Pertussis toxin (PTX) has been widely used as an adjuvant to induce Th1-mediated organ-specific autoimmune diseases in animal models. However, the cellular and molecular mechanisms remain to be defined. In this study, we showed that dendritic cells (DC) stimulated with PTX (PTX-DC) were able to substitute for PTX to promote experimental autoimmune uveitis (EAU). EAU induced by PTX-DC revealed a typical Th1 response, characterized by high uveitogenic retinal Ag interphotoreceptor retinoid-binding protein (IRBP)-specific IFN-γ and IL-12 production in the draining lymph nodes, as well as increased levels of anti-IRBP IgG2a and decreased levels of anti-IRBP IgG1 in the serum of IRBP-immunized mice. Furthermore, PTX-DC preferentially induced T cells to produce the Th1 cytokine, IFN-γ. After being stimulated with PTX, DC exhibited up-regulation of MHC class II, CD80, CD86, CD40, and DEC205. PTX-DC had also increased allostimulatory capacity and IL-12 and TNF-α production. Serum IL-12 was increased in naive mice that received PTX-DC i.p. In addition, PTX activated extracellular signal-regulated kinase in DC. Following the inhibition of extracellular signal-regulated kinase signaling, the maturation of PTX-DC was reduced. Subsequently, the ability of PTX-DC to promote IFN-γ production by T cells in vitro and to induce EAU in vivo was blocked. The results suggest that PTX might exert an adjuvant effect on DC to promote their maturation and the production of proinflammatory cytokines, thereby eliciting a Th1 response.

Pertussis toxin (PTX)3 is an exotoxin produced by Bordetella pertussis, known to have adjuvant properties that induce Th1 responses (1). PTX has been widely used to enhance Th1-mediated organ-specific autoimmune diseases, including experimental autoimmune encephalomyelitis, experimental autoimmune uveitis (EAU), and experimental autoimmune orchitis (2, 3, 4, 5, 6, 7). PTX has also been reported to augment delayed-type hypersensitivity reactions (8). One study demonstrated that PTX is able to induce Th1 responses by inhibiting Gi protein signaling that negatively regulates IL-12 production (9). It is noted that the adjuvant effect of PTX is associated with its ability to induce production of proinflammatory cytokines and expression of the costimulatory molecules B7-1 and B7-2 on APCs, which affects T cell differentiation (10, 11). However, the cells targeted by PTX in vivo that promote Th1-mediated organ-specific autoimmune diseases, and the molecular mechanisms of this regulation are not clear.

Dendritic cells (DC) are the most potent APCs for priming naive T cells and the main source of IL-12 production (12, 13, 14, 15). Upon contact with bacterial components or proinflammatory mediators, immature DC that have captured Ag in the periphery migrate to the T cell zone of lymphoid organs, where they present Ag in the context of MHC molecules. During migration from the periphery to the lymph nodes (LN), DC undergo a maturation process that results in morphological and functional changes. Expression of IL-12 by DC can be induced at the time of encounter with microbial compounds. This proinflammatory cytokine promotes cell-mediated response, stimulates IFN-γ secretion by NK and T cells, and favors the development of Th1 cells (16, 17).

Because DC are essential to generate and maintain an immune response, and because they play a crucial role in priming naive T cells and initiating autoimmunity (13, 18, 19), we hypothesize that the adjuvant effect of PTX results from a modulation of DC function. In this study, we show for the first time that DC stimulated with PTX (PTX-DC) are able to substitute for PTX to enhance Th1-mediated EAU in a syngeneic cell transfer model. Moreover, PTX-DC preferentially induced T cells to produce the Th1 cytokine, IFN-γ. Subsequent experiments demonstrated that the PTX-DC secreted high IL-12 in vitro and induced serum IL-12 production in vivo. The novel role of PTX-DC prompted us to assess PTX-mediated signaling and the correlation between PTX-DC maturation and its adjuvant effect on EAU induction.

Six- to 8-wk-old female B10.A and BALB/c mice were purchased from The Jackson Laboratory (Bar Harbor, ME). Animals were kept in a specific pathogen-free facility at the Chinese Academy of Sciences. Animal care and use were in compliance with institutional guidelines.

RPMI 1640 (Life Technologies, Grand Island, NY) supplemented with 10% heat-inactivated FCS, 100 U/ml penicillin, 100 μg/ml streptomycin, 1 mM sodium pyruvate, 2 mM l-glutamine, and 5 × 10−5 M 2-ME was used for DC culture, fibroblast culture, and primary DC-T cell coculture, as well as for secondary T cell stimulation culture. Interphotoreceptor retinoid-binding protein (IRBP) was a gift from R. Caspi, National Eye Institute, National Institutes of Health. Mouse rGM-CSF, BSA, α-methylmannopyranoside, PTX, and CFA were purchased from Sigma-Aldrich (St. Louis, MO). Mycobacterium tuberculosis strain H37RA and LPS (Escherichiacoli 0111:B4) were from Difco (Detroit, MI). Mitogen-activated protein/extracellular signal-regulated kinase (ERK) kinase inhibitor PD98059 was purchased from Biomol Research Laboratories (Plymouth Meeting, PA). PD98059 was dissolved in DMSO.

The method was modified from one initially described by Inaba et al. (20). Briefly, bone marrow cells were flushed from the femurs and tibias of B10.A mice and subsequently depleted of red cells with ammonium chloride. Cells were cultured at 2 × 106 cells/well in 24-well plates in medium supplemented with 20 ng/ml murine rGM-CSF. Nonadherent cells were removed carefully, and fresh medium was added every 2 days. On day 8, nonadherent cells released spontaneously from proliferating cell clusters were collected. Surface phenotype analysis by flow cytometry consistently revealed a typical immature DC surface phenotype (CD11c+, CD80low, CD86low, I-Aklow) in 80–90% cells and microscopically revealed a dendritic, veiled morphology. As expected, no contaminating B cells, macrophages, and T cells were generated under these conditions, as determined by FACS analysis using mAbs specific for B220, F4/80, and CD3 (data not shown). PTX-DC were generated in the same way, but with the addition of 10 ng/ml PTX for the final 24 h of incubation. For in vitro chemotaxis analysis, 100 ng/ml LPS was added to DC cultures. DC were harvested, washed twice, and used in subsequent experiments. For immunization with DC in a syngeneic cell transfer model, DC were resuspended in PBS and administered i.p. into syngeneic mice. In some experiments, DC were preincubated with different concentrations of PD98059 for 30 min and then with PTX (10 ng/ml) for 6 and 24 h. Because PD98059 was dissolved in DMSO, DC that had been incubated with DMSO (at the same concentration as the diluent for PD98059) alone were used as a control.

Lung fibroblasts were prepared according to a method described by Kumar et al. (21). Briefly, lung tissue from 6- to 8-wk-old female B10.A mice was cut into 0.5- to 1-mm fragments, and 20–40 pieces were dispersed over the surface of 25-cm2 culture flasks (Corning Costar, Cambridge, MA). The explants were allowed to adhere overnight in 1 ml medium, and then were brought to a final volume of 5 ml. The lung explants were removed after 3 days of culture. The fibroblasts that grew out of the lung explants were cultured, and the medium was replaced at 3-day intervals. The fibroblasts were grown to confluence and serially passaged by trypsinization at low temperature (22). Cells between passages 1 and 3 were used. For preparing PTX-stimulated fibroblasts (PTX-fibroblasts), 10 ng/ml PTX was added to the fibroblast cultures for the final 24 h of incubation. The PTX-fibroblasts were harvested, washed twice, and used in subsequent experiments.

In all experiments, B10.A mice were immunized s.c. with 50 μg of IRBP in 0.2-ml emulsion (1:1 v/v) with CFA (containing 2.5 mg/ml of M. tuberculosis). In addition, PTX (0.5 μg/0.1 ml) was injected i.p. as an additional adjuvant control to induce EAU. DC (1 × 106 cells/0.1 ml/mouse), PTX-fibroblasts (1 × 106 cells/0.1 ml/mouse), or PBS (0.1 ml/mouse) were injected i.p. into the other groups. Eyes were collected 21 days after immunization, and EAU score was evaluated by histopathology.

