Immunodominant tuberculosis CD8 antigens preferentially restricted by HLA-B

Deborah A Lewinsohn, Ervina Winata, Gwendolyn M Swarbrick, Katie E Tanner, Matthew S Cook, Megan D Null, Meghan E Cansler, Alessandro Sette, John Sidney, David M Lewinsohn, Deborah A Lewinsohn, Ervina Winata, Gwendolyn M Swarbrick, Katie E Tanner, Matthew S Cook, Megan D Null, Meghan E Cansler, Alessandro Sette, John Sidney, David M Lewinsohn

Abstract

CD8(+) T cells are essential for host defense to intracellular bacterial pathogens such as Mycobacterium tuberculosis (Mtb), Salmonella species, and Listeria monocytogenes, yet the repertoire and dominance pattern of human CD8 antigens for these pathogens remains poorly characterized. Tuberculosis (TB), the disease caused by Mtb infection, remains one of the leading causes of infectious morbidity and mortality worldwide and is the most frequent opportunistic infection in individuals with HIV/AIDS. Therefore, we undertook this study to define immunodominant CD8 Mtb antigens. First, using IFN-gamma ELISPOT and synthetic peptide arrays as a source of antigen, we measured ex vivo frequencies of CD8(+) T cells recognizing known immunodominant CD4(+) T cell antigens in persons with latent tuberculosis infection. In addition, limiting dilution was used to generate panels of Mtb-specific T cell clones. Using the peptide arrays, we identified the antigenic specificity of the majority of T cell clones, defining several new epitopes. In all cases, peptide representing the minimal epitope bound to the major histocompatibility complex (MHC)-restricting allele with high affinity, and in all but one case the restricting allele was an HLA-B allele. Furthermore, individuals from whom the T cell clone was isolated harbored high ex vivo frequency CD8(+) T cell responses specific for the epitope, and in individuals tested, the epitope represented the single immunodominant response within the CD8 antigen. We conclude that Mtb-specific CD8(+) T cells are found in high frequency in infected individuals and are restricted predominantly by HLA-B alleles, and that synthetic peptide arrays can be used to define epitope specificities without prior bias as to MHC binding affinity. These findings provide an improved understanding of immunodominance in humans and may contribute to a development of an effective TB vaccine and improved immunodiagnostics.

Conflict of interest statement

Competing interests. The authors have declared that no competing interests exist.

