Lymphocyte Activation Gene 3 (LAG-3) modulates the ability of CD4 T-cells to be suppressed in vivo

Nicholas M Durham, Christopher J Nirschl, Christopher M Jackson, Jimmy Elias, Christina M Kochel, Robert A Anders, Charles G Drake, Nicholas M Durham, Christopher J Nirschl, Christopher M Jackson, Jimmy Elias, Christina M Kochel, Robert A Anders, Charles G Drake

Abstract

Lymphocyte Activation Gene - 3 (LAG-3) is an immune checkpoint molecule that regulates both T-cell activation and homeostasis. However, the molecular mechanisms underlying LAG-3's function are generally unknown. Using a model in which LAG-3 blockade or absence reliably augmented homeostatic proliferation in vivo, we found that IL-2 and STAT5 are critical for LAG-3 function. Similarly, LAG-3 blockade was ineffective in the absence of regulatory T-cells (Treg), suggesting an important role for LAG-3 in either the responsiveness of conventional T-cells (Tconv) to regulation, or a relative defect in the ability of LAG-3 KO regulatory T-cells (Treg) to suppress the proliferation of Tconv. In this model, LAG-3 KO Treg suppressed proliferation in a manner fairly similar to wild-type (WT) Treg, but LAG-3 KO Tconv were relatively resistant to suppression. Further studies also identified a role for LAG-3 in the induction/expansion of Treg. Finally, we found that LAG-3 blockade (or knockout) led to a relative skewing of naïve CD4 T-cells toward a TH1 phenotype both in vitro and in in vivo. Together, these data suggest that LAG-3 expression on Tconv cells makes them more susceptible to Treg based suppression, and also regulates the development of a TH1 T-cell response.

Conflict of interest statement

Competing Interests: CGD has served as a paid consultant for Bristol Myers Squibb, Compugen and Roche/Genentech. CGD is a co-inventor on a patent entitled: “Manipulation of Regulatory T Cells Using LAG-3 Antibodies, Genes, Agonists and Antagonists”, filing case number: C04255. This patent is currently un-licensed. This does not alter the authors’ adherence to PLOS ONE policies on sharing data and materials.

