Supplementary Table 1. T-cell receptor sequences of HERV-K(HML-2)-specific CD8 + T cell clone.

Similar documents
Supplementary Table 1. Functional avidities of survivin-specific T-cell clones against LML-peptide

NK mediated Antibody Dependent Cellular Cytotoxicity in HIV infections

HERV-K specific T cells eliminate diverse HIV-1/2 and SIV primary isolates

Commercially available HLA Class II tetramers (Beckman Coulter) conjugated to

DEBATE ON HIV ENVELOPE AS A T CELL IMMUNOGEN HAS BEEN GAG-GED

Supporting Information

Mechanisms of antagonism of HIVspecific CD4+ T cell responses BSRI

Surface plasmon resonance (SPR) analysis

Cytotoxicity assays. Rory D. de Vries, PhD 1. Viroscience lab, Erasmus MC, Rotterdam, the Netherlands

Antigen Recognition by T cells

Supplementary Figure 1. Enhanced detection of CTLA-4 on the surface of HIV-specific

Potential cross reactions between HIV 1 specific T cells and the microbiome. Andrew McMichael Suzanne Campion

Supplementary Data 1. Alanine substitutions and position variants of APNCYGNIPL. Applied in

Supplementary Figure 1. Example of gating strategy

CD25-PE (BD Biosciences) and labeled with anti-pe-microbeads (Miltenyi Biotec) for depletion of CD25 +

T Cell Development. Xuefang Cao, MD, PhD. November 3, 2015

SUPPLEMENTARY INFORMATION

Significance of the MHC

staining and flow cytometry

Rajesh Kannangai Phone: ; Fax: ; *Corresponding author

Recombinant Vaccinia Virus-Induced T-Cell Immunity: Quantitation of the Response to the Virus Vector and the Foreign Epitope

Biomolecules: amino acids

RAISON D ETRE OF THE IMMUNE SYSTEM:

Supplementary Figure 1. Using DNA barcode-labeled MHC multimers to generate TCR fingerprints

Blocking antibodies and peptides. Rat anti-mouse PD-1 (29F.1A12, rat IgG2a, k), PD-

The Structure and Function of Large Biological Molecules Part 4: Proteins Chapter 5

DIRECT IDENTIFICATION OF NEO-EPITOPES IN TUMOR TISSUE

Rapid perforin upregulation directly ex vivo by CD8 + T cells is a defining characteristic of HIV elite controllers

TITLE: Development of Antigen Presenting Cells for adoptive immunotherapy in prostate cancer

Mutations and Disease Mutations in the Myosin Gene

Structural Analysis of TCRpMHC Complexes Using Computational Tools. Feroze Mohideen Briarcliff High School

Evidence for antigen-driven TCRβ chain convergence in the tumor infiltrating T cell repertoire

CD44

Introduction. and Department of Medical Oncology, Dana-Farber Cancer Institute, 77 Avenue Louis Pasteur, Boston, MA 02115, USA

Principles of Adaptive Immunity

Nature Biotechnology: doi: /nbt Supplementary Figure 1. Binding capacity of DNA-barcoded MHC multimers and recovery of antigen specificity

1,000 in silico simulated alpha, beta, gamma and delta TCR repertoires were created.

Defining kinetic properties of HIV-specific CD8 + T-cell responses in acute infection

How T cells recognize antigen: The T Cell Receptor (TCR) Identifying the TCR: Why was it so hard to do? Monoclonal antibody approach

A second type of TCR TCR: An αβ heterodimer

SUPPLEMENTARY INFORMATION

HD1 (FLU) HD2 (EBV) HD2 (FLU)

Therapeutic effect of baicalin on experimental autoimmune encephalomyelitis. is mediated by SOCS3 regulatory pathway

Why are validated immunogenicity assays important for HIV vaccine development?

