Cross-presentation enables dendritic cells to present on their MHC class I molecules antigenic peptides derived from exogenous material, through a mechanism that remains partly unclear. It is particularly efficient with long peptides, which are used in cancer vaccines. We studied the mechanism of long-peptide cross-presentation using human dendritic cells and specific CTL clones against melanoma Ags gp100 and Melan-A/MART1. We found that cross-presentation of those long peptides does not depend on the proteasome or the transporter associated with Ag processing, and therefore follows a vacuolar pathway. We also observed that it makes use of newly synthesized MHC class I molecules, through peptide exchange in vesicles distinct from the endoplasmic reticulum and classical secretory pathway, in an SEC22b- and CD74-independent manner. Our results indicate a nonclassical secretion pathway followed by nascent HLA-I molecules that are used for cross-presentation of those long melanoma peptides in the vacuolar pathway. Our results may have implications for the development of vaccines based on long peptides.

Cross-presentation is the process whereby antigenic peptides derived from exogenous Ags are presented by dendritic cells (DCs) on their MHC class I (MHC-I) molecules. This process is needed for the induction of CD8 T cell responses to Ags derived from tumors or from pathogens that do not infect DCs (13). Cross-presentation is key to the development of vaccines aimed at inducing CD8 responses against cancer or HIV. A promising vaccine approach pioneered by Melief et al. (4, 5) is based on synthetic long peptides, which need to be cross-presented and therefore results in efficient presentation of the antigenic peptide by DCs only, as opposed to short peptides, which can load any cell and therefore induce tolerance. Therapeutic vaccines based on long peptides have shown clear promise in preclinical models (6), and recently showed clinical efficacy in patients with human papillomavirus–induced neoplasia (7). Contrasting with the advanced clinical development of long-peptide vaccination, relatively little is known about the pathway used for cross-presentation of long peptides (811).

The pathway used for cross-presentation appears to vary according to the nature of the exogenous Ag. In the mouse, two main cross-presentation pathways have been proposed: cytosolic and vacuolar. In the cytosolic pathway, the Ag internalized by endocytosis is transferred to the cytosol, from which it follows the classical MHC-I processing route. This involves processing by the proteasome and translocation of the resulting peptide by transporter associated with Ag processing (TAP) into the endoplasmic reticulum (ER), where it combines with newly synthesized MHC-I molecules. A proposed variation of the cytosolic pathway involves TAP-mediated transport of the cytosolic peptide back to the phagosome or the endolysosome, where it would combine with MHC-I molecules. The latter can be routed to phagosomes or to endolysosomes by SEC22B or CD74 (invariant chain), respectively (12, 13). Until recently, the missing piece in the cytosolic pathway was the transporter in charge of the transfer of Ags from the endosome to the cytosol. However, recent evidence has implicated SEC61 in this process (14). In the vacuolar pathway, there is no need for Ag transfer across membranes, because the whole processing happens in vacuoles, including cleavage of the internalized protein by endolysosomal peptidases and loading of the resulting peptide onto MHC-I molecules. This pathway is therefore independent from the proteasome and from TAP. Mechanistic studies of the vacuolar pathway are limited. It is unclear whether MHC-I molecules used in the vacuolar pathway are recycled from the cell surface or are newly synthesized (13, 1519).

T2 cells were given by A. Hill (Oxford University), and T1 cells were given by P. Cresswell (Yale University). We used two gp100209–217-specific CTL clones: EB81-CTL-606C/2.1 (clone 7) (20), which was used in Figs. 1 and 2A–C, and LB2686-CTL-811/327.4 (given by P. Coulie, de Duve Institute), which was used in Figs. 2D–G, 3, and 4. The RU134 – 42-specific CTL clone 381/84 was described previously (21). The Melan-A26–35-specific CTL clone CTL CP50-549/18 was given by P. Coulie. CTL clone A10, which recognizes peptide MAGE-A3168–176 presented by HLA-A1, was derived in-house (22).

FIGURE 1.

Human Mo-iDCs cross-present the long gp100 peptide in the vacuolar pathway (A, left panel) HLA-A*0201–positive Mo-iDCs can stimulate gp100209–217-specific CTL clone 7 to produce IFN-γ after a 2-h pulse with long peptide gp100184–227 at 37°C but not at 4°C. As controls (right panel), Mo-iDCs pulsed with short epitope peptide gp100209–217 stimulate CTL clone 7 at both 37°C and 4°C. (B) Fixed Mo-iDCs cannot cross-present the long gp100 peptide. Mo-iDCs were fixed with 0.004% glutaraldehyde for 45 s and then pulsed for 2 h with the long gp100 peptide (left panel) or the final epitope (right panel). CTL was added and the production of IFN-γ was measured after an overnight incubation. (C) The processing of the long gp100 peptide is not performed by extracellular peptidases. The long gp100 peptide was first incubated with HLA-A*0201 DCs at 37°C for 2 h. Next, the supernatant was collected and pulsed onto HLA-A*0201+ DCs at 37°C or 4°C for 2 h. After washing, the supernatant-pulsed DCs were assayed for their ability to stimulate IFN-γ production by gp100209–217-specific CTL clone 7. (D) Proteasome inhibition by epoxomicin (0.5 μM) does not block cross-presentation of the long gp100 peptide (left panel), whereas it blocks endogenous presentation of the same epitope by DCs transfected with gp100 mRNA (right panel). (E) Mo-iDCs (both HLA-A*0201+ and HLA-B*51+) were transfected with ICP47 or EGFP mRNA by electroporation. Three hours after transfection, they were incubated with long peptide gp100184–227 and tested for their ability to stimulate CTL 7 two hours later (left panel). As a control for TAP inhibition by ICP47, endogenous presentation of the TAP-dependent epitope derived from ubiquitous self-protein RU1 was tested with an RU134–42/HLA-B51–specific CTL clone (right panel) (21). Values are means ± SD of triplicates from a representative experiment repeated at least three times. ND, not detected.

FIGURE 1.

Human Mo-iDCs cross-present the long gp100 peptide in the vacuolar pathway (A, left panel) HLA-A*0201–positive Mo-iDCs can stimulate gp100209–217-specific CTL clone 7 to produce IFN-γ after a 2-h pulse with long peptide gp100184–227 at 37°C but not at 4°C. As controls (right panel), Mo-iDCs pulsed with short epitope peptide gp100209–217 stimulate CTL clone 7 at both 37°C and 4°C. (B) Fixed Mo-iDCs cannot cross-present the long gp100 peptide. Mo-iDCs were fixed with 0.004% glutaraldehyde for 45 s and then pulsed for 2 h with the long gp100 peptide (left panel) or the final epitope (right panel). CTL was added and the production of IFN-γ was measured after an overnight incubation. (C) The processing of the long gp100 peptide is not performed by extracellular peptidases. The long gp100 peptide was first incubated with HLA-A*0201 DCs at 37°C for 2 h. Next, the supernatant was collected and pulsed onto HLA-A*0201+ DCs at 37°C or 4°C for 2 h. After washing, the supernatant-pulsed DCs were assayed for their ability to stimulate IFN-γ production by gp100209–217-specific CTL clone 7. (D) Proteasome inhibition by epoxomicin (0.5 μM) does not block cross-presentation of the long gp100 peptide (left panel), whereas it blocks endogenous presentation of the same epitope by DCs transfected with gp100 mRNA (right panel). (E) Mo-iDCs (both HLA-A*0201+ and HLA-B*51+) were transfected with ICP47 or EGFP mRNA by electroporation. Three hours after transfection, they were incubated with long peptide gp100184–227 and tested for their ability to stimulate CTL 7 two hours later (left panel). As a control for TAP inhibition by ICP47, endogenous presentation of the TAP-dependent epitope derived from ubiquitous self-protein RU1 was tested with an RU134–42/HLA-B51–specific CTL clone (right panel) (21). Values are means ± SD of triplicates from a representative experiment repeated at least three times. ND, not detected.

Close modal
FIGURE 2.

