Skip to main content
Advertisement
Browse Subject Areas
?

Click through the PLOS taxonomy to find articles in your field.

For more information about PLOS Subject Areas, click here.

  • Loading metrics

Enhancement of Oral Tolerance Induction in DO11.10 Mice by Lactobacillus gasseri OLL2809 via Increase of Effector Regulatory T Cells

  • Ayako Aoki-Yoshida,

    Affiliations Department of Applied Biological Chemistry, The University of Tokyo, Yayoi, Bunkyo-ku, Tokyo, Japan, National Institute of Livestock and Grassland Science, National Agriculture and Food Research Organization, Tsukuba, Ibaraki, Japan

  • Kiyoshi Yamada,

    Affiliation Department of Applied Biological Chemistry, The University of Tokyo, Yayoi, Bunkyo-ku, Tokyo, Japan

  • Satoshi Hachimura,

    Affiliation Research Center for Food Safety, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo, Japan

  • Toshihiro Sashihara,

    Affiliation Division of Research and Development, Meiji Co. Ltd., Odawara, Kanagawa, Japan

  • Shuji Ikegami,

    Affiliation Division of Research and Development, Meiji Co. Ltd., Odawara, Kanagawa, Japan

  • Makoto Shimizu,

    Affiliation Department of Applied Biological Chemistry, The University of Tokyo, Yayoi, Bunkyo-ku, Tokyo, Japan

  • Mamoru Totsuka

    atotuka@mail.ecc.u-tokyo.ac.jp

    Affiliation Department of Applied Biological Chemistry, The University of Tokyo, Yayoi, Bunkyo-ku, Tokyo, Japan

Abstract

Food allergy is a serious problem for infants and young children. Induction of antigen-specific oral tolerance is one therapeutic strategy. Enhancement of oral tolerance induction by diet is a promising strategy to prevent food allergy in infants. Thus, in this study, we evaluate the effect of probiotic Lactobacillus gasseri OLL2809 (LG2809) on oral tolerance induction in a mouse model. The degree of oral tolerance induction was evaluated by measuring the proliferation and level of IL-2 production of splenic CD4+ T cells from DO11.10 mice fed ovalbumin (OVA) alone or OVA with LG2809. Oral administration of LG2809 significantly decreased the rate of proliferation and IL-2 production by CD4+ T cells from OVA-fed mice. LG2809 increased a ratio of CD4+ T-cell population, producing high levels of IL-10 and having strong suppressive activity. Moreover, LG2809 increased a ratio of plasmacytoid dendritic cells (pDCs) among the lamina propria (LP) in small intestine. When used as antigen presenting cells to naïve CD4+ T cells from DO11.10 mice, LP cells from BALB/c mice fed LG2809 induced higher IL-10 production and stronger suppressive activity than those from non-treated mice. These results suggest that oral administration of LG2809 increases the population of pDCs in the LP, resulting in the enhancement of oral tolerance induction by increasing the ratio of effector regulatory T cells. LG2809 could, therefore, act as a potent immunomodulator to prevent food allergies by promoting oral tolerance.

Introduction

Probiotics were defined as “live microorganisms which, when administered in adequate amounts, confer a health benefit to the host” by Food and Agricultural Organization of the United Nations /World Health Organization [1]. A growing body of evidence is accumulating to show that administration of probiotics modulate intestinal immunity, improve the balance of the gut microbiota, enhance the recovery of a disturbed gut mucosal barrier, and prevent microbial translocation [2, 3]. Lactobacillus gasseri OLL2809 (LG2809) is a probiotics that can reduce serum antigen-specific IgE levels in mice, and reduce the symptoms of Japanese cedar pollinosis [47]. We have previously shown in vitro that LG2809 suppresses proliferation of CD4+ T cells through a myeloid differentiation primary response gene 88 (MyD88)-dependent signaling pathway and that its RNA suppresses the delayed-type hypersensitivity response in vivo [8]. Hence, LG2809 is likely to have the potential to modulate various immune responses.

In recent years, food allergy has become a serious problem in infants and young children. The general treatment is to remove food allergens from the diet [9]. However, because milk and egg, the most frequent allergens in most countries, are nutritionally important sources of dietary proteins, especially for infants, removal of allergenic foods leads to an increased risk of undernutrition [10]. In addition, the developmental progression of allergic disease during early childhood is often known as the atopic march [11]. Therefore, it is beneficial for infants to achieve an early remission from food allergy.

Oral tolerance is the antigen-specific immune hyporesponsiveness to protein antigens repeatedly administered by the oral route [12]. Induction of antigen-specific oral tolerance is a promising strategy for treating food allergy [13]. Thus, it would be useful to enhance oral tolerance induction for an early remission from or to prevent food allergy in infants. Oral tolerance is mediated by multiple mechanisms, such as anergy, clonal deletion, and regulatory T-cell induction [14]. Antigen-specific T-cell anergy by oral tolerance induction was demonstrated by the transfer of T cells and B cells from orally tolerized mice into SCID mice [15]. The clonal deletion process occurs by apoptosis of antigen-specific CD4+ T cells [16], which in oral tolerance induction is mediated by signaling via Fas antigen and p55 tumor necrosis factor (TNF) receptor [17, 18]. Various regulatory T cells are induced by oral tolerance induction. Oral administration of myelin basic protein induces regulatory transforming growth factor (TGF)-β-secreting T cells in Peyer's patches of mice [19]. Oral tolerance induction in ovalbumin (OVA)-specific T-cell receptor (TCR) transgenic mice (DO11.10 mice) leads to an increase in regulatory T cells, and they produce high levels of IL-10 and exert suppressive activity [20]. There are several reports of dendritic cell (DC) involvement in the induction of oral tolerance and T-cell differentiation [2124]. DCs capture dietary antigens in the intestinal mucosa and present them to T cells. DCs are a heterogeneous population of leucocytes that act as professional antigen-presenting cells (APCs) [25]. In particular, DCs in the intestinal lamina propria (LP) have been shown to play an essential role in oral tolerance induction [22, 26, 27]. There are two classes of DCs, myeloid (mDC) and plasmacytoid (pDC), which are functionally different; they differ in cytokine/chemokine secretion, expression of cell surface markers, and T-cell-polarizing ability [18, 26, 2832]. Interestingly, recent studies have shown that nutrients and food antigens can alter DC phenotypes and behaviors [3335], suggesting that intestinal luminal contents are directly involved in modulating mucosal DC function.

The intestinal microbiota play important roles in oral tolerance induction and its long-term persistence [36, 37]. Several studies suggest that specific-pathogen free (SPF), but not germ-free mice, are susceptible to induction of oral tolerance [3840]. Colonization of gut bacteria can restore oral tolerance in germ-free mice, and the effect of probiotics is dependent on the bacterial strain [38, 39]. Lactobacillus casei potentiates the induction of oral tolerance and suppresses the T helper (Th)1-type immune responses of inflammation in a SPF-rat model of experimental arthritis [41]. Furthermore, probiotic treatment can alter DC phenotype and function. Feeding with the probiotic strain Lactobacillus paracasei subsp. paracasei NTU 101 [42] or with L. casei DN-114001-fermented milk [42, 43] results in the up-regulation of the antigen-presenting ability of DCs. In addition, administration of the probiotic nutrient supplement VSL#3 has been shown to increase the proportion of mDCs within the LP [27]. These findings suggest that probiotics would affect oral tolerance induction via alteration of the LP DC phenotype and its function. However, the effect of probiotics on oral tolerance remains largely unknown.

In the present study, we investigated the effect of oral administration of LG2809 on the induction of oral tolerance to OVA by examining the proliferative response, IL-2 production, and suppressive activity of splenic CD4+ T cells from DO11.10 mice. To elucidate the mechanism of the enhancement of oral tolerance induction by LG2809, the effect of LG2809 on DC responses in the LP was examined.