Copyright 2008 Pearson Education, Inc., publishing as Pearson Benjamin Cummings

CS612 - Algorithms in Bioinformatics

MHC class I MHC class II Structure of MHC antigens:

Properties of amino acids in proteins

T cell Receptor. Chapter 9. Comparison of TCR αβ T cells

Narrowed TCR repertoire and viral escape as a consequence of heterologous immunity

Treatment with IL-7 Prevents the Decline of Circulating CD4 + T Cells during the Acute Phase of SIV Infection in Rhesus Macaques

Key Concept B F. How do peptides get loaded onto the proper kind of MHC molecule?

B F. Location of MHC class I pockets termed B and F that bind P2 and P9 amino acid side chains of the peptide

Significance of the MHC

Supplementary Figure 1.

Antigen Presentation to T lymphocytes

Supporting Information

Determinants of Immunogenicity and Tolerance. Abul K. Abbas, MD Department of Pathology University of California San Francisco

Section 1 Proteins and Proteomics

T Cell Development II: Positive and Negative Selection

Profiling HLA motifs by large scale peptide sequencing Agilent Innovators Tour David K. Crockett ARUP Laboratories February 10, 2009

Lecture 11. Immunology and disease: parasite antigenic diversity

READ THIS FIRST. Your Name

Supplementary Figure 1. IL-12 serum levels and frequency of subsets in FL patients. (A) IL-12

Chapter 22: The Lymphatic System and Immunity

Supporting Online Material for

Third line of Defense

Received 19 September 2007/Accepted 21 November 2007

Chapter 6. Antigen Presentation to T lymphocytes

Cellular Immunity in Aging and HIV: Correlates of Protection. Immune Senescence

Significance of the MHC

Cell Mediated Immunity (I) Dr. Aws Alshamsan Department of Pharmaceu5cs Office: AA87 Tel:

Supplementary Information. Supplementary Figure 1

There are 2 major lines of defense: Non-specific (Innate Immunity) and. Specific. (Adaptive Immunity) Photo of macrophage cell

Antigen presenting cells

7.012 Quiz 3 Answers

Innate and Cellular Immunology Control of Infection by Cell-mediated Immunity

SUPPLEMENTARY INFORMATION

Medical Virology Immunology. Dr. Sameer Naji, MB, BCh, PhD (UK) Head of Basic Medical Sciences Dept. Faculty of Medicine The Hashemite University

Use of BONSAI decision trees for the identification of potential MHC Class I peptide epitope motifs.

Nature Immunology: doi: /ni Supplementary Figure 1. Gene expression profile of CD4 + T cells and CTL responses in Bcl6-deficient mice.

Degenerate T-cell Recognition of Peptides on MHC Molecules Creates Large Holes in the T-cell Repertoire

Supplemental Materials for. Effects of sphingosine-1-phosphate receptor 1 phosphorylation in response to. FTY720 during neuroinflammation

Lecture 7: Signaling Through Lymphocyte Receptors

There are approximately 30,000 proteasomes in a typical human cell Each proteasome is approximately 700 kda in size The proteasome is made up of 3

RAISON D ETRE OF THE IMMUNE SYSTEM:

Supporting Information

Developing Understanding of CMI. Dr Tom Wilkinson Associate Professor of Respiratory Medicine Faculty of Medicine University of Southampton UK

Epitope discovery. Using predicted MHC binding CENTER FOR BIOLOGICAL SEQUENCE ANALYSIS

Antiviral CD8 + T Cells Restricted by Human Leukocyte Antigen Class II Exist during Natural HIV Infection and Exhibit Clonal Expansion

PBMC from each patient were suspended in AIM V medium (Invitrogen) with 5% human

On an individual level. Time since infection. NEJM, April HIV-1 evolution in response to immune selection pressures

Received 25 April 2002/Accepted 21 May 2002

MATERIALS AND METHODS. Neutralizing antibodies specific to mouse Dll1, Dll4, J1 and J2 were prepared as described. 1,2 All

Variable HIV peptide stability in human cytosol is critical to epitope presentation and immune escape

