Influence of 2,3,7,8-Tetrachlorodibenzo-p-dioxin on the Antigen-Presenting Activity of Dendritic Cells

Beth A. Vorderstrasse*,1, Erica A. Dearstyne*,2 and Nancy I. Kerkvliet*,3

* Department of Environmental and Molecular Toxicology and Environmental Health Sciences Center, Oregon State University, Corvallis, Oregon 97331

Received September 18, 2002; accepted December 5, 2002


    ABSTRACT
 TOP
 ABSTRACT
 INTRODUCTION
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
We have previously shown that exposure of mice to 2,3,7, 8-tetrachlorodibenzo-p-dioxin (TCDD) induces activation-like changes in splenic dendritic cells (DC) in the absence of antigen challenge. Since activation of DC reduces their ability to phagocytize antigen, we examined the effects of TCDD on the ability of DC to process and present antigen to antigen-specific T cells and to internalize latex beads. Additionally, the expression of costimulatory and adhesion molecules was examined on DC from TCDD-treated mice injected with allogeneic tumor cells. The ability of DC from C57Bl/6 mice to induce proliferation of keyhole limpet hemocyanin (KLH)-specific 10.5.17 T cells and production of IL-4 was not significantly altered by TCDD exposure, either when KLH was addedin vitro or when the mice were injected with KLH prior to DC isolation. In contrast, ovalbumin (OVA) presentation by DC from TCDD-treated Balb/c mice induced enhanced proliferation of OVA-specific D011.10 T cells, although the production of IL-2 and IFN-{gamma} was not affected. Enhancedin vivo proliferation of adoptively transferred, CFSE-labeled DO11.10 T cells was also observed in TCDD-treated Balb/c mice that were challenged with OVA. TCDD treatment modulated the expression of major histocompatibility complex (MHC) class II, CD24, ICAM-1, CD40, and LFA-1 on splenic DC from C57Bl/6 mice injected with allogeneic tumor cells; however, the effects of TCDD were identical to changes seen previously in nonimmune mice, suggesting that these effects were not antigen-dependent. Finally, TCDD treatment did not affect the ability of splenic DC to internalize latex beads administeredin vivo. Taken together, these results suggest that the activation-like changes induced in DC by TCDD do not suppress the ability of DC to process and present antigen, but may enhance their ability to provide activation signals to T cells. This, in turn, may alter the survival of the T cells, the DC, or both, and might lead to dysregulation of the immune response.

Key Words: dendritic cells; TCDD; immunotoxicity; antigen presentation; T cell activation.


    INTRODUCTION
 TOP
 ABSTRACT
 INTRODUCTION
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
Dendritic cells (DC) are the most potent antigen-presenting cells (APC) and are particularly important in initiating primary T cell-mediated immune responses (Banchereau and Steinman, 1998Go). DC exist in different maturation stages, which are characterized by the functional capacity of the cells and the expression of key accessory molecules. Unactivated or immature DC have a high capacity for antigen capture but express relatively low levels of major histocompatibility complex (MHC) class II and costimulatory molecules such as B7-2 (CD86) and CD40, and thus are poor stimulators of naive T cells. When DC are activated by exposure to antigen, inflammatory stimuli, or bacterial products, they migrate to the T cell areas of lymphoid tissues and upregulate their expression of MHC and other adhesion and costimulatory molecules required for optimal T-cell activation (Winzler et al., 1997Go). In this activated state, DC have a significantly reduced capacity for antigen capture (Henderson et al., 1997Go; Reis e Sousa et al., 1993Go; Sallusto et al., 1995Go) but are instead specialized to induce T cells to proliferate and differentiate (Heufler et al., 1988Go; Labeur et al., 1999Go).

Suppression of T cell-dependent immune responses is one of the primary toxicities associated with exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD) (reviewed by Kerkvliet, 1998Go). However, despite intensive study, the immunological mechanisms of suppression remain poorly understood. Until recently, DC have been overlooked as a potential target for TCDD. Yet, it has been known for some time that TCDD and related congeners must be given during the early stages of the immune response in order for suppression to result (Kerkvliet and Brauner, 1987Go; Kerkvliet et al., 1996Go; Luster et al., 1988Go). This suggests that events occurring early in the generation of the response, such as activation of T cells by DC, are altered by exposure to TCDD. Additionally, immune responses that require antigen processing appear to be more sensitive to suppression than are mitogen- or superantigen-induced responses, which partially or completely bypass the need for normal APC function (Kerkvliet and Brauner, 1987Go; Neumann et al., 1993Go; Prell and Kerkvliet, 1997Go; Vecchi et al., 1980Go). Taken together, these data support the hypothesis that DC function is altered by TCDD.

Previously, we reported that, in the absence of antigen, TCDD exposure caused an apparent activation of DC (Vorderstrasse and Kerkvliet, 2001Go). Specifically, splenic DC from TCDD-treated mice produced more IL-12 and showed enhanced expression of ICAM-1, B7-2, CD24, and CD40, cell surface molecules which are characteristically upregulated following activation of the cells. In addition, DC from TCDD-treated mice enhanced T-cell proliferation and cytokine production when cultured with allogeneic T cells. Due to the immunosuppressive properties of TCDD, and the highly sensitive nature of T cell-dependent immune responses in particular, these results were unexpected and difficult to explain.

One possible explanation is that inappropriate preactivation of DC by TCDD negatively affects the function of the cells when processing and presentation of antigen is required. For example, activation of DC with stimuli such as TNF-{alpha}, CD40L, IL-1ß, and LPS diminishes their ability to internalize antigen (Henderson et al., 1997Go;Reis e Sousa et al., 1993Go; Sallusto et al., 1995Go). Furthermore, DC that have been activated with TNF-{alpha} in vitro or LPS in vivo prior to antigen exposure are significantly suppressed in their ability to subsequently activate antigen-specific T cells (De Smedt et al., 1996Go; Sallusto and Lanzavecchia, 1994Go).

In the studies presented here we have examined the effects of TCDD on DC function, specifically those involving antigen processing and presentation. We hypothesized that TCDD would suppress the ability of DC to activate T cells when processing and presentation of antigen was required. To test this hypothesis, we isolated DC from TCDD-treated mice and examined their ability to stimulate antigen-specific T cells ex vivo. In addition, the phagocytic activity of DC was evaluated using fluorescent beads. Finally, DC from mice injected with allogeneic tumor cells were examined to determine if TCDD exposure affects the expression of costimulatory molecules on DC from mice responding to antigen differently than DC from naïve mice.


