Cells for Cross-Presentation to CTL Dendritic Cells Acquire Antigens ...

6 downloads 1013 Views 1MB Size Report
Receive free email-alerts when new articles cite this article. Sign up at: ... Immature DC were generated by culturing normal monkey CD14 mono- cytes in ...
Dendritic Cells Acquire Antigens from Live Cells for Cross-Presentation to CTL1 Larry A. Harshyne,* Simon C. Watkins,† Andrea Gambotto,‡ and Simon M. Barratt-Boyes2* Dendritic cells (DC) can readily capture Ag from dead and dying cells for presentation to MHC class I-restricted CTL. We now show by using a primate model that DC also acquire Ag from healthy cells, including other DC. Coculture assays showed that fluorescently labeled plasma membrane was rapidly and efficiently transferred between DC, and transfer of intracellular proteins was observed to a lesser extent. Acquisition of labeled plasma membrane and intracellular protein was cell contact-dependent and was primarily a function of immature DC, whereas both immature and CD40L-matured DC could serve as donors. Moreover, immature DC could acquire labeled plasma membrane and intracellular proteins from a wide range of hemopoietic cells, including macrophages, B cells, and activated T cells. Notably, macrophages, which readily phagocytose apoptotic bodies, were very inefficient at acquiring labeled plasma membrane and intracellular proteins from other live macrophages or DC. With live-cell imaging techniques, we demonstrate that individual DC physically extract plasma membrane from other DC, generating endocytic vesicles of up to 1 ␮m in diameter. Finally, DC but not macrophages acquired an endogenous melanoma Ag expressed by live DC and cross-presented Ag to MHC class I-restricted CTL, demonstrating the immunological relevance of our finding. These data show for the first time that DC readily acquire Ag from other live cells. We suggest that Ag acquisition from live cells may provide a novel mechanism whereby DC can present Ag in the absence of direct infection, and may serve to expand and regulate the immune response in vivo. The Journal of Immunology, 2001, 166: 3717–3723.

D

endritic cells (DC)3 are sentinels of the immune system that function by surveying peripheral tissues for Ag (1). To facilitate this function, DC acquire Ag from a variety of sources. DC can uptake Ag released from cells undergoing apoptosis or necrosis for presentation to MHC class I-restricted CTL (2– 8). Similarly, DC acquire soluble proteins by macropinocytosis for presentation in the MHC class I pathway (9, 10). It has recently been demonstrated that DC acquire Ag from other DC for presentation in MHC class II molecules, a process also thought to be mediated by apoptosis of donor cells (11). These Ag acquisition processes have in common an initiating event involving cell death, which is thought to provide danger signals for DC activation and the subsequent stimulation of CTL (6). Several reports have indicated that physiologic interactions between hemopoietic cells in the absence of cell death can result in transfer of membrane components between cells. Passive transfer of MHC molecules between allogeneic murine T cell clones has been reported (12), and the internalization by T cells of MHC molecules derived from the surface of APC has been demonstrated (13, 14). DC are highly interactive cells with extensive membrane

*Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261; and Departments of †Cell Biology and Physiology, and ‡Surgery and Molecular Genetics and Biochemistry, School of Medicine, University of Pittsburgh, Pittsburgh, PA 15261 Received for publication August 31, 2000. Accepted for publication January 8, 2001. The costs of publication of this article were defrayed in part by the payment of page charges. This article must therefore be hereby marked advertisement in accordance with 18 U.S.C. Section 1734 solely to indicate this fact. 1 This work was supported by National Institutes of Health Grants RR00119 (to S.M.B.-B.) and AI43664 (to S.M.B.-B., S.C.W., and A.G.). 2 Address correspondence and reprint requests to Dr. Simon M. Barratt-Boyes, Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pittsburgh, PA 15261. E-mail address: [email protected] 3

Abbreviations used in this paper: DC, dendritic cells; DiD, 1,1⬘-dioctadecyl3,3,3⬘,3⬘-tetramethylindodi-carbocyanine perchlorate; CMFDA, 5-chloromethylfluorescein diacetate; Ad-gp100, recombinant adenovirus expressing gp100; ELISPOT, enzyme-linked immunospot assay. Copyright © 2001 by The American Association of Immunologists

processes that facilitate cell clustering and interaction with other cells, including T cells and B cells. We reasoned that during such interactions DC might acquire Ag in the absence of apoptosis or necrosis of the donor cell. Ag transfer between live DC has been suggested after the observation that both Ag-naive and Ag-exposed DC are important in T cell proliferation, although transfer was not directly demonstrated (15). Here, we used flow cytometry and live cell imaging techniques to visualize interactions of DC with other cells in a well-characterized monkey model (16). We demonstrate that DC but not macrophages have a remarkable capacity to acquire plasma membrane and intracellular proteins from other live APC and lymphocytes, and that such transfer results in cross-presentation of Ag to MHC class I-restricted CTL.

