• Aucun résultat trouvé

Pleiotropic effects of post-translational modifications on the fate of viral glycopeptides as cytotoxic T cell epitopes.

N/A
N/A
Protected

Academic year: 2021

Partager "Pleiotropic effects of post-translational modifications on the fate of viral glycopeptides as cytotoxic T cell epitopes."

Copied!
31
0
0

Texte intégral

(1)

HAL Id: hal-00092646

https://hal.archives-ouvertes.fr/hal-00092646

Submitted on 22 Mar 2021

HAL is a multi-disciplinary open access archive for the deposit and dissemination of sci-entific research documents, whether they are pub-lished or not. The documents may come from teaching and research institutions in France or abroad, or from public or private research centers.

L’archive ouverte pluridisciplinaire HAL, est destinée au dépôt et à la diffusion de documents scientifiques de niveau recherche, publiés ou non, émanant des établissements d’enseignement et de recherche français ou étrangers, des laboratoires publics ou privés.

epitopes.

Denis Hudrisier, Joelle Riond, Honoré Mazarguil, Jean Edouard Gairin

To cite this version:

Denis Hudrisier, Joelle Riond, Honoré Mazarguil, Jean Edouard Gairin. Pleiotropic effects of post-translational modifications on the fate of viral glycopeptides as cytotoxic T cell epitopes.. Journal of Virology, American Society for Microbiology, 2001, 276(41), pp.38255-38260. �hal-00092646�

(2)

Pleiotropic Effects of Post-translational Modifications on the Fate of Viral Glycopeptides as Cytotoxic T Cell Epitopes.

Denis Hudrisier†,*, Joëlle Riond† , Honoré Mazarguil† and Jean Edouard Gairin†

Institut de Pharmacologie et de Biologie Structurale, UMR5089 CNRS/Université Paul Sabatier,

205 route de Narbonne, 31400 Toulouse – France.

*Institut National de la Santé et de la Recherche Médicale, U395, CHU Purpan, BP3028, 31024

Toulouse Cedex 03 – France.

Corresponding author: Dr. Jean Edouard Gairin

Laboratoire d’ImmunoPharmacologie Structurale, Institut de Pharmacologie et de Biologie Structurale, 205 route de Narbonne, 31400 Toulouse – France. tel.: +33 (0)561 175 530,

fax: +33 (0)561 175 532, e-mail: gairin@ipbs.fr

Running title: Fate of viral glycopeptides as CTL epitopes

Abbreviations: LCMV, lymphocytic choriomeningitis virus; ER, endoplasmic reticulum; GlcNac, N-acetyl-D-glucosamine

(3)

SUMMARY

The fate of viral glycopeptides as cytotoxic T lymphocytes (CTL) epitopes is unclear. We have dissected the mechanisms of antigen presentation and CTL recognition of the peptide GP392-400 (WLVTNGSYL) from the lymphocytic choriomeningitis virus (LCMV) and compared them with those of the previously reported GP92-101 antigen (CSANNSHHYI). Both GP392-400 and GP92-101 bear a glycosylation motif, are naturally N-glycosylated in the mature viral glycoproteins, bind to major histocompatibility complex H-2Db molecules and are immunogenic. However, post-translational modifications differentially affected GP92-101 and GP392-400. Upon N-glycosylation or de-N-glycosylation, a marked decrease in MHC binding was observed for GP392-400 but not for GP92-101. Further, under its N-glycosylated or de-N-N-glycosylated form, GP392-400 then lost its initial ability to generate a CTL response in mice whereas GP92-101 was still immunogenic under the same conditions. The genetically encoded form of GP392-400, which, on the basis of its immunogenicity, could still be presented with H-2Db during the course of LCMV infection does not in fact

appear at the surface of LCMV-infected cells. Our results show that post-translational modifications of viral glycopeptides can have pleiotropic effects on their presentation to and recognition by CTL that contribute to either creation of neo-epitopes or destruction of potential epitopes.

(4)

INTRODUCTION

Viral peptides presented by major histocompatibility complex (MHC) class I molecules at the surface of infected cells to CTL are key molecular signals allowing recognition of the infected cells and their subsequent destruction by the activated CTL. Those peptides commonly consist of octameric to undecameric antigenic peptides generated from viral proteins by a multistep, intracellular processing pathway involving components present in the cytoplasm and the endoplasmic reticulum (ER). Peptides generated in the cytosol from the degradation of ubiquitinated proteins by the proteasome and possibly by non proteasomal cytosolic proteases (1,2), are then translocated via peptide-specific transporters (TAP) into the ER where they assemble with the MHC class I molecules and are eventually subjected to further trimming (3,4). The capacity or contrastingly the inability of a given peptide to proceed through these steps has been shown to determine its presentation by MHC molecules (5-7). In contrast to the well studied processing pathway of peptides derived from cytosolic proteins, very little is known about the ability of glycopeptides originating from viral glycoproteins to successfully pass through the above mentioned steps. For example based on the use of model peptides chemically glycosylated (8-12) or affected by other post-translational modifications such as reduction of sulfhydryl groups (13), bond rearrangement (14) or phosphorylation (15), it has been proposed that these modifications could generate new epitopes and thus increase antigenic diversity (10,11,16-19). However given the failure to identify naturally processed N-glycopeptides as epitopes to date, the fate of viral glycopeptides as MHC class I-restricted antigens remains very unclear.

