Skip to main content
PLOS One logoLink to PLOS One
. 2025 May 30;20(5):e0324105. doi: 10.1371/journal.pone.0324105

Ovalbumin-specific regulatory T cells differentiated from the naïve phenotype (CD44loCD62Lhi) in mesenteric lymph nodes stably suppress enteropathy even in severe food-allergic mice

Kyoko Shibahara 1,#, Tomohiro Hoshino 1,#, Haruka Nakanishi 1, Kosuke Nishitsuji 1, Kohei Soga 1, Yoshiyo Bamba 1, Satoshi Hachimura 1, Haruyo Nakajima-Adachi 1,2,*
Editor: Masanori A Murayama3
PMCID: PMC12124522  PMID: 40446078

Abstract

Impaired expansion, stability, and function of regulatory T cells (Tregs) are reported in patients with severe allergy. Transfer of Tregs is a potential means of treating severe food allergy; however, methods to obtain allergen-specific Tregs with stable regulatory activities are needed. To achieve our goal, we examined the characteristics of allergen-specific Tregs by comparing two mouse strains transgenic for the ovalbumin (OVA)-specific T cell receptor gene: Rag23−3 and RagD10 mice (OVA23−3 and DO11.10 crossed with Rag2 knockout mice, respectively). RagD10 is a tolerant model, whereas Rag23−3 shows severe allergy when fed egg white (EW). To examine the differentiation of CD4+ T cells into Foxp3+ Tregs (induced Tregs; iTregs), CD4+ T cells or whole cells from mesenteric lymph nodes or spleens were cultured under Treg-polarization conditions and stimulated with either a combination of anti-CD3 and anti-CD28 antibodies or OVA plus antigen-presenting cells. After stimulation with the antibodies, iTregs were induced at comparable levels from CD4+ T cells from untreated Rag23−3 and RagD10 mice. Transfer of the resultant iTregs from untreated Rag23−3 mice suppressed allergic responses in EW-fed Rag23−3 mice. In contrast, stimulation with OVA plus antigen-presenting cells prevented the differentiation of iTregs from CD4+ T cells from untreated Rag23−3 mice, suggesting that OVA-induced T-cell receptor signaling inhibits effective Treg differentiation. Furthermore, antibody-mediated differentiation afforded significantly more iTregs differentiation of naïve (CD44loCD62Lhi) CD4+ T cells than of effector/effector memory (CD44hiCD62Llo) T cells isolated from the mesenteric lymph nodes of EW-fed Rag-23–3 mice. Excessive production of interleukin-4 and interferon-gamma by CD4+ T cells from EW-fed Rag23−3 mice significantly inhibited Treg induction in RagD10 mice, suggesting the severe allergic cytokine milieu likely prevents their differentiation. However, our study showed that allergen-specific Tregs with regulatory activity can be obtained from naïve CD4+ T cells from the intestinal immune system of mice even with severe allergy.

Introduction

Over the last two decades, the prevalence of food allergies has been increasing in developing countries; however, a standard treatment is yet to be established [1,2]. Oral immunotherapy, which involves feeding an allergic individual an increasing amount of allergen with the goal of increasing the threshold that triggers a reaction, has been shown to confer food-allergic patients with tolerance to allergens, but there are many issues related to safety (e.g., severe anaphylaxis) and efficacy that must be overcome before this approach can be more widespread use [39].

At the cellular level, allergen-specific regulatory T cells (Tregs) are considered to be key players in inhibiting allergic responses to food and maintaining tolerance [1012]. Indeed, several studies have shown that individuals with food allergies who were able to tolerate by oral immunotherapy, or children who had outgrown a food allergy, possess allergen-specific Tregs with stronger abilities to inhibit the development of allergic inflammation compared with those in control patients still with severe food allergy [3,12,13]. This suggests that induction of allergen-specific Tregs in individuals with food allergies may confer stable tolerance in these individuals. Thus, just as in other inflammatory disease [14,15], Treg-transfer may be a promising means of treating food allergy.

To understand more about the characteristics and roles of allergen-specific Tregs, methods to induce their differentiation and to analyze their roles in food-allergic patients are desired; however, many allergists are trying to find markers for stable Tregs specifically activated by antigens in peripheral blood, but the goal has not been achieved, although CD137 has been proposed as a candidate marker [1618]. In addition, Tregs induced under allergic inflammation conditions are highly plastic and easily induced to change into effector T cells, making their identification more difficult [1618]. Indeed, the differentiation of antigen-specific Tregs occurs continuously in the intestine [19], and it is hypothesized that food allergy develops as a result of shifts in the balance of the activities of these antigen-specific Tregs and effector T cells [11]. This hypothesis is supported by reports showing that components of the microbiome destabilized Tregs in the small intestine, causing them to be easily induced to change to effector T cells [20,21]. To achieve the goal of adoptive transfer of allergen-specific Tregs for the treatment of food allergies, a more detailed understanding of how allergen-specific Tregs differ between individuals with and without allergy and in the various tissues is needed.

Here, to examine the differences between allergen-specific Tregs in healthy and allergic individuals, we used two mouse models of human food-allergic symptoms (Rag23−3 and RagD10), as reported previously [22,23]. Briefly, the two models are both transgenic for the ovalbumin (OVA)-specific T-cell-receptor gene and lack recombination-activating gene 2 (Rag2). As such, these mice produce only OVA-specific T cells in response to OVA challenge, which allows for the study of the functions of T cells without the need to consider B cell responses. Rag23−3 mice are a cross between OVA23−3 mice and Rag2 knockout mice and are used as a model of food allergy, whereas RagD10 mice are a cross between DO11.10 mice and Rag2 knockout mice and are used as a model of oral tolerance. When fed a diet containing egg white (EW) protein, the most abundant component of which is OVA, RagD10 mice acquire oral tolerance to the EW diet that is characterized by induction of OVA-specific Tregs expressing Foxp3, a master regulator of Tregs, and a quick decline of immune responses. The mice predominantly produce IFN-γ by stimulation with anti-CD3 antibody or OVA, indicating Th1-biased responses [22]. In contrast, Rag23−3 mice develop food-allergic enteropathy that is characterized by a lack of OVA-specific Foxp3+ Tregs during the allergic inflammation phase and strong IL-4 responses, when they were stimulated with anti-CD3 antibody or OVA [22]. Although continuous EW-feeding increases the OVA-specific Foxp3+ Treg population and Rag23−3 mice recover from the allergic inflammation, our previous findings indicate that these Tregs are unable to prevent the reoccurrence of food allergy [22], suggesting that continuous allergen administration might not be a suitable approach for obtaining stable Tregs. Indeed, in the same study, we showed that the stability of OVA-specific Tregs induced during feeding with EW-diet was higher in DO11.10 mice than in OVA23−3 mice [22].

It has been reported that the functions and phenotypes of Tregs differentiated by using a combination of anti-CD3 and anti-CD28 monoclonal antibodies (mAbs) differ from those of Tregs differentiated with an antigen in the presence of antigen-presenting cells [24]. Thus, in the present study, we examined how to obtain stable and suppressive Tregs from the Rag23−3 food-allergic enteropathy model by comparing both the inducing means and their resultant differentiation levels and functions of Tregs induced in Rag23−3 and RagD10 mice when fed an EW diet. We found that OVA-specific Tregs differentiated from naïve T cells from the mesenteric lymph nodes (mLNs) and that, when activated under appropriate conditions using Abs, these OVA-specific Tregs prevented allergic inflammation, even when the cells were from the food-allergic model (EW-fed Rag23−3 mice). However, we also found that a severe allergic cytokine milieu, characterized by prominent production of IFN-γ and interleukin (IL)-4 by CD4+ T cells, prevented Treg induction, even in CD4+ T cells from the tolerant model (RagD10 mice).

Materials and methods

Animals

Rag23−3 mice (OVA23−3 mice [25] crossed with Rag2 knockout mice) were kindly provided by S. Habu (Tokai University School of Medicine, Kanagawa, Japan). RagD10 mice (DO11.10 mice crossed with Rag2 knockout mice) were kindly provided by Y. Wakatsuki (Kyoto University, Kyoto Japan). Both strains of mouse carried the BALB/cA background, possessed OVA-specific T-cell-receptor genes, and were bred at Sankyo Labo Service Corporation Inc. (Tokyo, Japan). The only lymphocytes that these strains of mice produce in response to OVA challenge are OVA-specific T cells; therefore, we can observe the antigen-specific T cell responses without consideration of B cell responses. When fed a diet containing EW protein for 9 days, RagD10 mice develop tolerance to the protein, whereas Rag23−3 mice show a severe allergic response and do not develop tolerance. In all experiments, sex-matched mice older than 8 weeks of age were used [22].

The mice were housed in cages (≤5 mice/cage; 182 × 260 × 128 mm; CL-0103–2; CLEA Japan, Inc.) at The University of Tokyo (Tokyo, Japan) and were maintained under specific-pathogen-free conditions at a room temperature of 22°C and under a 12:12-h light:dark cycle. Sterilized deionized drinking water and sterilized commercial chow were provided ad libitum. The maintenance conditions were monitored daily. When necessary for cellular and histological analysis, mice were euthanized by cervical dislocation by experts. During the adoptive transfer of Tregs, to facilitate the subsequent injection of cells into the caudal vein, a mouse was placed in a small box with only its tail sticking out through the hole in the box for a few minutes. The injection was performed without anesthetization as smoothly and quickly as possible by experts. All experiments were performed in accordance with the guidelines of the University of Tokyo for the care and use of animals.

Antigen administration

Antigen (OVA) was administered to the mice via their feed. The antigen-containing diet was a solid feed with the protein fraction consisting only of EW (Funabashi Farm Co., Chiba, Japan). A control diet was also used, which was a solid feed with the protein fraction consisting only of casein (CN, Funabashi Farm). The mice were fed one of these diets ad libitum for 7 days.

Cell preparation and sorting

To prepare single-cell suspensions from mLNs (whole cells), mLNs were collected and cut into small pieces in complete RPMI 1640 medium (Thermo Fisher Scientific, Rockford, IL) supplemented with fetal calf serum (FCS; 10%, Thermo Fisher Scientific), penicillin/streptomycin (100 U/mL, Thermo Fisher Scientific), and 2-mercaptoethanol (50 μM, Fujifilm Wako Pure Chemical Corp, Osaka, Japan). The small pieces of mLN were cultured in complete RPMI1640 medium supplemented with 10 mM HEPES (Thermo Fisher Scientific), 1 mg/mL collagenase (final concentration; Fujifilm Wako Pure Chemical Corp), and 1 mg/mL DNase I (final concentration, Roche, Mannheim, Germany) with stirring at 37°C for 70 min. After incubation, the resulting mixture was passed through a 100-µm cell strainer (Falcon, Corning, NY, USA), and the cells collected through the strainer were washed twice and then resuspended in the complete RPMI. To prepare single-cell suspensions from spleen (whole cells), spleens were harvested, ground in complete RPMI medium with the bottom of a plastic syringe piston (Terumo, Tokyo, Japan), and passed through an 86-µm Tetron mesh (Tokyo Screen, Tokyo). The cells that passed through the mesh were washed twice and resuspended in complete RPMI medium. CD4+ T cells, CD44loCD62LhiCD4+ T cells (naïve T cells), and effector/effector memory CD44hiCD62LloCD4+ T cells (effector/effector memory; EMTs) were isolated from the single-cell suspensions obtained from the mLNs and spleens of Rag23−3 mice fed with the CN- or EW-diet for 7 days. CD4+ T cells were isolated by using a magnetic-activated cell sorting system (Miltenyi Biotec, Bergisch Gladbach, Germany) in accordance with the manufacturer’s instructions. For cell sorting, the single cells (4–8 × 107 spleen or mLN cells) were suspended in 1 mL of phosphate-buffered saline containing 2% FCS (2% FCS–PBS) and then blocked by incubation with an anti-CD16/32 mAb (BD Pharmingen, Franklin Lakes, NJ, USA) at 4°C for 15 min. Next, the cell suspensions were centrifuged at 1400 rpm and 4°C for 5 min, resuspended in 1 mL of 2% FCS–PBS, and stained with allophycocyanin (APC)-conjugated anti-mouse CD4 (GK1.5; BioLegend, San Diego, CA, USA), APC-Cy7-conjugated anti-mouse CD44 (IM7; BD Pharmingen), and phycoerythrin (PE)-Cy7-conjugated anti-mouse CD62L (MEL-14; BioLegend) mAbs at 4°C for 20 min. After washes with 2% FCS–PBS and resuspension in 1 mL of 2% FCS–PBS, the cells were stained with 7-aminoactinomycin D (BioLegend) at room temperature for 5 min. After washes, the cells were resuspended in 2 mL of 2% FCS–PBS (2–4 × 107 cells/mL), and the naïve T cells and EMTs were sorted by using a FACSAria II cell sorter (BD Biosciences, Franklin lakes, NJ, USA).

Treg-polarization culture

To induce Foxp3+ Tregs, whole cells (2 × 105 cells/well in 96-well plates), CD4+ T cells (1 × 105 cells/well in 96-well plates or 1 × 106 cells/well in 24-well plates), naïve cells (1 × 105 cells/well in 96-well plates), or EMTs (1 × 105 cells/well in 96-well plates) were incubated under in vitro Treg-polarization culture conditions in the presence of transforming growth factor-beta (TGF-β) 1 (2 ng/mL; R&D Systems, Minneapolis, MN, USA), retinoic acid (1 µM; Fujifilm Wako Pure Chemical, Osaka, Japan), and recombinant (r) IL-2 (2 ng/mL; R&D Systems) for 48 or 72 h. Whole cells were stimulated with OVA (0.25 mg/mL; Sigma-Aldrich, St. Louis, MO, USA), whereas CD4+ T cells, naïve T cells, and EMTs were stimulated with plate-bound anti-CD3 (145-2C11; BD Biosciences) and anti-CD28 (37.51; BioLegend) mAbs. In some experiments, pooled whole cells or purified CD4+ T cells were cultured with plate-bound anti-CD3 and anti-CD28 mAbs and rIL-2 (2 ng/mL) as a control condition. The culture supernatant obtained from each culture was also used for analysis of cytokine production by enzyme-linked immunosorbent assay (ELISA).

Flow cytometry

Flow cytometry was used to identify the phenotype of CD4+ T cells by analyzing the expression of cell-surface molecules. Cells collected from the Treg-polarization culture were incubated with anti-CD16/32 mAb (BD Pharmingen) at 4°C for 15 min to block non-specific binding to Fc receptors. After washes, the cells were stained with the following fluorescent mAbs: fluorescein isothiocyanate (FITC)-conjugated anti-mouse CD4 mAb (H129.19; BD Pharmingen), APC-conjugated anti-mouse CD4 mAb (GK1.5; BioLegend), FITC-conjugated anti-mouse CD25 mAb (3C7; BioLegend), PE/Cy7-conjugated anti-mouse IL-4 receptor (IL-4R) mAb (1015F8; BioLegend), biotin-conjugated anti-mouse IFN-γ receptor (IFN-γR) mAb (2E2; BioLegend), anti-APC-Cy7-conjugated anti-mouse CD44 mAb (IM7; BD Pharmingen), and PE-Cy7-conjugated anti-mouse CD62L mAb (MEL-14; BioLegend). Foxp3 intracellular staining was performed by using an eBioscience Foxp3/Transcription Factor Staining Set (Invitrogen). Briefly, cells were fixed, permeabilized, and stained with APC- or PE-conjugated anti-mouse Foxp3 mAb (FJK-16s; eBioscience, Santa Clara, CA, USA). A FACSVerse cell analyzer (BD Biosciences) and FlowJo software (Ver 10.7.1, BD Biosciences) were used for analysis.

ELISA

The amounts of IL-2, IL-4, and IFN-γ in culture supernatants were measured by ELISA, as described previously [23]. Anti-mouse IL-2 (JES6-1A12; BD Biosciences), anti-mouse IL-4 (11B11; BD Biosciences), and anti-mouse IFN-γ (R4-6A2; BD Biosciences) mAbs were used as capture Abs; biotin-conjugated anti-mouse IL-2 (JES6-5H4; BD Biosciences), biotin-conjugated anti-mouse IL-4 (BVD6-24G2; BD Biosciences), and biotin-conjugated anti-mouse IFN-γ (XMG1.2; BD Biosciences) mAbs were used as detection Abs. Recombinant murine IL-2 (rIL-2; Peprotech, Rocky Hill, NJ, USA), IL-4 (rIL-4; R&D Systems), and IFN-γ (rIFN-γ; Peprotech) were used as standards. The amount of IL-10 production in culture supernatant was analyzed by using Mouse IL-10 Quantikine ELISA kit (R&D Systems).

Induction of Tregs by using CD4+ T cell culture supernatants

To prepare the supernatants, RagD10 and Rag23−3 mice were fed the EW or CN diet for 7 days and spleens were harvested and processed into single-cell suspensions. Then, CD4+ T cells (1 × 105 cells in 96 well plates) isolated from the suspensions were stimulated with plate-bound anti-CD3 (BD Biosciences) and anti-CD28 (BioLegend) mAbs for 48 h, and the supernatant was collected. To analyze the effects of cytokines contained in the supernatants on the induction of Tregs, CD4+ T cells from spleens of untreated RagD10 mice were stimulated with plate-bound anti-CD3 (BD Biosciences) and anti-CD28 (BioLegend) Abs and then cultured with the prepared supernatants under Treg-polarization culture conditions for 72 h. Each culture supernatant of the cells derived from CN- or EW-fed RagD10 or Rag23−3 mice described above was added to 50% of the total volume of the Treg-polarization culture medium of the cells from the untreated RagD10 mice. To identify cytokines inhibiting Treg induction from CD4+ T cells of untreated RagD10 mice, the supernatant derived from EW-fed R23-3 mice was added to the culture medium with or without anti-IL-4 Ab (αIL-4 Ab; 1 µg/mL; BioLegend) or anti-IFN-γ Ab (αIFN-γ Ab; 1 µg/mL; BioLegend) and cultured for 72 h. The volume of the supernatant, or the medium with or without Abs, was 25% of the total culture medium (200 µL) in each well.

Treg induction under rIL-4 or rIFN-γ supplemented conditions

CD4+ T cells (1 × 105 cells in 96-well plates) were prepared from splenocytes derived from Rag10 mice fed the CN-diet for 7 days and then stimulated with plate-bound anti-CD3 and anti-CD28 Abs under Treg-polarization conditions supplemented with mouse rIFN-γ(0, 1.0, 2.0, or 3.5 ng/mL; 315−05, Peprotech) and mouse rIL-4 (0, 0.5, 1.5, or 2.5 ng/mL; 214−14, Peprotech) for 72 h.