Materials and Methods

Mice

Female BALB/c mice (8–10 weeks old) were obtained from CLEA Japan (Tokyo, Japan). Female DO11.10 T-cell receptor transgenic mice (8–10 weeks old) were transgenic for OVA323–339-specific and I-Ad-restricted T-cell receptor αβ, with a BALB/c genetic background [44]. These mice were fed chow CE-2 (CLEA) ad libitum and housed in cages (6 mice per cage) maintained under a continuous 12 hour light: 12 hour dark cycle in conventional conditions at the Department of Applied Biological Chemistry, the University of Tokyo. Hair gloss of mice was checked daily to monitor their health. All of the mice used in this study were euthanized by cervical dislocation. All animal care and use protocols were conducted in accordance with the animal experimentation guidelines of the University of Tokyo. The research protocol was approved by the Animal Experimentation Committee of the University of Tokyo (Tokyo, Japan; permit number: P07-065, P08-242).

Reagents

The following antibodies were used: fluorescein isothiocyanate (FITC)-conjugated anti-mouse CD4 (clone H129.19), anti-DO11.10 clonotypic TCR (KJ-1.26), anti-mouse CD44 (clone IM7), peridinin chlorophyll protein (PerCP)-conjugated anti-mouse CD4 (clone RM4-5), allophycocyanin (APC)-conjugated anti-mouse CD4 (clone 145-2C11), biotinylated anti-mouse CD3ε (clone 145-2C11), biotinylated anti-mouse CD62L (clone MEL14), biotinylated anti-mouse CD103 (clone M290), biotinylated anti-mouse CD11b (clone M1/70), and streptavidin-phycoerythrin (PE)-cyanine (Cy)5 and PE-anti-mouse Foxp3 (clone FJK-16s) were purchased from BD Biosciences (San Diego, CA, USA). FITC-conjugated anti-mouse CD11c (clone N418), PE-anti mouse CD45R/B220 (clone RA3-6B2), and biotinylated anti-mouse Ly-6G (clone RB6-8C5) were purchased from eBioscience (San Diego, CA, USA).

Bacterial strain and preparation

LG2809 was provided by the Division of Research and Development, Meiji Co., Ltd. (Odawara, Japan). LG2809 was cultured in Lactobacilli MRS broth (DIFCO, Detroit, MI, USA) at 37°C for 18 hrs. Bacteria were harvested by centrifugation at 1,800 x g at 4°C for 15 mins, and then washed twice with 0.85% NaCl (saline). The cells were collected and suspended in saline at 5 mg dry weight/mL. The bacterial suspension was stored at -80°C until required.

Oral tolerance induction and treatment of mice with LG2809

DO11.10 mice were administered saline or live LG2809 (1 mg/day) by gavage for 12 days. For the last 5 days, the mice were fed a solution of 20% OVA (Wako; Albumin, from Eggs, 012–09885) ad libitum instead of sterile water. To examine the effect of LG2809 on the phenotype and function of LPDCs, BALB/c mice were orally administered LG2809 (1 mg/day) by gavage for 7 days.

Cell preparation and culture conditions

Cells were cultured in RPMI1640 (Nissui Pharmaceutical, Tokyo, Japan) containing 10% fetal calf serum (FCS; GIBCO, Grand Island, NY, USA), 2 g/L NaHCO3, 100 U/mL penicillin, 100 μg/mL streptomycin, 50 μM 2-mercaptoethanol, and 300 mg/L L-glutamine at 37°C in 5% CO2 in air.

Mouse spleen (SPL) CD4+ T cells were isolated as previously described [45] using magnetic activated-cell sorting (MACS) positive selection with CD4 microbeads (Miltenyi Biotec, Bergish Gladbach, Germany) according to the manufacturer’s instructions. CD4+ CD25- T cells were purified using a CD4+ CD25+ regulatory T-cell isolation kit (Miltenyi Biotec).

T-cell-depleted splenocytes were isolated by MACS negative selection using Thy-1.2 microbeads (Miltenyi Biotec), and the isolated cells were used as APCs.

Preparation of FACS-sorted cells from DO11.10 mice was performed as follows: MACS- sorted CD4+ T cells were washed with PBS. Then the cells (2–4 x 107 cells/mL) were incubated with biotinylated anti-CD62L antibody in PBS/1%FCS buffer for 20 mins on ice. After the cells were washed, they were incubated with anti-KJ-1.26-FITC, PE-conjugated anti-CD44 antibody, and streptavidin-PE-Cy5 for 20 mins on ice. KJ-1.26+ CD62Lhigh CD44high and KJ-1.26+ CD62Llow CD44high T cells were isolated from CD4+ T cells of LG2809 and OVA-fed mice using a FACS Vantage SE (BD Biosciences). The purity of the sorted populations was routinely >95%.

LP cells from BALB/c mice were isolated from the small intestines as follows: Peyer’s patches were removed from the small intestines to prevent their lymphocyte contamination in LP cells. After cutting open the intestines longitudinally, they were cut into pieces of approximately 4 cm and washed with Ca- and Mg-free Hanks’ balanced salt solution (HBSS; GIBCO) containing 0.04% NaHCO3, 5 mM EDTA (pH 8.0) and 5% FCS with shaking at 37°C in 5% CO2 in air for 20 mins, three times. The supernatant was discarded following filtration with gauze, and the intestines were minced into 5 mm pieces and treated with 1 mg/mL collagenase IV (Sigma-Aldrich, St Louis, MO, USA) in HBSS containing Ca, Mg, and 5% FCS (HBSS (+)) in a 100 mL flask with gentle stirring at 37°C in 5% CO2 in air for 60 mins. After collagenase treatment, the preparation was filtered with gauze and the cells were washed with HBSS (+) followed by centrifugation at 25°C, 300 x g for 5 mins. The supernatant was then removed and 3 mL of 100% Percoll (Amersham Biosciences, Uppsala, Sweden) was added to the cell pellets and made up to 10 mL with HBSS (+) (a 30% Percoll concentration). The cell suspension was mixed gently and centrifuged at 25°C, 580 x g for 20 mins. All but 1 mL of the supernatant was then removed, the cells were resuspended, and 4.1 mL of 100% Percoll and 10 mL of RPMI containing 10% FCS was added (a 44% Percoll concentration). Next, 2 mL of 70% Percoll was carefully added beneath the cell suspension and centrifuged at 20°C, 580 x g for 20 mins. Finally, the cells located at the interface between the 44% and 70% Percoll fractions were collected as LP cells.

CD3- cells among the LP cells were isolated using a MACS LD column (Miltenyi Biotec). The CD3-negative fraction was used as an APC-enriched fraction that included DCs. SPL CD4+ CD25- T cells from DO11.10 mice were stimulated with OVA323–339 and CD3- LP cells, and the activated CD4+ T cells were purified using MACS positive selection with CD4 microbeads.

CD4+ T-cell proliferation assay

After oral administration of LG2809, SPL cells were obtained from DO11.10 mice in each group. Isolated CD4+ T cells (1 x 105 cells/well) and APCs (1 x 105 cells/well) were cultured with OVA323–339 (1 μM) in 96-well flat-bottomed plates at 37°C in 5% CO2 in air. Proliferation of the CD4+ T cells was evaluated by measuring incorporation of 3H-thymidine (37 kBq/ well) (ICN Pharmaceuticals, Costa Mesa, CA, USA) added into the culture during the final 24 hrs of incubation.

Cytokine analysis

Isolated CD4+ T cells (1 x 105 cells/well) and APCs (1 × 105 cells/well) were cultured with OVA323–339 (1 μM) in U-bottomed 96-well plates at 37°C in 5% CO2 in air. After incubation for 2 days, culture supernatants were collected.

Determination of the IL-2 level in the culture supernatants was performed by a sandwich enzyme-linked immunosorbent assay (ELISA), as previously described [46]. Briefly, anti-mouse IL-2 (clone JES6-1A12, BD PharMingen, San Diego, CA, USA) was coated on ELISA plates. After washing and blocking the plates, samples and standards were added. After washing, biotinylated anti-mouse IL-2 (clone JES6-5H4, BD PharMingen) was added. The wells were washed, and streptavidin-conjugated alkaline phosphatase (Zymed, South San Francisco, CA, USA) was added. The wells were washed and incubated with disodium 4-nitrophenylphosphate hexahydrate solution. Optical densities were read at 405 nm on a BIO-RAD Model 550 Microplate Reader (Bio-Rad, Hercules, CA, USA). The IL-10 level in the culture supernatants was measured by sandwich ELISA using Mouse Opt EIA kits (BD Biosciences), as described previously [47].

