Dendritic cells (DCs) are important orchestrators of the immune response, ensuring that immunity against pathogens is generated, whereas immunity against healthy tissues is prevented. Using the tumor Ag MUC1, we previously showed that i.v. immunization of MUC1 transgenic mice, but not wild-type, with a MUC1 peptide resulted in transient tolerization of all splenic DCs. These DCs did not upregulate costimulatory molecules and induced regulatory T cells rather than effector T cells. They were characterized by suppressed expression of a cohort of pancreatic enzymes not previously reported in DCs, which were upregulated in DCs presenting the same MUC1 peptide as a foreign Ag. In this article, we examined the self-antigen–tolerized DC phenotype, function, and mechanisms responsible for inducing or maintaining their tolerized state. Tolerized DCs share some characteristics with immature DCs, such as a less inflammatory cytokine/chemokine profile, deficient activation of NF-κB, and sustained expression of zDC and CCR2. However, tolerized DCs demonstrated a novel inducible expression of aldehyde dehydrogenase 1/2 and phospho-STAT3. Suppressed expression of one of the pancreatic enzymes, trypsin, in these DC impeded their ability to degrade extracellular matrix, thus affecting their motility. Suppressed metallopeptidases, reflected in low expression of carboxypeptidase B1, prevented optimal Ag-specific CD4+ T cell proliferation suggesting their role in Ag processing. Tolerized DCs were not refractory to maturation after stimulation with a TLR3 agonist, demonstrating that this tolerized state is not terminally differentiated and that tolerized DCs can recover their ability to induce immunity to foreign Ags.

Dendritic cells (DC) can induce potent immunity by presenting foreign Ags derived from infectious agents or abnormal self-antigens derived from tumors (1). However, DCs can also maintain tolerance to self-antigen derived from healthy tissues, environmental agents, and commensal microbiota (2, 3). Ablation of DCs during homeostasis results in a breakdown of CD4+ T cell tolerance with ensuing fatal autoimmunity (4, 5). Multiple mechanisms assist DCs in maintaining peripheral tolerance to self-antigen, such as low expression of MHC class II (MHC II), costimulatory molecules CD40, CD80, and CD86, production of anti-inflammatory molecules such as IL-10, IDO, and retinoic acid (RA), exposure to low Ag dose, and expression of cell-surface molecules such as DEC-205 and CD103 that mark DCs that preferentially prime peripheral regulatory T cells (Tregs) (2, 3, 68).

Vaccines designed to induce immunity against tumors often use Ags that are closely related to self-antigens or are a combination of normal (self) and abnormal (foreign) epitopes, resulting in a suboptimal immune response (9). Immune responses to vaccines are controlled by many different mechanisms in the periphery. Better understanding of these mechanisms is important for designing vaccines that elicit the desired outcome: strong, protective immunity against pathogens and cancer, or tolerance of self-antigens to avoid autoimmunity.

We have been studying immune responses to cancer vaccines based on peptides derived from the human tumor Ag MUC1 that range from those closest to self (i.e., peptides with no sugar moieties) to those representing “abnormal self” (i.e., peptides decorated with tumor-specific, O-linked GalNAc adducts) (10, 11). We recently published that i.v. immunization of human MUC1 transgenic (MUC1.Tg) mice with an unglycosylated human MUC1 peptide (MUC1p) resulted in the transient tolerization (between 24 and 72 h) of the entire splenic DC population that failed to upregulate costimulatory molecules, lacked motility, and preferentially primed Foxp3+ Tregs. These vaccine-tolerized DCs also differed in their gene expression profile from vaccine-activated (immunogenic) DCs by suppressed expression of a group of “pancreatic” enzymes, specifically trypsin and carboxypeptidase B1 (CPB1) (12), which we proposed as predictive biomarkers of vaccine outcome. These results were consistent with the previously observed low level of vaccine-induced immunity against MUC1p in MUC1.Tg mice (10, 13, 14).

In this study, we elucidated several molecular and cellular pathways that characterize DCs tolerized by vaccination with MUC1p as a self-antigen. We show that, in contrast with endogenous splenic DCs recovered from vaccinated wild-type (WT) mice where MUC1p is a foreign Ag, splenic DCs from vaccinated MUC1.Tg mice where MUC1p is a self-antigen show inhibition of NF-κB whereas simultaneously increasing signaling through phospho-STAT3. As a result, they produce fewer inflammatory cytokines and chemokines. We also found that these DCs induce expression of aldehyde dehydrogenase 1/2 (Aldh1/2), the enzyme catalyzing the final step in the biosynthesis of immunosuppressive RA. Inhibition of Aldh before immunization of MUC1.Tg mice with MUC1p reverses DC tolerance as seen by increased expression of trypsin and CPB1 to levels observed in MUC1p vaccinated WT mice.

We also examined the functional consequence of the suppressed expression of “pancreatic” proteases in tolerized DCs. We show that increased expression by DCs of metallopeptidases, such as CBP1, is required for optimal proliferation of MUC1p-specific CD4+ T cells, suggesting a role for these enzymes in increasing efficiency of Ag processing and presentation. We found that trypsin expression, in contrast, is required for degradation of the extracellular matrix (ECM), facilitating DC motility. Suppressed expression of these enzymes in tolerized DCs contributes to their low T cell stimulatory capacity and low motility, resulting in low or no response to MUC1p when it is presented as a self-antigen.

C57BL/6 (WT) mice were purchased from The Jackson Laboratory. MUC1.Tg mice were originally obtained from Dr. Sandra Gendler (Mayo Clinic, Scottsdale, AZ) (15) and subsequently bred at the University of Pittsburgh. All colonies were housed under specific pathogen-free conditions. Experiments were approved by the Institutional Animal Care and Use Committee at the University of Pittsburgh.

A 100-mer MUC1p represents 5 repeats of the 20-aa sequence HGVTSAPDTRPAPGSTAPPA from the MUC1 VNTR region. It was synthesized as described previously by the University of Pittsburgh Genomics and Proteomics Core Laboratories (13).