Adoptive transfer of Tregs into Rag23-3 mice

CD4+ T cells isolated from whole-cell suspensions prepared from the spleens of untreated Rag23−3 mice (1 × 106 cells) were cultured under control conditions by being stimulated with plate-bound anti-CD3 and anti-CD28 mAbs in the presence of 2 ng/mL rIL-2, or cultured under Treg-polarization conditions by being stimulated with plate-bound anti-CD3 and anti-CD28 mAbs in the presence of rIL-2 (2 ng/mL), TGF-β1 (5 ng/mL), and retinoic acid (1 µM) in 24-well plates. Using the FlowJo software, we confirmed that the purity of Foxp3+ cells was > 90% in the Treg-polarization medium. Control cells and Treg-abundant cells were collected from each culture, re-suspended in PBS, and transferred via a caudal vein to untreated Rag23−3 mice (1 × 107 cells/head). As a positive control group, untreated Rag23−3 mice were injected with PBS. From 1 day after the injection of the cells or PBS, each group of Rag23−3 mice were fed the EW diet for 8 days and their weights were measured. On day 7 of EW feeding, the mice were euthanized. Jejunum was collected for histological analysis, and mLN and spleen were collected for analysis of CD4+ T cell proliferation and their cytokine (IL-4 and IFN-γ) production.

Histological analysis

A 3-cm section of jejunum, 10 cm away from the distal end of the stomach, was harvested and fixed in 10% formalin (Fujifilm Wako Pure Chemical). Tissues were embedded in paraffin, 4-μm section were obtained, and the sections were stained with hematoxylin and eosin. Morphological features were observed under a BX-51 microscope (Olympus, Tokyo, Japan).

Measurement of cytokine production by CD4+ T cells isolated from Rag23−3 mice that received adoptive transfer of Tregs

CD4+ T cells were isolated by magnetic-activated cell sorting from single-cell suspensions of mLN or spleen from the recipient mice used in the adoptive Treg transfer experiment. The CD4+ T cells (1 × 105 cells/well) were then stimulated with OVA (0.25 mg/mL; Sigma-Aldrich) plus splenocytes from BALB/cA mice (CLEA Japan Inc., Tokyo, Japan) treated with mitomycin C (Fujifilm Wako Pure Chemical Corp) as antigen-presenting cells (CD4+ T cell-culture) for 48 h. The amount of IL-4 or IFN-γ in the culture supernatant was measured by ELISA, as already described.

Proliferation assay

CD4+ T cells were stimulated with OVA and antigen-presenting cells for 24 h and then [3H]-thymidine (0.5 µCi/well; Moravek Biochemicals, Brea, California, USA) was added to each well. After incubation for 16 h, cells were harvested and the incorporation of [3H]-thymidine was measured by a scintillation counter (Perkin Elmer, Rodgau, Germany).

Statistical analysis

Unpaired student’s t-test was performed for comparison between two groups. To compare more than two groups of data, Tukey’s honestly significant difference (HSD) test or Dunnett’s test was performed following ANOVA by using R software (Ver 3.3.1). p values below 0.05 were considered significant. All data in the graphs are shown as mean or mean ± 1 SD.

Ethics statement

This study was carried out in strict accordance with the recommendations in the Guide for the Animal Use Committee of the Faculty of Agriculture at the University of Tokyo. Approval numbers: P15-022, P15-023, P17-044, and P19-038. All animal experiments were conducted at the University of Tokyo according to the ARRIVE guidelines regarding the care and use of experimental animals (https://www.u-tokyo.ac.jp/adm/lifescience/ja/doukisoku.html). Our experiments were approved without setting preemptive humane endpoints, because during the experimental period (7 days), the mice did not exhibit other clinical signs than weight loss.

Results

Weight change and intestinal histology in Rag23−3 and RagD10 mice

In our previous study using the Rag23−3 and RagD10 mice fed for 7 days with either the EW or CN diet, we reported that EW-fed Rag23−3 mice have a difficulty in inducing Foxp3 expression by CD4+ T cells, whereas EW-fed RagD10 mice readily induce the expression, which resulted in intestinal inflammation and weight loss in the Rag23−3 but not the RagD10 mice [22]. In the present study, we observed results consistent with those of our previous study, with the EW-fed Rag23−3 mice showing a greater loss of body weight compared with the CN-fed Rag23−3 mice or EW- or CN-fed RagD10 mice (S1A Fig). Hematoxylin and eosin staining of sections of jejunum revealed that the EW-fed Rag23−3 mice also developed intestinal inflammation during the experimental period, whereas the other mice did not (S1B Fig).

Stimulation with OVA impairs Treg induction but promotes EMT induction and IL-4 and IFN-γ production in Rag23−3 mice, but not in RagD10 mice

To investigate the abilities of the CD4+ T cells in the two mouse models to differentiate into Tregs, we firstly stimulated whole cells isolated from mLN cells (including OVA-specific T cells and antigen-presenting cells) with OVA under Treg-polarization culture conditions and examined by flow cytometry; the gating strategy used to identify the Treg and EMT subpopulations within the CD4+ T cell population is shown in Fig 1A. The Treg population was significantly larger in the EW-fed RagD10 mice than in those fed the CN-diet, accounting for around 22% and 5% of the CD4+ T cell population, respectively (Fig 1B). In contrast, the sizes of the Treg populations were comparable in the CN- and EW-fed Rag23−3 mice, with both also around 5%. The EMT populations in both mouse strains fed the EW-diet were significantly larger than those in the mice fed the CN-diet (RagD10: around 8% vs. 3%; Rag23−3: around 20% vs. 5%; Fig 1C). In both treatment groups for both mouse strains, around 80% of the Tregs expressed CD25, a common marker of murine Tregs and a component of the Treg IL-2 receptor (Fig 1B). The frequency of CD25+ T cells significantly increased in EW-fed mice in spite of the difference of the strains, showing that EW feeding even at low levels, induced Tregs, even at minor level under the severe allergic condition of Rag23−3 mice.

Fig 1. Ovalbumin (OVA) stimulation of mesenteric lymph node T cells impaired regulatory T cell differentiation under a characteristic cytokine milieu accompanied by greater differentiation of effector/effector memory T cells (EMTs) in Rag23−3 mice, but not RagD10 mice.

Fig 1

RagD10 mice and Rag23−3 mice were fed a diet containing egg white (EW) or casein (CN) for 7 days. Whole cells isolated from the mesenteric lymph nodes (mLNs) were cultured under Treg-polarization culture conditions and stimulated with OVA. A) Gating strategy used to identify the regulatory T cell (Treg; Foxp3+) and effector/effector memory T cell (EMT; CD44hiCD62lo) subpopulations within the CD4+ T cell population. B) Frequency of Tregs in the CD4+ T cell population and of CD25 expression in the Treg population. C) (left) Frequency of EMTs in the CD4+ T cell population. Each plot in b) and c-left) represents the value for a single well and the horizontal lines indicate the mean value (n= 4, mixture of cells from two to three mice/group). (right) Flow cytometric analysis of the EMTs of Rag23−3 and RagD10 mice. EMT fractions are shown within the black squares. D) Concentrations of interleukin (IL)-4, interferon gamma (IFN-γ), and IL-2 in the culture supernatants (n= 3, mixture of cells from three mice/group). Data are representative of two independent experiments. Statistical analysis: Tukey’s HSD test [#p < 0.05 (CN vs EW in each strain); * p < 0.05, ** p < 0.01, and *** p < 0.001 (between different groups)]. N.D., not detected.

Examination of the cytokine milieu produced by the mLN cells after stimulation with OVA revealed characteristic cytokine profiles for IL-4 production, a marker of Th2-type immune responses; IFN-γ production, a marker of Th1-type immune responses; and IL-2 production, a marker of T cell proliferation (Fig 1D). The RagD10 mice, irrespective of diet, showed no detectable IL-4 production, high IFN-γ production, and no IL-2 production. In contrast, the Rag23−3 mice fed the EW-diet showed high IL-4 production, whereas those fed the CN-diet showed no IL-4 production. For IFN-γ production, the EW-fed Rag23−3 mice showed the same high production as was observed for the RagD10 mice, but the CN-fed mice showed significantly less production. For IL-2, the Rag23−3 mice showed some production irrespective of diet, but the production in the CN-fed mice was significantly greater than that in the EW-fed mice.

Taken together, these results indicate that OVA stimulation resulted in greater T cell proliferation in the Rag23−3 mice than in the RagD10 mice, which is consistent with our previous findings [22]. In addition, these results suggest that the CD4+ T cell population in EW-fed Rag23−3 mice is considerably activated and that EMTs are promoted from the naïve to the Th2 phenotype. Given the observed increase of Treg frequency in the EW-fed RagD10 mice, we expected to also observe increased IL-2 production in these mice, but this was not found to be the case. Since these data were collected at 7 days after the start of EW feeding, these findings imply that the response to OVA in RagD10 mice is fully attenuated by day 7 of EW feeding. Although IFN-γ production tended to be suppressed by EW-feeding compared with those in CN-fed RagD10 mice, the RagD10 mice showed significantly greater IFN-γ production, indicating that the immune response to OVA in RagD10 mice is fundamentally of the Th1 type, which is consistent with previous reports [22,26]

The T cells of CN-fed Rag23−3 mice are reported to produce large amounts of IL-2, a cytokine needed for Treg differentiation, but not IL-4, which is suggested to suppress Treg induction [27]. These findings are inconsistent with our finding that the Treg population is very small in the CN-fed Rag23−3 mice (Fig 1B) despite high IL-2 production (Fig 1D). There are two possibilities for this inconsistency: 1) excessive activation of EMTs inhibit Treg differentiation, independently of IL-2 production [28] or 2) excess activation of the overall immune response through antigen-specific T cell receptor via antigen presenting cells and resultant suppression of their TGF-β1 receptor expression on T cells reduce the responsiveness of CD4+ T cells to TGF-β1, leading resultant inhibition of Treg differentiation [29]. However, the Treg population induced by responses of CD4+ T cells to TGF-β1 was rather larger in Rag 23−3 mice than in RagD10 mice (S2 Fig).

Antibodies stimulation induces CD4+ T cells from CN-fed, but not EW-fed Rag23−3 mice to differentiate into Tregs, whereas CD4+ T cells from RagD10 mice differentiate regardless of diet

The results presented in the previous section suggested that transcription factor activation via antigen-specific T cell receptors signaling for Foxp3 expression may differ in the presence (antigen plus antigen-presenting cell) or absence (no-antigen mediated) of OVA as reported by Zhao C. et al [24]. Therefore, we next examined the effects of no-antigen mediated induction of Tregs and EMTs in CD4+ T cells isolated from the spleens and mLNs of the model mice by using anti-CD3 and anti-CD28 mAbs. In contrast to the earlier data, the frequency of Tregs in the T cells isolated from the spleen or mLNs of CN-fed Rag23−3 mice was surprisingly comparable to those in T cells of CN-fed RagD10 mice (Fig 2A). However, the frequency of Tregs in the EW-fed Rag23−3 mice was significantly reduced compared with that in the CN-fed mice for both the spleen-derived and mLN-derived cells, furthermore is higher in the spleen derived compared with mLN-derived cells, which was consistent with the data presented in Fig 1. In RagD10 mice, the frequency of Tregs in the mLN-derived cells was significantly higher in the EW-fed mice than in the CN-fed mice, whereas that in the cells from the spleen was significantly lower in the EW-fed mice than in the CN-fed mice. We have reported that Foxp3 molecule is more easily induced in the mLN than in the spleen, but in the food-allergic intestinal inflammation model like Rag23−3 or OVA23−3 mice, this is not necessarily the cases [22]. This is because CD4+ T cells from the mLN of Rag23−3 mice produce high levels of IL-4 in response to OVA that invade into the intestinal tract after consumption of the EW diet, preventing the differentiation of CD4+ T cells into Tregs [30]. Tregs induced in RagD10 mice responded normally against antigen entry in the intestine as indicated by Traxinger, et al. [31]. In contrast, in the spleen, the number of CD4+ T cells is less and the level of IL-4 production is lower than those in mLN [30], which lead to higher induction of Tregs compared with the induction in mLN. In the present study, almost 100% of the induced Tregs in both models and with both diets expressed CD25 (Fig 2A), which is similar to what was observed for Tregs induced from mLN cells by antigen stimulation (Fig 1B).

Fig 2. Antibody (Ab)-mediated stimulation differentiated CD4.

Fig 2

+ T cells into regulatory T cells in casein (CN) diet-fed Rag23−3 like that that did in RagD10 mice. RagD10 and Rag23−3 mice were fed diets containing egg white (EW) or casein (CN) for 7 days. CD4+ T cells (1 × 105) from spleen (SPL) and mesenteric lymph nodes (mLN) were stimulated with plate-bound anti-CD3 and anti-CD28 mAb and cultured under Treg-polarization culture conditions. A) (Top) Frequencies of regulatory T cells (Tregs; Foxp3+) and effector/effector memory T cells (EMT; CD44hiCD62Llo) in the CD4+ T cell populations isolated from the spleen (SPL) and mesenteric lymph nodes (mLN). (Bottom) Frequencies of CD25+ cells in the Treg population and the associated flow cytometry histogram. Each plot indicates the value for an individual well and the horizontal bars indicate the mean values (n= 3, mixture of cells from three mice/group). B) Levels of interleukin (IL)-4, interferon gamma (IFN-γ), IL-2 and IL-10 in the culture supernatants (n= 3, mixture of cells from three mice/group). Data are representative of two independent experiments. Statistical analysis: Tukey’s HSD test [#p < 0.05 (CN vs EW in each strain); *p < 0.05, **p < 0.01, and ***p < 0.001 (between different groups)]. N.D. = not detected.

In both the spleen- and mLN-derived cells, the frequency of EMTs was significantly higher in the EW-fed Rag23−3 mice than in those fed the CN-diet (Fig 2A). The frequency in EW-fed Rag23−3 mice was significantly higher than that in the CN- and EW-fed RagD10 mice in both tissues. In addition, the frequency in CN-fed Rag23−3 mice was significantly lower than those in the CN- and EW-fed RagD10 mice for both tissues, indicating that Ab-mediated signaling inhibited differentiation of naïve CD4+ T cells into EMTs, even in the absence of an antigen.

Large amounts of IL-4 and IFN-γ were detected in the supernatants of the CD4+ T cells isolated from the spleen and mLN of EW-fed Rag23−3 mice, but not in the other three experimental groups, which showed no production (Fig 2B). This indicates the presence of excessive activation of CD4+ T cells in the EW group compared with the other groups IL-2 production was observed in all groups and was comparable for all groups for the cells derived from the spleen. However, IL-2 production was significantly (EW) or tended to be (CN) higher in the cells isolated from the mLN of the Rag23−3 mice than in those from the RagD10 mice. The level of IL-10 production in the spleen cells and in the mLN cells was significantly higher in the Rag23−3 mice than in the RagD10 mice when fed the EW-diet. The amount of IL-10 produced by CD4+ T cells in mLN was much more than that in spleen EW-fed mice. These results suggest that oral administration of EW induced overactivation of intestinal immune responses, as shown by the production of IL-4, IFN-γ, and IL-10 in EW-fed Rag23−3 mice. In addition, Tregs differentiated from CD4+ T cells by stimulation with mAbs in both strains of EW-fed mice probably have sufficient regulatory function producing IL-10, but suppressive function of the Tregs in both strains of CN-fed mice may be independent of IL-10.

The finding that stimulation with OVA did not fully induce Treg differentiation in CN-fed Rag23−3 mice, but stimulation with mAbs did, suggests the presence of one or more factors that prevent Treg induction, such as a molecule expressed by signal transduction in CD4+ T cells via interaction of T-cell receptor with the antigen in the presence of antigen-presenting cells.

Excessive IL-4 and IFN-γ production by CD4+ T cells from EW-fed Rag23−3 mice suppresses Treg differentiation of CD4+ T cells from RagD10 mice

Unlike in EW-fed RagD10 mice, the higher frequency of EMT induction in the EW-fed Rag23−3 mice was observed to have occurred concomitantly with greater cytokine production, suggesting that this higher cytokine production prevented the differentiation of CD4+ T cells into Tregs. To examine this further, we first examined whether the supernatants of CD4+ T cells isolated from the spleen of EW- or CN-fed Rag23−3 or RagD10 mice did indeed have the ability to prevent the differentiation of CD4+ T cells into Tregs. The cytokine profiles of the supernatants used in this experiment are shown in S3 Fig, and these profiles were almost consistent with those shown in Fig 2B. When CD4+ T cells from spleens of untreated RagD10 mice were stimulated with plate-bound anti-CD3 and anti-CD28 Abs and then cultured with the prepared supernatants under Treg-polarization culture conditions, the supernatant from the EW-fed Rag23−3 mice significantly suppressed the induction of Tregs compared with the no-treatment control (Fig 3A). Furthermore, when anti-IL-4 or anti- IFN-γ Abs were added to the supernatant from the EW-fed Rag23−3 mice, the differentiation of Tregs was partially restored (Fig 3B), confirming that IL-4 and IFN-γ production by EMTs inhibits the differentiation of CD4+ T cells into Tregs. The frequency of Tregs in the CD4+ T cell population was higher regardless of the mouse strain when the supernatants from the CN-fed mice were used (Fig 3A), with the supernatant from the CN-fed Rag23−3 mice inducing significantly more Tregs compared with that from CN-fed RagD10, suggesting that the greater amount of IL-2 in the former supernatant contributed to the greater Treg differentiation (S3 Fig), as mentioned in [16]. Neutralization of IL-4 in the supernatant significantly reduced IFN-γ production, whereas neutralization of IFN-γ in the supernatant did not affect the production of IL-4 (Fig 3C). However, while the decrease of IFN-γ concentration in the culture supernatant had no effect on the differentiation of EMTs and their activities compared with no Ab treatment, decreasing the concentration of IL-4 significantly reduced the differentiation of EMTs (Fig 3B), but the levels suppressing the Treg differentiation (Fig 3B) and inhibiting their expression of cytokines receptors (S4B Fig) are similar despite of either neutralized mAbs against the cytokines used. Almost Tregs induced were CD25+ (S4A Fig). These results suggest that in contrast to the equal contribution of these cytokines to impaired Treg induction, only IL-4, not IFN-γ is associated with EMT expansion. Therefore, because IFN-γ does not regulate EMT activities, excessive IL-4 production regulates IL-4 and IFN-γ production by EMT cells and their activities, leading to suppressed Treg differentiation with IL-4 alone or with IFN-γ produced by activated EMT through excessive IL-4 production. The excessive IFN-γ may promote further IL-4 production creating a feedback loop and inhibition of Tregs. Together, these findings indicate that CD4+ T cells of Rag23−3 mice, which are strongly pathogenic, can differentiate into Tregs in vitro if they are incubated with anti-CD3 and anti-CD28 mAbs but not if they are incubated with an allergen (in this case OVA) or in an environment with excessive IL-4 and IFN-γ, like which were produced in the culture supernatant of mLN Tregs shown in Fig 2B (IL-4; 2.5 ng/mL, IFN-γ; 3.5 ng/mL).