Figures

Figure 1. LAG-3 blockade augments homeostatic proliferation…
Figure 1. LAG-3 blockade augments homeostatic proliferation in vivo.
A) 1E6 WT or LAG-3 KO CD4+ T-cells were adoptively transferred into RAG KO mice and harvested on day 10. Splenocytes were then counted and analyzed. B) 1E6 WT CD4+ T-cells were transferred into RAG KO mice. Isotype control antibody or LAG-3 blocking antibody given every 2 days. Splenocytes were then counted and analyzed. C) LAG-3 antibody staining of LAG-3 in vivo. D) Serum IL-2 from RAG KO mice with 1E6 WT or LAG-3 KO CD4+ T-cells on Day 7. E) Percentage of CD4+ T-cells that express FOXP3. F) Total number of adoptively transferred CD4+ T-cells expressing FoxP3. Data shown are representative of at least two independent experiments with n = 3–6 mice per group.
Figure 2. IL-2 is required for LAG-3…
Figure 2. IL-2 is required for LAG-3 blockade to augment homeostatic proliferation in vivo.
A) 1E6 WT or IL-2 KO CD4 T-cells transferred into RAG KO mice. Isotype control antibody or LAG-3 blocking antibody given every 2 days. Splenocytes counted and analyzed. B) 1E6 WT or IL-2 KO CD4+ T-cells were transferred into RAG KO/IL-2 KO mice. C) 1E6 WT or STAT5 KO CD4+ T-cells transferred into RAG KO mice. Isotype control antibody or LAG-3 blocking antibody was given every 2 days. Splenocytes were then counted and analyzed. D) LAG-3 antibody staining of LAG-3 on IL-2 KO cells in vivo. E) LAG-3 antibody staining of LAG-3 on STAT5 KO cells in vivo. Data shown are representative of at least two independent experiments with n = 3 mice per group.
Figure 3. FOXP3 Treg are required for…
Figure 3. FOXP3 Treg are required for LAG-3 Blockade to augment homeostatic proliferation in vivo.
A) 1E6 FOXP3 DTR CD4+ T-cells transferred into RAG KO mice. Isotype control antibody or LAG-3 blocking antibody was given every 2 days with either PBS or Diphtheria Toxin. B) FOXP3 expression by flow cytometry ICS. C) Percent of CD4+ T-cells expressing FOXP3 D) Serum IL-2 levels by ELISA. Data shown are representative of at least two independent experiments with n = 4 mice per group.
Figure 4. LAG-3 KO Treg suppress homeostatic…
Figure 4. LAG-3 KO Treg suppress homeostatic proliferation, but LAG-3 KO responders are resistant to suppression.
A) WT or LAG-3 KO Treg were transferred into RAG KO mice at a ratio of 1∶4 with WT responders. Responders alone received 4E6 WT Tresp with no Treg. B) WT or LAG-3 KO Treg were transferred into RAG KO mice at a ratio of 1∶4 with KO responders. Responders alone received 4E6 LAG3 KO Tresp with no Treg. C) Representative plots of FOXP3 expression in adoptively transferred cells. D) Summary of FOXP3 expression in adoptively transferred cells (n = 4). E) CD25 MFI expression on Treg with either WT or LAG-3 KO Responders. F) WT or LAG-3 KO CD4 Tresp were mixed at a 4∶1 ratio with FOXP3 GFP Treg, stimulated with CD3/CD28, and pulsed with H3-Thymadine after 72 hours. Total CPM counts are shown. Baseline activation of WT or LAG-3 KO Tresp without Treg was not different and is reported by a dashed line. Data shown are representative of at least two independent experiments with n = 4–5 mice per group.
Figure 5. Decreased FOXP3 Treg induction in…
Figure 5. Decreased FOXP3 Treg induction in LAG-3 KO cells in vitro.
WT or LAG-3 KO 6.5 TCR Transgenic CD4+ T-cells skewed in Th1 or Treg Conditions and analyzed for A) FOXP3 or B) TBET Expression. C) 6.5 TCR transgenic CD4+ T-cells were isolated and mixed with matched splenocytes 1∶3 and stimulated with 1 or 10 µM HA peptide for 3 days. WT or LAG-3 KO 6.5 CD4+ T-cells were treated with αLAG-3 or isotype control antibody at 50 µg/mL and cells were stained for P-STAT5. D) Summary graph of two experiments. Data shown are representative of at least two independent experiments.
Figure 6. Decreased FOXP3 Treg induction in…
Figure 6. Decreased FOXP3 Treg induction in LAG-3 KO T-cells in an in vivo self-tolerance.
A) WT or LAG-3 KO 6.5 TCR Transgenic CD4+ T-cells adoptively transferred into C3-HA expressing mice. B) FOXP3 and C) Tbet expression was analyzed and representative graphs shown. C–D) Summary of FOXP3 expression in adoptively transferred cells. E–F) Summary of TBET expression in adoptively transferred cells. Data shown are representative of at least two independent experiments where n = 4 mice per group.
Figure 7. WT Treg Cannot Completely Protect…
Figure 7. WT Treg Cannot Completely Protect against LAG-3 KO Tresp in a Colitis Model.
A) WT or LAG-3 KO Tresp were transferred into RAG KO mice at a ratio of 4∶1 with WT Treg. Mice were weighed 3 times weekly for 50 days. Percentage of initial body weight is reported. B) Percentage of initial body weight at Day 49. C) H & E staining of histological sections of colons from the 4 groups of mice. D) Blinded histological score of colitis in mouse groups. E) Total splenocytes as well as total CD4+ T-cells were counted and analyzed. F) Percentage of CD4+ T-cells that were FOXP3 or TBET positive. Data shown are representative of at least two independent experiments with n = 8–10 mice per group.