Materials and Methods Cells Immature DC were generated by culturing normal monkey CD14⫹ monocytes in GM-CSF and IL-4 for 4 days, with an additional 48-h exposure to CD40L (generously provided by Elaine Thomas, Immunex, Seattle, WA) to induce maturation, as described previously (16). Human DC from HLAtyped donors were generated in the same manner. Macrophages were generated by culturing monkey CD14⫹ monocytes for 24 h in the absence of cytokines. T cells used in the transfer experiments were monocyte-depleted from monkey PBMC and cultured in the presence of 5 ␮g/ml Con A and 20 U/ml IL-2 for 3 days to generate activated T cells, or for an additional 5 days to generate resting T cells. The EBV-transformed human HLA-A2⫹ B cell line Croft was generously provided by Olivera Finn (University of Pittsburgh, Pittsburgh, PA). The oligoclonal T cell line TIL620, which recognizes two epitopes of the melanoma Ag gp100 (17), was derived in the laboratory of Dr. Steven Rosenberg (National Institutes of Health, Bethesda, MD).

Fluorescence labeling Cells were incubated with the lipophilic probe 1,1⬘-dioctadecyl-3,3,3⬘,3⬘tetramethylindodi-carbocyanine perchlorate (DiD; excitation/emission spectra ⫽ 644 nm/663 nm) as described (18) and the thiol-reactive chloromethyl probe 5-chloromethylfluorescein diacetate (CMFDA; 492 nm/ 516 nm; both from Molecular Probes, Eugene, OR) at a concentration of 0022-1767/01/$02.00

3718 0.5␮g/ml in 5 mM EDTA for 30 min at 37°C. Any dead cells were removed by centrifugation through sodium diatrizoate and Ficoll (Sigma, St. Louis, MO).

Transfer assays and flow cytometric analysis Unlabeled DC, macrophages, B cells, or T cells with or without pretreatment with 10 ␮g/ml cytochalasin D were cocultured with labeled DC at various ratios at 4 or 37°C in 24- or 48-well plates at a concentration of 1 ⫻ 105 cells/ml. For transwell studies, labeled DC were added to 0.4-␮m pore transwell chambers (Millipore, Bedford, MA) inserted into wells containing unlabeled DC. Cells were harvested at various intervals and maintained on ice until analyzed on a FACSCaliber flow cytometer (Becton Dickinson, Mountain View, CA) by using CellQuest software (Becton Dickinson). All analyses were done by using log10 fluorescence. In some experiments, cells were fixed in 2% paraformaldehyde before analysis by flow cytometry. We determined in preliminary studies that fixation did not lead to leakage of dye from DiD- and CMFDA-labeled cells (data not shown).

Detection of apoptosis DC were labeled with DiD and incubated with unlabeled DC for various time periods. Apoptosis was measured by using the TUNEL assay (Boehringer Mannheim, Indianapolis, IN) and staining with Alexa Fluor 488 annexin V (Molecular Probes) as per manufacturer’s instructions. As a positive control for apoptosis, T cells were treated with a UVB light source calibrated at 0.6 mJ/cm2/sec for 8 min and analyzed 12–16 h later.

Live cell microscopy DC labeled with DiD were introduced into a Bioptechs FCS2 (Butler, PA) closed microscope chamber maintained at 37°C and allowed to adhere. CMFDA-labeled DC were then added in medium containing Hoescht 33342 to label nuclei. Cells were imaged with a Zeiss Axiovert 135 microscope (Oberkochen, Germany) equipped with an XYZ stage, dual excitation filter wheels (Ludl, Hawthorne, NY), shuttered xenon and halogen light sources, and an Orca 12-bit cooled charge-coupled device camera (Hamamatsu, Tokyo, Japan). Images from five random fields were collected every 4 min for each of the illumination conditions with Metamorph software (Universal Imaging, West Chester, PA). The time-lapse sequences were integrated into movies and observed. Cell-cell interactions were tagged and tracked by particle tracking methods within the Metamorph software.

DENDRITIC CELLS ACQUIRE Ag FROM LIVE CELLS pretreated with cytochalasin D, an inhibitor of membrane ruffling, transfer of fluorescence was substantially inhibited. Similarly, unlabeled and labeled DC populations remained separate when cultured for 4 h at 4°C (Fig. 1a). In addition, coculture of labeled DC with resting T cells for 4 h at 37°C did not result in fluorescence transfer between cell populations (Fig. 1a). These experiments indicate that fluorescent labels remain cell associated and do not passively transfer between cells. In contrast to the control experiments, when labeled and unlabeled DC were cultured together at 37°C in the absence of phagocytic inhibitors transfer of DiD-labeled plasma membrane occurred rapidly, as by 1 h of coculture, 52% of unlabeled cells were DID⫹, and by 4 h, up to 95% were DiD⫹ (Fig. 1b). Transfer of CMFDA occurred in cells that were DiDbright, indicating that movement of plasma membrane was accompanied by movement of cytoplasmic protein to unlabeled DC. Labeled DC were viable at all time points of the experiment as measured by lack of staining using both the TUNEL assay and annexin V binding (Fig. 1b), indicating that transfer of membrane and cytoplasmic proteins between cells was not the result of uptake of apoptotic material. To investigate the mechanisms of transfer between DC we used a transwell system. Separation of labeled and unlabeled DC by a 0.4-␮m transwell membrane completely abrogated transfer of label when cocultured for 4 h at 37°C (Fig. 2a). This indicates that direct cell-cell contact is required and reinforces the earlier finding that