Figures

Figure 1. Determination of Human Effector Cell…
Figure 1. Determination of Human Effector Cell Frequencies Ex Vivo Using the IFN-γ ELISPOT Assay
Magnetic bead–purified CD8+ T cells from a single donor (D466) were cultured with DCs (20,000/well) either infected with Mtb (H37Rv, multiplicity of infection = 50) or pulsed with peptide pool representing CFP10 (5 μg/ml each peptide; 15-mer overlap by 11 aa) in an IFN-γ ELISPOT assay. Each responding T cell population was tested in duplicate at four different cell concentrations. To determine the effector cell frequency of antigen-specific T cells, the average number of spot forming units per well for each duplicate was plotted against the number of responder cells per well. Linear regression analysis was used to determine the slope of the line, which represents the frequency of antigen-specific T cells. The assay was considered positive, reflecting the presence of a primed T cell response, if the binomial probability for the number of spots was significantly different by experimental and control assays, i.e., if the experimental line is statistically significantly different from the control line. The frequency of Mtb-specific and CFP10-specific T cells demonstrated in this figure was 1/307 and 1/1676, respectively.
Figure 2. High Ex vivo CD8 +…
Figure 2. High Ex vivo CD8+ T Cell Frequencies to Mtb Antigens Are Associated with Mtb Infection
As in Figure 1, to determine ex vivo CD8+ T cell frequencies, autologous DCs either infected with Mtb (graph labeled “Mtb DC”) or pulsed with cognate peptide pools representing known Mtb CD4 antigens (all other graphs) were incubated with CD8+ T cells in an IFN-γ ELISPOT assay. The number of spot forming units (SFU)/250,000 cells is shown, a number which is derived from linear regression analysis of the frequency of T cells tested at concentrations of 5 × 105, 2.5 × 105, 1.2 × 105, and 6 × 104 cells/well. Individuals without evidence for Mtb infection (“normal”); those with LTBI, and those with active TB (“active” defined as culture confirmed pulmonary TB) were evaluated. “Mtb Infected” includes both the “LTBI” and “active” groups. The number of individuals tested for each antigen were: Mtb infection (normal, n = 11; LTBI, n = 11; active, n = 6); Mtb39 Pool A (normal, n = 14; LTBI, n = 20; active, n = 10); Mtb39 Pool B (normal, n = 14; LTBI, n = 20; active, n = 10); CFP10 (normal, n = 14; LTBI, n = 20; active, n = 10); Mtb8.4 (normal, n = 14; LTBI, n = 20; active, n = 10); Mtb9.9A (normal, n = 14; LTBI, n = 20; active, n = 5); ESAT-6 (normal, n = 14; LTBI, n = 20; active, n = 5); Ag85 Pool A (normal, n = 14; LTBI, n = 17; active, n = 5); Ag85 Pool B (normal, n = 14; LTBI, n = 17; active, n = 5); 19kDa (normal, n = 12; LTBI, n = 17; active, n = 5); and EsxG (normal, n = 14; LTBI, n = 17; active, n = 5). p-Values are noted where statistically significant differences between groups are noted (p = <0.05; Wilcoxon/Kruskal–Wallis).
Figure 3. Definition of Antigenic Specificity and…
Figure 3. Definition of Antigenic Specificity and HLA Restriction: Characterization of T Cell Clone D466 D6
(A–C) To identify the antigen and minimal epitope recognized by T cell clone D466 D6, T cells (5,000 cells/well) were incubated with autologous LCL (20,000/well) and antigen (5 μg/ml). Negative controls (media, no antigen) and positive controls (phytohemagglutanin [PHA], final, 10 μg/ml) are shown for each assay. IFN-γ was assessed by ELISPOT after 18 h of co-culture. Pictures of ELISPOT wells are shown. (A) Antigens consisted of peptide pools representing known CD4 Mtb antigens, made up of 15-aa peptides overlapping by 11 aa. (B) Antigens consisted of individual 15-aa CFP10 peptides that together constitute the peptide pool. (C) Antigens consisted of individual nested peptides within CFP101–15 (10 aa, 9 aa, or 8 aa), used to further map the epitope. (D) The restricting allele was identified using LCLs (20,000/well) that were either autologous or expressing HLA alleles matching D466 at one or two alleles (shown in bold font), pulsed with CFP102–10 (5 μg/ml) as APC. Autologous LCLs without peptide (control) are also shown. After 2 h, cells were washed and incubated with T cells (500 cells/well) in an IFN- γ ELISPOT assay. Pictures of ELISPOT wells are shown.
Figure 4. Confirmation of Minimal Epitope Mapping…
Figure 4. Confirmation of Minimal Epitope Mapping of D466 D6
To confirm the minimal epitope, autologous LCLs (20,000/well) were pulsed with peptide at the concentration indicated and co-cultured with T cells (1,000 cells/well). IFN-γ was assessed by ELISPOT after 18 h co-culture. Each point represents the mean of duplicate determinations.
Figure 5. Summary of Minimal Epitope Mapping…
Figure 5. Summary of Minimal Epitope Mapping Data
To determine the minimal epitope, autologous LCLs (20,000/well) were pulsed with peptide at the concentration indicated and co-cultured with T cells (1,000 cells/well). IFN-γ was assessed by ELISPOT after 18 h co-culture. Each point represents the mean of duplicate determinations.
Figure 6. Profiling of Immunodominance Pattern for…
Figure 6. Profiling of Immunodominance Pattern for CFP10
To determine the effector cell frequencies, autologous DCs (20,000/well) were pulsed either with each individual 15-mer peptide (5 μg/ml), the peptide pool (PP; 5 μg/each peptide), or the minimal epitope (ME) determined from T cell clones derived from each donor (D466:CFP102–11; D480:CFP103–11; D481:CFP1075–83; 5 μg/ml), and tested against 250,000 magnetic bead–purified CD8+ T cells. IFN-γ release was assessed by ELISPOT after 18 h of co-culture. Each point represents the mean of duplicate determinations.