Fig 3. Excessive production of interleukin (IL)-4 and interferon-gamma (IFN-.

Fig 3

γ) by CD4+ T cells from EW-fed Rag23−3 mice suppresses Treg differentiation of CD4+ T cells from RagD10 mice. CD4+T cells were prepared from splenocytes of RagD10 and Rag23−3 mice fed a diet containing egg white (EW) or casein (CN) for 7 days. The cells were then stimulated with plate-bound anti-CD3 and anti-CD28 antibodies (mAbs) under Treg-polarization culture conditions for 48 h, and the supernatants (Sup) were collected. A) Treg induction in spleen-derived CD4+ T cells of untreated RagD10 mice cultured under Treg-polarization conditions for 72 h with plate-bound anti-CD3 and anti-CD28 mAbs with or without (-) the indicated supernatants (Sup). B) Regulatory T cell (Treg; Foxp3+) and effector/effector memory T cell (EMT; CD44hiCD62Llo) induction in spleen-derived CD4+ T cells from untreated RagD10 mice. They were cultured without the supernatant of EW-fed Rag23−3 mice (-) or cultured with the Sup with or without anti IL-4 or IFN-γ Abs, under Treg-polarization culture conditions for 72 h. C) IL-4 and IFN-γ concentrations in the culture supernatants obtained in (B). D, E) Frequencies of Treg (Foxp3+) (D) or and EMT (CD44hiCD62Llo) (E) in CD4+ cells prepared from splenocytes derived from RagD10 mice fed a diet containing casein for 7 days and stimulated with plate-bound anti-CD3 and anti-CD28 mAbs supplemented with rIFN-γ and/or rIL-4 under Treg-polarization culture conditions for 72 h. Each plot in A), B), D), and E) represents the value for a single well. Bar (-) indicates the mean values (n= 4, mixture of cells from two to three mice/group). F, G) IL-4 or IFN-γ concentrations in the culture supernatants obtained in (D and ES), respectively. (n = 4, mixture of cells from two to three mice/group). Data are representative of two independent experiments. Means ± SD. Analysis: Unpaired Student’s t test or Dunnett’s test were performed in Fig 3F and 3G respectively. *p < 0.05 [[IFN-γ (0 ng/mL) vs [IFN-γ (3.5 ng/mL)] or [IL-4 (0 ng/mL)] vs [IL-4 (2.5 ng/mL)]] in Fig 3F; *p < 0.05 [[IFN-γ (0 ng/mL) compared with [IFN-γ (1.0 ng/mL)], [IFN-γ (2.0 ng/mL)] and [IFN-γ (3.5 ng/mL)] or [IL-4 (0 ng/mL)] compared with [IL-4 (0.5 ng/mL)], [IL-4 (1.5 ng/mL)]], and [IL-4 (2.5 ng/mL)]] in Fig 3G. Otherwise, Tukey’s HSD test was performed [#p < 0.05 [IFN-γ (0 ng/mL) vs [IFN-γ (3.5 ng/mL)] or [IL-4 (0 ng/mL)] vs [IL-4 (2.5 ng/mL)] and *p < 0.05, ** p < 0.01, and *** p < 0.001 (between different groups)]. N.D., not detected.

Next, we further examined the contributions of the excessive IL-4 and IFN-γ to the promotion of EMT differentiation and inhibition of Treg differentiation. To do this, we added rIL-4 and/or rIFN-γ to Treg-polarization culture of CD4+ T cells derived from the splenocyte population of CN-fed RagD10 mice. The Tregs induced were almost CD25+ independently of the concentration of rIL-4 and/or IFN-γ (S4C Fig). Consistent with the results shown in Fig 3B, rIL-4 and rIFN-γ both inhibited Treg induction in dose dependent manner (Fig 3D). Under excessive IL-4 conditions [Fig 3D-right: 0 and 3.5 ng/mL of rIFN-γ in rIL-4 (2.5 ng/mL)], Treg differentiation was significantly less in the presence of excessive rIFN-γ compared than that in the absence of the protein, clearly indicating that IFN-γ reduces Treg differentiation even under not only without but also with excessive IL-4 condition. For EMT induction, while rIL-4 alone significantly promoted EMT induction, treatment with rIFN-γ alone neither promoted nor suppressed EMT induction (Fig 3E). However, when supplemented with rIL-4 (Fig 3E-left) to excessive IFN-γ condition, the IFN-γ significantly weakened EMT inducive effect of rIL-4 (Fig 3E-left). In addition, under excessive IL-4 conditions (Fig 3E-right), the frequency of EMTs also significantly decreased with rIFN-γ concentration in a dose dependent manner. These results suggest that excessive production of IFN-γ by EMT suppresses IL-4-mediated EMT activation.

Thus, we next assessed the production and receptor expression of these cytokines. The downregulated IL-4 production under condition of excess rIFN-γ (Fig 3F-left, Fig 3G-left) with no change in IL-4R expression (S4D Fig-lower left) suggests that EMT induction, which is caused by excessive IL-4, is impaired not via regulation of sensitivity to IL-4 but by downregulation of IL-4 production. In addition, excessive IL-4 enhance IFN-γR expression compared to no IL-4 (S4D Fig-upper-right), but the difference was not significant. rIL-4 promoted IFN-γ production regardless of rIFN-γ supplementation (Fig 3F-right, Fig 3G-right) in agreement with the hypothesis that excessive IL-4 initiates EMT induction, which then causes IFN-γ overproduction resultant inhibition of Treg differentiation, although IL-4 itself can suppress Treg differentiation (Fig 3D). In addition, excessive rIL-4 itself did not promote (S4D-lower-right) or rather downregulated (S4D Fig-lower-left) IL-4R expression. However, by supplementation with excessive rIFN-γ, IL-4R expression was enhanced, suggesting that in the activation producing excessive IL-4 and IFN-γ production, EMTs raised to increase CD4+ T cell sensitivity to both cytokines most likely not via IFN-γR but rather IL-4R expression, resulting in increased Treg suppression.

Adoptive transfer of Ab-stimulated Tregs from untreated Rag23−3 mice to EW-fed Rag23−3 mice have sufficient regulatory activity to prevent allergic inflammation

We proved in Fig 2 that naïve CD4+ T cells had similar ability to differentiate into Tregs in Rag23−3 mice compared with those in RagD10 mice. Therefore, we next investigated whether the Tregs induced from Rag23−3 CD4+ T cells possess sufficient regulatory activity to inhibit intestinal allergic responses. Spleen-derived CD4+ T cells from untreated Rag23−3 mice were cultured under control conditions or under Treg-polarization conditions. Cells from each culture were collected and intravenously injected into untreated Rag23−3 mice. Flow cytometry analysis of the cultured cells before the adoptive cell transfer showed that 91.2% of the Tregs were Foxp3+, whereas only 4.36% of the control cells were (Fig 4A). From one day after administration of either the control cells, Tregs, or PBS to untreated Rag23−3 mice, the mice were fed the EW diet for 8 days. The mice that received the control treatments (administered control cells or PBS; Control or PBS) showed a significantly lower body weight from day 5 compared with the mice that received the Tregs (Fig 4B). Histological analysis of the jejunum showed a normal appearance in the Treg group, but morphological changes (i.e., crypt elongation, goblet cell hyperplasia, and cell infiltration into the villus) throughout the tissue in the control groups (Fig 4C). The body weight and histological data indicate that the transfer of the Tregs inhibited the induction of food-allergic enteropathy. On day 8 of EW feeding, spleen- and mLN-derived CD4+ T cells were isolated from the mice that received the adoptive transfer and stimulated with OVA. The proliferation of the CD4+ T cells from the two tissues was significantly inhibited in the Treg group compared with that in the control groups (Fig 4D). In addition, the excessive production of IL-4 and IFN-γ observed in the control groups was significantly reduced in the Treg group (Fig 4E). Together, these results indicate that adoptive transfer of the Tregs suppressed excessive activation of CD4+ T cells in the recipient EW-fed Rag23−3 mice, preventing the mice from developing food-allergic enteropathy. Thus, we conclude that Tregs derived from untreated Rag23−3 mice retain sufficient regulatory activity to suppress intestinal allergic responses.

Fig 4. Adoptive transfer of antibody (Ab)-stimulated Tregs from untreated Rag23−3 mice to EW-fed Rag23−3 mice have sufficient regulatory activity to prevent allergic inflammation.

Fig 4

A) Regulatory T cell (Treg; Foxp3+) population within the transferred CD4+ T cell population. Transferred CD4+ T cells were derived from spleen of untreated Rag23−3 mice cultured for 72 h under control conditions (Control; stimulated with plate-bound anti-CD3 and anti-CD28 mAbs and then incubated with recombinant IL-2 [rIL-2]) or under Treg-polarization conditions (Treg). After collection, the cells were intravenously injected into untreated Rag23−3 mice (1 × 107 cells/mouse). As a control without cell transfer, Rag23−3 mice received phosphate-buffered saline (PBS). B) Time course of body-weight changes relative to initial values (100%). Weight changes were compared between the Treg and control groups each day. C) Hematoxylin and eosin–stained jejunum. Scale bars, 100 µm. D) [H3]-thymidine uptake (in counts per minute; CPM) of spleen (SPL)- or mesenteric lymph node (mLN)-derived CD4+ T cells isolated from the indicated treatment groups after egg white feeding and stimulated with OVA and antigen-presenting cells. E) Concentrations of interleukin (IL)-4 and interferon gamma in the supernatant of SPL or mLN-derived CD4+ T cells isolated from the indicated treatment groups after egg white feeding and incubation for 48 h. Data are representative of two or three independent experiments. Statistical analysis: Tukey’s HSD test [*p < 0.05 (CN vs. each of the two groups) in Fig 4B; *p < 0.05, **p < 0.01, and ***p < 0.001 (between different groups) in Fig 4D and 4E].

CD44loCD62LhiCD4+ T cells (Naïve-like) in the mLN, but not in the spleen, of EW-fed Rag23−3 mice have the potential to differentiate into Tregs

We have shown that CD4+ T cells of CN-fed Rag23−3 mice have the potential to differentiate into Tregs (Fig 2) when they are stimulated and expanded with anti-CD3 and anti-CD28 mAbs, but it was unclear whether the naïve CD4+ T cells in EW-fed Rag23−3 mice with enteropathy also keep their ability to differentiate into Tregs. Therefore, we next examined if the naive CD4+ T cells purified from the spleens or mLNs cells isolated from EW-fed Rag23−3 mice can differentiate into Tregs when they were stimulated with anti-CD3 and anti-CD28 mAbs. To purify the cells, we sorted naïve cells (naïve; CD44loCD62LhiCD4+) or EMTs (effector/effector memory; CD44hiCD62LloCD4+) from the mLN and spleen cells (SPL) of Rag23−3 mice fed with the EW- or CN-diet for 7 days (Fig 5A). The cells were then stimulated with plate-bound anti-CD3 and anti-CD28 mAbs under Treg-polarization conditions. After incubation for 72 h, the Treg population within the CD4+ T cell population was examined by flow cytometry (Fig 5B and 5C). The frequency of Tregs within the CD4+ T cell population in the culture of mLN-derived naïve T cells from the EW-fed Rag23−3 mice (EW_naive) was significantly lower than that in the cells from the culture of mLN-derived naïve T cells from CN-fed mice (CN_naive), but was significantly higher than that under the Treg-polarization culture of EMTs from EW-fed mice (EW_EMT) (Fig 5C). In the spleen cells (SPL), the frequency of Tregs was comparable in the Treg-polarization cultures of naïve phenotype CD4+ T cells (EW_naive) and EMTs (EW_EMT groups), but these were statistically lower than that in the culture of naïve T cells from CN-fed mice (Fig 5C). In addition, the production of the IL-4 was not enhanced in the naïve CD4+ T cells of the EW-fed mice (EW_naive) and was mostly comparable to that in the culture of naïve T cells of CN-fed mice (CN_naive); in contrast, overproduction of IL-4 in addition to IFN-γ was observed in the Treg-polarization culture of EMTs from EW-fed mice (EW_EMT). IL-2 production in mLN was comparable among experimental groups, but that in EW_EMT in SPL showed the highest level compared to those in other groups, but the difference of IL-2 production was not affected to the induction of Tregs (Fig 5D). These results indicate that naïve CD4+ T cells in the spleen and mLNs of Rag23−3 mice with severe allergic enteritis had a significantly reduced potential to differentiate into Tregs compared with those in mice fed the CN-diet fed the mice; this decrease of potential might be the result of changes of surface molecules expression of the cells affected by the allergic environment in vivo but not by the cytokines. This is consistent with the absence of detectable IL-4 or IFN-γ in the culture supernatant of Tregs differentiated from naive CD4+ T cells under polarization conditions, in contrast to the supernatant of Tregs differentiated from EMTs. Thus, this naïve CD4+ T cells might be one of several lineages of naïve T cells, not a typical naïve. Therefore, we defined the naïve CD4+ T cells defines as naïve-like T cells, hereafter.

Fig 5. Naïve-like CD4.

Fig 5

+ T cells (CD44loCD62Lhi) from mesenteric lymph nodes (mLN) of EW-fed Rag23−3 mice differentiate into regulatory T cells (Treg) even under severe allergic conditions. A) Gating strategy to sort naïve (CD44loCD62Lhi) and effector/effector memory (EMT; CD44hiCD62Llo) CD4+ T cells. Naïve CD4+ T cells were prepared from Rag23−3 mice fed a diet containing casein (CN) or egg-white (EW), whereas EMTs were prepared only from Rag23−3 mice fed the EW-diet. B, C) Flow cytometry analysis of Treg population (Foxp3+) in CD4+ T cells differentiated from the naïve CD4+ T cell (naïve) or EMT populations stimulated with plate-bound anti-CD3 and anti-CD28 antibodies and cultured under Treg polarization conditions for 72 h. Each plot indicates the value for an individual well, and the horizontal lines indicate the mean values (CN_naive: n = 4, EW_naive: n = 4, EW_EMT: n = 4 using a mixture of cells from 6 [CN] or 3 [EW] mice/group). D), Interleukin (IL)-4 IFN-gamma (IFN-γ), and IL-2 levels in the culture supernatants (CN_naive: n = 4, EW_naive: n = 4, EW_EMT: n = 4 using a mixture of cells from 6 [CN] or 3 [EW] mice/group). Data are representative of two independent experiments. Statistical analysis: Tukey’s HSD test [*p < 0.05, **p < 0.01, and ***p < 0.001 (between different groups)]. N.D., not detected.

In previous studies on Treg stability during tolerance acquisition in individuals with food allergy, CD137 expression by Tregs has been suggested to be a marker of enhanced Foxp3 expression and stability [1416]. To examine the relationship between CD137 and Treg differentiation from naïve-like CD4+ T cells (EW_naive) and EMT phenotype (EW_EMT) from EW-fed mice, we examined the CD137 population within Tregs and CD44hiCD62LloTreg populations differentiated from naïve-like T cells from CN- or EW-fed Rag23−3 mice (CN_naive, EW_naive) or EMTs of EW-fed Rag23−3 mice (EW_EMT) (S5A and S5B Fig). The frequency of CD137+ cells within Tregs or CD44hiCD62LloTregs was significantly higher in the EW_naive and EW-EMT groups than those in the CN_naive group for both mLN and SPL, with almost 100% of the cells in the EW_EMT group being CD137+ cells (S5B Fig). In addition, the CD137+ population increased with increasing frequency of CD44hiCD62Llo within the Tregs population (S5C Fig), indicating that as the population of activated Tregs increased, so did the CD137+ population. However, the contribution of CD137 to the stability of the CD137+ population remains to be elucidated.

Discussion

In this study, we examined whether allergen-specific T cells from food-allergic model mice showing weight loss and enteropathy are able to differentiate into functional Tregs with optimal activation and a stable suppressive function in vitro. To investigate this aim, we used two strains of OVA-specific T-cell-receptor transgenic mice: Rag23−3 mice (OVA23−3 mice crossed with Rag2 knockout mice) and RagD10 mice (DO11.10 mice crossed with Rag2 knockout mice). When fed a diet containing EW, RagD10 mice acquire tolerance to OVA, whereas Rag23−3 mice manifest severe Th2 responses and food-allergic enteropathy. Overall, our findings suggest that to obtain allergen-specific Tregs with sufficient activity to suppress food-allergic inflammation, naïve (CD44loCD62hi) CD4+ T cells must be from the total lymphocyte population, be expanded by stimulation with anti-CD3 and anti-CD28 mAbs but not OVA plus antigen-presenting cells, and then incubated under a cytokine milieu with low concentrations of IL-4 and IFN-γ. Furthermore, our results indicate that different from mLN, naïve T cells from the spleen of EW-fed Rag23−3 mice manifesting severe allergic responses, but not those from CN-fed Rag23−3 mice, had lost their ability to differentiate into Tregs before they were subjected to Treg expansion in vitro. This indicates not only that the ability of naïve CD4+ T cells to differentiate into Tregs is dependent on the conditions in the tissues in which they were localized in vivo, but also that the intestinal immune system maintains conditions suitable for Treg expansion even under conditions of severe allergic inflammation [30].