Bone marrow–derived DCs (BMDCs) were generated according to a previously reported protocol (10). In brief, female C57BL/6 mice (Jackson) were sacrificed and their femurs and tibiae harvested. Marrow was flushed with RPMI 1640 and cells were passed through a 70-μM strainer and pelleted before RBC lysis using ACK buffer. Cells were resuspended in AIM-V (Life Technologies), counted, and plated at 1.25 × 106 cells/ml in AIM-V containing 20 ng/ml GM-CSF (Miltenyi Biotec). On days 3 and 5, half of the media was replaced with fresh AIM-V and GM-CSF. On day 6 of culture, DCs were harvested with 2 mM EDTA, counted, and (when indicated) loaded with 30 μg/ml MUC1 100-mer and matured overnight with 30 μg/ml polyinosinic-polycytidylic acid and poly-l-lysine (poly-ICLC; Hiltonol), a generous gift from Oncovir. On day 7, cells were harvested as described earlier. For immunizations, day 7 DCs were washed and resuspended in sterile PBS. Mice were immunized i.v. via the lateral tail vein with 1 × 106 DCs.

BMDCs were cultured overnight in the presence of 10 or 100 μM N-orthophenanthroline (Sigma) or 100 μg/ml trypsin inhibitor from chicken OVA (Sigma), 30 μg/ml MUC1p 100-mer, and 10 ng/ml GM-CSF (Miltenyi). The following day, MUC1p-specific TCR.Tg CD4+ T cells labeled with 5 μM CFSE (Invitrogen) were added to culture at a 5:1 CD4/DC ratio. After 3 d of coculture, cells were harvested and T cell proliferation determined by CFSE dilution on an LSR II cytometer (BD).

Mice were given 100 mg/kg 4-diethylaminobenzaldehyde (DEAB) (Sigma) in 100 μl DMSO (i.p.) to inhibit Aldh1 activity as previously described (16). Immediately afterward, mice were immunized with unloaded DCs or MUC1-loaded DCs as described earlier.

RNA was extracted from bead-isolated CD11c+ splenic DCs (Miltenyi) using an RNeasy Mini kit (Qiagen) according to the manufacturer’s protocol. Reverse transcription was performed using Oligo(dT) primes and SuperScript III reverse transcriptase (Invitrogen). cDNA was amplified using the following primers: CCR2 (forward: 5′-CTGCAAAGACCAGAAGAGGGC-3′, reverse: 5′-CCACCACCCAAGTGACTACA-3′), suppressor of cytokine signaling 1 (SOCS1; forward: 5′-CCAGCCTGCGCCTCAAGACC-3′, reverse: 5′-GCGTGCTTCGGGGGTCACTC-3′), Aldh1 (forward: 5′-CCATGCCGGGCGAGGTGAAG-3′, reverse: 5′-TCCGGGTGGAAAGCCAGCCT-3′), and zDC (forward: 5′-AGAGAGCACATGAAGCGACA-3′, reverse: 5′-CTGGCTGCAGACATGAACAC-3′). Quantitative PCR was conducted using a QuantiTect SYBR Green Kit (Qiagen). Reactions were run on a StepOne Plus cycler (Applied Biosystems) with data generated using ΔCT methodology.

Cells were lysed in RIPA buffer and proteins run on 10% Tris-HCL Mini-PROTEAN TGX gels (Bio-Rad) followed by transfer onto a polyvinylidene difluoride membrane. Membranes were blocked in 5% milk followed by overnight incubation at 4°C with one of the following Abs: Rb X-trypsin (M-60), Rb X-CPB1 (M-134), Ms X-Aldh1/2 (H-8), Rb X-IκB-α (C-21) or Rb X-p-STAT3 (Ser727) (Santa Cruz), and Rb X-p-p65 (93H1) (Cell Signaling).

DCs were MACS (Miltenyi) isolated from spleens 24 h post-MUC1p immunization of WT and MUC1.Tg mice. DCs were then cultured overnight in the presence of 500 ng/ml LPS (Sigma) and GolgiPlug (BD). The following day, cells were lysed and total DC protein queried for cytokine/chemokine expression using a Proteome Profiler Array (R&D Systems). Densitometry was conducted on blots using ImageJ (National Institutes of Health).

DCs were MACS isolated (Miltenyi) from spleens 24 h postimmunization of WT and MUC1.Tg mice. Cells (1 × 105) were immediately plated into eight-well chamber slides coated with FITC-gelatin using the QCM Gelatin Invadopodia Kit (Millipore). Images were collected on an Olympus Provis fluorescent microscope with a 40× objective. Areas of FITC-gelatin degradation were quantified at 4 and 24 h using binary thresholding in ImageJ (National Institutes of Health). Where indicated, DCs were cultured for indicated time points in the presence of 10 μM N-orthophenanthroline and/or 100 μg/ml trypsin inhibitor from chicken OVA (Sigma).

As we previously reported, splenic DCs recovered from MUC1.Tg mice immunized i.v. with MUC1p express lower levels of MHC II, CD40, and CD86, compared with DCs from WT mice immunized with the same peptide. They also preferentially prime naive CD4+ T cells into Tregs versus IFN-γ–producing effector T cells. In addition, there are fewer DCs 24 h postimmunization in the spleens of MUC1.Tg mice compared with WT (12). Low costimulatory molecule expression and priming of Tregs are hallmarks of immature DCs (iDCs) (2, 17, 18). We now show, using a cytokine blot array, that DCs recovered from MUC1p-immunized MUC1.Tg mice produce lower levels of proinflammatory cytokines (i.e., IL-1a, TNF-α, IL-6) relative to WT (Fig. 1A, 1B). These tolerized DCs also produced less chemokines such as MIP-1b, RANTES, and IL-16, as well as less CXCL1, MIP-2, and CCL2. Tolerized DCs failed to downregulate expression of CCR2, the receptor for CCL2 and a phenotypic marker of iDCs (19, 20) (Fig. 1C). CCR2 was functional as shown by the enhanced ex vivo migration of CCR2hi DCs in a linear CCL2 gradient (Supplemental Fig. 1).

FIGURE 1.

