#PAGE_PARAMS# #ADS_HEAD_SCRIPTS# #MICRODATA#

Aging boosts antiviral CD8+T cell memory through improvedengagement of diversified recall response determinants


Autoři: Bennett Davenport aff001;  Jens Eberlein aff001;  Tom T. Nguyen aff001;  Francisco Victorino aff001;  Kevin Jhun aff003;  Haedar Abuirqeba aff003;  Verena van der Heide aff003;  Peter Heeger aff004;  Dirk Homann aff001
Působiště autorů: Department of Anesthesiology & Barbara Davis Center for Childhood Diabetes, University of Colorado Denver, Aurora, Colorado, United States of America aff001;  Integrated Department of Immunology, University of Colorado Denver and National Jewish Health, Denver, Colorado, United States of America aff002;  Diabetes, Obesity & Metabolism Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America aff003;  Precision Immunology Institute, Icahn School of Medicine at Mount Sinai, New York, New York, United States of America aff004
Vyšlo v časopise: Aging boosts antiviral CD8+T cell memory through improvedengagement of diversified recall response determinants. PLoS Pathog 15(11): e32767. doi:10.1371/journal.ppat.1008144
Kategorie: Research Article
prolekare.web.journal.doi_sk: https://doi.org/10.1371/journal.ppat.1008144

Souhrn

The determinants of protective CD8+ memory T cell (CD8+TM) immunity remain incompletely defined and may in fact constitute an evolving agency as aging CD8+TM progressively acquire enhanced rather than impaired recall capacities. Here, we show that old as compared to young antiviral CD8+TM more effectively harness disparate molecular processes (cytokine signaling, trafficking, effector functions, and co-stimulation/inhibition) that in concert confer greater secondary reactivity. The relative reliance on these pathways is contingent on the nature of the secondary challenge (greater for chronic than acute viral infections) and over time, aging CD8+TM re-establish a dependence on the same accessory signals required for effective priming of naïve CD8+T cells in the first place. Thus, our findings reveal a temporal regulation of complementary recall response determinants that is consistent with the recently proposed “rebound model” according to which aging CD8+TM properties are gradually aligned with those of naïve CD8+T cells; our identification of a broadly diversified collection of immunomodulatory targets may further provide a foundation for the potential therapeutic “tuning” of CD8+TM immunity.

Klíčová slova:

Immune response – Blood – T cells – Cytotoxic T cells – Spleen – Aging – Memory recall – CD coreceptors


Zdroje

1. Sallusto F, Geginat J, Lanzavecchia A. Central memory and effector memory T cell subsets: function, generation, and maintenance. Annu Rev Immunol. 2004;22:745–63. doi: 10.1146/annurev.immunol.22.012703.104702 15032595

2. Harty JT, Badovinac VP. Shaping and reshaping CD8+ T-cell memory. Nat Rev Immunol. 2008;8(2):107–19. doi: 10.1038/nri2251 18219309

3. Mueller SN, Gebhardt T, Carbone FR, Heath WR. Memory T cell subsets, migration patterns, and tissue residence. Annu Rev Immunol. 2013;31:137–61. doi: 10.1146/annurev-immunol-032712-095954 23215646

4. Crotty S, Kaech SM, Schoenberger SP. Immunologic memory. In: Paul WE, editor. Fundamental Immunology. 7th ed. ed. Philadelphia, PA: Lippincott Williams & Wilkins; 2013. p. 741–64.

5. Roberts AD, Ely KH, Woodland DL. Differential contributions of central and effector memory T cells to recall responses. J Exp Med. 2005;202(1):123–33. doi: 10.1084/jem.20050137 15983064

6. Klinger A, Gebert A, Bieber K, Kalies K, Ager A, Bell EB, et al. Cyclical expression of L-selectin (CD62L) by recirculating T cells. Int Immunol. 2009;21(4):443–55. doi: 10.1093/intimm/dxp012 19240088

7. Rutishauser RL, Kaech SM. Generating diversity: transcriptional regulation of effector and memory CD8 T-cell differentiation. Immunol Rev. 2010;235(1):219–33. doi: 10.1111/j.0105-2896.2010.00901.x 20536566

8. Martin MD, Kim MT, Shan Q, Sompallae R, Xue HH, Harty JT, et al. Phenotypic and Functional Alterations in Circulating Memory CD8 T Cells with Time after Primary Infection. PLoS Pathog. 2015;11(10):e1005219. doi: 10.1371/journal.ppat.1005219 26485703

9. Eberlein J, Davenport B, Nguyen TT, Victorino F, Karimpour-Fard A, Hunter LE, et al. Aging promotes acquisition of naïve-like CD8+ memory T cell traits and enhanced functionalities. J Clin Invest. 2016;106(10):3942–60.