Lymphocytic choriomeningitis virus (LCMV) infection of H-2b mice is cleared by CTL that

(5)

(16,20,25,26). The presence of glycopeptides derived from the LCMV glycoproteins bearing the

H-2Db binding motif makes LCMV infection a model of choice to study the fate of glycopeptides

as CTL epitopes. We recently showed that the LCMV GP92-101 subdominant epitope (CSANNSHHYI), which bears the glycosylation motif NNS within its sequence, was

post-translationally modified in infected cells and presented at the cell surface by H-2Db molecules in

two different forms, a non-glycosylated form (CSANNSHHYI) and a de-N-glycosylated form (CSADNSHHYI) resulting from the N-glycanase mediated de-N-glycosylation of a previously N-glycosylated peptide sequence (16).

An additional H-2Db-restricted glycopeptide GP392-400 (WLVTNGSYL) was previously

identified in the LCMV GP2 protein (25). It shares similar biochemical properties with GP92-101. Like GP92-101, GP392-400 i) bears a NXS glycosylation motif, ii) is N-glycosylated in the

mature LCMV GP2 glycoprotein (27), and iii) binds efficiently to H-2Db (25,28). In contrast to

GP92-101, no CTL response against GP392-400 has been detected (28) and its cellular status remains unknown. The contrasting immunogenic properties of GP92-101 and GP392-400 suggest that the common biochemical events or intracellular mechanisms encountered by these two viral glycopeptides along the processing and presentation pathway lead to different fates as MHC class I-restricted CTL epitopes. This would not have been predicted on the basis of their biochemical properties. The aim of our study was therefore to explore and dissect the intracellular molecular mechanisms that may differentially affect the processing pathways of these two viral glycopeptides and explain their different antigenic properties.

(6)

EXPERIMENTAL PROCEDURES

CTL, cell lines and virus. CTL were obtained from C57BL/6 mice immunized s.c. at the base of

the tail with 50-100 µg of the indicated peptides mixed with 5 µg of P30 T-helper epitope from

tetanus toxoid in incomplete Freund’s adjuvant. After one week, draining lymph nodes were removed and CTL were restimulated at weekly intervals with irradiated (2,500 rads) C57BL/6

splenocytes and irradiated (10,000 rads) peptide-pulsed (1 µM) RMA cells in the presence of 30

UI/ml IL-2 (EL4 supernatant). Murine H-2b mutant RMA-S cells were used in binding experiments. The murine H-2b cell lines RMA and MC57 were used in in vitro cytotoxicity assays. Cells were grown in DMEM (RMA, RMA-S, MC57) containing 5% bovine serum, L-glutamine (2 mM) and antibiotics (Penicillin, 10 units/ml and Streptomycin, 10 µg/ml). The LCMV Armstrong strain (LCMV Arm) was used to infect mice or cells.

Peptide synthesis. Peptides were synthesized by the solid-phase method using Fmoc chemistry.

For Asn-glycosylated peptides, Fmoc-Asn(Ac3AcNH-ßGlc)-OH, a commercially available derivative of Asn bearing an N-acetyl-D-glucosamine moiety (GlcNac, Novabiochem) was used. After standard protocols for solid phase synthesis, cleavage and deprotection, the glycopeptide was de-O-acetylated with 0.1 M sodium hydroxide as previously described (16). Peptides were purified by HPLC on a RP300-C8 reversed-phase column (Brownlee Lab) and their identity confirmed by electrospray ionization mass spectrometry.

Cytotoxicity assays. RMA or MC57 cells were incubated for 1 h at 37°C in medium containing

(7)

prior to the assay. RMA (peptide-pulsed or not) or MC57 (LCMV-infected or not) cells were 51Cr

labeled and used as targets (5 x 103 / well) in chromium release assays. CTL (1.5 x 104 / well or

at the indicated E/T ratio) were added and after 4h incubation at 37°C, the 51Cr content of

supernatants was determined. The specific lysis was calculated as 100 x [(experimental - spontaneous release)/(total - spontaneous release)].

Binding studies. Binding studies were performed as previously described (25,29). Briefly,

RMA-S cells (5 x 105 cells/well) previously incubated at 26°C for 40 h were placed in U-bottomed

96-well plates for 4 h at 37°C with increasing concentrations (0 to 10-4 M) of unlabeled peptides.

Cells were then washed twice with BSA-PBS and incubated at 4°C for 45 min. with the 28-14-8S

anti-H-2Db monoclonal antibody (hybridoma supernatant) (30). After two washes in BSA-PBS,

an anti-mouse IgG antibody conjugated to FITC (Sigma) was added for 45 min. then cells were washed twice in BSA-PBS and analyzed by flow cytometry.

Peptide Extraction from LCMV-infected cells. Peptides were extracted from the surface of

LCMV-infected MC57 (H-2b) cells as previously described (16). Briefly, cells (1 - 2 x 109) were

washed in PBS, then resuspended in 0.1 M citrate/phosphate buffer at pH 3.0 for 2 mins. The eluted material was desalted on a SepPak column, transferred onto a Centricon 10 and centrifuged at 3500 rpm for 30 min at 4°C. Material less than 10 kDa was vacuum concentrated then resuspended in 20 µl of 0.08% trifluoroacetic acid (TFA). Peptides were separated by HPLC (Waters 600S controller system) on a reversed-phase C18 column (7 µm, 2.1x100 mm,

Aquapore, BrownleeTM) according to the following procedure: solution A, 0.08% TFA in H2O ;

(8)

Fractions (200 µl) were collected in U-bottomed 96-well plates, lyophilized, reconstituted in PBS and stored at -80 °C before analysis.