    MATERIALS AND METHODS
 TOP
 ABSTRACT
 INTRODUCTION
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
Animal treatments.
C57Bl/6 mice were purchased from the Jackson Laboratory (Bar Harbor, ME). Balb/c mice were purchased from B&K Universal, Inc. (Kent, WA). DO11.10 transgenic mice on a Balb/c background (Pape et al., 1997Go) were originally provided by Dr. M. Jenkins (University of Minnesota Medical School, Minneapolis, MN) and were subsequently bred and maintained in our animal facility. The CD4+ T cells in the DO11.10 animals express a T-cell receptor that is specific for a peptide of ovalbumin (OVA) presented by MHC class II (I-Ad). The transgenic T cells from these mice can be identified using the monoclonal antibody KJ1-26 (Haskins et al., 1983Go). Mice were housed singly, and were maintained in front of a laminar flow unit in accordance with National Research Council guidelines. Mice were used at 7–12 weeks of age and were killed by CO2 overdose.

TCDD exposure.
TCDD (Cambridge Isotope Laboratories, Inc., Woburn, MA) was dissolved in anisole and diluted in peanut oil. The vehicle control consisted of an equivalent amount of anisole in peanut oil. Mice were treated with vehicle or 15 µg/kg TCDD by gavage. This dose of TCDD is known to suppress immune responses in mice (Kerkvliet et al., 1996Go), and was chosen based on a previous dose-response study examining the effects of TCDD on DC phenotype (Vorderstrasse and Kerkvliet, 2001Go).

Preparation of DC
Depending on the experimental design, DC were isolated from the spleens of vehicle- and TCDD-treated mice by various procedures described below. No consistent effect of TCDD on DC recovery was observed with any of the methods. Since the cells were obtained from TCDD-treated mice within 5 days of exposure, the loss of DC that occurs later (Vorderstrasse and Kerkvliet, 2001Go) was not a factor in these studies. Likewise, the median channel fluorescence of CD11c expression on the positive cells was not affected by treatment with TCDD.

For flow cytometric analysis.
DC were enriched from spleens using the method of Swiggard et al. (1992)Go with modifications as described in Inaba et al. (1997)Go. Briefly, splenic tissue was digested with collagenase D (Boehringer Mannheim, Indianapolis, IN) at 37°C for 45–60 min to release DC from the capsule and increase recovery. Recovered cells were spun over a BSA gradient (1.080 g/ml) and cells in the low-density fraction were collected. These freshly isolated DC-enriched preparations were then stained for flow cytometric analysis. Typically, 15–25% of the low-density cells expressed high levels of CD11c (CD11cHI), a marker for splenic DC (Crowley et al., 1990bGo)

For use as APC for 10.5.17 cells.
Low density spleen cells were obtained from C57Bl/6 mice as indicated above, and were further enriched for DC by overnight culturing based on their property of transient adherence to plastic (Swiggard et al., 1992Go). In this procedure, the low density cells were cultured in plastic dishes for 90 min to allow for the DC to adhere. The nonadherent contaminating cells were then washed away, and the remaining adherent cells left in culture overnight. DC become nonadherent during this culture period and were collected from media the following day. Purity after this final enrichment was greater than 80% CD11cHI cells.

For use as APC for DO11.10 T cells.
Spleens from Balb/c mice were digested with collagenase as indicated above. DC were enriched by incubating cell preparations with CD11c MicroBeads (Miltenyi Biotec, Auburn, CA) and subjecting the cells to magnetic separation following the manufacturer’s standard protocol. The concentration of DC was determined by flow cytometry. CD11cHI cells comprised 13–47% of the recovered cells. Cell suspensions were irradiated (1500 rads) and cell concentrations were adjusted such that a constant number of DC were added to each well.

10.5.17 T Cells
A TH2 cloned T-cell line (10.5.17), derived from a C57Bl/6 mouse and specific for keyhole limpet hemocyanin (KLH), was obtained from Barbara Fox (Immunologic Pharmaceutical Corp., Waltham, MA). For normal maintenance, 10.5.17 cells were restimulated every 10–14 days with irradiated C57Bl/6 spleen cells and KLH as previously described (Li and Fox, 1993Go). For proliferation and cytokine analysis, 10.5.17 cells were used 11 days after the last activation.

DC-Mediated Activation of 10.5.17 Cells
DC (1 x 104 cells per well) were cultured with 10.5.17 cells (1 x 104 per well) in standard 96-well tissue culture plates. KLH (Calbiochem, San Diego, CA) was either provided in vitro at 20 µg/ml or in vivo. For in vivo studies, DC were isolated from mice which had been injected ip with 4 mg KLH (in PBS) 3 h prior to sacrifice, as previously described (Crowley et al., 1990aGo). T cell proliferation was measured by incorporation of 3H-TdR that was added (1 µCi/well) during the last 20 h of culture. For IL-4 analysis, 60 µl supernatant was removed from each well just prior to addition of 3H-TdR (in 60 µl).

DC-Mediated Activation of DO11.10 T Cells
T cells were purified from spleens of DO11.10 T cell receptor transgenic mice using Cellect T-cell enrichment columns (Cytovax Biotechnologies Inc., Edmonton, Canada). Final T-cell purity was 80% CD4+ and 11% CD8+, and no B cells or MHC class II+ cells were detected. Various numbers of DC-enriched cells from Balb/c mice were cultured with T cells (3 x 105) and OVA protein (100 µg/ml, Sigma, St. Louis, MO) in 96-well Costar Ultra Low Cluster plates (Corning, Inc., Corning, NY). T-cell proliferation was measured by incorporation of 3H-TdR (1 µCi/well) that was added during the last 20 h of culture.

In Vivo Proliferation of DO11.10 T Cells
CFSE-labeling of DO11.10 splenocytes.
The proliferation of OVAspecific DO11.10 cells in vivo was monitored by labeling cells with 5- and 6-carboxyfluorescein diacetate, succinimidyl ester (CFSE) (Lyons and Parish, 1994Go). This dye freely diffuses into cells and becomes fluorescent and nonpermeable upon cleavage of the carboxyl groups by cellular esterases. CFSE is equally divided among daughter cells during cell division, thereby decreasing the fluorescence by half with each division. Single-cell suspensions of spleen cells were prepared from DO11.10 mice and were labeled by incubating the cells in 10 µM CFSE (Molecular Probes, Eugene, OR) for 8 min at room temperature in the dark. After labeling, the cells were adoptively transferred into syngeneic Balb/c hosts as described below.