Long peptide cross-presentation is favored by suboptimally loaded HLA-A*0201 molecules in Mo-iDC. (A) T2 and T1 cells were pulsed with peptide gp100184–227 for the cross-presentation assay. (B) T2 cells were treated with 0.5 μM epoxomicin or vehicle (DMSO) before and during incubation with long peptide gp100184–227. After washing, they were tested for their ability to stimulate gp100-specific CTL to produce IFN-γ. (C) Mo-iDCs were treated overnight with (Z-LL)2-ketone (45 μM) or vehicle (DMSO) before transfection with mRNA encoding ICP47 or EGFP. Three hours after transfection, some of the cells were pulsed with peptide gp100184–227 for the cross-presentation assay (left). Another portion of the same cells was infected with a recombinant MAGE-A3–encoding vaccinia virus to verify efficient TAP blockade (right). (ZLL)2-ketone was maintained in the culture medium during transfection and infection. Two hours postinfection, cells were washed and tested for their ability to stimulate CTL clone A10, which recognizes peptide MAGE-A3168–176 presented by HLA-A*0101 (22). (D) T2 cells were treated with (ZLL)2-ketone (45 μM) or vehicle for 2 or 16 h at 37°C, then labeled with Alexa-633-conjugated anti-HLA-I murine mAb W6/32 at 4°C. HLA-class I expression level was represented by geometric mean fluorescence intensity (GMFI). (E) T2 and T1 cells were treated with (Z-LL)2-ketone (45 μM) or vehicle overnight before being pulsed with peptide gp100184–227 for the cross-presentation assay. (F) Mo-iDCs were treated with l-leucinethiol (30 μM) or vehicle (Tris(2-carboxyethyl)phosphine [TCEP]) for 3 h before pulsing with peptide gp100184–227 for cross-presentation (left) or electroporation with gp100 mRNA to test endogenous presentation (right). After 3 h of additional incubation in the presence of l-leucinethiol or vehicle, cells were tested for their ability to stimulate gp100-specific CTL. (G) Peptides of various affinities for HLA-A2 were targeted into the ER of T2 cells with lentiviral constructs. The constructs used IL-2 signal sequence (IL-2ss) as a targeting motif and IRES-mCherry as the selection marker. Forty-eight hours after transduction, mCherry-positive cells were sorted, cultured for 48 h, and used for a cross-presentation assay. The proper expression of the constructs was monitored by FACS for mCherry fluorescence, as indicated in the column at the right (MFI). Control non–HLA-A2–binding peptide EVDPIGHLY corresponds to peptide MAGE-A3168–176, which binds HLA-A1. The affinity of the peptides for HLA-A*0201 as predicted by algorithms BIMAS and SYFPEITHI are indicated. HLA-A2 binding affinity was also estimated from the peptide concentration (EC50) needed to achieve 50% of maximal stabilization of HLA-A2 molecules on T2 cells after acid strip, and from the maximal level of stabilization relative to peptide gp100209–217 (relative efficacy). EC50 shown are the mean of three experiments. Each graph is representative of at least three independent experiments. For all graphs, assays were performed in triplicate or more and repeated at least three times. MFI, mean fluorescence intensity; ND, not detected.

FIGURE 2.

Long peptide cross-presentation is favored by suboptimally loaded HLA-A*0201 molecules in Mo-iDC. (A) T2 and T1 cells were pulsed with peptide gp100184–227 for the cross-presentation assay. (B) T2 cells were treated with 0.5 μM epoxomicin or vehicle (DMSO) before and during incubation with long peptide gp100184–227. After washing, they were tested for their ability to stimulate gp100-specific CTL to produce IFN-γ. (C) Mo-iDCs were treated overnight with (Z-LL)2-ketone (45 μM) or vehicle (DMSO) before transfection with mRNA encoding ICP47 or EGFP. Three hours after transfection, some of the cells were pulsed with peptide gp100184–227 for the cross-presentation assay (left). Another portion of the same cells was infected with a recombinant MAGE-A3–encoding vaccinia virus to verify efficient TAP blockade (right). (ZLL)2-ketone was maintained in the culture medium during transfection and infection. Two hours postinfection, cells were washed and tested for their ability to stimulate CTL clone A10, which recognizes peptide MAGE-A3168–176 presented by HLA-A*0101 (22). (D) T2 cells were treated with (ZLL)2-ketone (45 μM) or vehicle for 2 or 16 h at 37°C, then labeled with Alexa-633-conjugated anti-HLA-I murine mAb W6/32 at 4°C. HLA-class I expression level was represented by geometric mean fluorescence intensity (GMFI). (E) T2 and T1 cells were treated with (Z-LL)2-ketone (45 μM) or vehicle overnight before being pulsed with peptide gp100184–227 for the cross-presentation assay. (F) Mo-iDCs were treated with l-leucinethiol (30 μM) or vehicle (Tris(2-carboxyethyl)phosphine [TCEP]) for 3 h before pulsing with peptide gp100184–227 for cross-presentation (left) or electroporation with gp100 mRNA to test endogenous presentation (right). After 3 h of additional incubation in the presence of l-leucinethiol or vehicle, cells were tested for their ability to stimulate gp100-specific CTL. (G) Peptides of various affinities for HLA-A2 were targeted into the ER of T2 cells with lentiviral constructs. The constructs used IL-2 signal sequence (IL-2ss) as a targeting motif and IRES-mCherry as the selection marker. Forty-eight hours after transduction, mCherry-positive cells were sorted, cultured for 48 h, and used for a cross-presentation assay. The proper expression of the constructs was monitored by FACS for mCherry fluorescence, as indicated in the column at the right (MFI). Control non–HLA-A2–binding peptide EVDPIGHLY corresponds to peptide MAGE-A3168–176, which binds HLA-A1. The affinity of the peptides for HLA-A*0201 as predicted by algorithms BIMAS and SYFPEITHI are indicated. HLA-A2 binding affinity was also estimated from the peptide concentration (EC50) needed to achieve 50% of maximal stabilization of HLA-A2 molecules on T2 cells after acid strip, and from the maximal level of stabilization relative to peptide gp100209–217 (relative efficacy). EC50 shown are the mean of three experiments. Each graph is representative of at least three independent experiments. For all graphs, assays were performed in triplicate or more and repeated at least three times. MFI, mean fluorescence intensity; ND, not detected.

Close modal
FIGURE 3.

Cross-presentation of the long gp100 peptide makes use of nascent HLA-I molecules. (A) Mo-iDCs were preincubated for 1 h at 20°C and then assayed for cross-presentation (2 h) of the long gp100 peptide at the indicated temperature. Degranulation assay performed at 20°C was used as read-out, which is quicker and less temperature-sensitive than the cytokine production assay. Values are means ± SD of triplicates. (B) Surface HLA-I (left) or CD206 (right) molecules of Mo-iDC were labeled with Alexa-633-conjugated W6/32 or Alexa-488-conjugated anti-CD206. After washing, cells were incubated at 37°C in the presence of 220 μM primaquine to prevent recycling. Internalization was stopped by transferring cells to 4°C at different time points. The remaining surface Ag/Ab complexes were stripped off by citric acid (pH 3.0) treatment. Internalized HLA-I or CD206 were then quantified by FACS analysis measuring geometric mean fluorescence intensity (GMFI) increase with time and shown as a ratio to the GMFI of Ab-labeled cells without acid-strip (100%). GMFI of cells labeled with isotype control Ab was considered as 0%. (C) HLA-I recycling was evaluated on Mo-iDCs at different temperatures. Anti–HLA-I murine mAb W6/32 labeled Mo-iDC were incubated for 12 min at the indicated temperature in the presence of primaquine (220 μM). After acid stripping with 50 mM citric acid (pH 3.0) and washing away of primaquine, the reappearance of HLA-I/Ab complexes at the cell surface was followed at different temperatures and analyzed by FACS using labeled anti-murine IgG Ab. Geometric mean fluorescent intensity (GMFI) of non–acid-stripped cells was considered as 100%. Values are means ± SD of duplicates. (D) Surface HLA-A2 of T2 cells were acid stripped and reconstituted at 20°C overnight in the presence of β2-microglobulin and short peptides with different HLA-A2 binding affinities (Fig. 2G). Cells were pulsed with long peptide gp100184–227 for the cross-presentation assay. In parallel, surface HLA-A2 expression was detected with conformation-dependent anti–HLA-A2 Ab BB7.2 and analyzed by FACS. The expression level is shown as mean fluorescence intensity. (E) Mo-iDCs were treated with cycloheximide (1 mg/ml) and tested for cross-presentation of the long gp100 peptide. Values are means ± SD of triplicates. Each representative experiment shown was repeated at least three times.

FIGURE 3.