References

    1. WHO report 2007. Geneva: World Health Organization; 2007. Global tuberculosis control: Surveillance, planning, financing.
    1. The global plan to stop TB, 2006–2015 / Stop TB Partnership. Geneva: World Health Organization; 2006.
    1. Flynn JL, Chan J. Immunology of tuberculosis. Annu Rev Immunol. 2001;19:93–129.
    1. North RJ, Jung YJ. Immunity to tuberculosis. Annu Rev Immunol. 2004;22:599–623.
    1. Oldstone MB. The role of cytotoxic T lymphocytes in infectious disease: History, criteria, and state of the art. Curr Top Microbiol Immunol. 1994;189:1–8.
    1. Orme IM. The dynamics of infection following BCG and Mycobacterium tuberculosis challenge in T-cell-deficient mice. Tubercle. 1987;68:277–283.
    1. Muller I, Cobbold SP, Waldmann H, Kaufmann SH. Impaired resistance to Mycobacterium tuberculosis infection after selective in vivo depletion of L3T4+ and Lyt-2+ T cells. Infect Immun. 1987;55:2037–2041.
    1. Silva CL, Silva MF, Pietro RC, Lowrie DB. Protection against tuberculosis by passive transfer with T-cell clones recognizing mycobacterial heat-shock protein 65. Immunology. 1994;83:341–346.
    1. Flynn JL, Goldstein MM, Triebold KJ, Koller B, Bloom BR. Major histocompatibility complex class I-restricted T cells are required for resistance to Mycobacterium tuberculosis infection. Proc Natl Acad Sci U S A. 1992;89:12013–12017.
    1. Ladel CH, Daugelat S, Kaufmann SH. Immune response to Mycobacterium bovis bacille Calmette Guerin infection in major histocompatibility complex class I- and II-deficient knock-out mice: Contribution of CD4 and CD8 T cells to acquired resistance. Eur J Immunol. 1995;25:377–384.
    1. Sousa AO, Mazzaccaro RJ, Russell RG, Lee FK, Turner OC, et al. Relative contributions of distinct MHC class I-dependent cell populations in protection to tuberculosis infection in mice. Proc Natl Acad Sci U S A. 2000;97:4204–4208.
    1. Behar SM, Dascher CC, Grusby MJ, Wang CR, Brenner MB. Susceptibility of mice deficient in CD1D or TAP1 to infection with Mycobacterium tuberculosis . J Exp Med. 1999;189:1973–1980.
    1. Rolph MS, Raupach B, Kobernick HH, Collins HL, Perarnau B, et al. MHC class Ia-restricted T cells partially account for beta2-microglobulin-dependent resistance to Mycobacterium tuberculosis . Eur J Immunol. 2001;31:1944–1949.
    1. Mogues T, Goodrich ME, Ryan L, LaCourse R, North RJ. The relative importance of T cell subsets in immunity and immunopathology of airborne Mycobacterium tuberculosis infection in mice. J Exp Med. 2001;193:271–280.
    1. Urdahl KB, Liggitt D, Bevan MJ. CD8+ T cells accumulate in the lungs of Mycobacterium tuberculosis-infected Kb-/-Db-/- mice, but provide minimal protection. J Immunol. 2003;170:1987–1994.
    1. Alderson MR, Bement T, Day CH, Zhu L, Molesh D, et al. Expression cloning of an immunodominant family of Mycobacterium tuberculosis antigens using human CD4(+) T cells. J Exp Med. 2000;191:551–560.
    1. Lewinsohn DM, Zhu L, Madison VJ, Dillon DC, Fling SP, et al. Classically restricted human CD8+ T lymphocytes derived from Mycobacterium tuberculosis-infected cells: Definition of antigenic specificity. J Immunol. 2001;166:439–446.