10. Davenport B, Eberlein J, van der Heide V, Jhun K, Nguyen TT, Victorino F, et al. Aging of Antiviral CD8(+) Memory T Cells Fosters Increased Survival, Metabolic Adaptations, and Lymphoid Tissue Homing. J Immunol. 2019;202(2):460–75. doi: 10.4049/jimmunol.1801277 30552164

11. Graef P, Buchholz VR, Stemberger C, Flossdorf M, Henkel L, Schiemann M, et al. Serial transfer of single-cell-derived immunocompetence reveals stemness of CD8(+) central memory T cells. Immunity. 2014;41(1):116–26. doi: 10.1016/j.immuni.2014.05.018 25035956

12. Stemberger C, Graef P, Odendahl M, Albrecht J, Dossinger G, Anderl F, et al. Lowest numbers of primary CD8(+) T cells can reconstitute protective immunity upon adoptive immunotherapy. Blood. 2014;124(4):628–37. doi: 10.1182/blood-2013-12-547349 24855206

13. Nolz JC, Harty JT. Protective capacity of memory CD8+ T cells is dictated by antigen exposure history and nature of the infection. Immunity. 2011;34(5):781–93. doi: 10.1016/j.immuni.2011.03.020 21549619

14. Boyman O, Krieg C, Homann D, Sprent J. Homeostatic maintenance of T cells and natural killer cells. Cell Mol Life Sci. 2012;69(10):1597–608. doi: 10.1007/s00018-012-0968-7 22460580

15. Mackall CL, Fry TJ, Gress RE. Harnessing the biology of IL-7 for therapeutic application. Nat Rev Immunol. 2011;11(5):330–42. doi: 10.1038/nri2970 21508983

16. Kim MT, Harty JT. Impact of Inflammatory Cytokines on Effector and Memory CD8+ T Cells. Frontiers in immunology. 2014;5:295. doi: 10.3389/fimmu.2014.00295 24995011

17. Pandey A, Ozaki K, Baumann H, Levin SD, Puel A, Farr AG, et al. Cloning of a receptor subunit required for signaling by thymic stromal lymphopoietin. Nat Immunol. 2000;1(1):59–64. doi: 10.1038/76923 10881176

18. Brown VI, Hulitt J, Fish J, Sheen C, Bruno M, Xu Q, et al. Thymic stromal-derived lymphopoietin induces proliferation of pre-B leukemia and antagonizes mTOR inhibitors, suggesting a role for interleukin-7Ralpha signaling. Cancer Res. 2007;67(20):9963–70. doi: 10.1158/0008-5472.CAN-06-4704 17942929

19. Richer MJ, Pewe LL, Hancox LS, Hartwig SM, Varga SM, Harty JT. Inflammatory IL-15 is required for optimal memory T cell responses. J Clin Invest. 2015;125(9):3477–90. doi: 10.1172/JCI81261 26241055

20. Becker TC, Wherry EJ, Boone D, Murali-Krishna K, Antia R, Ma A, et al. Interleukin 15 is required for proliferative renewal of virus-specific memory CD8 T cells. J Exp Med. 2002;195(12):1541–8. doi: 10.1084/jem.20020369 12070282

21. Tinoco R, Alcalde V, Yang Y, Sauer K, Zuniga EI. Cell-intrinsic transforming growth factor-beta signaling mediates virus-specific CD8+ T cell deletion and viral persistence in vivo. Immunity. 2009;31(1):145–57. doi: 10.1016/j.immuni.2009.06.015 19604493

22. Harker JA, Lewis GM, Mack L, Zuniga EI. Late interleukin-6 escalates T follicular helper cell responses and controls a chronic viral infection. Science. 2011;334(6057):825–9. doi: 10.1126/science.1208421 21960530

23. Lee A, Park SP, Park CH, Kang BH, Park SH, Ha SJ, et al. IL-4 Induced Innate CD8+ T Cells Control Persistent Viral Infection. PLoS Pathog. 2015;11(10):e1005193. doi: 10.1371/journal.ppat.1005193 26452143