References

    1. Sakaguchi S, Toda M, Asano M, Itoh M, Morse SS, et al. (1996) T cell-mediated maintenance of natural self-tolerance: its breakdown as a possible cause of various autoimmune diseases. J Autoimmun 9: 211–220 10.1006/jaut.1996.0026
    1. Sprent J, Cho J-H, Boyman O, Surh CD (2008) T cell homeostasis. Immunol Cell Biol 86: 312–319 10.1038/icb.2008.12
    1. Boyman O, Sprent J (2012) The role of interleukin-2 during homeostasis and activation of the immune system. Nat Rev Immunol 12: 180–190 10.1038/nri3156
    1. Sprent J, Surh CD (2011) Normal T cell homeostasis: the conversion of naive cells into memory-phenotype cells. Nat Immunol 131: 478–484 10.1038/ni.2018
    1. Surh CD (2000) Homeostatic T Cell Proliferation: How Far Can T Cells Be Activated to Self-Ligands? J Exp Med 192: 9F–14 10.1084/jem.192.4.F9
    1. Takeda S, Rodewald HR, Arakawa H, Bluethmann H, Shimizu T (1996) MHC class II molecules are not required for survival of newly generated CD4+ T cells, but affect their long-term life span. Immunity 5: 217–228.
    1. Hataye J, Moon JJ, Khoruts A, Reilly C, Jenkins MK (2006) Naive and memory CD4+ T cell survival controlled by clonal abundance. Science 312: 114–116 10.1126/science.1124228
    1. Rochman Y, Spolski R, Leonard WJ (2009) New insights into the regulation of T cells by gamma(c) family cytokines. Nat Rev Immunol 9: 480–490 10.1038/nri2580
    1. Leonard WJ, Shores EW, Love PE (1995) Role of the common cytokine receptor gamma chain in cytokine signaling and lymphoid development. Immunol Rev 148: 97–114.
    1. He Y, Nakajima H, Leonard W, Adkins B, Malek T (1997) The common gamma-chain of cytokine receptors regulates intrathymic T cell development at multiple stages. J Immunol 158: 2592–2599.
    1. DiSanto JP, Müller W, Guy-Grand D, Fischer A, Rajewsky K (1995) Lymphoid development in mice with a targeted deletion of the interleukin 2 receptor gamma chain. Proc Natl Acad Sci U S A 92: 377–381.
    1. Boyman O, Krieg C, Homann D, Sprent J (2012) Homeostatic maintenance of T cells and natural killer cells. Cell Mol Life Sci 69: 1597–1608 10.1007/s00018-012-0968-7
    1. Tan JT, Dudl E, LeRoy E, Murray R, Sprent J, et al. (2001) IL-7 is critical for homeostatic proliferation and survival of naive T cells. Proc Natl Acad Sci U S A 98: 8732–8737 10.1073/pnas.161126098
    1. Schluns KS, Kieper WC, Jameson SC, Lefrançois L (2000) Interleukin-7 mediates the homeostasis of naïve and memory CD8 T cells in vivo. Nat Immunol 1: 426–432 10.1038/80868
    1. Becker TC, Wherry EJ, Boone D, Murali-Krishna K, Antia R, et al. (2002) Interleukin 15 is required for proliferative renewal of virus-specific memory CD8 T cells. J Exp Med 195: 1541–1548.
    1. Bayer AL, Yu A, Adeegbe D, Malek TR (2005) Essential role for interleukin-2 for CD4(+)CD25(+) T regulatory cell development during the neonatal period. J Exp Med 201: 769–777 10.1084/jem.20041179
    1. Setoguchi R, Hori S, Takahashi T, Sakaguchi S (2005) Homeostatic maintenance of natural Foxp3(+) CD25(+) CD4(+) regulatory T cells by interleukin (IL)-2 and induction of autoimmune disease by IL-2 neutralization. J Exp Med 201: 723–735 10.1084/jem.20041982
    1. Malek TR, Castro I (2010) Interleukin-2 receptor signaling: at the interface between tolerance and immunity. Immunity 33: 153–165 10.1016/j.immuni.2010.08.004
    1. Fontenot JD, Rasmussen JP, Gavin MA, Rudensky AY (2005) A function for interleukin 2 in Foxp3-expressing regulatory T cells. Nat Immunol 6: 1142–1151 10.1038/ni1263
