#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Interferon-alpha Subtype 11 Activates NK Cells and Enables Control of Retroviral Infection


The innate immune response mediated by cells such as natural killer (NK) cells is critical for the rapid containment of virus replication and spread during acute infection. Here, we show that subtype 11 of the type I interferon (IFN) family greatly potentiates the antiviral activity of NK cells during retroviral infection. Treatment of mice with IFN-α11 during Friend retrovirus infection (FV) significantly reduced viral loads and resulted in long-term protection from virus-induced leukemia. The effect of IFN-α11 on NK cells was direct and signaled through the type I IFN receptor. Furthermore, IFN-α11-mediated activation of NK cells enabled cytolytic killing of FV-infected target cells via the exocytosis pathway. Depletion and adoptive transfer experiments illustrated that NK cells played a major role in successful IFN-α11 therapy. Additional experiments with Mouse Cytomegalovirus infections demonstrated that the therapeutic effect of IFN-α11 is not restricted to retroviruses. The type I IFN subtypes 2 and 5, which bind the same receptor as IFN-α11, did not elicit similar antiviral effects. These results demonstrate a unique and subtype-specific activation of NK cells by IFN-α11.


Vyšlo v časopise: Interferon-alpha Subtype 11 Activates NK Cells and Enables Control of Retroviral Infection. PLoS Pathog 8(8): e32767. doi:10.1371/journal.ppat.1002868
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1002868

Souhrn

The innate immune response mediated by cells such as natural killer (NK) cells is critical for the rapid containment of virus replication and spread during acute infection. Here, we show that subtype 11 of the type I interferon (IFN) family greatly potentiates the antiviral activity of NK cells during retroviral infection. Treatment of mice with IFN-α11 during Friend retrovirus infection (FV) significantly reduced viral loads and resulted in long-term protection from virus-induced leukemia. The effect of IFN-α11 on NK cells was direct and signaled through the type I IFN receptor. Furthermore, IFN-α11-mediated activation of NK cells enabled cytolytic killing of FV-infected target cells via the exocytosis pathway. Depletion and adoptive transfer experiments illustrated that NK cells played a major role in successful IFN-α11 therapy. Additional experiments with Mouse Cytomegalovirus infections demonstrated that the therapeutic effect of IFN-α11 is not restricted to retroviruses. The type I IFN subtypes 2 and 5, which bind the same receptor as IFN-α11, did not elicit similar antiviral effects. These results demonstrate a unique and subtype-specific activation of NK cells by IFN-α11.


Zdroje

1. TrinchieriG (1989) Biology of natural killer cells. Adv Immunol 47: 187–376.

2. VivierE, NunesJA, VelyF (2004) Natural killer cell signaling pathways. Science 306: 1517–1519.

3. BironCA, ByronKS, SullivanJL (1989) Severe herpesvirus infections in an adolescent without natural killer cells. N Engl J Med 320: 1731–1735.

4. VivierE, TomaselloE, BaratinM, WalzerT, UgoliniS (2008) Functions of natural killer cells. Nat Immunol 9: 503–510.

5. MartinMP, QiY, GaoX, YamadaE, MartinJN, et al. (2007) Innate partnership of HLA-B and KIR3DL1 subtypes against HIV-1. Nat Genet 39: 733–740.

6. FellayJ, ShiannaKV, GeD, ColomboS, LedergerberB, et al. (2007) A whole-genome association study of major determinants for host control of HIV-1. Science 317: 944–947.

7. CerboniC, NeriF, CasartelliN, ZingoniA, CosmanD, et al. (2007) Human immunodeficiency virus 1 Nef protein downmodulates the ligands of the activating receptor NKG2D and inhibits natural killer cell-mediated cytotoxicity. J Gen Virol 88: 242–250.

8. OgawaT, Tsuji-KawaharaS, YuasaT, KinoshitaS, ChikaishiT, et al. (2011) Natural killer cells recognize friend retrovirus-infected erythroid progenitor cells through NKG2D-RAE-1 interactions In Vivo. J Virol 85: 5423–5435.

9. ZelinskyyG, BalkowS, SchimmerS, WernerT, SimonMM, et al. (2007) The level of friend retrovirus replication determines the cytolytic pathway of CD8+ T-cell-mediated pathogen control. J Virol 81: 11881–11890.

10. FerlazzoG, PackM, ThomasD, PaludanC, SchmidD, et al. (2004) Distinct roles of IL-12 and IL-15 in human natural killer cell activation by dendritic cells from secondary lymphoid organs. Proc Natl Acad Sci U S A 101: 16606–16611.

11. TrinchieriG, SantoliD, GranatoD, PerussiaB (1981) Antagonistic effects of interferons on the cytotoxicity mediated by natural killer cells. Fed Proc 40: 2705–2710.