Generation of recombinant adenovirus and transduction of DC Recombinant adenovirus encoding gp100 (Ad-gp100) was generated by cloning a SalI-NotI fragment containing full-length human gp100 (generously provided by Dr. Stephan Wagner, University of Essen, Essen, Germany) into the pAdlox shuttle vector, with subsequent cotransfection with ⌿5 helper virus into the CRE8 packaging cell line as described previously (19). DC or macrophages were transduced with Ad-gp100 at a multiplicity of infection of 100 by adding virus directly to cells in culture as described (16). Cells were washed extensively at 24 h after infection to remove free virus and cultured for an additional 24 –36 h to allow for protein expression. We routinely achieve 90% transduction efficiency using this method (16).

IFN-␥ enzyme-linked immunospot (ELISPOT) assay TIL620 cells (3 ⫻ 104) were cultured with the same number of Ad-gp100transduced immature monkey DC or macrophages with and without immature human DC or macrophages on Multiscreen-HA plates (Millipore) coated with anti-human IFN-␥ Ab (1-D1K; MABTECH, Stockholm, Sweden). After 18 h of incubation, IFN-␥ production was detected by labeling with a second anti-human IFN-␥ Ab (7-B6-1; MABTECH) as described previously (19). Spots were enumerated by using a dissecting microscope.

Results Transfer of plasma membrane and intracellular proteins between live DC Our initial studies focused on interactions between immature DC, as these cells are actively phagocytic and function in Ag capture (16, 20). DC were dual-labeled with DiD and CMFDA to track movement of plasma membrane and intracellular proteins, respectively, and cocultured with unlabeled cells for various time periods before flow cytometric analysis. To establish the validity of the fluorescence transfer system, we did a number of control experiments. When labeled DC were cultured for 4 h with unlabeled DC

FIGURE 1. Specific transfer of fluorescently labeled plasma membrane and intracellular proteins between monkey DC in the absence of apoptosis. Immature DC were labeled with DiD and CMFDA to identify plasma membrane and cytoplasmic proteins, respectively, and cocultured with unlabeled DC or T cells. Fluorescence transfer was analyzed by flow cytometry. a, Equal numbers of labeled and unlabeled DC were cocultured and analyzed immediately after mixing (0 h), after 4 h at 37°C after pretreatment of unlabeled cells with cytochalasin D (CCD), and after 4 h incubation at 4°C. Alternatively, labeled DC were cultured with T cells for 4 h at 37°C. b, Labeled and unlabeled immature DC were cultured for indicated times at 37°C and then harvested. Numbers indicate proportion of unlabeled cells that acquired fluorescence by using a gate based on profiles at 0 h (top row). Labeled DC were analyzed by TUNEL assay and annexin V binding at the indicated times to rule out the presence of apoptotic cells (bottom row).

The Journal of Immunology

3719

FIGURE 2. Transfer of labeled plasma membrane and intracellular proteins between live DC is dependent on cell contact and is highly efficient. Immature DC were labeled with DiD and CMFDA as in Fig. 1. a, Labeled and unlabeled immature DC were analyzed immediately after mixing (0 h) or following coculture for 4 h at 37°C in the absence (⫺twell) or presence (⫹twell) of a 0.4-␮m transwell. b, Labeled (L) and unlabeled (U) immature DC were cultured together for 4 h at 37°C at the indicated ratios. Numbers indicate the proportion of unlabeled cells that acquired fluorescence by using a gate based on profiles at 0 h for the respective experiment.