Adoptive transfer.
Balb/c mice were treated with vehicle or TCDD 1 day prior to injection of DO11.10 cells as described previously (Kearney et al., 1994Go; Shepherd et al., 2000Go). Briefly, spleen cells from untreated DO11.10 mice were collected, and the percentage of CD4+/KJ1-26+ cells determined by flow cytometry. Balb/c recipient mice (sex and age matched) were injected iv with 5 x 106 CD4+/KJ1-26+ T cells (total spleen cells injected {approx} 2 x 107) in a volume of 0.5 ml. The adoptively transferred mice were rested for 2 days and then immunized ip with 2 mg OVA emulsified in complete Freund’s adjuvant (OVA-CFA).

Flow cytometric analysis of cell division.
Spleen cells from adoptively transferred mice were collected 1 or 3 days after injection of OVA-CFA. Cells were stained with anti-CD4 and KJ-126 antibodies to identify the DO11.10 cells, and analyzed by flow cytometry as described below. The CFSE content of the adoptively transferred cells was examined by establishing a gate on the CD4+/KJ1-26+ cells.

Flow Cytometry
Cells were incubated in 96-well plates with optimized concentrations of antibodies. Biotinylated-CD11c, as well as fluorochrome-conjugated antibodies to CD40, ICAM-1 (CD54), LFA-1 (CD11a), CD24, CD4, CD8, CD19, and I-Ab (MHCII) were obtained from BD PharMingen (San Diego, CA). Red670- and Red613-streptavidin were obtained from Gibco (Gaithersburg, MD). The KJ1-26 hybridoma (used to identify DO11.10 cells) was provided by Dr. M. Jenkins. KJ1-26 antibody was purified from culture supernatant using Protein A sepharose Fast Flow columns (Amersham Pharmacia) and biotinylated using N-hydroxysuccinimido-biotin (Sigma, St. Louis, MO). Pre-incubating the cells with rat, hamster, and/or mouse IgG blocked nonspecific antibody binding. Appropriately labeled isotype controls were used to determine nonspecific fluorescence. A viable cell gate was established based on propidium iodide exclusion. For DC cell surface molecule evaluation, 10,000 viable CD11cHI cells were analyzed. Listmode data were collected on a Coulter Epics XL flow cytometer and analyzed using WinList software (Verity Software House, Inc., Topsham, ME). WinList was also used occasionally for compensation during data analysis.

P815 Tumor Response
C57Bl/6 mice (H-2b) were injected ip with 1 x 107 allogeneic P815 mastocytoma cells (H-2d). Mice were sacrificed on days 1–4 postinjection for analysis of splenic DC. P815 cells were maintained by propagation in syngeneic DBA hosts.

Cytokine Analysis
Culture supernatants were analyzed using antibody sandwich ELISA techniques. IL-2, IFN-{gamma}, and IL-4 antibody pairs were purchased from BD PharMingen, and antibodies to the p40 subunit of mouse IL-12 were purchased from Genzyme (Cambridge, MA). The secondary biotinylated antibodies were complexed with avidin-peroxidase and visualized with 2,2'-azinobis[3-ethylbenzthiazoline-6-sulfonic acid] as substrate. Absorbance was read at 405 nm using a microplate reader (Bio-Tek Instruments, Inc., Winooski, VT).

In Vivo Uptake of Fluorescent Beads
FITC-fluorescent 2 µm latex beads (1 x 109; Polysciences, Warrington, PA) were injected via the tail vein. Twenty-four h later, spleens were digested with collagenase as described above, and uptake of beads by DC was analyzed by flow cytometry. Preliminary experiments were conducted to optimize the route of injection, dose, and optimum time for detection of the beads after injection.

Statistical Analysis
For most experiments, a Student’s t-test was used to compare means of vehicle-treated groups to TCDD-treated groups. For comparison of cell surface marker expression across multiple groups, analysis of variance modeling (ANOVA) was performed using SAS statistical software (SAS Institute, Inc., Cary, NC), and comparisons between means were made using the least significant difference (LSD) multiple comparison t-test. For all analyses, values of p <= 0.05 were considered significant.


    RESULTS
 TOP
 ABSTRACT
 INTRODUCTION
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
Expression of costimulatory molecules in DC from mice challenged with allogeneic tumor cells.
Previously we have shown that exposure of mice to TCDD, in the absence of antigen challenge, causes a phenotypic activation of splenic DC, resulting in increased expression of ICAM-1, CD24, CD40, and MHC class II. Among the markers examined, only LFA-1 was significantly decreased by TCDD (Vorderstrasse and Kerkvliet, 2001Go). Because the increased expression of such activation markers is generally considered to enhance the ability of DC to activate T cells, these observations were difficult to reconcile with the suppression of T cell-dependent immune responses observed in TCDD-treated mice. It was possible however, that TCDD exposure might affect DC differently when animals were also challenged with antigen. To address this possibility, we examined the effects of TCDD on DC from mice injected with allogeneic P815 tumor cells. The immune response to P815 tumor cells requires the activation of CD4+ T cells, presumably by DC, and is highly sensitive to suppression by TCDD (DeKrey and Kerkvliet, 1995Go; Kerkvliet et al., 1996Go).

Accessory molecule expression was evaluated on days 1 through 4 following tumor injection, as the activation of CD4+ T cells has been shown to occur during this time (Kerkvliet et al., 1996Go). Vehicle- and TCDD-treated nonimmune animals (not injected with P815 cells) were also included as controls.

Figure 1Go shows representative histograms of accessory molecule expression on DC evaluated 3 days after tumor injection. Similar results were observed on the other days examined. Injection of P815 cells alone did not significantly alter DC phenotype. Except for a small and transient increase in ICAM-1 expression on day 3, DC profiles from nonimmune mice treated with vehicle (Fig. 1AGo, dotted line) and P815-injected mice treated with vehicle (Fig. 2BGo, dotted line) were similar.



View larger version (34K):
[in this window]
[in a new window]
 
FIG. 1. Effect of TCDD on accessory molecule expression on splenic DC from P815-injected mice. C57Bl/6 mice were treated with vehicle (V, dotted line) or 15 µg/kg TCDD (T, thick solid line) 1 day prior to ip injection of allogeneic P815 tumor cells. Vehicle- and TCDD-treated nonimmune animals were included as controls. On days 1 through 4 following P815 injection, splenic tissue was digested with collagenase and DC were enriched by density gradient centrifugation. Low-density cell preparations were stained with antibodies to CD11c and indicated accessory molecules, and analyzed by flow cytometry. Representative histograms show the expression of each marker on DC from nonimmune mice (A), or on day 3 following tumor injection (B). All mice were sacrificed at the same time relative to TCDD exposure. A thin line shows staining of an isotype control antibody. Values on histograms indicate the staining intensity as median channel fluorescence (± SEM) (n = 3 per group). *Significant difference from vehicle.