    1. Heinzel AS, Grotzke JE, Lines RA, Lewinsohn DA, McNabb AL, et al. HLA-E-dependent presentation of Mtb-derived antigen to human CD8+ T cells. J Exp Med. 2002;196:1473–1481.
    1. Lalvani A, Brookes R, Wilkinson RJ, Malin AS, Pathan AA, et al. Human cytolytic and interferon gamma-secreting CD8+ T lymphocytes specific for Mycobacterium tuberculosis . Proc Natl Acad Sci U S A. 1998;95:270–275.
    1. Mohagheghpour N, Gammon D, Kawamura LM, van Vollenhoven A, Benike CJ, et al. CTL response to Mycobacterium tuberculosis: Identification of an immunogenic epitope in the 19-kDa lipoprotein. J Immunol. 1998;161:2400–2406.
    1. Lam KS. Application of combinatorial library methods in cancer research and drug discovery. Anticancer Drug Des. 1997;12:145–167.
    1. Turner J, Dockrell HM. Stimulation of human peripheral blood mononuclear cells with live Mycobacterium bovis BCG activates cytolytic CD8+ T cells in vitro. Immunology. 1996;87:339–342.
    1. Lewinsohn DM, Alderson MR, Briden AL, Riddell SR, Reed SG, et al. Characterization of human CD8+ T cells reactive with Mycobacterium tuberculosis-infected antigen-presenting cells. J Exp Med. 1998;187:1633–1640.
    1. Beckman EM, Melian A, Behar SM, Sieling PA, Chatterjee D, et al. CD1c restricts responses of mycobacteria-specific T cells. Evidence for antigen presentation by a second member of the human CD1 family. J Immunol. 1996;157:2795–2803.
    1. Rosat JP, Grant EP, Beckman EM, Dascher CC, Sieling PA, et al. CD1-restricted microbial lipid antigen-specific recognition found in the CD8+ alpha beta T cell pool. J Immunol. 1999;162:366–371.
    1. Moody DB, Ulrichs T, Muhlecker W, Young DC, Gurcha SS, et al. CD1c-mediated T-cell recognition of isoprenoid glycolipids in Mycobacterium tuberculosis infection. Nature. 2000;404:884–888.
    1. Yewdell JW, Bennink JR. Immunodominance in major histocompatibility complex class I-restricted T lymphocyte responses. Annu Rev Immunol. 1999;17:51–88.
    1. Louise R, Skjot V, Agger EM, Andersen P. Antigen discovery and tuberculosis vaccine development in the post-genomic era. Scand J Infect Dis. 2001;33:643–647.
    1. Reed S, Lobet Y. Tuberculosis vaccine development; from mouse to man. Microbes Infect. 2005;7:922–931.
    1. Geluk A, van Meijgaarden KE, Franken KL, Drijfhout JW, D'Souza S, et al. Identification of major epitopes of Mycobacterium tuberculosis AG85B that are recognized by HLA-A*0201-restricted CD8+ T cells in HLA- transgenic mice and humans. J Immunol. 2000;165:6463–6471.
    1. Klein MR, Smith SM, Hammond AS, Ogg GS, King AS, et al. HLA-B*35-Restricted CD8 T Cell Epitopes in the Antigen 85 Complex of Mycobacterium tuberculosis . J Infect Dis. 2001;183:928–934.
    1. Charo J, Geluk A, Sundback M, Mirzai B, Diehl AD, et al. The identification of a common pathogen-specific HLA class I A*0201-restricted cytotoxic T cell epitope encoded within the heat shock protein 65. Eur J Immunol. 2001;31:3602–3611.