The only lymphocytes that Rag23−3 and RagD10 mice produce in response to OVA challenge are OVA-specific T cells. Using these mouse models allowed us to compare the functions of the OVA-specific Tregs between the inflammatory and tolerant conditions in response to exposure to the same volume of OVA over the same period of time. This comparison revealed that signaling via OVA-specific T-cell receptors plays a critical role in controlling Treg differentiation and inducing immune responses under conditions that cause T cells to respond excessively to allergens. More specifically, we have found that there are some differences in how the immune responses are invoked between the Rag23−3 and RagD10 mice. One difference, reported elsewhere, is that the two strains possess different T-cell-receptor genes [26]. Another difference involves the signals that induce IL-4 production by CD4+ T cells when the cells are stimulated with OVA through T-cell receptor. The signals to produce IL-4 are likely stronger and more immediate in OVA-specific EMTs from EW-fed Rag23−3 mice than in those from RagD10 mice, especially in the mLNs. Indeed, as we have described in a previous report [22], although CD4+ T cells from the spleens of EW-fed RagD10 mice were able to produce high levels of IL-4 and show a Th2-type immune response, it was increased only in the first 3 days of EW-feeding and was decreased thereafter out to 7 days; furthermore, CD4+ T cells from these mice stimulated with OVA needed 72 h to release high levels of IL-4 into the culture supernatant [22]. We have also reported that the Treg function induced during that experimental period is stable, as evidenced by the mice not showing severe allergic inflammation after restarting the EW diet after being on the CN diet for 1 month [22]. In contrast, high IL-4 production in mLN cells was immediately induced and maintained for 7–9 days in EW-fed R23-3 mice, and the incubation time for CD4+ T cells stimulated with OVA to produce a large amount of IL-4 was less than 48 h [22]. Inhibitory function of Tregs was not stable as indicated in the recurrence of the intestinal inflammation. Excessive IL-4 responses in the mLNs of EW-fed Rag23−3 mice dramatically decreased Treg induction and caused intestinal inflammation [22]. However, the mLNs are suggested to not only be a major inflammatory tissue in food allergy but also an important regulatory tissue in food allergy [22,31]; therefore, their potential to induce Tregs for regulation of intestinal inflammation may be higher than that of other tissues.

Although in the present study we found that T cells from EW-fed Rag23−3 mice have lower ability to differentiate into Tregs than T cells from RagD10 mice, we also found that using a combination of anti-CD3 and anti-CD28 mAbs to obtain sufficient volume of Tregs for adoptive transfer to other mice was superior to using a combination of antigen (OVA) and antigen-presenting cells. Although we did not investigate in detail why the responses to the Ab-stimulation were different from those to the antigen stimulation, similar findings, albeit with a different capacity of Tregs, have been reported elsewhere: Zhao et al. reported that the inhibitory effects on Th1 responses, surface molecules, and levels of Foxp3 expression in hen-egg-lysozyme–specific Tregs induced by mAbs are not the same as those induced by a combination of antigen plus antigen-presenting cells [24]. The implication of this result is that when considering adoptive Treg transfer as a therapeutic option, we need to choose the most appropriate approach to obtain Tregs with sufficient suppressive function. Our present results suggest that using mAbs to stimulate Treg differentiation affords Tregs with superior suppressive function in the context of severe food allergy. In addition, if we were to use the combination of antigen and antigen-presenting cells, we would also have to deal with removing the antigen and antigen-presenting cells from the culture at the end of the process to increase the purity of Tregs and avoid any potentially harmful effects of leftover antigen. Thus, the present findings highlight the potential of using Tregs differentiated from intestinal naïve T cells using a combination of anti-CD3 and anti-CD28 mAbs to suppress severe food-allergic inflammation.

Further studies are needed to elucidate the mechanism underlying the induction of suppressive Tregs by using mAbs. In the present study, induction of Tregs using the Abs-combination produced highly stable and highly suppressive Tregs, even from splenic CD4+ T cells from untreated Rag23–3 mice, suggesting that activating naive T cells from untreated mice via both CD3 and CD28 mAbs can produce Tregs (iTregs) having persistent suppressive abilities in food allergy. However, in lung allergy, it has been reported that iTregs appear to enhance allergic responses [32]. Other reports have indicated that iTregs are stable, when they are expanded without the CD28 costimulatory signal [33]. Stimulation by anti-CD3 suppresses activation of EMTs induced by stimulation with co-stimulatory molecules such as anti-CD28 and Treg induction is promoted under Treg-polarization conditions [34]. Indeed, administration of anti-CD3 mAbs is already used clinically as a method to attenuate EMT activities for the treatment of some autoimmune diseases such as type 1 diabetes and multiple sclerosis [3537]. Therefore, we hypothesize that if our iTregs are expanded via anti-CD3 stimulation only, they may show an increased suppressive capability.

The present results are noteworthy in that they show that, in EW-fed Rag23−3 mice with severe allergic inflammation, mLN had higher capacity to induce Tregs than spleen, and the frequency of Foxp3+ Tregs in the CD4+ T cell population induced from naïve-like (CD44loCD62hi) CD4+ T cells from mLN was higher than that from EMT (CD44hiCD62lo) CD4+ T cells when the cells were cultured under Treg-polarization conditions and stimulated with anti-CD3 and anti-CD28 mAbs (Fig 5). This understanding will be useful for developing techniques to investigate the differences of Treg differentiation among different tissues in individuals with food allergy. However, the frequency of Tregs within the CD4+ T cell population was significantly lower in the Tregs induced from naïve-like (CD44loCD62hi) CD4+ T cells from EW-fed Rag23−3 mice than in those from CN-fed the mice, and the Tregs from both naïve-like CD4+ T cells from both types of mouse did not produce any cytokines, indicating that prior exposure of naïve CD4+ T cells to excessive Th2-cytokine circumstances (i.e., severe allergy) may change the characteristics of the cells to naïve-like CD4+ T cells, resulting in inhibition of Treg expansion in EW-fed mice compared with that in CN-fed mice. In previous studies examining Treg stability under allergic conditions [1618], CD137 expression in Tregs has been suggested to be a marker of enhanced Foxp3 expression. In our mouse model of food-allergy enteropathy, although CD137 expression indicates activated Tregs, it may not necessarily reflect the correlation between the enhanced Foxp3 expression and stability of Treg function. Our data show the frequency of CD137+ cells in CD44hiCD62LloFoxp3+CD4+ T cells (activated Tregs) was enhanced by the EW diet, with 100% of the Tregs with the EMT phenotype being differentiated from EMTs from EW-fed Rag23−3 mice which expressing CD137 molecule (S5 Fig). This result reminds us of previous studies that have shown that activated Tregs have a higher plasticity and can change into EMTs [11,1618]. Thus, our results suggest that naïve-like (CD44loCD62hi) CD4+ T cells expressing a surface marker to migrate to the intestinal tissues (e.g., the gut homing integrin, α4β7 [38] or C-C motif chemokine receptor 9 (CCR9), specific homing receptors for colon or small intestine [39]) may be better for producing stable Tregs. In addition, ROR-γt Tregs also may be one of the functional population [20,40], but the characteristics of the Tregs from individuals with severe food allergies should be carefully investigated.

Regarding cooperative inhibition of Treg-differentiation by IL-4 and IFN-γ,it has been previously reported that IL-4 inhibits Treg induction [27], but the role of IFN-γ, which is produced by Th1 cells, is yet to be clarified. Although several reports have indicated that IFN-γ suppresses the induction of Tregs [4143], IFN-γ is also reported to be required for the generation of Tregs in diseases, such as graft-versus-host disease or experimental autoimmune encephalomyelitis [44,45]. Although Th1 cells suppress the induction of Th2 cells, transplanting excess Th1 cells may not be an effective treatment method because of their ability to inhibit Treg differentiation (Fig 3). Because our results show that CD4+ T cells from a mouse model of allergy could still differentiate into Tregs under appropriate conditions of limited IL-4 and IFN-γ production, we consider that regulation of the cytokines may be useful for designing treatments using iTregs.

To clarify the impacts of the two cytokines, we performed the experiments adding rIL-4 and/or rIFN-γ or anti-IL-4 or IFN-γ mAbs into the Treg polarization culture; IFN-γ and IL-4 concentrations of the culture, the contribution of these cytokines to the inhibitory function of Treg differentiation and induction of EMTs were evaluated. We found that IFN-γ, IL-4, and EMTs worked together to create a feedback loop that works to balance the effects of the two cytokines; on the start of feeding Rag23−3 mice with EW, a greater amount of IL-4 might be strongly produced by EMT [22], excessive IL-4 production upregulated sensitivity to IFN-γ while also initiating EMT expansion, which results in IFN-γ overproduction of the EMT, which in turn promotes IL-4R expression, additional IL-4 overproduction, and further EMT expansion. Our results clearly indicated that during these inflammatory loops, Treg differentiation is strongly inhibited. Therefore, by the neutralization with each antibody in Fig 3B, the concentration of either cytokine was decreased, resulting that EMT activation was subsided, leading to recovery of Treg differentiation. In RagD10 mice, excessive IL-4 production can be induced, but it is produced more slowly and only for a short time during the early period of EW-feeding compared with what occurs in Rag23−3 mice [22] thereby preventing its inhibitory effects on Treg differentiation.

We also examined the expression of IL-4R and IFN-γR in Tregs differentiated from CD4+ T cells in RagD10 mice. We did not find any double-positive Tregs in the culture, but that might be the result of the low sensitivity of flow cytometry for small number of double-positive cells. Thus, these findings did not clarify whether the cytokines were produced by separate cells or there were cells expressing both cytokines. Adding anti IL-4 or IFN-γ Ab to the supernatant reduced the expression of both receptors compared with the situation without either of the Abs, and the frequency of Tregs in both groups was enhanced. That is, decreased cytokine production inhibited the receptor expression, showing that decreasing cytokine-mediated signaling improved the differentiation of Tregs. By adding of either IL-4 or IFN-γ or both, we clarified that CD4+ T cells from RagD10 mice are more sensitive to the IL-4–mediated than to the IFN-γ–mediated signal not to differentiate into Tregs.

It has been reported that excessive IL-4 production by naïve CD4+ T cells from the spleen of BALB/c mice induces IL-4R expression and phosphorylation of STAT6 and STAT3; the IL-4R expression is supplied by STAT3, and STAT3 suppresses IFN-γR1 through STAT6 [45] indicating that the receptor-mediated inter-regulatory relationship between the two transcription factors is provoked by excessive IL-4 and contribute to create the excessive IL-4 loop. Furthermore, IL-4 stimulation has been shown to downregulate CD28 and IFN-γR expression in CD3+CD4+CD8IL-4R+ T cells [46]. Although it is unclear whether the relationship can be established in our Tregs differentiation, the addition of anti-IL-4 Ab to the culture may increase CD28 expression, increase the inhibitory effect of anti-CD3 Ab that suppresses T cell activation, and increase Treg differentiation [35]. Although we cannot discuss the IFN-γ-mediated mechanism underlying Treg expansion and their expression of IL-4R, there is possibly an inter-relationship of transcription factor–induced IFN-γ overproduction like excess production of IL-4 involved in the differentiation of our Tregs.

Adoptive transfer of Tregs for the treatment of severe inflammatory diseases is an attractive approach because it has the potential to not only treat but also prevent severe inflammatory diseases including allergy [3], although there have not yet been any clinical trials of Treg transfer therapy. There are, however, currently three approaches that have been reported to obtain stable Tregs from peripheral blood mononuclear cells: naturally occurring regulatory T cells (nTregs) [47], chimeric antigen receptor regulatory T cells (CAR-Tregs) [48], and stable and functional induced regulatory T cells (S/F-iTregs) [49]. It has been clarified in individuals who have outgrown their food allergy naturally or by OIT that Tregs and mediation of their function through inhibitory cytokines play important roles [3]. Therefore, Treg transfer may be a suitable approach for the treatment of individuals with persistent food allergy.

Allergen-specific Tregs are considered a good target not only for monitoring of the immunological condition of individuals with severe food allergy but also as a platform for the development of safe and stable treatments for these patients. The present results suggest several areas where further research is needed before Treg transfer can be used in the clinic. However, although we need to analyze the effect of stability of Treg function using the BALB/c mouse model [50] and verify the generality of this method, if we improve our understanding of T cell phenotypes and are able to find a specific marker of the cells related to the intestinal immune system, using antibodies rather than allergens and a low cytokine milieu for Treg expansion may provide a safe and efficient means of treating inflammatory diseases.

Conclusions

Under severe allergic circumstances, induction of allergen-specific Tregs plays an important role in the inhibition of allergic inflammation, but there are many challenges to overcome before we will be able to obtain stable, suppressive Tregs for clinical use. Naïve (CD44lowCD62Lhi) CD4+ T cells attributed to the intestinal immune system are candidate cells for expansion into stable, suppressive Tregs, but only when they are stimulated with anti-CD3 and anti-CD28 mAbs and rather than allergens and expanded under Treg polarization culture conditions with a low cytokine milieu.

Supporting information

S1 Fig. Body weight changes and jejunum histology in RagD10 and Rag23−3 mice fed a diet containing egg white (EW) or casein (CN).

A) RagD10 and Rag23−3 mice were fed a diet containing EW or CN for 7 days, and body weights were measured on days 0, 2, 4, 5, and 7. Body weight changes were determined relative to those on day 0. Values are expressed as mean ± SD. B) Jejunum histology. Samples of jejunum were collected from the mice on day 7 and stained with hematoxylin and eosin. Scale bars, 100 µm. n = 3 per group. Data are representative of two independent experiments. Statistical analysis: Tukey’s HSD test [*p < 0.05 (Rag23−3 EW vs each of the three groups)].

(TIFF)

pone.0324105.s001.tiff (1.8MB, tiff)
S2 Fig. Sensitivity of CD4+ T cells from RagD10 and Rag23−3 mice to stimulation by transforming growth factor beta 1 (TGF-β1).

Spleen (SPL) and mesenteric lymph nodes (mLN) were harvested from untreated RagD10 or Rag23−3 mice, and CD4+ T cells were isolated by magnetic cell separation system. The cells were then stimulated with plate-bound anti-CD3 and anti-CD28 monoclonal antibodies and cultured in the presence of the indicated amounts of TGF-β1, retinoic acid (1 µM), and recombinant IL-2 (2 ng/mL) for 48 h, and the frequency of regulatory T cells (Foxp3+) within the CD4+ T cell population was determined. Each circle indicates the value for an individual well, and the horizontal lines indicate mean values (n = 3, mixture of cells from three mice/group). Data are representative of two independent experiments. Statistical analysis: Tukey’s HSD test [(#p < 0.05 (between different strains); * p < 0.05, ** p < 0.01, and *** p < 0.001 (between different groups in each strain)].

(TIFF)

pone.0324105.s002.tiff (341.2KB, tiff)
S3 Fig. Cytokine profiles for the supernatants of CD4+ T cells isolated from the spleen of RagD10 mice and Rag23−3 mice and cultured under regulatory T cell (Treg) polarization conditions.

RagD10 and Rag23−3 mice were fed a diet containing egg white (EW) or casein (CN; control) for 7 days and spleens were harvested; CD4+ T cells were isolated, stimulated with plate-bound anti-CD3 and anti-CD28 monoclonal antibodies, and cultured under Treg polarization conditions for 48 h, and the culture supernatants were collected. The concentrations of interleukin (IL)-2, IL-4, and interferon gamma (IFN-γ) in the supernatants were determined by enzyme-linked immunosorbent assay (n = 3, mixture of cells from three mice/group). Data are representative of two independent experiments. Statistical analysis: Tukey’s HSD test [#p < 0.05 (CN vs EW in each strain); * p < 0.05, ** p < 0.01, and *** p < 0.001 (between different groups)]. N.D. = not detected.

(TIFF)

pone.0324105.s003.tiff (103.9KB, tiff)
S4 Fig. Expression of interferon-gamma (IFN-γ) or interleukin (IL)-4 receptor on regulatory T cells differentiated from untreated RagD10 mice is regulated by excessive IFN-γ or IL-4.

A) Frequency of CD25+ cells in regulatory T cells (Tregs; Foxp3+CD4+ T cells) differentiated from splenocytes of untreated-RagD10 mice cultured under Treg-polarization culture conditions described in the caption to Fig 3B. B) Gating strategy for identifying IFN-γ receptor (IFN-γR+) or IL-4 receptor (IL-4R+) on CD4+ T cells (left) and the frequencies of each receptor expression cells in CD4+ T cells (right). Each plot indicates the value for an individual well and horizontal lines indicate mean values (n = 3, mixture of cells from three mice/group). C) Frequency of CD25+ cells in Tregs from spleen cells of untreated RagD10 mice cultured under the Treg-polarization culture conditions indicated in Fig 3D and 3E. D) Frequencies of IFN-γR+ or IL-4R+ on CD4+ cells supplemented with rIL-4 (left: 0, 0.5, 1.5, 2.5 ng/mL) or rIFN-γ (right: 0, 1.0, 2.0, 3.5 ng/mL). Error bars indicate means ± SD (n= 4, mixture of cells from two to three mice/group). Analysis: Tukey’s HSD test {#p < 0.05 [IFN-γ (0 ng/mL)] vs [IFN-γ (3.5 ng/mL)] or [IL-4 (0 ng/mL)] vs [IL-4 (2.5 ng/mL)] in Fig S4C and S4D; * p < 0.05, ** p < 0.01, and *** p < 0.001 (between different groups)}.

(TIFF)

pone.0324105.s004.tiff (1.1MB, tiff)
S5 Fig. Effector/effector memory T cells within the regulatory T cell population show enhanced CD137 expression.

Naïve-like CD4+ T cells (CD44loCD62Lhi) and effector/effector memory CD4+ T cells (EMT; CD44hiCD62Llo) from Rag23−3 mice fed a diet containing egg white (EW) or casein (CN; control) for 7 days were stimulated with plate-bound anti-CD3 and anti-CD28 monoclonal antibodies and cultured under Treg-polarization culture conditions for 72 h. A) Gating strategy used to identify the CD137+ subpopulation within the Foxp3+ CD4+ and CD44hiCD62LloFoxp3+CD4+ populations. B) Frequency of CD137+ cells within the Foxp3+CD4+ and CD44hiCD62LloFoxp3+CD4+ T cell populations. C) Frequency of CD44hiCD62Llo cells within the Foxp3+CD4+ T cell population. In panels 5B and 5C, plots indicate the values for individual wells and horizontal lines indicate mean values (CN_naive: n = 4, EW_naive: n = 4, EW_EMT: n = 4, using a mixture of cells from 6 (CN) or 3 (EW) mice/group). Data are representative of two independent experiments. Statistical analysis: Tukey’s HSD test [*p < 0.05, ** p < 0.01, and *** p < 0.001 (between different groups)].

(TIFF)

pone.0324105.s005.tiff (1.5MB, tiff)

Data Availability

All Excel files containing the data underlying the results presented in our study (Fig 1~Fig 5 and S1 Fig ~ S5 Fig) are available at the URL address of UTokyo Repository (http://hdl.handle.net/2261/0002013444).

