Adaptive immune features of natural killer cells

Joseph C Sun, Joshua N Beilke, Lewis L Lanier, Joseph C Sun, Joshua N Beilke, Lewis L Lanier

Abstract

In an adaptive immune response, naive T cells proliferate during infection and generate long-lived memory cells that undergo secondary expansion after a repeat encounter with the same pathogen. Although natural killer (NK) cells have traditionally been classified as cells of the innate immune system, they share many similarities with cytotoxic T lymphocytes. We use a mouse model of cytomegalovirus infection to show that, like T cells, NK cells bearing the virus-specific Ly49H receptor proliferate 100-fold in the spleen and 1,000-fold in the liver after infection. After a contraction phase, Ly49H-positive NK cells reside in lymphoid and non-lymphoid organs for several months. These self-renewing 'memory' NK cells rapidly degranulate and produce cytokines on reactivation. Adoptive transfer of these NK cells into naive animals followed by viral challenge results in a robust secondary expansion and protective immunity. These findings reveal properties of NK cells that were previously attributed only to cells of the adaptive immune system.

Figures

Figure 1. Preferential expansion of wild-type but…
Figure 1. Preferential expansion of wild-type but not DAP12-deficient NK cells during MCMV infection
Mixed bone marrow chimeric mice (1:1 mixture of wild-type (CD45.1+) and DAP12-deficient (CD45.2+) cells) were infected with MCMV. a, Upper, percentages of wild-type and DAP12-deficient NK cells (gated on CD3−, NK1.1+). Lower, Ly49H and KLRG1 on wild-type NK cells. b, Upper, BrdU incorporation (day 7 PI) of wild-type (solid lines) and DAP12-deficient (dotted lines) NK cells. Lower, BrdU incorporation by Ly49H+ (solid lines) and Ly49H− (dotted lines) wild-type NK cells. c, d Percentages of Ly49H+ cells within the wild-type NK cell population following infection with MCMV (c) or MCMV-Δm157 (d). Data are representative of 3 experiments with 3–5 mice per time point.
Figure 2. Robust proliferation of adoptively transferred…
Figure 2. Robust proliferation of adoptively transferred wild-type NK cells in DAP12-deficient mice following MCMV infection
a, 105 wild-type NK cells (CD45.1+) were transferred into DAP12-deficient mice (CD45.2+) and infected with MCMV. b, Transferred NK cells (CD45.1+) within the total CD3− NK1.1+ gated population analyzed for Ly49H, CD69, NKG2D, and intracellular IFN-γ. c, Percentages of transferred CD45.1+ Ly49H+ NK cells within the total CD3− NK1.1+ population after infection. d, CFSE-labeled wild-type NK cells (5×105) were transferred into DAP12-deficient hosts. Ly49H+ and Ly49H− NK cells were analyzed after infection. e, Percentages (left graph) and absolute numbers (right graph) of transferred CD45.1+ NK cells in DAP12-deficient or wild-type B6 recipients after infection. Error bars display s.e.m. (n = 3–5). Data are representative of 5 experiments.
Figure 3. Expansion and contraction of NK…
Figure 3. Expansion and contraction of NK cells in lymphoid and non-lymphoid tissues results in “memory” NK cells
a–b, Following adoptive transfer of 105 (squares) or 104 (circles) wild-type NK cells (CD45.1+) into DAP12-deficient mice and MCMV infection, percentages (left) and absolute numbers (right) of Ly49H+ NK cells in spleen (a) and liver (b). Error bars display s.e.m. (n = 3–5). Data are representative of 3 experiments. c, Fold expansions of Ly49H+ NK cells over 7 days of infection were calculated in B6 mice, in 1:1 and 1:5 wild-type:DAP12-deficient bone marrow chimeric mice, and following adoptive transfer of wild-type NK cells into DAP12-deficient mice. Error bars display s.e.m. from 3 experiments in each group of mice indicated.