 


View larger version (21K):
[in this window]
[in a new window]
 
FIG. 2. Effect of TCDD on DC activation of 10.5.17 T cells with antigen provided in vitro. Spleens were removed from C57BL/6 mice 3 days after treatment with vehicle (open bars) or 15 µg/kg TCDD (solid bars). Spleens from 3 animals were pooled and dendritic cells were isolated by collagenase digestion, density gradient centrifugation, and overnight plastic adherence. DC (1 x 104) were cultured with10.5.17 T-cell clones (1 x 104) and KLH (20 µg/ml). Proliferation (A) was measured at 48 and 72 h by 3H-TdR incorporation. IL-4 production (B) was measured by ELISA, using supernatant removed from well immediately prior to addition of 3H-TdR. No IL-4 was detected in control wells that lacked DC, T cells, and/or KLH. Data points represent the mean ± SEM (n = 4 per group). Data are representative of 3 independent experiments.

 
As expected, TCDD treatment of nonimmune mice (Fig. 1AGo) enhanced the expression of ICAM-1, CD24, CD40, and MHCII and decreased the expression of LFA-1 on DC, similar to previously published results (Vorderstrasse and Kerkvliet, 2001Go). Interestingly, nearly identical effects of TCDD were seen when DC from P815-injected animals were evaluated (Fig. 1BGo), suggesting that the presence of antigen per se does not modulate the effects of TCDD on DC phenotype.

Effect of TCDD on ability of DC to stimulate KLH-specific 10.5.17 cells.
In previous studies we found that TCDD enhanced the ability of DC to stimulate T-cell proliferation and cytokine production in an allogeneic mixed lymphocyte response (MLR), a response which does not require antigen processing. To evaluate DC function in a response that requires processing and presentation of antigen, we examined the ability of DC to activate KLH-specific cloned T cells (10.5.17). DC from vehicle- or TCDD-treated mice were cultured with 10.5.17 cells and KLH, and proliferation and IL-4 production were measured. As shown in Figure 2Go, TCDD exposure did not influence the ability of DC to process and present KLH to 10.5.17 T cells. Neither the proliferation of antigen-specific T cells nor the production of IL-4 was significantly different in cultures containing DC from vehicle- or TCDD-treated mice.

The method used to isolate DC in these experiments included an overnight culturing step, a procedure that has been shown to activate DC and reduce their ability to internalize antigen (Inaba et al., 1994Go;Reis e Sousa et al., 1993Go). Therefore it was possible that T-cell activation resulted from presentation of peptide fragments on surface MHC, and did not reflect the ability of DC to internalize antigen. To better assess the potential for TCDD to alter the uptake of antigen, we also tested the APC function of DC which had been exposed to KLH antigen in vivo. In these experiments, vehicle- and TCDD-treated mice were injected with PBS or KLH and sacrificed 3 h later. Splenic DC were isolated and cultured with 10.5.17 cells without additional antigen. As shown in Figure 3Go, DC from KLH-, but not PBS-injected mice stimulated proliferation of 10.5.17 cells, indicating that the DC had effectively phagocytized and processed KLH antigens in vivo. As in the experiments where KLH was provided in vitro, TCDD did not suppress the ability of DC from KLH-injected mice to stimulate proliferation of 10.5.17 cells. These results confirm that TCDD does not suppress the uptake and processing of antigen by DC.



View larger version (12K):
[in this window]
[in a new window]
 
FIG. 3. Effect of TCDD on the ability of DC from antigen-treated mice to activate 10.5.17 T cells. C57Bl/6 mice were treated with vehicle (open bars) or 15 µg/kg TCDD (black bars). One day later, mice were ip injected with 4 mg KLH in PBS, and spleens were removed after 3 h. Spleens from 3 animals were pooled and dendritic cells were isolated by collagenase digestion, density gradient centrifugation, and overnight plastic adherence. DC (1 x 104) were cultured with 10.5.17 T cells (1 x 104) without additional antigen, and proliferation was measured by 3H-TdR incorporation. Data points represent the mean ± SEM (n = 4 per group). Data are representative of five independent experiments.

 
TCDD enhances the ability of DC to stimulate naive OVA-specific DO11.10 T cells.
The 10.5.17 cloned T cells used in the previous experiments are maintained in long-term culture by repeated stimulation with APC and antigen, and therefore are representative of memory T cells. Because the requirements for activation of memory T cells appear to be less stringent than for naïve T cells (reviewed by Croft and Dubey, 1997Go), it is possible that activation of naive T cells might be impacted differently by TCDD-treated DC. To evaluate this possibility, we tested the ability of DC to activate naïve T cells purified from spleens of unimmunized, untreated DO11.10 transgenic mice. In addition, to reduce the activation of DC caused by the isolation method, magnetic beads were used for DC enrichment in order to avoid the overnight adherence procedure, and low adherence tissue culture plates were used for the T-cell stimulation assays.

As shown in Figure 4Go, the proliferation of OVA-specific T cells was not suppressed in the presence of DC from TCDD-treated mice, but was instead significantly enhanced compared to the response with DC from vehicle-treated mice. This enhancement of proliferation occurred independent of the time of assessment or number of DC added to the cultures. On the other hand, as shown in Figure 5Go, the production of T cell-derived cytokines, IL-2 and IFN-{gamma}, and the production of DC-derived IL-12 were not significantly different in cultures containing DC from vehicle- or TCDD-treated mice



View larger version (15K):
[in this window]
[in a new window]
 
FIG. 4. DC from TCDD-treated mice augment the proliferation of DO11.10 T cells. Balb/c mice were treated with vehicle control (open bars) or 15 µg/kg TCDD (black bars). Three days later, spleens were digested with collagenase and enriched for DC using CD11c MicroBeads. Resulting cell suspensions were analyzed by flow cytometry and adjusted such that indicated numbers of DC were added to each well. DC-enriched cells were cultured with DO11.10 T cells (3 x 105) from untreated mice, and OVA (100 µg/ml), and proliferation was measured by 3H-TdR incorporation. Proliferation in control wells (T cells + OVA), (DC-enriched cells + OVA), and (T cells + DC-enriched cells, no OVA), was below 800 cpm. Data points represent the mean ± SEM (n = 4 per group); *p <= 0.05 when compared to vehicle control.

