#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Location of the CD8 T Cell Epitope within the Antigenic Precursor Determines Immunogenicity and Protection against the Parasite


CD8 T cells protect the host from disease caused by intracellular pathogens, such as the Toxoplasma gondii (T. gondii) protozoan parasite. Despite the complexity of the T. gondii proteome, CD8 T cell responses are restricted to only a small number of peptide epitopes derived from a limited set of antigenic precursors. This phenomenon is known as immunodominance and is key to effective vaccine design. However, the mechanisms that determine the immunogenicity and immunodominance hierarchy of parasite antigens are not well understood.

Here, using genetically modified parasites, we show that parasite burden is controlled by the immunodominant GRA6-specific CD8 T cell response but not by responses to the subdominant GRA4- and ROP7-derived epitopes. Remarkably, optimal processing and immunodominance were determined by the location of the peptide epitope at the C-terminus of the GRA6 antigenic precursor. In contrast, immunodominance could not be explained by the peptide affinity for the MHC I molecule or the frequency of T cell precursors in the naive animals. Our results reveal the molecular requirements for optimal presentation of an intracellular parasite antigen and for eliciting protective CD8 T cells.


Vyšlo v časopise: Location of the CD8 T Cell Epitope within the Antigenic Precursor Determines Immunogenicity and Protection against the Parasite. PLoS Pathog 9(6): e32767. doi:10.1371/journal.ppat.1003449
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1003449

Souhrn

CD8 T cells protect the host from disease caused by intracellular pathogens, such as the Toxoplasma gondii (T. gondii) protozoan parasite. Despite the complexity of the T. gondii proteome, CD8 T cell responses are restricted to only a small number of peptide epitopes derived from a limited set of antigenic precursors. This phenomenon is known as immunodominance and is key to effective vaccine design. However, the mechanisms that determine the immunogenicity and immunodominance hierarchy of parasite antigens are not well understood.

Here, using genetically modified parasites, we show that parasite burden is controlled by the immunodominant GRA6-specific CD8 T cell response but not by responses to the subdominant GRA4- and ROP7-derived epitopes. Remarkably, optimal processing and immunodominance were determined by the location of the peptide epitope at the C-terminus of the GRA6 antigenic precursor. In contrast, immunodominance could not be explained by the peptide affinity for the MHC I molecule or the frequency of T cell precursors in the naive animals. Our results reveal the molecular requirements for optimal presentation of an intracellular parasite antigen and for eliciting protective CD8 T cells.


Zdroje

1. van EndertP (2011) Providing ligands for MHC class I molecules. Cellular and molecular life sciences : CMLS 68: 1467–1469.

2. ChenW, McCluskeyJ (2006) Immunodominance and immunodomination: critical factors in developing effective CD8+ T-cell-based cancer vaccines. Adv Cancer Res 95: 203–247.

3. YewdellJW (2006) Confronting complexity: real-world immunodominance in antiviral CD8+ T cell responses. Immunity 25: 533–543.

4. TenzerS, WeeE, BurgevinA, Stewart-JonesG, FriisL, et al. (2009) Antigen processing influences HIV-specific cytotoxic T lymphocyte immunodominance. Nat Immunol 10: 636–646.

5. BlanchardN, KanasekiT, EscobarH, DelebecqueF, NagarajanNA, et al. (2010) Endoplasmic reticulum aminopeptidase associated with antigen processing defines the composition and structure of MHC class I peptide repertoire in normal and virus-infected cells. J Immunol 184: 3033–3042.

6. Le GallS, StamegnaP, WalkerBD (2007) Portable flanking sequences modulate CTL epitope processing. J Clin Invest 117: 3563–3575.

7. La GrutaNL, KedzierskaK, PangK, WebbyR, DavenportM, et al. (2006) A virus-specific CD8+ T cell immunodominance hierarchy determined by antigen dose and precursor frequencies. Proceedings of the National Academy of Sciences of the United States of America 103: 994–999.