Bioinformation Volume 5

Supplementary Materials for

EMERGING ISSUES IN THE HUMORAL IMMUNE RESPONSE TO HIV. (Summary of the recommendations from an Enterprise Working Group)

Supplementary Figure 1. Normal T lymphocyte populations in Dapk -/- mice. (a) Normal thymic development in Dapk -/- mice. Thymocytes from WT and Dapk

Defensive mechanisms include :

Transcription:

Supplementary Table 1. T-cell receptor sequences of HERV-K(HML-2)-specific CD8 + T cell clone. alpha beta ATGCTCCTGCTGCTCGTCCCAGTGCTCGAGGTGATTTTTACTCTGGGAGGAACCAGAGCC CAGTCGGTGACCCAGCTTGACAGCCACGTCTCTGTCTCTGAAGGAACCCCGGTGCTGCTG AGGTGCAACTACTCATCTTCTTATTCACCATCTCTCTTCTGGTATGTGCAACACCCCAAC AAAGGACTCCAGCTTCTCCTGAAGTACACATCAGCGGCCACCCTGGTTAAAGGCATCAAC GGTTTTGAGGCTGAATTTAAGAAGAGTGAAACCTCCTTCCACCTGACGAAACCCTCAGCC CATATGAGCGACGCGGCTGAGTACTTCTGTGTTGTGAGTACTCTCAAGATCATCTTTGGA AAAGGGACACGACTTCATATTCTCCCCAATATCCAGAACCCTGACCCTGCCGTGTACCAG CTGAGAGACTCTAAATCCAGTGACAAGTCTGTCTGCCTATTCACCGATTTTGATTCTCAA ACAAATGTGTCACAAAGTAAGGATTCTGATGTGTATATCACAGACAAAACTGTGCTAGAC ATGAGGTCTATGGACTTCAAGAGCAACAGTGCTGTGGCCTGGAGCAACAAATCTGACTTT GCATGTGCAAACGCCTTCAACAACAGCATTATTCCAGAAGACACCTTCTTCCCCAGCCCA GAAAGTTCCTGTGATGTCAAGCTGGTCGAGAAAAGCTTTGAAACAGATACGAACCTAAA CTTTCAAAACCTGTCAGTGATTGGGTTCCGAATCCTCCTCCTGAAAGTGGCCGGGTTTAAT CTGCTCATGACGCTGCGGCTGTGGTCCAGCTGA ATGGGCACCAGCCTCCTCTGCTGGATGGCCCTGTGTCTCCTGGGGGCAGATCACGCAGAT ACTGGAGTCTCCCAGGACCCCAGACACAAGATCACAAAGAGGGGACAGAATGTAACTTT CAGGTGTGATCCAATTTCTGAACACAACCGCCTTTATTGGTACCGACAGACCCTGGGGCA GGGCCCAGAGTTTCTGACTTACTTCCAGAATGAAGCTCAACTAGAAAAATCAAGGCTGCT CAGTGATCGGTTCTCTGCAGAGAGGCCTAAGGGATCTTTCTCCACCTTGGAGATCCAGCG CACAGAGCAGGGGGACTCGGCCATGTATCTCTGTGCCAGCAGCATAGGCCCGTCTGAAG CTTTCTTTGGACAAGGCACCAGACTCACAGTTGTAGAGGACCTGAACAAGGTGTTCCCAC CCGAGGTCGCTGTGTTTGAGCCATCAGAAGCAGAGATCTCCCACACCCAAAAGGCCACA CTGGTGTGCCTGGCCACAGGCTTCTTCCCTGACCACGTGGAGCTGAGCTGGTGGGTGAAT GGGAAGGAGGTGCACAGTGGGGTCAGCACGGACCCGCAGCCCCTCAAGGAGCAGCCCGC CCTCAATGACTCCAGATACTGCCTGAGCAGCCGCCTGAGGGTCTCGGCCACCTTCTGGCA GAACCCCCGCAACCACTTCCGCTGTCAAGTCCAGTTCTACGGGCTCTCGGAGAATGACGA GTGGACCCAGGATAGGGCCAAACCCGTCACCCAGATCGTCAGCGCCGAGGCCTGGGGTA GAGCAGACTGTGGCTTTACCTCGGTGTCCTACCAGCAAGGGGTCCTGTCTGCCACCATCC TCTATGAGATCCTGCTAGGGAAGGCCACCCTGTATGCTGTGCTGGTCAGCGCCCTTGTGTT GATG GCCATGGTCAAGAGAAAGGATTTCTGA 1