Molecular modeling. The three-dimensional models for LCMV GP92-101 (CSANNSHHYI) and

LCMV GP392-400 (WLVTNGSYL) interacting with H-2Db have been produced using the online

modeling facility SwissModel (GlaxoWellcome) at the following address http://www.expasy.ch/spdbv/.

(9)

RESULTS

The genetically encoded form of LCMV GP392-400 but neither its N-glycosylated nor de-N-glycosylated forms can generate a CTL response in H-2b mice. Peptides bearing a

N-glycosylation motif NXS/T can theoretically exist in three different forms: a non-glycosylated one bearing an unmodified NXS/T sequence, a N-glycosylated one in which a glycan moiety is attached to N and a de-N-glycosylated one resulting in the conversion of N to D. In order to analyse their fate as CTL epitopes, we synthesized peptides corresponding to the non-glycosylated (genetically encoded), N-non-glycosylated and de-N-non-glycosylated forms of LCMV GP392-400 (named GP392-400, [GlcNac-N396]-GP392-400 and [D396]-GP392-400 respectively), and of LCMV GP92-101 (named GP92-101, [GlcNac-N95]-GP92-101 and [D95]-GP92-101 respectively). The N-acetyl-D-glucosamine moiety was chosen to mimic the glycosylated form because it is the first carbohydrate attached to N and common to all eukaryotic glycosylation processes. Previous studies have failed to reveal a response to LCMV GP392-400 following LCMV infection of mice (28) but in these studies the fact that GP392-400 bears a N-glycan in the mature viral protein was not taken into account. Our first aim was therefore to analyze the ability of the different unmodified (GP392-400) and post-translationally modified ([GlcNac-N396]-GP392-400, [D396]-GP392-400) forms of LCMV GP392-400 to generate a CTL response upon immunization. After several rounds of in vitro restimulation, CTL from peptide-immunized B6 mice were obtained against the GP392-400 form but not against either the [GlcNac-N396]-GP392-400 or [D396]-GP392-400 forms. This indicated that only GP392-400 was immunogenic, a result that contrasted with those obtained with LCMV GP92-101, where all three forms (GP92-101, [GlcNac-N95]-GP92-101 and [D95]-GP92-101) were found to be immunogenic under the same experimental conditions (16). As shown in Figure 1,

(10)

anti-GP392-400 CTL specifically recognized the H-2b RMA (panel A) or MC57 (panel B) cells pulsed with GP392-400, in an effector:target (E:T) ratio-dependent manner. Unpulsed targets were not lysed (RMA) or were lysed weakly at the highest E:T ratios (MC57).

A CTL clone was then derived from the LCMV GP392-400-specific CTL line by limiting dilution. We tested the ability of these CTL to kill target cells coated with the immunizing peptide GP392-400 or its post-translationally modified forms [GlcNac-N396]-GP392-400 or [D396]-GP392-400. Both the CTL line (Figure 2A) and the CTL clone (Figure 2B) recognized and lysed GP392-400-pulsed target cells with comparable efficacy (half maximal lysis in the pM range). In contrast, three to four-log higher concentrations of [GlcNac-N396]-GP392-400 or of

[D396]-GP392-400 (half maximal lysis in the 10-9-10-8 M range) were required to obtain the

same lysis by both the CTL line and the clone. In a comparative analysis shown in Figure 2C, LCMV GP92-101-specific CTL generated against the GP92-101 peptide recognized the [D95]-GP92-101 form more efficiently than the [D95]-GP92-101 form used for the immunization (half

maximal lysis at 10-12 M and 10-10 M respectively). Higher concentrations of

[GlcNac-N95]-GP92-101 (half maximal lysis in the 10-8 M range, comparable to that of

[GlcNac-N396]-GP392-400 by anti-GP392-[GlcNac-N396]-GP392-400 specific CTL) were necessary to sensitize target cells to CTL killing, confirming previous results (16).

Post-translational modifications (N-glycosylation or de-N-glycosylation) differentially affect the binding of GP392-400 and of GP92-101 to H-2Db. As one of the factors determining the

immunogenicity of peptides is their ability to be presented by MHC class-I molecules (31,32), we hypothesized that post-translational modifications could differentially affect the MHC class I presentation and could result in different immunogenic properties of the LCMV viral

(11)

glycopeptides. Peptide binding to H-2Db was measured in a previously described MHC stabilization assay on RMA-S cells (25,29). As shown in Figure 3A, GP392-400 bound

efficiently to H-2Db (half maximal binding in the 0.1-0.2 µM range), in agreement with previous

observations (25). In contrast binding of the two modified analogues to H-2Db was strongly

reduced: [D396]-GP392-400 showed a 2 log decrease in H-2Db binding properties (half maximal

binding at 10 µM). Half maximal binding was even not measurable for

[GlcNac-N396]-GP392-400 (only ~25% maximal binding was observed at the highest concentration tested (10-4 M)).

Deficiency in MHC binding of these two peptides is therefore likely one of the reasons which may explain their lack of immunogenicity. A different situation was observed with the LCMV

GP92-101 subdominant antigen for which the three forms bound efficiently to H-2Db (Figure

3B). GP92-101 and [D95]-GP92-101 showed superimposed curves (half maximal binding < 0.1 µM) whereas the [GlcNac-N395]-GP92-101 form exhibited a 1 log higher binding (half maximal binding < 0.01 µM). The differences in the efficiency with which they are recognized by CTL may thus likely reflect differences in TCR recognition rather than MHC binding (16).

