#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

T-bet and Eomes Are Differentially Linked to the Exhausted Phenotype of CD8+ T Cells in HIV Infection


CD8+ T cells display numerous traits of severe dysfunction in both treated and untreated HIV infection. Previous studies have demonstrated that HIV-specific CD8+ T cells in most individuals possess poor polyfunctionality, and an immature/skewed maturation phenotype. However, it remains unclear which transcriptional programming governs the regulation of CD8+ T cell differentiation and exhaustion in HIV infection. T-bet and Eomes represent two key transcription factors for CD8+ T cell differentiation and function, but surprisingly little is known about their influence of effector immunity following chronic viral infections in humans. In this study, we demonstrate that HIV-specific CD8+ T cells possess highly elevated levels of Eomes, but low T-bet expression. This differential relationship is linked to the up-regulation of several inhibitory receptors, impaired functional characteristics and a transitional memory differentiation phenotype for virus-specific CD8+ T cells. Importantly, these characteristics of HIV-specific CD8+ T cells remained stable despite suppressive ART for many years. These results implicate that reinvigoration of these cells might fail to elicit efficient responses to eradicate the viral reservoir.


Vyšlo v časopise: T-bet and Eomes Are Differentially Linked to the Exhausted Phenotype of CD8+ T Cells in HIV Infection. PLoS Pathog 10(7): e32767. doi:10.1371/journal.ppat.1004251
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1004251

Souhrn

CD8+ T cells display numerous traits of severe dysfunction in both treated and untreated HIV infection. Previous studies have demonstrated that HIV-specific CD8+ T cells in most individuals possess poor polyfunctionality, and an immature/skewed maturation phenotype. However, it remains unclear which transcriptional programming governs the regulation of CD8+ T cell differentiation and exhaustion in HIV infection. T-bet and Eomes represent two key transcription factors for CD8+ T cell differentiation and function, but surprisingly little is known about their influence of effector immunity following chronic viral infections in humans. In this study, we demonstrate that HIV-specific CD8+ T cells possess highly elevated levels of Eomes, but low T-bet expression. This differential relationship is linked to the up-regulation of several inhibitory receptors, impaired functional characteristics and a transitional memory differentiation phenotype for virus-specific CD8+ T cells. Importantly, these characteristics of HIV-specific CD8+ T cells remained stable despite suppressive ART for many years. These results implicate that reinvigoration of these cells might fail to elicit efficient responses to eradicate the viral reservoir.


Zdroje

1. IntlekoferAM, TakemotoN, KaoC, BanerjeeA, SchambachF, et al. (2007) Requirement for T-bet in the aberrant differentiation of unhelped memory CD8+ T cells. The Journal of experimental medicine 204: 2015–2021.

2. IntlekoferAM, TakemotoN, WherryEJ, LongworthSA, NorthrupJT, et al. (2005) Effector and memory CD8+ T cell fate coupled by T-bet and eomesodermin. Nature immunology 6: 1236–1244.

3. PearceEL, MullenAC, MartinsGA, KrawczykCM, HutchinsAS, et al. (2003) Control of effector CD8+ T cell function by the transcription factor Eomesodermin. Science 302: 1041–1043.

4. BanerjeeA, GordonSM, IntlekoferAM, PaleyMA, MooneyEC, et al. (2010) Cutting edge: The transcription factor eomesodermin enables CD8+ T cells to compete for the memory cell niche. Journal of immunology 185: 4988–4992.

5. JoshiNS, CuiW, ChandeleA, LeeHK, UrsoDR, et al. (2007) Inflammation directs memory precursor and short-lived effector CD8(+) T cell fates via the graded expression of T-bet transcription factor. Immunity 27: 281–295.

6. JoshiNS, CuiW, DominguezCX, ChenJH, HandTW, et al. (2011) Increased numbers of preexisting memory CD8 T cells and decreased T-bet expression can restrain terminal differentiation of secondary effector and memory CD8 T cells. Journal of immunology 187: 4068–4076.

7. ZhouX, YuS, ZhaoDM, HartyJT, BadovinacVP, et al. (2010) Differentiation and persistence of memory CD8(+) T cells depend on T cell factor 1. Immunity 33: 229–240.

8. KaechSM, CuiW (2012) Transcriptional control of effector and memory CD8+ T cell differentiation. Nature reviews Immunology 12: 749–761.

9. WherryEJ, BlattmanJN, Murali-KrishnaK, van der MostR, AhmedR (2003) Viral persistence alters CD8 T-cell immunodominance and tissue distribution and results in distinct stages of functional impairment. Journal of virology 77: 4911–4927.

10. ZajacAJ, BlattmanJN, Murali-KrishnaK, SourdiveDJ, SureshM, et al. (1998) Viral immune evasion due to persistence of activated T cells without effector function. The Journal of experimental medicine 188: 2205–2213.