Freshly enucleated eyes were fixed for 1 h in 4% phosphate-buffered glutaraldehyde and transferred into 10% phosphate-buffered formaldehyde until processing. Fixed and dehydrated tissue was embedded in methacrylate, and 4- to 6-mm sections, cut through the pupillary-optic nerve plane, were stained by standard H&E. Severity of EAU was scored on a scale of 0–4 in half-point increments, according to a semiquantitative system described previously (23). In brief, the minimal criterion to score an animal as positive by histopathology was inflammatory cell infiltration of the ciliary body, choroid, or retina. Progressively higher grades were assigned for presence of discrete lesions in the tissue, such as vasculitis, granuloma formation, retinal folding and/or detachment, photoreceptor damage, etc. The grading system takes into account lesion type, size, and number.

Cell suspensions were prepared from draining LN of mice that had been immunized with IRBP 21 days previously. The cells (5 × 106 cells/ml/well) were cultured for 48 h with or without IRBP (30 μg/ml) in 24-well plates in complete RPMI 1640 medium containing 0.5% syngeneic normal mouse serum (24). The supernatants were collected after 48 h, and IRBP-specific IFN-γ, IL-4, and IL-12 secretion was measured by ELISA (R&D Systems, Minneapolis, MN). The sensitivity for IFN-γ, IL-4, and IL-12p70 was 10 pg/ml.

Serum levels of anti-IRBP IgG2a and IgG1 subclasses were determined by ELISA in serum collected 21 days after immunization, as described previously (25). In brief, 96-well microtiter plates (Corning Costar) were coated with IRBP (1 μg/ml). After blocking the plates with BSA (Sigma-Aldrich), and an overnight incubation with diluted serum samples, the plates were developed using HRP-conjugated goat anti-IgG subclass-specific Abs (BD PharMingen, San Diego, CA). A series of serum dilutions were examined in the preliminary experiments. The OD values obtained positively correlated with the concentration of anti-IRBP Abs. In each assay, IgG subclass was measured in duplicate on a single plate. Data are mean ± SD of three separate experiments.

DC or fibroblasts were resuspended in PBS, as mentioned above, and used to immunize mice in a syngeneic cell transfer model. Naive B10.A mice were injected with fresh DC or fibroblasts (1 × 106 cells/0.1 ml/mouse) via i.p. injection. The injection of PBS alone served as the negative control. Mice were sacrificed at various time points following injection, and the serum of individual mice was collected for IL-12 determinations.

DC were labeled with the tracking fluorochrome CFSE (Molecular Probes, Eugene, OR), as previously described (26). Briefly, either DC not stimulated with PTX (unstimulated DC), PTX-DC, or PTX-fibroblasts were incubated with CFSE at a final concentration of 5 μM in PBS for 10 min, and the labeling was terminated by the addition of FCS (10% of the total volume). DC were washed twice with RPMI 1640/10% FCS medium, washed once with PBS, and resuspended in PBS for i.p. injection. B10.A mice were injected with labeled DC or fibroblasts (1 × 106 cells/0.1 ml/mouse, i.p.). Control mice received i.p. injection of PBS alone. After 48 h, the mice were sacrificed, and the inguinal LN was removed. Single cell suspensions of LN cells were prepared and were stained with CD11c-PE (BD PharMingen). CFSE+ and CD11c+ double-positive DC cells were detected by flow cytometry.

The chemotaxis analysis in vitro was performed, as described previously (27), with minor modifications. Recombinant macrophage-inflammatory protein (MIP)-3β (100 ng/ml; R&D Systems) was diluted in medium without FCS, and 500 μl was added to 24-well tissue culture plates (Corning Costar). Transwell culture inserts (5.0-μm pore size; Corning Costar) were placed in each well, and 80 μl of DC suspension (4.0 × 105 cells/well) was added to the top chamber. After incubation at 37°C in 5% CO2 for 3 h, the cells that had migrated to the bottom chamber were recovered and counted by light microscopy.

DC were incubated with the following primary mAbs (conjugated or uncoupled): FITC-labeled anti-mouse I-Ak (clone 10-3.6; BD PharMingen), FITC-labeled anti-mouse CD40 (clone 5C3; BD PharMingen), rat anti-mouse CD80 (clone 1G10; BD PharMingen), FITC-labeled anti-mouse CD86 (clone GL1; BD PharMingen), PE-labeled anti-mouse CD11c (clone HL3; BD PharMingen), and rat anti-mouse DEC205/NLDC145 (a gift from P. Allavena, Istituto di Ricerche Farmacologiche Mario Negri, Milan, Italy; 10% culture supernatant). If possible, cells were incubated with anti-CD16/CD32 (2.4G2) to reduce nonspecific binding of mAbs. Unconjugated rat mAbs were revealed by FITC-conjugated goat anti-rat IgG and IgM Ab (Southern Biotechnology, Birmingham, AL) as the second reagent. Isotype controls were conjugated or uncoupled rat IgG2a, and hamster IgG (all from BD PharMingen). All staining reactions were performed on ice. After each step, cells were washed twice with 1% FCS and 0.1% sodium azide in PBS. After the final incubation, cells were fixed with paraformaldehyde (1%). Cells were analyzed on a FACSCalibur cytometer using CellQuest software (BD Biosciences, San Jose, CA).

For analysis of intracellular IFN-γ and IL-4 production, T cells from BALB/c mice were cocultured with allogeneic DC from B10.A mice (DC-T cell ratio of 1:20; 2 × 105 cells/well) in 96-well plates for 5 days. T cells were purified using Dynabeads M-280 (Dynal) with the indirect method using an Ab cocktail composed of anti-I-Ak, CD45RO/B220 (BD PharMingen), with a resulting purity of >98%. Primed cells were stimulated with anti-mouse anti-CD3ε mAb (5 μg/ml; clone 145.2C11; BD PharMingen) and anti-mouse CD28 mAb (3 μg/ml; clone 37.51; BD PharMingen) for 6 h. Brefeldin A (10 μg/ml; Sigma-Aldrich) was added for the last 4 h, and cells were collected, washed, fixed, saponin permeabilized (perm/fix solution; BD PharMingen), and stained with FITC-labeled anti-IFN-γ and PE-labeled anti-IL-4 mAbs (BD PharMingen), according to the manufacturer’s instructions. Appropriate fluorochrome-conjugated, isotype-matched, irrelevant mAbs were used as negative controls. Cells were analyzed on a FACSCalibur cytometer using CellQuest software (BD Biosciences).

For analysis of intracellular IL-12 and TNF-α production, on day 7, DC were incubated in the presence or absence of PTX for 6 h. Brefeldin A (10 μg/ml) was added for the last 4 h. Cells were stained as described above with PE-labeled anti-IL-12p40/p70 (clone C15.6; BD PharMingen) or FITC-labeled anti-TNF-α (clone MP6-XT22; BD PharMingen). Appropriate fluorochrome-conjugated, isotype-matched, irrelevant mAbs were used as negative controls.

For analysis of cytokines by ELISA, cultures were set up as described above. On day 5, IFN-γ and IL-4 were measured using commercially available ELISA kits (R&D Systems). The supernatants from DC cultures in the presence or absence of PTX for 24 h were collected for determining IL-12p70 production (R&D Systems).

Unstimulated DC or PTX-DC from B10.A mice were exposed to 15 Gy of x-ray irradiation, and various dilutions from 3 × 103 to 1 × 104 cells/well were added to round-bottom 96-well microtiter plates. Allogeneic responder T cells from BALB/c mice were purified using Dynabeads M-280 (Dynal) with the indirect method using an Ab cocktail composed of anti-I-Ak, CD45RO/B220 (BD PharMingen), with a resulting purity of >98%, and added at a final concentration of 2 × 105 cells/well. The cultures were then incubated for 96 h, followed by an 18-h pulse with 0.5 μCi of [3H]thymidine/well. Results are expressed as mean cpm ± SD for triplicate wells.

DC at a concentration of 1 × 106 cells/ml were challenged with or without 10 ng/ml PTX in modified Eagle’s medium containing 0.1% BSA at 37°C for 15 min. The cells were then lysed in 10 mM Tris-HCl, pH 7.4, 5 mM EDTA, 2% SDS, and 1% 2-ME. Aliquots (50 μg of protein) of the whole cell extract were prepared and subjected to 10% SDS-PAGE and then electroblotted onto nitrocellulose membranes. The total amounts of ERK and phosphorylated ERK were detected by anti-total p44/p42 mitogen-activated protein kinase (MAPK) Abs and anti-phospho-p44/p42 MAPK Abs (New England Biolabs, Beverly, MA), respectively.