DCs isolated from MUC1p-immunized MUC1.Tg mice are less inflammatory than DCs from immunized WT mice. (A) WT and MUC1.Tg mice (n = 3/group) were immunized (i.v.) with DCs loaded with MUC1p (1 × 106 cells). Twenty-four hours later, isolated splenic DCs were cultured overnight in the presence of 500 ng/ml LPS and brefeldin A. DC lysates were then incubated on a mouse cytokine blot array that was developed according to the manufacturer’s instructions. Blots show global expression of all 40 chemokines and cytokines queried with a global preponderance of decreased expression in DCs from immunized MUC1.Tg mice. (B) Densitometry of the blots from (A) quantified with ImageJ. (C) Quantitative PCR performed for CCR2 on DCs 24 h postvaccination. Data are normalized to MUC1p-immunized WT mice, with bars representing the mean ± SEM. Data are representative of two (A) and three (C) independent experiments.

FIGURE 1.

DCs isolated from MUC1p-immunized MUC1.Tg mice are less inflammatory than DCs from immunized WT mice. (A) WT and MUC1.Tg mice (n = 3/group) were immunized (i.v.) with DCs loaded with MUC1p (1 × 106 cells). Twenty-four hours later, isolated splenic DCs were cultured overnight in the presence of 500 ng/ml LPS and brefeldin A. DC lysates were then incubated on a mouse cytokine blot array that was developed according to the manufacturer’s instructions. Blots show global expression of all 40 chemokines and cytokines queried with a global preponderance of decreased expression in DCs from immunized MUC1.Tg mice. (B) Densitometry of the blots from (A) quantified with ImageJ. (C) Quantitative PCR performed for CCR2 on DCs 24 h postvaccination. Data are normalized to MUC1p-immunized WT mice, with bars representing the mean ± SEM. Data are representative of two (A) and three (C) independent experiments.

Close modal

SOCS1/3 and histone deacetylase 11 (HDAC11) have been implicated in suppressing proinflammatory cytokine production and promoting IL-10 expression, respectively, in DCs (21, 22). Although SOCS1 expression was decreased by ∼50% in DCs recovered from MUC1p-vaccinated WT mice, as would be expected of a DC developing an immunostimulatory phenotype, its expression was unchanged in DCs from immunized MUC1.Tg mice (Fig. 2A). HDAC11 levels in immunized WT or MUC1.Tg mice were not significantly different (Fig. 2B). This suggested that these two transcription factors, which play a role in DCs that are actively immunosuppressive, do not have the same role in either inducing or maintaining the transient tolerized state of DCs from vaccinated MUC1.Tg mice. We also examined the transcription factor Zbtb46 (zDC) that has recently been identified as a conventional DC-lineage marker whose expression is inversely correlated with DC maturational status (5, 23, 24). Expression of zDC was∼2.5-fold higher in tolerized DCs recovered from immunized MUC1.Tg mice compared with WT (Fig. 2C). Expression of zDC and CCR2 decreased after stimulation of DC with poly-ICLC ex vivo demonstrating that these DCs are not refractory to maturation (Fig. 2C, 2D).

FIGURE 2.

SOCS1 and HDAC11 do not contribute to the tolerized state, whereas sustained expression of Zbtb46 and CCR2 maintains an iDC phenotype. WT and MUC1.Tg mice (n = 3/group) were immunized (i.v.) with DC:MUC1p (MUC1p; 1 × 106 cells) or unloaded DCs (Ctrl). One day later, isolated DCs were pooled for quantitative RT-PCR for SOCS1 (A) or HDAC11 (B). Bars represent mean ± SEM after normalization to respective control vaccinations and are representative of three independent experiments. (C and D) Mice were immunized with MUC1p as in (A); splenic DCs were isolated at 24 h and were left untreated (No Tx) or treated with 30 μg/ml poly-ICLC for 4 h before performing quantitative PCR for zDC (C) or CCR2 (D).

FIGURE 2.

SOCS1 and HDAC11 do not contribute to the tolerized state, whereas sustained expression of Zbtb46 and CCR2 maintains an iDC phenotype. WT and MUC1.Tg mice (n = 3/group) were immunized (i.v.) with DC:MUC1p (MUC1p; 1 × 106 cells) or unloaded DCs (Ctrl). One day later, isolated DCs were pooled for quantitative RT-PCR for SOCS1 (A) or HDAC11 (B). Bars represent mean ± SEM after normalization to respective control vaccinations and are representative of three independent experiments. (C and D) Mice were immunized with MUC1p as in (A); splenic DCs were isolated at 24 h and were left untreated (No Tx) or treated with 30 μg/ml poly-ICLC for 4 h before performing quantitative PCR for zDC (C) or CCR2 (D).

Close modal

Activation of STAT3 characterizes DCs that are unable to prime efficient Th1 responses and is considered a negative regulator of DC function (25, 26). Splenic DCs isolated from MUC1p-immunized MUC1.Tg mice upregulated phospho-STAT3 24 h postimmunization (Fig. 3A). Conversely, NF-κB pathway activation that results in degradation of IκBα and phosphorylation of p65 is critical for DC phenotypic maturation and immunogenic function (27, 28). Tolerized DCs from immunized MUC1.Tg mice express less phospho-p65 (Fig. 3B) with a concurrent increase in total IκBα (Fig. 3C).

FIGURE 3.

Deficient NF-κB activation and enhanced STAT3 signaling in tolerized DCs. WT and MUC1.Tg mice (n = 3/group) were immunized (i.v.) with DC:MUC1p (MUC1p; 1 × 106 cells) or unloaded DCs (Ctrl). Twenty-four hours later, isolated splenic DCs were pooled and whole-cell lysates were analyzed by Western blots for phospho-STAT3 (A), phospho-p65, (B) or total IκBα (C). β-Actin is shown as a loading control for each blot. Data are representative of two to three independent experiments for each molecule.

FIGURE 3.

Deficient NF-κB activation and enhanced STAT3 signaling in tolerized DCs. WT and MUC1.Tg mice (n = 3/group) were immunized (i.v.) with DC:MUC1p (MUC1p; 1 × 106 cells) or unloaded DCs (Ctrl). Twenty-four hours later, isolated splenic DCs were pooled and whole-cell lysates were analyzed by Western blots for phospho-STAT3 (A), phospho-p65, (B) or total IκBα (C). β-Actin is shown as a loading control for each blot. Data are representative of two to three independent experiments for each molecule.