 


View larger version (20K):
[in this window]
[in a new window]
 
FIG. 5. The production of IL-2, IFN-{gamma}, and IL-12 are similar in cultures containing DO11.10 T cells and DC from vehicle- or TCDD-treated mice. DC were enriched from vehicle-(open bars) or TCDD-(black bars) treated Balb/c mice as described in Figure 4Go. Resulting cell suspensions were analyzed by flow cytometry and adjusted such that 1 x 104 DC were added to wells with DO11.10 T cells (3 x 105) from untreated animals, and OVA (100 µg/ml). Cytokine production was measured by ELISA, and the dashed line indicates the limit of detection. For IL-2 and IFN-{gamma}, no cytokine production was detected in control cultures (DC + T cells, no OVA), (DC + OVA), or (T cells + OVA, no DC). IL-12 levels in control cultures were also at or below detection limits, except for the 90 h cultures of DC and T cells without OVA (V = 415 ± 49, T = 662 ± 40). Data points represent the mean ± SEM (n = 4 per group).

 
TCDD enhances DO11.10 T cell cycling in vivo.
To determine whether the increased proliferation of DO11.10 T cells observed in vitro also occurred in vivo, we tracked the cycling of CSFE-labeled D011.10 spleen cells following their adoptive transfer into syngeneic Balb/c recipients. Balb/c mice were treated with vehicle or TCDD 1 day prior to injection of CFSE-labeled spleen cells from untreated DO11.10 mice. Recipient mice were challenged with OVA-CFA, and CFSE fluorescence associated with the adoptively transferred CD4+/KJ1-26+ cells was examined by flow cytometry. As shown in representative histograms in Figure 6Go, on day 1 after antigen challenge the majority of the CD4+/KJ1-26+ cells fell in a single peak representing maximum CFSE fluorescence, indicating that the cells had not yet divided. In contrast, by day 3 after antigen challenge, fluorescence intensity associated with the CD4+/KJ1-26+ cells had diminished and discrete peaks representing increasing rounds of cell division were evident. When the percentage of cells in each peak was compared between vehicle- and TCDD-treated mice, TCDD exposure significantly increased the percentage of antigen-specific T cells that had undergone 6 or 7 rounds of division by day 3, while the percentage of cells that had divided only 4 or 5 times was significantly reduced. Overall, TCDD exposure resulted in a small but statistically significant increase in the mean number of cell divisions experienced by the antigen-specific T cells within the first 3 days following antigen exposure. Because treatment with TCDD did not alter the total number of CD4+/KJ1-26+ cells recovered on day 3, similar results were obtained when the number of cells in each division cycle was calculated (data not shown). These results are similar to the enhanced proliferation observed in vitro when DC from TCDD-treated mice were used to present antigen to DO11.10 T cells. However, since both the DC and CD4+ DO11.10 T cells, as well as other potential target cells, were exposed to TCDD in this in vivo model, it remains to be proven whether or not the in vivo enhancement is mediated by the DC.



View larger version (20K):
[in this window]
[in a new window]
 
FIG. 6. In vivo proliferation of DO11.10 T cells is enhanced in TCDD-treated mice. Balb/c mice were treated with vehicle or 15 µg/kg TCDD 1 day prior to injection of CFSE-labeled DO11.10 cells as described in Materials and Methods. OVA emulsified in CFA was injected ip, and spleen cells were collected on day 1 or 3 following antigen challenge. Cell division in the antigen-specific D011.10 T cells was examined by flow cytometry. Histograms representative of 4 mice per group show the CFSE staining on day 1 and day 3. The numbers above individual peaks indicate the number of cell divisions. The table shows the percentage of cells in each division cycle on day 3 following antigen challenge. Data in the table represent the mean of 4 mice per group; *p <= 0.05 when compared to vehicle control.

 
TCDD does not suppress the ability of DC to take up latex beads in vivo.
To further evaluate the potential for TCDD to affect phagocytosis, we measured the ability of DC in the spleen to take up fluorescently labeled latex beads that were injected intravenously. This in vivo approach also eliminated the complicating factor that phagocytic activity can be influenced by the DC isolation procedure (Inaba et al., 1994Go; Reis e Sousa et al., 1993Go). In the experiment shown in Figure 7Go, mice were injected iv with 1 x 109 FITC-beads in PBS, and were sacrificed after 24 h. Flow cytometric analysis of bead uptake was performed by gating on CD11cHI cells in BSA-enriched DC preparations. The number of beads associated with each cell was measured by flow cytometry, based on the linear relationship between the amount of fluorescence and the number of beads present. Figure 7AGo shows a representative histogram of the FITC-bead fluorescence in the CD11cHI cells, and indicates that bead-positive DC were usually found associated with a single bead. In vehicle-treated animals, approximately 4% of DC were associated with beads (Fig. 7BGo). TCDD exposure slightly, but significantly, increased the percentage of bead-positive cells to 5.4%; however, the number of bead-positive DC was not altered. To address the possibility that uptake of beads could affect the density of the DC, causing them to segregate differently in the BSA gradient, DC in unfractionated spleen cell preparations were also examined. TCDD did not alter the percentage or number of bead-positive DC in the unfractionated spleen cells (data not shown). Taken together with the results from studies where KLH was administered in vivo, these results demonstrate that internalization of antigen by DC is not suppressed by TCDD exposure.



View larger version (18K):
[in this window]
[in a new window]
 
FIG. 7. TCDD does not alter the ability of DC to internalize latex beads administered in vivo. C57Bl/6 mice were treated with vehicle or 15 µg/kg TCDD 3 days prior to iv injection of 1 x 109 FITC-fluorescent latex beads. Twenty-four h later, spleens were digested with collagenase and resulting cells fractionated by density gradient centrifugation. Cells in the low density BSA fraction were stained with CD11c, and analyzed by flow cytometry. (A) Representative histogram showing FITC fluorescence of bead-positive, CD11cHI cells. (B) Summary of percent and number of total bead-positive CD11cHI cells. (n = 3 or 4 animals per group); *p <= 0.05 when compared to vehicle control.