passive movement of dye does not contribute to transfer of fluorescence between cells. In addition, by using different ratios of labeled and unlabeled DC, we demonstrate that transfer of plasma membrane and intracellular proteins between DC is an extremely efficient process, as 43% of unlabeled DC acquired fluorescence within 4 h when mixed with labeled DC at a 4:1 ratio (Fig. 2b). Together, these data indicate that DC can readily transfer plasma membrane and, to a lesser extent, cytoplasmic proteins to other live DC. Immature DC but not macrophages efficiently acquire labeled membrane and cytoplasmic proteins from DC, macrophages, B cells, and activated T cells To determine whether the capacity to acquire labeled plasma membrane and intracellular proteins from live cells was unique to immature DC, we performed similar studies with different DC populations as well as macrophages, which are highly effective at acquiring cellular Ag via uptake of apoptotic material (4). We first compared immature DC with DC that had been treated with CD40L to induce maturation, which is known to down-regulate phagocytic function (16, 21). Immature DC were able to acquire plasma membrane and intracellular labels with similar efficiency irrespective of the maturation state of the labeled population. When DC were matured with CD40L, the capacity to acquire labeled membrane and intracellular proteins from either immature or mature DC was suppressed by ⬃50%, indicating that acquisition was primarily a function of immature DC (Fig. 3a). The process of transfer did not result in maturation of DC, as coculture of unlabeled immature DC with DiD- and CMFDA-labeled DC for 6 h did not result up-regulation of CD86 or expression of the maturation marker CD83 (data not shown). Expression of CD83 is induced on immature DC after exposure to CD40L for the same time period (data not shown). In contrast to DC, macrophages were extremely limited in the capacity to acquire labeled plasma membrane and intracellular proteins from either live macrophages or immature DC (Fig. 3b). Conversely, immature DC readily acquired Ag from labeled macrophages (Fig. 3b). To determine whether cells other than APC could serve as donors, we cocultured immature DC with DiD- and CMFDA-labeled

FIGURE 3. Immature DC but not macrophages efficiently acquire labeled plasma membrane and intracellular proteins from DC, macrophages, B cells, and activated T cells. a, DiD- and CMFDA-labeled immature (iDCL) or mature monkey DC (mDCL) were cocultured with equal numbers of unlabeled immature (iDCU) or mature DC (mDCU) for 4 h at 37°C before analysis by flow cytometry. b, DiD and CMFDA-labeled (M␾L) or unlabeled macrophages (M␾U) were cocultured together or with labeled and unlabeled immature DC for 4 h at 37°C. c, DiD and CMFDA-labeled (BL) or unlabeled EBV-transformed human B cells (BU) and DiD and CMFDA-labeled (TL) or unlabeled IL-2 and ConA-activated monkey T cells (TU) were cultured with labeled or unlabeled immature DC for 4 h at 37°C. Numbers indicate proportion of unlabeled cells that acquire fluorescence by using a gate based on profiles at 0 h. Differences in the gates used in c reflect different levels of autofluorescence of unlabeled DC, B cells, and T cells.

activated T cells and B cells. Although neither of these cell populations was able to acquire label from DC, immature DC readily acquired labeled plasma membrane and cytoplasmic proteins from both activated T cells and B cells (Fig. 3c). Similar findings were generated when immature DC were cultured with labeled resting monkey T cells, the Jurkat T cell line, and the 174XCEM.T1 T-B cell hybrid (data not shown). These experiments indicate that DC are unique in the capacity to acquire cellular components from other APC and from lymphocytes. Direct visualization of plasma membrane transfer between live DC To study the dynamics of transfer between individual DC, we used live cell fluorescence microscopy, which allows real-time imaging of cellular interactions in culture. DC labeled with DiD were seeded into the microscope chamber and an equal number of CMFDA-labeled DC then were added (Fig. 4, 0 min). Detection of double-labeled cells over time signaled transfer of cellular components between cells. DC were highly motile and frequently moved in and out of small clusters, while rapidly extending and retracting plasma membrane processes (see supplemental video at

3720

DENDRITIC CELLS ACQUIRE Ag FROM LIVE CELLS

FIGURE 4. Direct visualization of transfer of labeled plasma membrane between live monkey DC with real-time cell imaging. Immature DC were labeled with either DiD (falsely colored green) or CMFDA (falsely colored red) and cocultured in a live cell microscopy chamber. Images were collected every 4 min from which a montage of frames 24 min apart was generated. Fluorescence (left) and differential interference contrast (right) images for each time point are shown. A DiD-labeled DC (arrowhead) and a CMFDA-labeled DC (arrow) are identified at the onset of the experiment (0 min) which then come together (24 min). DiD-labeled plasma membrane is physically transferred to the CMFDA-labeled DC (48 –168 min). The cells separate, leaving the recipient cell containing multiple DiD-labeled endocytic vesicles (192–216 min). The full complement of images from which the montage was made can be viewed on real time at www.sbic.pitt.edu/harshyne/figure 4.

www.sbic.pitt.edu/harshyne/figure 4). During some of these interactions, large portions of DiD-labeled plasma membrane were physically pulled from one DC to another (Fig. 4, 48 –168 min). Transferred membrane appeared to be contained in endocytic ves-

icles of up to 1 ␮m in diameter, and frequently multiple vesicles were present within single DC (Fig. 4, 192–216 min). DC that had acquired membrane could be seen in isolation as double-labeled cells less than 4 h after the initiation of the experiment (Fig. 4, 216