Flow cytometric analysis

Flow cytometric analysis was performed using a FACS LSR with CellQuest software (BD Biosciences). The cells were harvested and washed with PBS containing 1% fetal calf serum and 0.1% NaN3 (FACS buffer). The cells were incubated with anti-CD16/CD32 monoclonal antibody (mAb; clone 2.4G2, BD PharMingen) on ice to block non-specific binding to Fc receptors. Then, the cells were stained with the appropriate PerCP-, FITC-, or PE-conjugated antibodies or biotinylated antibody for 30 mins on ice. When stained with the biotinylated antibody, the cells were further stained with streptavidin-APC conjugated antibody for 30 mins on ice. The cells were analyzed by flow cytometry.

For apoptotic analysis, the cells were washed in FACS buffer and incubated with anti-CD16/CD32 mAb (BD PharMingen) on ice. The cells were then stained with biotinylated CD4 antibodies for 30 mins on ice, followed by staining with anti-KJ-1.26-FITC (BD PharMingen) and anti-CD4-APC (BD PharMingen) for 30 mins on ice. Cells were further stained using an Annexin V-PE Apoptosis Detection kit I (BD Biosciences) and analyzed.

For intracellular analysis of the expression of Foxp3, the cells were fixed with Fixation/Permeabilization Concentrate (eBioscience) and Fixation/Permeabilization Diluent (eBioscience) overnight at 4°C. The cells were washed with Permeabilization Buffer (eBioscience) and incubated with anti-CD16/CD32 mAb (BD PharMingen) on ice. Then the cells were stained with anti-Foxp3-PE mAb (clone FJK-16s; BD Biosciences) for 30 min on ice, and the cells were subjected to the flow cytometric analysis.

Assays for T-cell suppressive activity

To determine the T-cell suppressive activity of the groups of splenic CD4+ T cells, i.e. CD62Lhigh CD44high CD4+ T cells and CD62Llow CD44high CD4+ T cells from DO11.10 mice fed OVA and LG2809, and the CD4+ T cells differentiated in vitro through activation with antigen-presentation by CD3- LP cells, the ability of these groups of T cells to inhibit IL-2 production by responder SPL CD4+ CD25- T cells was measured. CD4+ T cells (1 x 105 cells, 5 x 104 cells, or 2.5 x 104 cells/well) were co-cultured with CD4+ CD25- T cells from untreated DO11.10 mice (5 x 104 cells/well), APCs (1 x 105 cells/well), and 0.3 μM OVA323–339 in 96-well U-bottomed plates at 37°C in 5% CO2 in air. Then the amount of IL-2 in the culture supernatants was analyzed by ELISA.

Statistical analysis

All experimental data were expressed as the mean ± standard deviation (SD). Statistical differences were analyzed by Student’s t-tests.

Results

Oral administration of live LG2809 enhanced oral tolerance induction in DO11.10 mice

Female DO11.10 mice were administered 1 mg of live LG2809 in saline or saline alone by gavage every day for 12 days. The mice were fed 20% OVA in drinking water ad libitum for the last 5 days. Consistent with a phenotype of oral tolerance, the proliferation rate and level of IL-2 secretion were lower in SPL T cells from OVA-fed mice (saline/OVA group) than in T cells from untreated mice (saline/water group; Fig 1A and 1B). Oral administration of live LG2809 and OVA (LG2809/OVA group) further decreased proliferation and IL-2 secretion to levels significantly below those of the saline/OVA group, indicating that oral administration of LG2809 enhanced oral tolerance induction. Clonal deletion of antigen-specific CD4+ T cells is known to occur with oral tolerance induction [16]. Thus, we examined whether oral administration of LG2809 affected apoptosis of OVA-specific T cells. OVA-specific T cells from DO11.10 mice can be detected with the KJ-1.26 mAb specific for the transgenic TCR. The ratio of CD4+ KJ-1.26+ T cells among SPL cells and the ratio of viable cells (annexin V- 7-AAD-) among CD4+ KJ-1.26+ T cells from DO11.10 mice fed OVA were lower compared to those found in DO11.10 mice fed no OVA, and the ratio of annexin V+ cells among CD4+ KJ-1.26+ cells from the saline/OVA group was higher compared to those from saline/water group. However, there was no significant difference between the groups fed or not fed LG2809 (Fig 1C–1E). These results suggest that clonal deletion of antigen-specific T cells is not responsible for the decrease in proliferation and IL-2 production by SPL CD4+ T cells induced by feeding LG2809. Since it has been reported that Tregs play a crucial role for the induction of oral tolerance [19], and thus we examined the effect of LG2809 on the induction of Foxp3+ Treg. Although the ratio of Foxp3+ cells among CD4+ KJ-1.26+ T cells was increased by oral tolerance induction, no effect was observed by the administration of LG2809 (Fig 1F).

thumbnail
Fig 1. Oral administration of live LG2809 enhanced oral tolerance induction in DO11.10 mice via non-apoptotic pathways.

DO11.10 mice were fed live LG2809 (1 mg/day; treated) or 0.85% NaCl (saline; untreated) for 12 days. For the last 5 days, the mice did (oral tolerance group) or did not (control group) have OVA added to the drinking water. According to feeding regimes, the groups were as follows: saline/water (untreated control group), saline/OVA (untreated oral tolerance group), LG2809/water (treated control group), and LG2809/OVA (treated oral tolerance group). SPL CD4+ T cells from each group were cultured with APCs and 1 μM OVA323–339. (A) The cultures were pulsed with [3H]-thymidine for the last 24 hrs of the 96 hrs culture period and [3H]-thymidine incorporation was measured. (B) IL-2 in the supernatant after 48 hrs in culture was measured by ELISA. (C, D and E) Whole SPLs from DO11.10 mice of each group were stained with anti-CD4-PerCP, anti- KJ-1.26-FITC, and annexin V-PE. The ratio of apoptotic cells was determined by flow cytometric analysis. The ratio of CD4+ KJ-1.26+ cells (C), of live (CD4+ KJ-1.26+ annexin V- 7-AAD-) cells (D), of CD4+ KJ-1.26+ annexin V+ apoptotic cells (E) and CD4+ KJ-1.26+ Foxp3+ cells (F) is shown for each group. Data are shown as the means ± SD (n = 6). Data are representative of two independent experiments. Statistical differences were analyzed by Student’s t-test and were considered significant (*) when p was <0.05. n.s., not significant.

https://doi.org/10.1371/journal.pone.0158643.g001

Oral administration of live LG2809 enhanced IL-10 secretion and T-cell suppressive activity in splenocytes from orally tolerized mice

Since IL-10-secreting T cells are induced in the SPL of orally tolerized mice [20, 48], IL-10 secretion in the culture medium of the SPL CD4+ T cells from the different groups was examined. Consistent with previous reports, the level of IL-10 secretion by orally tolerized T cells was higher than that of the control cells. Furthermore, oral administration of LG2809 significantly enhanced IL-10 secretion from the T cells of OVA-fed mice (Fig 2A). Next, we examined the effect of LG2809 on suppressive activity of CD4+ T cells from OVA-fed DO11.10 mice. SPL CD4+ T cells from each test group of mice, responder CD4+ CD25- T cells from untreated DO11.10 mice, and APCs were cultured in the presence of OVA323–339, and then the levels of IL-2 secretion were examined. As shown in Fig 2B, SPL CD4+ T cells from the saline/OVA group caused a decrease in the level of IL-2 secretion from the responder CD4+ CD25- T cells. Moreover, feeding LG2809 significantly enhanced the suppressive effect of the T cells on IL-2 secretion.

thumbnail
Fig 2. LG2809 enhanced IL-10 secretion and T-cell suppressive activity in splenocytes from orally tolerized mice.

