Natural Killer Cells Regulate Th17 Cells After Autologous Hematopoietic Stem Cell Transplantation for Relapsing Remitting Multiple Sclerosis

Peter J Darlington, Brandon Stopnicki, Tarik Touil, Jean-Sebastien Doucet, Lama Fawaz, Morgan E Roberts, Marie-Noëlle Boivin, Nathalie Arbour, Mark S Freedman, Harold L Atkins, Amit Bar-Or, Peter J Darlington, Brandon Stopnicki, Tarik Touil, Jean-Sebastien Doucet, Lama Fawaz, Morgan E Roberts, Marie-Noëlle Boivin, Nathalie Arbour, Mark S Freedman, Harold L Atkins, Amit Bar-Or

Abstract

In autoimmunity, the balance of different helper T (Th) cell subsets can influence the tissue damage caused by autoreactive T cells. Pro-inflammatory Th1 and Th17 T cells are implicated as mediators of several human autoimmune conditions such as multiple sclerosis (MS). Autologous hematopoietic stem cell transplantation (aHSCT) has been tested in phase 2 clinical trials for MS patients with aggressive disease. Abrogation of new clinical relapses and brain lesions can be seen after ablative aHSCT, accompanied by significant reductions in Th17, but not Th1, cell populations and activity. The cause of this selective decrease in Th17 cell responses following ablative aHSCT is not completely understood. We identified an increase in the kinetics of natural killer (NK) cell reconstitution, relative to CD4+ T cells, in MS patients post-aHSCT, resulting in an increased NK cell:CD4+ T cell ratio that correlated with the degree of decrease in Th17 responses. Ex vivo removal of NK cells from post-aHSCT peripheral blood mononuclear cells resulted in higher Th17 cell responses, indicating that NK cells can regulate Th17 activity. NK cells were also found to be cytotoxic to memory Th17 cells, and this toxicity is mediated through NKG2D-dependent necrosis. Surprisingly, NK cells induced memory T cells to secrete more IL-17A. This was preceded by an early rise in T cell expression of RORC and IL17A mRNA, and could be blocked with neutralizing antibodies against CD58, a costimulatory receptor expressed on NK cells. Thus, NK cells provide initial co-stimulation that supports the induction of a Th17 response, followed by NKG2D-dependent cytotoxicity that limits these cells. Together these data suggest that rapid reconstitution of NK cells following aHSCT contribute to the suppression of the re-emergence of Th17 cells. This highlights the importance of NK cells in shaping the reconstituting immune system following aHSCT in MS patients.

Trial registration: ClinicalTrials.gov NCT01099930.

Keywords: CD58; NKG2D; Th17 cells; aHSCT; multiple sclerosis; natural killer cells.