24. White JT, Cross EW, Kedl RM. Antigen-inexperienced memory CD8+ T cells: where they come from and why we need them. Nat Rev Immunol. 2017;17(6):391–400. doi: 10.1038/nri.2017.34 28480897

25. Bonilla WV, Frohlich A, Senn K, Kallert S, Fernandez M, Johnson S, et al. The alarmin interleukin-33 drives protective antiviral CD8(+) T cell responses. Science. 2012;335(6071):984–9. doi: 10.1126/science.1215418 22323740

26. Garidou L, Heydari S, Gossa S, McGavern DB. Therapeutic blockade of transforming growth factor beta fails to promote clearance of a persistent viral infection. J Virol. 2012;86(13):7060–71. doi: 10.1128/JVI.00164-12 22553324

27. Boettler T, Cheng Y, Ehrhardt K, von Herrath M. TGF-beta blockade does not improve control of an established persistent viral infection. Viral Immunol. 2012;25(3):232–8. doi: 10.1089/vim.2011.0079 22620718

28. Wherry EJ, Teichgraber V, Becker TC, Masopust D, Kaech SM, Antia R, et al. Lineage relationship and protective immunity of memory CD8 T cell subsets. Nat Immunol. 2003;4(3):225–34. doi: 10.1038/ni889 12563257

29. Feau S, Arens R, Togher S, Schoenberger SP. Autocrine IL-2 is required for secondary population expansion of CD8(+) memory T cells. Nat Immunol. 2011;12(9):908–13. doi: 10.1038/ni.2079 21804558

30. Whitmire JK, Tan JT, Whitton JL. Interferon-gamma acts directly on CD8+ T cells to increase their abundance during virus infection. J Exp Med. 2005;201(7):1053–9. doi: 10.1084/jem.20041463 15809350

31. Balkow S, Kersten A, Tran TT, Stehle T, Grosse P, Museteanu C, et al. Concerted action of the FasL/Fas and perforin/granzyme A and B pathways is mandatory for the development of early viral hepatitis but not for recovery from viral infection. J Virol. 2001;75(18):8781–91. doi: 10.1128/JVI.75.18.8781-8791.2001 11507223

32. Rode M, Balkow S, Sobek V, Brehm R, Martin P, Kersten A, et al. Perforin and Fas act together in the induction of apoptosis, and both are critical in the clearance of lymphocytic choriomeningitis virus infection. J Virol. 2004;78(22):12395–405. doi: 10.1128/JVI.78.22.12395-12405.2004 15507626

33. Weant AE, Michalek RD, Khan IU, Holbrook BC, Willingham MC, Grayson JM. Apoptosis regulators Bim and Fas function concurrently to control autoimmunity and CD8+ T cell contraction. Immunity. 2008;28(2):218–30. doi: 10.1016/j.immuni.2007.12.014 18275832

34. Hughes PD, Belz GT, Fortner KA, Budd RC, Strasser A, Bouillet P. Apoptosis regulators Fas and Bim cooperate in shutdown of chronic immune responses and prevention of autoimmunity. Immunity. 2008;28(2):197–205. doi: 10.1016/j.immuni.2007.12.017 18275830

35. Klebanoff CA, Scott CD, Leonardi AJ, Yamamoto TN, Cruz AC, Ouyang C, et al. Memory T cell-driven differentiation of naive cells impairs adoptive immunotherapy. J Clin Invest. 2016;126(1):318–34. doi: 10.1172/JCI81217 26657860

36. Hallermalm K, De Geer A, Kiessling R, Levitsky V, Levitskaya J. Autocrine secretion of Fas ligand shields tumor cells from Fas-mediated killing by cytotoxic lymphocytes. Cancer Res. 2004;64(18):6775–82. doi: 10.1158/0008-5472.CAN-04-0508 15374996

37. Rai D, Pham NL, Harty JT, Badovinac VP. Tracking the total CD8 T cell response to infection reveals substantial discordance in magnitude and kinetics between inbred and outbred hosts. J Immunol. 2009;183(12):7672–81. doi: 10.4049/jimmunol.0902874 19933864

38. Hogg N, Patzak I, Willenbrock F. The insider’s guide to leukocyte integrin signalling and function. Nat Rev Immunol. 2011;11(6):416–26. doi: 10.1038/nri2986 21597477