Close modal

Aldh catalyzes the final biosynthetic step in RA production in multiple populations of DCs residing in the gut, skin, and lung (29). RA plays an important role in maintaining tolerance to oral and commensal-derived Ags (30). We found that splenic DCs from immunized MUC1.Tg mice expressed both Aldh1 and Aldh1/2 (Fig. 4A–C), whereas those recovered from immunized WT mice did not. This Aldh was biologically active as determined by its ability to oxidize aminoacetaldehyde to aminoacetate (data not shown). Inhibition of Aldh1 activity before MUC1p vaccination of MUC1.Tg mice using the specific inhibitor DEAB led to reconstitution of normal expression of trypsin and CPB1 to levels observed in WT mice immunized with MUC1p as a foreign Ag (Fig. 4D). These data implicate Aldh activity and likely the subsequent production of RA as being required for self-antigen vaccine-induced tolerization of splenic DCs.

FIGURE 4.

Production of Aldh by tolerized DCs. WT and MUC1.Tg mice (n = 3/group) were immunized (i.v.) with DC:MUC1p (MUC1p; 1 × 106 cells) or unloaded DCs (Ctrl). Twenty-four hours later, isolated DCs were pooled and quantitative RT-PCR (qRT-PCR) performed for Aldh1 (A). Concurrently, whole-cell lysates from recovered DCs were Western blotted for Aldh1/2 (B), and bands were quantified via densitometry (C). (D) MUC1.Tg mice were immunized as described earlier immediately after injection of the Aldh inhibitor DEAB (MUC1p+DEAB). Twenty-four hours later, isolated splenic DCs were pooled and qRT-PCR performed for Trypsin and CPB1. Bars represent mean ± SEM after normalization to respective control vaccinations (A, D). Data are representative of two (D) and three (A–C) independent experiments.

FIGURE 4.

Production of Aldh by tolerized DCs. WT and MUC1.Tg mice (n = 3/group) were immunized (i.v.) with DC:MUC1p (MUC1p; 1 × 106 cells) or unloaded DCs (Ctrl). Twenty-four hours later, isolated DCs were pooled and quantitative RT-PCR (qRT-PCR) performed for Aldh1 (A). Concurrently, whole-cell lysates from recovered DCs were Western blotted for Aldh1/2 (B), and bands were quantified via densitometry (C). (D) MUC1.Tg mice were immunized as described earlier immediately after injection of the Aldh inhibitor DEAB (MUC1p+DEAB). Twenty-four hours later, isolated splenic DCs were pooled and qRT-PCR performed for Trypsin and CPB1. Bars represent mean ± SEM after normalization to respective control vaccinations (A, D). Data are representative of two (D) and three (A–C) independent experiments.

Close modal

Expression of pancreatic proteases by splenic DCs is coordinately regulated such that immunization of WT mice with MUC1p results in a 10- to 40-fold increase in their expression by 24 h, whereas the same immunization in MUC1.Tg mice results in their profound suppression. We found that this expression profile is a predictive biomarker of DC immunogenicity and the ensuing T effector or Treg responses (Supplemental Fig. 2) (10, 12). We explored the potential function of these enzymes, focusing on CBP1 and trypsin as representatives of two main families of enzymes, metallopeptidases (represented by CBP1) and serine proteases (represented by trypsin). We hypothesized that both might be involved in the processing and/or presentation of MHC II–restricted peptides derived from MUC1p, because a number of serine, aspartyl, and cysteine peptidases, as well as asparaginyl endopeptidase, have been reported to be involved in this pathway (31). We loaded BMDCs overnight with MUC1p in the presence of n-orthophenanthroline, an inhibitor of all metallopeptidases including CPB1, or a trypsin-specific inhibitor derived from chicken OVA. Neither inhibitor had an effect on DC viability as determined by Annexin V/propidium iodide staining (Supplemental Fig. 3). MUC1p-loaded DCs were then cocultured for 3 d with MUC1p-specific TCR.Tg CD4+ T cells (10) labeled with CFSE to quantify proliferation. Fig. 5A shows that inhibition of the metallopeptidase CPB1 inhibited CD4+ T cell proliferation, suggesting less efficient Ag processing and/or presentation. Inhibition of trypsin did not have any effect.

FIGURE 5.

Activity of the metallopeptidase CPB1 in DCs is required for optimal proliferation of MUC1p-specific CD4+ T cells. (A) BMDCs were cultured overnight in the presence of 30 μg/ml MUC1p and 30 μg/ml poly-ICLC with or without protease inhibitors (no inhibition, 10 or 100 μM orthophenanthroline, and 100 μg/ml trypsin inhibitor, respectively). The following day, naive CFSE-labeled, MUC1p-specific CD4+ T cells were added at a 1:5 DC/T cell ratio and cultured for 3 d. Cells were then harvested and percent proliferation determined by CFSE dilution. Histograms are from one experiment and are representative of two independent experiments. (B) Quantification of pooled data from two independent experiments. Bars represent mean ± SEM with **p ≤ .001, and ***p ≤ .0001.

FIGURE 5.

Activity of the metallopeptidase CPB1 in DCs is required for optimal proliferation of MUC1p-specific CD4+ T cells. (A) BMDCs were cultured overnight in the presence of 30 μg/ml MUC1p and 30 μg/ml poly-ICLC with or without protease inhibitors (no inhibition, 10 or 100 μM orthophenanthroline, and 100 μg/ml trypsin inhibitor, respectively). The following day, naive CFSE-labeled, MUC1p-specific CD4+ T cells were added at a 1:5 DC/T cell ratio and cultured for 3 d. Cells were then harvested and percent proliferation determined by CFSE dilution. Histograms are from one experiment and are representative of two independent experiments. (B) Quantification of pooled data from two independent experiments. Bars represent mean ± SEM with **p ≤ .001, and ***p ≤ .0001.