Cross-presentation of the long gp100 peptide makes use of nascent HLA-I molecules. (A) Mo-iDCs were preincubated for 1 h at 20°C and then assayed for cross-presentation (2 h) of the long gp100 peptide at the indicated temperature. Degranulation assay performed at 20°C was used as read-out, which is quicker and less temperature-sensitive than the cytokine production assay. Values are means ± SD of triplicates. (B) Surface HLA-I (left) or CD206 (right) molecules of Mo-iDC were labeled with Alexa-633-conjugated W6/32 or Alexa-488-conjugated anti-CD206. After washing, cells were incubated at 37°C in the presence of 220 μM primaquine to prevent recycling. Internalization was stopped by transferring cells to 4°C at different time points. The remaining surface Ag/Ab complexes were stripped off by citric acid (pH 3.0) treatment. Internalized HLA-I or CD206 were then quantified by FACS analysis measuring geometric mean fluorescence intensity (GMFI) increase with time and shown as a ratio to the GMFI of Ab-labeled cells without acid-strip (100%). GMFI of cells labeled with isotype control Ab was considered as 0%. (C) HLA-I recycling was evaluated on Mo-iDCs at different temperatures. Anti–HLA-I murine mAb W6/32 labeled Mo-iDC were incubated for 12 min at the indicated temperature in the presence of primaquine (220 μM). After acid stripping with 50 mM citric acid (pH 3.0) and washing away of primaquine, the reappearance of HLA-I/Ab complexes at the cell surface was followed at different temperatures and analyzed by FACS using labeled anti-murine IgG Ab. Geometric mean fluorescent intensity (GMFI) of non–acid-stripped cells was considered as 100%. Values are means ± SD of duplicates. (D) Surface HLA-A2 of T2 cells were acid stripped and reconstituted at 20°C overnight in the presence of β2-microglobulin and short peptides with different HLA-A2 binding affinities (Fig. 2G). Cells were pulsed with long peptide gp100184–227 for the cross-presentation assay. In parallel, surface HLA-A2 expression was detected with conformation-dependent anti–HLA-A2 Ab BB7.2 and analyzed by FACS. The expression level is shown as mean fluorescence intensity. (E) Mo-iDCs were treated with cycloheximide (1 mg/ml) and tested for cross-presentation of the long gp100 peptide. Values are means ± SD of triplicates. Each representative experiment shown was repeated at least three times.

Close modal
FIGURE 4.

HLA-I molecules used for cross-presentation traffic to the cross-presenting vesicle through a nonclassical secretion pathway. (A) Low temperature blocks conventional secretion of HLA-I. After acid stripping, reappearance of HLA-I at the cell surface of Mo-iDCs was followed by FACS at the indicated temperature with Alexa-633–conjugated anti–HLA-I mAb W6/32. Values are means ± SD of three independent experiments. (B) Low temperature blocks endogenous Ag presentation by HLA-I molecules. HLA-B51–positive or –negative Mo-iDCs were acid-stripped and incubated at the indicated temperature for 2 h and assayed for their ability to stimulate RU134–42/HLA-B51–specific CTL clone CTL381/84 in a degranulation assay performed at 20°C. (C and D) Mo-iDCs were transfected with (C) Sec22b- or (D) CD74-targeting siRNA by electroporation on day 3 (CD74) or day 4 (Sec22b) after differentiation and assayed for their ability to cross-present the long gp100 peptide to gp100209–217-specific CTL after 72 h (for CD74) or 48 h (Sec22b) of transfection. Efficiency of knocking down was analyzed by Western blot with cells collected at the same time as the cross-presentation assay. The black lines indicate where parts of the image were joined. Values shown in (C) and (D) are means ± SD of triplicates. The experiment in (C) was repeated three times, and the experiment in (D) was repeated twice.

FIGURE 4.

HLA-I molecules used for cross-presentation traffic to the cross-presenting vesicle through a nonclassical secretion pathway. (A) Low temperature blocks conventional secretion of HLA-I. After acid stripping, reappearance of HLA-I at the cell surface of Mo-iDCs was followed by FACS at the indicated temperature with Alexa-633–conjugated anti–HLA-I mAb W6/32. Values are means ± SD of three independent experiments. (B) Low temperature blocks endogenous Ag presentation by HLA-I molecules. HLA-B51–positive or –negative Mo-iDCs were acid-stripped and incubated at the indicated temperature for 2 h and assayed for their ability to stimulate RU134–42/HLA-B51–specific CTL clone CTL381/84 in a degranulation assay performed at 20°C. (C and D) Mo-iDCs were transfected with (C) Sec22b- or (D) CD74-targeting siRNA by electroporation on day 3 (CD74) or day 4 (Sec22b) after differentiation and assayed for their ability to cross-present the long gp100 peptide to gp100209–217-specific CTL after 72 h (for CD74) or 48 h (Sec22b) of transfection. Efficiency of knocking down was analyzed by Western blot with cells collected at the same time as the cross-presentation assay. The black lines indicate where parts of the image were joined. Values shown in (C) and (D) are means ± SD of triplicates. The experiment in (C) was repeated three times, and the experiment in (D) was repeated twice.

Close modal

PBMCs were isolated from whole blood of hemochromatosis patients with Ficoll gradient (Lymphoprep; Axis-Shield PoC), after approval from the Commission d’Ethique Biomédicale Hospitalo-Facultaire from the Université Catholique de Louvain institutional review board. Monocytes were enriched on Percoll as described (23) and isolated by adherence. After washing away nonadherent cells with PBS, adherent cells were kept in culture in RPMI 1640 medium supplemented with 10% FCS, 200 U/ml human IL-4 (made in-house) and 70 ng/ml GM-CSF (Leukine [sargramostim]). The cultures were fed with fresh medium and cytokines every 2–3 d. The monocyte-derived immature DCs (Mo-iDCs) were harvested and used on days 5 to 7 of differentiation.

Fluorochrome-conjugated secondary Abs were obtained from Life Technologies. Alexa-633 (Life Technologies) conjugated and nonconjugated murine mAb W6/32 were produced in-house. FITC-conjugated mouse anti-human CD107a/CD107b, and FITC-conjugated anti-HLA-A2 murine mAb BB7.2 were from BD Pharmingen. Mouse anti-human β-actin AC15 and mouse anti-human SEC22B were from Sigma-Aldrich. Rabbit anti-human CD74 polyclonal Ab Matilda was given by P. Cresswell (24). All inhibitors were obtained from Sigma-Aldrich. Long peptide gp100184–227 was given by P. Cresswell. The other peptides were synthesized in-house.

Target cells were pulsed with long peptide (5 μM) for 2–4 h at the indicated temperatures in complete medium, and cocultured with CTL after washing. CTL activation was evaluated in a cytokine production assay or a degranulation assay. For IFN-γ and TNF production assay, 40,000–50,000 Ag-loaded target cells were cocultured with 10,000–20,000 CTLs in round-bottom 96-well plates in triplicate. Then IFN-γ or TNF was quantified by ELISA in the supernatant after 18 h. Degranulation assay was done by coculturing target cells with CTLs in the presence of monensin and FITC-labeled mouse anti-human CD107a and CD107b Ab at 20°C. The percentage of FITC-positive T cells among the CD8+ T cells was determined by FACS after 3 h of coculture. To study the effect of inhibitors, unless stated otherwise, DCs were treated with drugs for 30 min before and during the 2 h of pulse with the long precursor peptides.

EGFP-, gp100-, and ICP47-encoding cDNAs were first cloned into the backbone of pST1-A(120) (25). Next, the plasmids were linearized with SapI, and mRNA was prepared as described (26). Electroporation was performed in a 4-mm gap electroporation cuvette using gene pulse Xcell electroporation system (Bio-Rad) with 2.5 million DCs resuspended in 200 μl OPTI-MEM I no phenol red (Life Technologies) containing 10 μg mRNA, under the following conditions: mode, exponential decay; voltage, 300 V; capacitance, 150 μF; resistance, ∞ Ω, resulting in a pulse time of ∼10 ms.

CD74-targeted 3-RNAi mix (CD74HSS190576, CD74HSS190577, CD74HSS190578, equally mixed) and its control (nontargeting negative control medium GC 12935-300) stealth small interfering RNA (siRNA) were from Invitrogen. Sec22b-targeted siRNA (on-target plus SMARTpool siRNA L-011963-00-0005) and its control (non-targeting SiControl) were from Dharmacon. To knock down CD74, DCs were transfected with 1 μM siRNA on day 3 after differentiation and assayed for their ability of cross-presentation 72 h after transfection. To knock down Sec22b, DCs were transfected with 1 μM siRNA on day 4 after differentiation and assayed for their ability of cross-presentation 48 h after transfection. Efficacy of knocking down was analyzed by Western blot with cells collected at the same time as the cross-presentation assay. siRNA were delivered into cells by electroporation as described above for mRNA transfection.

Mo-iDCs were first incubated with murine mAb W6/32 (40 μg/ml) at 4°C. After thorough washing, cells were incubated at 20°C or 37°C for 12 min in the presence of 220 μM of primaquine. Next, the cells were stripped with 50 mM citric acid (pH 3) for 2 min on ice and neutralized immediately with 150 mM NaH2PO4 (pH 10.5). After washing, cells were incubated at 37°C or 20°C. Aliquots were removed at various intervals and placed on ice. Recycled HLA-I/Ab complexes were detected with flow cytometry using Alexa-647 conjugated Donkey anti-mouse IgG.