8. CroweSR, TurnerSJ, MillerSC, RobertsAD, RappoloRA, et al. (2003) Differential antigen presentation regulates the changing patterns of CD8+ T cell immunodominance in primary and secondary influenza virus infections. J Exp Med 198: 399–410.

9. KotturiMF, ScottI, WolfeT, PetersB, SidneyJ, et al. (2008) Naive precursor frequencies and MHC binding rather than the degree of epitope diversity shape CD8+ T cell immunodominance. J Immunol 181: 2124–2133.

10. TanAC, La GrutaNL, ZengW, JacksonDC (2011) Precursor Frequency and Competition Dictate the HLA-A2-Restricted CD8+ T Cell Responses to Influenza A Infection and Vaccination in HLA-A2.1 Transgenic Mice. J Immunol 187: 1895–1902.

11. St LegerAJ, PetersB, SidneyJ, SetteA, HendricksRL (2011) Defining the herpes simplex virus-specific CD8+ T cell repertoire in C57BL/6 mice. J Immunol 186: 3927–3933.

12. La GrutaNL, RothwellWT, CukalacT, SwanNG, ValkenburgSA, et al. (2010) Primary CTL response magnitude in mice is determined by the extent of naive T cell recruitment and subsequent clonal expansion. J Clin Invest 120: 1885–1894.

13. DolanBP, BenninkJR, YewdellJW (2011) Translating DRiPs: progress in understanding viral and cellular sources of MHC class I peptide ligands. Cellular and molecular life sciences : CMLS 68: 1481–1489.

14. BlanchardN, ShastriN (2010) Cross-presentation of peptides from intracellular pathogens by MHC class I molecules. Annals of the New York Academy of Sciences 1183: 237–250.

15. KumarKA, SanoG, BoscardinS, NussenzweigRS, NussenzweigMC, et al. (2006) The circumsporozoite protein is an immunodominant protective antigen in irradiated sporozoites. Nature 444: 937–940.

16. MacHughND, WeirW, BurrellsA, LizundiaR, GrahamSP, et al. (2011) Extensive polymorphism and evidence of immune selection in a highly dominant antigen recognized by bovine CD8 T cells specific for Theileria annulata. Infect Immun 79: 2059–2069.

17. BlanchardN, GonzalezF, SchaefferM, JonckerNT, ChengT, et al. (2008) Immunodominant, protective response to the parasite Toxoplasma gondii requires antigen processing in the endoplasmic reticulum. Nat Immunol 9: 937–944.

18. MontoyaJG, LiesenfeldO (2004) Toxoplasmosis. Lancet 363: 1965–1976.

19. JohnsonJJ, RobertsCW, PopeC, RobertsF, KirisitsMJ, et al. (2002) In vitro correlates of Ld-restricted resistance to toxoplasmic encephalitis and their critical dependence on parasite strain. J Immunol 169: 966–973.

20. FrickelEM, SahooN, HoppJ, GubbelsMJ, CraverMP, et al. (2008) Parasite stage-specific recognition of endogenous Toxoplasma gondii-derived CD8+ T cell epitopes. J Infect Dis 198: 1625–1633.

21. RammenseeH, BachmannJ, EmmerichNP, BachorOA, StevanovicS (1999) SYFPEITHI: database for MHC ligands and peptide motifs. Immunogenetics 50: 213–219.

22. KamauE, MeehanT, LavineMD, ArrizabalagaG, Mustata WilsonG, et al. (2011) A novel benzodioxole-containing inhibitor of Toxoplasma gondii growth alters the parasite cell cycle. Antimicrobial agents and chemotherapy 55: 5438–5451.

23. MoonJJ, ChuHH, PepperM, McSorleySJ, JamesonSC, et al. (2007) Naive CD4(+) T cell frequency varies for different epitopes and predicts repertoire diversity and response magnitude. Immunity 27: 203–213.