Funding Statement

This work was supported by grants from the Kieikai Research Foundation (HNA, Grant number; 2017S063, https://www.nakashima-foundation.org/kieikai/) and Grant-in-Aid for Scientific Research (B) from the Japan Society for the Promotion of Science (JSPS)(SH, Grant number; 26292065, https://www.jsps.go.jp/j-grantsinaid/index.html). The analysis for Fig. 5 and S5 Fig was funded by Meiji Holdings Co., Ltd (HNA and SH). Meiji Holdings Co., Ltd., we have no grant number assigned to this program grant in accordance with the nature of the grant, because The University of Tokyo's Corporate Sponsored Research Programs are programs established to conduct research on common issues that are of highly public nature in collaboration with the University of Tokyo, using funds received from the private sector and other external organizations. [Corporate Sponsored Research Programs | The University of Tokyo (u-tokyo.ac.jp)].The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

References

  • 1.Peters RL, Krawiec M, Koplin JJ, Santos AF. Update on food allergy. Pediatr Allergy Immunol. 2021;32(4):647–57. doi: 10.1111/pai.13443 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 2. Sicherer SH, Sampson HA. Food allergy: a review and update on epidemiology, pathogenesis, diagnosis, prevention, and management. J. Allergy Clin Immunol. 2018;141:41–58. 10.1016/j.jaci.2017.11.003 [DOI] [PubMed] [Google Scholar]
  • 3.Kazmi W, Berin MC. Oral tolerance and oral immunotherapy for food allergy: evidence for common mechanisms?. Cell Immunol. 2023;383:104650. doi: 10.1016/j.cellimm.2022.104650 [DOI] [PubMed] [Google Scholar]
  • 4.Hussey Freeland DM, Fan-Minogue H, Spergel JM, Chatila TA, Nadeau KC. Advances in food allergy oral immunotherapy: toward tolerance. Curr Opin Immunol. 2016;42:119–23. doi: 10.1016/j.coi.2016.08.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 5.Kim EH, Burks AW. Food allergy immunotherapy: Oral immunotherapy and epicutaneous immunotherapy. Allergy. 2020;75(6):1337–46. doi: 10.1111/all.14220 [DOI] [PubMed] [Google Scholar]
  • 6.Nowak-Wegrzyn A, Sato S, Fiocchi A, Ebisawa M. Oral and sublingual immunotherapy for food allergy. Curr Opin Allergy Clin Immunol. 2019;19(6):606–13. doi: 10.1097/ACI.0000000000000587 [DOI] [PubMed] [Google Scholar]
  • 7.Sabouraud M, Biermé P, Andre-Gomez S-A, Villard-Truc F, Corréard A-K, Garnier L, et al. Oral immunotherapy in food allergies: a practical update for pediatricians. Arch Pediatr. 2021;28(4):319–24. doi: 10.1016/j.arcped.2021.03.006 [DOI] [PubMed] [Google Scholar]
  • 8.Bertolini TB, Biswas M, Terhorst C, Daniell H, Herzog RW, Piñeros AR. Role of orally induced regulatory T cells in immunotherapy and tolerance. Cell Immunol. 2021;359:104251. doi: 10.1016/j.cellimm.2020.104251 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9.Jones SM, Burks AW, Dupont C. State of the art on food allergen immunotherapy: oral, sublingual, and epicutaneous. J Allergy Clin Immunol. 2014;133(2):318–23. doi: 10.1016/j.jaci.2013.12.1040 [DOI] [PubMed] [Google Scholar]
  • 10.Kim KS, Hong S-W, Han D, Yi J, Jung J, Yang B-G, et al. Dietary antigens limit mucosal immunity by inducing regulatory T cells in the small intestine. Science. 2016;351(6275):858–63. doi: 10.1126/science.aac5560 [DOI] [PubMed] [Google Scholar]
  • 11.Noval Rivas M, Burton OT, Wise P, Charbonnier L-M, Georgiev P, Oettgen HC, et al. Regulatory T cell reprogramming toward a Th2-cell-like lineage impairs oral tolerance and promotes food allergy. Immunity. 2015;42(3):512–23. doi: 10.1016/j.immuni.2015.02.004 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Syed A, Garcia MA, Lyu S-C, Bucayu R, Kohli A, Ishida S, et al. Peanut oral immunotherapy results in increased antigen-induced regulatory T-cell function and hypomethylation of forkhead box protein 3 (FOXP3). J Allergy Clin Immunol. 2014;133(2):500–10. doi: 10.1016/j.jaci.2013.12.1037 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 13.Karlsson MR, Rugtveit J, Brandtzaeg P. Allergen-responsive CD4+ CD25+ regulatory T cells in children who have outgrown cow’s milk allergy. J Exp Med. 2004;199(12):1679–88. doi: 10.1084/jem.20032121 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14.Mazzarella G. Effector and suppressor T cells in celiac disease. World J Gastroenterol. 2015;21(24):7349–56. doi: 10.3748/wjg.v21.i24.7349 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 15.Eggenhuizen PJ, Ng BH, Ooi JD. Treg enhancing therapies to treat autoimmune diseases. Int J Mol Sci. 2020;21(19):7015. doi: 10.3390/ijms21197015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Lozano-Ojalvo D, Tyler SR, Aranda CJ, Wang J, Sicherer S, Sampson HA, et al. Allergen recognition by specific effector Th2 cells enables IL-2-dependent activation of regulatory T-cell responses in humans. Allergy. 2023;78(3):697–713. doi: 10.1111/all.15512 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17.Zhang Y, Li L, Genest G, Zhao W, Ke D, Bartolucci S, et al. Successful milk oral immunotherapy promotes generation of casein-specific CD137+ FOXP3+ regulatory T cells detectable in peripheral blood. Front Immunol. 2021;12:705615. doi: 10.3389/fimmu.2021.705615 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 18.Weissler KA, Rasooly M, DiMaggio T, Bolan H, Cantave D, Martino D, et al. Identification and analysis of peanut-specific effector T and regulatory T cells in children allergic and tolerant to peanut. J Allergy Clin Immunol. 2018;141(5):1699-1710.e7. doi: 10.1016/j.jaci.2018.01.035 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19.Fukaya T, Takagi H, Sato Y, Sato K, Eizumi K, Taya H, et al. Crucial roles of B7-H1 and B7-DC expressed on mesenteric lymph node dendritic cells in the generation of antigen-specific CD4+Foxp3+ regulatory T cells in the establishment of oral tolerance. Blood. 2010;116(13):2266–76. doi: 10.1182/blood-2009-10-250472 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 20.Kedmi R, Najar TA, Mesa KR, Grayson A, Kroehling L, Hao Y, et al. A RORγt+ cell instructs gut microbiota-specific Treg cell differentiation. Nature. 2022;610(7933):737–43. doi: 10.1038/s41586-022-05089-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21.Jacobse J, Li J, Rings EHHM, Samsom JN, Goettel JA. Intestinal regulatory T cells as specialized tissue-restricted immune cells in intestinal immune homeostasis and disease. Front Immunol. 2021;12:716499. doi: 10.3389/fimmu.2021.716499 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 22.Nakajima-Adachi H, Shibahara K, Fujimura Y, Takeyama J, Hiraide E, Kikuchi A, et al. Critical role of intestinal interleukin-4 modulating regulatory T cells for desensitization, tolerance, and inflammation of food allergy. PLoS One. 2017;12(2):e0172795. doi: 10.1371/journal.pone.0172795 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Nakajima-Adachi H, Ebihara A, Kikuchi A, Ishida T, Sasaki K, Hirano K, et al. Food antigen causes TH2-dependent enteropathy followed by tissue repair in T-cell receptor transgenic mice. J Allergy Clin Immunol. 2006;117(5):1125–32. doi: 10.1016/j.jaci.2006.01.016 [DOI] [PubMed] [Google Scholar]
  • 24.Zhao C, Shi G, Vistica BP, Hinshaw SJH, Wandu WS, Tan C, et al. Induced regulatory T-cells (iTregs) generated by activation with anti-CD3/CD28 antibodies differ from those generated by the physiological-like activation with antigen/APC. Cell Immunol. 2014;290(2):179–84. doi: 10.1016/j.cellimm.2014.06.004 [DOI] [PubMed] [Google Scholar]
  • 25.Sato T, Sasahara T, Nakamura Y, Osaki T, Hasegawa T, Tadakuma T, et al. Naive T cells can mediate delayed‐type hypersensitivity response in T cell receptor transgenic mice. Eur J Immunol. 1994;24(7):1512–6. doi: 10.1002/eji.1830240708 [DOI] [PubMed] [Google Scholar]
  • 26.Nakajima-Adachi H, Koike E, Totsuka M, Hiraide E, Wakatsuki Y, Kiyono H, et al. Two distinct epitopes on the ovalbumin 323-339 peptide differentiating CD4⁺T cells into the Th2 or Th1 phenotype. Biosci Biotechnol Biochem. 2012;76(10):1979–81. doi: 10.1271/bbb.120349 [DOI] [PubMed] [Google Scholar]
  • 27.Dardalhon V, Awasthi A, Kwon H, Galileos G, Gao W, Sobel RA, et al. IL-4 inhibits TGF-β-induced Foxp3+ T cells and, together with TGF-β, generates IL-9+ IL-10+ Foxp3− effector T cells. Nat Immunol. 2008;9(12):1347–55. doi: 10.1038/ni.1677 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Hill JA, Hall JA, Sun C-M, Cai Q, Ghyselinck N, Chambon P, et al. Retinoic acid enhances Foxp3 induction indirectly by relieving inhibition from CD4+CD44hi Cells. Immunity. 2008;29(5):758–70. doi: 10.1016/j.immuni.2008.09.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 29.Tu E, Chia CPZ, Chen W, Zhang D, Park SA, Jin W, et al. T cell receptor-regulated TGF-β type I receptor expression determines T cell quiescence and activation. Immunity. 2018;48(4):745–759.e6. doi: 10.1016/j.immuni.2018.03.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 30.Nakajima-Adachi H, Kikuchi A, Fujimura Y, Shibahara K, Makino T, Goseki-Sone M, et al. Peyer’s patches and mesenteric lymph nodes cooperatively promote enteropathy in a mouse model of food allergy. PLoS One. 2014;9(10):e107492. doi: 10.1371/journal.pone.0107492 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 31.Traxinger BR, Richert-Spuhler LE, Lund JM. Mucosal tissue regulatory T cells are integral in balancing immunity and tolerance at portals of antigen entry. Mucosal Immunol. 2022;15(3):398–407. doi: 10.1038/s41385-021-00471-x [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Joetham A, Schedel M, O’Connor BP, Kim S, Takeda K, Abbott J, et al. Inducible and naturally occurring regulatory T cells enhance lung allergic responses through divergent transcriptional pathways through divergent transcriptional pathways. J Allergy Clin Immunol. 2017;139:1331–42. https://doi:10.1016/j.jaci.2016.06.051. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 33.Mikami N, Kawakami R, Chen KY, Sugimoto A, Ohkura N, Sakaguchi S. Epigenetic conversion of conventional T cells into regulatory T cells by CD28 signal deprivation. Proc Natl Acad Sci U S A. 2020;117(22):12258–68. doi: 10.1073/pnas.1922600117 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Wallberg M, Recino A, Phillips J, Howie D, Vienne M, Paluch C, et al. Anti-CD3 treatment up-regulates programmed cell death protein-1 expression on activated effector T cells and severely impairs their inflammatory capacity. Immunology. 2017;151(2):248–60. doi: 10.1111/imm.12729 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35.Tian J, Dang H, O’Laco KA, Song M, Tiu BC, Gilles S, et al. Homotaurine treatment enhances CD4+ and CD8+ regulatory T cell responses and synergizes with low-dose Anti-CD3 to enhance diabetes remission in type 1 diabetic mice. Immunohorizons. 2019;3(10):498–510. doi: 10.4049/immunohorizons.1900019 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Kuhn C, Weiner HL. Therapeutic anti-CD3 monoclonal antibodies: from bench to bedside. Immunotherapy. 2016;8(8):889–906. doi: 10.2217/imt-2016-0049 [DOI] [PubMed] [Google Scholar]
  • 37.Chitnis T, Kaskow BJ, Case J, Hanus K, Li Z, Varghese JF, et al. Nasal administration of anti-CD3 monoclonal antibody modulates effector CD8+ T cell function and induces a regulatory response in T cells in human subjects. Front Immunol. 2022;13:956907. doi: 10.3389/fimmu.2022.956907 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 38.Zundler S, Becker E, Schulze LL, Neurath MF. Immune cell trafficking and retention in inflammatory bowel disease: mechanistic insights and therapeutic advances. Gut. 2019;68(9):1688–700. doi: 10.1136/gutjnl-2018-317977 [DOI] [PubMed] [Google Scholar]
  • 39.Larson JH, Jin S, Loschi M, Bolivar Wagers S, Thangavelu G, Zaiken MC, et al. Enforced gut homing of murine regulatory T cells reduces early graft-versus-host disease severity. Am J Transplant. 2023;23(8):1102–15. doi: 10.1016/j.ajt.2023.01.030 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Stephen-Victor E, Crestani E, Chatila TA. Dietary and microbial determinants in food allergy. Immunity. 2020;53(2):277–89. doi: 10.1016/j.immuni.2020.07.025 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Olalekan SA, Cao Y, Hamel KM, Finnegan A. B cells expressing IFN-γ suppress Treg-cell differentiation and promote autoimmune experimental arthritis. Eur J Immunol. 2015;45(4):988–98. doi: 10.1002/eji.201445036 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 42.Dominguez-Villar M, Baecher-Allan CM, Hafler DA. Identification of T helper type 1-like, Foxp3+ regulatory T cells in human autoimmune disease. Nat Med. 2011;17(6):673–5. doi: 10.1038/nm.2389 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Caretto D, Katzman SD, Villarino AV, Gallo E, Abbas AK. Cutting edge: the Th1 response inhibits the generation of peripheral regulatory T cells. J Immunol. 2010;184(1):30–4. doi: 10.4049/jimmunol.0903412 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 44.Koenecke C, Lee C-W, Thamm K, Föhse L, Schafferus M, Mittrücker H-W, et al. IFN-γ production by allogeneic Foxp3+ regulatory T cells is essential for preventing experimental graft-versus-host disease. J Immunol. 2012;189(6):2890–6. doi: 10.4049/jimmunol.1200413 [DOI] [PubMed] [Google Scholar]
  • 45.Wang Z, Hong J, Sun W, Xu G, Li N, Chen X, et al. Role of IFN-gamma in induction of Foxp3 and conversion of CD4+ CD25- T cells to CD4+ Tregs. J Clin Invest. 2006;116(9):2434–41. doi: 10.1172/JCI25826 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Deimel LP, Li Z, Roy S, Ranasinghe C. STAT3 determines IL-4 signalling outcomes in naïve T cells. Sci Rep. 2021;11(1). doi: 10.1038/s41598-021-89860-7 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 47.Morikawa H, Sakaguchi S. 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;259(1):192–205. doi: 10.1111/imr.12174 [DOI] [PubMed] [Google Scholar]
  • 48.Abdeladhim M, Zhang A-H, Kropp LE, Lindrose AR, Venkatesha SH, Mitre E, et al. Engineered ovalbumin-expressing regulatory T cells protect against anaphylaxis in ovalbumin-sensitized mice. Clin Immunol. 2019;207:49–54. doi: 10.1016/j.clim.2019.07.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Akamatsu M, Mikami N, Ohkura N, Kawakami R, Kitagawa Y, Sugimoto A, et al. Conversion of antigen-specific effector/memory T cells into Foxp3-expressing Treg cells by inhibition of CDK8/19. Sci Immunol. 2019;4(40):eaaw2707. doi: 10.1126/sciimmunol.aaw2707 [DOI] [PubMed] [Google Scholar]
  • 50.Burggraf M, Nakajima-Adachi H, Hachimura S, Ilchmann A, Pemberton AD, Kiyono H, et al. Oral tolerance induction does not resolve gastrointestinal inflammation in a mouse model of food allergy. Mol Nutr Food Res. 2011;55(10):1475–83. doi: 10.1002/mnfr.201000634 [DOI] [PubMed] [Google Scholar]

Decision Letter 0

Masanori A Murayama

24 Jun 2024

PONE-D-24-17797Naïve intestinal T cells in food-allergic model mice, expanded with anti-CD3 and anti-CD28 antibodies while under regulatory T cell-polarization culture condition, can exhibit suppressive activityPLOS ONE

Dear Dr. Nakajima-Adachi,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process.

Please submit your revised manuscript by Aug 08 2024 11:59PM. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plosone@plos.org . When you're ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

If applicable, we recommend that you deposit your laboratory protocols in protocols.io to enhance the reproducibility of your results. Protocols.io assigns your protocol its own identifier (DOI) so that it can be cited independently in the future. For instructions see: https://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols . Additionally, PLOS ONE offers an option for publishing peer-reviewed Lab Protocol articles, which describe protocols hosted on protocols.io. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols .

We look forward to receiving your revised manuscript.

Kind regards,

Masanori A. Murayama

Academic Editor

PLOS ONE

Journal requirements:

When submitting your revision, we need you to address these additional requirements.

1. Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at 

https://journals.plos.org/plosone/s/file?id=wjVg/PLOSOne_formatting_sample_main_body.pdf and 

https://journals.plos.org/plosone/s/file?id=ba62/PLOSOne_formatting_sample_title_authors_affiliations.pdf

2. To comply with PLOS ONE submissions requirements, in your Methods section, please provide additional information regarding the experiments involving animals and ensure you have included details on (1) methods of sacrifice, (2) methods of anesthesia and/or analgesia, and (3) efforts to alleviate suffering.

3. We note that the grant information you provided in the ‘Funding Information’ and ‘Financial Disclosure’ sections do not match. 

When you resubmit, please ensure that you provide the correct grant numbers for the awards you received for your study in the ‘Funding Information’ section.

4. Thank you for stating the following financial disclosure: 

 [This work was supported by grants from the Kieikai Research Foundation (HNA, Grant number; 2017S063, https://www.nakashima-foundation.org/kieikai/) and 

Grant-in-Aid for Scientific Research (B) from

the Japan Society for the Promotion of Science (JSPS)(SH, Grant number; 26292065, https://www.jsps.go.jp/j-grantsinaid/index.html) .

The analysis for Fig. 5 and S5 Fig was funded by Meiji Holdings Co., Ltd (HNA and SH). ].  

Please state what role the funders took in the study.  If the funders had no role, please state: ""The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript."" 

If this statement is not correct you must amend it as needed. 

Please include this amended Role of Funder statement in your cover letter; we will change the online submission form on your behalf.

5. Thank you for stating the following in the Competing Interests section: 

[I have read the journal's policy and the authors of this manuscript have the following competing interests: [Meiji Holdings Co.,Ltd.]. 

Please confirm that this does not alter your adherence to all PLOS ONE policies on sharing data and materials, by including the following statement: ""This does not alter our adherence to  PLOS ONE policies on sharing data and materials.” (as detailed online in our guide for authors http://journals.plos.org/plosone/s/competing-interests).  If there are restrictions on sharing of data and/or materials, please state these. Please note that we cannot proceed with consideration of your article until this information has been declared. 

Please include your updated Competing Interests statement in your cover letter; we will change the online submission form on your behalf.