Figure 4. Function and phenotype of “memory”…
Figure 4. Function and phenotype of “memory” NK cells
a, 105 wild-type NK cells (CD45.1+) transferred into DAP12-deficient mice (CD45.2+) were analyzed 70 days after MCMV infection. Left, percentage of CD45.1+ Ly49H+ cells within total NK cell population. Right, percentages of CD45.1+ NK cells producing IFN-γ after stimulation. b, “Memory” NK cells compared to naïve NK cells from uninfected mice after anti-NK1.1 stimulation. Percentages of Ly49H+ NK cells (gated on total NK cells) producing IFN-γ. Right, percentages of Ly49H+ NK cells producing IFN-γ (4 mice/group, horizontal bar is mean, p = 0.0009). c, LAMP-1 on “memory” NK cells (day 55 PI) versus naïve NK cells (gated on Ly49H+ NK cells) after anti-NK1.1 stimulation. d, Surface markers on “memory” NK cells (day 45 PI) versus naïve NK cells. e, NK cells from day 7 MCMV-infected Yeti mice were transferred into DAP12-deficient Rag2−/− mice. YFP in “memory” Ly49H+ NK cells after 28 days compared to Ly49H+ NK cells and T cells in uninfected Yeti mouse.
Figure 5. Secondary expansion and protective immunity…
Figure 5. Secondary expansion and protective immunity in “memory” NK cells
a, 105 wild-type NK cells (CD45.1+) transferred to DAP12-deficient mice (CD45.2+) were isolated 40–50 days after primary MCMV infection and transferred into DAP12-deficient mice (CD45.2+). Following infection of the second host, Ly49H+ NK cells were analyzed. b, Percentages of transferred “memory” Ly49H+ NK cells in the second host. c, Percentages (left) and absolute number (right) of Ly49H+ NK cells within the total NK cell population in second host. Error bars display s.e.m. (n = 3–5). Data are representative of 5 experiments. d, Analysis of CSFE-labeled “memory” and naïve NK cells transferred into DAP12-deficient recipient mice (day 3 and 6 PI). e, Expansion and contraction of “memory” and naïve NK cells (1×104 input) shown as a percentages (left) and absolute number (right) of Ly49H+ NK cells within the total NK cell population. Error bars display s.e.m. (n = 3–5). Data are representative of 3 experiments. f, Survival of DAP12-deficient neonatal mice receiving 1×104 or 1×105 naive, or 1×104 “memory” NK cells (or PBS as control) followed by MCMV infection, with or without anti-Ly49H blocking. Data were pooled from 3 experiments.
Figure 5. Secondary expansion and protective immunity…
Figure 5. Secondary expansion and protective immunity in “memory” NK cells
a, 105 wild-type NK cells (CD45.1+) transferred to DAP12-deficient mice (CD45.2+) were isolated 40–50 days after primary MCMV infection and transferred into DAP12-deficient mice (CD45.2+). Following infection of the second host, Ly49H+ NK cells were analyzed. b, Percentages of transferred “memory” Ly49H+ NK cells in the second host. c, Percentages (left) and absolute number (right) of Ly49H+ NK cells within the total NK cell population in second host. Error bars display s.e.m. (n = 3–5). Data are representative of 5 experiments. d, Analysis of CSFE-labeled “memory” and naïve NK cells transferred into DAP12-deficient recipient mice (day 3 and 6 PI). e, Expansion and contraction of “memory” and naïve NK cells (1×104 input) shown as a percentages (left) and absolute number (right) of Ly49H+ NK cells within the total NK cell population. Error bars display s.e.m. (n = 3–5). Data are representative of 3 experiments. f, Survival of DAP12-deficient neonatal mice receiving 1×104 or 1×105 naive, or 1×104 “memory” NK cells (or PBS as control) followed by MCMV infection, with or without anti-Ly49H blocking. Data were pooled from 3 experiments.