Figures

Figure 1
Figure 1
Rapid reconstitution kinetics of natural killer (NK) cell counts compared with CD4 T cells in aHSCT-treated multiple sclerosis (MS) patient blood. From freshly drawn blood samples, absolute number of NK cell counts (A) and CD4+ T cell counts (B) were determined at baseline (BL), and at 3 weeks (3 W), 3 months (3 M), 12 months (12 M), and 21 months (21 M) post-aHSCT. The ratio of NK cells to CD4+ T cells was calculated (C). N = 7 patients.
Figure 2
Figure 2
The proportions of CD56dim and CD56bright natural killer (NK) cells are higher post-aHSCT compared witho baseline (BL). Cryopreserved peripheral blood mononuclear cell (PBMC) from aHSCT-treated MS patients was analyzed by flow cytometry. Representative plots for BL (A,C), and month 12 [M12; (B,D)] stained for CD3 and CD56 (A,B), gated on CD3−CD56+, then shown as CD56 with CD16 (C,D). Example gates for CD56bright and CD56dim are shown. Additional time points include BL, and serial samples approximately every 3 months for up to 24 months post-aHSCT. The average proportions of total NK cells as well as CD56dim and CD56bright NK cell subsets expressed as a proportion of total PBMC (E). The ratio of NK cells with the CD56bright phenotype calculated as CD56bright/CD56dim NK cells (F). The proportions of NK cells with CD16+ or CD16− phenotype expressed as a proportion of total PBMC (G). The ratio of gated NK cells with the CD16− phenotype calculated as CD16−/CD16+(H). N = 7 patients. For statistical analysis, the time points were grouped in M3–M6, M9–M12, M15–M18, and M21–M24, followed by univariate one-way ANOVA with pairwise comparisons with the BL values.
Figure 3
Figure 3
Changes in blood natural killer (NK) cell populations correlate with changes in helper T (Th) cells populations following aHSCT in MS patients. MS patient peripheral blood mononuclear cell (PBMC) samples from baseline (BL) and 12 month post-aHSCT were activated in vitro with anti-CD3, anti-CD28, and Th17 polarizing factors for 4 days. Th17 and Th1 cells were assessed by analysis of cytokine production by intracellular flow cytometry (CD3+CD4+IL-17A+IFN-γ− or CD3+CD4+IL-17A−IFN-γ+, respectively). The change in frequency of Th17 cells (A) or Th1 cells (B) was plotted against the change in NK cell frequency, and linear regression was performed on the data points. N = 7 patients.
Figure 4
Figure 4
CD56+ cells suppress Th17 and Th1 cell responses in aHSCT. CD56+ cells were depleted from MS patient PBMC samples collected at 12 months post-aHSCT. Representative plots are shown for complete (A,B) and CD56-depleted samples (C,D). Cells were activated in vitro with anti-CD3, anti-CD28, and Th17 polarizing factors (Act) for 4 days. The proportions of Th17 and Th1 cells were assessed by analysis of cytokine production by intracellular flow cytometry. Representative plots are shown for complete samples (B) and CD56-depleted samples (D). The average proportion of Th17 (E) or Th1 cells (F) is shown. N = 3 patients.
Figure 5
Figure 5
Natural killer (NK) cells reduce the proportion of Th17 and Th1 cells in vitro. Memory CD4+ T cells and NK cells were isolated from healthy subject PBMC. Memory CD4+ T cells were CD3+CD4+CD45RO+CD45RA− as shown in representative dot plots (A,B). NK cells were CD3−CD56+ as shown in a representative dot plot (C). Memory CD4+ T cells were cultured without (D,E), or with NK cells at a 1:1 ratio (F,G) for 4 days with anti-CD3, anti-CD28, and Th17 polarizing factors for 4 days. Plots are shown for CD4 × CD56, which was used to gate CD4+ helper T (Th) cells. Expression of IL-17 and IFN-γ by CD4+ T cells was assessed by intracellular flow cytometry (Th17 = CD3+CD4+IL-17A+IFN-γ−, Th1 = CD3+CD4+IL-17A−IFN-γ+, and Th1/17 = CD3+CD4+IL-17A+IFN-γ+). Data pooled from 12 experiments showing the proportion (H) and absolute number of Th cells (I). A time-course analysis for Th17 cells (J) and Th1 cells (K) was performed for 5 days using intracellular cytokine staining. Open diamond = T cells and closed square = T cells + NK cells.
Figure 6
Figure 6