DO11.10 mice were treated as described in the legend to Fig 1. (A) SPL CD4+ T cells from DO11.10 mice of each group were cultured with APCs and 1 μM OVA323–339. IL-10 in the supernatant after 48 hrs in culture was measured by ELISA. (B) SPL CD4+ T cells from DO11.10 mice of the saline/OVA or LG2809/OVA groups (effector cells) were cultured with APCs, 1 μM OVA323–339, and responder DO11.10 CD4+ CD25- T cells at the ratio indicated in the figure. IL-2 in the supernatant was measured by ELISA. Data are shown as means ± SD. (n = 6). Data are representative of two independent experiments. Statistical differences were analyzed by Student’s t-test. *p<0.05 and **p<0.01, significantly different vs. saline/OVA group; ##p<0.01 and ###p<0.001, significantly different vs. responder DO11.10 CD4+ CD25- T cells alone.

https://doi.org/10.1371/journal.pone.0158643.g002

Oral administration of live LG2809 increased the ratio of CD62Llow CD44high CD4+ T cells with IL-10-producing and suppressive activities

T cells can be divided into three phenotypes (naïve T cells, memory T cells, and effector T cells) according to the expression of CD44 and CD62L molecules, and therefore to further analyze the subsets involved in tolerance induction, we examined the expression of CD44 and CD62L in CD4+ KJ-1.26+ T cells from LG2809 and OVA-fed mice by flow cytometry. The results indicated that the ratio of CD62Llow CD44high CD4+ T cells, considered to be the effector phenotype, was higher among SPL T cells from OVA-fed mice, compared to those from untreated mice. The ratio of this subset was significantly increased in the LG2809/OVA group compared to that in the saline/OVA group (Fig 3A and 3B). To elucidate the relationship between the effector T cells and IL-10 production, the ratio of KJ-1.26+ CD62Llow CD44high cells among CD4+ T cells was plotted against IL-10 concentration in the culture supernatant of CD4+ T cells. We found a positive correlation between the ratio of KJ-1.26+ CD62Llow CD44high cells among CD4+ T cells and the IL-10 concentration in the culture supernatant of CD4+ T cells in both saline/OVA and LG2809/OVA groups (Fig 3C). To assess whether CD62Llow CD44high CD4+ T cells produce IL-10 and have suppressive activity, IL-10 production and suppressive activity of sorted CD62Llow CD44high CD4+ T cells and CD62Lhigh CD44high CD4+ T cells isolated from DO11.10 mice of LG2809/OVA group were also investigated. Cultured sorted CD62Llow CD44high CD4+ T cells secreted high concentrations of IL-10 into the supernatant (Fig 3D). Furthermore, sorted CD62Llow CD44high T cells had stronger suppressive activity against co-cultured responder T cells than CD62Lhigh CD44high CD4+ T cells (Fig 3E). Proliferative response of the sorted cells was also examined. CD62Llow CD44high CD4+ T cells and CD62Lhigh CD44high CD4+ T cells proliferated much more weakly than CD4+ CD25- T cells, indicating that they were both anergic cells (Fig 3F). These results suggest that the enhanced IL-10 production and suppressive activity of SPL CD4+ T cells from LG2809-fed orally tolerized mice is caused by an increase in the ratio of CD62Llow CD44high CD4+ T cells.

thumbnail
Fig 3. LG2809 increased the ratio of CD62Llow CD44high CD4+ T cells with IL-10-producing and suppressive activities.

DO11.10 mice were treated as described in the legend to Fig 1. (A and B) Whole SPLs from DO11.10 mice of each group were stained with anti-CD4-PerCP, KJ-1.26-FITC, anti-CD44-PE, and anti-CD62L-APC. The expression of CD44 and CD62L on CD4+ KJ-1.26+ T cells was analyzed by flow cytometric analysis. The mean ratio of CD62Llow CD44high cells in CD4+ KJ-1.26+ T cells is shown (B). Data are shown as means ± SD (n = 6). Data are representative of two independent experiments. Statistical differences were analyzed by Student’s t-test. *p<0.05. (C) The correlation between the concentration of IL-10 in the culture supernatant of CD4+ T cells shown in Fig 1C and the ratio of KJ-1.26+ CD62Llow CD44high cells among CD4+ T cells. Closed circles, LG2809/OVA group; open circles, LG2809/water group; closed triangles, saline/OVA group; open triangles, saline/water group. The continuous line is a linear regression for LG2809/OVA group and the dashed line is a linear regression for saline/OVA group. R, Pearson correlation coefficient. Data are representative of two independent experiments. (D) CD62Lhigh CD44high and CD62Llow CD44high cells sorted from CD4+ T cells isolated from DO11.10 mice of the LG2809/OVA group were cultured with APCs and 0.3 μM OVA323–339. After 48 hrs, IL-10 in the supernatant was measured by ELISA. (E) The sorted CD62Lhigh CD44high and CD62Llow CD44high CD4+ T cells from DO11.10 mice of the LG2809/OVA group were incubated with responder CD4+ T cells from DO11.10 SPL (at a ratio of 1:1), plus APCs and 0.3 μM OVA323–339. IL-2 in the supernatant was measured by ELISA. (F) The sorted CD62Lhigh CD44high and CD62Llow CD44high CD4+ T cells from DO11.10 mice of the saline/OVA, LG2809/OVA group, and CD4+ CD25- T cells were incubated with APCs and 0.3 μM OVA323-339. The cultures were pulsed with [3H]-thymidine for the last 24 hrs of the 96 hrs culture periods and [3H]-thymidine incorporation was measured. Data are representative of two independent experiments.

https://doi.org/10.1371/journal.pone.0158643.g003

Next, we plotted the ratio of KJ-1.26+ CD62Llow Foxp3+ cells among CD4+ T cells or that of KJ-1.26+ CD62Llow Foxp3+ cells against IL-10 concentration in the culture supernatant of each population of CD4+ T cells, respectively (S1 Fig). Interestingly, we found very good positive correlation between the ratio of KJ-1.26+ CD62Llow Foxp3- cells among CD4+ T cells and the IL-10 concentration in the culture supernatant of CD4+ T cells in LG2809/OVA group. It is known that not only Foxp3 Tregs but also Foxp3 negative T regulatory type 1 (Tr1) cells are induced in oral tolerance [49]. Our results suggested that LG2809 might increase Tr1 cells but not Foxp3 Tregs in orally tolerized mice.

Oral administration of live LG2809 increased the population of pDCs in the LP

The two classes of functionally different DCs, mDC and pDC, are defined by the surface expression of CD11c, CD11b and CD103, and CD11c, B220 and Ly-6G, respectively [27, 30, 31, 50, 51]. DCs in the LP play important roles in oral tolerance induction [22, 26, 27], and nutrients and food antigens can alter the DC phenotype [3335]. To investigate whether oral administration of LG2809 affects DCs in the LP, we examined the expression of DC surface molecules by LP cells of BALB/c mice fed live LG2809. BALB/c mice were fed LG2809 or saline for 7 days, and CD3- LP cells were then isolated and analyzed for the expression of CD11c, B220, Ly-6G, CD11b and CD103 by flow cytometry (Fig 4). Feeding LG2809 significantly increased the ratio of CD11c+ B220+ and CD11c+ Ly-6G+ cells among the CD3- LP cells (Fig 4A and 4B). On the other hand, LG2809 had no effect on the ratio of CD11c+ CD11b+ cells (Fig 4C) but significantly reduced the ratio of CD11c+ CD103+ cells among the CD3- LP cells (Fig 4D). pDCs express CD11c, B220 and Ly-6G, and mDCs express CD11c, CD11b and CD103. Thus, the oral administration of LG2809 had led to an increase in the ratio of pDCs in the LP.

thumbnail
Fig 4. LG2809 enhanced plasmacytoid dendritic cells in the lamina propria.