    1. Malek TR, Yu A, Vincek V, Scibelli P, Kong L (2002) CD4 regulatory T cells prevent lethal autoimmunity in IL-2Rbeta-deficient mice. Implications for the nonredundant function of IL-2. Immunity 17: 167–178.
    1. Jameson SC (2002) Maintaining the norm: T-cell homeostasis. Nat Rev Immunol 2: 547–556 10.1038/nri853
    1. Yadav M, Stephan S, Bluestone JA (2013) Peripherally induced tregs - role in immune homeostasis and autoimmunity. Front Immunol 4: 232 10.3389/fimmu.2013.00232
    1. Workman CJ, Vignali DAA (2005) Negative regulation of T cell homeostasis by lymphocyte activation gene-3 (CD223). J Immunol 174: 688–695.
    1. Hannier S, Tournier M, Bismuth G, Triebel F (1998) CD3/TCR complex-associated lymphocyte activation gene-3 molecules inhibit CD3/TCR signaling. J Immunol 161: 4058–4065.
    1. Blackburn SD, Shin H, Haining WN, Zou T, Workman CJ, et al. (2009) Coregulation of CD8+ T cell exhaustion by multiple inhibitory receptors during chronic viral infection. Nat Immunol 10: 29–37 Available: Accessed 2013 March 4..
    1. Huard B, Prigent P, Tournier M, Bruniquel D, Triebel F (1995) CD4/major histocompatibility complex class II interaction analyzed with CD4- and lymphocyte activation gene-3 (LAG-3)-Ig fusion proteins. Eur J Immunol 25: 2718–2721 10.1002/eji.1830250949
    1. Workman CJ, Dugger KJ, Vignali DAA (2002) Cutting edge: molecular analysis of the negative regulatory function of lymphocyte activation gene-3. J Immunol 169: 5392–5395.
    1. Nirschl CJ, Drake CG (2013) Molecular pathways: coexpression of immune checkpoint molecules: signaling pathways and implications for cancer immunotherapy. Clin Cancer Res 19: 4917–4924 Available: Accessed 2013 Nov 9..
    1. Grosso JF, Kelleher CC, Harris TJ, Maris CH, Hipkiss EL, et al. (2007) LAG-3 regulates CD8+ T cell accumulation and effector function in murine self- and tumor-tolerance systems. J Clin Invest 117: 3383–3392 Available: Accessed 2014 Feb 24..
    1. Grosso JF, Goldberg M V, Getnet D, Bruno TC, Yen H-R, et al. (2009) Functionally distinct LAG-3 and PD-1 subsets on activated and chronically stimulated CD8 T cells. J Immunol 182: 6659–6669 Available: Accessed 2014 March 9..
    1. Huang C-T, Workman CJ, Flies D, Pan X, Marson AL, et al. (2004) Role of LAG-3 in regulatory T cells. Immunity 21: 503–513 Available: Accessed 2014 March 9..
    1. Workman CJ, Wang Y, El Kasmi KC, Pardoll DM, Murray PJ, et al. (2009) LAG-3 regulates plasmacytoid dendritic cell homeostasis. J Immunol 182: 1885–1891 10.4049/jimmunol.0800185
    1. Lanoue A, Bona C, von Boehmer H, Sarukhan A (1997) Conditions that induce tolerance in mature CD4+ T cells. J Exp Med 185: 405–414.
    1. Workman CJ, Collison LW, Bettini M, Pillai MR, Rehg JE, et al. (2011) In vivo Treg suppression assays. Methods Mol Biol 707: 119–156 10.1007/978-1-61737-979-69
    1. Lin JX, Leonard WJ (2000) The role of Stat5a and Stat5b in signaling by IL-2 family cytokines. Oncogene 19: 2566–2576 10.1038/sj.onc.1203523
    1. Moriggl R, Topham DJ, Teglund S, Sexl V, McKay C, et al. (1999) Stat5 Is Required for IL-2-Induced Cell Cycle Progression of Peripheral T Cells. Immunity 10: 249–259 10.1016/S1074-7613(00)80025-4
    1. Antov A, Yang L, Vig M, Baltimore D, Van Parijs L (2003) Essential role for STAT5 signaling in CD25+CD4+ regulatory T cell homeostasis and the maintenance of self-tolerance. J Immunol 171: 3435–3441.
    1. Burchill MA, Yang J, Vang KB, Farrar MA (2007) Interleukin-2 receptor signaling in regulatory T cell development and homeostasis. Immunol Lett 114: 1–8 10.1016/j.imlet.2007.08.005
