tTreg and RFP+GFP pTreg cells in peripheral lymphoid tissues of otherwise nonmanipulated Foxp3RFP/GFP mice (Figure 3B). Consistently, Foxp3+ Treg cells with a RFP+GFP phenotype are enriched in Peyer’s patches of the small intestine of Foxp3RFP/GFP mice [7], a site known to support pTreg cell induction [28]. Consistent with their distinct developmental origins, global transcriptional signatures of RFP+GFP+ and RFP+GFP Treg cells from the same anatomical location (e.g. spleen or lymph nodes) exhibit considerable differences in the expression of genes encoding T-cell transcription factors, chemokine receptors, and effector molecules implicated in suppressor function [7]. Thus, the sub-phenotypes of tTreg and pTreg cells are largely specified by their developmental origin, rather than by microenvironmental imprinting. Another key requisite for exploiting differential GFP expression among RFP+ Treg cells in Foxp3RFP/GFP mice as a universal marker to discriminate tTreg and pTreg cells is the preservation of their GFP expression status under pro-inflammatory conditions. In fact, the overwhelming majority of FACS-purified populations of RFP+GFP+ and RFP+GFP Treg cells stably maintained their fluorochrome expression status under TCR stimulatory conditions, both in vitro (APCs + anti-CD3 mAbs or titrating amounts of anti-CD3/CD28 mAb-coated beads ± IL-2, either alone or in the presence of cytokine-secreting CD4+ T effector cells) and under highly inflammatory conditions in vivo (lymphopenia-induced proliferation, autoimmune diabetes, etc.) [7]. Additionally, although RFP+GFP+ tTreg and RFP+GFP pTreg cells were similarly effective in controlling the activation and proliferation of co-cultured conventional CD4+ T effector cells in vitro, the assessment of their suppressor function in vivo revealed marked qualitative differences between both developmental Treg cell sub-lineages: the in vivo accumulation of adoptively transferred RFP+GFP+ tTreg and RFP+GFP pTreg cell populations was largely comparable, but RFP+GFP+ tTreg cells more potently suppressed lymphopenic proliferative expansion of co-transferred conventional CD4+ T cells, and RFP+GFP pTreg cells were far more efficient in the autoimmune protection of insulin-producing cells in an BDC2.5-based adoptive transfer model of type 1 diabetes [7].

Conclusion and Future Perspectives

While the exact mechanistic cause for the restricted BAC-Foxp3Cre-GFP reporter activity during tTreg cell differentiation remains to be determined, the differential expression of GFP in Foxp3RFP/GFP mice provides a unique opportunity to track viable populations of RFP+GFP+ tTreg cells and RFP+GFP pTreg cells in otherwise nonmanipulated mice. Importantly, the GFP expression status (either GFP+ or GFP) of Foxp3+RFP+ Treg cells is preserved under pro-inflammatory conditions, which represents a key advantage of the Foxp3RFP/GFP model. Future studies on the synergistic action of tTreg and pTreg cells [4-6] under physiological conditions [7] will be facilitated by the recent generation of Foxp3RFP/GFP mice on genetically well-defined backgrounds that are commonly used for immunological research (e.g. C57BL/6, BALB/c, NOD, etc.). This may include studies on the relative contribution of tTreg and pTreg cells in clinical settings of exacerbated (chronic infections by microbes and parasites, cancer) and insufficient (autoimmunity, allergies, transplant rejection) Treg cell suppressor activity. Additionally, genome-wide single-cell transcriptomics of RFP+GFP+ tTreg cells and RFP+GFP pTreg cells may help identifying a surface marker (or a combination of surface markers) suitable to discriminate developmental CD4+FOXP3+ Treg cell sub-lineages in humans.