    1. Klein MR, Hammond AS, Smith SM, Jaye A, Lukey PT, et al. HLA-B*35-restricted CD8(+)-T-cell epitope in Mycobacterium tuberculosis Rv2903c. Infect Immun. 2002;70:981–984.
    1. Caccamo N, Milano S, Di Sano C, Cigna D, Ivanyi J, et al. Identification of epitopes of Mycobacterium tuberculosis 16-kDa protein recognized by human leukocyte antigen-A*0201 CD8(+) T lymphocytes. J Infect Dis. 2002;186:991–998.
    1. Lewinsohn DA, Lines RA, Lewinsohn DM. Human dendritic cells presenting adenovirally expressed antigen elicit Mycobacterium tuberculosis-specific CD8+ T cells. Am J Respir Crit Care Med. 2002;166:843–848.
    1. Lewinsohn DM, Briden AL, Reed SG, Grabstein KH, Alderson MR. Mycobacterium tuberculosis-reactive CD8+ T lymphocytes: The relative contribution of classical versus nonclassical HLA restriction. J Immunol. 2000;165:925–930.
    1. Wong P, Pamer EG. CD8 T cell responses to infectious pathogens. Annu Rev Immunol. 2003;21:29–70.
    1. Klenerman P, Hill A. T cells and viral persistence: Lessons from diverse infections. Nat Immunol. 2005;6:873–879.
    1. Kiepiela P, Leslie AJ, Honeyborne I, Ramduth D, Thobakgale C, et al. Dominant influence of HLA-B in mediating the potential co-evolution of HIV and HLA. Nature. 2004;432:769–775.
    1. Crubezy E, Ludes B, Poveda JD, Clayton J, Crouau-Roy B, et al. Identification of Mycobacterium DNA in an Egyptian Pott's disease of 5,400 years old. C R Acad Sci III. 1998;321:941–951.
    1. Salo WL, Aufderheide AC, Buikstra J, Holcomb TA. Identification of Mycobacterium tuberculosis DNA in a pre-Columbian Peruvian mummy. Proc Natl Acad Sci U S A. 1994;91:2091–2094.
    1. Sette A, Fleri W, Peters B, Sathiamurthy M, Bui HH, et al. A roadmap for the immunomics of category A-C pathogens. Immunity. 2005;22:155–161.
    1. Peters B, Bui HH, Frankild S, Nielson M, Lundegaard C, et al. A community resource benchmarking predictions of peptide binding to MHC-I molecules. PLoS Comput Biol. 2006;2:e65.
    1. Kamath AB, Woodworth J, Xiong X, Taylor C, Weng Y, et al. Cytolytic CD8+ T cells recognizing CFP10 are recruited to the lung after Mycobacterium tuberculosis infection. J Exp Med. 2004;200:1479–1489.
    1. Sylwester AW, Mitchell BL, Edgar JB, Taormina C, Pelte C, et al. Broadly targeted human cytomegalovirus-specific CD4+ and CD8+ T cells dominate the memory compartments of exposed subjects. J Exp Med. 2005;202:673–685.
    1. Noss EH, Pai RK, Sellati TJ, Radolf JD, Belisle J, et al. Toll-like receptor 2-dependent inhibition of macrophage class II MHC expression and antigen processing by 19-kDa lipoprotein of Mycobacterium tuberculosis . J Immunol. 2001;167:910–918.
    1. Tobian AA, Potter NS, Ramachandra L, Pai RK, Convery M, et al. Alternate class I MHC antigen processing is inhibited by Toll-like receptor signaling pathogen-associated molecular patterns: Mycobacterium tuberculosis 19-kDa lipoprotein, CpG DNA, and lipopolysaccharide. J Immunol. 2003;171:1413–1422.
    1. Gehring AJ, Rojas RE, Canaday DH, Lakey DL, Harding CV, et al. The Mycobacterium tuberculosis 19-kilodalton lipoprotein inhibits gamma interferon-regulated HLA-DR and Fc gamma R1 on human macrophages through Toll-like receptor 2. Infect Immun. 2003;71:4487–4497.