Close modal

DCs in lymphoid and nonlymphoid tissues are known to remodel ECM proteins such as collagen, elastin, and fibronectin by using secreted or membrane-bound proteases (32). Because the optimal pH of both trypsin and CPB1 activity are within physiologic range (7.5, and 7.9, respectively) (33, 34), we cultured DCs and assayed the supernatants for secreted trypsin and CPB1 by Western blotting. Both proteins were secreted by 40 h with the majority of CPB1 being the 45-kDa proenzyme form. There were multiple trypsin bands detected with at least one of them corresponding to an active 23-kDa enzyme (Fig 6A). To determine whether there was a difference in the ability to degrade ECM between DCs recovered from MUC1p-immunized WT mice that upregulate these enzymes and DCs from MUC1p-immunized MUC1.Tg mice that do not, we plated both DC populations on FITC-gelatin–coated slides and examined them 4 h later. The regions of the slide devoid of FITC fluorescence correspond to the amount of gelatin degraded by the DCs. DCs from immunized WT mice were able to degrade more FITC-gelatin compared with those from immunized MUC1.Tg mice (Fig. 6B). When we then cultured DCs on FITC-gelatin in the presence of a specific trypsin inhibitor derived from chicken OVA, the ability of DCs from immunized WT mice to degrade the matrix was impeded (Fig. 6C).

FIGURE 6.

Trypsin activity in DCs enhances degradation of the ECM. (A) Splenic DCs were isolated from naive WT mice and cultured for 40 h in the presence or absence of 30 μg/ml poly-ICLC. Cell-free supernatants were collected and Western blotted for the presence of trypsin and CPB1. Data are representative of three independent experiments. (B) WT and MUC1.Tg mice (n = 2/group) were immunized (i.v.) with DC:MUC1p (MUC1p; 1 × 106 cells) or unloaded DCs (Ctrl). Two days later, splenic DCs were isolated, pooled, and plated onto chamber slides coated with an FITC-gelatin matrix in the presence or absence of 100 μg/ml trypsin inhibitor. Cells were fixed after 4 h, and images were collected on an epifluorescent microscope and quantified with ImageJ (C). Bars represent mean ± SEM with ***p ≤ .0001. Data are representative of two independent experiments.

FIGURE 6.

Trypsin activity in DCs enhances degradation of the ECM. (A) Splenic DCs were isolated from naive WT mice and cultured for 40 h in the presence or absence of 30 μg/ml poly-ICLC. Cell-free supernatants were collected and Western blotted for the presence of trypsin and CPB1. Data are representative of three independent experiments. (B) WT and MUC1.Tg mice (n = 2/group) were immunized (i.v.) with DC:MUC1p (MUC1p; 1 × 106 cells) or unloaded DCs (Ctrl). Two days later, splenic DCs were isolated, pooled, and plated onto chamber slides coated with an FITC-gelatin matrix in the presence or absence of 100 μg/ml trypsin inhibitor. Cells were fixed after 4 h, and images were collected on an epifluorescent microscope and quantified with ImageJ (C). Bars represent mean ± SEM with ***p ≤ .0001. Data are representative of two independent experiments.

Close modal

We previously showed that immunization with a self-antigen inhibits, within the first 24 h, the maturation and immunogenicity of all splenic DCs in the target lymphoid organ and not just the relatively few DCs that present the Ag (12). This rapid tolerization of all DCs is likely due to the increased frequency of pre-existing self-antigen–specific Tregs (12, 35). In this article, we report that early and transiently tolerized DCs have a less inflammatory cytokine/chemokine profile that results in impaired DC-mediated inflammation and chemotaxis. Specifically, tolerized DCs produced less CCL2, suggesting an inability to recruit additional CD11c+CCR2+ cells. DCs have been reported to rely on β-catenin, SOCS1/3, NF-κB, RAR/RXR, HDAC11, zDC, and STAT3 to induce and sustain various anti-inflammatory programs (2, 5, 21, 3638). We found that activated DCs isolated from MUC1p-immunized WT mice downregulated SOCS1 as expected, but tolerized DCs from MUC1.Tg mice did not increase expression, suggesting that SOCS signaling was not involved in maintaining their tolerized state. Similarly, expression of HDAC11, known to inversely correlate with IL-10 transcription (22), did not decrease in MUC1.Tg DCs and there was no increase of IL-10 relative to WT DCs (Supplemental Fig. 4).

The transcription factor Zbtb46 (zDC), a repressor of DC maturation, was expressed ∼2.5-fold higher in tolerized DCs. Importantly, the expression of zDC decreased when tolerized DCs were stimulated with a TLR3 agonist directly ex vivo, indicating that these cells maintain the potential for maturation when removed from in vivo suppressive signals, and are thus only transiently tolerized. This would allow the host to control a potentially autoimmune response against self-antigen whereas preserving the ability to prime a T cell response against foreign Ag.

DCs from immunized MUC1.Tg mice did not produce more IL-10 than those from WT (Supplemental Fig. 4), yet they demonstrated increased phosphorylation of STAT3, suggesting that a paracrine source of IL-10 was contributing to their tolerized state. We previously showed that both Tregs and IL-10 actively suppress expression of trypsin and CPB1 in splenic DCs and, by extension, contribute to tolerizing those DCs. Because MUC1.Tg mice have a higher frequency of MUC1p-specific Tregs (35), Tregs and paracrine IL-10 may act on DCs to induce STAT3 phosphorylation. IL-6 also signals through STAT3; however, tolerized DCs produce less IL-6 compared with immunogenic DCs and there is no increase in the IL-6 transcript in the spleens of WT or MUC1.Tg mice postimmunization (data not shown). The cellular source of this IL-10 remains to be identified. STAT3 can also directly inhibit NF-κB signaling, as well as the production of IL-6 and TNF-α, as observed in DCs recovered from immunized MUC1.Tg mice (39).