Experiments were repeated at least twice, and usually three or more times. Figures show data compiled from several experiments, or from a representative experiment, as specified. Results represent the mean ± SD, where applicable. Statistical significance of differences was analyzed using the independent Student’s t test. Values of p ≤ 0.05 were considered significant. The results of statistical analyses are given in the figure legends. Statistical analysis of EAU scores was determined by Snedecor and Cochran’s (28) test for linear trend in proportions (nonparametric, frequency based). Each mouse (average of both eyes) was treated as one statistical event.

We hypothesized that DC represent the cellular targets for PTX, and the adjuvant effect of PTX results from a modulation of DC function. To determine whether DC are the target cells for PTX in vivo in inducing EAU, we designed a syngeneic cell transfer model. In these experiments, B10.A mice were immunized with IRBP in CFA. At the same time of immunization, the mice were treated by i.p. injection of DC, fibroblasts, or PTX, as described in Materials and Methods. For the unstimulated DC control, the mice were injected i.p. with syngeneic DC that were cultured in the absence of PTX. For PTX-DC group, the mice were injected i.p. with syngeneic DC that were cultured in the presence of PTX for the final 24 h of incubation. For PTX group (EAU positive control), the mice were given PTX (0.5 μg/mouse) via i.p. injection. To exclude the possibility that PTX may be absorbed by DC in vitro and free PTX released following DC injection in vivo, the mice were injected i.p. with syngeneic PTX-fibroblasts as an irrelevant cell type control. Our earlier experiments show that without PTX as an additional adjuvant, B10.A mice fail to develop EAU (29). Therefore, the immunization with IRBP, CFA, and PTX is required to induce EAU (30). Preliminary experiments were performed to determine the numbers of DC that were needed to induce the disease. A total of 1 × 106 PTX-DC/mouse were optimal numbers for inducing EAU. The eyes from recipient mice were collected 21 days following immunization and cell transfer, and EAU severity was evaluated by histopathology. As expected, mice receiving unstimulated DC or PTX-fibroblasts did not develop EAU (Fig. 1, A and C), and EAU disease was restored in the mice receiving PTX-DC treatment (Fig. 1, B and C). Both incidence and severity of disease in PTX-DC group were comparable to the EAU-positive control mice (PTX group) (Fig. 1 C). The results suggest that PTX-DC is capable of substituting for PTX’s adjuvant effect on EAU induction in vivo.

FIGURE 1.

B10.A mice develop EAU after immunization with IRBP plus PTX-DC, but not with unstimulated DC. B10.A mice were immunized with s.c. IRBP in CFA, and were then injected i.p. with freshly isolated syngeneic unstimulated DC or PTX-DC suspended in PBS, as described in Materials and Methods. Twenty-one days later, the eyes were processed for histopathology. A, Normal retina (R) with intact photoreceptors (∗) and choroid (C) were observed in the eye of a B10.A mouse immunized with IRBP plus unstimulated DC (disease grade 0) (×400). B, In contrast, disorganized retina with retinal vasculitis (arrows) and thickening of the choroid are observed in the eye of a B10.A mouse immunized with IRBP plus PTX-DC (disease grade 2–3) (×400). C, EAU scores in mice receiving PTX-DC or PTX at all points differ significantly from mice receiving unstimulated DC or PTX-fibroblast controls (p < 0.05). Each symbol represents the pathology score for one mouse (average of both eyes). The average EAU score of each group is denoted by the horizontal bar. The results are a pool of two separate experiments.

FIGURE 1.

B10.A mice develop EAU after immunization with IRBP plus PTX-DC, but not with unstimulated DC. B10.A mice were immunized with s.c. IRBP in CFA, and were then injected i.p. with freshly isolated syngeneic unstimulated DC or PTX-DC suspended in PBS, as described in Materials and Methods. Twenty-one days later, the eyes were processed for histopathology. A, Normal retina (R) with intact photoreceptors (∗) and choroid (C) were observed in the eye of a B10.A mouse immunized with IRBP plus unstimulated DC (disease grade 0) (×400). B, In contrast, disorganized retina with retinal vasculitis (arrows) and thickening of the choroid are observed in the eye of a B10.A mouse immunized with IRBP plus PTX-DC (disease grade 2–3) (×400). C, EAU scores in mice receiving PTX-DC or PTX at all points differ significantly from mice receiving unstimulated DC or PTX-fibroblast controls (p < 0.05). Each symbol represents the pathology score for one mouse (average of both eyes). The average EAU score of each group is denoted by the horizontal bar. The results are a pool of two separate experiments.

Close modal

Our previous work has shown that a dominant Th1-like response is required for EAU development in B10.A mice (29). Because the recipients developed EAU after PTX-DC treatment, we examined whether the recipients produced an IRBP-specific Th1 response. Twenty-one days after immunization with IRBP in CFA, draining LN cells were collected from mice that received different treatments. The cells were restimulated in vitro for 48 h in the absence or presence of IRBP. The supernatants were collected, and IFN-γ, IL-4, and IL-12p70 were measured by ELISA. Upon rechallenging with IRBP in vitro, draining LN cells from PTX-DC-treated mice secreted significantly higher amounts of IFN-γ and IL-12p70 than mice that received PBS or unstimulated DC (Fig. 2, A and B). IL-4 was undetectable in cell culture supernatants in all groups (data not shown). This finding is consistent with our previous observation that a Th1-like response correlates with susceptibility to EAU in B10.A mice (29). To confirm the presence of a dominant Th1 response in vivo, we determined IRBP-specific IgG2a and IgG1 Abs in serum of individual mice 21 days after immunization. IFN-γ and IL-4 serve as switch factors for IgG2a and IgG1, respectively. Thus, an increased IgG2a-IgG1 ratio is evidence of a response shifting toward Th1. The data showed that in PTX-DC group, IgG2a levels were increased and IgG1 levels were decreased as compared with unstimulated DC group (Fig. 2, C and D), consistent with a Th1-dependent isotype switch. Taken together, these results suggest that PTX-DC induce a Th1 response in vivo, characterized by high IFN-γ and IL-12p70 secretion, increased levels of anti-IRBP IgG2a, and decreased levels of anti-IRBP IgG1.

FIGURE 2.

IRBP-specific cytokine production by draining LN and serum concentrations of anti-IRBP IgG2a and IgG1 Abs in B10.A mice immunized with IRBP. B10.A mice were immunized with IRBP in CFA, and received an i.p. injection of PBS, unstimulated DC, or PTX-DC. Cells were prepared from draining LN of mice 21 days after immunization and were cultured for 48 h with or without (−) IRBP (30 μg/ml). The supernatants were collected, and IFN-γ, IL-4, and IL-12p70 were determined by ELISA. Sera were collected from IRBP-immunized individual mice, as mentioned above, and IRBP-specific IgG2a (1/400 dilution) and IgG1 (1/1600 dilution) were determined by ELISA. A, IFN-γ production. B, IL-12p70 production. IL-4 production is undetectable (data not shown). C, IRBP-specific IgG2a. D, IRBP-specific IgG1. Statistically significant differences from control values are indicated by asterisks (∗, p < 0.05; ∗∗, p < 0.01). Data are presented as the mean ± SD, n = 5, and are representative of two separate experiments. N.D., not detectable.

FIGURE 2.

IRBP-specific cytokine production by draining LN and serum concentrations of anti-IRBP IgG2a and IgG1 Abs in B10.A mice immunized with IRBP. B10.A mice were immunized with IRBP in CFA, and received an i.p. injection of PBS, unstimulated DC, or PTX-DC. Cells were prepared from draining LN of mice 21 days after immunization and were cultured for 48 h with or without (−) IRBP (30 μg/ml). The supernatants were collected, and IFN-γ, IL-4, and IL-12p70 were determined by ELISA. Sera were collected from IRBP-immunized individual mice, as mentioned above, and IRBP-specific IgG2a (1/400 dilution) and IgG1 (1/1600 dilution) were determined by ELISA. A, IFN-γ production. B, IL-12p70 production. IL-4 production is undetectable (data not shown). C, IRBP-specific IgG2a. D, IRBP-specific IgG1. Statistically significant differences from control values are indicated by asterisks (∗, p < 0.05; ∗∗, p < 0.01). Data are presented as the mean ± SD, n = 5, and are representative of two separate experiments. N.D., not detectable.

