#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

IFNγ/IL-10 Co-producing Cells Dominate the CD4 Response to Malaria in Highly Exposed Children


Although evidence suggests that T cells are critical for immunity to malaria, reliable T cell correlates of exposure to and protection from malaria among children living in endemic areas are lacking. We used multiparameter flow cytometry to perform a detailed functional characterization of malaria-specific T cells in 78 four-year-old children enrolled in a longitudinal cohort study in Tororo, Uganda, a highly malaria-endemic region. More than 1800 episodes of malaria were observed in this cohort, with no cases of severe malaria. We quantified production of IFNγ, TNFα, and IL-10 (alone or in combination) by malaria-specific T cells, and analyzed the relationship of this response to past and future malaria incidence. CD4+ T cell responses were measurable in nearly all children, with the majority of children having CD4+ T cells producing both IFNγ and IL-10 in response to malaria-infected red blood cells. Frequencies of IFNγ/IL10 co-producing CD4+ T cells, which express the Th1 transcription factor T-bet, were significantly higher in children with ≥2 prior episodes/year compared to children with <2 episodes/year (P<0.001) and inversely correlated with duration since malaria (Rho = −0.39, P<0.001). Notably, frequencies of IFNγ/IL10 co-producing cells were not associated with protection from future malaria after controlling for prior malaria incidence. In contrast, children with <2 prior episodes/year were significantly more likely to exhibit antigen-specific production of TNFα without IL-10 (P = 0.003). While TNFα-producing CD4+ T cells were not independently associated with future protection, the absence of cells producing this inflammatory cytokine was associated with the phenotype of asymptomatic infection. Together these data indicate that the functional phenotype of the malaria-specific T cell response is heavily influenced by malaria exposure intensity, with IFNγ/IL10 co-producing CD4+ T cells dominating this response among highly exposed children. These CD4+ T cells may play important modulatory roles in the development of antimalarial immunity.


Vyšlo v časopise: IFNγ/IL-10 Co-producing Cells Dominate the CD4 Response to Malaria in Highly Exposed Children. PLoS Pathog 10(1): e32767. doi:10.1371/journal.ppat.1003864
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1003864

Souhrn

Although evidence suggests that T cells are critical for immunity to malaria, reliable T cell correlates of exposure to and protection from malaria among children living in endemic areas are lacking. We used multiparameter flow cytometry to perform a detailed functional characterization of malaria-specific T cells in 78 four-year-old children enrolled in a longitudinal cohort study in Tororo, Uganda, a highly malaria-endemic region. More than 1800 episodes of malaria were observed in this cohort, with no cases of severe malaria. We quantified production of IFNγ, TNFα, and IL-10 (alone or in combination) by malaria-specific T cells, and analyzed the relationship of this response to past and future malaria incidence. CD4+ T cell responses were measurable in nearly all children, with the majority of children having CD4+ T cells producing both IFNγ and IL-10 in response to malaria-infected red blood cells. Frequencies of IFNγ/IL10 co-producing CD4+ T cells, which express the Th1 transcription factor T-bet, were significantly higher in children with ≥2 prior episodes/year compared to children with <2 episodes/year (P<0.001) and inversely correlated with duration since malaria (Rho = −0.39, P<0.001). Notably, frequencies of IFNγ/IL10 co-producing cells were not associated with protection from future malaria after controlling for prior malaria incidence. In contrast, children with <2 prior episodes/year were significantly more likely to exhibit antigen-specific production of TNFα without IL-10 (P = 0.003). While TNFα-producing CD4+ T cells were not independently associated with future protection, the absence of cells producing this inflammatory cytokine was associated with the phenotype of asymptomatic infection. Together these data indicate that the functional phenotype of the malaria-specific T cell response is heavily influenced by malaria exposure intensity, with IFNγ/IL10 co-producing CD4+ T cells dominating this response among highly exposed children. These CD4+ T cells may play important modulatory roles in the development of antimalarial immunity.


Zdroje

1. LanghorneJ, NdunguFM, SponaasAM, MarshK (2008) Immunity to malaria: more questions than answers. Nat Immunol 9: 725–732.

2. MarshK, KinyanjuiS (2006) Immune effector mechanisms in malaria. Parasite Immunol 28: 51–60.

3. TranTM, LiS, DoumboS, DoumtabeD, HuangCY, et al. (2013) An intensive longitudinal cohort study of Malian children and adults reveals no evidence of acquired immunity to Plasmodium falciparum infection. Clin Infect Dis 57: 40–47.