    1. Fulton SA, Reba SM, Pai RK, Pennini M, Torres M, et al. Inhibition of major histocompatibility complex II expression and antigen processing in murine alveolar macrophages by Mycobacterium bovis BCG and the 19-kilodalton mycobacterial lipoprotein. Infect Immun. 2004;72:2101–2110.
    1. Berthet FX, Rasmussen PB, Rosenkrands I, Andersen P, Gicquel B. A Mycobacterium tuberculosis operon encoding ESAT-6 and a novel low- molecular-mass culture filtrate protein (CFP-10) Microbiology. 1998;144:3195–3203.
    1. Dillon DC, Alderson MR, Day CH, Bement T, Campos-Neto A, et al. Molecular and immunological characterization of Mycobacterium tuberculosis CFP-10, an immunodiagnostic antigen missing in Mycobacterium bovis BCG. J Clin Microbiol. 2000;38:3285–3290.
    1. Sorensen AL, Nagai S, Houen G, Andersen P, Andersen AB. Purification and characterization of a low-molecular-mass T-cell antigen secreted by Mycobacterium tuberculosis . Infect Immun. 1995;63:1710–1717.
    1. Dillon DC, Alderson MR, Day CH, Lewinsohn DM, Coler R, et al. Molecular characterization and human T-cell responses to a member of a novel Mycobacterium tuberculosis mtb39 gene family. Infect Immun. 1999;67:2941–2950.
    1. Coler RN, Skeiky YA, Vedvick T, Bement T, Ovendale P, et al. Molecular cloning and immunologic reactivity of a novel low molecular mass antigen of Mycobacterium tuberculosis . J Immunol. 1998;161:2356–2364.
    1. Gey Van Pittius NC, Gamieldien J, Hide W, Brown GD, Siezen RJ, et al. The ESAT-6 gene cluster of Mycobacterium tuberculosis and other high G+C Gram-positive bacteria. Genome Biol. 2001;2:RESEARCH0044.
    1. Rosenkrands I, King A, Weldingh K, Moniatte M, Moertz E, et al. Towards the proteome of Mycobacterium tuberculosis . Electrophoresis. 2000;21:3740–3756.
    1. Collins ME, Patki A, Wall S, Nolan A, Goodger J, et al. Cloning and characterization of the gene for the ‘19 kDa' antigen of Mycobacterium bovis . J Gen Microbiol. 1990;136:1429–1436.
    1. Borremans M, de Wit L, Volckaert G, Ooms J, de Bruyn J, et al. Cloning, sequence determination, and expression of a 32-kilodalton-protein gene of Mycobacterium tuberculosis . Infect Immun. 1989;57:3123–3130.
    1. Romani N, Gruner S, Brang D, Kampgen E, Lenz A, et al. Proliferating dendritic cell progenitors in human blood. J Exp Med. 1994;180:83–93.
    1. Lewinsohn DA, Heinzel AS, Gardner JM, Zhu L, Alderson MR, et al. Mycobacterium tuberculosis-specific CD8+ T cells preferentially recognize heavily infected cells. Am J Respir Crit Care Med. 2003;168:1346–1352.
    1. Sidney J, Southwood S, Oseroff C, Del Guercio MF, Sette A, et al. Current protocols in immunology. New York: John Wiley & Sons, Inc; 1999. Unit 18.3: Measurement of MHC/peptide interactions by gel filtration.
    1. Lewinsohn DM, Tydeman IS, Frieder M, Grotzke JE, Lines RA, et al. High resolution radiographic and fine immunologic definition of TB disease progression in the rhesus macaque. Microbes Infect. 2006;8:2587–2598.

Source: PubMed

3
Suscribir