Supplementary Table 2. Results summary from IMGT/V-QUEST tool alpha chain of HERV-K(HML-2)-specific CD8 + T cell clone TCR. Result summary: Productive TRA rearranged sequence (no stop codon and in-frame junction) V-GENE and allele Homsap TRAV8-2*01 F score = 1360 identity = 100,00% (273/273 nt) J-GENE and allele Homsap TRAJ30*01 F score = 209 identity = 91,84% (45/49 nt) FR-IMGT lengths, CDR-IMGT lengths and AA JUNCTION [26.17.34.11] [6.8.8] CVVSTLKIIF Supplementary Table 3. Results summary from IMGT/V-QUEST tool beta chain of HERV-K(HML-2)-specific CD8 + T cell clone TCR. Result summary: Productive TRB rearranged sequence (no stop codon and in-frame junction) V-GENE and allele Homsap TRBV7-9*03 F score = 1375 identity = 100,00% (276/276 nt) J-GENE and allele Homsap TRBJ1-1*01 F score = 186 identity = 87,50% (42/48 nt) D-GENE and allele by IMGT/JunctionAnalysis Homsap TRBD1*01 F D-REGION is in reading frame 1 FR-IMGT lengths, CDR-IMGT lengths and AA JUNCTION [26.17.38.10] [5.6.10] CASSIGPSEAFF 2

Supplementary Table 4. HIV-1 viruses. Clade/CRF Catalog # Reference ID Full Length Accession Coreceptor A 2521 ELI K03454 X4 A 11245 99KE_KNH1135 AF47605 R5 A 11248 00KE_KNH1209 AF457069 R5 A 11244 99KE_KNH1088 AF457063 R5 A 11247 00KE_KNH1207 AF47068 R5 B 11250 94US_33931N AY713410 R5 B 7686 91_US1 AY173952 R5 B 7691 90TH_BK132 AY173951 X4 B 510 85US_Ba_L AY713409 R5 C 11254 02ET_14 AY255825 R5 C 7697 89SM_145 AY713415 R5 C 11258 02_ET288 AY713417 R5 C 11253 98US_MSC5016 AY444801 R5 C 2900 93IN_101 N/A R5 D 11259 98UG_57128 AF484502 R5 D 11263 00UG_J32228M4 AF484516 R5 D 10047 1165MB N/A Not tested G 3187 G3 NA R5 CRF02AG 11283 01CM_1475MV AY371138 R5 CRF02AG 7685 91DJ_263 AF063223 R5 3