The Journal of Immunology

3721

FIGURE 5. DC remain viable during release and uptake of labeled plasma membrane. Labeled DC were coincubated and examined by live cell microscopy as described in Fig. 4. Top, Images collected at 0, 96, and 216 min after mixing were analyzed by overlapping DiD (green) and CMFDA (red) with Hoescht 33342 (blue). Bottom, These same images are shown with Hoescht 33342 staining in black and white to emphasize nuclei. Arrowheads and arrows identify donor and recipient cells, respectively, as described in Fig. 4.

min). Importantly, DC that had donated membrane also moved away and interacted with other DC, indicating continued viability of these cells (Fig. 4, 192–216 min). To definitively rule out the contribution of apoptotic cell death in transfer of fluorescent label between DC, we used Hoescht 33342 to label nuclei during live cell imaging. Nuclei of cells remained intact and noncondensed during all stages of cell interaction and transfer, as highlighted by the images collected at 0, 96, and 216 min (Fig. 5). These data provide direct evidence that DC involved in plasma membrane transfer were viable. Endogenous tumor Ag expressed in live DC is acquired by DC but not macrophages for cross-presentation to MHC class Irestricted CTL To ascertain the immunological significance of the observed transfer between DC, we developed a xenogeneic T cell stimulation assay with TIL620, an HLA-A2-restricted CTL line specific for melanoma Ag gp100 (17). Ad-gp100 was used to express gp100 protein as an endogenous cytoplasmic Ag in DC. Immature monkey DC, confirmed to be HLA-A2⫺ by Ab staining (data not shown), were transduced with Ad-gp100 and cocultured in equal numbers with immature HLA-A2⫹ human DC. TIL620 cells were added to the culture and IFN-␥ production was assayed 18 h later by using ELISPOT. As expected, culturing Ad-gp100 transduced HLA-A2⫹ DC with TIL620 resulted in vigorous IFN-␥ release (Fig. 6a). However, coculture of Ad-gp100 transduced monkey DC with HLA-A2⫹ human DC was also effective at inducing cytokine release from TIL620 (Fig. 6a). Cytokine production was not attributable to direct stimulation of TIL620 by transduced monkey DC, as coculture with HLA-A2⫺ human DC did not elicit significant IFN-␥ production. Similarly, coculture of monkey DC and HLA-A2⫹ human DC in the absence of gp100 resulted in negligible stimulation of TIL620 (Fig. 6a). When HLA-A2⫹ macro-

phages were used as acceptor cells with Ad-gp100-transduced monkey DC minimal IFN-␥ release by TIL620 was observed (Fig. 6b), consistent with the flow cytometry data indicating that macrophages cannot acquire plasma membrane or intracellular protein from live cells. Directly transduced HLA-A2⫹ macrophages efficiently stimulated TIL620 (Fig. 6b). To determine whether Ad-gp100-transduced cells were dying in the ELISPOT assays, which could provide Ag to DC in the form of apoptotic bodies, we performed TUNEL staining at the conclusion of the 18-h coculture. Monkey DC were TUNELneg at 18 h, ruling out this possibility (Fig. 6c). Apoptosis of human DC directly transfected with Ad-gp100 did occur after 18 h of coincubation with TIL620 (Fig. 6c). In summary, the ELISPOT data are consistent with direct transfer of intracellular Ag between live DC resulting in processing and presentation in MHC class I.

Discussion The results of this study demonstrate for the first time that simple interactions between DC and other healthy cells lead to substantial acquisition of plasma membrane and cytoplasmic proteins by DC. Moreover, the data show that such transfer is immunologically relevant, resulting in cross-presentation of endogenous cytoplasmic Ag to MHC class I-restricted CTL. These data challenge the current view that DC only acquire Ag from dead and dying cells. Several findings provide conclusive evidence that transfer occurred independently of apoptosis, the common mechanism cited for acquisition of cellular Ag by DC (2, 3). First, labeled DC used in the 4 h coculture experiments had intact DNA as measured by TUNEL assay. Importantly, DC did not undergo apoptosis during the assays as measured by annexin V staining, which detects the early apoptotic event of phosphatidylserine membrane translocation. Second, live cell imaging experiments clearly demonstrated that DC maintained intact nuclear structure, cell morphology, and