Activated memory CD4+ T cells express MICA and are sensitive to NKG2D-mediated natural killer (NK) cell cytotoxicity. Memory CD4+ T cells were obtained from healthy subject PBMC, as described in Figure 5, and activated with anti-CD3, anti-CD28, and Th17 polarizing factors for 4 days. Expression of CD4, MICA (Zenon labeled), IL-17A, and IFN-γ were assessed by intracellular cytokine staining and flow cytometry. Representative plots of FSC × SSC (A), CD4 × SSC (B), and IL-17A × IFN-γ (C) are shown. MICA expression on Th17 (D), Th1 (E), and the Th0 cells (F) is shown. Open histogram indicate MICA stained cells and closed histograms indicate an isotype control. The average mean fluorescent intensity of MICA minus the isotype control is shown [ΔMFI; (G)]. Expression of CD56 and NKG2D by NK cells was assessed by flow cytometry and a representative plot is shown (H). NK cells were cultured with memory CD4+ T cells and activated with anti-CD3, anti-CD28, and Th17 polarizing factors, at the same time treated without antibody, (NIL), anti-NKG2D neutralizing antibody, or isotype control antibody (I). N = 7 samples.
Figure 7
Figure 7
Natural killer (NK) cells are cytotoxic toward helper T (Th) cells through necrosis but not apoptosis. Memory CD4+ T cells from healthy subject PBMC were activated with anti-CD3, anti-CD28, and Th17 polarizing factors, either in the absence or presence of NK cells for 4 days. Samples were stained with CD3, CD56, 7AAD, and annexin V. Representative plots of T cells (CD4+CD56−) cultured without (A) or with NK cells (B) are shown. The average proportion of necrotic (7-AAD+annexinV+) T cells is shown (C). Open bars = memory CD4 T cells, closed bars = memory CD4+ T cells plus NK cells. N = 8 samples. A time-course analysis of apoptotic (7-AAD−annexin V+) T cells is shown (D). Open circles indicate memory CD4 T cells alone and filled squares indicate T cells plus NK cells.
Figure 8
Figure 8
Natural killer (NK) cells augment IL-17A and RORC levels in memory CD4 T cells. Purified memory CD4+ T cells from healthy subject PBMCs were activated with anti-CD3, anti-CD28, and Th17 polarizing factors without (open diamond) or with NK cells (closed square). Cytokines IL-17A (A) and IFN-γ (B) were measured in the supernatant by enzyme-linked immunosorbent assay. Expression of RORC(C) and IL17A(D) mRNA was measured by qPCR at the indicated time points. Data are representative of three samples. Groups included non-activated memory CD4+ T cells (T nil; closed circles), activated memory CD4+ T cells (T; open circle), activated memory CD4+ T cells with NK cells (T NK; closed squares), and NK cells cultured alone with IL-2 (NK IL-2; open squares). Representative plots of IL-17A and IFN-γ expression in NK cells (CD3−CD56+) are shown (E,F).
Figure 9
Figure 9
Natural killer (NK) cells support IL-17A expression by helper T (Th) cells by CD58 co-stimulation. A representative plot of CD58 expression by CD3−CD56+ NK cells is shown from healthy subject peripheral blood mononuclear cell (PBMC) (A). Memory CD4+ T cells from healthy subjects PBMC were activated with anti-CD3, anti-CD28, and Th17 polarizing factors with NK cells in the presence or absence of CD58 neutralizing or isotype control antibody for 4 days. The cytokine IL-17A was assessed by enzyme-linked immunosorbent assay from cell culture supernatants (B). Graph indicates mean IL-17A concentration for N = 3 samples.
Figure 10
Figure 10
CD58 expression levels on natural killer (NK) cells before and after aHSCT treatment of multiple sclerosis (MS) patients. Cryopreserved peripheral blood mononuclear cell (PBMC) from the aHSCT cohort of MS patients was stained for CD3, CD56, and CD58. Representative plots for CD56 and CD58 are shown for baseline (BL) (A) and month 21 [M21; (B)]. A time series of samples from BL until month 24 (M24) is presented (C). For statistical analysis, the time points were grouped in M3–M6, M9–M12, M15–M18, and M21–M24, followed by univariate one-way ANOVA with pairwise comparisons with the BL values. N = 7 patients.