A 3512 HIV-2 7924A N/A Not tested A 3513 HIV-2 60415K N/A Not tested N/A 3060 SIVhu N/A Not tested N/A 253 SIVmac251 N/A Not tested Supplementary Table 5. Quantitative PCR primers and probes. Forward Primer (5'-3') Reverse Primer (5'-3') Probe (5'-3') HERV-K-HML-2-gag TCGGGAAACGAGCAAAGG GAATTGGGAATGCCCCAGTT 6FAM-CTCAGGCCCCACAAC-MGBNFQ HERV-K-HML-2-pol GGGAATGCTTAATAGTCCAACTATTTG TGAAAACTTGTCTCTAACTGGTTGAAG 6FAM-CAGACTTTTGTAGCTCAAG-MGBNFQ HERV-K-HML-2-env GGGTACCTGGCCCCATAGA CATCATCCCTTCTTCCTCAGGTT 6FAM-ATCGCTGCCCTGCC-MGBNFQ HERV-K-HML-2-rec GTGACACAAACCCCAGAGAGTATG CTGCAGACACCATTGATACAATCA 6FAM-TGCTTGCAGCCTTG-MGBNFQ TBP AAGTTGGGTTTTCCAGCTAAGTTC CATCACAGCTCCCCACCATAT 6FAM-TGGACTTCAAGATTCAG-MGBNFQ PP1A TCTGCCCACCTTACAGACC GATCAAATCCGCCACCTCTA N/A GAPDH TTGACCTCAGCTGCACATTC AGGATGGTCTCGAGTGCTTG N/A β-actin CACAGGGGAGGTGATAGCAT CACGAAGGCTCATCATTCAA N/A Supplementary Results. Additional Characterization of HERV-K(HML-2)-Env-specific T cell clone. Additional Characterization of T-cell Determinant. The EC 50 of the HERV-K(HML-2)-Env-specific T cell clone to the optimal epitope, at approximately 4µM, is considerably higher than that typically observed with virus-specific T cell responses. One possible explanation for this is that this T cell clone is of relatively low avidity. However it is also possible that this response was primarily raised against a distinct HERV-K-Env sequence containing mismatches with the consensus HERV-K(HML-2)-Env CIDSTFNWHQRI sequence. We assembled an alignment of HERV-K-Env sequences and selected the following variants of this sequence: CIDLTFNWQHRI, CIDSTFDWQHHI, CIDSTFDWQHYI, CIDSTSDWQHHI. Peptides corresponding to these sequences were manufactured, but 4

each of these completely failed to stimulate the clone (data not shown). This does not represent an exhaustive list of HERV-K variant sequences, however, and we feel that it may be important for future studies to identify precisely which subset of HERV-K insertions are induced upon HIV-1 infection. A third possible explanation for the high EC 50 of the clone recognizes is that it may recognize only a modified version of the determinant that is present at low abundance in our synthetic peptide. It is important to note that we can strictly rule out the possibility that the clone could be recognizing a minor foreign contaminating peptide by virtue of the consistent recognition of 6 different batches of the CIDSTFNWQHRI peptide (from two different manfacturers), by the logical fine-mapping of the epitope where any peptide containing the minimal epitope is recognized while shorter peptides are not, and by the very similar titration curves observed with different peptide preparations (ex. CIDSTFNWQHRILLV vs CIDSTFNWQHRI, Figure 3D). One possibility is that the clone recognizes the native peptide sequence but that this sequence is present at low abundance in the synthetic peptide. It has previously been demonstrated that sulfhydryl modification of cysteine is responsible for reducing the antigenicity of the influenza virus nuclear protein determinants NP 39-47 and NP 218-226 and that replacing cysteine residues in these determinants with either serine or alanine resulted in substantial increases in antigenicity (1). Similarly, substitution of the cysteine residue in the lympocytic choriomeningitis virus T cell determinant KAVYNFATC with methionine, to give KAVYNFATM, results in a decrease in the EC 50 from >5µM to 21nM by preventing the formation of cysteine dimers (2). We tested the effect of replacing the N-terminal cysteine in CIDSTFNWQHRI with either alanine, serine methionine or tyrosine 5