Close modal

Because PTX-DC are able to support the induction of EAU and promote a characteristic Th1 response in vivo, we asked whether PTX-DC acquired the functional features to induce Th1-like responses (IFN-γ) in vitro. Unstimulated DC or PTX-DC were cocultured with purified naive BALB/c T cells (DC-T cell ratio of 1:20) for 5 days. For intracellular cytokine staining, after 5-day culture, T cells were restimulated with plate-bound anti-CD3ε mAb and soluble anti-CD28 mAb in the presence of brefeldin A. As shown in Fig. 3,A, IFN-γ-producing T cells were significantly increased when allogeneic PTX-DC served as the stimulator cell as compared with unstimulated DC. Both PTX-DC and unstimulated DC elicited few IL-4-producing T cells (Fig. 3,A). To further confirm this observation, the supernatants were collected, and IFN-γ and IL-4 levels were determined by ELISA. As shown in Fig. 3 B, PTX-DC induced higher IFN-γ secretion as compared with unstimulated DC. IL-4 was undetectable by ELISA (data not shown). These results clearly demonstrate that PTX-DC preferentially induce IFN-γ production by T cells in vitro.

FIGURE 3.

PTX-DC stimulate T cells to produce IFN-γ. A, The proportion of IFN-γ+ cells and IL-4+ cells was measured by intracellular staining of alloreactive BALB/c T cells. After 5 days in MLR, the cells were collected, counted, and restimulated by plate-bound anti-mouse CD3ε mAb and anti-mouse CD28 mAb for 6 h, as described in Materials and Methods. Thereafter, the cells were fixed and stained for detection of intracellular cytokine using primary specific mAbs. DC were gated out according to their high side scatter. B, The supernatants from a 5-day primary MLR were assayed for IFN-γ by ELISA. Statistically significant differences from control values are indicated by asterisks (∗, p < 0.05). Results are representative of three separate experiments.

FIGURE 3.

PTX-DC stimulate T cells to produce IFN-γ. A, The proportion of IFN-γ+ cells and IL-4+ cells was measured by intracellular staining of alloreactive BALB/c T cells. After 5 days in MLR, the cells were collected, counted, and restimulated by plate-bound anti-mouse CD3ε mAb and anti-mouse CD28 mAb for 6 h, as described in Materials and Methods. Thereafter, the cells were fixed and stained for detection of intracellular cytokine using primary specific mAbs. DC were gated out according to their high side scatter. B, The supernatants from a 5-day primary MLR were assayed for IFN-γ by ELISA. Statistically significant differences from control values are indicated by asterisks (∗, p < 0.05). Results are representative of three separate experiments.

Close modal

To efficiently prime naive T cells, immature DC must be stimulated and acquire a mature phenotype, as defined by the up-regulation of MHC class II, costimulatory molecules, and the secretion of cytokines, such as IL-12 and TNF-α (13, 31). The phenotype and cytokine secretion profiles of unstimulated DC and PTX-DC were analyzed by flow cytometry. The data showed that the expression of MHC class II, DEC205, CD80, CD86, and CD40 was all increased in PTX-DC (Fig. 4,A). This implies that DC are activated by PTX. PTX-DC also secreted higher levels of IL-12 and TNF-α (Fig. 4, B and C). IL-6 and CCR7 mRNA expression were also up-regulated in PTX-DC (data not shown). To test their allostimulatory ability, unstimulated DC or PTX-DC were cocultured with naive BALB/c T cells in a standard 96-h primary MLR. PTX-DC had a higher ability to stimulate T cell proliferation as compared with unstimulated DC (Fig. 4 D). Again, the stimulatory effect of PTX-DC on MLR was consistent with the ability of PTX to drive DC maturation.

FIGURE 4.

PTX induces DC maturation. A, The phenotype of unstimulated DC and PTX-DC was analyzed by flow cytometry. As compared with unstimulated DC, PTX-DC showed high expression of MHC class II, CD86, CD80, CD40, and DEC205. PTX enhances IL-12 and TNF-α production by DC. B, The frequencies of IL-12- or TNF-α-positive DC were determined by intracellular cytokine staining. C, IL-12p70 was determined by ELISA, as described in Materials and Methods. D, PTX-DC increase allostimulatory capacity. Statistically significant differences from control values are indicated by asterisks (∗, p < 0.05). Results are representative of three separate experiments.

FIGURE 4.

PTX induces DC maturation. A, The phenotype of unstimulated DC and PTX-DC was analyzed by flow cytometry. As compared with unstimulated DC, PTX-DC showed high expression of MHC class II, CD86, CD80, CD40, and DEC205. PTX enhances IL-12 and TNF-α production by DC. B, The frequencies of IL-12- or TNF-α-positive DC were determined by intracellular cytokine staining. C, IL-12p70 was determined by ELISA, as described in Materials and Methods. D, PTX-DC increase allostimulatory capacity. Statistically significant differences from control values are indicated by asterisks (∗, p < 0.05). Results are representative of three separate experiments.

Close modal

Because IL-12 is an initiator needed to switch on Th1 responses and because mature PTX-DC are able to produce IL-12 in vitro, we hypothesized that PTX-DC have a potential to alter the systemic environment in vivo and favor the development of a Th1 response. To address this hypothesis, serum IL-12 production was determined in naive mice that received unstimulated DC, PTX-DC, or PTX-fibroblasts. Injecting PBS served as the negative control. Serum samples were collected from the mice at 4 and 24 h after cell transfer. IL-12p70 protein was determined by ELISA. The data showed that mice receiving PTX-DC injection produced significantly higher levels of IL-12p70 than mice receiving unstimulated DC or PTX-fibroblasts. IL-12p70 peaked at 4 h and lasted at 24 h after PTX-DC transfer (Fig. 5). The results suggest that PTX-DC might increase systemic IL-12 production and in turn promote the development of Th1 responses in vivo.

FIGURE 5.

Enhanced serum IL-12 production in naive mice receiving PTX-DC injection. B10.A mice were given PBS, syngeneic unstimulated DC, PTX-DC, or PTX-fibroblasts via i.p. injection. Mice were sacrificed 4 or 24 h following injection, blood was taken, and the serum was collected for IL-12 determination by ELISA. Statistically significant differences from control values are indicated by asterisks (∗, p < 0.05). Data are presented as the mean ± SD values that were derived from three animals in each group, and are representative of two separate experiments.

FIGURE 5.

Enhanced serum IL-12 production in naive mice receiving PTX-DC injection. B10.A mice were given PBS, syngeneic unstimulated DC, PTX-DC, or PTX-fibroblasts via i.p. injection. Mice were sacrificed 4 or 24 h following injection, blood was taken, and the serum was collected for IL-12 determination by ELISA. Statistically significant differences from control values are indicated by asterisks (∗, p < 0.05). Data are presented as the mean ± SD values that were derived from three animals in each group, and are representative of two separate experiments.

Close modal

To initiate specific T cell-mediated immune responses, the direct migration of DC to draining LN appears to be essential. Because PTX-DC are able to enhance Th1 responses and induce EAU in vivo, it is likely that PTX-DC have a potential to migrate after adoptive transfer in vivo. To confirm this, unstimulated DC, PTX-DC, and PTX-fibroblasts were labeled with the fluorescent cell tracker CFSE and injected i.p. into naive mice. The inguinal LN cells were harvested 48 h later. The migratory capacities of DC from the peritoneal cavity to the draining LN were evaluated by measuring CFSE-labeled DC in inguinal LN by flow cytometry. To exclude the possibility that fluorescent cells within the LN preparation were derived from host phagocytic cells that had ingested CFSE-labeled cells, the mice were injected i.p. with syngeneic CFSE-labeled PTX-fibroblasts as a control. The data showed that CFSE+ and CD11c+ double-positive cells were undetectable in inguinal LN from the mice injected with either PBS or PTX-fibroblasts (Fig. 6,A). In contrast, these cells were detectable in inguinal LN of mice that had received either unstimulated DC or PTX-DC, although their percentages were low and there were no significant difference between two groups (Fig. 6,A). To directly address whether PTX-DC can migrate, we designed the following experiment. Because CCR7 is highly expressed on the surface of mature DC and its ligand (MIP-3β)-triggered response plays critical roles in mature DC migration in mice (32), we measured the chemotaxis of PTX-DC in response to MIP-3β in vitro. As shown in Fig. 6,B, PTX-DC had a higher migratory capability in response to MIP-3β than that of unstimulated DC. In the same experimental system, LPS, a known DC maturation-enhancing factor (100 ng/ml), was used as a positive control. As expected, DC stimulated with LPS (LPS-DC) and PTX-DC had a similar effect on MIP-3β-induced chemotaxis (Fig. 6 B). The data clearly demonstrated that under our experimental conditions, PTX-DC were capable of migration in vitro and in vivo.