For HLA-class I secretion analysis, DCs were first stripped with 50 mM citric acid for 2 min. After neutralization and wash, cells were incubated at 20°C or 37°C in culture medium. Aliquots were removed at various intervals and placed on ice. Reappearance of HLA-class I molecules was detected with Alexa-633 conjugated mAb W6/32 and analyzed by flow cytometry.

Lentiviral vectors were derived from pCCLsin.PPT.hPGK.GFP (27). Control pTM895 was generated by inserting, in the backbone pCCLsin.PPT.hPGK.GFP, an IRES from TMEV (28) and the eGFP coding sequence downstream of the PGK promoter. The ICP47-coding cDNA was cloned from pBJ1-Neo (gifted by H.G. Rammensee, University of Tübingen) into pTM895 upstream the eGFP-IRES sequence. pTM945 vector, which was used to deliver peptides into the ER in a TAP-independent manner, was generated by inserting in the backbone of the same vector: a CMV promoter, a multicloning site, an IRES from TMEV and an mCherry coding sequence (28). The coding sequences of IL-2 signal sequence-peptide fusion polypeptide were cloned into the multicloning site of pTM945. Vectors pTM897, pTM895, and pTM945 were provided by Prof. Thomas Michiels (de Duve Institute, Brussels, Belgium). Lentiviral particles were produced upon cotransfection of HEK-293T cells with vector plasmid, and plasmids pMDLg/pRRE#54, pRSV-Rev and pMD2.VSVG (provided by Luigi Naldini, Ospedale San Raffaele, Milan, Italy) (27). Helper lentiviral particles SIVmac were produced upon cotransfection of HEK-293T cells with envelope plasmid pMD2.VSVG and SIVmac package construct pSIV3+ (gifted by Prof. Cimarelli, Ecole Normale Supérieure de Lyon, France) (23). To transduce Mo-iDCs, cells were incubated with both viral particles on day 4 of differentiation. Next, cells were harvested and analyzed on day 7. T2 cells were transduced with lentiviral particles by spin infection at 2,400 rpm and 32°C for 90 min in the presence of 8 μg/ml of polybrene. Forty-eight hours after transduction, mCherry-positive cells were sorted, seeded to 2.5 105 cells/ml, and cultured for 24 or 48 h. Cross-presentation assay was then performed.

For the reconstitution of cell surface HLA-A2, T2 cells were first stripped with 50 mM citric acid for 2 min. After neutralization and washing, cells were incubated with β2-microglobulin (2.5 μM) and short peptides (10 μM) at 18°C for 20 h. For peptide affinity estimation after acid-stripping, T2 cells were incubated with β2-microglobulin (2.5 μM) and serially diluted peptides at 18°C for 3 h. Next, surface HLA-A2 was detected with FITC-conjugated conformation-dependent anti–HLA-A2 murine mAb BB7.2, and analyzed by flow cytometry. Peptide concentrations were then plotted against the mean fluorescence intensity of surface HLA-A2. The concentration needed to restore surface HLA-A2 molecules to 50% of the maximum was calculated (EC50). The maximum level of HLA-A2 stabilization achieved with each peptide relative to that obtained with peptide gp100205-217 was also calculated (relative efficacy).

We used a 44-aa-long peptide derived from melanocytic protein gp100 (gp100184–227) to study cross-presentation of long peptides by hDCs (Fig. 1). After a 2-h incubation with this peptide at 37°C, HLA-A2–positive Mo-iDCs stimulated the production of IFN-γ by a CTL clone recognizing the HLA-A*0201–restricted epitope gp100209–217 comprised in long peptide gp100184–227. We observed no cross-presentation when the long peptide was incubated with Mo-iDC at 4°C, or with fixed Mo-iDC (Fig. 1A, B). We performed a two-step experiment to exclude the possibility that the long peptide was processed in the extracellular milieu and loaded directly onto surface HLA-A*0201 molecules in our experimental setup. We first incubated the long peptide for 2 h at 37°C with HLA-A*0201–negative Mo-iDCs. We then collected and incubated the supernatant with HLA-A*0201–positive Mo-iDCs in a cross-presentation assay performed at 4°C or 37°C. If the long peptide was processed in the extracellular milieu during the first incubation, the supernatant of this first step should activate the CTL when loaded at 4°C onto HLA-A*0201-positive DC in the second step. This was not the case (Fig. 1C). This result therefore excluded the processing of the long peptide by extracellular proteases. To strengthen this conclusion further, we blocked endocytosis by inhibiting actin polymerization with cytochalasin B (Supplemental Fig. 1A). We observed a dose-dependent inhibition of cross-presentation of the long peptide. Because cytochalasin B inhibits endocytosis but not proteolysis, this result further confirmed that cross-presentation of the long peptide in our system depends on endocytosis and involves an intracellular processing step.

To determine whether long-peptide cross-presentation followed a cytosolic or a vacuolar pathway, we tested the effect of proteasome and TAP inhibition. We observed that cross-presentation of the long gp100 peptide was not reduced by inhibition of the proteasome with epoxomicin (Fig. 1D) or MG132 (Supplemental Fig. 1B), whereas endogenous presentation of the same gp100 peptide introduced by mRNA electroporation was blocked by epoxomicin (Fig. 1D). TAP blockade with viral inhibitor ICP47, which was introduced into the cells by mRNA electroporation, did not inhibit the cross-presentation of the long gp100 peptide either, whereas endogenous presentation of an ubiquitous self-peptide (RU134–42) was blocked by ICP47 (Fig. 1E). We concluded that cross-presentation of the long peptide occurred in the vacuolar pathway.

We were intrigued by the consistent 2-fold increase of cross-presentation observed after TAP inhibition (Fig. 1E). We performed an extensive phenotyping of Mo-iDCs to verify that ICP47 transfection did not activate the production of IL-10 and IL-12, nor did it modify the expression of costimulatory molecules (Supplemental Fig. 2). Although it is well established that DCs are the most efficient cross-presenting cells, other cells might cross-present with a low efficiency. We reasoned that if the cross-presentation of long peptides was indeed favored by TAP blockade, it might be increased or revealed in other cells by the loss of TAP. We therefore compared the cross-presentation of the long gp100 peptide by TAP-deficient T2 cells and their parental TAP-competent T-B lymphoblast hybrid cells T1 (29). We found that T2 cells cross-presented long peptide gp100184–227 quite efficiently in a proteasome-independent manner, and much better than its parental cells T1 (Fig. 2A, B). The cross-presentation ability of T2 cells was not modified by ICP47 transfection, which excluded off-target effects of ICP47 and further validated TAP inhibition as responsible for the increased cross-presentation observed after transfection of ICP47 in Mo-iDCs (Supplemental Fig. 3A). Because TAP blockade results in a shortage of peptide supply in the ER, we considered two possible explanations for the increased cross-presentation: either more empty HLA-A*0201 molecules became available in the ER to load the cross-presented epitope, provided it can reach the ER, or the ER contained more HLA-A*0201 molecules loaded with suboptimal peptides derived from signal peptides (3033). Those suboptimal HLA-peptide complexes might then exit the ER and reach another compartment, where the preloaded suboptimal peptides would be exchanged for the cross-presented epitope.

If the increased cross-presentation after TAP inhibition was due to more empty HLA-A*0201 molecules in the ER, then blocking the release of signal peptides into the ER lumen by inhibiting signal peptide peptidase (SPP) should result in even more empty HLA-A*0201 molecules and a further increased cross-presentation. Conversely, if increased cross-presentation was due to more HLA-A*0201 molecules loaded with suboptimal signal peptides, inhibiting signal peptide release from the ER membrane should reduce suboptimally loaded HLA-A*0201 molecules and reduce cross-presentation in TAP-deficient cells. To inhibit SPP, we treated Mo-iDCs with (Z-LL)2-ketone before electroporating ICP47 or EGFP mRNA and incubating with long peptide gp100184–227. SPP inhibition completely prevented the increase of cross-presentation induced by TAP inhibition (Fig. 2C, left), which was confirmed by the failure of the very same ICP47-transfected DCs to present the HLA-A*0101–restricted epitope MAGE-A3168–176 provided endogenously with a vaccinia vector (22, 34) (Fig. 2C, right). The lack of effect of (Z-LL)2-ketone on control cells transfected with EGFP instead of ICP47 indicated that the effect of (Z-LL)2-ketone on ICP47-transfected DC was not caused by toxicity or inhibition of the secretion of HLA-I or costimulatory signals. This was further confirmed by analyzing the expression of costimulatory molecules such as CD80 and CD86, and the production of cytokines such as IL-10 and IL-12 by (Z-LL)2-ketone treated cells (Supplemental Fig. 2). That (Z-LL)2-ketone efficiently reduced the number of suboptimally loaded HLA-A2 molecules in TAP-deficient cells was confirmed by the decreased expression of HLA class I—which are mainly HLA-A2 in these cells—at the surface of T2 cells treated with (Z-LL)2-ketone (Fig. 2D). Thus, these results supported the second model positing that increased cross-presentation observed after TAP inhibition resulted from increased abundance of suboptimally loaded HLA-A*0201 molecules available for peptide exchange. The cross-presented epitope is seemingly not delivered into the ER, otherwise it would be able to load empty HLA-A*0201 molecules in the ER and cross-presentation should be increased by (Z-LL)2-ketone. It follows that peptide exchange on suboptimally loaded HLA-A*0201 molecules should occur in a post-ER compartment. In line with this, the cross-presentation of long peptide gp100184–227 by T2 cells was also inhibited dramatically by (ZLL)2-ketone (Fig. 2E), indicating that, like in DC, it relied on suboptimally loaded HLA-A*0201 molecules.