12. StaceyAR, NorrisPJ, QinL, HaygreenEA, TaylorE, et al. (2009) Induction of a striking systemic cytokine cascade prior to peak viremia in acute human immunodeficiency virus type 1 infection, in contrast to more modest and delayed responses in acute hepatitis B and C virus infections. J Virol 83: 3719–3733.

13. GerlachN, SchimmerS, WeissS, KalinkeU, DittmerU (2007) Effects of Type I Interferons on Friend Retrovirus Infection (Erratum). J Virol 81: 6160.

14. GerlachN, SchimmerS, WeissS, KalinkeU, DittmerU (2006) Effects of type I interferons on Friend retrovirus infection. J Virol 80: 3438–3444.

15. AhlenstielG, EdlichB, HogdalLJ, RotmanY, NoureddinM, et al. (2011) Early changes in natural killer cell function indicate virologic response to interferon therapy for hepatitis C. Gastroenterology. 141: 1231–1239.

16. StegmannKA, BjorkstromNK, VeberH, CiesekS, RieseP, et al. (2010) Interferon-alpha-induced TRAIL on natural killer cells is associated with control of hepatitis C virus infection. Gastroenterology 138: 1885–1897.

17. MiyazawaM, NishioJ, ChesebroB (1992) Protection against Friend retrovirus-induced leukemia by recombinant vaccinia viruses expressing the gag gene. J Virol 66: 4497–4507.

18. van PeschV, LanayaH, RenauldJC, MichielsT (2004) Characterization of the murine alpha interferon gene family. J Virol 78: 8219–8228.

19. HardyMP, OwczarekCM, JermiinLS, EjdebackM, HertzogPJ (2004) Characterization of the type I interferon locus and identification of novel genes. Genomics 84: 331–345.

20. AguetM, GrobkeM, DreidingP (1984) Various human interferon alpha subclasses cross-react with common receptors: their binding affinities correlate with their specific biological activities. Virology 132: 211–216.

21. JaksE, GavutisM, UzeG, MartalJ, PiehlerJ (2007) Differential receptor subunit affinities of type I interferons govern differential signal activation. J Mol Biol 366: 525–539.

22. CullVS, TilbrookPA, BartlettEJ, BrekaloNL, JamesCM (2003) Type I interferon differential therapy for erythroleukemia: specificity of STAT activation. Blood 101: 2727–2735.

23. MollHP, MaierT, ZommerA, LavoieT, BrostjanC (2011) The differential activity of interferon-alpha subtypes is consistent among distinct target genes and cell types. Cytokine 53: 52–59.

24. AustinBA, JamesC, SilvermanRH, CarrDJ (2005) Critical role for the oligoadenylate synthetase/RNase L pathway in response to IFN-beta during acute ocular herpes simplex virus type 1 infection. J Immunol 175: 1100–1106.

25. HarleP, CullV, AgbagaMP, SilvermanR, WilliamsBR, et al. (2002) Differential effect of murine alpha/beta interferon transgenes on antagonization of herpes simplex virus type 1 replication. J Virol 76: 6558–6567.

26. JamesCM, AbdadMY, MansfieldJP, JacobsenHK, VindAR, et al. (2007) Differential activities of alpha/beta IFN subtypes against influenza virus in vivo and enhancement of specific immune responses in DNA vaccinated mice expressing haemagglutinin and nucleoprotein. Vaccine 25: 1856–1867.

27. YeowWS, LaiCM, BeilharzMW (1997) The in vivo expression patterns of individual type I interferon genes in murine cytomegalovirus infections. Antiviral Res 34: 17–26.

28. MullerU, SteinhoffU, ReisLF, HemmiS, PavlovicJ, et al. (1994) Functional role of type I and type II interferons in antiviral defense. Science 264: 1918–1921.

29. HeuselJW, WesselschmidtRL, ShrestaS, RussellJH, LeyTJ (1994) Cytotoxic lymphocytes require granzyme B for the rapid induction of DNA fragmentation and apoptosis in allogeneic target cells. Cell 76: 977–987.

30. RobertsonSJ, AmmannCG, MesserRJ, CarmodyAB, MyersL, et al. (2008) Suppression of acute anti-friend virus CD8+ T-cell responses by coinfection with lactate dehydrogenase-elevating virus. J Virol 82: 408–418.

31. BukowskiJF, WodaBA, HabuS, OkumuraK, WelshRM (1983) Natural killer cell depletion enhances virus synthesis and virus-induced hepatitis in vivo. J Immunol 131: 1531–1538.