39. Berlin-Rufenach C, Otto F, Mathies M, Westermann J, Owen MJ, Hamann A, et al. Lymphocyte migration in lymphocyte function-associated antigen (LFA)-1-deficient mice. J Exp Med. 1999;189(9):1467–78. doi: 10.1084/jem.189.9.1467 10224287

40. Bose TO, Pham QM, Jellison ER, Mouries J, Ballantyne CM, Lefrancois L. CD11a regulates effector CD8 T cell differentiation and central memory development in response to infection with Listeria monocytogenes. Infect Immun. 2013;81(4):1140–51. doi: 10.1128/IAI.00749-12 23357382

41. Gerard A, Khan O, Beemiller P, Oswald E, Hu J, Matloubian M, et al. Secondary T cell-T cell synaptic interactions drive the differentiation of protective CD8+ T cells. Nat Immunol. 2013;14(4):356–63. doi: 10.1038/ni.2547 23475183

42. Ford ML, Adams AB, Pearson TC. Targeting co-stimulatory pathways: transplantation and autoimmunity. Nature reviews Nephrology. 2014;10(1):14–24. doi: 10.1038/nrneph.2013.183 24100403

43. Krummey SM, Ford ML. Heterogeneity within T Cell Memory: Implications for Transplant Tolerance. Frontiers in immunology. 2012;3:36. doi: 10.3389/fimmu.2012.00036 22566919

44. Griffith JW, Sokol CL, Luster AD. Chemokines and chemokine receptors: positioning cells for host defense and immunity. Annu Rev Immunol. 2014;32:659–702. doi: 10.1146/annurev-immunol-032713-120145 24655300

45. Guarda G, Hons M, Soriano SF, Huang AY, Polley R, Martin-Fontecha A, et al. L-selectin-negative CCR7- effector and memory CD8+ T cells enter reactive lymph nodes and kill dendritic cells. Nat Immunol. 2007;8(7):743–52. doi: 10.1038/ni1469 17529983

46. Hu JK, Kagari T, Clingan JM, Matloubian M. Expression of chemokine receptor CXCR3 on T cells affects the balance between effector and memory CD8 T-cell generation. Proc Natl Acad Sci U S A. 2011;108(21):E118–27. doi: 10.1073/pnas.1101881108 21518913

47. Kurachi M, Kurachi J, Suenaga F, Tsukui T, Abe J, Ueha S, et al. Chemokine receptor CXCR3 facilitates CD8(+) T cell differentiation into short-lived effector cells leading to memory degeneration. J Exp Med. 2011;208(8):1605–20. doi: 10.1084/jem.20102101 21788406

48. Kohlmeier JE, Reiley WW, Perona-Wright G, Freeman ML, Yager EJ, Connor LM, et al. Inflammatory chemokine receptors regulate CD8(+) T cell contraction and memory generation following infection. J Exp Med. 2011;208(8):1621–34. doi: 10.1084/jem.20102110 21788409

49. Sung JH, Zhang H, Moseman EA, Alvarez D, Iannacone M, Henrickson SE, et al. Chemokine guidance of central memory T cells is critical for antiviral recall responses in lymph nodes. Cell. 2012;150(6):1249–63. doi: 10.1016/j.cell.2012.08.015 22980984

50. Hikono H, Kohlmeier JE, Takamura S, Wittmer ST, Roberts AD, Woodland DL. Activation phenotype, rather than central- or effector-memory phenotype, predicts the recall efficacy of memory CD8+ T cells. J Exp Med. 2007;204(7):1625–36. doi: 10.1084/jem.20070322 17606632

51. Uppaluri R, Sheehan KC, Wang L, Bui JD, Brotman JJ, Lu B, et al. Prolongation of cardiac and islet allograft survival by a blocking hamster anti-mouse CXCR3 monoclonal antibody. Transplantation. 2008;86(1):137–47. doi: 10.1097/TP.0b013e31817b8e4b 18622291

52. Abboud G, Desai P, Dastmalchi F, Stanfield J, Tahiliani V, Hutchinson TE, et al. Tissue-specific programming of memory CD8 T cell subsets impacts protection against lethal respiratory virus infection. J Exp Med. 2016;213(13):2897–911. doi: 10.1084/jem.20160167 27879287

53. van der Heide V, Homann D. CD28 days later: Resurrecting costimulation for CD8(+) memory T cells. Eur J Immunol. 2016;46(7):1587–91. doi: 10.1002/eji.201646500 27401871