References

    1. Bar-Or A, Darlington PJ. The immunology of MS. In: Cohen JA, editor. MS Therapeutics. Cambridge UK: Cambridge University Press; (2011). p. 20–34.
    1. Kebir H, Kreymborg K, Ifergan I, Dodelet-Devillers A, Cayrol R, Bernard M, et al. Human TH17 lymphocytes promote blood-brain barrier disruption and central nervous system inflammation. Nat Med (2007) 13:1173–5.10.1038/nm1651
    1. Kebir H, Ifergan I, Alvarez JI, Bernard M, Poirier J, Arbour N, et al. Preferential recruitment of interferon-gamma-expressing TH17 cells in multiple sclerosis. Ann Neurol (2009) 66:390–402.10.1002/ana.21748
    1. Deniz G, Erten G, Kucuksezer UC, Kocacik D, Karagiannidis C, Aktas E, et al. Regulatory NK cells suppress antigen-specific T cell responses. J Immunol (2008) 180:850–7.10.4049/jimmunol.180.2.850
    1. Phan M, Chun S, Kim S, Ali AK, Lee S, Kim S, et al. Natural killer cell subsets and receptor expression in peripheral blood mononuclear cells of a healthy Korean population: reference range, influence of age and sex, and correlation between NK cell receptors and cytotoxicity. Hum Immunol (2017) 78:103–12.10.1016/j.humimm.2016.11.006
    1. Bernardini G, Antonangeli F, Bonanni V, Santoni A. Dysregulation of chemokine/chemokine receptor axes and NK cell tissue localization during diseases. Front Immunol (2016) 7:402.10.3389/fimmu.2016.00402
    1. Poli A, Michel T, Thérésine M, Andres E, Hentges F, Zimmer J. CD56bright natural killer (NK) cells: an important NK cell subset. Immunology (2009) 126:458–65.10.1111/j.1365-2567.2008.03027.x
    1. Melsen JE, Lugthart G, Lankester AC, Schilham MW. Human circulating and tissue-resident CD56bright natural killer cell populations. Front Immunol (2016) 7:262.10.3389/fimmu.2016.00262
    1. Lo CKC, Lam QLK, Sun L, Wang S, Ko K, Xu H, et al. Natural killer cell degeneration exacerbates experimental arthritis in mice via enhanced interleukin-17 production. Arthritis Rheum (2008) 58:2700–11.10.1002/art.23760
    1. Leavenworth JW, Wang X, Wenander CS, Spee P, Cantor H. Mobilization of natural killer cells inhibits development of collagen-induced arthritis. Proc Natl Acad Sci U S A (2011) 108:14584–9.10.1073/pnas.1112188108
    1. Fort MM, Leach MW, Rennick DM. A role for NK cells as regulators of CD4+ T cells in a transfer model of colitis. J Immunol (1998) 161:3256–61.
    1. Zhang B, Yamamura T, Kondo T, Fujiwara M, Tabira T. Regulation of experimental autoimmune encephalomyelitis by natural killer (NK) cells. J Exp Med (1997) 186:1677–87.10.1084/jem.186.10.1677
    1. Matsumoto Y, Kohyama K, Aikawa Y, Shin T, Kawazoe Y, Suzuki Y, et al. Role of natural killer cells and TCR gamma delta T cells in acute autoimmune encephalomyelitis. Eur J Immunol (1998) 28:1681–8.10.1002/(SICI)1521-4141(199805)28:05<1681::AID-IMMU1681>;2-T
    1. Xu W, Fazekas G, Hara H, Tabira T. Mechanism of natural killer (NK) cell regulatory role in experimental autoimmune encephalomyelitis. J Neuroimmunol (2005) 163:24–30.10.1016/j.jneuroim.2005.02.011
    1. Hao J, Liu R, Piao W, Zhou Q, Vollmer TL, Campagnolo DI, et al. Central nervous system (CNS)-resident natural killer cells suppress Th17 responses and CNS autoimmune pathology. J Exp Med (2010) 207:1907–21.10.1084/jem.20092749
    1. Benczur M, Petranyl GG, Palffy G, Varga M, Talas M, Kotsy B, et al. Dysfunction of natural killer cells in multiple sclerosis: a possible pathogenetic factor. Clin Exp Immunol (1980) 39:657–62.
    1. Vraneš Z, Poljaković Z, Marušić M. Natural killer cell number and activity in multiple sclerosis. J Neurol Sci (1989) 94:115–23.10.1016/0022-510X(89)90222-0
    1. Munschauer F, Hartrich L, Stewart C, Jacobs L. Circulating natural killer cells but not cytotoxic T lymphocytes are reduced in patients with active relapsing multiple sclerosis and little clinical disability as compared to controls. J Neuroimmunol (1995) 62:177–81.10.1016/0165-5728(95)00115-9