BALB/c mice were fed live LG2809 (1 mg/day) or saline for 7 days. The ratio of CD11c+ B220+ (A), CD11c+ Ly-6G+ (B), CD11c+ CD11b+ (C), and CD11c+ CD103+ (D) cells among CD3- LP cells was determined by flow cytometric analysis. Data are shown as the means ± SD. (n = 4). Data are representative of two independent experiments. Statistical differences were analyzed by Student’s t-test. *p<0.05. n.s., not significant.

https://doi.org/10.1371/journal.pone.0158643.g004

Regulatory CD4+ T cells differentiated through activation with cells from the LP of mice fed LG2809 had stronger T-cell suppressive activity

We next investigated whether LG2809 administration altered the antigen-presenting function of the LP cells. CD3- LP cells were prepared from BALB/c mice administered saline or LG2809 for 7 days. SPL CD4+ CD25- T cells from DO11.10 were stimulated with OVA323–339 and CD3- LP cells as APCs. The CD4+ T cells activated with the antigen and CD3- LP cells from mice fed LG2809 had significantly stronger suppressive activity (Fig 5A). In addition, the differentiated CD4+ T cells through activation with CD3- LP cells from LG2809-fed mice produced marginally higher levels of IL-10 (Fig 5B), but no difference was found in the ratio of Foxp3+ cells (data not shown). These results suggest that LG2809 had altered the antigen-presenting function of LP cells and enhanced the suppressive activity of differentiated T cells.

thumbnail
Fig 5. CD4+ T cells differentiated with lamina propria APCs from mice fed LG2809 had stronger suppressive activity.

(A and B) BALB/c mice were fed live LG2809 (1 mg/day) or saline for 7 days. CD4+ CD25- T cells from DO11.10 mice were cultured with 1 μM OVA323–339 and CD3- LP cells from BALB/c mice fed LG2809 or saline as APCs. After 72 hrs, the CD4+ T cells were purified by MACS. The activated CD4+ T cells were incubated with responder CD4+ CD25- T cells from DO11.10 SPL cells (at ratios of 0.4:1 and 1:1), plus APCs and 1 μM OVA323–339. After 48 hrs, IL-2 in the supernatant was measured by ELISA (A). The activated CD4+ T cells were incubated with APCs and 1 μM OVA323–339. After 48 hrs, IL-10 in the supernatant was measured by ELISA (B). Data are shown as means ± SD (n = 4). Data are representative of two independent experiments. Statistical differences were analyzed by Student’s t-test. *p<0.05, significantly different vs. saline group; n.s., not significant.; ###p<0.001, significantly different vs. responder DO11.10 CD4+ CD25- T cells alone.

https://doi.org/10.1371/journal.pone.0158643.g005

Discussion

Intestinal microbiota have been proved to play critical roles in the induction of oral tolerance [52, 53]. However, little is known about the effect of orally administered probiotic bacteria on the induction of oral tolerance. In this study, we investigated the effect of oral administration of live LG2809 in a mouse model of oral tolerance induction. We found that LG2809 enhanced the suppression of proliferation and IL-2 production of SPL CD4+ T cells from OVA-fed DO11.10 mice. LG2809 increased the population of CD62Llow CD44high CD4+ T cells, which was found to be anergic, to secrete a large amount of IL-10 and to have suppressive activity. Moreover, LG2809 increased the ratio of pDCs in the LP and enhanced their ability to induce regulatory T cells. These results suggested that LG2809 increased the population of pDCs in the LP and enhanced the induction of oral tolerance by increasing the ratio of CD62Llow CD44high CD4+ T cells. To our knowledge, this is the first report to show how probiotics enhance the induction of oral tolerance.

The effect of probiotics on oral tolerance induction has been shown with L. casei. So et al. showed in a rat model of experimental arthritis that oral administration of L. casei suppresses arthritic inflammation by increasing the ratio of Foxp3+ CD4+ T cells, and increasing the secretion of IL-10 and TGF-β by CD4+ T cells [41]. We found an analogous increase in IL-10 production by the T cells but we did not observe an increase in the ratio of Foxp3+ T cells by oral administration of LG2809 in our experimental system. Moreover, LG2809 did not enhance TGF-β production (data not shown). Although it is difficult to compare the results because of differences in experimental conditions, we suggest that LG2809 might induce types of IL-10-producing Tr1 cells that are different from those induced by L. casei.

Oral administration of LG2809 increased the ratio of T cells with a CD62Llow CD44high CD4+ effector phenotype. CD44 is a hyaluronan receptor and CD62L is L-selectin. Previous reports show that the expression of CD44 increases and the expression of CD62L decreases when CD4+ T cells are stimulated with antigen [54, 55]. Therefore, the loss of expression of CD62L is known as a marker of activation in CD4+ T cells. Taking these and our results into consideration, we suggest that feeding LG2809 enhances oral tolerance by promoting the proliferation and activation of antigen-specific CD4+ T cells.

In this study, we found that administering LG2809 led to an increase in the ratio of pDCs in the LP. Wittman et al. reported that feeding of Bifidobacterium adolescentis protected the infection of Yersinia enterocolitica probably by increase of pDC in intestine [56]. They discussed that the increase of pDCs by administration of B. adolescentis might depend on invariant NKT cells (iNKT). iNKT are responsible for the recruitment of pDCs to the pancreas during lymphocytic choriomeningitis virus infection [57]. Recently, it was reported that iNKT cells recognize glycolipid antigens from Gram-positive bacteria presented by CD1d [58]. The release of glycolipids by LG2809 might result in the activation of iNKT cells by which pDCs might be recruited to the intestine. Wang et al. reported that feeding mice the probiotic preparation VSL#3, which contains 8 strains of live bacteria namely Bifidobacterium breve, Bifidobacterium longum, Bifidobacterium infantis, Lactobacillus acidophilus, Lactobacillus plantarum, Lactobacillus paracasei, Lactobacillus bulgaricus and Streptococcus thermophilus, alters the population of DCs in the LP [27], but they observed a decrease in pDCs in the LP and found that the ratio of mDCs in the LP was greater in VSL#3-fed mice than in the controls. This result is the inverse of ours, suggesting that the effect of probiotics on the induction of LP DCs depends on the probiotic strains used. In addition, we found that regulatory CD4+ T cells differentiated through antigen presentation by LP cells from LG2809-administered mice produced a higher level of IL-10 and had stronger suppressive activity. Since LG2809 increased the ratio of pDCs but not mDCs in the LP, the enhancement of IL-10 production and T-cell suppressive activity of the CD4+ T cells was probably caused by the increased ratio of pDCs. The finding that pDCs induced IL-10-producing regulatory CD4+ T cells [26] strongly supports this hypothesis. It remains to be elucidated, however, whether an increase in LP pDCs can increase the population of anergic, IL-10 producing, and suppressive CD62Llow CD44high CD4+ T cells in SPL.

Not only DC cells but also B cells can induce T cell tolerance via direct or indirect mechanisms [59, 60]. It is considered to be depending on experimental conditions whether B cells are essential for tolerance induction of T cells [59]. Oral tolerance of Th1 immune response was reported to be observed in B cell-depleted mice, suggesting that resting B cells are not essential for the induction of oral tolerance [61]. However, there are several studies showing B cells play a role in the oral tolerance induction [62]. In addition, chimeric mice specifically lacking IL-10 producing B cells were reported to develop an exacerbated collagen-induced arthritis and to have a reduction in IL-10 secreting Tr1 cells compared to wild type mice [63]. This suggests the importance of B cells in the differentiation and maintenance of Tr1 cells in vivo. It remains to be elucidated whether B cells are involved in the mechanisms by which LG2809 enhances the induction of oral tolerance.

Recently, the use of bispecific antibodies [6466] and antibody multimers [67], and the use of regulatory T cells, Foxp3 Tregs [68] and Tr1 cells [69], have come to be proposed as a new modality to prevent and treat autoimmune and inflammatory diseases. Especially in the treatment of inflammatory bowel disease, Tr1 cells are believed to be a very promising because they have the strong anti-inflammatory effect [70]. Induction of antigen-specific Tr1 cells in vivo is possible using oral tolerance as described above. In fact, even up to now, application of the oral tolerance has been attempted to treat the autoimmune and inflammatory disease [12]. Probiotics, which promotes the induction of Tr1 cells in oral tolerance induction, may be useful in treating autoimmune and inflammatory diseases using oral tolerance.