6. We note that you have included the phrase “data not shown” in your manuscript. Unfortunately, this does not meet our data sharing requirements. PLOS does not permit references to inaccessible data. We require that authors provide all relevant data within the paper, Supporting Information files, or in an acceptable, public repository. Please add a citation to support this phrase or upload the data that corresponds with these findings to a stable repository (such as Figshare or Dryad) and provide and URLs, DOIs, or accession numbers that may be used to access these data. Or, if the data are not a core part of the research being presented in your study, we ask that you remove the phrase that refers to these data.

7. Please include captions for your Supporting Information files at the end of your manuscript, and update any in-text citations to match accordingly. Please see our Supporting Information guidelines for more information: http://journals.plos.org/plosone/s/supporting-information. 

Additional Editor Comments:

Thank you for submitting manuscript. This research is very interesting, however, this manuscript is open to discussion in this time.

As reviewer indicated, title is too difficult to understand the contents of this manuscript. To be honest, this manuscript is hard to understand in whole. So this manuscript need to help with English language editing.

As major point, this study just focused on the difference of R23-3 and RagD10, and the experiments and discussion was lack for the development of clinical application. Thus, this manuscript requires significant revision.

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Partly

Reviewer #2: Yes

Reviewer #3: No

**********

2. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: I Don't Know

Reviewer #2: Yes

Reviewer #3: Yes

**********

3. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: No

Reviewer #2: Yes

Reviewer #3: Yes

**********

4. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: No

**********

5. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: General comments

This study tried to find the difference in T cell characteristics between food allergic and non-allergic animal models. The article includes several interesting findings although there are some points which are requested to be improved to publish in PLOS ONE.

Specific comments

1. The title is difficult to understand the aim of this study.

2. I do not understand the reason why the authors stimulated T cells by anti-CD3 and anti-CD28 antibodies. What does the stimulation with the antibodies physiologically mean?

3. Related to above comment, I do not understand the reason why the responses to anti-CD3 and anti-CD28 were different from that to antigen stimulation.

4. The authors mentioned the Tregs from the mice as ‘allergen-specific Tregs”. I was wondering if the Tregs inhibited allergy in an allergen-specific manner. The authors should confirm the allergen specificity of the suppressive activity.

5. The authors wrote “ Naïve CD4+ T cells isolated from CN-fed Rag23-3 mice and stimulated with anti-CD3 and anti-CD28 mAbs differentiate into Tregs” (line 369). However, the authors used not only CD-fed mice but also EW-fed mice in the section. I do not understand the reason why title of the section was different from the content.

6. The position of markers in the graph right bottom of Fig. 2a seems out of alignment.

7. The authors wrote “Fig. 2c (data not shown).” (line 434). The authors should provide all data you used in this manuscript.

8. Fig. 3b shows that each of the anti-IL-4 or anti-IFN-g antibody almost completely inhibited the activity of the Sup by itself. I was wondering why IFN-g in anti-IL-4 condition or IL-4 in anti-IFN-g condition did not affect Treg induction.

9. I was wondering why the characteristics of naïve T cells of EW-fed mice differed from those of CN-fed mice in Fig. 5c. Were the naïve T cells of EW-fed mice really naïve?

10. It was very difficult to understand the purpose of each experiment throughout the manuscript. Please give it a clear title easy to understand the purpose of each section of results and explain the reason why the experiments were adopted for the purpose of each section.

Reviewer #2: <major comment="">

This manuscript showed a potential of ex-vivo polarization of functional Treg cells from the naïve intestinal T cells of on-going allergic enteropathy model mouse. Furthermore, overexpression of IL-4 and IFN-� from activated T cells suppressed the induction of Treg cells in the allergic mouse model. The experiments were conducted appropriately, and the detail of methods and results are clearly shown.

Although the length of manuscript is not limited in PONE journal, the text in method, result and figure legends is overlapping, that makes the redundant impression to the readers. Especially, the text in the figure legends may be shorter, because they use almost an equal volume as the text.

<minor points="">

1. Reference

The Ref. 1,2,4 should be updated to represent the recent clinical situations regarding food allergy and oral immunotherapy.

2. L. 371-373

Immobilized anti-CD3 stimulation should be TCR-mediated, although not antigen-specific stimulation.

3. L. 434

Fig 2c might be Fig 2b?

4. Statement of S3 Fig is missing in the text.

5. L.439-442

Blocking either IL-4 or IFN-� alone almost completely restored the suppressive effect of the Rag23-3_EW_Sup, suggesting these two cytokines are affecting to one lymphocyte simultaneously. How do authors explain this mechanism? Does a naïve T cell express both IL-4 receptor and IFN-� receptor on the surface? I’m wondering if direct supplementation of IL-4 and/or IFN-� shows the same effect?

6. L. 658-660

In this TCR-transgenic mouse model, how authors consider the difference between OVA+APC stimulation and anti-CD3+anti-CD28 stimulation? The difference of TCR signal transduction, or the other signals through co-stimulatory molecules on the APC? In other words, does OVA+APC stimulation with Treg-polarization condition on EW-fed Rag23-3 cells have a potential to induce Tregs?</minor></major>

Reviewer #3: In their manuscript "Naïve intestinal T cells in food-allergic model mice, expanded with anti-CD3 and anti-CD28 antibodies while under regulatory T cell-polarization culture conditions, can exhibit suppressive activity," Haruyo Nakajima-Adachi et al. compared two strains of OVA-specific TCR transgenic mice and found the distinct induction of Tregs and effector memory T cells in food allergen-mediated enteropathy.

1. This manuscript is difficult to follow and understand the emphasized points in its current version. The manuscript could benefit from English language editing to enhance clarity and readability. I strongly suggest this is necessary in its current format.

2. The study begins with the introduction of potential therapy using Treg administration for food allergy patients. Is there any clinical perspective on that? To avoid confusion for the reader, the authors should cite adequate references for Treg therapy in food allergies. Compared to other inflammatory diseases, cellular interventions such as cell therapy for food allergies represent an ultimate approach. The importance of how this research can be substantiated is in question.

3. Two strains of OVA-specific TCR tg mice are used in the manuscript, and they published the differential function of Tregs between these mouse strains in a 2017 PLOS One paper. In that paper, the amount of IL-4 was shown to be a causative factor in the regulation of Treg function. In this manuscript, their aim is to understand the stability of Tregs, but the study lacks analysis of how the cells are “stabilized”, for instance, how epigenetic regulations differ in the two mice lines of iTregs. How about RORgt+ Treg which is important cell population regulating allergic disorder. Is this caused by intrinsic control based on the strength of TCR signaling? How are Tregs differently induced in the two lines? The authors can experimentally define this by measuring the strength of TCR signaling.

4. Counter gating should be shown in Figure 1a of effector memory T cells with adequate control.

5. Upon activation, CD4+ T cells transiently express FOXP3, especially in activated CD4+CD25– T cells, thus CD25 expression should be shown in the panel throughout the manuscript.

6. I don’t understand why IL-2 is significantly induced in the control of Rag23-3 mice. Fully discuss this.

7. TCR is specific for OVA; however, EMT cells are significantly induced in the mice without OVA. What is the underlying mechanism of these phenotypic discrepancies?

8. The author indicated the importance of IL-4 for the induction of Treg. Does IL-4 administration induce Tregs in Rag23-3 CN mice?

9. TGF-beta should be referred to as “TGF-beta1” throughout the manuscript.

10. In terms of Treg induction via non-TCR mediated stimulation, I do not understand why the authors stimulated with an anti-CD3 antibody, which mimics TCR stimulation. The authors should reconsider the fundamental concept of this study and rearrange the experimental design.

11. The authors stop explaining the details of the results obtained about the different induction rates of Tregs between spleen and MLN by citing the Mucosal Immunology paper from Traxinger BR. The authors should fully explain and conclude the results based on their data.

12. IL-4 and IFNg are produced at higher levels in the Rag23-3 mice, thus Th1 and Th2 cells may be induced in the Treg-induction experiments. The authors should show if IL-4 and IFNg suppresses or skews Th1/Th2 cells by analyzing intracellular FACS of Th1/2 in mice.

13. Line 403: “The finding that OVA did not fully induce Treg differentiation, but stimulation with antibodies induced Treg differentiation in CN-fed Rag23-3 mice suggests the presence of factors” – however, they mimic TCR signaling by anti-CD3 and anti-CD28. I don’t understand this part.

14. The authors indicate Tregs derived from Rag23-3 mice have sufficient regulatory activity; however, it is important to examine the functional differences in the suppressive roles of RagDO10 and Rag23-3 derived Tregs in vivo and in vitro. Cytokine productions are different?

15. Lines 552 to 557 are hard to understand. Why do naïve T cells ameliorate differentiation to Tregs? This part is complicate to the reader. Please reorganize this part.

**********

6. PLOS authors have the option to publish the peer review history of their article (what does this mean? ). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy .

Reviewer #1: No

Reviewer #2: No

Reviewer #3: No

**********

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/ . PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org . Please note that Supporting Information files do not need this step.

PLoS One. 2025 May 30;20(5):e0324105. doi: 10.1371/journal.pone.0324105.r003

Author response to Decision Letter 1


9 Nov 2024

Responses to the Editor’s Comments

Comment�

Please ensure that your manuscript meets PLOS ONE's style requirements, including those for file naming. The PLOS ONE style templates can be found at https://journals.plos.org/plosone/s/file?id=ba62/PLOSOne_formatting_sample_title_authors_affiliations.pdf and https://journals.plos.org/plosone/s/file?id=wjVg/PLOSOne_formatting_sample_main_body.pdf

Responses

As requested, we have formatted the manuscript in line with PLOS ONE’s style requirements.

Comment 2

To comply with PLOS ONE submissions requirements, in your Methods section, please provide additional information regarding the experiments involving animals and ensure you have included details on (1) methods of sacrifice, (2) methods of anesthesia and/or analgesia, and (3) efforts to alleviate suffering.

Responses

As requested, we have added the following information regarding the experiments involving animals in Methods section:

“When necessary for cellular and histological analysis, mice were euthanized by cervical dislocation by experts. During the adoptive transfer of Tregs, to facilitate the subsequent injection of cells into the caudal vein, a mouse was placed in a small box with only its tail sticking out through the hole in the box for a few minutes. The injection was performed without anesthetization as smoothly and quickly as possible by experts.” (lines 147–152 in the revised manuscript)

“Our experiments were approved without setting preemptive humane endpoints, because during the experimental period (7 days), the mice did not exhibit other clinical signs than weight loss.” (lines 325–327 in the revised manuscript)

Comments 3 and 4

No. 3

We note that the grant information you provided in the ‘Funding Information’ and ‘Financial Disclosure’ sections do not match. When you resubmit, please ensure that you provide the correct grant numbers for the awards you received for your study in the ‘Funding Information’ section.

Responses

We provided the correct grant numbers for the awards we received for our study in the “Funding Information” section in our corrected manuscript as follows:

“This work was supported by grants from the Kieikai Research Foundation (HNA, Grant number; 2017S063, https://www.nakashima-foundation.org/kieikai/) and Grant-in-Aid for Scientific Research (B) from the Japan Society for the Promotion of Science (JSPS) (SH, Grant number; 26292065, https://www.jsps.go.jp/j-grantsinaid/index.html). The analysis for Fig 2a (CD25), Fig 3b–3g, Fig 5, S4 Fig, and S5 Fig was funded by Meiji Holdings Co., Ltd (HNA and SH), but we have no grant number assigned to this program grant in accordance with the nature of the grant, because The University of Tokyo's Corporate Sponsored Research Programs are programs established to conduct research on common issues that are of highly public nature in collaboration with the University of Tokyo, using funds received from the private sector and other external organizations. [Corporate Sponsored Research Programs | The University of Tokyo (u-tokyo.ac.jp)]”

No. 4

Thank you for stating the following financial disclosure:

[This work was supported by grants from the Kieikai Research Foundation (HNA, Grant number; 2017S063, https://www.nakashima-foundation.org/kieikai/) and

Grant-in-Aid for Scientific Research (B) from

the Japan Society for the Promotion of Science (JSPS) (SH, Grant number; 26292065, https://www.jsps.go.jp/j-grantsinaid/index.html) .

The analysis for Fig. 5 and S5 Fig was funded by Meiji Holdings Co., Ltd (HNA and SH).

Please state what role the funders took in the study. If the funders had no role, please state: ""The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.""

If this statement is not correct you must amend it as needed.

Please include this amended Role of Funder statement in your cover letter; we will change the online submission form on your behalf.

Responses

We have included the following Role of Funder statement in the cover letter to this response document:

“The funders had no role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript.”

In addition, in this revision, we have added several data obtained by some experiments. These experiments have been performed by the funding support by Meiji Holdings Co., Ltd. Thus, we would like to update this financial disclosure as follows:

“This work was supported by grants from the Kieikai Research Foundation (HNA, Grant number; 2017S063, https://www.nakashima-foundation.org/kieikai/) and Grant-in-Aid for Scientific Research (B) from the Japan Society for the Promotion of Science (JSPS) (SH, Grant number; 26292065, https://www.jsps.go.jp/j-grantsinaid/index.html). The analysis for Fig 2a (CD25), Fig 3b–3g, Fig 5, S4 Fig, and S5 Fig was funded by Meiji Holdings Co., Ltd (HNA and SH), but we have no grant number assigned to this program grant in accordance with the nature of the grant, because The University of Tokyo's Corporate Sponsored Research Programs are programs established to conduct research on common issues that are of highly public nature in collaboration with the University of Tokyo, using funds received from the private sector and other external organizations. [Corporate Sponsored Research Programs | The University of Tokyo (u-tokyo.ac.jp)].

The funders had no role in the study design, data collection and analysis, decision to publish, or preparation of the manuscript.”

This update of financial disclosure is included in our cover letter.

Comment 5

Thank you for stating the following in the Competing Interests section:

[I have read the journal's policy and the authors of this manuscript have the following competing interests: [Meiji Holdings Co.,Ltd.].

Please confirm that this does not alter your adherence to all PLOS ONE policies on sharing data and materials, by including the following statement: ""This does not alter our adherence to PLOS ONE policies on sharing data and materials.” (as detailed online in our guide for authors http://journals.plos.org/plosone/s/competing-interests). If there are restrictions on sharing of data and/or materials, please state these. Please note that we cannot proceed with consideration of your article until this information has been declared.

Please include your updated Competing Interests statement in your cover letter; we will change the online submission form on your behalf.

Response

We have added the following text to the Competing Interests section of our revised manuscript:

“all of the authors of this manuscript had read the journal’s policy and that the authors had a competent interest with Meiji Holdings Co., Ltd. This does not alter our adherence to PLOS ONE policies on sharing data and materials.”

This statement is also declared in the cover letter to this response document.

Comment 6

We note that you have included the phrase “data not shown” in your manuscript. Unfortunately, this does not meet our data sharing requirements. PLOS does not permit references to inaccessible data. We require that authors provide all relevant data within the paper, Supporting Information files, or in an acceptable, public repository. Please add a citation to support this phrase or upload the data that corresponds with these findings to a stable repository (such as Figshare or Dryad) and provide and URLs, DOIs, or accession numbers that may be used to access these data. Or, if the data are not a core part of the research being presented in your study, we ask that you remove the phrase that refers to these data.

Response

As requested, we have removed the phrase “data not shown” from the revised manuscript. In addition, we have corrected the citation for Fig 2c to Fig 2b and now cite S3 Fig (corrected S4 Fig) as shown in line 496–497 in the revised manuscript as follows:

“The cytokine profiles of the supernatants used in this experiment are shown in S3 Fig, and these profiles were consistent with those shown in Fig 2b.”

Comment 7

Please include captions for your Supporting Information files at the end of your manuscript, and update any intext citations to match accordingly. Please see our Supporting Information guidelines for more information: http://journals.plos.org/plosone/s/supporting-information.

Response

As requested, we have added captions for our Supporting Information files (lines 1093–1158) at the end of our revised manuscript meeting PLOS ONE's style requirements.

Comment 8

Thank you for submitting manuscript. This research is very interesting, however, this manuscript is open to discussion in this time. As Reviewer indicated, title is too difficult to understand the contents of this manuscript. To be honest, this manuscript is hard to understand in whole. So, this manuscript needs to help with English language editing. As major point, this study just focused on the difference of R23-3 and RagD10, and the experiments and discussion were lack for the development of clinical application. Thus, this manuscript requires significant revision.

Response

We appreciate the opportunity to revise and resubmit our manuscript. We have updated the title to more simply describe our study to readers. In addition, the English in the revised manuscript has been edited by two native-English-speaking professional scientific editors from ELSS, Inc. (http://www.elss.co.jp), who also provided suggestions on ways to improve the clarity of the revised manuscript and according to the suggestion, the original manuscript have been throughout updated. In the Discussion, we now discuss the clinical application (line 889–899 in the revised manuscript). The remaining changes that we have made in response to the Reviewer’s comments are explained on the following pages. In addition, to describe the possibility of clinical application of treatment of Treg-transfer in the intestinal diseases, we added (ref. 15: Int J Mol Sci. 2020;21:7015).

Responses to Reviewer 1

Comment 1

The title is difficult to understand the aim of this study.

Response

We have updated the title as follows:

Old title: “Naïve intestinal T cells in food-allergic model mice, expanded with anti-CD3 and anti-CD28 antibodies while under regulatory T cell-polarization culture condition, can exhibit suppressive activity”

New title: “Ovalbumin-specific regulatory T cells with the naïve phenotype (CD62LloCD44hi) from mesenteric lymph nodes suppress food-allergic enteropathy in mice, when expanded by anti-CD3/CD28 antibodies but not by ovalbumin plus antigen-presenting cells”

Comment 2

I do not understand the reason why the authors stimulated T cells by anti-CD3 and anti-CD28 antibodies. What does the stimulation with the antibodies physiologically mean?

Response

First, we observed that T cells from Rag23-3 mice have lower ability to differentiate into Foxp3+ regulatory T cells (Tregs) than T cells from RagD10 mice. However, to promote T cell differentiation into Tregs, we needed an approach for stimulating and expanding the T cells. In addition, in performing an adoptive transfer of Tregs as a treatment, we have to differentiate and expand CD4+ T cells into Tregs and to generate “poly-clonal Tregs” in vitro after isolating T cells from the individuals. If we used an antigen for the expansion in the culture, we would have to remove both the antigen-presenting cells and the antigen from the culture to increase the purity of the Tregs and to prevent side effects caused by contamination with the antigen. In addition, it has been reported that the functions and phenotype of Tregs differ depending on whether their differentiation was induced by using an antigen plus antigen-presenting cells or by using a combination of anti-CD3 and anti-CD28 antibodies (ref. 24: Cellular Immunol: 2014;290:179–184). Thus, in the present study, we examined the stimulation of T cell differentiation into Tregs by using anti-CD3 and anti-CD28 antibodies as well as OVA plus antigen-presenting cells. We now describe this rationale and a comparison of the data obtained with both approaches (Fig 1: antigen/antigen-presenting cell approach; Fig 2: antibody approach) in the revised Introduction (line 113–119), Discussion (lines 769–788) and at the beginning of the Results (lines 422–427). See also our response to Comment 3.