4. DeloronP, ChougnetC (1992) Is immunity to malaria really short-lived? Parasitol Today 8: 375–378.

5. Di PerriG, SolbiatiM, VentoS, De ChecchiG, LuzzatiR, et al. (1994) West African Immigrants and New Patterns of Malaria Imported to North Eastern Italy. Journal of Travel Medicine 1: 147–151.

6. BarryAE, SchultzL, BuckeeCO, ReederJC (2009) Contrasting population structures of the genes encoding ten leading vaccine-candidate antigens of the human malaria parasite, Plasmodium falciparum. PLoS ONE 4: e8497.

7. AdkinsB (1999) T-cell function in newborn mice and humans. Immunol Today 20: 330–335.

8. PassRF, StagnoS, BrittWJ, AlfordCA (1983) Specific cell-mediated immunity and the natural history of congenital infection with cytomegalovirus. J Infect Dis 148: 953–961.

9. AdkinsB, LeclercC, Marshall-ClarkeS (2004) Neonatal adaptive immunity comes of age. Nat Rev Immunol 4: 553–564.

10. HoltPG (2003) Functionally mature virus-specific CD8(+) T memory cells in congenitally infected newborns: proof of principle for neonatal vaccination? J Clin Invest 111: 1645–1647.

11. WilsonCB, LewisDB (1990) Basis and implications of selectively diminished cytokine production in neonatal susceptibility to infection. Rev Infect Dis 12 (Suppl 4) S410–420.

12. WilsonCB, LewisDB, EnglishBK (1991) T cell development in the fetus and neonate. Adv Exp Med Biol 310: 17–27.

13. HoM, WebsterHK, LooareesuwanS, SupanaranondW, PhillipsRE, et al. (1986) Antigen-specific immunosuppression in human malaria due to Plasmodium falciparum. J Infect Dis 153: 763–771.

14. PlebanskiM, FlanaganKL, LeeEA, ReeceWH, HartK, et al. (1999) Interleukin 10-mediated immunosuppression by a variant CD4 T cell epitope of Plasmodium falciparum. Immunity 10: 651–660.

15. WaltherM, TongrenJE, AndrewsL, KorbelD, KingE, et al. (2005) Upregulation of TGF-beta, FOXP3, and CD4+CD25+ regulatory T cells correlates with more rapid parasite growth in human malaria infection. Immunity 23: 287–296.

16. BejonP, MwacharoJ, KaiO, TodrykS, KeatingS, et al. (2007) The induction and persistence of T cell IFN-gamma responses after vaccination or natural exposure is suppressed by Plasmodium falciparum. J Immunol 179: 4193–4201.

17. ButlerNS, MoebiusJ, PeweLL, TraoreB, DoumboOK, et al. (2012) Therapeutic blockade of PD-L1 and LAG-3 rapidly clears established blood-stage Plasmodium infection. Nat Immunol 13: 188–195.

18. Freitas do RosarioAP, LambT, SpenceP, StephensR, LangA, et al. (2012) IL-27 promotes IL-10 production by effector Th1 CD4+ T cells: a critical mechanism for protection from severe immunopathology during malaria infection. J Immunol 188: 1178–1190.

19. Freitas do RosarioAP, LanghorneJ (2012) T cell-derived IL-10 and its impact on the regulation of host responses during malaria. Int J Parasitol 42: 549–555.

20. JagannathanP, MuhindoMK, KakuruA, ArinaitweE, GreenhouseB, et al. (2012) Increasing incidence of malaria in children despite insecticide-treated bed nets and prompt anti-malarial therapy in Tororo, Uganda. Malar J 11: 435.

21. Roca-FeltrerA, KwizombeCJ, SanjoaquinMA, SesaySS, FaragherB, et al. (2012) Lack of decline in childhood malaria, Malawi, 2001–2010. Emerging infectious diseases 18: 272–278.

22. TrapeJF, TallA, DiagneN, NdiathO, LyAB, et al. (2011) Malaria morbidity and pyrethroid resistance after the introduction of insecticide-treated bednets and artemisinin-based combination therapies: a longitudinal study. Lancet Infectious Diseases 11: 925–932.