FIGURE 6.

Donor PTX-DC are detectable in inguinal LN from recipient mice after i.p. injection in vivo. A, A representative FACS profile is shown for groups of mice receiving PBS, unstimulated DC, PTX-DC, or PTX-fibroblasts. Numbers represent the percentage of CFSE+ and CD11c+ cells in the total LN DC. Similar results were obtained in separate experiments. No statistically significant differences in migration were observed between unstimulated DC and PTX-DC. B, PTX-DC and LPS-DC have a similar chemotaxis response to MIP-3β. The unstimulated DC, PTX-DC, and LPS-DC were prepared and recovered, as described in Materials and Methods, and their migratory abilities in response to MIP-3β (100 ng/ml) were determined in vitro. DC were placed in the upper chambers of transwell culture inserts, and DC migration was evaluated after 3-h incubation by assessing the number of cells in the bottom chamber. Statistically significant differences from control values are indicated by asterisks (∗, p < 0.05; ∗∗, p < 0.01). Data are presented as the mean ± SD values obtained from triplicate cultures and are representative of two separate experiments.

FIGURE 6.

Donor PTX-DC are detectable in inguinal LN from recipient mice after i.p. injection in vivo. A, A representative FACS profile is shown for groups of mice receiving PBS, unstimulated DC, PTX-DC, or PTX-fibroblasts. Numbers represent the percentage of CFSE+ and CD11c+ cells in the total LN DC. Similar results were obtained in separate experiments. No statistically significant differences in migration were observed between unstimulated DC and PTX-DC. B, PTX-DC and LPS-DC have a similar chemotaxis response to MIP-3β. The unstimulated DC, PTX-DC, and LPS-DC were prepared and recovered, as described in Materials and Methods, and their migratory abilities in response to MIP-3β (100 ng/ml) were determined in vitro. DC were placed in the upper chambers of transwell culture inserts, and DC migration was evaluated after 3-h incubation by assessing the number of cells in the bottom chamber. Statistically significant differences from control values are indicated by asterisks (∗, p < 0.05; ∗∗, p < 0.01). Data are presented as the mean ± SD values obtained from triplicate cultures and are representative of two separate experiments.

Close modal

One study has demonstrated the involvement of ERK signaling pathways in PTX-induced endothelial cell activation (33). To address whether ERK signaling plays an important role in PTX-stimulated DC activation, the activation of the ERK signaling pathway was determined. When DC were exposed to PTX for 15 min, the level of phospho-p44/p42 MAPK was increased (Fig. 7,A). The data indicated that ERK signaling pathway was activated in PTX-DC. To further confirm whether ERK signaling pathway has a regulatory role in the maturation of PTX-DC, PD98059, a highly specific inhibitor of the ERK signaling pathway (34), was used to treat PTX-DC. Preliminary experiments were performed to determine an optimal concentration of PD98059. The selected concentrations (50 or 100 μM) of PD98059 had a strong inhibition on ERK signaling, but had little influence on DC viability as determined by trypan blue dye exclusion (data not shown). When DC were pretreated with 50 μM PD98059 for 30 min before PTX was added, the up-regulation of CD86 expression was significantly diminished (Fig. 7,B) and PTX-induced up-regulation of MHC class II exhibited minor changes (data not shown). When DC were pretreated with 50 or 100 μM PD98059 for 30 min before PTX stimulation, IL-12 and TNF-α production was reduced (Fig. 7,C). These results revealed that ERK signaling pathway might be involved in the regulation of maturation and proinflammatory cytokine (IL-12 and TNF-α) production in PTX-DC. To address whether ERK signaling pathway is involved in the ability of PTX-DC to induce IFN-γ production by T cells, DC were pretreated with 50 μM PD98059 for 30 min before PTX stimulation, and then cocultured with naive allogeneic T cells. Intracellular IFN-γ and IL-4 in T cells induced by PTX-DC were analyzed by flow cytometry. It was found that once ERK signaling was inhibited in PTX-DC, the ability of PTX-DC to induce IFN-γ production by T cells was significantly eliminated (Fig. 7 D). Taken together, these results demonstrated that ERK signaling pathway might play an important role in PTX-stimulated DC activation, contribute to DC maturation and proinflammatory cytokine secretion, and in turn promote IFN-γ induction by T cells in vitro.

FIGURE 7.

Inhibition of ERK signaling in PTX-DC reduces its maturation and cytokine production. A, PTX activates ERK kinases. DC were incubated with or without 10 ng/ml PTX for 15 min. The total amounts of ERK and phosphorylated ERK were detected by anti-total p44/p42 MAPK Abs and anti-phospho-p44/p42 MAPK Abs, respectively, by Western blot analysis. Lane 1, Unstimulated DC; lane 2, PTX-DC. DC were preincubated with the indicated concentrations of PD98059 for 30 min and then with 10 ng/ml PTX for 6 and 24 h. B, The up-regulation of CD86 by PTX is inhibited by 50 μM PD98059. After 24 h, cell surface expression of CD86 was determined by flow cytometry. Line 1, Isotype control; line 2, unstimulated DC; line 3, PTX-DC; and line 4, PTX-DC plus 50 μM PD98059. C, PD98059 inhibits PTX-induced up-regulation of IL-12 and TNF-α of DC. After 6 h, IL-12 and TNF-α production was determined by intracellular cytokine staining. D, PD98059 suppresses PTX-DC-induced IFN-γ production by T cells. After 24 h, DC were harvested, washed twice, and then cocultured with alloreactive T cells, as mentioned in Fig. 3 A. Results are representative of three separate experiments.

FIGURE 7.

Inhibition of ERK signaling in PTX-DC reduces its maturation and cytokine production. A, PTX activates ERK kinases. DC were incubated with or without 10 ng/ml PTX for 15 min. The total amounts of ERK and phosphorylated ERK were detected by anti-total p44/p42 MAPK Abs and anti-phospho-p44/p42 MAPK Abs, respectively, by Western blot analysis. Lane 1, Unstimulated DC; lane 2, PTX-DC. DC were preincubated with the indicated concentrations of PD98059 for 30 min and then with 10 ng/ml PTX for 6 and 24 h. B, The up-regulation of CD86 by PTX is inhibited by 50 μM PD98059. After 24 h, cell surface expression of CD86 was determined by flow cytometry. Line 1, Isotype control; line 2, unstimulated DC; line 3, PTX-DC; and line 4, PTX-DC plus 50 μM PD98059. C, PD98059 inhibits PTX-induced up-regulation of IL-12 and TNF-α of DC. After 6 h, IL-12 and TNF-α production was determined by intracellular cytokine staining. D, PD98059 suppresses PTX-DC-induced IFN-γ production by T cells. After 24 h, DC were harvested, washed twice, and then cocultured with alloreactive T cells, as mentioned in Fig. 3 A. Results are representative of three separate experiments.

Close modal

Because ERK signaling pathway might be involved in PTX-DC activation and function, then we asked whether PTX-stimulated DC activation was related to its adjuvant effect on EAU induction. To answer this question, we evaluated the induction of EAU conferred by injection of PTX-DC that had been pretreated with 50 μM PD98059 or DMSO (vehicle control) for 30 min before PTX stimulation (under this condition, the maturation and the cytokine secretion of PTX-DC were reduced). B10.A mice were immunized with IRBP in CFA concurrently with the injection of PD98059-pretreated PTX-DC. EAU score was evaluated by histopathology 21 days following immunization. The results demonstrated that EAU was strikingly diminished in the mice receiving PD98059-pretreated PTX-DC (Fig. 8). The data suggest that the PTX-stimulated DC maturation and cytokine secretion might be involved in the adjuvant effect of PTX-DC on EAU induction.

FIGURE 8.