We show for the first time, to our knowledge, that self-antigen vaccine tolerized DCs express Aldh1/2, the enzymes responsible for oxidizing retinaldehyde to immunosuppressive RA. CD11c+CD103+ cells from the gut have been shown to produce RA for the maintenance of tolerance to commensal flora, and RA plays a role in oral tolerance to food Ags (30). RA is also produced by CD11c+CD103 in the lung and dermis (29), demonstrating that this molecule is not restricted to the gut and that CD103 may not be a marker of RA-producing DCs. Induction of Aldh or RA by DCs in a secondary lymphoid organ in response to vaccination with a self-antigen has not, to our knowledge, been demonstrated. Importantly, we identified a novel mechanistic link between Aldh activity and the relative immunogenicity of splenic DCs after immunization with self-antigen. In the absence of Aldh activity, DCs from MUC1p-immunized MUC1.Tg mice are not tolerized, as determined by levels of trypsin and CPB1 that resemble those observed after vaccination with a foreign rather than a self-antigen. Because enhanced expression of trypsin and CPB1 by splenic DCs is a biomarker of DCs that induces effective ensuing immunity (12), we conclude that Aldh, and likely subsequent production of RA, actively suppress the development of immunogenic DCs. These data show that Aldh production in the spleen is required for vaccine-induced tolerance in secondary lymphoid organs in addition to tolerance to oral and commensal Ags.

Our finding that metallopeptidases, such as CPB1, are necessary for DCs to prime an optimal Ag-specific CD4+ T cell response suggested that CPB1 might be involved in the generation of MHC II–restricted MUC1ps. The majority of endolysosomal peptidases involved in generating peptides to be loaded onto MHC II are cathepsins, consisting of cysteine, serine, and aspartyl peptidases. It is possible that the metallopeptidase inhibitor we used, n-orthophenanthroline, also affected other susceptible enzymes involved in the MHC II pathway. However, there are currently no known metallopeptidases reported to be involved in MHC II processing and presentation, and only two carboxypeptidases, suggesting that in this particular pathway, CPB1 can be implicated as playing a role directly or indirectly in increasing the efficiency of MHC II–restricted MUC1p presentation (40, 41).

Our previous data showed that DCs recovered from MUC1p-immunized MUC1.Tg mice were less motile ex vivo compared with those from WT. We wanted to determine whether this was due to a defect in the ability of DCs to migrate through the ECM. Because ECM degradation is dependent on extracellular protease expression, we examined DCs and found that they were able to secrete trypsin and CPB1, making them candidates for remodeling ECM proteins. Inhibition of trypsin decreased the ability of DCs recovered from immunized WT mice (immunogenic DCs) to degrade gelatin, suggesting that it is necessary for remodeling of the ECM. In vivo it is likely that suppressed expression of trypsin in tolerized DCs from immunized MUC1.Tg mice retards their ability to migrate to T cell areas within the spleen, as the majority of DCs are initially localized in the red pulp and marginal zone (42). We also observed that the 45-kDa proenzyme form of CPB1 was secreted into DC supernatant, whereas an active 23-kDa trypsin isoform was secreted. Because trypsin activates the CPB1 zymogen (43), it is likely that both enzymes are functional in the extracellular space. The active suppression of trypsin and CPB1 expression, and the resulting effects on DC gelatinolysis and T cell priming, also constitute part of the novel phenotype of tolerized DCs.

Together, our data demonstrate that vaccination with self-antigen has a rapid and global tolerizing effect on DCs within the draining lymphoid tissue. Once tolerized, these DCs possess some phenotypic hallmarks of iDCs as well as other novel features that preclude their immunogenicity, such as expression of Aldh1/2 and phospho-STAT3, and suppressed expression of trypsin and CPB1. Understanding of early events after immunization will ultimately allow for better rational design of vaccines for cancer and autoimmune diseases.

We thank Douglas M. Marvel for insightful discussions, Dr. Sandra Cascio for expert advice, and Jia Xue for technical assistance. We also gratefully acknowledge the expertise and instrumentation provided by Dr. Simon Watkins at the University of Pittsburgh Center for Biologic Imaging.

This work was supported by National Institutes of Health Grants RO1 CA168392 (to O.J.F.) and T32 CA082084 (to A.M.F.) and by University of Pittsburgh Cancer Center Core Support Grant 2P30 CA47904.

The online version of this article contains supplemental material.

Abbreviations used in this article:

Aldh

aldehyde dehydrogenase

BMDC

bone marrow–derived dendritic cell

CPB1

carboxypeptidase B1

DC

dendritic cell

DEAB

4-diethylaminobenzaldehyde

ECM

extracellular matrix

HDAC

histone deacetylase

iDC

immature DC

MHC II

MHC class II

MUC1p

MUC1 peptide

poly-ICLC

polyinosinic-polycytidylic acid and poly-l-lysine

RA

retinoic acid

SOCS

suppressor of cytokine signaling

Tg

transgenic

Treg

regulatory T cell

WT

wild-type.