23. SchofieldL, VillaquiranJ, FerreiraA, SchellekensH, NussenzweigR, et al. (1987) Gamma interferon, CD8+ T cells and antibodies required for immunity to malaria sporozoites. Nature 330: 664–666.

24. RomeroP, MaryanskiJL, CorradinG, NussenzweigRS, NussenzweigV, et al. (1989) Cloned cytotoxic T cells recognize an epitope in the circumsporozoite protein and protect against malaria. Nature 341: 323–326.

25. RodriguesM, NussenzweigRS, ZavalaF (1993) The relative contribution of antibodies, CD4+ and CD8+ T cells to sporozoite-induced protection against malaria. Immunology 80: 1–5.

26. TsujiM, BergmannCC, Takita-SonodaY, MurataK, RodriguesEG, et al. (1998) Recombinant Sindbis viruses expressing a cytotoxic T-lymphocyte epitope of a malaria parasite or of influenza virus elicit protection against the corresponding pathogen in mice. J Virol 72: 6907–6910.

27. StephensR, AlbanoFR, QuinS, PascalBJ, HarrisonV, et al. (2005) Malaria-specific transgenic CD4(+) T cells protect immunodeficient mice from lethal infection and demonstrate requirement for a protective threshold of antibody production for parasite clearance. Blood 106: 1676–1684.

28. SchmidtNW, PodyminoginRL, ButlerNS, BadovinacVP, TuckerBJ, et al. (2008) Memory CD8 T cell responses exceeding a large but definable threshold provide long-term immunity to malaria. Proc Natl Acad Sci U S A 105: 14017–14022.

29. OverstreetMG, CockburnIA, ChenYC, ZavalaF (2008) Protective CD8 T cells against Plasmodium liver stages: immunobiology of an ‘unnatural’ immune response. Immunol Rev 225: 272–283.

30. StephensR, LanghorneJ (2010) Effector memory Th1 CD4 T cells are maintained in a mouse model of chronic malaria. PLoS Pathog 6: e1001208.

31. NussenzweigRS, VanderbergJ, MostH, OrtonC (1967) Protective immunity produced by the injection of x-irradiated sporozoites of plasmodium berghei. Nature 216: 160–162.

32. ClydeDF, MostH, McCarthyVC, VanderbergJP (1973) Immunization of man against sporozite-induced falciparum malaria. Am J Med Sci 266: 169–177.

33. HoffmanSL, DoolanDL (2000) Malaria vaccines-targeting infected hepatocytes. Nat Med 6: 1218–1219.

34. RoestenbergM, McCallM, HopmanJ, WiersmaJ, LutyAJ, et al. (2009) Protection against a malaria challenge by sporozoite inoculation. N Engl J Med 361: 468–477.

35. FriesenJ, SilvieO, PutriantiED, HafallaJC, MatuschewskiK, et al. (2010) Natural immunization against malaria: causal prophylaxis with antibiotics. Sci Transl Med 2: 40ra49.

36. RoestenbergM, TeirlinckAC, McCallMB, TeelenK, MakamdopKN, et al. (2011) Long-term protection against malaria after experimental sporozoite inoculation: an open-label follow-up study. Lancet 377: 1770–1776.

37. ReeceWH, PinderM, GothardPK, MilliganP, BojangK, et al. (2004) A CD4(+) T-cell immune response to a conserved epitope in the circumsporozoite protein correlates with protection from natural Plasmodium falciparum infection and disease. Nat Med 10: 406–410.

38. TodrykSM, BejonP, MwangiT, PlebanskiM, UrbanB, et al. (2008) Correlation of memory T cell responses against TRAP with protection from clinical malaria, and CD4 CD25 high T cells with susceptibility in Kenyans. PLoS ONE 3: e2027.

39. KurtisJD, HollingdaleMR, LutyAJ, LanarDE, KrzychU, et al. (2001) Pre-erythrocytic immunity to Plasmodium falciparum: the case for an LSA-1 vaccine. Trends Parasitol 17: 219–223.

40. HoffmanSL, OsterCN, MasonC, BeierJC, SherwoodJA, et al. (1989) Human lymphocyte proliferative response to a sporozoite T cell epitope correlates with resistance to falciparum malaria. J Immunol 142: 1299–1303.