    1. Kastrukoff LF, Morgan NG, Zecchini D, White R, Petkau AJ, Satoh J, et al. A role for natural killer cells in the immunopathogenesis of multiple sclerosis. J Neuroimmunol (1998) 86:123–33.10.1016/S0165-5728(98)00014-9
    1. Takahashi K, Aranami T, Endoh M, Miyake S, Yamamura T. The regulatory role of natural killer cells in multiple sclerosis. Brain (2004) 127:1917–27.10.1093/brain/awh219
    1. Aranami T, Miyake S, Yamamura T. Differential expression of CD11c by peripheral blood NK cells reflects temporal activity of multiple sclerosis. J Immunol (2006) 177:5659–67.10.4049/jimmunol.177.8.5659
    1. Su N, Shi SX, Zhu X, Borazanci A, Shi F, Gan Y. Interleukin-7 expression and its effect on natural killer cells in patients with multiple sclerosis. J Neuroimmunol (2014) 276:180–6.10.1016/j.jneuroim.2014.08.618
    1. Caruana P, Lemmert K, Ribbons K, Lea R, Lechner-Scott J. Natural killer cell subpopulations are associated with MRI activity in a relapsing-remitting multiple sclerosis patient cohort from Australia. Mult Scler (2016) 23(11):1479–87.10.1177/1352458516679267
    1. Gross CC, Schulte-Mecklenbeck A, Runzi A, Kuhlmann T, Posevitz-Fejfar A, Schwab N, et al. Impaired NK-mediated regulation of T-cell activity in multiple sclerosis is reconstituted by IL-2 receptor modulation. Proc Natl Acad Sci U S A (2016) 113:E2973–82.10.1073/pnas.1524924113
    1. Laroni A, Armentani E, de Rosbo NK, Ivaldi F, Marcenaro E, Sivori S, et al. Dysregulation of regulatory CD56 bright NK cells/T cells interactions in multiple sclerosis. J Autoimmun (2016) 72:8–18.10.1016/j.jaut.2016.04.003
    1. Rabinovich BA, Li J, Shannon J, Hurren R, Chalupny J, Cosman D, et al. Activated, but not resting, T cells can be recognized and killed by syngeneic NK cells. J Immunol (2003) 170:3572–6.10.4049/jimmunol.170.7.3572
    1. Molinero LL, Domaica CI, Fuertes MB, Girart MV, Rossi LE, Zwirner NW. Intracellular expression of MICA in activated CD4 T lymphocytes and protection from NK cell-mediated MICA-dependent cytotoxicity. Hum Immunol (2006) 67:170–82.10.1016/j.humimm.2006.02.010
    1. Cerboni C, Zingoni A, Cippitelli M, Piccoli M, Frati L, Santoni A. Antigen-activated human T lymphocytes express cell-surface NKG2D ligands via an ATM/ATR-dependent mechanism and become susceptible to autologous NK-cell lysis. Blood (2007) 110:606–15.10.1182/blood-2006-10-052720
    1. Smeltz RB, Wolf NA, Swanborg RH. Inhibition of autoimmune T cell responses in the DA rat by bone marrow-derived NK cells in vitro: implications for autoimmunity. J Immunol (1999) 163:1390–7.
    1. Bielekova B, Catalfamo M, Reichert-Scrivner S, Packer A, Cerna M, Waldmann TA, et al. Regulatory CD56(bright) natural killer cells mediate immunomodulatory effects of IL-2Ralpha-targeted therapy (daclizumab) in multiple sclerosis. Proc Natl Acad Sci U S A (2006) 103:5941–6.10.1073/pnas.0601335103
    1. Gross CC, Ahmetspahic D, Ruck T, Schulte-Mecklenbeck A, Schwarte K, Jörgens S, et al. Alemtuzumab treatment alters circulating innate immune cells in multiple sclerosis. Neurol Neuroimmunol Neuroinflammation (2016) 3:e289.10.1212/NXI.0000000000000289
    1. Smith MD, Calabresi PA, Bhargava P. Dimethyl fumarate treatment alters NK cell function in multiple sclerosis. Eur J Immunol (2017) 48(2):380–3.10.1002/eji.201747277
    1. Saraste M, Irjala H, Airas L. Expansion of CD56bright natural killer cells in the peripheral blood of multiple sclerosis patients treated with interferon-beta. Neurol Sci (2007) 28:121.10.1007/s10072-007-0803-3
    1. Darlington PJ, Podjaski C, Horn KE, Costantino S, Blain M, Saikali P, et al. Innate immune-mediated neuronal injury consequent to loss of astrocytes. J Neuropathol Exp Neurol (2008) 67:590–9.10.1097/NEN.0b013e3181772cf6
    1. Lagumersindez-Denis N, Wrzos C, Mack M, Winkler A, van der Meer F, Reinert MC, et al. Differential contribution of immune effector mechanisms to cortical demyelination in multiple sclerosis. Acta Neuropathol (2017) 134(1):15–34.10.1007/s00401-017-1706-x