 

    DISCUSSION
 TOP
 ABSTRACT
 INTRODUCTION
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
In a previous report, we showed that in the absence of antigen, exposure to TCDD increases the expression of MHC class II molecules, the costimulatory molecules B7-2, CD40, and CD24, and the adhesion molecule ICAM-1 on DC. These phenotypic changes suggested that TCDD activates DC, a scenario which was consistent with our observations that DC from TCDD-treated mice produced more IL-12 and were better at stimulating the proliferation of allogeneic T cells in vitro than DC from vehicle-treated animals. Because the activation of DC reduces their ability to internalize antigen, we hypothesized that TCDD-induced activation of DC would inhibit the processing and presentation of antigen and thereby suppress antigen-dependent T cell activation.

In the current studies we examined the effects of TCDD on DC surface markers in mice challenged with antigen, and tested the ability of DC from TCDD-treated mice to present antigen to antigen-specific T cells. Interestingly, the results of the current studies do not support the hypothesis that exposure to TCDD suppresses the ability of DC to process and present antigen to T cells. Antigen-dependent T-cell proliferation and cytokine production was either unaffected or enhanced in the presence of DC from TCDD-treated mice. Likewise, the ability of DC to internalize latex beads was not affected by TCDD. These observations are consistent with results from other recent studies where TCDD did not suppress the early proliferation or phenotypic activation of OVA-specific CD4+ T cells in vivo in adoptively transferred mice (Shepherd et al., 2000Go).

Contrary to suppression of T-cell activation, exposure to TCDD enhanced the ability of DC to stimulate proliferation of antigen-specific T cells in vitro when naïve DO11.10 T cells were used as responders. Similarly, enhanced OVA-specific T-cell proliferation was observed in vivo when CFSE-labeled DO11.10 T cells were adoptively transferred into TCDD-treated Balb/c mice. Interestingly, recent studies indicate that following the initial T-cell expansion, an increased percentage of the DO11.10 T cells from TCDD-treated mice undergo apoptosis (Funatake et al., manuscript in preparation), suggesting that TCDD may induce a type of activation-induced cell death (AICD). If true, this conclusion would be consistent with the significantly reduced numbers of D011.10 T cells observed in TCDD-treated mice at later time points during the anti-OVA response (Shepherd et al., 2000Go). Similar results have been shown in mice treated in vivo with polyclonal T-cell activators (anti-CD3 or Staphyloccus enterotoxin A), where TCDD exposure resulted in enhanced T-cell proliferation followed by an apparent increase in death of CD4+ T cells (Camacho et al., 2001Go, 2002Go; Dearstyne and Kerkvliet, 2002Go; Prell et al., 1995Go). It is also possible that TCDD enhances the deletion of DC, since, once activated, DC are committed to undergo apoptosis (De Smedt et al., 1996Go, 1998Go; Winzler et al., 2000). Since DC and T cells must remain in close association for T cells to become fully activated, premature death of DC may contribute to premature death or incomplete activation of T cells (Josien et al., 2000Go; Miga et al., 2001Go). This possibility is supported by previous studies showing that exposure to TCDD significantly reduces the number of DC in the spleen (Vorderstrasse and Kerkvliet, 2001Go).

Although DC from TCDD-treated Balb/c mice enhanced the proliferation of the DO11.10 T cells relative to DC from vehicle-treated animals, the production of cytokines was not altered. This finding is in agreement with in vivo studies, where TCDD had little effect on the production of various T cell cytokines (Shepherd et al., 2000Go). However, these results contrast with our previous observations where DC from TCDD-treated C57Bl/6 mice produced more IL-12 and stimulated enhanced production of IFN-{gamma} and IL-2 when cultured with allogeneic T cells (Vorderstrasse and Kerkvliet, 2001Go). The reasons for these conflicting results are not clear. However, C57Bl/6 and Balb/c mice may respond to TCDD differently in terms of cytokine production since these strains are considered to be predisposed toward Th1 (C57Bl/6) and Th2 (Balb/c) responses (Brenner et al., 1994Go). Further studies will be needed to clarify such differential effects. Alternatively, different methods were used to enrich DC in the current study (magnetic beads) than in the previous studies using cells from C57Bl/6 mice (adherence to plastic). Thus it is possible that the method by which the DC were isolated in the different experiments contributed to the difference in cytokine production. For example, Vremec et al. (2000)Go recently reported that particular subsets of DC were selectively depleted when splenic DC were isolated by adherence to plastic. It is therefore possible that TCDD enhances IL-12 production in a subpopulation of DC that was selectively enriched in the experiments with C57Bl/6 mice. Alternatively, cytokine production may have been influenced by some other cell population, which contaminated the DC preparations used to activate the DO11.10 T cells. However, this is unlikely as we failed to find a relationship between T cell activation and the other potential antigen-presenting cells present in the DC preparations.

TCDD exposure did not suppress the expression of costimulatory molecules on splenic DC isolated from mice responding to challenge with allogeneic P815 tumor cells. This result was somewhat unexpected since previous studies have shown that TCDD suppresses the expression of CD86 on B220+ and Mac-1+ cells in the same model (Prell and Kerkvliet, 1997Go; Shepherd et al., 2001Go). However, the effect of TCDD on CD86 expression was not seen until later in the response, suggesting that the changes were not associated with the disruption of early T-cell activation events. In contrast to suppression, in the current studies, TCDD significantly increased the expression of CD40, CD24, ICAM-1, and MHC class II on DC on days 1–4 after P815 tumor challenge. This increased expression was identical to the increased expression of accessory molecules on DC from naïve mice treated with TCDD, suggesting that the changes noted were not antigen-induced. Since injection of P815 cells alone did not alter accessory molecule expression, it also suggests that the subpopulation of DC that are directly involved in processing and presenting P815 alloantigens to CD4+ T cells was below detection limits. Therefore it remains possible that TCDD influences accessory molecule expression on the actual antigen-bearing DC, which might influence T-cell activation in this model.

In summary, although we hypothesized that the inappropriate activation of DC by TCDD would suppress their ability to internalize antigen and activate T cells, results from the current studies do not support this conclusion. Instead we have found that TCDD-induced activation of DC occurs both with and without antigen and does not appear to inhibit the ability of the DC to activate T cells. Because treatment with TCDD caused a phenotypic activation of DC and enhanced their ability to stimulate the proliferation of naïve DO11.10 T cells, further studies are warranted to determine whether TCDD-induced activation of DC plays a role in suppression of T cell-dependent immune responses.


    ACKNOWLEDGMENTS
 
We thank Linda Steppan and Julie Oughton for outstanding technical support. In addition, we thank B. Paige Lawrence for comments on this manuscript, Jennifer Ferguson and Cliff Pereira for assistance with statistical analysis, and the Environmental Health Sciences Center for use of the flow cytometer. This research was supported by grants from the National Institute of Environmental Health Sciences (ES03966, ES00040, and NIEHS Center grant ES00210), and Training grant ES07060 (B.A.V. and E.A.D.).