(AIDSTFNWQHRI, SIDSTFNWQHRI, MIDSTFNWQHRI and YIDSTFNWQHRI) on the antigenicity of the peptide. Substitution with either alanine, serine, or threonine completely abolished recognition by the HERV-K(HML-2)-Env-specific T cell clone, while the methionine-substituted peptide was recognized at a reduced level (data not shown). We further tested the response of the T cell clone to reduced (TCEP treated) CIDSTFNWQHRI peptide as well as oxidized dimeric peptide and observed lesser antigenicity of both of these modified peptides as compared to the untreated peptide (data not shown). Taken together, these data suggest that a modification of the cysteine residue in this peptide may be required for optimal immunogenicity but that this is not simply related to redox state. Next, we tested the possibility that a minor species of the CIDSTFNWQHRILLV peptide bearing an unknown modification is the primary T cell determinant recognized by the HERV-K(HML-2)-Env specific T cell clone, 50mg of crude peptide was divided into 60 fractions by C18 reversed-phase HPLC (JPT Peptide Technologies) and each fraction was tested for its ability to stimulate the T cell clone in IFN-γ ELISPOT assays. The T cell clone responded maximally to fraction 8 with lesser responses to fractions 9-11 and no response observed to later fraction. This overlapped only partially with the elution profile of the native CIDSTFNWQHRILLV peptide (identified by a 923 mw/z peak) that was the primary product in fractions 8 18 (data not shown). The elution of CIDSTFNWQHRILLV peaked at fractions 12 and 13, while the clone did not respond to these fractions (data not shown). These data indicate that the T cell determinant of the HERV-K(HML-2)-Env specific T cell clone is a modified version of CIDSTFNWQHRILLV, which is slightly less hydrophobic than the native peptide (elutes 6

earlier). Peptide titration experiments performed on fraction 8 demonstrated an EC 50 of 2µM, only slightly lower than the 4µM of the previously tested >98% pure CIDSTFNWQHRILLV. This corresponds with the observation that fraction 8 does not represent an isolated modified species but rather is still comprised primarily of native peptide. In fact, no unique peak could be observed in fractions 8-11 on the corresponding mass spectra, suggesting that the T-cell agonist is still present as only a minor component in these fractions. This has prevented us from using these data to identify the peptide modification needed for antigenicity. Deamidation of asparagine or glutamine residues represents one common peptide modification that has been shown to alter antigenicity(3). We therefore tested whether demamidation of the asparagine (N) residue in CIDSTFNWQHRI to asparatate or iso-aspartate (CIDSTFDWHQRI or CIDSTF-iso-Asp- WQHRI) resulted in a more antigenic peptide. Both of these modified peptides completely failed to stimulate the HERV-K(HML-2)-Env-specific T-cell clone (data not shown). Taken together, these data indicate that the HERV-K(HML-2)-Env-specific T-cell clone is remarkably specific for its cognate determinant, as a number of very subtle changes in the peptide sequence abolished recognition. The peptide fractionation experiment clearly demonstrates that the unmodified CIDSTFNWQHRILLV peptide is not the determinant recognized by the clone however, we have been unable to identify the precise identity of the modification required to render this peptide antigenic. The fact that the exact chemical composition of determinants recognized by T-cells is generally unknown has been highlighted by others (4) 7

Recognition of low-level HIV-1 infections at 16 hours post-infection by HERV- K(HML-2)-Env-specific T-cells. The majority of recognition assays presented in this manuscript tested the ability of HERV-K(HML-2)-Env-specific T-cells to recognize high-levels of HIV-1 infection. One of the side observations from the ARV suppression recognition assay presented in Figure 6 was that even the small amount of HIV-1 infection that occurred in nevirapinetreated cells was sufficient to induce measurable recognition by the T-cell clone, although this was reduced as compared to cells infected to high-levels in the absence of ARVs. We repeated this experiment and again observed that even a low-level HIV-1 infection defined by both a low frequency of Gag + cells and low-levels of Gag expression on a percell basis was sufficient to trigger robust recognition by the HERV-K(HML-2)-Envspecific T-cell clone. Also of note, the co-culture between clone and target was initiated at 16 hours post-infection. The addition of brefledin A at the time of co-culture would have blocked additional MHC-I antigen presentation over the course of this co-culture. Thus, low levels of HIV-1 infection are detectable by HERV-K(HML-2)-Env-specific T- cell clones within at least 16 hours of infection. 8