41. LutyAJ, LellB, Schmidt-OttR, LehmanLG, LucknerD, et al. (1999) Interferon-gamma responses are associated with resistance to reinfection with Plasmodium falciparum in young African children. J Infect Dis 179: 980–988.

42. MoormannAM, SumbaPO, ChelimoK, FangH, TischDJ, et al. (2013) Humoral and Cellular Immunity to Plasmodium falciparum Merozoite Surface Protein 1 and Protection From Infection With Blood-Stage Parasites. J Infect Dis 208: 149–158.

43. D'OmbrainMC, RobinsonLJ, StanisicDI, TaraikaJ, BernardN, et al. (2008) Association of early interferon-gamma production with immunity to clinical malaria: a longitudinal study among Papua New Guinean children. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America 47: 1380–1387.

44. McCallMB, HopmanJ, DaouM, MaigaB, DaraV, et al. (2010) Early interferon-gamma response against Plasmodium falciparum correlates with interethnic differences in susceptibility to parasitemia between sympatric Fulani and Dogon in Mali. J Infect Dis 201: 142–152.

45. BejonP, WarimweG, MackintoshCL, MackinnonMJ, KinyanjuiSM, et al. (2009) Analysis of immunity to febrile malaria in children that distinguishes immunity from lack of exposure. Infect Immun 77: 1917–1923.

46. GreenhouseB, HoB, HubbardA, Njama-MeyaD, NarumDL, et al. (2011) Antibodies to Plasmodium falciparum antigens predict a higher risk of malaria but protection from symptoms once parasitemic. J Infect Dis 204: 19–26.

47. DouekDC, RoedererM, KoupRA (2009) Emerging concepts in the immunopathogenesis of AIDS. Annual Review of Medicine 60: 471–484.

48. DarrahPA, PatelDT, De LucaPM, LindsayRW, DaveyDF, et al. (2007) Multifunctional TH1 cells define a correlate of vaccine-mediated protection against Leishmania major. Nature medicine 13: 843–850.

49. AndersonCF, OukkaM, KuchrooVJ, SacksD (2007) CD4(+)CD25(−)Foxp3(−) Th1 cells are the source of IL-10-mediated immune suppression in chronic cutaneous leishmaniasis. J Exp Med 204: 285–297.

50. JankovicD, KullbergMC, FengCG, GoldszmidRS, CollazoCM, et al. (2007) Conventional T-bet(+)Foxp3(−) Th1 cells are the major source of host-protective regulatory IL-10 during intracellular protozoan infection. J Exp Med 204: 273–283.

51. OlotuA, MorisP, MwacharoJ, VekemansJ, KimaniD, et al. (2011) Circumsporozoite-Specific T Cell Responses in Children Vaccinated with RTS,S/AS01(E) and Protection against P falciparum Clinical Malaria. PLoS ONE 6: e25786.

52. WaltherM, JeffriesD, FinneyOC, NjieM, EbonyiA, et al. (2009) Distinct roles for FOXP3 and FOXP3 CD4 T cells in regulating cellular immunity to uncomplicated and severe Plasmodium falciparum malaria. PLoS Pathog 5: e1000364.

53. ZhangG, ManacaMN, McNamara-SmithM, MayorA, NhabombaA, et al. (2012) Interleukin-10 (IL-10) polymorphisms are associated with IL-10 production and clinical malaria in young children. Infect Immun 80: 2316–2322.

54. Epstein JE, Tewari K, Lyke KE, Sim BK, Billingsley PF, et al. (2011) Live Attenuated Malaria Vaccine Designed to Protect through Hepatic CD8+ T Cell Immunity. Science.

55. OlotuA, MorisP, MwacharoJ, VekemansJ, KimaniD, et al. (2011) Circumsporozoite-specific T cell responses in children vaccinated with RTS,S/AS01E and protection against P falciparum clinical malaria. PLoS ONE 6: e25786.

56. CopeA, Le FriecG, CardoneJ, KemperC (2011) The Th1 life cycle: molecular control of IFN-gamma to IL-10 switching. Trends in Immunology 32: 278–286.

57. O'GarraA, VieiraP (2007) T(H)1 cells control themselves by producing interleukin-10. Nature reviews Immunology 7: 425–428.

58. WenischC, ParschalkB, NarztE, LooareesuwanS, GraningerW (1995) Elevated serum levels of IL-10 and IFN-gamma in patients with acute Plasmodium falciparum malaria. Clinical Immunology and Immunopathology 74: 115–117.