LCMV-infected MC57 cells are not lysed by LCMV GP392-400- specific CTL. It can be

deduced from the experiments presented above that only the N396 form of GP392-400 is likely to

be presented by H-2Db at the surface of infected cells while all three forms of GP92-101 could be

presented. The cytolytic activity of the anti-GP392-400 CTL was thus tested against LCMV-infected or unLCMV-infected MC57 target cells. Figure 4 shows that neither the CTL line (panel A) nor the CTL clone (panel B) generated against GP392-400 were able to kill LCMV-infected cells, even at the highest E:T ratio tested. In comparison (panel C), efficient killing was observed when the CTL line generated against the unmodified form of the subdominant LCMV epitope

(12)

GP92-101 was used, in accord with a previous study (16). Further, when HPLC-fractionated material eluted from the surface of LCMV-infected MC57 cells was used to pulse uninfected targets, no lysis by anti-GP392-400 CTL was observed (Figure 5B). This observation contrasted to that made with cells coated with mock-extracted synthetic GP392-400 peptide (Figure 5A) and results obtained with anti-GP92-101 CTL directed against cells pulsed with the same pool of peptides extracted from LCMV-infected MC57 cells (16). Since a very low number of copies (100 or less) of an antigenic peptide present at the cell surface is sufficient to trigger CTL lysis, we can conclude from these results that GP392-400 is not naturally presented at the surface of LCMV-infected cells. This observation contrasts with that made with LCMV GP92-101 for which we found that both the genetically encoded GP92-101 and post-translationally modified [D95]-GP92-101 forms were co-presented to CTL at the surface of LCMV-infected cells while the N-glycosylated form [GlcNac-N95]-GP92-101 was absent (16).

(13)

DISCUSSION

In this study, by analyzing two H-2Db-restricted LCMV glycopeptides, GP392-400 and

GP92-101 (25,26), we showed that post-translational modifications of MHC class I-restricted viral peptides that bear a glycosylation motif -NXS- can have pleiotropic effects on their presentation by MHC and CTL recognition, depending on the peptide sequence (Table 1). We previously reported that synthetic peptides corresponding to the unmodified (genetically encoded), N-glycosylated or de-N-N-glycosylated analogs of GP92-101 share the ability to bind strongly to

H-2Db, are both immunogenic and are naturally processed and presented at the surface of infected

cells (16). We now show that post-translational modifications of so called “high affinity peptides” do not always have a positive effect on the diversity of antigen presentation. Results presented in this paper show that N-glycosylation or de-N-glycosylation of GP392-400 led to peptides with reduced MHC binding properties at physiological concentrations while unmodified GP392-400 efficiently bound to this molecule. This may be one of the reasons why only the unmodified form of GP392-400 is immunogenic in C56BL/6 mice and could generate epitope-specific CTL that efficiently lysed peptide-pulsed target cells. However GP392-400-epitope-specific CTL

did not recognise LCMV-infected cells, showing that GP392-400 is not presented by H-2Db in a

natural situation. The lack of presentation of the [GlcNac-N396]-GP392-400 and

[D396]-GP392-400 forms can be easily explained by our observation that those peptides are poor H-2Db binders

and show no immunogenicity. Indeed above a certain threshold of binding to the MHC, the immunogenicity of peptides rapidly drops, preventing these peptides from acting as T cell epitopes (33). That LCMV-infected cells were not recognized by anti-GP392-400 CTL tells us that not only [GlcNac-N396]-GP392-400 and [D396]-GP392-400 but also GP392-400, despite its

(14)

Why is the genetically encoded form of GP92-101 but not that of GP392-400 naturally presented at the surface of infected cells while both GP92-101 and GP392-400 are known to be N-glycosylated in their respective proteins GP1 and GP2 (27)? One explanation is that some of the glycosylation sites of LCMV GP1 but none in GP2 may be statistically left unglycosylated within the viral glycoprotein. Conversely it is conceivable that some GP1 but not GP2 is aberrantly translated on free ribosomes in the cytosol (34). Whatever the explanation, GP2 protein containing N at position 396 would require further processing mediated by the proteasome before being presented by MHC class I molecules. Interestingly we observed that purified proteasomes were unable to cleave the GP392-400 precursor at the correct C-terminus of the potential CTL epitope, therefore suggesting that even though position 396 was left unmodified no production of GP392-400 would occur (S. Claverol, B. Monsarrat and J.E. Gairin, unpublished observations). The inability of the proteasome to generate an antigenic peptide or a precursor extended by a few a.a. at its N-terminus has been shown to represent one of the main blocks in antigen presentation (5-7).

What may explain the differences between GP92-101 and GP392-400 with regard to the effect of post-translational modifications? In terms of MHC presentation, the loss of MHC binding capacity of the [GlcNac-N396]-GP392-400 and [D396]-GP392-400 forms of GP392-400 is likely to be due to the role played by N396 as a MHC anchor (N at position 5 of the sequence for

H-2Db). Indeed we and others have previously shown that one important property of MHC anchor

residues is their limited or null tolerance to substitution (29,35,36). However in some instances glycosylation of residues at anchor position has been shown to preserve the ability of the modified peptide to bind to MHC molecules and to be recognized by CTL (9,11). In the case of GP92-101, this peptide contains two adjacent N residues. It is likely that N96 and not N95 serves