To determine whether suboptimally loaded HLA-A*0201 molecules were also needed for cross-presentation of long peptides in TAP-competent cells, we took advantage of the fact that ERAP, an aminopeptidase that trims antigenic peptides in the ER, is required to produce an optimal repertoire of stable peptide–MHC-I complexes (35). We observed that ERAP inhibition by l-leucinethiol dramatically increased cross-presentation of the gp100 epitope by Mo-iDCs, whereas endogenous presentation of the same gp100 epitope after electroporation of Mo-iDCs with gp100 mRNA was almost completely blocked (Fig. 2F). Again, no significant difference was observed concerning the expression of costimulatory signals by cells after l-leucincethiol or vehicle treatment (Supplemental Fig. 2). These results further supported the essential role of suboptimally loaded HLA-A*0201 molecules for cross-presentation of long peptides.

To confirm further the role of suboptimally loaded HLA-I molecules, we used lentiviral constructs enabling the delivery of peptides into the ER in a TAP-independent manner, through fusion to the signal sequence of IL-2 (36). From the list of HLA-A*0201-binding peptides eluted from TAP-deficient T2 cells (37), we selected two short peptides with low/intermediate affinity for HLA-A*0201 and one peptide with high affinity (Fig. 2G). As a negative control, we also used a peptide known to bind to HLA-A1 but not to HLA-A2 (MAGE-A3168-176; EVDPIGHLY). We reasoned that after introduction of the constructs in T2 cells, the high amounts of ER-delivered peptides should modulate cross-presentation efficiency, with the low–intermediate HLA-A2 binders (suboptimal peptides) favoring cross-presentation, as opposed to the high HLA-A2 binder and the nonbinder peptides. This is exactly what we observed (Fig. 2G). As a control for the ER delivery of the peptides, we observed the HLA-A1–restricted peptide was efficiently presented to specific CTL in a TAP-independent manner (Supplemental Fig. 4). These results supported our model that cross-presentation occurs by peptide exchange on suboptimally loaded HLA-I molecules.

The results obtained so far suggest that cross-presentation of the long gp100 peptide occurs through peptide exchange on suboptimally loaded HLA-A*0201 molecules in a post-ER compartment containing the processed epitope derived from the engulfed long peptide. Because the vacuolar pathway of cross-presentation has been considered to make use of recycling MHC-I molecules that are derived from the cell surface (1518), we asked whether those suboptimally loaded HLA-I were recycled or were newly synthesized. To avoid unspecific effects of inhibitors like primaquine on vesicle trafficking, we chose to block HLA-I recycling by incubating cells at low temperature, which is reported to block recycling of surface molecules (38). We observed that cross-presentation was not reduced at 20°C (Fig. 3A). We then confirmed that low temperature efficiently blocked HLA-I recycling in our Mo-iDCs cellular system using a two-step approach. We first showed that internalization of HLA-I, which conditions recycling, was almost completely blocked at 20°C (Fig. 3B, left), in line with the observation that endocytosis of MHC-I depends on membrane rafts, which are highly temperature sensitive (39), as opposed to the clathrin-dependent endocytosis of mannose receptor CD206 (38, 40, 41) (Fig. 3B, right). We then evaluated the recycling step at different temperatures (Fig. 3C). Surface HLA-I molecules of Mo-iDCs were first labeled with conformation-dependent anti–HLA-I murine mAb W6/32. Next, HLA-I/Ab complexes were allowed to internalize for 12 min at 37°C or 20°C in the presence of primaquine (220 μM) to prevent recycling. After stripping non-internalized HLA-I/Ab complexes and washing away primaquine, we followed the reappearance of HLA-I/Ab complexes at the cell surface at 20°C or 37°C by FACS using labeled anti-murine IgG Ab. When both internalization and recycling were performed at 37°C, we observed a fast kinetics of recycling, which was consistent with the observation that HLA-I are recycled through the RAB-35–dependent fast recycling pathway (42, 43). When recycling was performed at 20°C, we observed a striking reduction in the number of HLA-I recycled to the cell surface. When both internalization and recycling were performed at 20°C, HLA-I recycling to the cell surface was completely blocked. Because cross-presentation was not blocked at 20°C but recycling of HLA-I was, we concluded that cross-presentation of long peptides did not use recycled HLA-I for peptide exchange. To confirm further that cell surface HLA-I molecules are dispensable for long-peptide cross-presentation, we acid-stripped T2 cells and reconstituted cell surface HLA-A2 with short peptides of different HLA-A2 binding affinities before the cross-presentation assay. Although the extent of surface HLA-A2 reconstitution varied according to the peptide affinity for HLA-A2, the cross-presentation ability of the cells remained unaffected (Fig. 3D). If cross-presentation does not use recycled HLA-I molecules, it must use nascent HLA-I. This was confirmed by the inhibitory effect of protein synthesis inhibitor cycloheximide on cross-presentation of the long gp100 peptide (Fig. 3E).

Low temperature incubation not only blocks recycling of surface molecules; it also blocks the exit of secretory and surface proteins from the trans-Golgi-network in the classical secretion pathway (44, 45). We confirmed in our cellular system that incubation at 20°C prevented classical secretion of HLA-I molecules (Fig. 4A) and the presentation of endogenously derived class I–restricted Ags (Fig. 4B). Because cross-presentation was not affected by low temperature, we concluded that the nascent HLA-I molecules used for cross-presentation followed a nonclassical secretion pathway.

The ER-Golgi Intermediate Compartment is an important early secretion compartment involved in the sorting of ER-synthesized proteins. Cebrian et al. (12) reported a role for SEC22B in escorting ER-resident molecules from the ER-Golgi Intermediate Compartment to the phagosome, making it a good candidate to escort HLA-I for long-peptide cross-presentation. However, we observed that knocking down Sec22b with siRNA in Mo-iDCs did not prevent cross-presentation (Fig. 4C). Recently, CD74 was reported to escort MHC-I from the ER to the endolysosome compartment to promote cross-presentation (13). However, knocking down CD74 in Mo-iDC did not prevent cross-presentation of the long peptide either (Fig. 4D).

Taken together, the results reported above suggest that cross-presentation of the long gp100 peptide makes use of peptide-loaded nascent HLA-I molecules that follow an alternative secretory pathway to reach the cross-presenting vacuole, where preloaded peptides are exchanged for the cross-presented peptide before transfer of the final peptide/HLA complexes to the cell surface.

We used another long peptide, derived from melanocytic protein Melan-A to determine whether other long peptides followed the same cross-presenting pathway as gp100184–227. Long peptide Melan-A15-40A27L (15KGHGHSYTTAE26ELAGIGILTV35ILGVL40), which comprises the heteroclitic antigenic peptide Melan-A26-35A27L with an alanine-to-leucine substitution in position 27 to increase binding to HLA-A*0201 (46, 47), was efficiently cross-presented to Melan-A26–35-specific CTL by Mo-iDCs in a proteasome- and TAP-independent manner (Fig. 5A, B). Although ICP47 efficiently blocked TAP, as indicated by the reduced HLA-I surface expression (Supplemental Fig. 3B), it did not increase cross-presentation of the long Melan-A peptide as it did for the long gp100 peptide. Consistently, there was no effect of l-leucinethiol on the cross-presentation either (Fig. 5C). This might result from the higher affinity of Melan-A26-35A27L for HLA-A*0201 as compared with gp100209–217 (Supplemental Fig. 3C). The higher the affinity of the cross-presented epitope, the easier peptide exchange can take place, so that the limited pool of suboptimally loaded HLA-I molecules available in TAP- or ERAP-competent cells might become sufficient for optimal cross-presentation. This is supported by the fact that l-leucinethiol strongly increased cross-presentation of the wild-type Melan-A long peptide (Melan-A15–40) (Fig. 5C), whose epitope (Melan-A26–35) has a much lower affinity for HLA-A2 (Supplemental Fig. 3C) and is otherwise not cross-presented efficiently. As with the gp100 peptide, low temperature (20°C) did not block cross-presentation of the long Melan-A peptide (Fig. 5D), further supporting the involvement of an alternative secretion pathway of the HLA-I molecules used for cross-presentation. These results indicate that the cross-presentation of other long peptides follow the same pathway as the gp100 long peptide, involving vacuolar peptide exchange on nascent suboptimally loaded HLA-I molecules, which follow a nonclassical secretion pathway.