11. BarberDL, WherryEJ, MasopustD, ZhuB, AllisonJP, et al. (2006) Restoring function in exhausted CD8 T cells during chronic viral infection. Nature 439: 682–687.

12. BlackburnSD, ShinH, HainingWN, ZouT, WorkmanCJ, et al. (2009) Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nature immunology 10: 29–37.

13. TrautmannL, JanbazianL, ChomontN, SaidEA, GimmigS, et al. (2006) Upregulation of PD-1 expression on HIV-specific CD8+ T cells leads to reversible immune dysfunction. Nature medicine 12: 1198–1202.

14. DayCL, KaufmannDE, KiepielaP, BrownJA, MoodleyES, et al. (2006) PD-1 expression on HIV-specific T cells is associated with T-cell exhaustion and disease progression. Nature 443: 350–354.

15. PetrovasC, CasazzaJP, BrenchleyJM, PriceDA, GostickE, et al. (2006) PD-1 is a regulator of virus-specific CD8+ T cell survival in HIV infection. The Journal of experimental medicine 203: 2281–2292.

16. YamamotoT, PriceDA, CasazzaJP, FerrariG, NasonM, et al. (2011) Surface expression patterns of negative regulatory molecules identify determinants of virus-specific CD8+ T-cell exhaustion in HIV infection. Blood 117: 4805–4815.

17. UrbaniS, AmadeiB, TolaD, MassariM, SchivazappaS, et al. (2006) PD-1 expression in acute hepatitis C virus (HCV) infection is associated with HCV-specific CD8 exhaustion. Journal of virology 80: 11398–11403.

18. RadziewiczH, IbegbuCC, FernandezML, WorkowskiKA, ObideenK, et al. (2007) Liver-infiltrating lymphocytes in chronic human hepatitis C virus infection display an exhausted phenotype with high levels of PD-1 and low levels of CD127 expression. Journal of virology 81: 2545–2553.

19. PennaA, PilliM, ZerbiniA, OrlandiniA, MezzadriS, et al. (2007) Dysfunction and functional restoration of HCV-specific CD8 responses in chronic hepatitis C virus infection. Hepatology 45: 588–601.

20. KroyDC, CiuffredaD, CooperiderJH, TomlinsonM, HauckGD, et al. (2013) Liver Environment and HCV Replication Affect Human T-Cell Phenotype and Expression of Inhibitory Receptors. Gastroenterology 146: 550–61.

21. BoniC, FisicaroP, ValdattaC, AmadeiB, Di VincenzoP, et al. (2007) Characterization of hepatitis B virus (HBV)-specific T-cell dysfunction in chronic HBV infection. Journal of virology 81: 4215–4225.

22. PengG, LiS, WuW, TanX, ChenY, et al. (2008) PD-1 upregulation is associated with HBV-specific T cell dysfunction in chronic hepatitis B patients. Molecular immunology 45: 963–970.

23. RaziorrouhB, SchrautW, GerlachT, NowackD, GrunerNH, et al. (2010) The immunoregulatory role of CD244 in chronic hepatitis B infection and its inhibitory potential on virus-specific CD8+ T-cell function. Hepatology 52: 1934–1947.

24. AlmeidaJR, PriceDA, PapagnoL, ArkoubZA, SauceD, et al. (2007) Superior control of HIV-1 replication by CD8+ T cells is reflected by their avidity, polyfunctionality, and clonal turnover. J Exp Med 204: 2473–2485.

25. BettsMR, NasonMC, WestSM, De RosaSC, MiguelesSA, et al. (2006) HIV nonprogressors preferentially maintain highly functional HIV-specific CD8+ T cells. Blood 107: 4781–4789.

26. AppayV, DunbarPR, CallanM, KlenermanP, GillespieGM, et al. (2002) Memory CD8+ T cells vary in differentiation phenotype in different persistent virus infections. Nature medicine 8: 379–385.

27. ChampagneP, OggGS, KingAS, KnabenhansC, EllefsenK, et al. (2001) Skewed maturation of memory HIV-specific CD8 T lymphocytes. Nature 410: 106–111.

28. DoeringTA, CrawfordA, AngelosantoJM, PaleyMA, ZieglerCG, et al. (2012) Network analysis reveals centrally connected genes and pathways involved in CD8+ T cell exhaustion versus memory. Immunity 37: 1130–1144.

29. QuigleyM, PereyraF, NilssonB, PorichisF, FonsecaC, et al. (2010) Transcriptional analysis of HIV-specific CD8+ T cells shows that PD-1 inhibits T cell function by upregulating BATF. Nature medicine 16: 1147–1151.