Comment 3

Related to above comment, I do not understand the reason why the responses to anti-CD3 and anti-CD28 were different from that to antigen stimulation.

Response

As Reviewer 1 indicates, we did not clarify in our original manuscript the reasons for the different outcomes when Treg differentiation is induced by using the antigen/antigen-presenting cell approach or the antibody approach. As mentioned in our response to the previous comment, there is a report showing that there are functional and phenotypic differences between the iTreg lineages induced using either an antigen (HEL)/antigen-presenting cell approach or the combination of anti-CD3/anti-CD28 antibodies (ref. 24: Cellular Immunol. 2014;290:179–184). Therefore, our result has a precedent. However, considering together the present and previous comments from the Reviewer, we have added the following sentences to the revised Discussion to explain the different outcomes from the different approaches to induce Treg differentiation (lines 769–788); the reference mentioned in our responses to comments 2 and 3 has also been added to the revised manuscript as reference 24:

“Although in the present study we found that T cells from EW-fed Rag23-3 mice have lower ability to differentiate into Tregs than T cells from RagD10 mice, we also found that using a combination of anti-CD3 and anti-CD28 mAbs to obtain sufficient volume of Tregs for adoptive transfer to other mice was superior to using a combination of antigen (OVA) and antigen-presenting cells. Although we did not investigate in detail why the responses to the Ab-stimulation were different from those to the antigen stimulation, similar findings have been reported elsewhere: Zhao et al. reported that the inhibitory effects on Th1 responses, surface molecules, and levels of Foxp3 expression in hen-egg-lysozyme–specific Tregs induced by mAbs are not the same as those induced by a combination of antigen plus antigen-presenting cells [24]. The implication of this when considering adoptive Treg transfer as a therapeutic option is that we have to choose the most appropriate approach to obtain Tregs with sufficient suppressive function. Our present results show that using mAbs to stimulate Treg differentiation affords Tregs with superior suppressive function in the context of severe food allergy. In addition, if we were to use the combination of antigen and antigen-presenting cells, we would also have to deal with removing the antigen and antigen-presenting cells from the culture at the end of the process to increase the purity of Tregs and avoid any potentially harmful effects of leftover antigen. Thus, the present findings highlight the potential of using Tregs differentiated from intestinal naïve T cells using a combination of anti-CD3 and anti-CD28 mAbs to suppress severe food allergic inflammation.”

In addition, in the Introduction, we now clearly indicate the reason for using antibody-mediated stimulation rather than antigen-mediated stimulation as follows (lines 113–119 in the revised manuscript):

“It has been reported that the functions and phenotypes of Tregs differentiated by using a combination of anti-CD3 and anti-CD28 monoclonal antibodies (mAbs) differ from those of Tregs differentiated with an antigen in the presence of antigen-presenting cells [24]. Thus, in the present study, we examined how to obtain stable suppressive Tregs from the Rag23-3 food-allergic enteropathy model by comparing both the inducing means and their resultant differentiation levels and functions of Tregs induced in Rag23-3 and RagD10 mice when fed EW.”

Comment 4

The authors mentioned the Tregs from the mice as ‘allergen-specific Tregs”. I was wondering if the Tregs inhibited allergy in an allergen-specific manner. The authors should confirm the allergen specificity of the suppressive activity.

Response

Both the Rag23-3

Attachment

Submitted filename: Responses to Commentsfinal.pdf

pone.0324105.s007.pdf (219.9KB, pdf)

Decision Letter 1

Masanori A Murayama

6 Jan 2025

PONE-D-24-17797R1Ovalbumin-specific regulatory T cells with the naïve phenotype (CD62LloCD44hi) from mesenteric lymph nodes suppress food-allergic enteropathy in mice, when expanded by anti-CD3/CD28 antibodies but not by ovalbumin plus antigen-presenting cellsPLOS ONE

Dear Dr. Nakajima-Adachi,

Thank you for submitting your manuscript to PLOS ONE. After careful consideration, we feel that it has merit but does not fully meet PLOS ONE’s publication criteria as it currently stands. Therefore, we invite you to submit a revised version of the manuscript that addresses the points raised during the review process.

Please submit your revised manuscript by Feb 20 2025 11:59PM. If you will need more time than this to complete your revisions, please reply to this message or contact the journal office at plosone@plos.org . When you're ready to submit your revision, log on to https://www.editorialmanager.com/pone/ and select the 'Submissions Needing Revision' folder to locate your manuscript file.

Please include the following items when submitting your revised manuscript:

  • A rebuttal letter that responds to each point raised by the academic editor and reviewer(s). You should upload this letter as a separate file labeled 'Response to Reviewers'.

  • A marked-up copy of your manuscript that highlights changes made to the original version. You should upload this as a separate file labeled 'Revised Manuscript with Track Changes'.

  • An unmarked version of your revised paper without tracked changes. You should upload this as a separate file labeled 'Manuscript'.

If you would like to make changes to your financial disclosure, please include your updated statement in your cover letter. Guidelines for resubmitting your figure files are available below the reviewer comments at the end of this letter.

If applicable, we recommend that you deposit your laboratory protocols in protocols.io to enhance the reproducibility of your results. Protocols.io assigns your protocol its own identifier (DOI) so that it can be cited independently in the future. For instructions see: https://journals.plos.org/plosone/s/submission-guidelines#loc-laboratory-protocols . Additionally, PLOS ONE offers an option for publishing peer-reviewed Lab Protocol articles, which describe protocols hosted on protocols.io. Read more information on sharing protocols at https://plos.org/protocols?utm_medium=editorial-email&utm_source=authorletters&utm_campaign=protocols .

We look forward to receiving your revised manuscript.

Kind regards,

Masanori A. Murayama, Ph.D.

Academic Editor

PLOS ONE

Additional Editor Comments:

Thank you for re-submission of your manuscript. This revised manuscript is interesting, but it has open issues. Please check comments from reviewer2. And I have some comments; In introduction section, please explain why authors use different OVA-TCR Tg mice. At least, authors should explain what is the difference OVA23-3 and DO11.10 mice at basic levels.

In Results section, authors should delete the supplementary explanation about fundamental knowledge, and some sentence should move discussion section. To promote understanding the contents, authors need concise result section. I think the result section is too long. As pointed out by reviewer 2, conclusions are unclear.

At all figures, it was very difficult to understand what is the significantly difference. Please use * or #, instead of a, b, c, and d, and use lines between graphs. And please change small to large alphabet, Fig1a -> Fig1A. And please change IFN-gamma -> IFNγ, TGF beta1 - TGFβ1.

In result section, authors investigated some cytokine from helper T cells, but not IL-10 and IL-17. Why authors did not investigate IL-10 expression?

[Note: HTML markup is below. Please do not edit.]

Reviewers' comments:

Reviewer's Responses to Questions

Comments to the Author

1. If the authors have adequately addressed your comments raised in a previous round of review and you feel that this manuscript is now acceptable for publication, you may indicate that here to bypass the “Comments to the Author” section, enter your conflict of interest statement in the “Confidential to Editor” section, and submit your "Accept" recommendation.

Reviewer #1: All comments have been addressed

Reviewer #2: All comments have been addressed

Reviewer #3: (No Response)

**********

2. Is the manuscript technically sound, and do the data support the conclusions?

The manuscript must describe a technically sound piece of scientific research with data that supports the conclusions. Experiments must have been conducted rigorously, with appropriate controls, replication, and sample sizes. The conclusions must be drawn appropriately based on the data presented.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: Partly

**********

3. Has the statistical analysis been performed appropriately and rigorously?

Reviewer #1: (No Response)

Reviewer #2: Yes

Reviewer #3: Yes

**********

4. Have the authors made all data underlying the findings in their manuscript fully available?

The PLOS Data policy requires authors to make all data underlying the findings described in their manuscript fully available without restriction, with rare exception (please refer to the Data Availability Statement in the manuscript PDF file). The data should be provided as part of the manuscript or its supporting information, or deposited to a public repository. For example, in addition to summary statistics, the data points behind means, medians and variance measures should be available. If there are restrictions on publicly sharing data—e.g. participant privacy or use of data from a third party—those must be specified.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: Yes

**********

5. Is the manuscript presented in an intelligible fashion and written in standard English?

PLOS ONE does not copyedit accepted manuscripts, so the language in submitted articles must be clear, correct, and unambiguous. Any typographical or grammatical errors should be corrected at revision, so please note any specific errors here.

Reviewer #1: Yes

Reviewer #2: Yes

Reviewer #3: No

**********

6. Review Comments to the Author

Please use the space provided to explain your answers to the questions above. You may also include additional comments for the author, including concerns about dual publication, research ethics, or publication ethics. (Please upload your review as an attachment if it exceeds 20,000 characters)

Reviewer #1: The authors appropriately answered to my questions and comments, and revised the manuscript according to the answers.

Reviewer #2: The manuscript has been appropriately edited, and the methods and results sound much clearer. The role of excessive amount of IL-4 and IFN-gamma, either autologous production or supplementation, on the suppression of Treg differentiation has now been clearly shown.

Reviewer #3: In their manuscript, "Naïve intestinal T cells in food-allergic model mice, expanded with anti-CD3 and anti-CD28 antibodies under regulatory T cell-polarization culture conditions, can exhibit suppressive activity," the authors currently describe, "Ovalbumin-specific regulatory T cells with the naïve phenotype (CD62L^lo^CD44^hi^) from mesenteric lymph nodes suppress food-allergic enteropathy in mice, when expanded by anti-CD3/CD28 antibodies but not by ovalbumin plus antigen-presenting cells." Haruyo Nakajima-Adachi et al. compared two strains of OVA-specific TCR transgenic mice and found distinct differences in the induction of regulatory T cells (Tregs) and effector memory T cells during food allergen-mediated enteropathy.

The comparison between anti-CD3/CD28 antibodies and ovalbumin plus antigen-presenting cells is a critical aspect of the paper. I suggest a revised title for the manuscript: "Stable and suppressive regulatory T cells from mesenteric lymph nodes suppress food-allergic enteropathy in mice" as the current title is long and somewhat complex.

Extracting Tregs from mesenteric lymph nodes (MLN) for therapeutic use in food allergy patients seems impractical. Would it not be possible to use Tregs derived from peripheral blood instead as an experimental setting which strength the requirement of their study? The manuscript appears to lack appropriate control experiments to address this question. The authors mention in line 79 that “it is difficult to separate allergen-specific Tregs from the total population of peripheral blood mononuclear cells.” However, obtaining Tregs from MLN seems even more challenging than isolation from peripheral blood mononuclear cells (PBMCs).

The authors highlight that iTregs derived from naïve CD4+ T cells of OVA-transgenic mice strongly suppress allergies. However, their explanation about how antigen-specific naïve T cell-derived Tregs could be obtained in non-transgenic models is overly complex for a general understanding of immunological mechanisms.

There are several established mouse models for food allergy, such as OVA-Alum. It would be more robust to test the expansion of Tregs using these models, comparing the two methods described in the manuscript. Demonstrating the inhibition of allergic symptoms with the authors’ method would strengthen the study’s findings.

Additionally, the manuscript cites a new reference (Reference 24). However, the cited paper’s results seem contradictory to the authors’ findings, stating that “iTregs generated by activation with antigen/APC are more suppressive than iTregs generated by anti-CD3/CD28 antibodies.” The cited manuscript attributes these functional differences to chemokine-related gene expression. If the authors choose to reference this paper, they should address this apparent discrepancy in their discussion.

Overall, the manuscript leaves its conclusions unclear. It does not effectively explain what was discovered regarding the differences between the two OVA-TCR mouse strains or the mechanisms of Treg induction by different antigen stimulation methods. While the stated aim is to evaluate the stability of Tregs, the study lacks critical analysis, such as how epigenetic regulation might contribute to stabilization in the two strains of iTregs. To provide generalizable insights, I recommend additional experimental comparisons of Tregs derived from these strains to clarify their cellular properties.

**********

7. PLOS authors have the option to publish the peer review history of their article (what does this mean? ). If published, this will include your full peer review and any attached files.

If you choose “no”, your identity will remain anonymous but your review may still be made public.

Do you want your identity to be public for this peer review? For information about this choice, including consent withdrawal, please see our Privacy Policy .

Reviewer #1: No

Reviewer #2: No

Reviewer #3: No

**********

[NOTE: If reviewer comments were submitted as an attachment file, they will be attached to this email and accessible via the submission site. Please log into your account, locate the manuscript record, and check for the action link "View Attachments". If this link does not appear, there are no attachment files.]

While revising your submission, please upload your figure files to the Preflight Analysis and Conversion Engine (PACE) digital diagnostic tool, https://pacev2.apexcovantage.com/ . PACE helps ensure that figures meet PLOS requirements. To use PACE, you must first register as a user. Registration is free. Then, login and navigate to the UPLOAD tab, where you will find detailed instructions on how to use the tool. If you encounter any issues or have any questions when using PACE, please email PLOS at figures@plos.org . Please note that Supporting Information files do not need this step.

PLoS One. 2025 May 30;20(5):e0324105. doi: 10.1371/journal.pone.0324105.r005

Author response to Decision Letter 2


17 Apr 2025

Responses to the Editor’s Comments

Comment 1

In introduction section, please explain why authors use different OVA-TCR Tg mice. At least, authors should explain what is the difference OVA23-3 and DO11.10 mice at basic levels.

Responses

As requested, we have added the following information about the differences of basic immune responses in these OVA-TCR Tg mice in the Introduction section in the revised manuscripts:

“The mice predominantly produce IFN-γ by stimulation with anti-CD3 antibody or OVA, indicating Th1-biased responses [22].” (lines 103 – 105 in the revised manuscript)

“and strong IL-4 responses, when they were stimulated with anti-CD3 antibody or OVA [22].” (lines 107 – 108 in the revised manuscript).

In addition, to clarify our aim and the differences of the 2 strains of mice, we corrected some words in the Abstract section and highlighted by yellow.

Comment 2

In Results section, authors should delete the supplementary explanation about fundamental knowledge, some sentence should move discussion section.

Responses

As the Editor commented, it might be better to move some sentences to the Discussion section.

However, we felt that it is difficult to understand our results, especially Figure 2 and 3, without the supplementary explanation of the results and the subsequent discussion of the results, so we have added the explanation at the site after the description of the results of the figures, especially Figure 2 and 3. However, in the Results section of Figure 2, we agree that the description explaining the results was not appropriate. Therefore, we have corrected some sentences (lines 434 – 435, 441– 445 as highlighted by yellow.

Therefore, we would like to submit our revised manuscript without any changes regarding the supplementary explanation, although we can understand the editor's comment.

Comment 3

it was very difficult to understand what is the significantly difference. Please use * or #, instead of a, b, c, and d, and use lines between graphs. And please change small to large alphabet, Fig1a -> Fig1A. And please change IFN-gamma -> IFNγ, TGF beta1 - TGFβ1.

Responses

We have corrected the way to show the significances by different letters, such as a, b, c, and d to * or # in all figures and each legend (highlighted by yellow) and corrected small to large alphabet like from Fig1a to Fig 1A in the revised manuscript. In addition, we have corrected IFN-gamma to IFNγ, and TGF beta1 - TGFβ1.

Comment 4

In result section, authors investigated some cytokine from helper T cells, but not IL-10 and IL-17. Why authors did not investigate IL-10 expression?

Responses

As the editor suggested, we further analyzed IL-10 production in the supernatant of MLN or spleen CD4+ T cells incubated under in vitro Treg-polarization culture condition stimulated with anti-CD3 and anti-CD28 antibodies. The results were added in Fig. 2B. The results showed that Treg-population induced in Rag23-3 or RagD10 mice differed in IL-10 production when stimulated and differentiated with anti-CD3 and CD28 antibodies.

Therefore, we have corrected the legend of Figure 2 (line 491), have added the ELISA kit used in this assay in the Methods section (lines 243 – 244) and further added the following sentences in the Results section of our revised manuscript:

“The level of IL-10 production in the spleen cells and in the mLN cells was significantly higher in the Rag23-3 mice than in the RagD10 mice when fed the EW-diet. The amount of IL-10 produced by CD4+ T cells in mLN was much more than that in spleen EW-fed mice.” (lines 465 – 468 in the revised manuscript)

“the production of IL-4, IFN-γ, and IL-10 in EW-fed Rag23-3 mice. In addition, Tregs differentiated from CD4+ T cells by stimulation with mAbs in both strains of EW-fed mice probably have sufficient regulatory function producing IL-10, but suppressive function of the Tregs in both strains of CN-fed mice may be independent of IL-10.” (lines 470 – 473 in the revised manuscript)

Others

We apologize for the lack of description of the result describing IL-2 production in the supernatant in Fig 5D of our previous manuscript. Therefore, we have added the following sentences in the Results section (lines 685 – 688) of the revised manuscript as follows;

“IL-2 production in mLN was comparable among experimental groups, but that in EW_EMT in SPL showed the highest level compared to those in other groups, but the difference of IL-2 production was not affected to the induction of Tregs (Fig 5D).”

Responses to Reviewer 3

Comment1

I suggest a revised title for the manuscript: "Stable and suppressive regulatory T cells from mesenteric lymph nodes suppress food-allergic enteropathy in mice" as the current title is long and somewhat complex.

Responses

Following the suggestion of Reviewer3, we have changed the title of our revised manuscript.

Old title: "Ovalbumin-specific regulatory T cells with the naïve phenotype (CD62LloCD44hi) from mesenteric lymph nodes suppress food-allergic enteropathy in mice, when expanded by anti-CD3/CD28 antibodies but not by ovalbumin plus antigen-presenting cells".

We agree with the critical comments of Reviewer3 that our manuscript should reduce the generality but emphasize the specificity of the method inducing regulatory T cells from the mouse model of severe intestinal allergy. However, the unique way in which we obtained the suppressive and stable regulatory T cells in this study even under severe food allergic conditions, is that by using anti-CD3 and anti-CD28 antibodies for stimulation, naive antigen-specific CD4+ T cells which are likely to originate from the intestinal tissues, are differentiated into Tregs. Therefore, if Reviewer3 agrees with our proposal, we think it would be better to add the words "ovalbumin-specific regulatory T cells differentiated from naïve phenotype (CD44loCD62Lhi)”, “mesenteric lymph nodes”, and “severe food allergy” in the title. Furthermore, we would like to clearly state our claim in the title, as the title suggested by Reviewer3 is similar to our short title “Stable and suppressive regulatory T cell induction in a food-allergic enteropathy mouse model”.