FIGURE 5.

Cross-presentation of the long Melan-A peptide. (A) Mo-iDCs were treated with 0.5 μM epoxomicin or with vehicle (DMSO) before and during incubation with peptide Melan-A15–40 or the heteroclitic peptide Melan-A15–40A27L. Melan-A26–35–specific CTL clone 549/18 was added, and cytokine production was measured after 18 h. (B) Mo-iDCs were transfected with mRNA encoding ICP47 or EGFP. Three hours after transfection, they were incubated with peptide Melan-A15–40A27L for 2 h. Melan-A26–35–specific CTL was added and cytokine production was measured. (C) Mo-iDCs were pulsed with peptide Melan-A15–40 or Melan-A15–40A27L in the presence of l-leucinethiol (30 μM) or vehicle (Tris[2-carboxyethyl]phosphine) after pretreatment with the inhibitor for 3 h. Melan-A26–35–specific CTL was added, and cytokine production was measured. (D) Cross-presentation of the long peptide Melan-A15–40A27L was performed at the indicated temperature and evaluated in a degranulation assay performed at 20°C. For all experiments, values are means ± SD of triplicates from a representative experiment repeated at least three times. ND, not detected.

FIGURE 5.

Cross-presentation of the long Melan-A peptide. (A) Mo-iDCs were treated with 0.5 μM epoxomicin or with vehicle (DMSO) before and during incubation with peptide Melan-A15–40 or the heteroclitic peptide Melan-A15–40A27L. Melan-A26–35–specific CTL clone 549/18 was added, and cytokine production was measured after 18 h. (B) Mo-iDCs were transfected with mRNA encoding ICP47 or EGFP. Three hours after transfection, they were incubated with peptide Melan-A15–40A27L for 2 h. Melan-A26–35–specific CTL was added and cytokine production was measured. (C) Mo-iDCs were pulsed with peptide Melan-A15–40 or Melan-A15–40A27L in the presence of l-leucinethiol (30 μM) or vehicle (Tris[2-carboxyethyl]phosphine) after pretreatment with the inhibitor for 3 h. Melan-A26–35–specific CTL was added, and cytokine production was measured. (D) Cross-presentation of the long peptide Melan-A15–40A27L was performed at the indicated temperature and evaluated in a degranulation assay performed at 20°C. For all experiments, values are means ± SD of triplicates from a representative experiment repeated at least three times. ND, not detected.

Close modal

The first major conclusion from our work is that long-peptide cross-presentation follows a vacuolar pathway. This conclusion contrasts with a number of previous reports supporting a cytosolic pathway (811). Our proposed model implying a key role of suboptimally loaded HLA-I molecules provides a potential explanation for those discrepancies. Indeed, the support for a cytosolic pathway usually comes from the observation that cross-presentation is impaired by TAP inhibition. This is interpreted to indicate that the cross-presented epitope needs to be transported across membranes, in line with the cytosolic pathway (13). However, if, as we propose, cross-presentation critically depends on suboptimally loaded MHC molecules, TAP inhibition may block cross-presentation indirectly by reducing the availability of suboptimally loaded MHC-I. In fact, HLA-A2 is an exception among MHC-I in the fact that it can load signal peptides in the absence of TAP and therefore increase its pool of suboptimal peptides. Other MHC class I molecules need TAP to load suboptimal peptides. Therefore, in non–HLA-A2–restricted cross-presenting model systems, TAP inhibition may indirectly—and paradoxically—block long-peptide cross-presentation even though it follows a vacuolar pathway.

This alternative interpretation may apply to the study reported by Rosalia et al. (11) who studied cross-presentation of long peptides by mouse and human DC. They studied the role of the proteasome and TAP in murine bone marrow-derived DCs, and observed reduced cross-presentation using epoxomicin (1 μM) or using cells derived from TAP-KO mice. Although it is difficult to compare results obtained in different species using different peptides and DC types, the latter observation might be explained by an indirect effect of TAP inhibition in a vacuolar pathway, as discussed above.

Other reports studied cross-presentation of HLA-A2–restricted peptides similar or identical to the ones we analyzed (811). Faure et al. (10) used proteasome inhibitor lactacystin (40 μM) to evaluate cross-presentation of Melan-A and gp100 long peptides by human Mo-DCs. In line with our results, they observed no inhibition of the cross-presentation of the gp100 peptide. For Melan-A, they observed partial inhibition, with high donor-to-donor variation. They did not evaluate the effect of TAP inhibition. In Segura et al. (9),the same group used lactacystin (2.5 μM) to evaluate cross-presentation of a long Melan-A peptide by various DC, using an IFN-γ production assay. They observed some inhibition, again with donor-to-donor variation, but the IFN-γ signal was weak (∼100 pg/ml), probably at the limit of sensitivity of the IFN-γ ELISA. Again, they did not evaluate the effect of TAP inhibition. If the effects reported by Segura et al. (9) are confirmed, the difference with our data might result from the fact that different DC types were used.

Contrary to these studies, Ménager et al. (8) did evaluate the effect of TAP inhibition on the cross-presentation of a long HLA-A2–restricted Melan-A peptide. They did so using a synthetic peptide corresponding to the N-terminal 35-amino acid residues of TAP inhibitor ICP47. They reported inhibition of cross-presentation using this long ICP47 peptide at 50 or 100 μM. However, this experiment was lacking a control with an irrelevant peptide. We have observed competition effects between long peptides for cross-presentation. Because the TAP inhibitor used by Ménager et al. (8) was, in essence, a long peptide, it is expected to compete with the long peptide for cross-presentation. Therefore, it is likely that the inhibition observed was due to competition rather than TAP inhibition. Ménager et al. (8) also studied the effect of proteasome inhibitor epoxomicin at high doses (1–5 μM), and observed significant inhibition only at 5 μM. We used epoxomicin at 0.5 μM, and provided controls demonstrating complete inhibition of endogenous presentation of gp100 at this dose (Fig. 1). It is known that epoxomicin at high doses induces a number of other effects in the cell, including ER stress. Therefore, the effect observed by Ménager et al. (8) might be unspecific because of the high dose used. In line with this conclusion is the fact that the authors did not observe any modulation of cross-presentation when they changed the proteasomal subunit composition of DCs, whereas the Melan-A antigenic peptide is known to be processed differently by the standard proteasome and the immunoproteasome (21, 4850).

To our knowledge, our study on long-peptide cross-presentation is the first to evaluate both the effect of TAP inhibition and the effect of proteasome inhibition in carefully controlled conditions. Our results clearly indicate a vacuolar pathway of cross-presentation of these long peptides by human monocyte-derived DCs.

The second major conclusion of our work is that long-peptide cross-presentation depends on newly synthesized HLA-I molecules that are loaded with suboptimal peptides. In our view, suboptimal peptides mean peptides that are able to bind HLA-I, but with a low affinity so that they can be readily exchanged. The binding of a low-affinity peptide may be needed to stabilize the MHC-I and allow it to exit to ER and reach the post-ER compartment, where the low-affinity peptide would be exchanged for the cross-presented peptide. The exact nature of the cross-presenting compartment is unclear at this stage and is under study in our laboratory. Candidates include the late endosomal MHC class II compartment, the phagosome and the IRAP-containing vesicles. Another highly relevant question we are currently addressing is to delineate the exact secretion pathway followed by the nascent MHC-I molecules used for cross-presentation. Answering such questions will bring new insights into intracellular trafficking pathways and may influence the development of vaccines based on long peptides to trigger CD8 T cell responses in patients with cancer.

We thank the Centre d’hématologie of the Cliniques universitaires Saint-Luc (Brussels) for providing blood samples from hemochromatosis patients; Aline Depasse for technical help; Aude Bonehill and Carlo Heirman (Vrije Universiteit Brussel, Brussels) for help with mRNA preparation for electroporation; Zhaojun Sun, Didier Colau, and Florence Depontieu for help at various stages of the project; Julie Klein and Mandy Macharis for editorial assistance; and Pierre Coulie for critical reading of the manuscript.

This work was supported by grants from the Ludwig Institute for Cancer Research, the Walloon Region (Programme d’Excellence CIBLES), the Fonds National de la Recherche Scientifique, the Fondation contre le Cancer, the Fonds Maisin, and the Walloon Excellence in Life Sciences and Biotechnology Program. N.V. was a postdoctoral researcher with the Fonds National de la Recherche Scientifique.