3722

FIGURE 6. DC but not macrophages cross-present endogenous tumor Ag derived from live DC to MHC class I-restricted CTL. a, The HLAA2-restricted, gp100-specific CTL line TIL620 was cultured with various combinations of equal numbers of Ad-gp100-transduced immature HLAA2⫹ human DC (HLA-A2⫹ DC/gp100), Ad-gp100 transduced immature monkey DC (monkey DC/gp100), uninfected monkey DC, or HLA-A2⫹ or HLA-A2⫺ human DC on plates coated with anti-IFN-␥ Ab for 18 h before determination of IFN-␥ release. b, TIL620 cells were cultured with various combinations of HLA-A2⫹ human DC and monkey DC as in a, or with Ad-gp100 transduced HLA-A2⫹ human macrophages (HLA-A2⫹M␾/ gp100) or uninfected HLA-A2⫹ macrophages (HLA-A2⫹ M␾). The frequencies of cytokine-producing TIL620 cells were determined by staining with a second IFN-␥-specific Ab and enumerated using a dissecting microscope. Results are presented as mean spot number ⫾ SD of duplicate determinations. c, TUNEL staining was done after the 18-h coincubation of cells as indicated and DC analyzed by using a gate exclusive of lymphocytes.

motility before, during, and after transfer of fluorescent label to other DC. Third, gp100-transduced monkey DC were TUNELneg for the duration of the 18-h ELISPOT assay, indicating that apoptosis of these cells did not contribute to Ag acquisition by human DC with subsequent stimulation of T cells. TIL620 were clearly capable of killing Ag-bearing DC as evidenced by apoptosis in gp100-transduced HLA-A2⫹ DC. We hypothesize that apoptosis would become apparent in HLA-A2⫹ DC cocultured with gp100transduced monkey DC had a longer time period elapsed to allow for sufficient Ag transfer and processing in recipient DC. Finally and most notably, macrophages did not acquire fluorescently labeled membrane and cytoplasm from DC and other macrophages,

DENDRITIC CELLS ACQUIRE Ag FROM LIVE CELLS nor did they cross-present Ag when cocultured with live Ag-expressing DC, despite the exquisite capacity of these cells to phagocytose apoptotic material (4). This finding highlights an apparent distinction between DC and macrophages in that both cell types internalize particulate material, but only DC are able to acquire significant levels of Ag from other live cells. The observed mechanism of Ag acquisition by DC also appears to be independent of exosomes, which are 50- to 90-nm diameter vesicles released from DC and other cells that have been shown to function as Ag-presenting moieties (22, 23). Studies using DC pulsed with FITC have suggested a role for secreted exosomes in uptake by other DC, although Ag transfer was more effective after direct cell contact (15). In our experiments, acquisition of fluorescently labeled membrane and cytoplasm was completely abrogated by a 0.4-␮m transwell filter, a pore size that would not restrict flow of exosomes. The live cell microscopy images revealed that intimate DC-DC contact resulted in physical stripping of membrane from one cell by another, leading to the formation of relatively large vesicles up to 1 ␮m in diameter. DC appeared to interact via plasma membrane extensions, suggesting that these characteristic cell processes play an important role in Ag transfer between DC. Hence, DC can secrete small exosomes into the supernatant but have the capacity to readily produce substantially larger vesicles after direct interaction with other DC. The functional studies indicated that endogenous tumor Ag expressed in one DC could be acquired by another DC for crosspresentation to class I-restricted CTL. Cross-presentation by DC of cell-associated Ag in MHC class I previously only had been demonstrated for Ag derived from dead or dying cells (2– 8). In our studies, it is likely that cytoplasmic Ag was carried within vesicles that were observed being transferred between DC by live-cell imaging. This is consistent with the flow cytometry data showing that cells that had acquired substantial membrane label also acquire labeled intracellular proteins. Whether Ag is transferred intact and requires further processing in the recipient DC before loading on MHC class I molecules is not known. An alternative mechanism for cross-presentation could be trafficking of plasma membrane containing MHC molecules in a reverse direction, supported by recent findings that T cells can acquire MHC-peptide complexes from APC after TCR engagement (13, 14). However, transferred HLA-A2 would have to be internalized by the monkey DC, loaded with processed gp100 Ag, and trafficked intact to the cell surface for subsequent recognition by TIL620. We currently are investigating the precise mechanism of cross-presentation and whether reciprocal transfer of Ag and MHC molecules occurs in this system. The marked degree of Ag transfer detected in this study suggests that Ag acquisition by DC from other viable cells may be an important process in vivo. Acquisition of Ag was significantly more efficient by immature than mature DC, indicating that Ag transfer in vivo may occur at the level of the skin and other peripheral tissues where immature DC reside. Hence, DC directly infected with a pathogen could provide a continuous source of Ag for resident Langerhan’s cells or immature DC recruited to the site through release of inflammatory cytokines (24), thereby rapidly and effectively amplifying the immune response. Transfer of Ag between DC in skin would enable subpopulations that are apparently nonmigratory to nevertheless stimulate robust CTL responses in vivo, as has been suggested for CD8␣⫹ lymphoid DC in the mouse (25). This may be the case for mature DC administered as vaccines, which we have shown in the primate model have a tendency to remain at the site of injection (16). Alternatively, Ag transfer could occur after migration of Ag-exposed DC to lymph nodes as has been suggested by others (11), mediated by lymph