59. PeyronF, BurdinN, RingwaldP, VuillezJP, RoussetF, et al. (1994) High levels of circulating IL-10 in human malaria. Clinical and experimental immunology 95: 300–303.

60. WilsonNO, BythwoodT, SolomonW, JollyP, YatichN, et al. (2010) Elevated levels of IL-10 and G-CSF associated with asymptomatic malaria in pregnant women. Infectious Diseases in Obstetrics and Gynecology 2010: pii: 317430.

61. Pinzon-CharryA, WoodberryT, KienzleV, McPhunV, MinigoG, et al. (2013) Apoptosis and dysfunction of blood dendritic cells in patients with falciparum and vivax malaria. J Exp Med 210: 1635–1646.

62. O'ConnorRA, JensonJS, OsborneJ, DevaneyE (2003) An enduring association? Microfilariae and immunosuppression [correction of immunosupression] in lymphatic filariasis. Trends Parasitol 19: 565–570.

63. SteelC, GuineaA, McCarthyJS, OttesenEA (1994) Long-term effect of prenatal exposure to maternal microfilaraemia on immune responsiveness to filarial parasite antigens. Lancet 343: 890–893.

64. WammesLJ, HamidF, WiriaAE, WibowoH, SartonoE, et al. (2012) Regulatory T cells in human lymphatic filariasis: stronger functional activity in microfilaremics. PLoS Neglected Tropical Diseases 6: e1655.

65. MaizelsRM, YazdanbakhshM (2003) Immune regulation by helminth parasites: cellular and molecular mechanisms. Nature reviews Immunology 3: 733–744.

66. McNeilAC, ShupertWL, IyasereCA, HallahanCW, MicanJA, et al. (2001) High-level HIV-1 viremia suppresses viral antigen-specific CD4(+) T cell proliferation. Proc Natl Acad Sci U S A 98: 13878–13883.

67. ThimmeR, OldachD, ChangKM, SteigerC, RaySC, et al. (2001) Determinants of viral clearance and persistence during acute hepatitis C virus infection. J Exp Med 194: 1395–1406.

68. HaringerB, LozzaL, SteckelB, GeginatJ (2009) Identification and characterization of IL-10/IFN-gamma-producing effector-like T cells with regulatory function in human blood. J Exp Med 206: 1009–1017.

69. BoussiotisVA, TsaiEY, YunisEJ, ThimS, DelgadoJC, et al. (2000) IL-10-producing T cells suppress immune responses in anergic tuberculosis patients. J Clin Invest 105: 1317–1325.

70. MeilerF, ZumkehrJ, KlunkerS, RuckertB, AkdisCA, et al. (2008) In vivo switch to IL-10-secreting T regulatory cells in high dose allergen exposure. J Exp Med 205: 2887–2898.

71. O'GarraA, VieiraP (2007) T(H)1 cells control themselves by producing interleukin-10. Nat Rev Immunol 7: 425–428.

72. LiC, CorralizaI, LanghorneJ (1999) A defect in interleukin-10 leads to enhanced malarial disease in Plasmodium chabaudi chabaudi infection in mice. Infect Immun 67: 4435–4442.

73. WinklerS, WillheimM, BaierK, SchmidD, AichelburgA, et al. (1998) Reciprocal regulation of Th1- and Th2-cytokine-producing T cells during clearance of parasitemia in Plasmodium falciparum malaria. Infect Immun 66: 6040–6044.

74. FlanaganKL, PlebanskiM, OdhiamboK, SheuE, MwangiT, et al. (2006) Cellular reactivity to the p. Falciparum protein trap in adult kenyans: novel epitopes, complex cytokine patterns, and the impact of natural antigenic variation. Am J Trop Med Hyg 74: 367–375.

75. GitauEN, TujuJ, StevensonL, KimaniE, KaranjaH, et al. (2012) T-cell responses to the DBLalpha-tag, a short semi-conserved region of the Plasmodium falciparum membrane erythrocyte protein 1. PLoS ONE 7: e30095.

76. RoetynckS, OlotuA, SimamJ, MarshK, StockingerB, et al. (2013) Phenotypic and functional profiling of CD4 T cell compartment in distinct populations of healthy adults with different antigenic exposure. PLoS ONE 8: e55195.