    1. Muraro PA, Pasquini M, Atkins HL, Bowen JD, Farge D, Fassas A, et al. Long-term outcomes after autologous hematopoietic stem cell transplantation for multiple sclerosis. JAMA Neurol (2017) 74:459–69.10.1001/jamaneurol.2016.5867
    1. Lee H, Narayanan S, Brown RA, Chen JT, Atkins HL, Freedman MS, et al. Brain atrophy after bone marrow transplantation for treatment of multiple sclerosis. Mult Scler (2017) 23:420–31.10.1177/1352458516650992
    1. Lee H, Nakamura K, Narayanan S, Brown R, Chen J, Atkins HL, et al. Impact of immunoablation and autologous hematopoietic stem cell transplantation on gray and white matter atrophy in multiple sclerosis. Mult Scler (2017).10.1177/1352458517715811
    1. Atkins HL, Bowman M, Allan D, Anstee G, Arnold DL, Bar-Or A, et al. Immunoablation and autologous haemopoietic stem-cell transplantation for aggressive multiple sclerosis: a multicentre single-group phase 2 trial. Lancet (2016) 388(10044):576–85.10.1016/S0140-6736(16)30169-6
    1. Darlington PJ, Touil T, Doucet JS, Gaucher D, Zeidan J, Gauchat D, et al. Diminished Th17 (not Th1) responses underlie multiple sclerosis disease abrogation after hematopoietic stem cell transplantation. Ann Neurol (2013) 73:341–54.10.1002/ana.23784
    1. Ullah MA, Hill GR, Tey SK. Functional reconstitution of natural killer cells in allogeneic hematopoietic stem cell transplantation. Front Immunol (2016) 7:144.10.3389/fimmu.2016.00144
    1. Karnell FG, Lin D, Motley S, Duhen T, Lim N, Campbell DJ, et al. Reconstitution of immune cell populations in multiple sclerosis patients after autologous stem cell transplantation. Clin Exp Immunol (2017) 189(3):268–78.10.1111/cei.12985
    1. Larochelle C, Cayrol R, Kebir H, Alvarez JI, Lecuyer M, Ifergan I, et al. Melanoma cell adhesion molecule identifies encephalitogenic T lymphocytes and promotes their recruitment to the central nervous system. Brain (2012) 135:2906–24.10.1093/brain/aws212
    1. Pittet CL, Newcombe J, Antel JP, Arbour N. The majority of infiltrating CD8 T lymphocytes in multiple sclerosis lesions is insensitive to enhanced PD-L1 levels on CNS cells. Glia (2011) 59:841–56.10.1002/glia.21158
    1. Trivedi PP, Roberts PC, Wolf NA, Swanborg RH. NK cells inhibit T cell proliferation via p21-mediated cell cycle arrest. J Immunol (2005) 174:4590–7.10.4049/jimmunol.174.8.4590
    1. Abrahamsson SV, Angelini DF, Dubinsky AN, Morel E, Oh U, Jones JL, et al. Non-myeloablative autologous haematopoietic stem cell transplantation expands regulatory cells and depletes IL-17 producing mucosal-associated invariant T cells in multiple sclerosis. Brain (2013) 136:2888–903.10.1093/brain/awt182
    1. Muraro PA, Douek DC, Packer A, Chung K, Guenaga FJ, Cassiani-Ingoni R, et al. Thymic output generates a new and diverse TCR repertoire after autologous stem cell transplantation in multiple sclerosis patients. J Exp Med (2005) 201:805–16.10.1084/jem.20041679
    1. Schuster IS, Wikstrom ME, Brizard G, Coudert JD, Estcourt MJ, Manzur M, et al. TRAIL NK cells control CD4 T cell responses during chronic viral infection to limit autoimmunity. Immunity (2014) 41:646–56.10.1016/j.immuni.2014.09.013
    1. Roncarolo MG, Bigler M, Haanen JB, Yssel H, Bacchetta R, de Vries JE, et al. Natural killer cell clones can efficiently process and present protein antigens. J Immunol (1991) 147:781–7.
    1. Zingoni A, Sornasse T, Cocks BG, Tanaka Y, Santoni A, Lanier LL. Cross-talk between activated human NK cells and CD4+ T cells via OX40-OX40 ligand interactions. J Immunol (2004) 173:3716–24.10.4049/jimmunol.173.6.3716
    1. Nielsen N, Ødum N, Ursø B, Lanier LL, Spee P. Cytotoxicity of CD56bright NK cells towards autologous activated CD4 T cells is mediated through NKG2D, LFA-1 and TRAIL and dampened via CD94/NKG2A. PLoS One (2012) 7:e31959.10.1371/journal.pone.0031959

Source: PubMed

Подписаться