    NOTES
 
1 Present address: Department of Pharmaceutical Sciences, Wegner Hall, Washington State University, Pullman, WA 99164. Back

2 Present address: NeoRx Corporation, 300 Elliott Avenue West, Suite 500, Seattle, WA 98119. Back

3 To whom correspondence should be sent at 1007 Agricultural and Life Sciences Building, Oregon State University, Corvallis, OR 97331. Fax: (541) 737-0497. E-mail: nancy.kerkvliet{at}orst.edu. Back


    REFERENCES
 TOP
 ABSTRACT
 INTRODUCTION
 MATERIALS AND METHODS
 RESULTS
 DISCUSSION
 REFERENCES
 
Banchereau, J., and Steinman, R. M. (1998). Dendritic cells and the control of immunity. Nature 392, 245–252.[CrossRef][ISI][Medline]

Brenner, G. J., Cohen, N., and Moynihan, J. A. (1994). Similar immune response to nonlethal infection with herpes simplex virus-1 in sensitive (BALB/c) and resistant (C57BL/6) strains of mice. Cell. Immunol. 157, 510–524.[CrossRef][ISI][Medline]

Camacho, I. A., Hassuneh, M. R., Nagarkatti, M., and Nagarkatti, P. S. (2001). Enhanced activation-induced cell death as a mechanism of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced immunotoxicity in peripheral T cells. Toxicology 165, 51–63.[CrossRef][ISI][Medline]

Camacho, I.A., Nagarkatti, M., and Nagarkatti, P.S. (2002). 2,3,7,8-Tetrachlorodibenzo-p-dioxin (TCDD) induces Fas-dependent activation-induced cell death in superantigen-primed T cells. Arch. Toxicol. 76, 570–580.[CrossRef][ISI][Medline]

Croft, M., and Dubey, C. (1997). Accessory molecule and costimulation requirements for CD4 T cell response. Crit. Rev. Immunol. 17, 89–118.[ISI][Medline]

Crowley, M., Inaba, K., and Steinman, R. M. (1990a). Dendritic cells are the principal cells in mouse spleen bearing immunogenic fragments of foreign proteins. J. Exp. Med. 172, 383–386.[Abstract]

Crowley, M. T., Inaba, K., Witmer-Pack, M. D., Gezelter, S., and Steinman, R. M. (1990b). Use of the fluorescence activated cell sorter to enrich dendritic cells from mouse spleen. J. Immunol. Methods 133, 55–66.[CrossRef][ISI][Medline]

De Smedt, T., Pajak, B., Klaus, G. G. B., Noelle, R. J., Urbain, J., Leo, O., and Moser, M. (1998). Antigen-specific T lymphocytes regulate lipopolysaccharide-induced apoptosis of dendritic cells in vivo. J. Immunol. 161, 4476–4479.[Abstract/Free Full Text]

De Smedt, T., Pajak, B., Muraille, E., Lespagnard, L., Heinen, E., Debaetselier, P., Urbain, J., Leo, O., and Moser, M. (1996). Regulation of dendritic cell numbers and maturation by lipopolysaccharide in vivo. J. Exp. Med. 184, 1413–1424.[Abstract]

Dearstyne, E. A., and Kerkvliet, N. I. (2002). Mechanism of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)-induced decrease in anti-CD3-activated CD4(+) T cells: The roles of apoptosis, Fas, and TNF. Toxicology 170, 139–151.[CrossRef][ISI][Medline]

DeKrey, G. K., and Kerkvliet, N. I. (1995). Suppression of cytotoxic T lymphocyte activity by 2,3,7,8-tetrachlorodibenzo-p-dioxin occurs in vivo but not in vitro, and is independent of corticosterone elevation. Toxicology 97, 105–112.[CrossRef][ISI][Medline]

Haskins, K., Kubo, R., White, J., Pigeon, M., Kappler, J., and Marrack, P. (1983). The major histocompatibility complex-restricted antigen receptor on T cells: I. Isolation with a monoclonal antibody. J. Exp. Med. 157, 1149–1169.[Abstract]

Henderson, R. A., Watkins, S. C., and Flynn, J. L. (1997). Activation of human dendritic cells following infection with Mycobacterium tuberculosis. J. Immunol. 159, 635–643.[Abstract]

Heufler, C., Koch, F., and Schuler, G. (1988). Granulocyte/macrophage colony-stimulating factor and interleukin 1 mediate the maturation of murine epidermal Langerhans cells into potent immunostimulatory dendritic cells. J. Exp. Med. 167, 700–705.[Abstract]

Inaba, K., Pack, M., Inaba, M., Sakuta, H., Isdell, F., and Steinman, R. M. (1997). High levels of a major histocompatibility complex II-self peptide complex on dendritic cells from the T-cell areas of lymph nodes. J. Exp. Med. 186, 665–672.[Abstract/Free Full Text]

Inaba, K., Witmer Pack, M., Inaba, M., Hathcock, K. S., Sakuta, H., Azuma, M., Yagita, H., Okumura, K., Linsley, P. S., Ikehara, S., Muramatsu, S., Hodes, R. J., and Steinman, R. M. (1994). The tissue distribution of the B7–2 costimulator in mice: Abundant expression on dendritic cells in situ and during maturation in vitro. J. Exp. Med. 180, 1849–1860.[Abstract]

Josien, B. R., Li, H. L., Ingulli, E., Sarma, S., Wong, R., Vologodskaia, M., Steinman, R. M., and Choi, Y. (2000). TRANCE, a tumor necrosis factor family member, enhances the longevity and adjuvant properties of dendritic cells in vivo. J. Exp. Med. 191, 495–502.[Abstract/Free Full Text]

Kerkvliet, N. I. (1998). T-lymphocyte subpopulations and TCDD immunotoxicity. In T-Lymphocyte Subpopulations in Immunotoxicology. (I. Kimber and M. K. Selgrade, Eds.), pp. 55–72. John Wiley & Sons, New York.