30. PeretzY, HeZ, ShiY, Yassine-DiabB, GouletJP, et al. (2012) CD160 and PD-1 co-expression on HIV-specific CD8 T cells defines a subset with advanced dysfunction. PLoS pathogens 8: e1002840.

31. PaleyMA, KroyDC, OdorizziPM, JohnnidisJB, DolfiDV, et al. (2012) Progenitor and terminal subsets of CD8+ T cells cooperate to contain chronic viral infection. Science 338: 1220–1225.

32. KaoC, OestreichKJ, PaleyMA, CrawfordA, AngelosantoJM, et al. (2011) Transcription factor T-bet represses expression of the inhibitory receptor PD-1 and sustains virus-specific CD8+ T cell responses during chronic infection. Nature immunology 12: 663–671.

33. HerspergerAR, MartinJN, ShinLY, ShethPM, KovacsCM, et al. (2011) Increased HIV-specific CD8+ T-cell cytotoxic potential in HIV elite controllers is associated with T-bet expression. Blood 117: 3799–3808.

34. BettsMR, BrenchleyJM, PriceDA, De RosaSC, DouekDC, et al. (2003) Sensitive and viable identification of antigen-specific CD8+ T cells by a flow cytometric assay for degranulation. Journal of immunological methods 281: 65–78.

35. BuggertM, NorstromMM, SalemiM, HechtFM, KarlssonAC (2014) Functional Avidity and IL-2/Perforin Production Is Linked to the Emergence of Mutations within HLA-B*5701-Restricted Epitopes and HIV-1 Disease Progression. Journal of immunology 192: 4685–4696.

36. NorstromMM, BuggertM, TauriainenJ, HartogensisW, ProsperiMC, et al. (2012) Combination of immune and viral factors distinguishes low-risk versus high-risk HIV-1 disease progression in HLA-B*5701 subjects. Journal of virology 86: 9802–9816.

37. R Core Team (2012). R: A language and environment for statistical computing. R Foundation for Statistical Computing V, Austria. ISBN 3-900051-07-0, URL http://www.R-project.org/.

38. RoedererM, NozziJL, NasonMC (2011) SPICE: exploration and analysis of post-cytometric complex multivariate datasets. Cytometry Part A : the journal of the International Society for Analytical Cytology 79: 167–174.

39. PorichisF, KwonDS, ZupkoskyJ, TigheDP, McMullenA, et al. (2011) Responsiveness of HIV-specific CD4 T cells to PD-1 blockade. Blood 118: 965–974.

40. JonesRB, NdhlovuLC, BarbourJD, ShethPM, JhaAR, et al. (2008) Tim-3 expression defines a novel population of dysfunctional T cells with highly elevated frequencies in progressive HIV-1 infection. The Journal of experimental medicine 205: 2763–2779.

41. SmithC, ElhassenD, GrasS, WynnKK, DasariV, et al. (2012) Endogenous antigen presentation impacts on T-box transcription factor expression and functional maturation of CD8+ T cells. Blood 120: 3237–3245.

42. BuggertM, NorstromMM, CzarneckiC, TupinE, LuoM, et al. (2012) Characterization of HIV-specific CD4+ T cell responses against peptides selected with broad population and pathogen coverage. PloS one 7: e39874.

43. HarariA, EndersFB, CelleraiC, BartPA, PantaleoG (2009) Distinct profiles of cytotoxic granules in memory CD8 T cells correlate with function, differentiation stage, and antigen exposure. Journal of virology 83: 2862–2871.

44. MakedonasG, HutnickN, HaneyD, AmickAC, GardnerJ, et al. (2010) Perforin and IL-2 upregulation define qualitative differences among highly functional virus-specific human CD8 T cells. PLoS pathogens 6: e1000798.

45. PerezCL, LarsenMV, GustafssonR, NorstromMM, AtlasA, et al. (2008) Broadly immunogenic HLA class I supertype-restricted elite CTL epitopes recognized in a diverse population infected with different HIV-1 subtypes. Journal of immunology 180: 5092–5100.

46. BuggertM, FrederiksenJ, NoyanK, SvardJ, BarqashoB, et al. (2014) Multiparametric bioinformatics distinguish the CD4/CD8 ratio as a suitable laboratory predictor of combined T cell pathogenesis in HIV infection. Journal of immunology 192: 2099–2108.

47. TakemotoN, IntlekoferAM, NorthrupJT, WherryEJ, ReinerSL (2006) Cutting Edge: IL-12 inversely regulates T-bet and eomesodermin expression during pathogen-induced CD8+ T cell differentiation. Journal of immunology 177: 7515–7519.

48. GrangeM, VerdeilG, ArnouxF, GriffonA, SpicugliaS, et al. (2013) Active STAT5 regulates T-bet and eomesodermin expression in CD8 T cells and imprints a T-bet-dependent Tc1 program with repressed IL-6/TGF-beta1 signaling. Journal of immunology 191: 3712–3724.