Acknowledgments

KK was supported by the Competence Network for Diabetes mellitus funded by the Federal Ministry of Education and Research (BMBF; FKZ 01GI0805-07) and by a BMBF grant to the German Center for Diabetes Research (DZD e.V., FKZ01GI0924). Additionally, KK and AIG were supported by the FZT 111 (DFG, Center for Regenerative Therapies Dresden, Cluster of Excellence). AIG received a DFG grant (GA 1576/1-2) within the SPP 1468 IMMUNOBONE, and K.K and A.C. received a DFG grant (KR 2316/3-1, CH 1862/2-1) within the FOR 2599. M.S. was supported by the Dresden International Graduate School for Biomedicine and Bioengineering (DIGS-BB) and funded by a DIGS-BB Fellow Award.

References

  1. Petzold C, Schallenberg S, Stern JNH, Kretschmer K (2012) Targeted Antigen vDelivery to DEC-205(+) Dendritic Cells for Tolerogenic Vaccination. Rev Diabet Stud 9: 305-318.
  2. Lio CW, Hsieh CS (2008) A two-step process for thymic regulatory T cell development. Immunity 28: 100-111.
  3. Schallenberg S, Tsai PY, Riewaldt J, Kretschmer K (2010) Identification of an immediate Foxp3- precursor to Foxp3+ regulatory T cells in peripheral lymphoid organs of nonmanipulated mice. J Exp Med 207: 1393-1407.
  4. Haribhai D, Williams Jason B, Jia S, Nickerson D, Schmitt Erica G, Edwards B, et al. (2011) A Requisite Role for Induced Regulatory T Cells in Tolerance Based on Expanding Antigen Receptor Diversity. Immunity 35: 109-122.
  5. Josefowicz SZ, Niec RE, Kim HY, Treuting P, Chinen T, et al. (2012) Extrathymically generated regulatory T cells control mucosal TH2 inflammation. Nature 482: 395-399.
  6. Samstein Robert M, Josefowicz Steven Z, Arvey A, Treuting Piper M, Rudensky Alexander Y (2012) Extrathymic Generation of Regulatory T Cells in Placental Mammals Mitigates Maternal-Fetal Conflict. Cell 150: 29-38.
  7. Petzold C, Steinbronn N, Gereke M, Strasser RH, Sparwasser T, Bruder D, et al. (2014) Fluorochrome-based definition of naturally occurring Foxp3+ regulatory T cells of intra- and extrathymic origin. Eur J Immunol 44: 3632-3645.
  8. Feuerer M, Hill JA, Kretschmer K, von Boehmer H, Mathis D, et al. (2010) Genomic definition of multiple ex vivo regulatory T cell subphenotypes. Proc Natl Acad Sci U S A 107: 5919-5924.
  9. Thornton AM, Korty PE, Tran DQ, Wohlfert EA, Murray PE, Belkaid Y, et al. (2010) Expression of Helios, an Ikaros transcription factor family member, differentiates thymic-derived from peripherally induced Foxp3+ T regulatory cells. J Immunol 184: 3433-3441.
  10. Weiss JM, Bilate AM, Gobert M, Ding Y, Curotto de Lafaille MA, Parkhurst CN, et al. (2012) Neuropilin 1 is expressed on thymus-derived natural regulatory T cells, but not mucosa-generated induced Foxp3+ T reg cells. J Exp Med 209: 1723-1742.
  11. Yadav M, Louvet C, Davini D, Gardner JM, Martinez-Llordella M, Bailey-Bucktrout S, et al. (2012) Neuropilin-1 distinguishes natural and inducible regulatory T cells among regulatory T cell subsets in vivo. J Exp Med 209: 1713-1722
  12. Gottschalk RA, Corse E, Allison JP (2012) Expression of Helios in Peripherally Induced Foxp3+ Regulatory T Cells. J Immunol 188: 976-980.
  13. Verhagen J, Wraith DC (2010) Comment on "Expression of Helios, an Ikaros Transcription Factor Family Member, Differentiates, Thymic-Derived from Peripherally Induced Foxp3+ T Regulatory Cells". J Immunol 185: 7129.