The ability of PTX-DC to induce EAU is blocked by inhibiting ERK signaling. B10.A mice were immunized s.c. with 50 μg IRBP in CFA and, in addition, the mice were injected i.p. with unstimulated DC, vehicle (DMSO)-treated PTX-DC, or 50 μM PD98059-treated PTX-DC. Eyes were harvested 21 days after immunization. EAU score is evaluated by histopathology. Each symbol represents the pathology score for one mouse (average of both eyes). The average EAU score in each group is denoted by the horizontal bar. EAU scores are compared between vehicle-treated PTX-DC and PD98059-treated PTX-DC (p < 0.05). The results are a pool of two separate experiments.

FIGURE 8.

The ability of PTX-DC to induce EAU is blocked by inhibiting ERK signaling. B10.A mice were immunized s.c. with 50 μg IRBP in CFA and, in addition, the mice were injected i.p. with unstimulated DC, vehicle (DMSO)-treated PTX-DC, or 50 μM PD98059-treated PTX-DC. Eyes were harvested 21 days after immunization. EAU score is evaluated by histopathology. Each symbol represents the pathology score for one mouse (average of both eyes). The average EAU score in each group is denoted by the horizontal bar. EAU scores are compared between vehicle-treated PTX-DC and PD98059-treated PTX-DC (p < 0.05). The results are a pool of two separate experiments.

Close modal

PTX serves as an adjuvant to induce Th1-mediated experimental autoimmune disease. Previous work indicates that PTX treatment strongly enhances Th1 responses to the immunizing Ag in parallel to enhancing disease scores (1, 30). One report confirms that PTX treatment strongly enhances Th1 responses and suggests that the activation of APCs and the induction of IL-12 are involved in these processes (9). However, it has been difficult to unambiguously characterize the cellular targets of PTX in vivo. In this study, we have shown that DC treated with PTX can substitute for PTX in the induction of Th1-mediated EAU. It was interesting to suggest that PTX may have a novel role in promoting DC maturation and secreting proinflammatory cytokines, which might be involved in EAU induction.

The enhancement of PTX on Th1 response was seen not only in EAU, but also in experimental autoimmune encephalomyelitis and orchitis. Therefore, this enhancement may be a general characteristic of the animal models in which PTX is used as an adjuvant to elicit a Th1-mediated autoimmune disease.

In this study, we focused on whether DC are target cells for PTX in promoting a Th1 response in vivo. We initially examined the possibility that PTX was able to stimulate DC and consequently favor the development of a Th1 response. This possibility was confirmed by in vivo and in vitro experiments, because PTX-DC were capable of enhancing EAU (Th1-mediated disease) in vivo and promoting IFN-γ production by T cells in vitro.

Because mature DC not only have the capacity to prime naive T cells, but also to provide different signals to induce different types of T cell response (35), we then asked whether the adjuvant role of PTX was correlated with its ability to drive DC maturation. The data reveal that PTX-DC are likely to be mature DC, because MHC class II, CD80, CD86, DEC205, and CD40 were highly expressed on the cell surface of PTX-DC. The cells also secreted high amounts of TNF-α and IL-12, the latter of which is critical in inducing Th1 responses (13, 31). Thus, PTX-DC provide stronger costimulatory signals and/or the proinflammatory cytokines needed for T cell activation and Th1 development. In humans, PTX has been proved to induce human monocyte-derived DC maturation and increase IL-12 production as well as to promote Th1 cell development in vitro (36, 37). In mice, Hofstetter et al. (38) have observed that PTX enhances the expression of MHC class II and costimulatory molecules on cells isolated from spleen or spinal cords. He et al. (9) have shown that inoculation of PTX in mice enhances IL-12 production by splenocytes in response to both microbial and nonmicrobial stimuli. All these experiments suggest that PTX has a potential to drive DC maturation.

Tarrant et al. (39) have reported that endogenous IL-12 is required to generate uveitogenic effector cells and the development of EAU. Yokoi et al. (40) have also observed that the anti-IL-12 treatment during the IRBP-priming phase prevents EAU induction. Our data showed that serum IL-12 was increased in naive mice that received PTX-DC injection. This finding implies the possibility that an increase in systemic IL-12 production might promote autoreactive T cell activation, favor Th1 development, and finally elicit EAU. The question raised by this new finding is that it is not clear for what cell types to produce IL-12 in vivo. It is most likely that IL-12 probably originates from donor PTX-DC because IL-12 secretion reached a peak 4 h after cell transfer. In our experimental model, it is possible that PTX-DC could initially secrete proinflammatory cytokines which might result in the activation of recipient APCs or NK cells, in turn to secrete IL-12 or IFN-γ, respectively, and drive T cell differentiation toward the Th1 pathway (41, 42).

In addition to secreting IL-12, PTX-DC may interact with host T cells. This communication between the cells might also contribute to the ability of PTX-DC to induce EAU. To address this possibility, the ability of PTX-DC to migrate was tested in vivo and in vitro. In vivo, both injected unstimulated DC and PTX-DC were detectable in draining inguinal LN, although the percentages were very low. The data suggest that PTX-DC are able to migrate from the peritoneal cavity into the draining inguinal LN, where autoreactive T cells can be activated and differentiate into Th1 cells. In vitro, we also found that PTX-DC could be stimulated to migrate in response to MIP-3β, suggesting that PTX-DC are likely to be mature DC that have the potential to migrate in vitro. Labeur et al. (43) have reported that murine bone marrow-derived DC appear to migrate very inefficiently into regional LN after s.c. injection, regardless of their maturation state; however, their Ag-presenting capacity in vivo correlates with their maturation stage. Eggert et al. (44) have demonstrated that bone marrow-derived DC are able to accumulate in the T cell areas of draining LN when DC are delivered by s.c. administration or i.p. injection.

In our experimental model, because PTX-DC were not pulsed with Ag in vitro, PTX-DC might affect the cytokine milieu in draining LN by secreting proinflammatory cytokines. This could favor the priming of newly Ag-exposed naive T cells to become Th1 cells or further induce activated Ag-specific T cells toward the Th1 phenotype in an Ag-independent manner (45, 46). In contrast, donor PTX-DC in draining LN might activate local DC and NK cells to secrete IL-12 and IFN-γ, respectively, further driving T cells to differentiate toward the Th1 pathway (41, 42). In addition, if autoantigen, IRBP would be presented by donor PTX-DC before naive T cells primed in vivo; PTX-DC could influence the Th1/Th2 balance as the major APCs involved in priming naive T cells in an Ag-dependent manner.

Eggert et al. (44) have presented evidence that after i.p. injection, most bone marrow-derived DC remain at the injection site. Labeur et al. (43) have reported that the vast majority of bone marrow-derived DC that remain at the site of injection exhibit equal in vitro survival, random migration in collagen gels, and allostimulatory capacity. In this study, we also observed that most injected PTX-DC did not reach draining LN. These data suggest that, in our experimental system, donor PTX-DC at the site of injection may also shift the Th1/Th2 balance through proinflammatory cytokine production without playing any direct effect on priming naive T cells, which are dependent on cell-cell contact in draining LN. This interpretation is consistent with the observation by Maldonado et al. (47), in which s.c. administration of CD8α DC combined with parenteral injection of IL-12 switches a Th2-type response to a Th1-type response. Smith and de St. Groth (48) have also reported that Ag-pulsed CD8α+ DC generate an immune response after s.c. injection without homing to draining LN.

Thus, there are three possibilities for PTX-DC in enhancing Th1 responses in vivo. First, PTX-DC might directly generate an immune response as APCs after homing to draining LN. Second, PTX-DC act as cytokine-producing accessory cells and indirectly deliver a proinflammatory message to T cells to enhance a Th1 response. Third, both the direct and indirect role of PTX-DC could be involved in its adjuvant effect.

PTX has the A-B architecture type of many bacterial toxins similar to cholera toxin and E. coli heat-labile toxin (49). Evidence shows that LPS and PTX bind to different domains on the same p73 receptor on murine splenocytes (50). Recent data imply that CD14 may be a binding site for PTX on myelomonocytic cells (51). These data suggest that PTX may share some endotoxin signal with LPS-mediated signaling (52). A prior report has shown that LPS activates ERK and regulates DC function (53). A recent study shows that PTX directly activates endothelial cell ERK kinases (33). We therefore propose that ERK kinases are key factors mediating the activation of DC by PTX. In this study, we found that PTX activated ERK signaling pathway. It is interesting to note that when ERK signaling was inhibited in PTX-DC, the expression of costimulatory molecules and the production of proinflammatory cytokines were reduced. Similarly, the ability of PTX-DC to promote IFN-γ production by T cells in vitro and its adjuvant effect on the EAU induction was blocked when the ERK signaling was inhibited. Bouchon et al. (54) have demonstrated that owing to selective activation of the ERK pathway, TREM-2/DAP12 promotes a unique maturation program on human monocyte-derived DC. Thus, this DC activation pathway might be responsible for DC maturation and enhanced Ag presentation, which may result in T cell priming and differentiation in vivo.