(15)

that shifted N95 to position 5 of the sequence resulted in a marked loss of H-2Db binding (25) as

would be expected if N96 is the anchor residue (an increase in H-2Db binding would have been

expected if N95 played this role), ii) the high H-2Db binding properties of GP92-101 was left

unaffected by either N-glycosylation or de-N-glycosylation that in turn dramatically altered TCR

recognition, consistent with previous data showing that position 4 of H-2Db-restricted peptides

tolerates a wide variety of substitutions and is directed toward the TCR. This is well supported by

3D structure and molecular models of GP392-H-2Db and GP92-H-2Db complexes, which clearly

place position 4 and position 5 as TCR contact and MHC anchor residues respectively (see Figure 6 for illustration). Also the data presented in Figures 2 and 3 show that the CTL recognition properties of GP392-400, [GlcNac-N396]-GP392-400 and [D396]-GP392-400 forms of GP392-400 were directly correlated to their MHC binding properties while the CTL recognition of GP92-101, [GlcNac-N95]-GP92-101 and [D95]-GP92-101 were not. Therefore, the beneficial or detrimental effects of post-translational modification of peptides on T cell activation will greatly depend on the nature and location of the acceptor site within the peptide sequence [(9,11,16) and this report]. If beneficial, these modifications may be seen as an advantage to the host in its fight against viral infection by creating neo-epitopes (16). Conversely, if detrimental, they may be considered as a means for a virus to achieve CTL escape by destroying potential immunogenic sequences (37,38).

It is interest to note that while artificially N-glycosylated model peptides can efficiently bind classical MHC class I molecules and be recognized by CTL (8-12) no such naturally processed N-glycopeptides have yet been identified. However natural peptides bearing a N-glycosylation motif appeared, as presented by classical MHC class I molecules, at the surface of target cells in their non-glycosylated or de-N-glycosylated forms (16-18). In contrast several N-glycopeptides were found to be naturally presented by MHC class II or non classical MHC class I (39-49). In

(16)

addition, experimental evidence exists that O-glycosylated peptides (50) or peptides bearing other post-translational modifications such as cysteinylation (51-53) or phosphorylation (19) can be naturally presented by classical MHC class I. It is possible that N-glycopeptides could not be naturally presented as a result of constraints imposed by the processing pathway (54,55). Altogether key parameters that contribute to the creation or conversely to the disruption of potent antiviral epitopes such as the ones presented here must be taken into account in approaches to predicting viral or tumor antigens of therapeutic interest.

ACKNOWLEDGMENTS

We thank S. Claverol and B. Monsarrat for sharing unpublished observations, P. Borrow for helpful discussions and critical reading of the manuscript, Y. Barascud and C. Grégoire for help in MHC binding assays. This work was supported in part by grants from the Centre National de la Recherche Scientifique, the Association pour la Recherche sur le Cancer (contract n° 5485). D.H. was a recipient of a post-doctoral fellowship from the Association pour la Recherche sur le Cancer.

(17)

REFERENCES

1. Nandi, D., Marusina, K., and Monaco, J. (1998) Curr. Top. Microbiol. Immunol. 232,

15-48

2. Groettrup, M., Soza, A., Kuckelhorn, U., and Kloetzel, P.-M. (1996) Immunol. Today 17,

429-435

3. Elliott, T., Willis, A., Cerundolo, W., and Townsend, A. (1995) J. Exp. Med. 181,

1481-1491

4. Snyder, H. L., Yewdell, J. W., and Bennink, J. R. (1994) J. Exp. Med. 180(6), 2389-2394

5. Luckey, C. J., King, G. M., Marto, J. A., Venketeswaran, S., Maier, B. F., Crotzer, V. L.,

Colella, T. A., Shabanowitz, J., Hunt, D. F., and Engelhard, V. H. (1998) J Immunol

161(1), 112-21.

6. Morel, S., Levy, F., Burlet-Schiltz, O., Brasseur, F., Probst-Kepper, M., Peitrequin, A. L.,

Monsarrat, B., Van Velthoven, R., Cerottini, J. C., Boon, T., Gairin, J. E., and Van den Eynde, B. J. (2000) Immunity 12(1), 107-17

7. Valmori, D., Gileadi, U., Servis, C., Dunbar, P. R., Cerottini, J. C., Romero, P.,

Cerundolo, V., and Levy, F. (1999) J Exp Med 189(6), 895-906

8. Speir, J. A., Abdel-Motal, U. M., Jondal, M., and Wilson, I. A. (1999) Immunity 10(1),

51-61.

9. Glithero, A., Tormo, J., Haurum, J. S., Arsequell, G., Valencia, G., Edwards, J., Springer,

S., Townsend, A., Pao, Y. L., Wormald, M., Dwek, R. A., Jones, E. Y., and Elliott, T. (1999) Immunity 10(1), 63-74.

10. Haurum, J. S., Arsequell, G., Lellouch, A. C., Wong, S. Y., Dwek, R. A., McMichael, A.

(18)

11. Haurum, J. S., Tan, L., Arsequell, G., Frodsham, P., Lellouch, A. C., Moss, P. A., Dwek, R. A., McMichael, A. J., and Elliott, T. (1995) Eur J Immunol 25(12), 3270-6.

12. Abdel-Motal, U., Berg, L., Rosen, A., Bengtsson, M., Thorpe, C., Kihlberg, J., Dahmen,

J., Magnusson, G., Karlsson, K., and Jondal, M. (1996) Eur. J. immunol. 26, 544-551

13. Chen, W., Yewdell, J. W., Levine, R. L., and Bennink, J. R. (1999) J Exp Med 189(11),

1757-64.