In conclusion, we demonstrate that feeding LG2809 enhanced oral tolerance induction and suggest that the effect of LG2809 is exerted via an increase of pDCs in the LP and an increase of effector regulatory CD4+ T cells. The enhancement of oral tolerance induction by feeding LG2809 may be beneficial for the prevention of or early remission from food allergy.

Supporting Information

S1 Fig. The ratio of KJ-1.26+ CD62Llow Foxp3- cells was correlated with IL-10 production in LG2809/OVA group.

DO11.10 mice were treated as described in the legend of Fig 1. The correlation between the concentration of IL-10 in the culture supernatant of CD4+ T cells from LG2809/OVA group and the ratio of KJ-1.26+ CD62Llow Foxp3- (A) and KJ-1.26+ CD62Llow Foxp3+ cells (B) among CD4+ T cells from LG2809/OVA group. R, Pearson correlation coefficient.

https://doi.org/10.1371/journal.pone.0158643.s001

(TIFF)

Acknowledgments

This work was supported by a Grant-in-Aid for Scientific Research (B) from the Japan Society for the Promotion of Science. The funders had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Author Contributions

  1. Conceived and designed the experiments: AA MT.
  2. Performed the experiments: AA KY SH TS SI MS MT.
  3. Analyzed the data: AA.
  4. Contributed reagents/materials/analysis tools: AA KY SH TS SI MS MT.
  5. Wrote the paper: AA MT.