The Journal of Immunology node DC subpopulations that have Ag-acquiring capacity (26). Moreover, our data indicate that cells other than DC can serve as Ag donors, including macrophages, T cells, and B cells. Hence, macrophages containing phagocytosed material could serve as a source of Ag for DC in peripheral tissues. Similarly, lymphocytes or macrophages infected with pathogens could donate Ag to DC. This latter scenario suggests a mechanism whereby DC could acquire Ag derived from pathogens that preferentially infect macrophages and lymphocytes without necessarily causing cell death, such as HIV and EBV. It is believed that maturation is required for DC to become efficient stimulators of Ag-specific T cells (6, 27). Studies indicate that uptake of apoptotic material by itself may not provide sufficient stimulus for maturation of DC and that additional stimuli provided by necrotic cells may be required for effective Ag presentation (7). In addition, uptake of apoptotic material by DC that have migrated from the gut has been reported, suggesting that DC exposed to Ag in this manner may serve to induce T cell tolerance to intestinal Ag in draining lymph nodes (28). However, in our studies, immature DC readily acquired Ag from other immature DC and were able to cross-present Ag to CTL, suggesting that maturation signals may not be essential for effective immunostimulatory function. It remains to be determined whether Ag transfer between live DC is involved in induction of tolerance to self. In summary, our studies reveal that interactions between DC and other healthy cells result in acquisition of membrane and cytoplasmic components by the DC, and subsequent cross-presentation to T cells. We suggest that this previously unrecognized means of Ag transfer might be important in the development and regulation of immune responses in vivo.

Acknowledgments We thank M. Zimmer for stimulating discussion, O. Finn and S. Rosenberg for cell lines, Immunex Corporation and Schering-Plough Research Institute for cytokines, M. Murphey-Corb and the University of Pittsburgh Primate Facility for Infectious Disease Research for monkey samples, C. Castillo for technical assistance, and J. Ahearn for access to the flow cytometer.

References 1. Banchereau, J., and R. M. Steinman. 1998. Dendritic cells and the control of immunity. Nature 392:245. 2. Rubartelli, A., A. Poggi, and M. R. Zocchi. 1997. The selective engulfment of apoptotic bodies by dendritic cells is mediated by the ␣v␤3 integrin and requires intracellular and extracellular calcium. Eur. J. Immunol. 27:1893. 3. Albert, M. L., B. Sauter, and N. Bhardwaj. 1998. Dendritic cells acquire antigen from apoptotic cells and induce class I-restricted CTLs. Nature 392:86. 4. Albert, M. L., S. F. Pearce, L. M. Francisco, B. Sauter, P. Roy, R. L. Silverstein, and N. Bhardwaj. 1998. Immature dendritic cells phagocytose apoptotic cells via ␣v␤5 and CD36, and cross-present antigens to cytotoxic T lymphocytes. J. Exp. Med. 188:1359. 5. Chiodoni, C., P. Paglia, A. Stoppacciaro, M. Rodolfo, M. Parenza, and M. P. Colombo. 1999. Dendritic cells infiltrating tumors cotransduced with granulocyte/macrophage colony-stimulating factor (GM-CSF) and CD40 ligand genes take up and present endogenous tumor-associated antigens, and prime naive mice for a cytotoxic T lymphocyte response. J. Exp. Med. 190:125. 6. Gallucci, S., M. Lolkema, and P. Matzinger. 1999. Natural adjuvants: endogenous activators of dendritic cells. Nat. Med. 5:1249. 7. Sauter, B., M. L. Albert, L. Francisco, M. Larsson, S. Somersan, and N. Bhardwaj. 2000. Consequences of cell death: exposure to necrotic tumor cells,

3723

8.

9.

10.

11.

12.

13.

14.

15.

16.

17.

18. 19.

20.

21.

22.

23.

24.

25.

26.

27.

28.