32. GerlachN, GibbertK, AlterC, NairS, ZelinskyyG, et al. (2009) Anti-retroviral effects of type I IFN subtypes in vivo. Eur J Immunol 39: 136–146.

33. Bollati-FogolinM, MullerW (2005) Virus free, cell-based assay for the quantification of murine type I interferons. J Immunol Methods 306: 169–175.

34. DittmerU, BrooksDM, HasenkrugKJ (1998) Characterization of a live-attenuated retroviral vaccine demonstrates protection via immune mechanisms. J Virol 72: 6554–6558.

35. ChenHD, FraireAE, JorisI, WelshRM, SelinLK (2003) Specific history of heterologous virus infections determines anti-viral immunity and immunopathology in the lung. Am J Pathol 163: 1341–1355.

36. ChenW, QinH, ChesebroB, CheeverMA (1996) Identification of a gag-encoded cytotoxic T-lymphocyte epitope from FBL-3 leukemia shared by Friend, Moloney, and Rauscher murine leukemia virus-induced tumors. J Virol 70: 7773–7782.

37. ShimizuT, UenishiH, TeramuraY, IwashiroM, KuribayashiK, et al. (1994) Fine structure of a virus-encoded helper T-cell epitope expressed on FBL-3 tumor cells. J Virol 68: 7704–7708.

38. ZelinskyyG, KraftAR, SchimmerS, ArndtT, DittmerU (2006) Kinetics of CD8+ effector T cell responses and induced CD4+ regulatory T cell responses during Friend retrovirus infection. Eur J Immunol 36: 2658–2670.

39. ZelinskyyG, DietzeKK, HuseckenYP, SchimmerS, NairS, et al. (2009) The regulatory T-cell response during acute retroviral infection is locally defined and controls the magnitude and duration of the virus-specific cytotoxic T-cell response. Blood 114: 3199–3207.

40. CikesM, FribergSJr, KleinG (1973) Progressive loss of H-2 antigens with concomitant increase of cell-surface antigen(s) determined by Moloney leukemia virus in cultured murine lymphomas. J Natl Cancer Inst 50: 347–362.

41. McCoyJL, FeferA, GlynnJP (1967) Influence of infectious virus on the induction of transplantation resistance in the Friend tumor system. Cancer Res 27: 2267–2271.

42. KataokaT, ShinoharaN, TakayamaH, TakakuK, KondoS, et al. (1996) Concanamycin A, a powerful tool for characterization and estimation of contribution of perforin- and Fas-based lytic pathways in cell-mediated cytotoxicity. J Immunol 156: 3678–3686.

43. NeumannAU, LamNP, DahariH, GretchDR, WileyTE, et al. (1998) Hepatitis C viral dynamics in vivo and the antiviral efficacy of interferon-alpha therapy. Science 282: 103–107.

44. FeldJJ, HoofnagleJH (2005) Mechanism of action of interferon and ribavirin in treatment of hepatitis C. Nature 436: 967–972.

45. HerrmannE, LeeJH, MarinosG, ModiM, ZeuzemS (2003) Effect of ribavirin on hepatitis C viral kinetics in patients treated with pegylated interferon. Hepatology 37: 1351–1358.

46. GrahamGJ (2009) D6 and the atypical chemokine receptor family: novel regulators of immune and inflammatory processes. Eur J Immunol 39: 342–351.

47. ZidarDA, ViolinJD, WhalenEJ, LefkowitzRJ (2009) Selective engagement of G protein coupled receptor kinases (GRKs) encodes distinct functions of biased ligands. Proc Natl Acad Sci U S A 106: 9649–9654.

48. GrumbachIM, FishEN, UddinS, MajchrzakB, ColamoniciOR, et al. (1999) Activation of the Jak-Stat pathway in cells that exhibit selective sensitivity to the antiviral effects of IFN-beta compared with IFN-alpha. J Interferon Cytokine Res 19: 797–801.

49. DerSD, ZhouA, WilliamsBR, SilvermanRH (1998) Identification of genes differentially regulated by interferon alpha, beta, or gamma using oligonucleotide arrays. Proc Natl Acad Sci U S A 95: 15623–15628.

50. LeamanDW, Chawla-SarkarM, JacobsB, VyasK, SunY, et al. (2003) Novel growth and death related interferon-stimulated genes (ISGs) in melanoma: greater potency of IFN-beta compared with IFN-alpha2. J Interferon Cytokine Res 23: 745–756.

51. SeveraM, RemoliME, GiacominiE, RagimbeauJ, LandeR, et al. (2006) Differential responsiveness to IFN-alpha and IFN-beta of human mature DC through modulation of IFNAR expression. J Leukoc Biol 79: 1286–1294.