1
Steinman
R. M.
,
Banchereau
J.
.
2007
.
Taking dendritic cells into medicine.
Nature
449
:
419
426
.
2
Manicassamy
S.
,
Pulendran
B.
.
2011
.
Dendritic cell control of tolerogenic responses.
Immunol. Rev.
241
:
206
227
.
3
Coombes
J. L.
,
Siddiqui
K. R.
,
Arancibia-Cárcamo
C. V.
,
Hall
J.
,
Sun
C. M.
,
Belkaid
Y.
,
Powrie
F.
.
2007
.
A functionally specialized population of mucosal CD103+ DCs induces Foxp3+ regulatory T cells via a TGF-beta and retinoic acid-dependent mechanism.
J. Exp. Med.
204
:
1757
1764
.
4
Ohnmacht
C.
,
Pullner
A.
,
King
S. B. S.
,
Drexler
I.
,
Meier
S.
,
Brocker
T.
,
Voehringer
D.
.
2009
.
Constitutive ablation of dendritic cells breaks self-tolerance of CD4 T cells and results in spontaneous fatal autoimmunity.
J. Exp. Med.
206
:
549
559
.
5
Meredith
M. M.
,
Liu
K.
,
Kamphorst
A. O.
,
Idoyaga
J.
,
Yamane
A.
,
Guermonprez
P.
,
Rihn
S.
,
Yao
K. H.
,
Silva
I. T.
,
Oliveira
T. Y.
, et al
.
2012
.
Zinc finger transcription factor zDC is a negative regulator required to prevent activation of classical dendritic cells in the steady state.
J. Exp. Med.
209
:
1583
1593
.
6
Steinman
R. M.
,
Nussenzweig
M. C.
.
2002
.
Avoiding horror autotoxicus: the importance of dendritic cells in peripheral T cell tolerance.
Proc. Natl. Acad. Sci. USA
99
:
351
358
.
7
Turner
M. S.
,
Kane
L. P.
,
Morel
P. A.
.
2009
.
Dominant role of antigen dose in CD4+Foxp3+ regulatory T cell induction and expansion.
J. Immunol.
183
:
4895
4903
.
8
Yamazaki
S.
,
Dudziak
D.
,
Heidkamp
G. F.
,
Fiorese
C.
,
Bonito
A. J.
,
Inaba
K.
,
Nussenzweig
M. C.
,
Steinman
R. M.
.
2008
.
CD8+ CD205+ splenic dendritic cells are specialized to induce Foxp3+ regulatory T cells.
J. Immunol.
181
:
6923
6933
.
9
Banchereau
J.
,
Klechevsky
E.
,
Schmitt
N.
,
Morita
R.
,
Palucka
K.
,
Ueno
H.
.
2009
.
Harnessing human dendritic cell subsets to design novel vaccines.
Ann. N. Y. Acad. Sci.
1174
:
24
32
.
10
Ryan
S. O.
,
Turner
M. S.
,
Gariépy
J.
,
Finn
O. J.
.
2010
.
Tumor antigen epitopes interpreted by the immune system as self or abnormal-self differentially affect cancer vaccine responses.
Cancer Res.
70
:
5788
5796
.
11
Vlad
A. M.
,
Muller
S.
,
Cudic
M.
,
Paulsen
H.
,
Otvos
L.
 Jr.
,
Hanisch
F.-G.
,
Finn
O. J.
.
2002
.
Complex carbohydrates are not removed during processing of glycoproteins by dendritic cells: processing of tumor antigen MUC1 glycopeptides for presentation to major histocompatibility complex class II-restricted T cells.
J. Exp. Med.
196
:
1435
1446
.
12
Farkas
A. M.
,
Marvel
D. M.
,
Finn
O. J.
.
2013
.
Antigen choice determines vaccine-induced generation of immunogenic versus tolerogenic dendritic cells that are marked by differential expression of pancreatic enzymes.
J. Immunol.
190
:
3319
3327
.
13
Turner
M. S.
,
Cohen
P. A.
,
Finn
O. J.
.
2007
.
Lack of effective MUC1 tumor antigen-specific immunity in MUC1-transgenic mice results from a Th/T regulatory cell imbalance that can be corrected by adoptive transfer of wild-type Th cells.
J. Immunol.
178
:
2787
2793
.
14
Rowse
G. J.
,
Tempero
R. M.
,
VanLith
M. L.
,
Hollingsworth
M. A.
,
Gendler
S. J.
.
1998
.
Tolerance and immunity to MUC1 in a human MUC1 transgenic murine model.
Cancer Res.
58
:
315
321
.
15
Peat
N.
,
Gendler
S. J.
,
Lalani
N.
,
Duhig
T.
,
Taylor-Papadimitriou
J.
.
1992
.
Tissue-specific expression of a human polymorphic epithelial mucin (MUC1) in transgenic mice.
Cancer Res.
52
:
1954
1960
.
16
Mahmoud
M. I.
,
Potter
J. J.
,
Colvin
O. M.
,
Hilton
J.
,
Mezey
E.
.
1993
.
Effect of 4-(diethylamino)benzaldehyde on ethanol metabolism in mice.
Alcohol. Clin. Exp. Res.
17
:
1223
1227
.
17
Yamazaki
S.
,
Bonito
A. J.
,
Spisek
R.
,
Dhodapkar
M.
,
Inaba
K.
,
Steinman
R. M.
.
2007
.
Dendritic cells are specialized accessory cells along with TGF- for the differentiation of Foxp3+ CD4+ regulatory T cells from peripheral Foxp3 precursors.
Blood
110
:
4293
4302
.
18
Benson
M. J.
,
Pino-Lagos
K.
,
Rosemblatt
M.
,
Noelle
R. J.
.
2007
.
All-trans retinoic acid mediates enhanced T reg cell growth, differentiation, and gut homing in the face of high levels of co-stimulation.
J. Exp. Med.
204
:
1765
1774
.
19
Sallusto
F.
,
Schaerli
P.
,
Loetscher
P.
,
Schaniel
C.
,
Lenig
D.
,
Mackay
C. R.
,
Qin
S.
,
Lanzavecchia
A.
.
1998
.
Rapid and coordinated switch in chemokine receptor expression during dendritic cell maturation.
Eur. J. Immunol.
28
:
2760
2769
.
20
Kriegel
M. A.
,
Rathinam
C.
,
Flavell
R. A.
.
2012
.
Pancreatic islet expression of chemokine CCL2 suppresses autoimmune diabetes via tolerogenic CD11c+ CD11b+ dendritic cells.
Proc. Natl. Acad. Sci. USA
109
:
3457
3462
.
21
Li
Y.
,
Chu
N.
,
Rostami
A.
,
Zhang
G. X.
.
2006
.
Dendritic cells transduced with SOCS-3 exhibit a tolerogenic/DC2 phenotype that directs type 2 Th cell differentiation in vitro and in vivo.
J. Immunol.
177
:
1679
1688
.
22
Villagra
A.
,
Cheng
F.
,
Wang
H. W.
,
Suarez
I.
,
Glozak
M.
,
Maurin
M.
,
Nguyen
D.
,
Wright
K. L.
,
Atadja
P. W.