24. ObarJJ, KhannaKM, LefrancoisL (2008) Endogenous naive CD8+ T cell precursor frequency regulates primary and memory responses to infection. Immunity 28: 859–869.

25. GendrinC, BittameA, MercierC, Cesbron-DelauwMF (2010) Post-translational membrane sorting of the Toxoplasma gondii GRA6 protein into the parasite-containing vacuole is driven by its N-terminal domain. Int J Parasitol 40: 1325–1334.

26. WilsonDC, GrotenbregGM, LiuK, ZhaoY, FrickelEM, et al. (2010) Differential regulation of effector- and central-memory responses to Toxoplasma gondii Infection by IL-12 revealed by tracking of Tgd057-specific CD8+ T cells. PLoS Pathog 6: e1000815.

27. GroverHS, BlanchardN, GonzalezF, ChanS, RobeyEA, et al. (2012) The Toxoplasma gondii Peptide AS15 Elicits CD4 T Cells That Can Control Parasite Burden. Infect Immun 80: 3279–3288.

28. NewbergMH, McEversKJ, GorgoneDA, LiftonMA, BaumeisterSH, et al. (2006) Immunodomination in the evolution of dominant epitope-specific CD8+ T lymphocyte responses in simian immunodeficiency virus-infected rhesus monkeys. J Immunol 176: 319–328.

29. TzelepisF, de AlencarBC, PenidoML, ClaserC, MachadoAV, et al. (2008) Infection with Trypanosoma cruzi restricts the repertoire of parasite-specific CD8+ T cells leading to immunodominance. J Immunol 180: 1737–1748.

30. WangX, KangH, KikuchiT, SuzukiY (2004) Gamma interferon production, but not perforin-mediated cytolytic activity, of T cells is required for prevention of toxoplasmic encephalitis in BALB/c mice genetically resistant to the disease. Infect Immun 72: 4432–4438.

31. DzierszinskiF, PepperM, StumhoferJS, LaRosaDF, WilsonEH, et al. (2007) Presentation of Toxoplasma gondii antigens via the endogenous major histocompatibility complex class I pathway in nonprofessional and professional antigen-presenting cells. Infect Immun 75: 5200–5209.

32. SaeijJP, CollerS, BoyleJP, JeromeME, WhiteMW, et al. (2007) Toxoplasma co-opts host gene expression by injection of a polymorphic kinase homologue. Nature 445: 324–327.

33. SteinfeldtT, Konen-WaismanS, TongL, PawlowskiN, LamkemeyerT, et al. (2010) Phosphorylation of Mouse Immunity-Related GTPase (IRG) Resistance Proteins Is an Evasion Strategy for Virulent Toxoplasma gondii. PLoS Biol 8: e1000576.

34. FentressSJ, BehnkeMS, DunayIR, MashayekhiM, RommereimLM, et al. (2010) Phosphorylation of Immunity-Related GTPases by a Toxoplasma gondii-Secreted Kinase Promotes Macrophage Survival and Virulence. Cell Host Microbe 8: 484–495.

35. RosowskiEE, LuD, JulienL, RoddaL, GaiserRA, et al. (2011) Strain-specific activation of the NF-kappaB pathway by GRA15, a novel Toxoplasma gondii dense granule protein. J Exp Med 208: 195–212.

36. SousaS, AjzenbergD, MarleM, AubertD, VillenaI, et al. (2009) Selection of polymorphic peptides from GRA6 and GRA7 sequences of Toxoplasma gondii strains to be used in serotyping. Clinical and vaccine immunology : CVI 16: 1158–1169.

37. PeyronF, LobryJR, MussetK, FerrandizJ, Gomez-MarinJE, et al. (2006) Serotyping of Toxoplasma gondii in chronically infected pregnant women: predominance of type II in Europe and types I and III in Colombia (South America). Microbes Infect 8: 2333–2340.

38. KongJT, GriggME, UyetakeL, ParmleyS, BoothroydJC (2003) Serotyping of Toxoplasma gondii infections in humans using synthetic peptides. J Infect Dis 187: 1484–1495.