52. BironCA, NguyenKB, PienGC, CousensLP, Salazar-MatherTP (1999) Natural killer cells in antiviral defense: function and regulation by innate cytokines. Annu Rev Immunol 17: 189–220.

53. EdwardsBS, HawkinsMJ, BordenEC (1984) Comparative in vivo and in vitro activation of human natural killer cells by two recombinant alpha-interferons differing in antiviral activity. Cancer Res 44: 3135–3139.

54. OrtaldoJR, HerbermanRB, HarveyC, OsheroffP, PanYC, et al. (1984) A species of human alpha interferon that lacks the ability to boost human natural killer activity. Proc Natl Acad Sci U S A 81: 4926–4929.

55. VerhagenA, MackayIR, RowleyM, TymmsM (1990) Comparison of augmentation of human natural killer cell cytotoxicity by interferon-alpha subtypes. Nat Immun Cell Growth Regul 9: 325–333.

56. IwanamiN, NiwaA, YasutomiY, TabataN, MiyazawaM (2001) Role of natural killer cells in resistance against friend retrovirus-induced leukemia. J Virol 75: 3152–3163.

57. ZelinskyyG, BalkowS, SchimmerS, SchepersK, SimonMM, et al. (2004) Independent roles of perforin, granzymes, and Fas in the control of Friend retrovirus infection. Virology 330: 365–374.

58. JansDA, JansP, BriggsLJ, SuttonV, TrapaniJA (1996) Nuclear transport of granzyme B (fragmentin-2). Dependence of perforin in vivo and cytosolic factors in vitro. J Biol Chem 271: 30781–30789.

59. SimonHG, FruthU, KramerMD, SimonMM (1987) A secretable serine proteinase with highly restricted specificity from cytolytic T lymphocytes inactivates retrovirus-associated reverse transcriptase. FEBS Lett 223: 352–360.

60. SatoK, HidaS, TakayanagiH, YokochiT, KayagakiN, et al. (2001) Antiviral response by natural killer cells through TRAIL gene induction by IFN-alpha/beta. Eur J Immunol 31: 3138–3146.

61. KayagakiN, YamaguchiN, NakayamaM, EtoH, OkumuraK, et al. (1999) Type I interferons (IFNs) regulate tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) expression on human T cells: A novel mechanism for the antitumor effects of type I IFNs. J Exp Med 189: 1451–1460.

62. KayagakiN, YamaguchiN, NakayamaM, TakedaK, AkibaH, et al. (1999) Expression and function of TNF-related apoptosis-inducing ligand on murine activated NK cells. J Immunol 163: 1906–1913.

63. WalczakH, MillerRE, AriailK, GliniakB, GriffithTS, et al. (1999) Tumoricidal activity of tumor necrosis factor-related apoptosis-inducing ligand in vivo. Nat Med 5: 157–163.

64. ZamaiL, AhmadM, BennettIM, AzzoniL, AlnemriES, et al. (1998) Natural killer (NK) cell-mediated cytotoxicity: differential use of TRAIL and Fas ligand by immature and mature primary human NK cells. J Exp Med 188: 2375–2380.

65. AltfeldM, FaddaL, FrletaD, BhardwajN (2011) DCs and NK cells: critical effectors in the immune response to HIV-1. Nat Rev Immunol 11: 176–186.

66. HosmalinA, LebonP (2006) Type I interferon production in HIV-infected patients. J Leukoc Biol 80: 984–993.

67. HarmanAN, LaiJ, TurvilleS, SamarajiwaS, GrayL, et al. (2011) HIV infection of dendritic cells subverts the IFN induction pathway via IRF-1 and inhibits type 1 IFN production. Blood 118: 298–308.

68. YanN, Regalado-MagdosAD, StiggelboutB, Lee-KirschMA, LiebermanJ (2010) The cytosolic exonuclease TREX1 inhibits the innate immune response to human immunodeficiency virus type 1. Nat Immunol 11: 1005–1013.

69. KamgaI, KahiS, DeveliogluL, LichtnerM, MaranonC, et al. (2005) Type I interferon production is profoundly and transiently impaired in primary HIV-1 infection. J Infect Dis 192: 303–310.

70. FeldmanS, SteinD, AmruteS, DennyT, GarciaZ, et al. (2001) Decreased interferon-alpha production in HIV-infected patients correlates with numerical and functional deficiencies in circulating type 2 dendritic cell precursors. Clin Immunol 101: 201–210.

71. KhatissianE, ToveyMG, CumontMC, MonceauxV, LebonP, et al. (1996) The relationship between the interferon alpha response and viral burden in primary SIV infection. AIDS Res Hum Retroviruses 12: 1273–1278.

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2012 Číslo 8
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#