References

  1. 1. FAO/WHO, editor Guidelines for the evaluation of probiotics in food. Report of a Joint FAO/WHO Working Group on Drafting Guidelines for the Evaluation of Probiotics in Food.2002 April 30, May 1; Ontario, Canada.
  2. 2. Cong Y, Konrad A, Iqbal N, Elson CO. Probiotics and immune regulation of inflammatory bowel diseases. Curr Drug Targets Inflamm Allergy. 2003;2(2):145–54. Epub 2003/10/17. pmid:14561167.
  3. 3. Isolauri E, Kirjavainen PV, Salminen S. Probiotics: a role in the treatment of intestinal infection and inflammation? Gut. 2002;50 Suppl 3:III54–9. Epub 2002/04/16. pmid:11953334; PubMed Central PMCID: PMC1867676.
  4. 4. Sashihara T, Sueki N, Ikegami S. An analysis of the effectiveness of heat-killed lactic acid bacteria in alleviating allergic diseases. J Dairy Sci. 2006;89(8):2846–55. Epub 2006/07/15. S0022-0302(06)72557-7 [pii] pmid:16840600.
  5. 5. Gotoh M, Sashihara T, Ikegami S, Yamaji T, Kino K, Orii N, et al. Efficacy of oral administration of a heat-killed Lactobacillus gasseri OLL2809 on patients of Japanese cedar pollinosis with high Japanese-cedar pollen-specific IgE. Biosci Biotechnol Biochem. 2009;73(9):1971–7. Epub 2009/09/08. JST.JSTAGE/bbb/90144 [pii]. pmid:19734682.
  6. 6. Sashihara T, Ikegami S, Sueki N, Yamaji T, Kino K, Taketomo N, et al. Oral administration of heat-killed Lactobacillus gasseri OLL2809 reduces cedar pollen antigen-induced peritoneal eosinophilia in Mice. Allergol Int. 2008;57(4):397–403. Epub 2008/10/24. 057040397 [pii] pmid:18946235.
  7. 7. Sashihara T, Sueki N, Furuichi K, Ikegami S. Effect of growth conditions of Lactobacillus gasseri OLL2809 on the immunostimulatory activity for production of interleukin-12 (p70) by murine splenocytes. Int J Food Microbiol. 2007;120(3):274–81. Epub 2007/10/16. S0168-1605(07)00509-0 [pii] pmid:17936392.
  8. 8. Yoshida A, Yamada K, Yamazaki Y, Sashihara T, Ikegami S, Shimizu M, et al. Lactobacillus gasseri OLL2809 and its RNA suppress proliferation of CD4(+) T cells through a MyD88-dependent signalling pathway. Immunology. 2011;133(4):442–51. Epub 2011/06/02. pmid:21627651; PubMed Central PMCID: PMCPMC3143356.
  9. 9. Zeiger RS. Food allergen avoidance in the prevention of food allergy in infants and children. Pediatrics. 2003;111(6 Pt 3):1662–71. Epub 2003/06/05. pmid:12777607.
  10. 10. Isolauri E, Sutas Y, Salo MK, Isosomppi R, Kaila M. Elimination diet in cow's milk allergy: risk for impaired growth in young children. The Journal of pediatrics. 1998;132(6):1004–9. Epub 1998/06/17. pmid:9627594.
  11. 11. Hahn EL, Bacharier LB. The atopic march: the pattern of allergic disease development in childhood. Immunology and allergy clinics of North America. 2005;25(2):231–46, v. Epub 2005/05/10. pmid:15878453.
  12. 12. Faria AM, Weiner HL. Oral tolerance: therapeutic implications for autoimmune diseases. Clin Dev Immunol. 2006;13(2–4):143–57. Epub 2006/12/13. pmid:17162357; PubMed Central PMCID: PMCPmc2270752.
  13. 13. Tang ML, Martino DJ. Oral immunotherapy and tolerance induction in childhood. Pediatric allergy and immunology: official publication of the European Society of Pediatric Allergy and Immunology. 2013;24(6):512–20. Epub 2013/08/03. pmid:23905867.
  14. 14. Park KS, Park MJ, Cho ML, Kwok SK, Ju JH, Ko HJ, et al. Type II collagen oral tolerance; mechanism and role in collagen-induced arthritis and rheumatoid arthritis. Mod Rheumatol. 2009;19(6):581–9. Epub 2009/08/22. pmid:19697097.
  15. 15. Hirahara K, Hisatsune T, Nishijima K, Kato H, Shiho O, Kaminogawa S. CD4+ T cells anergized by high dose feeding establish oral tolerance to antibody responses when transferred in SCID and nude mice. J Immunol. 1995;154(12):6238–45. Epub 1995/06/15. pmid:7759861.
  16. 16. Chen Y, Inobe J, Marks R, Gonnella P, Kuchroo VK, Weiner HL. Peripheral deletion of antigen-reactive T cells in oral tolerance. Nature. 1995;376(6536):177–80. Epub 1995/07/13. pmid:7603570.
  17. 17. Laegreid A, Thommesen L, Jahr TG, Sundan A, Espevik T. Tumor necrosis factor induces lipopolysaccharide tolerance in a human adenocarcinoma cell line mainly through the TNF p55 receptor. J Biol Chem. 1995;270(43):25418–25. Epub 1995/10/27. pmid:7592709.
  18. 18. Martin P, Del Hoyo GM, Anjuere F, Arias CF, Vargas HH, Fernandez LA, et al. Characterization of a new subpopulation of mouse CD8alpha+ B220+ dendritic cells endowed with type 1 interferon production capacity and tolerogenic potential. Blood. 2002;100(2):383–90. Epub 2002/07/02. pmid:12091326.
  19. 19. Chen Y, Kuchroo VK, Inobe J, Hafler DA, Weiner HL. Regulatory T cell clones induced by oral tolerance: suppression of autoimmune encephalomyelitis. Science. 1994;265(5176):1237–40. Epub 1994/08/26. pmid:7520605.
  20. 20. Shiokawa A, Tanabe K, Tsuji NM, Sato R, Hachimura S. IL-10 and IL-27 producing dendritic cells capable of enhancing IL-10 production of T cells are induced in oral tolerance. Immunol Lett. 2009;125(1):7–14. Epub 2009/05/19. S0165-2478(09)00117-5 [pii] pmid:19446579.
  21. 21. Chang JH, Lee JM, Youn HJ, Lee KA, Chung Y, Lee AY, et al. Functional maturation of lamina propria dendritic cells by activation of NKT cells mediates the abrogation of oral tolerance. Eur J Immunol. 2008;38(10):2727–39. Epub 2008/10/01. pmid:18825753.
  22. 22. Goubier A, Dubois B, Gheit H, Joubert G, Villard-Truc F, Asselin-Paturel C, et al. Plasmacytoid dendritic cells mediate oral tolerance. Immunity. 2008;29(3):464–75. Epub 2008/09/16. S1074-7613(08)00372-5 [pii] pmid:18789731.
  23. 23. Min SY, Park KS, Cho ML, Kang JW, Cho YG, Hwang SY, et al. Antigen-induced, tolerogenic CD11c+,CD11b+ dendritic cells are abundant in Peyer's patches during the induction of oral tolerance to type II collagen and suppress experimental collagen-induced arthritis. Arthritis Rheum. 2006;54(3):887–98. Epub 2006/03/02. pmid:16508971.
  24. 24. Worbs T, Bode U, Yan S, Hoffmann MW, Hintzen G, Bernhardt G, et al. Oral tolerance originates in the intestinal immune system and relies on antigen carriage by dendritic cells. J Exp Med. 2006;203(3):519–27. Epub 2006/03/15. jem.20052016 [pii] pmid:16533884; PubMed Central PMCID: PMC2118247.
  25. 25. Steinman RM, Banchereau J. Taking dendritic cells into medicine. Nature. 2007;449(7161):419–26. Epub 2007/09/28. nature06175 [pii] pmid:17898760.
  26. 26. Bilsborough J, George TC, Norment A, Viney JL. Mucosal CD8alpha+ DC, with a plasmacytoid phenotype, induce differentiation and support function of T cells with regulatory properties. Immunology. 2003;108(4):481–92. Epub 2003/04/02. 1606 [pii]. pmid:12667210; PubMed Central PMCID: PMC1782923.
  27. 27. Wang X, O'Gorman MR, Bu HF, Koti V, Zuo XL, Tan XD. Probiotic preparation VSL#3 alters the distribution and phenotypes of dendritic cells within the intestinal mucosa in C57BL/10J mice. J Nutr. 2009;139(8):1595–602. Epub 2009/06/25. jn.109.109934 [pii] pmid:19549755; PubMed Central PMCID: PMC2709306.
  28. 28. Skripak JM, Nash SD, Rowley H, Brereton NH, Oh S, Hamilton RG, et al. A randomized, double-blind, placebo-controlled study of milk oral immunotherapy for cow's milk allergy. J Allergy Clin Immunol. 2008;122(6):1154–60. Epub 2008/10/28. S0091-6749(08)01725-9 [pii] pmid:18951617.
  29. 29. Boonstra A, Asselin-Paturel C, Gilliet M, Crain C, Trinchieri G, Liu YJ, et al. Flexibility of mouse classical and plasmacytoid-derived dendritic cells in directing T helper type 1 and 2 cell development: dependency on antigen dose and differential toll-like receptor ligation. J Exp Med. 2003;197(1):101–9. Epub 2003/01/08. pmid:12515817; PubMed Central PMCID: PMC2193804.
  30. 30. Coombes JL, Siddiqui KR, Arancibia-Carcamo CV, Hall J, Sun CM, Belkaid Y, et al. A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-beta and retinoic acid-dependent mechanism. J Exp Med. 2007;204(8):1757–64. Epub 2007/07/11. jem.20070590 [pii] pmid:17620361; PubMed Central PMCID: PMC2118683.
  31. 31. Sun CM, Hall JA, Blank RB, Bouladoux N, Oukka M, Mora JR, et al. Small intestine lamina propria dendritic cells promote de novo generation of Foxp3 T reg cells via retinoic acid. J Exp Med. 2007;204(8):1775–85. Epub 2007/07/11. pmid:17620362; PubMed Central PMCID: PMC2118682.
  32. 32. Wu L, Liu YJ. Development of dendritic-cell lineages. Immunity. 2007;26(6):741–50. Epub 2007/06/22. pmid:17582346.
  33. 33. Lackey DE, Ashley SL, Davis AL, Hoag KA. Retinoic acid decreases adherence of murine myeloid dendritic cells and increases production of matrix metalloproteinase-9. J Nutr. 2008;138(8):1512–9. Epub 2008/07/22. 138/8/1512 [pii]. pmid:18641199; PubMed Central PMCID: PMC2522314.
  34. 34. Ren Z, Guo Z, Meydani SN, Wu D. White button mushroom enhances maturation of bone marrow-derived dendritic cells and their antigen presenting function in mice. J Nutr. 2008;138(3):544–50. Epub 2008/02/22. 138/3/544 [pii]. pmid:18287364.
  35. 35. Szeles L, Keresztes G, Torocsik D, Balajthy Z, Krenacs L, Poliska S, et al. 1,25-dihydroxyvitamin D3 is an autonomous regulator of the transcriptional changes leading to a tolerogenic dendritic cell phenotype. J Immunol. 2009;182(4):2074–83. Epub 2009/02/10. 182/4/2074 [pii] pmid:19201860.