,
Bhalla
K.
, et al
.
2009
.
The histone deacetylase HDAC11 regulates the expression of interleukin 10 and immune tolerance.
Nat. Immunol.
10
:
92
100
.
23
Meredith
M. M.
,
Liu
K.
,
Darrasse-Jeze
G.
,
Kamphorst
A. O.
,
Schreiber
H. A.
,
Guermonprez
P.
,
Idoyaga
J.
,
Cheong
C.
,
Yao
K. H.
,
Niec
R. E.
,
Nussenzweig
M. C.
.
2012
.
Expression of the zinc finger transcription factor zDC (Zbtb46, Btbd4) defines the classical dendritic cell lineage.
J. Exp. Med.
209
:
1153
1165
.
24
Satpathy
A. T.
,
Kc
W.
,
Albring
J. C.
,
Edelson
B. T.
,
Kretzer
N. M.
,
Bhattacharya
D.
,
Murphy
T. L.
,
Murphy
K. M.
.
2012
.
Zbtb46 expression distinguishes classical dendritic cells and their committed progenitors from other immune lineages.
J. Exp. Med.
209
:
1135
1152
.
25
Nefedova
Y.
,
Nagaraj
S.
,
Rosenbauer
A.
,
Muro-Cacho
C.
,
Sebti
S. M.
,
Gabrilovich
D. I.
.
2005
.
Regulation of dendritic cell differentiation and antitumor immune response in cancer by pharmacologic-selective inhibition of the janus-activated kinase 2/signal transducers and activators of transcription 3 pathway.
Cancer Res.
65
:
9525
9535
.
26
Melillo
J. A.
,
Song
L.
,
Bhagat
G.
,
Blazquez
A. B.
,
Plumlee
C. R.
,
Lee
C.
,
Berin
C.
,
Reizis
B.
,
Schindler
C.
.
2010
.
Dendritic cell (DC)-specific targeting reveals Stat3 as a negative regulator of DC function.
J. Immunol.
184
:
2638
2645
.
27
Rescigno
M.
,
Martino
M.
,
Sutherland
C. L.
,
Gold
M. R.
,
Ricciardi-Castagnoli
P.
.
1998
.
Dendritic cell survival and maturation are regulated by different signaling pathways.
J. Exp. Med.
188
:
2175
2180
.
28
Akira
S.
,
Uematsu
S.
,
Takeuchi
O.
.
2006
.
Pathogen recognition and innate immunity.
Cell
124
:
783
801
.
29
Guilliams
M.
,
Crozat
K.
,
Henri
S.
,
Tamoutounour
S.
,
Grenot
P.
,
Devilard
E.
,
de Bovis
B.
,
Alexopoulou
L.
,
Dalod
M.
,
Malissen
B.
.
2010
.
Skin-draining lymph nodes contain dermis-derived CD103(-) dendritic cells that constitutively produce retinoic acid and induce Foxp3(+) regulatory T cells.
Blood
115
:
1958
1968
.
30
Hall
J. A.
,
Grainger
J. R.
,
Spencer
S. P.
,
Belkaid
Y.
.
2011
.
The role of retinoic acid in tolerance and immunity.
Immunity
35
:
13
22
.
31
Jensen
P. E.
2007
.
Recent advances in antigen processing and presentation.
Nat. Immunol.
8
:
1041
1048
.
32
Ratzinger
G.
,
Stoitzner
P.
,
Ebner
S.
,
Lutz
M. B.
,
Layton
G. T.
,
Rainer
C.
,
Senior
R. M.
,
Shipley
J. M.
,
Fritsch
P.
,
Schuler
G.
,
Romani
N.
.
2002
.
Matrix metalloproteinases 9 and 2 are necessary for the migration of Langerhans cells and dermal dendritic cells from human and murine skin.
J. Immunol.
168
:
4361
4371
.
33
Farmer
W. H.
,
Yuan
Z. Y.
.
1991
.
A continuous fluorescent assay for measuring protease activity using natural protein substrate.
Anal. Biochem.
197
:
347
352
.
34
Folk, J. E. 1970. [32] Carboxypeptidase B (porcine pancreas). In Methods in Enzymology. G. E. Perlmann and L. Lorand, eds. Academic Press, San Diego, CA, p. 504–508
.
35
Cloosen
S.
,
Arnold
J.
,
Thio
M.
,
Bos
G. M.
,
Kyewski
B.
,
Germeraad
W. T.
.
2007
.
Expression of tumor-associated differentiation antigens, MUC1 glycoforms and CEA, in human thymic epithelial cells: implications for self-tolerance and tumor therapy.
Cancer Res.
67
:
3919
3926
.
36
Manicassamy
S.
,
Reizis
B.
,
Ravindran
R.
,
Nakaya
H.
,
Salazar-Gonzalez
R. M.
,
Wang
Y.-C.
,
Pulendran
B.
.
2010
.
Activation of beta-catenin in dendritic cells regulates immunity versus tolerance in the intestine.
Science
329
:
849
853
.
37
Hanada
T.
,
Tanaka
K.
,
Matsumura
Y.
,
Yamauchi
M.
,
Nishinakamura
H.
,
Aburatani
H.
,
Mashima
R.
,
Kubo
M.
,
Kobayashi
T.
,
Yoshimura
A.
.
2005
.
Induction of hyper Th1 cell-type immune responses by dendritic cells lacking the suppressor of cytokine signaling-1 gene.
J. Immunol.
174
:
4325
4332
.
38
Medzhitov
R.
,
Preston-Hurlburt
P.
,
Janeway
C. A.
 Jr.
1997
.
A human homologue of the Drosophila Toll protein signals activation of adaptive immunity.
Nature
388
:
394
397
.
39
Takeda
K.
,
Clausen
B. E.
,
Kaisho
T.
,
Tsujimura
T.
,
Terada
N.
,
Förster
I.
,
Akira
S.
.
1999
.
Enhanced Th1 activity and development of chronic enterocolitis in mice devoid of Stat3 in macrophages and neutrophils.
Immunity
10
:
39
49
.
40
Neefjes
J.
,
Jongsma
M. L.
,
Paul
P.
,
Bakke
O.
.
2011
.
Towards a systems understanding of MHC class I and MHC class II antigen presentation.
Nat. Rev. Immunol.
11
:
823
836
.
41
Zavasnik-Bergant
T.
,
Turk
B.
.
2006
.
Cysteine cathepsins in the immune response.
Tissue Antigens
67
:
349
355
.
42
Shortman
K.
,
Naik
S. H.
.
2007
.
Steady-state and inflammatory dendritic-cell development.
Nat. Rev. Immunol.
7
:
19
30
.
43
Burgos
F. J.
,
Salvà
M.
,
Villegas
V.
,
Soriano
F.
,
Mendez
E.
,
Avilés
F. X.
.
1991
.
Analysis of the activation process of porcine procarboxypeptidase B and determination of the sequence of its activation segment.
Biochemistry
30
:
4082
4089
.

The authors have no financial conflicts of interest.

Supplementary data