54. Wortzman ME, Clouthier DL, McPherson AJ, Lin GH, Watts TH. The contextual role of TNFR family members in CD8(+) T-cell control of viral infections. Immunol Rev. 2013;255(1):125–48. doi: 10.1111/imr.12086 23947352

55. Schildknecht A, Miescher I, Yagita H, van den Broek M. Priming of CD8+ T cell responses by pathogens typically depends on CD70-mediated interactions with dendritic cells. Eur J Immunol. 2007;37(3):716–28. doi: 10.1002/eji.200636824 17295392

56. Penaloza-MacMaster P, Ur Rasheed A, Iyer SS, Yagita H, Blazar BR, Ahmed R. Opposing effects of CD70 costimulation during acute and chronic lymphocytic choriomeningitis virus infection of mice. J Virol. 2011;85(13):6168–74. doi: 10.1128/JVI.02205-10 21507976

57. Munitic I, Kuka M, Allam A, Scoville JP, Ashwell JD. CD70 deficiency impairs effector CD8 T cell generation and viral clearance but is dispensable for the recall response to lymphocytic choriomeningitis virus. J Immunol. 2013;190(3):1169–79. doi: 10.4049/jimmunol.1202353 23269247

58. Matter MS, Claus C, Ochsenbein AF. CD4+ T cell help improves CD8+ T cell memory by retained CD27 expression. Eur J Immunol. 2008;38(7):1847–56. doi: 10.1002/eji.200737824 18506879

59. Grujic M, Bartholdy C, Remy M, Pinschewer DD, Christensen JP, Thomsen AR. The role of CD80/CD86 in generation and maintenance of functional virus-specific CD8+ T cells in mice infected with lymphocytic choriomeningitis virus. J Immunol. 2010;185(3):1730–43. doi: 10.4049/jimmunol.0903894 20601595

60. Eberlein J, Davenport B, Nguyen TT, Victorino F, Sparwasser T, Homann D. Multiple Layers of CD80/86-Dependent Costimulatory Activity Regulate Primary, Memory, and Secondary Lymphocytic Choriomeningitis Virus-Specific T Cell Immunity. J Virol. 2012;86(4):1955–70. doi: 10.1128/JVI.05949-11 22156513

61. Suresh M, Whitmire JK, Harrington LE, Larsen CP, Pearson TC, Altman JD, et al. Role of CD28-B7 interactions in generation and maintenance of CD8 T cell memory. J Immunol. 2001;167(10):5565–73. doi: 10.4049/jimmunol.167.10.5565 11698427

62. Durlanik S, Loyal L, Stark R, Sercan Alp O, Hartung A, Radbruch A, et al. CD40L expression by CD4+ but not CD8+ T cells regulates antiviral immune responses in acute LCMV infection in mice. Eur J Immunol. 2016;46(11):2566–73. doi: 10.1002/eji.201646420 27562840

63. Frentsch M, Stark R, Matzmohr N, Meier S, Durlanik S, Schulz AR, et al. CD40L expression permits CD8+ T cells to execute immunologic helper functions. Blood. 2013;122(3):405–12. doi: 10.1182/blood-2013-02-483586 23719298

64. Shugart JA, Bambina S, Alice AF, Montler R, Bahjat KS. A self-help program for memory CD8+ T cells: positive feedback via CD40-CD40L signaling as a critical determinant of secondary expansion. PLoS One. 2013;8(5):e64878. doi: 10.1371/journal.pone.0064878 23717671

65. Homann D, Jahreis A, Wolfe T, Hughes A, Coon B, van Stipdonk MJ, et al. CD40L blockade prevents autoimmune diabetes by induction of bitypic NK/DC regulatory cells. Immunity. 2002;16(3):403–15. doi: 10.1016/s1074-7613(02)00290-x 11911825

66. West EE, Youngblood B, Tan WG, Jin HT, Araki K, Alexe G, et al. Tight Regulation of Memory CD8(+) T Cells Limits Their Effectiveness during Sustained High Viral Load. Immunity. 2011;35(2):285–98. doi: 10.1016/j.immuni.2011.05.017 21856186

67. Marzo AL, Vezys V, Klonowski KD, Lee SJ, Muralimohan G, Moore M, et al. Fully functional memory CD8 T cells in the absence of CD4 T cells. J Immunol. 2004;173(2):969–75. doi: 10.4049/jimmunol.173.2.969 15240684