49. DemersKR, ReuterMA, BettsMR (2013) CD8(+) T-cell effector function and transcriptional regulation during HIV pathogenesis. Immunological reviews 254: 190–206.

50. McLaneLM, BanerjeePP, CosmaGL, MakedonasG, WherryEJ, et al. (2013) Differential localization of T-bet and Eomes in CD8 T cell memory populations. Journal of immunology 190: 3207–3215.

51. Ribeiro-dos-SantosP, TurnbullEL, MonteiroM, LegrandA, ConrodK, et al. (2012) Chronic HIV infection affects the expression of the 2 transcription factors required for CD8 T-cell differentiation into cytolytic effectors. Blood 119: 4928–4938.

52. MiguelesSA, OsborneCM, RoyceC, ComptonAA, JoshiRP, et al. (2008) Lytic granule loading of CD8+ T cells is required for HIV-infected cell elimination associated with immune control. Immunity 29: 1009–1021.

53. HerspergerAR, PereyraF, NasonM, DemersK, ShethP, et al. (2010) Perforin expression directly ex vivo by HIV-specific CD8 T-cells is a correlate of HIV elite control. PLoS pathogens 6: e1000917.

54. MiguelesSA, LaboricoAC, ShupertWL, SabbaghianMS, RabinR, et al. (2002) HIV-specific CD8+ T cell proliferation is coupled to perforin expression and is maintained in nonprogressors. Nature immunology 3: 1061–1068.

55. WolintP, BettsMR, KoupRA, OxeniusA (2004) Immediate cytotoxicity but not degranulation distinguishes effector and memory subsets of CD8+ T cells. The Journal of experimental medicine 199: 925–936.

56. JennerRG, TownsendMJ, JacksonI, SunK, BouwmanRD, et al. (2009) The transcription factors T-bet and GATA-3 control alternative pathways of T-cell differentiation through a shared set of target genes. Proceedings of the National Academy of Sciences of the United States of America 106: 17876–17881.

57. MuellerSN, AhmedR (2009) High antigen levels are the cause of T cell exhaustion during chronic viral infection. Proceedings of the National Academy of Sciences of the United States of America 106: 8623–8628.

58. WhittallT, PetersB, RahmanD, KingsleyCI, VaughanR, et al. (2011) Immunogenic and tolerogenic signatures in human immunodeficiency virus (HIV)-infected controllers compared with progressors and a conversion strategy of virus control. Clinical and experimental immunology 166: 208–217.

59. MiguelesSA, WeeksKA, NouE, BerkleyAM, RoodJE, et al. (2009) Defective human immunodeficiency virus-specific CD8+ T-cell polyfunctionality, proliferation, and cytotoxicity are not restored by antiretroviral therapy. Journal of virology 83: 11876–11889.

60. TrautmannL, Mbitikon-KoboFM, GouletJP, PeretzY, ShiY, et al. (2012) Profound metabolic, functional, and cytolytic differences characterize HIV-specific CD8 T cells in primary and chronic HIV infection. Blood 120: 3466–3477.

61. HasleyRB, HongC, LiW, FriesenT, NakamuraY, et al. (2013) HIV immune activation drives increased Eomes expression in memory CD8 T cells in association with transcriptional downregulation of CD127. AIDS 27: 1867–1877.

62. WherryEJ, HaSJ, KaechSM, HainingWN, SarkarS, et al. (2007) Molecular signature of CD8+ T cell exhaustion during chronic viral infection. Immunity 27: 670–684.

63. PetrovasC, ChaonB, AmbrozakDR, PriceDA, MelenhorstJJ, et al. (2009) Differential association of programmed death-1 and CD57 with ex vivo survival of CD8+ T cells in HIV infection. Journal of immunology 183: 1120–1132.

64. UtzschneiderDT, LegatA, Fuertes MarracoSA, CarrieL, LuescherI, et al. (2013) T cells maintain an exhausted phenotype after antigen withdrawal and population reexpansion. Nature immunology 14: 603–610.

65. YoungbloodB, NotoA, PorichisF, AkondyRS, NdhlovuZM, et al. (2013) Cutting edge: Prolonged exposure to HIV reinforces a poised epigenetic program for PD-1 expression in virus-specific CD8 T cells. Journal of immunology 191: 540–544.

66. HansenSG, FordJC, LewisMS, VenturaAB, HughesCM, et al. (2011) Profound early control of highly pathogenic SIV by an effector memory T-cell vaccine. Nature 473: 523–527.

67. HansenSG, PiatakMJr, VenturaAB, HughesCM, GilbrideRM, et al. (2013) Immune clearance of highly pathogenic SIV infection. Nature 502: 100–104.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2014 Číslo 7
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#