Supplementary Figure 1 CD4+ Targets ARV Mix Nevirapine No ARVs 10 4 10 4 10 1.39 17.2 4 55 10 2 10 2 10 2 CD4 10 2 10 4 10 2 10 4 10 2 10 4 HIV-1-Gag HERV-K(HML-2)-Env-specific clone ARV Mix Nevirapine No ARVs 10 4 10 5.63 2.68 4 10 20.7 35.7 4 13.4 70.7 10 2 2 10 2 CD107a 87.4 4.29 10 2 10 4 39.8 3.79 10 2 10 4 15.1 0.74 10 2 10 4 IFN-! Supplementary Figure 1. HERV-K(HML-2)-Env-specific T-cells recognize lowlevels of HIV-1 infection within 16 hours of infection. Primary CD4 + T-cells autologous to the HERV-K(HML-2)-Env-specific T-cell clone were treated with either nevirapine, a mix of ARVs (see Figure 6 legend), or maintained as no ARV controls. These cells were subsequently infected with HIV-1. At 16 hours post-infection, these infected targets were co-cultured with the HERV-K(HML-2)-Env-specific T-cell clone in the presence of brefeldin A for 12 hours. Shown are flow cytometry plots gated on CD8 + cells and depicting CD107a by IFN-γ. 9

Elimination of SIV-infected cells by HERV-K(HML-2)-Env-specific T-cells. Here we present additional data supporting that the HERV-K(HML-2)-Envspecific T-cell clone specifically eliminates SIV-infected cells. The percentage of SIVinfected primary CD4 + T-cells was substantially reduced upon co-culture with HERV- K(HML-2)-specific, but not with CMV-pp65- or HIV-1-Gag-specific CD8 + T-cell clones (Supplementary Figure 2A, B). Pretreatment of these target cells with the anti-mhc-i antibody DX17 reduced this effect. We also observed dose-dependent elimination of SIVmac251-infected targets upon the co-culture with different clone (effector) to target ratios of HERV-K(HML-2)-Env-specific T-cell clone (Supplementary Figure 2C). Supplementary Figure 2 10

Supplementary Figure 2. HERV-K(HML-2)-Env-specific T-cells eliminate SIVinfected cells. Primary CD4+ T-cells from subject OM9 were infected with the indicated SIV viruses. Infections were allowed to proceed for 72 hours and then either CMV-pp65-, HIV-1-Gag, or HERV-K(HML-2)-Env-specific T-cell clones were added at a ratio of 1 clone:10 targets (A, B) or at the indicated effector (clone) to target ratios and cultured for 24 hours. Where indicated, these target cells were pre-incubated with the anti-mhc-i antibody DX17 at 10µg/ml prior to the addition of T-cell clone. (C) SIVmac239-infected target cells were co-cultured with the HERV-K(HML-2)-Env-specific T-cell clone at the indicated effector (clone) to target ratios. Levels of infection were measured by flow cytometry staining for CD4 and intracellular HIV-1-Gag. Supplementary References. 1. Chen, W., Yewdell, J.W., Levine, R.L., and Bennink, J.R. 1999. Modification of cysteine residues in vitro and in vivo affects the immunogenicity and antigenicity of major histocompatibility complex class I- restricted viral determinants. J Exp Med 189:1757-1764. 2. Puglielli, M.T., Zajac, A.J., van der Most, R.G., Dzuris, J.L., Sette, A., Altman, J.D., and Ahmed, R. 2001. In vivo selection of a lymphocytic choriomeningitis virus variant that affects recognition of the GP33-43 epitope by H- 2Db but not H- 2Kb. J Virol 75:5099-5107. 3. Arentz- Hansen, H., Korner, R., Molberg, O., Quarsten, H., Vader, W., Kooy, Y.M., Lundin, K.E., Koning, F., Roepstorff, P., Sollid, L.M., et al. 2000. The intestinal T cell response to alpha- gliadin in adult celiac disease is focused on a single deamidated glutamine targeted by tissue transglutaminase. J Exp Med 191:603-612. 4. Yewdell, J.W. 2005. The seven dirty little secrets of major histocompatibility complex class I antigen processing. Immunol Rev 207:8-18. 11