14. Chen, W., Ede, N. J., Jackson, D. C., McCluskey, J., and Purcell, A. W. (1996) J.

Immunol. 157, 1000-1005

15. Andersen, M. H., Bonfill, J. E., Neisig, A., Arsequell, G., Sondergaard, I., Valencia, G.,

Neefjes, J., Zeuthen, J., Elliott, T., and Haurum, J. S. (1999) J Immunol 163(7), 3812-8.

16. Hudrisier, D., Riond, J., Mazarguil, H., Oldstone, M. B. A., and Gairin, J. E. (1999) J.

Biol. Chem. 274, 36274-36280

17. Ferris, R. L., Buck, C., Hammond, S. A., Woods, A. S., Cotter, R. J., Takiguchi, M.,

Igarashi, Y., Ichikawa, Y., and Siliciano, R. F. (1996) J. Immunol. 156, 834-840

18. Skipper, J. C. A., Hendrickson, R. C., Gulden, P. H., Brichard, V., VanPel, A., Chen, Y.,

Shabanowitz, J., Wolfel, T., Slinguff, C. L. J., Boon, T., Hunt, D. F., and Engelhard, V. H. (1996) J. Exp. Med. 183, 527-534

19. Zarling, A., Ficarro, S., White, F., Shabanowitz, J., Hunt, D., and Engelhard, V. (2000) J.

Exp. Med. 192, 1755-1762

20. Murali-Krishna, K., Altman, J. D., Suresh, M., Sourdive, D. J., Zajac, A. J., Miller, J. D.,

Slansky, J., and Ahmed, R. (1998) Immunity 8(2), 177-87

21. Schulz, M., Aichele, P., Wollenweider, M., Bobe, F. W., Cardinaux, F., Hengartner, H.,

(19)

22. Gairin, J. E., Mazarguil, H., Hudrisier, D., and Oldstone, M. B. A. (1995) J. Virol. 69(4), 2297-2305

23. Klavinskis, L. S., Whitton, J. L., Joly, E., and Oldstone, M. B. (1990) Virology 178(2),

393-400

24. Oldstone, M. B. A., Whitton, J. L., Lewicki, H., and Tishon, A. (1988) J. Exp. Med. 168,

559-570

25. Hudrisier, D., Mazarguil, H., Laval, F., Oldstone, M. B. A., and Gairin, J. E. (1996) J.

Biol. Chem. 271, 17829-17836

26. Vandermost, R., Muralikrishna, K., Whitton, J., Oseroff, C., Alexander, J., Southwood,

S., Sidney, J., Chesnut, R., Sette, A., and Ahmed, R. (1998) Virology 240, 158-167

27. Wright, K. E., Spiro, R. C., Burns, J. W., and Buchmeier, M. J. (1990) Virology 177(1),

175-83

28. Oldstone, M. B. A., Lewicki, H., Borrow, P., Hudrisier, D., and Gairin, J. E. (1995) J.

Virol. 69, 7423-7429

29. Hudrisier, D., Mazarguil, H., Oldstone, M. B. A., and Gairin, J. E. (1995) Mol. Immunol.

32, 895-907

30. Ozato, K., Hansen, T. H., and Sachs, D. H. (1980) J.Immunol. 125, 2473-2477

31. vandenBurg, S. H., Visseren, M. J. W., Brandt, R. M. P., Kast, W. M., and Melief, C. J.

M. (1996) J. Immunol. 156, 3308-3314

32. Hudrisier, D., Riond, J., and Gairin, J. (2001) J. Virol. 75, 2468-2471

33. Barber, L. D., and Parham, P. (1994) J. Exp. Med. 180(4), 1191-1194

34. Yewdell, J. W., Anton, L. C., and Bennink, J. R. (1996) J. Immunol. 157(5), 1823-1826

(20)

36. Saito, Y., Peterson, P. A., and Matsumura, M. (1993) J. Biol. Chem. 268(28), 21309-21317

37. Theobald, M., Ruppert, T., Kuckelkorn, U., Hernandez, J., Haussler, A., Ferreira, E. A.,

Liewer, U., Biggs, J., Levine, A. J., Huber, C., Koszinowski, U. H., Kloetzel, P. M., and Sherman, L. A. (1998) J Exp Med 188(6), 1017-28.

38. Ossendorp, F., Eggers, M., Neisig, A., Ruppert, T., Groettrup, M., Sijts, A., Mengede, E.,

Kloetzel, P. M., Neefjes, J., Koszinowski, U., and Melief, C. (1996) Immunity 5(2), 115-124

39. Harding, C. V., Kihlberg, J., Elofsson, M., Magnusson, G., and Unanue, E. R. (1993) J.

Immunol. 151(5), 2419-2425

40. Harding, C., Roof, R., Allen, P., and Unanue, E. (1991) Proc. Natl. Acad. Sci. USA 88,

2740-2744

41. Deck, B., Elofsson, M., Kihlberg, J., and Unanue, E. (1995) J. Immunol. 155, 1074-1078

42. Dudler, T., Altmann, F., Carballido, J., and Blaser, k. (1995) Eur. J. Immunol. 25,

538-542

43. Corthay, A., Backlund, J., Broddefalk, J., Michaelsson, E., Goldschmidt, T. J., Kihlberg,

J., and Holmdahl, R. (1998) Eur J Immunol 28(8), 2580-90.

44. Michaelsson, E., Malmstrom, V., Reis, S., Engstrom, A., Burkhardt, H., and Holmdahl, R.

(1994) J Exp Med 180(2), 745-9.