We would appreciate it if Reviewer3 and the Editor could reconsider and accept the title we changed based on Reviewer 3's comments in the revised manuscript as follows:

New title: “Ovalbumin-specific regulatory T cells differentiated from the naïve phenotype (CD44loCD62Lhi) in mesenteric lymph nodes stably suppress enteropathy even in severe food-allergic mice "

Comment2

Extracting Tregs from mesenteric lymph nodes (MLN) for therapeutic use in food allergy patients seems impractical. Would it not be possible to use Tregs derived from peripheral blood instead as an experimental setting which strength the requirement of their study? The manuscript appears to lack appropriate control experiments to address this question. The authors mention in line 79 that “it is difficult to separate allergen-specific Tregs from the total population of peripheral blood mononuclear cells.” However, obtaining Tregs from MLN seems even more challenging than isolation from peripheral blood mononuclear cells (PBMCs).

Responses

As Reviewer3 suggested, we agree that extracting Tregs from MLN for therapeutic use would be impossible in food allergic patients. Therefore, we proposed the use of marker molecule (such as alpha4beta7) migrating to the intestine, expressed on naïve phenotype T cells to enable the method possible in clinical use; as described in lines 828 – 830 in the discussion section of our previous manuscript;

“Therefore, our results suggest that naïve-like (CD44loCD62hi) CD4+ T cells expressing a surface marker to migrate to the intestinal tissues (e.g., the gut homing integrin, alpha4beta7 [38]) may be best for producing stable Tregs”.

By considering Reviewer3’s comment, we thought that the description (line 78 – 79) in the Introduction section of the previous manuscript may give a misunderstanding to the readers of PLOS ONE, because allergic researchers have made efforts to find the maker of Tregs in peripheral blood. In addition, in the Discussion section, we mentioned about the marker migrating to the intestinal tissue of Tregs in the peripheral blood. Actually, although it was in graft-versus-host disease, C-C motif chemokine receptor 9 (CCR9), specific homing receptors for colon or small intestine, was presented as a candidate of transplantation Treg marker migrating to the intestine and reducing the symptom (American J Transplantation, 2023:23;1102-1115, https://doi.org/10.1016/j.ajt.2023.01.030), although they were obtained from spleen. However, they are trying to transplant Tregs migrating to the intestine and cure the disease. Therefore, we have corrected the sentence in the Introduction section and added CCR9 molecule as a candidate molecule and this paper as a reference in the Discussion section of the revised manuscript.

<Introduction section>

Old

*lines 78 – 81: however, it is difficult to separate allergen-specific Tregs from the total population of peripheral blood mononuclear cells, because of the lack of specific surface markers of Tregs with stable inhibitory functions that will not produce off-target effects, although CD137 has been proposed as a candidate marker [16-18].

New

*lines 78 – 80 in the revised manuscript: “many allergists are trying to find markers for stable Tregs specifically activated by antigens in peripheral blood, but the goal has not been achieved, although CD137 has been proposed as a candidate marker [16-18]”.

<Discussion section>

Old

*lines 828 – 830: Thus, our results suggest that naïve-like (CD44loCD62hi) CD4+ T cells expressing a surface marker to migrate to the intestinal tissues (e.g., the gut homing integrin, alpha4beta7 [38]) may be best for producing stable Tregs.

New

*lines 844 – 847 in the revised manuscript: “Thus, our results suggest that naïve-like (CD44loCD62hi) CD4+ T cells expressing a surface marker to migrate to the intestinal tissues (e.g., the gut homing integrin, α4β7 [38] or C-C motif chemokine receptor 9 specific homing receptors for colon or small intestine, CCR9 [39]) may be better for producing stable Tregs.”

*Reference:

39. Larson JH, Jin S, Loschi M, Bolivar Wagers S, Thangavelu G, Zaiken MC, et al. Enforced gut homing of murine regulatory T cells reduces early graft-versus-host disease severity. Am J Transplant. 2023;23:1102-1115. https:// doi: 10.1016/j.ajt.2023.01.030

Comment3

The authors highlight that iTregs derived from naïve CD4+ T cells of OVA-transgenic mice strongly suppress allergies. However, their explanation about how antigen-specific naïve T cell-derived Tregs could be obtained in non-transgenic models is overly complex for a general understanding of immunological mechanisms.

There are several established mouse models for food allergy, such as OVA-Alum. It would be more robust to test the expansion of Tregs using these models, comparing the two methods described in the manuscript. Demonstrating the inhibition of allergic symptoms with the authors’ method would strengthen the study’s findings.

Responses

We are grateful for Reviewer3’s suggestion to make our results more robust as a better way to induce functional Tregs. However, we have confirmed that the food-allergic inflammation and recovery from the severe inflammation observed in the EW-fed TCR-transgenic mouse model of OVA23-3, such as enteropathy, bone loss, tolerance acquisition, and mast cell infiltration into the intestinal tissues are reproduced by a food-allergic enteropathy model established by using BALB/c mice fed with EW-diet after intraperitoneal sensitization with OVA and alum. (Burggraf M., Nakajima-Adachi H., et al. Mol. Nutr. Food Res. 2011;55:1475–1483). In our manuscript, we have not yet analyzed in detail the characteristics of Tregs in the BALB/c mouse model and therefore, we agree to confirm the reproducibility of the induction of functional Tregs in the BALB/c mouse model. Indeed, we can analyze OVA-specific Treg responses by using the BALB/c mouse model (Morinaga H., Allergol Int. 2020;69:622-625). However, even if the method of inducing Tregs used in the present study worked in the BALB/c mouse model, we would raise the question whether it was the effect of alum immunity and whether the results might be different if we used the other adjuvant. Also, in allergic diseases, effective treatments vary from person to person and may not work for everyone. Therefore, we have added a sentence in the Discussion section suggesting this Treg induction method as one of them in severe allergy and added a reference in our revised manuscript as follows;

“although we need to analyze the effect of stability of Treg function using the BALB/c mouse model [50] and verify the generality of this method,” (lines 920 – 921 in the revised manuscript)

*Reference:

50. Burggraf M, Nakajima-Adachi H, Hachimura S, Ilchmann A, Pemberton AD, Kiyono H., et al. Mol. Nutr. Food Res. 2011;55:1475-1483. doi 10.1002/mnfr.201000634)

Comment4

Additionally, the manuscript cites a new reference (Reference 24). However, the cited paper’s results seem contradictory to the authors’ findings, stating that “iTregs generated by activation with antigen/APC are more suppressive than iTregs generated by anti-CD3/CD28 antibodies.” The cited manuscript attributes these functional differences to chemokine-related gene expression. If the authors choose to reference this paper, they should address this apparent discrepancy in their discussion.

Responses

Thank you for your suggestion. Although the study in reference 24 showed that the iTregs generated by anti-CD3 and anti-CD28 antibodies were less suppressive than iTregs generated by stimulation with HEL, which was the antigen of the Tregs, and APC, the situation and aim of the experiments were different from ours. We indicated in our previous manuscript that we could induce iTregs from severe allergic mice by using anti-CD3 and anti-CD28 antibodies, not antigen/APCs by the following mechanism that the mice were unable to induce Tregs when they were stimulated with antigen and APC, because of overproduction of IL-4 and IFN-γ. Therefore, we don’t think there is a discrepancy with reference24, but to avoid any misunderstanding of our aim, we have added some words in line 790 and corrected sentences in lines 794 – 796 in the revised manuscript, and both corrections are highlighted in yellow as follows;

“Although we did not investigate in detail why the responses to the Ab-stimulation were different from those to the antigen stimulation, similar findings, albeit with a different capacity of Tregs, have been reported elsewhere: Zhao et al. reported that the inhibitory effects on Th1 responses, surface molecules, and of Foxp3 expression levels in hen-egg-lysozyme–specific Tregs induced by mAbs are not the same as those induced by a combination of antigen plus antigen-presenting cells [24]. The implication of this result is that when considering adoptive Treg transfer as a therapeutic option, we need to choose the most appropriate approach to obtain Tregs with sufficient suppressive function. Our present results suggest that” 

Comment 5

Overall, the manuscript leaves its conclusions unclear. It does not effectively explain what was discovered regarding the differences between the two OVA-TCR mouse strains or the mechanisms of Treg induction by different antigen stimulation methods. While the stated aim is to evaluate the stability of Tregs, the study lacks critical analysis, such as how epigenetic regulation might contribute to stabilization in the two strains of iTregs. To provide generalizable insights, I recommend additional experimental comparisons of Tregs derived from these strains to clarify their cellular properties.

Responses

We are grateful for suggesting the importance comparing epigenetic status between Tregs from Rag23-3 as a severe food-allergic enteropathy mouse model and those from RagD10 as a tolerance acquisition mouse model. However, our study got started at the finding that in Rag23-3 mice, iTregs could not be induced by stimulation with OVA plus APCs. Therefore, it is the most important finding in

Attachment

Submitted filename: Responses to Reviewers#2.docx

pone.0324105.s008.docx (40.1KB, docx)

Decision Letter 2

Masanori A Murayama

22 Apr 2025

Ovalbumin-specific regulatory T cells differentiated from the naïve phenotype (CD44loCD62Lhi) in mesenteric lymph nodes stably suppress enteropathy even in severe food-allergic mice

PONE-D-24-17797R2

Dear Dr. Haruyo Nakajima-Adachi,

We’re pleased to inform you that your manuscript has been judged scientifically suitable for publication and will be formally accepted for publication once it meets all outstanding technical requirements.

Within one week, you’ll receive an e-mail detailing the required amendments. When these have been addressed, you’ll receive a formal acceptance letter and your manuscript will be scheduled for publication.

An invoice will be generated when your article is formally accepted. Please note, if your institution has a publishing partnership with PLOS and your article meets the relevant criteria, all or part of your publication costs will be covered. Please make sure your user information is up-to-date by logging into Editorial Manager at Editorial Manager®  and clicking the ‘Update My Information' link at the top of the page. If you have any questions relating to publication charges, please contact our Author Billing department directly at authorbilling@plos.org.

If your institution or institutions have a press office, please notify them about your upcoming paper to help maximize its impact. If they’ll be preparing press materials, please inform our press team as soon as possible -- no later than 48 hours after receiving the formal acceptance. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

Kind regards,

Masanori A. Murayama, Ph.D.

Academic Editor

PLOS ONE

Additional Editor Comments (optional):

Thank you for submitting revised manuscript. I am pleased for sending this mail. This manuscript is completely revised according to reviewer's comments. Thus, my decision is accept in this revision. Congratulations.

Reviewers' comments:

Acceptance letter

Masanori A Murayama

PONE-D-24-17797R2

PLOS ONE

Dear Dr. Nakajima-Adachi,

I'm pleased to inform you that your manuscript has been deemed suitable for publication in PLOS ONE. Congratulations! Your manuscript is now being handed over to our production team.

At this stage, our production department will prepare your paper for publication. This includes ensuring the following:

* All references, tables, and figures are properly cited

* All relevant supporting information is included in the manuscript submission,

* There are no issues that prevent the paper from being properly typeset

You will receive further instructions from the production team, including instructions on how to review your proof when it is ready. Please keep in mind that we are working through a large volume of accepted articles, so please give us a few days to review your paper and let you know the next and final steps.

Lastly, if your institution or institutions have a press office, please let them know about your upcoming paper now to help maximize its impact. If they'll be preparing press materials, please inform our press team within the next 48 hours. Your manuscript will remain under strict press embargo until 2 pm Eastern Time on the date of publication. For more information, please contact onepress@plos.org.

If we can help with anything else, please email us at customercare@plos.org.

Thank you for submitting your work to PLOS ONE and supporting open access.

Kind regards,

PLOS ONE Editorial Office Staff

on behalf of

Dr. Masanori A. Murayama

Academic Editor

PLOS ONE

Associated Data

    This section collects any data citations, data availability statements, or supplementary materials included in this article.

    Supplementary Materials

    S1 Fig. Body weight changes and jejunum histology in RagD10 and Rag23−3 mice fed a diet containing egg white (EW) or casein (CN).

    A) RagD10 and Rag23−3 mice were fed a diet containing EW or CN for 7 days, and body weights were measured on days 0, 2, 4, 5, and 7. Body weight changes were determined relative to those on day 0. Values are expressed as mean ± SD. B) Jejunum histology. Samples of jejunum were collected from the mice on day 7 and stained with hematoxylin and eosin. Scale bars, 100 µm. n = 3 per group. Data are representative of two independent experiments. Statistical analysis: Tukey’s HSD test [*p < 0.05 (Rag23−3 EW vs each of the three groups)].

    (TIFF)

    pone.0324105.s001.tiff (1.8MB, tiff)
    S2 Fig. Sensitivity of CD4+ T cells from RagD10 and Rag23−3 mice to stimulation by transforming growth factor beta 1 (TGF-β1).

    Spleen (SPL) and mesenteric lymph nodes (mLN) were harvested from untreated RagD10 or Rag23−3 mice, and CD4+ T cells were isolated by magnetic cell separation system. The cells were then stimulated with plate-bound anti-CD3 and anti-CD28 monoclonal antibodies and cultured in the presence of the indicated amounts of TGF-β1, retinoic acid (1 µM), and recombinant IL-2 (2 ng/mL) for 48 h, and the frequency of regulatory T cells (Foxp3+) within the CD4+ T cell population was determined. Each circle indicates the value for an individual well, and the horizontal lines indicate mean values (n = 3, mixture of cells from three mice/group). Data are representative of two independent experiments. Statistical analysis: Tukey’s HSD test [(#p < 0.05 (between different strains); * p < 0.05, ** p < 0.01, and *** p < 0.001 (between different groups in each strain)].

    (TIFF)

    pone.0324105.s002.tiff (341.2KB, tiff)
    S3 Fig. Cytokine profiles for the supernatants of CD4+ T cells isolated from the spleen of RagD10 mice and Rag23−3 mice and cultured under regulatory T cell (Treg) polarization conditions.

    RagD10 and Rag23−3 mice were fed a diet containing egg white (EW) or casein (CN; control) for 7 days and spleens were harvested; CD4+ T cells were isolated, stimulated with plate-bound anti-CD3 and anti-CD28 monoclonal antibodies, and cultured under Treg polarization conditions for 48 h, and the culture supernatants were collected. The concentrations of interleukin (IL)-2, IL-4, and interferon gamma (IFN-γ) in the supernatants were determined by enzyme-linked immunosorbent assay (n = 3, mixture of cells from three mice/group). Data are representative of two independent experiments. Statistical analysis: Tukey’s HSD test [#p < 0.05 (CN vs EW in each strain); * p < 0.05, ** p < 0.01, and *** p < 0.001 (between different groups)]. N.D. = not detected.

    (TIFF)

    pone.0324105.s003.tiff (103.9KB, tiff)
    S4 Fig. Expression of interferon-gamma (IFN-γ) or interleukin (IL)-4 receptor on regulatory T cells differentiated from untreated RagD10 mice is regulated by excessive IFN-γ or IL-4.

    A) Frequency of CD25+ cells in regulatory T cells (Tregs; Foxp3+CD4+ T cells) differentiated from splenocytes of untreated-RagD10 mice cultured under Treg-polarization culture conditions described in the caption to Fig 3B. B) Gating strategy for identifying IFN-γ receptor (IFN-γR+) or IL-4 receptor (IL-4R+) on CD4+ T cells (left) and the frequencies of each receptor expression cells in CD4+ T cells (right). Each plot indicates the value for an individual well and horizontal lines indicate mean values (n = 3, mixture of cells from three mice/group). C) Frequency of CD25+ cells in Tregs from spleen cells of untreated RagD10 mice cultured under the Treg-polarization culture conditions indicated in Fig 3D and 3E. D) Frequencies of IFN-γR+ or IL-4R+ on CD4+ cells supplemented with rIL-4 (left: 0, 0.5, 1.5, 2.5 ng/mL) or rIFN-γ (right: 0, 1.0, 2.0, 3.5 ng/mL). Error bars indicate means ± SD (n= 4, mixture of cells from two to three mice/group). Analysis: Tukey’s HSD test {#p < 0.05 [IFN-γ (0 ng/mL)] vs [IFN-γ (3.5 ng/mL)] or [IL-4 (0 ng/mL)] vs [IL-4 (2.5 ng/mL)] in Fig S4C and S4D; * p < 0.05, ** p < 0.01, and *** p < 0.001 (between different groups)}.

    (TIFF)

    pone.0324105.s004.tiff (1.1MB, tiff)
    S5 Fig. Effector/effector memory T cells within the regulatory T cell population show enhanced CD137 expression.

    Naïve-like CD4+ T cells (CD44loCD62Lhi) and effector/effector memory CD4+ T cells (EMT; CD44hiCD62Llo) from Rag23−3 mice fed a diet containing egg white (EW) or casein (CN; control) for 7 days were stimulated with plate-bound anti-CD3 and anti-CD28 monoclonal antibodies and cultured under Treg-polarization culture conditions for 72 h. A) Gating strategy used to identify the CD137+ subpopulation within the Foxp3+ CD4+ and CD44hiCD62LloFoxp3+CD4+ populations. B) Frequency of CD137+ cells within the Foxp3+CD4+ and CD44hiCD62LloFoxp3+CD4+ T cell populations. C) Frequency of CD44hiCD62Llo cells within the Foxp3+CD4+ T cell population. In panels 5B and 5C, plots indicate the values for individual wells and horizontal lines indicate mean values (CN_naive: n = 4, EW_naive: n = 4, EW_EMT: n = 4, using a mixture of cells from 6 (CN) or 3 (EW) mice/group). Data are representative of two independent experiments. Statistical analysis: Tukey’s HSD test [*p < 0.05, ** p < 0.01, and *** p < 0.001 (between different groups)].

    (TIFF)

    pone.0324105.s005.tiff (1.5MB, tiff)
    Attachment

    Submitted filename: Responses to Commentsfinal.pdf

    pone.0324105.s007.pdf (219.9KB, pdf)
    Attachment

    Submitted filename: Responses to Reviewers#2.docx

    pone.0324105.s008.docx (40.1KB, docx)

    Data Availability Statement

    All Excel files containing the data underlying the results presented in our study (Fig 1~Fig 5 and S1 Fig ~ S5 Fig) are available at the URL address of UTokyo Repository (http://hdl.handle.net/2261/0002013444).


    Articles from PLOS One are provided here courtesy of PLOS

    RESOURCES