References

    1. Harty JT, Badovinac VP. Shaping and reshaping CD8+ T-cell memory. Nat Rev Immunol. 2008;8(2):107.
    1. Kaech SM, Wherry EJ, Ahmed R. Effector and memory T-cell differentiation: implications for vaccine development. Nat Rev Immunol. 2002;2(4):251.
    1. Sprent J, Surh CD. T cell memory. Annu Rev Immunol. 2002;20:551.
    1. Williams MA, Bevan MJ. Effector and memory CTL differentiation. Annu Rev Immunol. 2007;25:171.
    1. Butz EA, Bevan MJ. Massive expansion of antigen-specific CD8+ T cells during an acute virus infection. Immunity. 1998;8(2):167.
    1. Goldrath AW, Bevan MJ. Selecting and maintaining a diverse T-cell repertoire. Nature. 1999;402(6759):255.
    1. Murali-Krishna K, et al. Counting antigen-specific CD8 T cells: a reevaluation of bystander activation during viral infection. Immunity. 1998;8(2):177.
    1. Badovinac VP, Porter BB, Harty JT. Programmed contraction of CD8(+) T cells after infection. Nat Immunol. 2002;3(7):619.
    1. Jameson SC. Maintaining the norm: T-cell homeostasis. Nat Rev Immunol. 2002;2(8):547.
    1. Marrack P, Kappler J. Control of T cell viability. Annu Rev Immunol. 2004;22:765.
    1. Schluns KS, Lefrancois L. Cytokine control of memory T-cell development and survival. Nat Rev Immunol. 2003;3(4):269.
    1. Lefrancois L, Masopust D. T cell immunity in lymphoid and non-lymphoid tissues. Curr Opin Immunol. 2002;14(4):503.
    1. Masopust D, Vezys V, Marzo AL, Lefrancois L. Preferential localization of effector memory cells in nonlymphoid tissue. Science. 2001;291(5512):2413.
    1. Lanier LL. NK cell recognition. Annu Rev Immunol. 2005;23:225.
    1. Lanier LL. Back to the future--defining NK cells and T cells. Eur J Immunol. 2007;37(6):1424.
    1. Glas R, et al. Recruitment and activation of natural killer (NK) cells in vivo determined by the target cell phenotype. An adaptive component of NK cell-mediated responses. J Exp Med. 2000;191(1):129.
    1. O'Leary JG, Goodarzi M, Drayton DL, von Andrian UH. T cell- and B cell-independent adaptive immunity mediated by natural killer cells. Nat Immunol. 2006;7(5):507.
    1. Arase H, et al. Direct recognition of cytomegalovirus by activating and inhibitory NK cell receptors. Science. 2002;296(5571):1323.
    1. Brown MG, et al. Vital involvement of a natural killer cell activation receptor in resistance to viral infection. Science. 2001;292(5518):934.
    1. Daniels KA, et al. Murine cytomegalovirus is regulated by a discrete subset of natural killer cells reactive with monoclonal antibody to Ly49H. J Exp Med. 2001;194(1):29.
    1. Smith HR, et al. Recognition of a virus-encoded ligand by a natural killer cell activation receptor. Proc Natl Acad Sci U S A. 2002;99(13):8826.
    1. Vidal SM, Lanier LL. NK cell recognition of mouse cytomegalovirus-infected cells. Curr Top Microbiol Immunol. 2006;298:183.
    1. Dokun AO, et al. Specific and nonspecific NK cell activation during virus infection. Nat Immunol. 2001;2(10):951.
    1. Yokoyama WM, Kim S, French AR. The dynamic life of natural killer cells. Annu Rev Immunol. 2004;22:405.
    1. Bakker AB, et al. DAP12-deficient mice fail to develop autoimmunity due to impaired antigen priming. Immunity. 2000;13(3):345.
    1. Homann D, Teyton L, Oldstone MB. Differential regulation of antiviral T-cell immunity results in stable CD8+ but declining CD4+ T-cell memory. Nat Med. 2001;7(8):913.
    1. Williams MA, Ravkov EV, Bevan MJ. Rapid culling of the CD4+ T cell repertoire in the transition from effector to memory. Immunity. 2008;28(4):533.
    1. Stetson DB, et al. Constitutive cytokine mRNAs mark natural killer (NK) and NK T cells poised for rapid effector function. J Exp Med. 2003;198(7):1069.
    1. Badovinac VP, Messingham KA, Hamilton SE, Harty JT. Regulation of CD8+ T cells undergoing primary and secondary responses to infection in the same host. J Immunol. 2003;170(10):4933.
    1. Belz GT, et al. Minimal activation of memory CD8+ T cell by tissue-derived dendritic cells favors the stimulation of naive CD8+ T cells. Nat Immunol. 2007;8(10):1060.
    1. Grayson JM, et al. Differential sensitivity of naive and memory CD8+ T cells to apoptosis in vivo. J Immunol. 2002;169(7):3760.
    1. Jabbari A, Harty JT. Secondary memory CD8+ T cells are more protective but slower to acquire a central-memory phenotype. J Exp Med. 2006;203(4):919.
    1. Zimmermann C, Prevost-Blondel A, Blaser C, Pircher H. Kinetics of the response of naive and memory CD8 T cells to antigen: similarities and differences. Eur J Immunol. 1999;29(1):284.
    1. Bukowski JF, Warner JF, Dennert G, Welsh RM. Adoptive transfer studies demonstrating the antiviral effect of natural killer cells in vivo. J Exp Med. 1985;161(1):40.
    1. Sun JC, Bevan MJ. Cutting edge: long-lived CD8 memory and protective immunity in the absence of CD40 expression on CD8 T cells. J Immunol. 2004;172(6):3385.
    1. Bubic I, et al. Gain of virulence caused by loss of a gene in murine cytomegalovirus. J Virol. 2004;78(14):7536.

Source: PubMed

3
订阅