77. Freitas do RosarioAP, LanghorneJ (2012) T cell-derived IL-10 and its impact on the regulation of host responses during malaria. International Journal for Parasitology 42: 549–555.

78. MetenouS, DembeleB, KonateS, DoloH, CoulibalyYI, et al. (2011) Filarial infection suppresses malaria-specific multifunctional Th1 and Th17 responses in malaria and filarial coinfections. Journal of immunology 186: 4725–4733.

79. BrustoskiK, MollerU, KramerM, PetelskiA, BrennerS, et al. (2005) IFN-gamma and IL-10 mediate parasite-specific immune responses of cord blood cells induced by pregnancy-associated Plasmodium falciparum malaria. Journal of immunology 174: 1738–1745.

80. BrockmanMA, KwonDS, TigheDP, PavlikDF, RosatoPC, et al. (2009) IL-10 is up-regulated in multiple cell types during viremic HIV infection and reversibly inhibits virus-specific T cells. Blood 114: 346–356.

81. BrooksDG, TrifiloMJ, EdelmannKH, TeytonL, McGavernDB, et al. (2006) Interleukin-10 determines viral clearance or persistence in vivo. Nat Med 12: 1301–1309.

82. DepinayN, FranetichJF, GrunerAC, MauduitM, ChavatteJM, et al. (2011) Inhibitory effect of TNF-alpha on malaria pre-erythrocytic stage development: influence of host hepatocyte/parasite combinations. PLoS ONE 6: e17464.

83. NusslerA, PiedS, GomaJ, ReniaL, MiltgenF, et al. (1991) TNF inhibits malaria hepatic stages in vitro via synthesis of IL-6. International Immunology 3: 317–321.

84. KatrakS, GasasiraA, ArinaitweE, KakuruA, WanziraH, et al. (2009) Safety and tolerability of artemether-lumefantrine versus dihydroartemisinin-piperaquine for malaria in young HIV-infected and uninfected children. Malaria Journal 8: 272.

85. SandisonTG, HomsyJ, ArinaitweE, WanziraH, KakuruA, et al. (2011) Protective efficacy of co-trimoxazole prophylaxis against malaria in HIV exposed children in rural Uganda: a randomised clinical trial. BMJ 342: d1617.

86. VoraN, HomsyJ, KakuruA, ArinaitweE, WanziraH, et al. (2010) Breastfeeding and the risk of malaria in children born to HIV-infected and uninfected mothers in rural Uganda. Journal of Acquired Immune Deficiency Syndromes 55: 253–261.

87. ArinaitweE, SandisonTG, WanziraH, KakuruA, HomsyJ, et al. (2009) Artemether-lumefantrine versus dihydroartemisinin-piperaquine for falciparum malaria: a longitudinal, randomized trial in young Ugandan children. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America 49: 1629–1637.

88. (2010) Uganda Clinical Guidelines 2010. National Guidelines on Management of Common Conditions. Kampala, Uganda: Uganda Ministry of Health.

89. WipasaJ, OkellL, SakkhachornphopS, SuphavilaiC, ChawansuntatiK, et al. (2011) Short-lived IFN-gamma effector responses, but long-lived IL-10 memory responses, to malaria in an area of low malaria endemicity. PLoS pathogens 7: e1001281.

90. HorowitzA, NewmanKC, EvansJH, KorbelDS, DavisDM, et al. (2010) Cross-talk between T cells and NK cells generates rapid effector responses to Plasmodium falciparum-infected erythrocytes. J Immunol 184: 6043–6052.

91. MaeckerHT, TrotterJ (2006) Flow cytometry controls, instrument setup, and the determination of positivity. Cytometry Part A 69: 1037–1042.

92. LamoreauxL, RoedererM, KoupR (2006) Intracellular cytokine optimization and standard operating procedure. Nat Protoc 1: 1507–1516.

93. McLaughlinBE, BaumgarthN, BigosM, RoedererM, De RosaSC, et al. (2008) Nine-color flow cytometry for accurate measurement of T cell subsets and cytokine responses. Part I: Panel design by an empiric approach. Cytometry Part A 73: 400–410.

94. RoedererM, NozziJL, NasonMX (2011) SPICE: Exploration and analysis of post-cytometric complex multivariate datasets. Cytometry Part A [Epub ahead of print].

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2014 Číslo 1
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#