Kerkvliet, N. I., Baecher-Steppan, L., Shepherd, D. M., Oughton, J. A., Vorderstrasse, B. A., and Dekrey, G. K. (1996). Inhibition of TC-1 cytokine production, effector cytotoxic T lymphocyte development, and alloantibody production by 2,3,7,8- tetrachlorodibenzo-p-dioxin. J. Immunol. 157, 2310–2319.[Abstract]

Kerkvliet, N. I., and Brauner, J. A. (1987). Mechanisms of 1,2,3,4,6,7,8-heptachlorodibenzo-p-dioxin (HpCDD)-induced humoral immune suppression: Evidence of primary defect in T-cell regulation. Toxicol. Appl. Pharmacol. 87, 18–31.[CrossRef][ISI][Medline]

Kearney, E. R., Pape, K. A., Loh, D. Y., and Jenkins, M. K. (1994). Visualization of peptide-specific T-cell immunity and peripheral tolerance induction in vivo. Immunity 1, 327–339.[ISI][Medline]

Labeur, M. S., Roters, B., Pers, B., Mehling, A., Luger, T. A., Schwarz, T., and Grabbe, S. (1999). Generation of tumor immunity by bone marrow-derived dendritic cells correlates with dendritic cell maturation stage. J. Immunol. 162, 168–175.[Abstract/Free Full Text]

Li, T., and Fox, B. S. (1993). Effect of prostaglandin E2 (PGE2) on IL-3/granulocyte-macrophage, colony-stimulating factor production by T helper cells. J. Immunol. 150, 1680–1690.[Abstract/Free Full Text]

Luster, M. I., Germolec, D. R., Clark, G., Wiegand, G., and Rosenthal, G. J. (1988). Selective effects of 2,3,7,8-tetrachlorodibenzo-p-dioxin and corticosteroid on in vitro lymphocyte maturation. J. Immunol. 140, 928–935.[Abstract/Free Full Text]

Lyons, A. B., and Parish, C. R. (1994). Determination of lymphocyte division by flow cytometry. J. Immunol. Methods 171, 131–137.[CrossRef][ISI][Medline]

Miga, A. J., Masters, S. R., Durell, B. G., Gonzales, M., Jenkins, M. K., Maliszewski, C., Kikutani, H., Wade, W. F., and Noelle, R. J. (2001). Dendritic-cell longevity and T-cell persistence is controlled by CD154-CD40 interactions. Eur. J. Immunol. 31, 959–965.[CrossRef][ISI][Medline]

Neumann, C. M., Oughton, J. A., and Kerkvliet, N. I. (1993). Anti-CD3-induced T-cell activation–II. Effect of 2,3,7,8- tetrachlorodibenzo-p-dioxin (TCDD). Int. J. Immunopharmacol. 15, 543–550.[CrossRef][ISI][Medline]

Pape, K. A., Kearney, E. R., Khoruts, A., Mondino, A., Merica, R., Chen, Z. M., Ingulli, E., White, J., Johnson, J. G., and Jenkins, M. K. (1997). Use of adoptive transfer of T-cell-antigen-receptor-transgenic T cells for the study of T-cell activation in vivo. Immunol. Rev. 156, 67–78.[ISI][Medline]

Prell, R. A., and Kerkvliet, N. I. (1997). Involvement of altered B7 expression in dioxin immunotoxicity: B7 transfection restores the CTL but not the alloantibody response to the P815 mastocytoma. J. Immunol. 158, 2695–2703.[Abstract]

Prell, R. A., Oughton, J. A., and Kerkvliet, N. I. (1995). Effect of 2,3,7,8-tetrachlorodibenzo-p-dioxin on anti-CD3-induced changes in T-cell subsets and cytokine production. Int. J. Immunopharmacol. 17, 951–961.[CrossRef][ISI][Medline]

Reis e Sousa, C., Stahl, P. D., and Austyn, J. M. (1993). Phagocytosis of antigens by Langerhans cells in vitro. J. Exp. Med. 178, 509–519.[Abstract]

Sallusto, F., Cella, M., Danieli, C., and Lanzavecchia, A. (1995). Dendritic cells use macropinocytosis and the mannose receptor to concentrate macromolecules in the major histocompatibility complex class II compartment: Downregulation by cytokines and bacterial products. J. Exp. Med. 182, 389–400.[Abstract]

Sallusto, F., and Lanzavecchia, A. (1994). Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor {alpha}. J. Exp. Med. 179, 1109–1118.[Abstract]

Shepherd, D. M., Dearstyne, E. D., and Kerkvliet, N. I. (2000). The effects of TCDD on the activation of ovalbumin (OVA)-specific DO11. 10 transgenic T cells in adoptively transferred mice. Toxicol. Sci. 56, 340–350.[Abstract/Free Full Text]

Shepherd, D. M., Steppan, L. B., Hedstrom, O. R., and Kerkvliet, N. I. (2001). Anti-CD40 treatment of 2,3,7,8-tetrachlorodibenzo-p-dioxin (TCDD)- exposed C57Bl/6 mice induces activation of antigen presenting cells yet fails to overcome TCDD-induced suppression of allograft immunity. Toxicol. Appl. Pharmacol. 170, 10–22.[CrossRef][ISI][Medline]

Swiggard, W. J., Nonacs, R. M., Witmer Pack, M. D., Steinman, R. M., and Inaba, K. (1992). Enrichment of dendritic cells by plastic adherence and EA rosetting. In Current Protocols in Immunology (J. E. Coligan, A. M. Kruisbeck, D. H. Margulies, E. M. Shevach, and W. Strober, Eds.), pp. 3.7.1–3.7.11. John Wiley & Sons, New York.

Vecchi, A., Mantovani, A., Sironi, M., Luini, W., Cairo, M., and Garattini, S. (1980). Effect of acute exposure to 2,3,7,8-tetrachlorodibenzo-p-dioxin on humoral antibody production in mice. Chem. Biol. Interact. 30, 337–342.[CrossRef][ISI][Medline]

Vorderstrasse, B. A., and Kerkvliet, N. I. (2001).2,3,7,8-Tetrachlorodibenzo-p-dioxin affects the number and function of murine splenic dendritic cells and their expression of accessory molecules. Toxicol. Appl. Pharmacol. 171, 117–125.[CrossRef][ISI][Medline]

Vremec, D., Pooley, J., Hochrein, H., Wu, L., and Shortman, K. (2000). CD4 and CD8 expression by dendritic cell subtypes in mouse thymus and spleen. J .Immunol. 164, 2978–2986.[Abstract/Free Full Text]

Winzler, C., Rovere, P., Rescigno, M., Granucci, F., Penna, G., Adorini, L., Zimmerman, V. S., Davoust, J., and Ricciardi-Castagnoli, P. (1997). Maturation stages of mouse dendritic cells in growth factor-dependent long-term cultures. J. Exp. Med. 185, 317–328.[Abstract/Free Full Text]