The online version of this article contains supplemental material.

Abbreviations used in this article:

DC

dendritic cell

MHC-I

MHC class I

Mo-iDC

monocyte-derived immature DC

siRNA

small interfering RNA

SPP

signal peptide peptidase

TAP

transporter associated with Ag processing.

1
Ackerman
A. L.
,
Cresswell
P.
.
2004
.
Cellular mechanisms governing cross-presentation of exogenous antigens.
Nat. Immunol.
5
:
678
684
.
2
Joffre
O. P.
,
Segura
E.
,
Savina
A.
,
Amigorena
S.
.
2012
.
Cross-presentation by dendritic cells.
Nat. Rev. Immunol.
12
:
557
569
.
3
Wagner
C. S.
,
Grotzke
J. E.
,
Cresswell
P.
.
2012
.
Intracellular events regulating cross-presentation.
Front. Immunol.
3
:
138
.
4
Melief
C. J.
,
van der Burg
S. H.
.
2008
.
Immunotherapy of established (pre)malignant disease by synthetic long peptide vaccines.
Nat. Rev. Cancer
8
:
351
360
.
5
Bijker
M. S.
,
van den Eeden
S. J.
,
Franken
K. L.
,
Melief
C. J.
,
Offringa
R.
,
van der Burg
S. H.
.
2007
.
CD8+ CTL priming by exact peptide epitopes in incomplete Freund’s adjuvant induces a vanishing CTL response, whereas long peptides induce sustained CTL reactivity.
J. Immunol.
179
:
5033
5040
.
6
Vambutas
A.
,
DeVoti
J.
,
Nouri
M.
,
Drijfhout
J. W.
,
Lipford
G. B.
,
Bonagura
V. R.
,
van der Burg
S. H.
,
Melief
C. J. M.
.
2005
.
Therapeutic vaccination with papillomavirus E6 and E7 long peptides results in the control of both established virus-induced lesions and latently infected sites in a pre-clinical cottontail rabbit papillomavirus model.
Vaccine
23
:
5271
5280
.
7
Kenter
G. G.
,
Welters
M. J.
,
Valentijn
A. R.
,
Lowik
M. J.
,
Berends-van der Meer
D. M.
,
Vloon
A. P.
,
Essahsah
F.
,
Fathers
L. M.
,
Offringa
R.
,
Drijfhout
J. W.
, et al
.
2009
.
Vaccination against HPV-16 oncoproteins for vulvar intraepithelial neoplasia.
N. Engl. J. Med.
361
:
1838
1847
.
8
Ménager
J.
,
Ebstein
F.
,
Oger
R.
,
Hulin
P.
,
Nedellec
S.
,
Duverger
E.
,
Lehmann
A.
,
Kloetzel
P. M.
,
Jotereau
F.
,
Guilloux
Y.
.
2014
.
Cross-presentation of synthetic long peptides by human dendritic cells: a process dependent on ERAD component p97/VCP but Not sec61 and/or Derlin-1.
PLoS One
9
:
e89897
.
9
Segura
E.
,
Durand
M.
,
Amigorena
S.
.
2013
.
Similar antigen cross-presentation capacity and phagocytic functions in all freshly isolated human lymphoid organ-resident dendritic cells.
J. Exp. Med.
210
:
1035
1047
.
10
Faure
F.
,
Mantegazza
A.
,
Sadaka
C.
,
Sedlik
C.
,
Jotereau
F.
,
Amigorena
S.
.
2009
.
Long-lasting cross-presentation of tumor antigen in human DC.
Eur. J. Immunol.
39
:
380
390
.
11
Rosalia
R. A.
,
Quakkelaar
E. D.
,
Redeker
A.
,
Khan
S.
,
Camps
M.
,
Drijfhout
J. W.
,
Silva
A. L.
,
Jiskoot
W.
,
van Hall
T.
,
van Veelen
P. A.
, et al
.
2013
.
Dendritic cells process synthetic long peptides better than whole protein, improving antigen presentation and T-cell activation.
Eur. J. Immunol.
43
:
2554
2565
.
12
Cebrian
I.
,
Visentin
G.
,
Blanchard
N.
,
Jouve
M.
,
Bobard
A.
,
Moita
C.
,
Enninga
J.
,
Moita
L. F.
,
Amigorena
S.
,
Savina
A.
.
2011
.
Sec22b regulates phagosomal maturation and antigen crosspresentation by dendritic cells.
Cell
147
:
1355
1368
.
13
Basha
G.
,
Omilusik
K.
,
Chavez-Steenbock
A.
,
Reinicke
A. T.
,
Lack
N.
,
Choi
K. B.
,
Jefferies
W. A.
.
2012
.
A CD74-dependent MHC class I endolysosomal cross-presentation pathway.
Nat. Immunol.
13
:
237
245
.
14
Zehner
M.
,
Marschall
A. L.
,
Bos
E.
,
Schloetel
J. G.
,
Kreer
C.
,
Fehrenschild
D.
,
Limmer
A.
,
Ossendorp
F.
,
Lang
T.
,
Koster
A. J.
, et al
.
2015
.
The translocon protein Sec61 mediates antigen transport from endosomes in the cytosol for cross-presentation to CD8(+) T cells.
Immunity
42
:
850
863
.
15
Basha
G.
,
Lizée
G.
,
Reinicke
A. T.
,
Seipp
R. P.
,
Omilusik
K. D.
,
Jefferies
W. A.
.
2008
.
MHC class I endosomal and lysosomal trafficking coincides with exogenous antigen loading in dendritic cells.
PLoS One
3
:
e3247
.
16
Grommé
M.
,
Uytdehaag
F. G.
,
Janssen
H.
,
Calafat
J.
,
van Binnendijk
R. S.
,
Kenter
M. J.
,
Tulp
A.
,
Verwoerd
D.
,
Neefjes
J.
.
1999
.
Recycling MHC class I molecules and endosomal peptide loading.
Proc. Natl. Acad. Sci. USA
96
:
10326
10331
.
17
Song
R.
,
Harding
C. V.
.
1996
.
Roles of proteasomes, transporter for antigen presentation (TAP), and beta 2-microglobulin in the processing of bacterial or particulate antigens via an alternate class I MHC processing pathway.
J. Immunol.
156
:
4182
4190
.
18
Zou
L.
,
Zhou
J.
,
Zhang
J.
,
Li
J.
,
Liu
N.
,
Chai
L.
,
Li
N.
,
Liu
T.
,
Li
L.
,
Xie
Z.
, et al
.
2009
.
The GTPase Rab3b/3c-positive recycling vesicles are involved in cross-presentation in dendritic cells.
Proc. Natl. Acad. Sci. USA
106
:
15801
15806
.
19
MacAry
P. A.
,
Lindsay
M.
,
Scott
M. A.
,
Craig
J. I.
,
Luzio
J. P.
,
Lehner
P. J.
.
2001
.
Mobilization of MHC class I molecules from late endosomes to the cell surface following activation of CD34-derived human Langerhans cells.
Proc. Natl. Acad. Sci. USA
98
:
3982
3987
.
20
Germeau
C.
,
Ma
W.
,
Schiavetti
F.
,
Lurquin
C.
,
Henry
E.
,
Vigneron
N.
,
Brasseur
F.
,
Lethé
B.
,
De Plaen
E.
,
Velu
T.
, et al
.
2005
.
High frequency of antitumor T cells in the blood of melanoma patients before and after vaccination with tumor antigens.
J. Exp. Med.
201
:
241
248
.
21
Morel
S.
,
Lévy
F.
,
Burlet-Schiltz
O.
,
Brasseur
F.
,
Probst-Kepper
M.
,
Peitrequin
A.-L.
,
Monsarrat
B.
,
Van Velthoven
R.
,
Cerottini
J.-C.
,
Boon
T.
, et al
.
2000
.
Processing of some antigens by the standard proteasome but not by the immunoproteasome results in poor presentation by dendritic cells.
Immunity
12
:
107
117
.
22
Parmentier
N.
,
Stroobant
V.
,
Colau
D.
,
de Diesbach
P.
,
Morel
S.
,
Chapiro
J.
,
van Endert
P.
,
Van den Eynde
B. J.
.
2010
.
Production of an antigenic peptide by insulin-degrading enzyme.
Nat. Immunol.
11
:
449
454
.
23
Berger
G.
,
Durand
S.
,
Goujon
C.
,
Nguyen
X. N.
,
Cordeil
S.
,
Darlix
J. L.
,
Cimarelli
A.
.
2011
.
A simple, versatile and efficient method to genetically modify human monocyte-derived dendritic cells with HIV-1-derived lentiviral vectors.
Nat. Protoc.
6
:
806
816
.
24
Marks
M. S.
,
Blum
J. S.
,
Cresswell
P.
.
1990