but not primary tissue cells or apoptotic cells, induces the maturation of immunostimulatory dendritic cells. J. Exp. Med. 191:423. Bonnotte, B., N. Favre, M. Moutet, A. Fromentin, E. Solary, M. Martin, and F. Martin. 2000. Role of tumor cell apoptosis in tumor antigen migration to the draining lymph nodes. J. Immunol. 164:1995. Norbury, C. C., B. J. Chambers, A. R. Prescott, H. G. Ljunggren, and C. Watts. 1997. Constitutive macropinocytosis allows TAP-dependent major histocompatibility complex class I presentation of exogenous soluble antigen by bone marrow-derived dendritic cells. Eur. J. Immunol. 27:280. Shen, Z., G. Reznikoff, G. Dranoff, and K. L. Rock. 1997. Cloned dendritic cells can present exogenous antigens on both MHC class I and class II molecules. J. Immunol. 158:2723. Inaba, K., S. Turley, F. Yamaide, T. Iyoda, K. Mahnke, M. Inaba, M. Pack, M. Subklewe, B. Sauter, D. Sheff, et al. 1998. Efficient presentation of phagocytosed cellular fragments on the major histocompatibility complex class II products of dendritic cells. J. Exp. Med. 188:2163. Lorber, M. I., M. R. Loken, A. M. Stall, and F. W. Fitch. 1982. I-A antigens on cloned alloreactive murine T lymphocytes are acquired passively. J. Immunol. 128:2798. Huang, J. F., Y. Yang, H. Sepulveda, W. Shi, I. Hwang, P. A. Peterson, M. R. Jackson, J. Sprent, and Z. Cai. 1999. TCR-mediated internalization of peptide-MHC complexes acquired by T cells. Science 286:952. Patel, D. M., P. Y. Arnold, G. A. White, J. P. Nardella, and M. D. Mannie. 1999. Class II MHC/peptide complexes are released from APC and are acquired by T cell responders during specific antigen recognition. J. Immunol. 163:5201. Knight, S. C., S. Iqball, M. S. Roberts, S. Macatonia, and P. A. Bedford. 1998. Transfer of antigen between dendritic cells in the stimulation of primary T cell proliferation. Eur. J. Immunol. 28:1636. Barratt-Boyes, S. M., M. I. Zimmer, L. A. Harshyne, E. M. Meyer, S. C. Watkins, S. Capuano, III, M. Murphey-Corb, L. D. Falo, Jr., and A. D. Donnenberg. 2000. Maturation and trafficking of monocyte-derived dendritic cells in monkeys: implications for dendritic cell-based vaccines. J. Immunol. 164:2487. Kawakami, Y., S. Eliyahu, C. Jennings, K. Sakaguchi, X. Kang, S. Southwood, P. F. Robbins, A. Sette, E. Appella, and S. A. Rosenberg. 1995. Recognition of multiple epitopes in the human melanoma antigen gp100 by tumor-infiltrating T lymphocytes associated with in vivo tumor regression. J. Immunol. 154:3961. Barratt-Boyes, S. M., S. C. Watkins, and O. J. Finn. 1997. In vivo migration of dendritic cells differentiated in vitro: a chimpanzee model. J. Immunol. 158:4543. Ranieri, E., W. Herr, A. Gambotto, W. Olson, D. Rowe, P. D. Robbins, L. S. Kierstead, S. C. Watkins, L. Gesualdo, and W. J. Storkus. 1999. Dendritic cells transduced with an adenovirus vector encoding Epstein-Barr virus latent membrane protein 2B: a new modality for vaccination. J. Virol. 73:10416. Inaba, K., M. Inaba, M. Naito, and R. M. Steinman. 1993. Dendritic cell progenitors phagocytose particulates, including bacillus Calmette-Guerin organisms, and sensitize mice to mycobacterial antigens in vivo. J. Exp. Med. 178:479. Sallusto, F., M. Cella, C. Danieli, and A. Lanzavecchia. 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. Zitvogel, L., A. Regnault, A. Lozier, J. Wolfers, C. Flament, D. Tenza, P. Ricciardi-Castagnoli, G. Raposo, and S. Amigorena. 1998. Eradication of established murine tumors using a novel cell-free vaccine: dendritic cell-derived exosomes. Nat. Med. 4:594. Thery, C., A. Regnault, J. Garin, J. Wolfers, L. Zitvogel, P. Ricciardi-Castagnoli, G. Raposo, and S. Amigorena. 1999. Molecular characterization of dendritic cell-derived exosomes: selective accumulation of the heat shock protein hsc73. J. Cell Biol. 147:599. Dieu, M. C., B. Vanbervliet, A. Vicari, J. M. Bridon, E. Oldham, S. Ait-Yahia, F. Briere, A. Zlotnik, S. Lebecque, and C. Caux. 1998. Selective recruitment of immature and mature dendritic cells by distinct chemokines expressed in different anatomic sites. J. Exp. Med. 188:373. Smith, A. L., and B. F. de St Groth. 1999. Antigen-pulsed CD8␣⫹ dendritic cells generate an immune response after subcutaneous injection without homing to the draining lymph node. J. Exp. Med. 189:593. Liu, L., M. Zhang, C. Jenkins, and G. G. MacPherson. 1998. Dendritic cell heterogeneity in vivo: two functionally different dendritic cell populations in rat intestinal lymph can be distinguished by CD4 expression. J. Immunol. 161:1146. Steinman, R. M., S. Turley, I. Mellman, and K. Inaba. 2000. The induction of tolerance by dendritic cells that have captured apoptotic cells. J. Exp. Med. 191: 411. Huang, F. P., N. Platt, M. Wykes, J. R. Major, T. J. Powell, C. D. Jenkins, and G. G. MacPherson. 2000. A discrete subpopulation of dendritic cells transports apoptotic intestinal epithelial cells to T cell areas of mesenteric lymph nodes. J. Exp. Med. 191:435.