In this study, we have shown DC activation evoked by PTX. PTX-DC acquired a feature of mature DC and secreted proinflammatory cytokines, which may be involved in its adjuvant effect in the EAU induction. Our data certainly do not exclude that pertussis toxin may be also affecting DC physiology by mechanisms that are not currently appreciated. In addition, our findings do not address the question as to whether PTX exerts effects on additional cell types that may contribute to its adjuvant activity in normal mice. NK cells or macrophages, which are the sources of IFN-γ and IL-12, respectively (55, 56), may also play a role in the enhancement of Th1-mediated EAU after PTX injection in vivo.

Overall, our results suggest that DC play a crucial role in the adjuvant effect of PTX on the induction of Th1 responses. The data show that this property of PTX is probably due to its ability to promote DC maturation and secrete proinflammatory cytokines. This mechanism might explain why microbial adjuvants or compounds, such as PTX, signal DC to undergo the coordinate maturation events that underlie efficient T cell priming and differentiation. Therefore, strategies that target DC activation and modulate DC function in vivo may have significant implications for vaccine design and the treatment of autoimmune disease.

We are grateful to Drs. Rachel R. Caspi, Stephen E. Ullrich, Bing Su, Chen Dong, and Ming Yeh for critical review of the manuscript and for helpful comments.

1

This work was supported by 973 Project (G1999053907), National Natural Science Foundation of China (30170888), National Key Basic Program of China (2001CB510006), and key project of Chinese Academy of Sciences (KSCX2-2-08-02).

3

Abbreviations used in this paper: PTX, pertussis toxin; DC, dendritic cell; EAU, experimental autoimmune uveitis; ERK, extracellular signal-regulated kinase; IRBP, interphotoreceptor retinoid-binding protein; LN, lymph node; MAPK, mitogen-activated protein kinase; MIP, macrophage-inflammatory protein.