  14. Zabransky DJ, Nirschl CJ, Durham NM, Park BV, Ceccato CM, Bruno TC, et al. (2012) Phenotypic and functional properties of Helios+ regulatory T cells. PLoS One 7: e34547.
  15. Schallenberg S, Petzold C, Riewaldt J, Kretschmer K (2013) Regulatory T cell-based immunotherapy: Prospects of antigen-specific tolerance induction. In Medical Advancements in Aging and Regenerative Technologies: Clinical Tools and Applications. Edited by A D. Hershey, USA: IGI Global: 112-136
  16. Kretschmer K, Apostolou I, Hawiger D, Khazaie K, Nussenzweig MC, et al. (2005) Inducing and expanding regulatory T cell populations by foreign antigen. Nat Immunol 6: 1219-1227.
  17. Walker MR, Carson BD, Nepom GT, Ziegler SF, Buckner JH. (2006) De novo generation of antigen-specific CD4+CD25+ regulatory T cells from human CD4+CD25– cells. Nat Protoc 1: 653-661.
  18. Polansky JK, Kretschmer K, Freyer J, Floess S, Garbe A, Baron U, et al. (2008) DNA methylation controls Foxp3 gene expression. Eur J Immunol 38: 1654-1663.
  19. Akimova T, Beier UH, Wang L, Levine MH, Hancock WW (2011) Helios Expression Is a Marker of T Cell Activation and Proliferation. PLoS ONE 6: e24226.
  20. Shevach EM, Thornton AM (2014) tTregs, pTregs, and iTregs: similarities and differences. Immunol Rev 259: 88-102.
  21. Zheng Y, Josefowicz S, Chaudhry A, Peng XP, Forbush K, et al. (2010) Role of conserved non-coding DNA elements in the Foxp3 gene in regulatory T-cell fate. Nature 463: 808-812.
  22. Zhou X, Jeker LT, Fife BT, Zhu S, Anderson MS, et al. (2008) Selective miRNA disruption in T reg cells leads to uncontrolled autoimmunity. J Exp Med 205: 1983-1991.
  23. Schallenberg S, Petzold C, Tsai P-Y, Sparwasser T, Kretschmer K (2012) Vagaries of Fluorochrome Reporter Gene Expression in Foxp3+ Regulatory T Cells. PLoS ONE 7: e41971.
  24. Srinivas S, Watanabe T, Lin CS, William C, Tanabe Y, et al. (2001) Cre reporter strains produced by targeted insertion of EYFP and ECFP into the ROSA26 locus. BMC Dev Biol 1: 4.
  25. Wan YY, Flavell RA (2005) Identifying Foxp3-expressing suppressor T cells with a bicistronic reporter. Proc Natl Acad Sci U S A 102: 5126-5131.
  26. Klein L, Khazaie K, von Boehmer H (2003) In vivo dynamics of antigen-specific regulatory T cells not predicted from behavior in vitro. Proc Natl Acad Sci U S A 100: 8886-8891.
  27. Freudenberg K, Lindner N, Dohnke S, Garbe A, Kretschmer K (2018) Critical role of TGF-beta and IL-2 receptor signaling in Foxp3 induction by an inhibitor of DNA methylation. Front Immunol in press.
  28. Curotto de Lafaille MA, Lafaille JJ (2009) Natural and Adaptive Foxp3+ Regulatory T Cells: More of the same or a Division of Labor? Immunity 30: 626-635.
Citation: Dohnke S, Schreiber M, Schallenberg S, Simonetti M, Fischer L, et al. (2018) Approaches to Discriminate Naturally Induced Foxp3+ Treg cells of Intra-and Extrathymic Origin: Helios, Neuropilin-1, and Foxp3RFP/GFP. J Clin Cell Immunol 9: 540.

Copyright: © 2018 Dohnke S, et al. This is an open-access article distributed under the terms of the Creative Commons Attribution License, which permits unrestricted use, distribution, and reproduction in any medium, provided the original author and source are credited.
Top