45. Michaelsson, E., Broddefalk, J., Engstrom, A., Kihlberg, J., and Holmdahl, R. (1996) Eur

J Immunol 26(8), 1906-10.

46. Jensen, T., Hansen, P., Galli-Stampino, L., Mouritsen, S., Frische, K., Meinjohanns, E.,

(21)

47. Jensen, t., Hansen, P., Faurskov-Nielsen, A., Meldal, M., Komba, S., and Werdelin, O. (1999) Eur. J. Immunol. 29, 2759-2768

48. Jensen, T., Galli-Stampino, L., Mouritsen, S., Frische, K., Peters, S., Meldal, M., and

Werdelin, O. (1996) Eur J Immunol 26(6), 1342-9.

49. Galli-Stampino, L., Meinjohanns, E., Frische, K., Meldal, M., Jensen, T., Werdelin, O.,

and Mouritsen, S. (1997) Cancer Res 57(15), 3214-22.

50. Haurum, J. S., Hoier, I. B., Arsequell, G., Neisig, A., Valencia, G., Zeuthen, J., Neefjes,

J., and Elliott, T. (1999) J Exp Med 190(1), 145-50.

51. Kittlesen, D. J., Thompson, L. W., Gulden, P. H., Skipper, J. C., Colella, T. A.,

Shabanowitz, J., Hunt, D. F., Engelhard, V. H., Slingluff, C. L., Jr., and Shabanowitz, J. A. (1998) J Immunol 160(5), 2099-106.

52. Meadows, L., Wang, W., den Haan, J. M., Blokland, E., Reinhardus, C., Drijfhout, J. W.,

Shabanowitz, J., Pierce, R., Agulnik, A. I., Bishop, C. E., Hunt, D. F., Goulmy, E., and Engelhard, V. H. (1997) Immunity 6(3), 273-81.

53. Pierce, R. A., Field, E. D., den Haan, J. M., Caldwell, J. A., White, F. M., Marto, J. A.,

Wang, W., Frost, L. M., Blokland, E., Reinhardus, C., Shabanowitz, J., Hunt, D. F., Goulmy, E., and Engelhard, V. H. (1999) J Immunol 163(12), 6360-4.

54. Wood, P., and Elliott, T. (1998) J Exp Med 188(4), 773-8.

55. Bacik, I., Snyder, H. L., Anton, L. C., Russ, G., Chen, W., Bennink, J. R., Urge, L.,

Otvos, L., Dudkowska, B., Eisenlohr, L., and Yewdell, J. W. (1997) J Exp Med 186(4), 479-87.

(22)

LEGENDS TO FIGURES

Figure 1. GP392-400 is immunogenic in C57BL/6 mice. CTL were obtained from C57BL/6

mice immunized with 50-100 µg of peptides and restimulated as described in the Materials and

Methods section. A) 51Cr-labeled RMA cells or B) 51Cr-labeled MC57 cells were pulsed with 10-6

M of synthetic GP392-400 (closed circles) or not (open circles) and were incubated with the anti-GP392-400 CTL line at the indicated E :T ratio for 4 h at 37°C.

Figure 2. Ability of GP392-400-specific or GP92-101-specific CTL to recognise post-translationally modified analogs of GP392-400 or GP92-101. 51Cr-labeled RMA cells were

pulsed with increasing concentrations (0 to 10-6 M) of synthetic peptides GP392-400 (black

circles), [GlcNac-N396]-GP392-400 (open circles) and [D396]-GP392-400 (grey circles) and were incubated with the anti-GP392-400 CTL line (panel A) or anti-GP392-400 CTL clone (panel B) at an E:T ratio of 10:1 for 4 h. A similar analysis was performed for GP92-101 (black squares), [GlcNac-N95]-GP92-101 (open squares) and [D95]-GP92-101 (grey squares) testing recognition by a GP92-101-specific CTL line (panel C).

Figure 3. Binding of GP392-400, GP92-101 and their glycosylated and de-glycosylated analogs to H-2Db. Binding to H-2Db was measured using a previously described stabilization assay. Increasing concentrations (0 to 10-4 M) of unmodified (black circles), N-glycosylated (open circles) and de-N-glycosylated (grey circles) forms of GP392-400 (panel A) or GP92-101 (panel B) were incubated for 4 h at 37°C with RMA-S cells previously cultured for 40 h at 26°C.

(23)

cells were analyzed by flow cytometry. Results are representative of those obtained in three independent experiments.

Figure 4. Recognition of LCMV-infected MC57 cells by anti-GP392-400 specific CTL.

MC57 (H-2b) cells were previously infected (closed circles) or not (open circles) with LCMV

(m.o.i = 2) 48 h before the assay. 51Cr-labeled MC57 target cells were then incubated for 4 h with

the anti-GP392-400 CTL line (panel A), anti-GP392-400 CTL clone (panel B) or anti-GP92-101 CTL, (panel C) at the indicated E:T ratio. Specific lysis was measured as indicated in the Materials and Methods section.

Figure 5. Recognition of acid-eluted peptides from LCMV-infected MC57 cells by anti-GP392-400 specific CTL. Each HPLC fraction corresponding to mock-extracted synthetic

LCMV GP396-400 control peptide (solid circles, panel A) or to material eluted from the surface

of LCMV-infected MC57 cells (2 x 108 eq) (open circles, panel B) was tested for its capacity to

sensitize RMA cells to lysis by anti-GP392-400 CTL in a classical CTL assay as described in Materials and Methods. Arrows numbered 1, 2 and 3 indicate the retention times of synthetic peptides [GlcNac-N396]-GP392-400, GP392-400 and [D396]-GP392-400, respectively. Results are expressed as percentage (%) of specific lysis, and are representative of two independent experiments.