.
Invariant chain trimers are sequestered in the rough endoplasmic reticulum in the absence of association with HLA class II antigens.
J. Cell Biol.
111
:
839
855
.
25
Holtkamp
S.
,
Kreiter
S.
,
Selmi
A.
,
Simon
P.
,
Koslowski
M.
,
Huber
C.
,
Türeci
O.
,
Sahin
U.
.
2006
.
Modification of antigen-encoding RNA increases stability, translational efficacy, and T-cell stimulatory capacity of dendritic cells.
Blood
108
:
4009
4017
.
26
Bonehill
A.
,
Heirman
C.
,
Tuyaerts
S.
,
Michiels
A.
,
Breckpot
K.
,
Brasseur
F.
,
Zhang
Y.
,
Van Der Bruggen
P.
,
Thielemans
K.
.
2004
.
Messenger RNA-electroporated dendritic cells presenting MAGE-A3 simultaneously in HLA class I and class II molecules.
J. Immunol.
172
:
6649
6657
.
27
Follenzi
A.
,
Ailles
L. E.
,
Bakovic
S.
,
Geuna
M.
,
Naldini
L.
.
2000
.
Gene transfer by lentiviral vectors is limited by nuclear translocation and rescued by HIV-1 pol sequences.
Nat. Genet.
25
:
217
222
.
28
Shaw-Jackson
C.
,
Michiels
T.
.
1999
.
Absence of internal ribosome entry site-mediated tissue specificity in the translation of a bicistronic transgene.
J. Virol.
73
:
2729
2738
.
29
Salter
R. D.
,
Howell
D. N.
,
Cresswell
P.
.
1985
.
Genes regulating HLA class I antigen expression in T-B lymphoblast hybrids.
Immunogenetics
21
:
235
246
.
30
Baas
E. J.
,
van Santen
H. M.
,
Kleijmeer
M. J.
,
Geuze
H. J.
,
Peters
P. J.
,
Ploegh
H. L.
.
1992
.
Peptide-induced stabilization and intracellular localization of empty HLA class I complexes.
J. Exp. Med.
176
:
147
156
.
31
De Silva
A. D.
,
Boesteanu
A.
,
Song
R.
,
Nagy
N.
,
Harhaj
E.
,
Harding
C. V.
,
Joyce
S.
.
1999
.
Thermolabile H-2Kb molecules expressed by transporter associated with antigen processing-deficient RMA-S cells are occupied by low-affinity peptides.
J. Immunol.
163
:
4413
4420
.
32
Leonhardt
R. M.
,
Keusekotten
K.
,
Bekpen
C.
,
Knittler
M. R.
.
2005
.
Critical role for the tapasin-docking site of TAP2 in the functional integrity of the MHC class I-peptide-loading complex.
J. Immunol.
175
:
5104
5114
.
33
Wei
M. L.
,
Cresswell
P.
.
1992
.
HLA-A2 molecules in an antigen-processing mutant cell contain signal sequence-derived peptides.
Nature
356
:
443
446
.
34
Gaugler
B.
,
Van den Eynde
B.
,
van der Bruggen
P.
,
Romero
P.
,
Gaforio
J. J.
,
De Plaen
E.
,
Lethé
B.
,
Brasseur
F.
,
Boon
T.
.
1994
.
Human gene MAGE-3 codes for an antigen recognized on a melanoma by autologous cytolytic T lymphocytes.
J. Exp. Med.
179
:
921
930
.
35
Hammer
G. E.
,
Gonzalez
F.
,
Champsaur
M.
,
Cado
D.
,
Shastri
N.
.
2006
.
The aminopeptidase ERAAP shapes the peptide repertoire displayed by major histocompatibility complex class I molecules.
Nat. Immunol.
7
:
103
112
.
36
Bacik
I.
,
Cox
J. H.
,
Anderson
R.
,
Yewdell
J. W.
,
Bennink
J. R.
.
1994
.
TAP (transporter associated with antigen processing)-independent presentation of endogenously synthesized peptides is enhanced by endoplasmic reticulum insertion sequences located at the amino- but not carboxyl-terminus of the peptide.
J. Immunol.
152
:
381
387
.
37
Weinzierl
A. O.
,
Rudolf
D.
,
Hillen
N.
,
Tenzer
S.
,
van Endert
P.
,
Schild
H.
,
Rammensee
H. G.
,
Stevanović
S.
.
2008
.
Features of TAP-independent MHC class I ligands revealed by quantitative mass spectrometry.
Eur. J. Immunol.
38
:
1503
1510
.
38
Baravalle
G.
,
Schober
D.
,
Huber
M.
,
Bayer
N.
,
Murphy
R. F.
,
Fuchs
R.
.
2005
.
Transferrin recycling and dextran transport to lysosomes is differentially affected by bafilomycin, nocodazole, and low temperature.
Cell Tissue Res.
320
:
99
113
.
39
Knorr
R.
,
Karacsonyi
C.
,
Lindner
R.
.
2009
.
Endocytosis of MHC molecules by distinct membrane rafts.
J. Cell Sci.
122
:
1584
1594
.
40
Kirchhausen
T.
2000
.
Clathrin.
Annu. Rev. Biochem.
69
:
699
727
.
41
Tomoda
H.
,
Kishimoto
Y.
,
Lee
Y. C.
.
1989
.
Temperature effect on endocytosis and exocytosis by rabbit alveolar macrophages.
J. Biol. Chem.
264
:
15445
15450
.
42
Allaire
P. D.
,
Marat
A. L.
,
Dall’Armi
C.
,
Di Paolo
G.
,
McPherson
P. S.
,
Ritter
B.
.
2010
.
The Connecdenn DENN domain: a GEF for Rab35 mediating cargo-specific exit from early endosomes.
Mol. Cell
37
:
370
382
.
43
Kouranti
I.
,
Sachse
M.
,
Arouche
N.
,
Goud
B.
,
Echard
A.
.
2006
.
Rab35 regulates an endocytic recycling pathway essential for the terminal steps of cytokinesis.
Curr. Biol.
16
:
1719
1725
.
44
Brand
M.
,
Jansen
E.
,
Ploegh
H. L.
.
1985
.
Effect of reduced temperature on glycoprotein (Ig, HLA) processing and transport in lymphoid cells.
Mol. Immunol.
22
:
787
794
.
45
Braulke
T.
,
Hasilik
A.
,
von Figura
K.
.
1988
.
Low temperature blocks transport and sorting of cathepsin D in fibroblasts.
Biol. Chem. Hoppe Seyler
369
:
441
449
.
46
Rivoltini
L.
,
Squarcina
P.
,
Loftus
D. J.
,
Castelli
C.
,
Tarsini
P.
,
Mazzocchi
A.
,
Rini
F.
,
Viggiano
V.
,
Belli
F.
,
Parmiani
G.
.
1999
.
A superagonist variant of peptide MART1/Melan A27-35 elicits anti-melanoma CD8+ T cells with enhanced functional characteristics: implication for more effective immunotherapy.
Cancer Res.
59
:
301
306
.
47
Valmori
D.
,
Fonteneau
J. F.
,
Valitutti
S.
,
Gervois
N.
,
Dunbar
R.
,
Liénard
D.
,
Rimoldi
D.
,
Cerundolo
V.
,
Jotereau
F.
,
Cerottini
J. C.
, et al
.
1999
.
Optimal activation of tumor-reactive T cells by selected antigenic peptide analogues.
Int. Immunol.
11
:
1971
1980
.
48
Chapatte
L.
,
Ayyoub
M.
,
Morel
S.
,
Peitrequin
A. L.
,
Lévy
N.
,
Servis
C.
,
Van den Eynde
B. J.
,
Valmori
D.
,
Lévy
F.
.
2006
.
Processing of tumor-associated antigen by the proteasomes of dendritic cells controls in vivo T-cell responses.
Cancer Res.
66
:
5461
5468
.
49
Dannull
J.
,
Lesher
D. T.
,
Holzknecht
R.
,
Qi
W.
,
Hanna
G.
,
Seigler
H.
,
Tyler
D. S.
,
Pruitt
S. K.
.
2007
.
Immunoproteasome down-modulation enhances the ability of dendritic cells to stimulate antitumor immunity.
Blood
110
:
4341
4350
.
50
Guillaume
B.
,
Stroobant
V.
,
Bousquet-Dubouch
M. P.
,
Colau
D.
,
Chapiro
J.
,
Parmentier
N.
,
Dalet
A.
,
Van den Eynde
B. J.
.
2012
.
Analysis of the processing of seven human tumor antigens by intermediate proteasomes.
J. Immunol.
189
:
3538
3547
.

The authors have no financial conflicts of interest.

Supplementary data