  36. 36. Moreau MC, Gaboriau-Routhiau V. The absence of gut flora, the doses of antigen ingested and aging affect the long-term peripheral tolerance induced by ovalbumin feeding in mice. Res Immunol. 1996;147(1):49–59. Epub 1996/01/01. 0923249496815483 [pii]. pmid:8739328.
  37. 37. Prioult G, Fliss I, Pecquet S. Effect of probiotic bacteria on induction and maintenance of oral tolerance to beta-lactoglobulin in gnotobiotic mice. Clin Diagn Lab Immunol. 2003;10(5):787–92. Epub 2003/09/11. pmid:12965905; PubMed Central PMCID: PMC193892.
  38. 38. Maeda Y, Noda S, Tanaka K, Sawamura S, Aiba Y, Ishikawa H, et al. The failure of oral tolerance induction is functionally coupled to the absence of T cells in Peyer's patches under germfree conditions. Immunobiology. 2001;204(4):442–57. Epub 2002/01/05. pmid:11776399.
  39. 39. Sudo N, Sawamura S, Tanaka K, Aiba Y, Kubo C, Koga Y. The requirement of intestinal bacterial flora for the development of an IgE production system fully susceptible to oral tolerance induction. J Immunol. 1997;159(4):1739–45. Epub 1997/08/15. pmid:9257835.
  40. 40. Wannemuehler MJ, Kiyono H, Babb JL, Michalek SM, McGhee JR. Lipopolysaccharide (LPS) regulation of the immune response: LPS converts germfree mice to sensitivity to oral tolerance induction. J Immunol. 1982;129(3):959–65. Epub 1982/09/01. pmid:6980928.
  41. 41. So JS, Lee CG, Kwon HK, Yi HJ, Chae CS, Park JA, et al. Lactobacillus casei potentiates induction of oral tolerance in experimental arthritis. Mol Immunol. 2008;46(1):172–80. Epub 2008/09/23. S0161-5890(08)00598-1 [pii] pmid:18804867.
  42. 42. Tsai YT, Cheng PC, Fan CK, Pan TM. Time-dependent persistence of enhanced immune response by a potential probiotic strain Lactobacillus paracasei subsp. paracasei NTU 101. Int J Food Microbiol. 2008;128(2):219–25. Epub 2008/09/24. S0168-1605(08)00456-X [pii] pmid:18809220.
  43. 43. de Moreno de LeBlanc A, Dogi CA, Galdeano CM, Carmuega E, Weill R, Perdigon G. Effect of the administration of a fermented milk containing Lactobacillus casei DN-114001 on intestinal microbiota and gut associated immune cells of nursing mice and after weaning until immune maturity. BMC Immunol. 2008;9:27. Epub 2008/06/17. 1471-2172-9-27 [pii] pmid:18554392; PubMed Central PMCID: PMC2459154.
  44. 44. Murphy KM, Heimberger AB, Loh DY. Induction by antigen of intrathymic apoptosis of CD4+CD8+TCRlo thymocytes in vivo. Science. 1990;250(4988):1720–3. Epub 1990/12/21. pmid:2125367.
  45. 45. Ise W, Totsuka M, Sogawa Y, Ametani A, Hachimura S, Sato T, et al. Naive CD4+ T cells exhibit distinct expression patterns of cytokines and cell surface molecules on their primary responses to varying doses of antigen. J Immunol. 2002;168(7):3242–50. Epub 2002/03/22. pmid:11907078.
  46. 46. Ise W, Totsuka M, Takato R, Hachimura S, Sato T, Ametani A, et al. Primary response of naive CD4(+) T cells to amino acid-substituted analogs of an antigenic peptide can show distinct activation patterns: Th1- and Th2-type cytokine secretion, and helper activity for antibody production without apparent cytokine secretion. FEBS Lett. 2000;465(1):28–33. Epub 2000/01/06. S0014-5793(99)01716-0 [pii]. pmid:10620701.
  47. 47. Yoshida A, Aoki R, Kimoto-Nira H, Kobayashi M, Kawasumi T, Mizumachi K, et al. Oral administration of live Lactococcus lactis C59 suppresses IgE antibody production in ovalbumin-sensitized mice via the regulation of interleukin-4 production. FEMS Immunol Med Microbiol. 2011;61(3):315–22. Epub 2011/01/06. pmid:21205006.
  48. 48. Tsuji NM, Mizumachi K, Kurisaki J. Interleukin-10-secreting Peyer's patch cells are responsible for active suppression in low-dose oral tolerance. Immunology. 2001;103(4):458–64. Epub 2001/09/01. imm1265 [pii]. pmid:11529936; PubMed Central PMCID: PMC1783258.
  49. 49. Battaglia M, Gianfrani C, Gregori S, Roncarolo MG. IL-10-producing T regulatory type 1 cells and oral tolerance. Annals of the New York Academy of Sciences. 2004;1029:142–53. Epub 2005/02/01. pmid:15681753.
  50. 50. Kang SG, Lim HW, Andrisani OM, Broxmeyer HE, Kim CH. Vitamin A metabolites induce gut-homing FoxP3+ regulatory T cells. J Immunol. 2007;179(6):3724–33. Epub 2007/09/06. 179/6/3724 [pii]. pmid:17785809.
  51. 51. Mucida D, Park Y, Kim G, Turovskaya O, Scott I, Kronenberg M, et al. Reciprocal TH17 and regulatory T cell differentiation mediated by retinoic acid. Science. 2007;317(5835):256–60. Epub 2007/06/16. 1145697 [pii] pmid:17569825.
  52. 52. Michalek SM, Kiyono H, Wannemuehler MJ, Mosteller LM, McGhee JR. Lipopolysaccharide (LPS) regulation of the immune response: LPS influence on oral tolerance induction. J Immunol. 1982;128(5):1992–8. Epub 1982/05/01. pmid:6460815.
  53. 53. Tanaka K, Ishikawa H. Role of intestinal bacterial flora in oral tolerance induction. Histol Histopathol. 2004;19(3):907–14. Epub 2004/05/29. pmid:15168353.
  54. 54. Bradley LM, Atkins GG, Swain SL. Long-term CD4+ memory T cells from the spleen lack MEL-14, the lymph node homing receptor. J Immunol. 1992;148(2):324–31. Epub 1992/01/15. pmid:1345918.
  55. 55. DeGrendele HC, Estess P, Siegelman MH. Requirement for CD44 in activated T cell extravasation into an inflammatory site. Science. 1997;278(5338):672–5. Epub 1997/10/24. pmid:9381175.
  56. 56. Wittmann A, Autenrieth IB, Frick JS. Plasmacytoid dendritic cells are crucial in Bifidobacterium adolescentis-mediated inhibition of Yersinia enterocolitica infection. PloS one. 2013;8(8):e71338. Epub 2013/08/27. pmid:23977019; PubMed Central PMCID: PMCPMC3748105.
  57. 57. Diana J, Griseri T, Lagaye S, Beaudoin L, Autrusseau E, Gautron AS, et al. NKT cell-plasmacytoid dendritic cell cooperation via OX40 controls viral infection in a tissue-specific manner. Immunity. 2009;30(2):289–99. Epub 2009/02/17. pmid:19217323.
  58. 58. Kinjo Y, Illarionov P, Vela JL, Pei B, Girardi E, Li X, et al. Invariant natural killer T cells recognize glycolipids from pathogenic Gram-positive bacteria. Nat Immunol. 2011;12(10):966–74. Epub 2011/09/06. pmid:21892173; PubMed Central PMCID: PMCPmc3178673.
  59. 59. Ashour HM, Seif TM. The role of B cells in the induction of peripheral T cell tolerance. Journal of leukocyte biology. 2007;82(5):1033–9. Epub 2007/07/28. pmid:17656652.
  60. 60. Ashour HM, Niederkorn JY. Expansion of B cells is necessary for the induction of T-cell tolerance elicited through the anterior chamber of the eye. Int Arch Allergy Immunol. 2007;144(4):343–6. Epub 2007/08/03. pmid:17671393.
  61. 61. Peng HJ, Chang ZN, Lee CC, Kuo SW. B-cell depletion fails to abrogate the induction of oral tolerance of specific Th1 immune responses in mice. Scandinavian journal of immunology. 2000;51(5):454–60. Epub 2000/05/03. pmid:10792836.
  62. 62. Sun JB, Flach CF, Czerkinsky C, Holmgren J. B lymphocytes promote expansion of regulatory T cells in oral tolerance: powerful induction by antigen coupled to cholera toxin B subunit. J Immunol. 2008;181(12):8278–87. Epub 2008/12/04. pmid:19050244.
  63. 63. Carter NA, Rosser EC, Mauri C. Interleukin-10 produced by B cells is crucial for the suppression of Th17/Th1 responses, induction of T regulatory type 1 cells and reduction of collagen-induced arthritis. Arthritis Res Ther. 2012;14(1):R32. Epub 2012/02/10. pmid:22315945; PubMed Central PMCID: PMCPmc3392827.
  64. 64. Bhattacharya P, Fan J, Haddad C, Essani A, Gopisetty A, Elshabrawy HA, et al. A novel pancreatic beta-cell targeting bispecific-antibody (BsAb) can prevent the development of type 1 diabetes in NOD mice. Clinical immunology (Orlando, Fla). 2014;153(1):187–98. Epub 2014/05/06. pmid:24792135; PubMed Central PMCID: PMCPmc4077286.
  65. 65. Buckland J. Rheumatoid arthritis: Anti-TNF and anti-IL-17 antibodies—better together! Nature reviews Rheumatology. 2014;10(12):699. Epub 2014/10/29. pmid:25348041.
  66. 66. Li Q, Ren G, Xu L, Wang Q, Qi J, Wang W, et al. Therapeutic efficacy of three bispecific antibodies on collagen-induced arthritis mouse model. Int Immunopharmacol. 2014;21(1):119–27. Epub 2014/05/08. pmid:24800661.
  67. 67. Thiruppathi M, Sheng JR, Li L, Prabhakar BS, Meriggioli MN. Recombinant IgG2a Fc (M045) multimers effectively suppress experimental autoimmune myasthenia gravis. Journal of autoimmunity. 2014;52:64–73. Epub 2014/01/07. pmid:24388113; PubMed Central PMCID: PMCPmc4518541.
  68. 68. Lan Q, Fan H, Quesniaux V, Ryffel B, Liu Z, Zheng SG. Induced Foxp3(+) regulatory T cells: a potential new weapon to treat autoimmune and inflammatory diseases? Journal of molecular cell biology. 2012;4(1):22–8. Epub 2011/11/24. pmid:22107826; PubMed Central PMCID: PMCPmc3491614.
  69. 69. Zeng H, Zhang R, Jin B, Chen L. Type 1 regulatory T cells: a new mechanism of peripheral immune tolerance. Cellular & molecular immunology. 2015;12(5):566–71. Epub 2015/06/09. pmid:26051475; PubMed Central PMCID: PMCPmc4579656.
  70. 70. Jin JO, Han X, Yu Q. Interleukin-6 induces the generation of IL-10-producing Tr1 cells and suppresses autoimmune tissue inflammation. Journal of autoimmunity. 2013;40:28–44. Epub 2012/08/28. pmid:22921334; PubMed Central PMCID: PMCPmc3524403.