Figure 6. Molecular models of LCMV GP92-101 and GP392-400 complexed with H-2Db.

The three-dimensional models for LCMV GP92-101 (CSANNSHHYI) (A) and LCMV

GP392-400 (WLVTNGSYL) (B) interacting with H-2Db have been produced using the online modeling

(24)

The three residues N, X and S of the NXS glycosylation motif are shown in red (N95 or N396 as acceptor residue of N-glycosylation and de-N-glycosylation), yellow (N96 or G397) and blue

(S97 or S298), respectively. Arrows indicate the H-2Db anchor positions. Post–translational

modifications of N95 do not affect MHC presentation of GP92-101 but influence T cell recognition (this study, ref. 16), whereas post-translational modifications of N396 are detrimental

(25)

Sequence Peptide Cellular Status CTL epitope H-2Db binding (a) Immunogenicity (b) Presented by LCMV-infected cells GP392-400 WLVTNGSYL GP392-400 genetically encoded No

(this study, ref. 28)

Efficient (this study, ref. 25, 28)

Yes (this study) Not detected (this study) N-Glycan I WLVTNGSYL [GlcNac-N396]-GP392-400 glycosylated in the mature viral protein No (this study) Weak (this study) No (this study) Not detected (this study) WLVTDGSYL [D396]-

GP392-400 post-translationnaly modified (this study) No (this study) Moderate (this study) No Not detected (this study)

GP92-101 CSANNSHHYI GP92-101 genetically encoded Yes (ref. 16, 26) Efficient (this study, ref. 16, 25)

Yes

(this study, ref. 16)

Yes (ref. 16) N-Glycan I CSANNSHHYI [GlcNac-N95]-GP92-101 glycosylated in the mature viral protein No (ref. 16) Efficient (this study, ref. 16)

Yes

(this study, ref. 16)

Not detected (ref. 16)

CSADNSHHYI [D95]-

GP92-101 post-translationnaly modified (ref. 16) Yes (this study, ref. 16) Efficient (this study, ref. 16) Yes (ref. 16) Yes

a) Binding was considered as efficient, moderate or weak when measured half maximal binding values were ≤ 1µM, ≤ 100 µM or > 100 µM respectively. b) Peptides were considered as immunogenic or not when a CTL response was generated or not following injection of synthetic peptide as described under Material and Methods.

(26)

0 10 20 30 40 1 10 100 0 10 20 30 40 1 10 100 E:T ratio %s p ec if icl ys by guest on November 15, 2015 http://www.jbc.org/

(27)

CTL line CTL clone CTL line peptide (- Log M) 0 20 40 60 80 0 12 10 8 6 0 20 40 60 0 12 10 8 6 % specific lysis 0 20 40 60 80 0 12 10 8 6 by guest on November 15, 2015 http://www.jbc.org/

(28)

30 50 70 90 110 130 0 9 8 7 6 5 4 30 50 70 90 110 130 0 9 8 7 6 5 4 peptide (- Log M) mean fluor esc ence inte by guest on November 15, 2015 http://www.jbc.org/

(29)

0 20 40 60 80 0.1 0 20 40 60 80 0.1 0 20 40 60 80 0.1 % specific lysis 1 10 100 1 10 100 1 10 100 E:T ratio by guest on November 15, 2015 http://www.jbc.org/

(30)

0 % specific l ysis 3 1 0 5

B/ peptides eluted from LCMV-infected MC57 cells 10 1 2 3 1: [GlcNac-N396]-GP392-400 2: GP392-400 3: [D396]-GP392-400 15 10 5 0 10 20 30 40 50 60 70 80 90 HPLC fractions by guest on November 15, 2015 http://www.jbc.org/ Downloaded from

(31)

N N G S B/ GP392-400 Figure 6 by guest on November 15, 2015 http://www.jbc.org/

Références

Documents relatifs

When using spinner bottles with Hela S3 cells, the method can be run in the background, and requires only a couple of hours per week for cell harvesting and re-inoculation of

Encountering grammatically correct and incorrect three- word se- quences (e.g., ‘this horse fell’ versus ‘fell horse this’, respectively), readers had to make syntactic

L’archive ouverte pluridisciplinaire HAL, est destinée au dépôt et à la diffusion de documents scientifiques de niveau recherche, publiés ou non, émanant des

10 3 kilo- k 10 –1 deci- d 10 –2 centi- c 10 –3 milli- m 10 –6 micro- μ 10 –9 nano- n 10 –12 pico- p. Symbol

GM8 : Comparer, estimer, mesurer des durées (Unités de mesure usuelles de durées : j, semaine, mois, année, siècle, millénaire.) Relations entre ces unités.. GM9 : Dans des

The following results were obtained: (1) complex bilateral SOL and TAR muscle EMG responses were evoked with spinal onset latencies (2) leg muscle EMG activity did not change within

In order to determine how dependent the reproducibil- ity of the results of the prediction methods YKW, SVMHC and DynaPred POS are on the size of the avail- able allele datasets

L ü n z Hücher: Der foll nur kommen; dem wollen wir die Leviten verlesen. Was soll das heißen: Ein wohlhabender Bauer, gesund und kräftig, dem ist das Arbeiten verleidet in