39. JenkinsMK, MoonJJ (2012) The role of naive T cell precursor frequency and recruitment in dictating immune response magnitude. Journal of immunology 188: 4135–4140.

40. MoAX, van LelyveldSF, CraiuA, RockKL (2000) Sequences that flank subdominant and cryptic epitopes influence the proteolytic generation of MHC class I-presented peptides. J Immunol 164: 4003–4010.

41. Yellen-ShawAJ, WherryEJ, DuboisGC, EisenlohrLC (1997) Point mutation flanking a CTL epitope ablates in vitro and in vivo recognition of a full-length viral protein. J Immunol 158: 3227–3234.

42. ShastriN, SerwoldT, GonzalezF (1995) Presentation of endogenous peptide/MHC class I complexes is profoundly influenced by specific C-terminal flanking residues. Journal of immunology 155: 4339–4346.

43. MaX, SernaA, XuRH, SigalLJ (2009) The amino acid sequences flanking an antigenic determinant can strongly affect MHC class I cross-presentation without altering direct presentation. J Immunol 182: 4601–4607.

44. LabruyereE, LingnauM, MercierC, SibleyLD (1999) Differential membrane targeting of the secretory proteins GRA4 and GRA6 within the parasitophorous vacuole formed by Toxoplasma gondii. Mol Biochem Parasitol 102: 311–324.

45. BlanchardN, ShastriN (2010) Topological journey of parasite-derived antigens for presentation by MHC class I molecules. Trends in immunology 31: 414–421.

46. StarnbachMN, LoomisWP, OvendaleP, ReganD, HessB, et al. (2003) An inclusion membrane protein from Chlamydia trachomatis enters the MHC class I pathway and stimulates a CD8+ T cell response. J Immunol 171: 4742–4749.

47. CebrianI, VisentinG, BlanchardN, JouveM, BobardA, et al. (2011) Sec22b regulates phagosomal maturation and antigen crosspresentation by dendritic cells. Cell 147: 1355–1368.

48. GoldszmidRS, CoppensI, LevA, CasparP, MellmanI, et al. (2009) Host ER-parasitophorous vacuole interaction provides a route of entry for antigen cross-presentation in Toxoplasma gondii-infected dendritic cells. J Exp Med 206: 399–410.

49. KimY, YewdellJW, SetteA, PetersB (2013) Positional Bias of MHC Class I Restricted T-Cell Epitopes in Viral Antigens Is Likely due to a Bias in Conservation. PLoS computational biology 9: e1002884.

50. JohnB, HarrisTH, TaitED, WilsonEH, GreggB, et al. (2009) Dynamic Imaging of CD8(+) T cells and dendritic cells during infection with Toxoplasma gondii. PLoS pathogens 5: e1000505.

51. GubbelsMJ, StriepenB, ShastriN, TurkozM, RobeyEA (2005) Class I major histocompatibility complex presentation of antigens that escape from the parasitophorous vacuole of Toxoplasma gondii. Infect Immun 73: 703–711.

52. BoyleJP, SaeijJP, BoothroydJC (2007) Toxoplasma gondii: inconsistent dissemination patterns following oral infection in mice. Experimental parasitology 116: 302–305.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2013 Číslo 6
Najčítanejšie tento týždeň
Najčítanejšie v tomto čísle
Kurzy

Zvýšte si kvalifikáciu online z pohodlia domova

Získaná hemofilie - Povědomí o nemoci a její diagnostika
nový kurz

Eozinofilní granulomatóza s polyangiitidou
Autori: doc. MUDr. Martina Doubková, Ph.D.

Všetky kurzy
Prihlásenie
Zabudnuté heslo

Zadajte e-mailovú adresu, s ktorou ste vytvárali účet. Budú Vám na ňu zasielané informácie k nastaveniu nového hesla.

Prihlásenie

Nemáte účet?  Registrujte sa

#ADS_BOTTOM_SCRIPTS#