1
Caspi, R. R., P. B. Silver, C. C. Chan, B. Sun, R. K. Agarwal, J. Wells, S. Oddo, Y. Fujino, F. Najafian, R. L. Wilder.
1996
. Genetic susceptibility to experimental autoimmune uveoretinitis in the rat is associated with an elevated Th1 response.
J. Immunol.
157
:
2668
2
Raine, C. S..
1984
. Biology of disease: analysis of autoimmune demyelination: its impact upon multiple sclerosis.
Lab. Invest.
50
:
608
3
Tuohy, V. K..
1994
. Peptide determinants of myelin proteolipid protein (PLP) in autoimmune demyelinating disease: a review.
Neurochem. Res.
19
:
935
4
Kerlero de Rosbo, N., I. Mendel, A. Ben-Nun.
1995
. Chronic relapsing experimental autoimmune encephalomyelitis with a delayed onset and an atypical clinical course, induced in PL/J mice by myelin oligodendrocyte glycoprotein (MOG)-derived peptide: preliminary analysis of MOG T cell epitopes.
Eur. J. Immunol.
25
:
985
5
Mendel, I., N. Kerlero de Rosbo, A. Ben-Nun.
1995
. A myelin oligodendrocyte glycoprotein peptide induces typical chronic experimental autoimmune encephalomyelitis in H-2b mice: fine specificity and T cell receptor Vβ expression of encephalitogenic T cells.
Eur. J. Immunol.
25
:
1951
6
Caspi, R. R., F. G. Roberge, C. C. Chan, B. Wiggert, G. J. Chader, L. A. Rozenszajn, Z. Lando, R. B. Nussenblatt.
1988
. A new model of autoimmune disease: experimental autoimmune uveoretinitis induced in mice with two different retinal antigens.
J. Immunol.
140
:
1490
7
Hart, M. N., D. S. Linthicum, M. M. Waldschmidt, S. K. Tassell, R. L. Schelper, R. A. Robinson.
1987
. Experimental autoimmune inflammatory myopathy.
J. Neuropathol. Exp. Neurol.
46
:
511
8
Sewell, W. A., J. J. Munoz, M. A. Vadas.
1983
. Enhancement of the intensity, persistence, and passive transfer of delayed-type hypersensitivity lesions by pertussigen in mice.
J. Exp. Med.
157
:
2087
9
He, J., S. Gurunathan, A. Iwasaki, B. Ash-Shaheed, B. L. Kelsall.
2000
. Primary role for Gi protein signaling in the regulation of interleukin 12 production and the induction of T helper cell type 1 responses.
J. Exp. Med.
191
:
1605
10
Shive, C. L., H. Hofstetter, L. Arredondo, C. Shaw, T. G. Forsthuber.
2000
. The enhanced antigen-specific production of cytokines induced by pertussis toxin is due to clonal expansion of T cells and not to altered effector functions of long-term memory cells.
Eur. J. Immunol.
30
:
2422
11
Ryan, M., L. McCarthy, R. Rappuoli, B. P. Mahon, K. H. G. Mills.
1998
. Pertussis toxin potentiates Th1 and Th2 responses to co-injected antigen: adjuvant action is associated with enhanced regulatory cytokine production and expression of the co-stimulatory molecules B7-1, B7-2 and CD28.
Int. Immunol.
10
:
651
12
Limmer, A., J. Ohl, C. Kurts, H. G. Ljunggren, Y. Reiss, M. Groettrup, F. Momburg, B. Arnold, P. A. Knolle.
2000
. Efficient presentation of exogenous antigen by liver endothelial cells to CD8+ T cells results in antigen-specific T-cell tolerance.
Nat. Med.
6
:
1348
13
Steinman, R. M..
1991
. The dendritic cell system and its role in immunogenicity.
Annu. Rev. Immunol.
9
:
271
14
De Smedt, T., B. Pajak, E. Muraille, L. Lespagnard, E. Heinen, P. De Baetselier, J. Urbain, O. Leo, M. Moser.
1996
. Regulation of dendritic cell numbers and maturation by lipopolysaccharide in vivo.
J. Exp. Med.
184
:
1413
15
Sousa, C. R., S. Hieny, T. Scharton-Kersten, D. Jankovic, H. Charest, R. N. Germain, A. Sher.
1997
. In vivo microbial stimulation induces rapid CD40 ligand-independent production of interleukin 12 by dendritic cells and their redistribution to T cell areas.
J. Exp. Med.
186
:
1819
16
Reis e Sousa, C., A. Sher, P. Kaye.
1999
. The role of dendritic cells in the induction and regulation of immunity to microbial infection.
Curr. Opin. Immunol.
11
:
392
17
Trinchieri, G..
1998
. Interleukin-12: a cytokine at the interface of inflammation and immunity.
Adv. Immunol.
70
:
83
18
Ludewig, B., B. Odermatt, S. Landmann, H. Hengartner, R. M. Zinkernagel.
1998
. Dendritic cells induce autoimmune diabetes and maintain disease via de novo formation of local lymphoid tissue.
J. Exp. Med.
188
:
1493
19
Dittel, B. N., I. Visintin, R. M. Merchant, C. A. Janeway, Jr.
1999
. Presentation of the self antigen myelin basic protein by dendritic cells leads to experimental autoimmune encephalomyelitis.
J. Immunol.
163
:
32
20
Inaba, K., M. Inaba, N. Romani, H. Aya, M. Deguchi, S. Ikehara, S. Muramatsu, R. M. Steinman.
1992
. Generation of large numbers of dendritic cells from mouse bone marrow cultures supplemented with granulocyte/macrophage colony-stimulating factor.
J. Exp. Med.
176
:
1693
21
Kumar, R. K., R. O’Grady, W. Li, L. W. Smith, G. C. Rhodes.
1991
. Primary culture of adult mouse lung fibroblasts in serum-free medium: responses to growth factors.
Exp. Cell Res.
193
:
398
22
McKeehan, W. L..
1977
. The effect of temperature during trypsin treatment on viability and multiplication potential of single normal human and chicken fibroblasts.
Cell Biol. Int. Rep.
1
:
335
23
Linthicum, D. S., J. A. Frelinger.
1982
. Acute autoimmune encephalomyelitis in mice. II. Susceptibility is controlled by the combination of H-2 and histamine sensitization genes.
J. Exp. Med.
156
:
31
24
Caspi, R. R., F. G. Roberge, C. G. McAllister, M. el-Saied, T. Kuwabara, I. Gery, E. Hanna, R. B. Nussenblatt.
1986
. T cell lines mediating experimental autoimmune uveoretinitis (EAU) in the rat.
J. Immunol.
136
:
928
25
Rizzo, L. V., R. H. DeKruyff, D. T. Umetsu, R. R. Caspi.
1995
. Regulation of the interaction between Th1 and Th2 T cell clones to provide help for antibody production in vivo.
Eur. J. Immunol.
25
:
708
26
Lyons, A. B., C. R. Parish.
1994
. Determination of lymphocyte division by flow cytometry.
J. Immunol. Methods
171
:
131
27
Takayama, T., A. E. Morelli, N. Onai, M. Hirao, K. Matsushima, H. Tahara, A. W. Thomson.
2001
. Mammalian and viral IL-10 enhance C-C chemokine receptor 5 but down-regulate C-C chemokine receptor 7 expression by myeloid dendritic cells: impact on chemotactic responses and in vivo homing ability.
J. Immunol.
166
:
7136
28
Snedecor, G. W., W. G. Cochran.
1967
.
Statistical Methods
248
Iowa State University Press, Ames.
29
Sun, B., L. V. Rizzo, S. H. Sun, C. C. Chan, B. Wiggert, R. L. Wilder, R. R. Caspi.
1997
. Genetic susceptibility to experimental autoimmune uveitis involves more than a predisposition to generate a T helper-1-like or a T helper-2-like response.
J. Immunol.
159
:
1004
30
Silver, P. B., C. C. Chan, B. Wiggert, R. R. Caspi.
1999
. The requirement for pertussis to induce EAU is strain-dependent: B10.RIII, but not B10.A mice, develop EAU and Th1 responses to IRBP without pertussis treatment.
Invest. Ophthalmol. Visual Sci.
40
:
2898
31
Banchereau, J., R. M. Steinman.
1998
. Dendritic cells and the control of immunity.
Nature
392
:
245
32
Vecchi, A., L. Massimiliano, S. Ramponi, W. Luini, S. Bernasconi, R. Bonecchi, P. Allavena, M. Parmentier, A. Mantovani, S. Sozzani.
1999
. Differential responsiveness to constitutive vs. inducible chemokines of immature and mature mouse dendritic cells.
J. Leukocyte Biol.
66
:
489
33
Garcia, J. G., P. Wang, F. Liu, M. B. Hershenson, T. Borbiev, A. D. Verin.
2001
. Pertussis toxin directly activates endothelial cell p42/p44 MAP kinases via a novel signaling pathway.
Am. J. Physiol. Cell. Physiol.
280
:
C1233
34
Dudley, D. T., L. Pang, S. J. Decker, A. J. Bridges, A. R. Saltiel.
1995
. A synthetic inhibitor of the mitogen-activated protein kinase cascade.
Proc. Natl. Acad. Sci. USA
92
:
7686
35
Kalinski, P., C. M. Hilkens, E. A. Wierenga, M. L. Kapsenberg.
1999
. T-cell priming by type-1 and type-2 polarized dendritic cells: the concept of a third signal.
Immunol. Today
20
:
561
36
Ausiello, C. M., G. Fedele, F. Urbani, R. Lande, B. Di Carlo, A. Cassone.
2002
. Native and genetically inactivated pertussis toxins induce human dendritic cell maturation and synergize with lipopolysaccharide in promoting T helper type 1 responses.
J. Infect. Dis.
186
:
351
37
Jong, E. C., P. L. Vieira, P. Kalinski, J. H. N. Schuitemaker, Y. Tanaka, E. A. Wierenga, M. Yazdanbakhsh, M. L. Kapsenberg.
2002
. Microbial compounds selectively induce Th1 cell-promoting or Th2 cell-promoting dendritic cells in vitro with diverse Th cell-polarizing signals.
J. Immunol.
168
:
1704
38
Hofstetter, H. H., C. L. Shive, T. G. Forsthuber.
2002
. Pertussis toxin modulates the immune response to neuroantigens injected in incomplete Freund’s adjuvant: induction of Th1 cells and experimental autoimmune encephalomyelitis in the presence of high frequencies of Th2 cells.
J. Immunol.
169
:
117
39
Tarrant, T. K., P. S. Silver, C. C. Chan, B. Wiggert, R. R. Caspi.
1998
. Endogenous IL-12 required for induction and expression of experimental autoimmune uveitis.
J. Immunol.
161
:
122
40
Yokoi, H., K. Kato, T. Kezuka, J. I. Sakai.
1997
. Prevention of experimental autoimmune uveoretinitis by monclonal antibody to interleukin-12.
Eur. J. Immunol.
27
:
641
41
Kelleher, P., S. C. Knight.
1998
. IL-12 increases CD80 expression and the stimulatory capacity of bone marrow-derived dendritic cells.
Int. Immunol.
10
:
749
42
Zitvogel, L..
2002
. Dendritic and natural killer cells cooperate in the control/switch of innate immunity.
J. Exp. Med.
195
:
F9
43
Labeur, M. S., B. Roters, B. Pers, A. Mehling, T. A. Luger, T. Schwarz, S. Grabbe.
1999
. Generation of tumor immunity by bone marrow-derived dendritic cells correlates with dendritic cell maturation stage.
J. Immunol.
162
:
168
44
Eggert, A. A., M. W. Schreurs, O. C. Boerman, W. J. Oyen, A. J. de Boer, C. J. Punt, C. G. Figdor, G. J. Adema.
1999
. Biodistribution and vaccine efficiency of murine dendritic cells are dependent on the route of administration.
Cancer Res.
59
:
3340
45
Kondo, T., I. Cortese, S. Markovic-Plese, K. P. Wandinger, C. Carter, M. Brown, S. Leitman, R. Martin.
2001
. Dendritic cells signal T cells in the absence of exogenous antigen.
Nat. Immun.
2
:
932
46
Revy, P., M. Sospedra, B. Barbour, A. Trautmann.
2001
. Functional antigen-independent synapses formed between T cells and dendritic cells.
Nat. Immun.
2
:
925
47
Maldonado-López, R., T. De Smedt, P. Michel, J. Godfroid, B. Pajak, C. Heirman, K. Thielemans, O. Leo, J. Urbain, M. Moser.
1999
. CD8α+ and CD8α subclasses of dendritic cells direct the development of distinct T helper cells in vivo.
J. Exp. Med.
189
:
587
48
Smith, A. L., B. F. de St. Groth.
1999
. Antigen-pulsed CD8α+ dendritic cells generate an immune response after subcutaneous injection without homing to the draining lymph node.
J. Exp. Med.
189
:
593
49
Kaslow, H. R., D. L. Burns.
1992
. Pertussis toxin and target eukaryotic cells: binding, entry, and activation.
FASEB J.
6
:
2684
50
Lei, M. G., D. C. Morrison.
1993
. Evidence that lipopolysaccharide and pertussis toxin bind to different domains on the same p73 receptor on murine splenocytes.
Infect. Immun.
61
:
1359
51
Li, H., W. S. Wong.
2000
. Mechanisms of pertussis toxin-induced myelomonocytic cell adhesion: role of CD14 and urokinase receptor.
Immunology
100
:
502
52
Guha, M., N. Mackman.
2001
. LPS induction of gene expression in human monocytes.
Cell. Signal.
13
:
85
53
Rescigno, M., M. Martino, C. L. Sutherland, M. R. Gold, P. Ricciardi-Castagnoli.
1998
. Dendritic cell survival and maturation are regulated by different signaling pathways.
J. Exp. Med.
188
:
2175
54
Bouchon, A., C. Hernández-Munain, M. Cella, M. Colonna.
2001
. A DAP12-mediated pathway regulates expression of CC chemokine receptor 7 and maturation of human dendritic cells.
J. Exp. Med.
194
:
1111
55
Romagnani, S..
1992
. Induction of Th1 and Th2 responses: a key role for the “natural” immune responses?.
Immunol. Today
13
:
379
56
Douglas, T. F..
1996
. The instructive role of innate immunity in the acquired immune response.
Science
272
:
50