68. Veillette A, Dong Z, Latour S. Consequence of the SLAM-SAP signaling pathway in innate-like and conventional lymphocytes. Immunity. 2007;27(5):698–710. doi: 10.1016/j.immuni.2007.11.005 18031694

69. Chen G, Tai AK, Lin M, Chang F, Terhorst C, Huber BT. Increased proliferation of CD8+ T cells in SAP-deficient mice is associated with impaired activation-induced cell death. Eur J Immunol. 2007;37(3):663–74. doi: 10.1002/eji.200636417 17266174

70. Waggoner SN, Taniguchi RT, Mathew PA, Kumar V, Welsh RM. Absence of mouse 2B4 promotes NK cell-mediated killing of activated CD8+ T cells, leading to prolonged viral persistence and altered pathogenesis. J Clin Invest. 2010;120(6):1925–38. doi: 10.1172/JCI41264 20440077

71. Guo H, Cranert SA, Lu Y, Zhong MC, Zhang S, Chen J, et al. Deletion of Slam locus in mice reveals inhibitory role of SLAM family in NK cell responses regulated by cytokines and LFA-1. J Exp Med. 2016;213(10):2187–207. doi: 10.1084/jem.20160552 27573813

72. Duttagupta PA, Boesteanu AC, Katsikis PD. Costimulation signals for memory CD8+ T cells during viral infections. Crit Rev Immunol. 2009;29(6):469–86. 20121696

73. Morris AB, Adams LE, Ford ML. Influence of T Cell Coinhibitory Molecules on CD8(+) Recall Responses. Frontiers in immunology. 2018;9:1810. doi: 10.3389/fimmu.2018.01810 30135685

74. Kahan SM, Wherry EJ, Zajac AJ. T cell exhaustion during persistent viral infections. Virology. 2015.

75. Curtsinger JM, Mescher MF. Inflammatory cytokines as a third signal for T cell activation. Curr Opin Immunol. 2010;22(3):333–40. doi: 10.1016/j.coi.2010.02.013 20363604

76. Badovinac VP, Porter BB, Harty JT. CD8+ T cell contraction is controlled by early inflammation. Nat Immunol. 2004;5(8):809–17. doi: 10.1038/ni1098 15247915

77. Schroder K, Hertzog PJ, Ravasi T, Hume DA. Interferon-gamma: an overview of signals, mechanisms and functions. J Leukoc Biol. 2004;75(2):163–89. doi: 10.1189/jlb.0603252 14525967

78. Homann D, Tishon A, Berger DP, Weigle WO, von Herrath MG, Oldstone MB. Evidence for an underlying CD4 helper and CD8 T-cell defect in B-cell-deficient mice: failure to clear persistent virus infection after adoptive immunotherapy with virus-specific memory cells from muMT/muMT mice. J Virol. 1998;72(11):9208–16. 9765468

79. Lenz DC, Kurz SK, Lemmens E, Schoenberger SP, Sprent J, Oldstone MB, et al. IL-7 regulates basal homeostatic proliferation of antiviral CD4+T cell memory. Proc Natl Acad Sci U S A. 2004;101(25):9357–62. doi: 10.1073/pnas.0400640101 15197277

80. Marzo AL, Klonowski KD, Le Bon A, Borrow P, Tough DF, Lefrancois L. Initial T cell frequency dictates memory CD8+ T cell lineage commitment. Nat Immunol. 2005;6(8):793–9. doi: 10.1038/ni1227 16025119

81. Badovinac VP, Haring JS, Harty JT. Initial T cell receptor transgenic cell precursor frequency dictates critical aspects of the CD8(+) T cell response to infection. Immunity. 2007;26(6):827–41. doi: 10.1016/j.immuni.2007.04.013 17555991

82. Eberlein J, Nguyen TT, Victorino F, Golden-Mason L, Rosen HR, Homann D. Comprehensive assessment of chemokine expression profiles by flow cytometry. J Clin Invest. 2010;120(3):907–23. doi: 10.1172/JCI40645 20197626

83. Hildemann SK, Eberlein J, Davenport B, Nguyen TT, Victorino F, Homann D. High efficiency of antiviral CD4(+) killer T cells. PLoS One. 2013;8(4):e60420. doi: 10.1371/journal.pone.0060420 23565245

Štítky
Hygiena a epidemiológia Infekčné lekárstvo Laboratórium

Článok vyšiel v časopise

PLOS Pathogens


2019 Číslo 11
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#