    1. Kim JM, Rasmussen JP, Rudensky AY (2007) Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice. Nat Immunol 8: 191–197 10.1038/ni1428
    1. Zabransky DJ, Nirschl CJ, Durham NM, Park B V, Ceccato CM, et al. (2012) Phenotypic and functional properties of Helios+ regulatory T cells. PLoS One 7: e34547 Available: Accessed 2013 March 28..
    1. Thornton AM, Korty PE, Tran DQ, Wohlfert EA, Murray PE, et al. (2010) Expression of Helios, an Ikaros transcription factor family member, differentiates thymic-derived from peripherally induced Foxp3+ T regulatory cells. J Immunol 184: 3433–3441 Available: Accessed 2013 June 10..
    1. Yadav M, Louvet C, Davini D, Gardner JM, Martinez-Llordella M, et al.. (2012) Neuropilin-1 distinguishes natural and inducible regulatory T cells among regulatory T cell subsets in vivo. J Exp Med 209: 1713–22, S1–19. Available: . Accessed 2013 May 27.
    1. Hansen W, Hutzler M, Abel S, Alter C, Stockmann C, et al. (2012) Neuropilin 1 deficiency on CD4+Foxp3+ regulatory T cells impairs mouse melanoma growth. J Exp Med 209: 2001–2016 Available: Accessed 2013 May 29..
    1. Sarris M, Andersen KG, Randow F, Mayr L, Betz AG (2008) Neuropilin-1 expression on regulatory T cells enhances their interactions with dendritic cells during antigen recognition. Immunity 28: 402–413 Available: 10.1016/j.immuni.2008.01.012 Accessed 2013 May 27..
    1. Delgoffe GM, Woo S-R, Turnis ME, Gravano DM, Guy C, et al. (2013) Stability and function of regulatory T cells is maintained by a neuropilin-1-semaphorin-4a axis. Nature 501: 252–256 Available: 10.1038/nature12428 Accessed 2013 May 26..
    1. Adler AJ, Marsh DW, Yochum GS, Guzzo JL, Nigam A, et al. (1998) CD4+ T cell tolerance to parenchymal self-antigens requires presentation by bone marrow-derived antigen-presenting cells. J Exp Med 187: 1555–1564.
    1. Andreae S, Buisson S, Triebel F (2003) MHC class II signal transduction in human dendritic cells induced by a natural ligand, the LAG-3 protein (CD223). Blood 102: 2130–2137 10.1182/blood-2003-01-0273
    1. Camisaschi C, Casati C, Rini F, Perego M, De Filippo A, et al. (2010) LAG-3 expression defines a subset of CD4(+)CD25(high)Foxp3(+) regulatory T cells that are expanded at tumor sites. J Immunol 184: 6545–6551 10.4049/jimmunol.0903879
    1. Okamura T, Fujio K, Shibuya M, Sumitomo S, Shoda H, et al. (2009) CD4+CD25-LAG3+ regulatory T cells controlled by the transcription factor Egr-2. Proc Natl Acad Sci U S A 106: 13974–13979 10.1073/pnas.0906872106
    1. Sega EI, Leveson-Gower DB, Florek M, Schneidawind D, Luong RH, et al. (2014) Role of lymphocyte activation gene-3 (Lag-3) in conventional and regulatory T cell function in allogeneic transplantation. PLoS One 9: e86551 Available: Accessed 2014 June 18..
    1. Butler NS, Moebius J, Pewe LL, Traore B, Doumbo OK, et al. (2012) Therapeutic blockade of PD-L1 and LAG-3 rapidly clears established blood-stage Plasmodium infection. Nat Immunol 13: 188–195 Available: Accessed 2013 March 21..
    1. Okazaki T, Okazaki I, Wang J, Sugiura D, Nakaki F, et al. (2011) PD-1 and LAG-3 inhibitory co-receptors act synergistically to prevent autoimmunity in mice. J Exp Med 208: 395–407 Available: Accessed 2013 March 29..
    1. Woo S-R, Turnis ME, Goldberg M V, Bankoti J, Selby M, et al. (2012) Immune inhibitory molecules LAG-3 and PD-1 synergistically regulate T-cell function to promote tumoral immune escape. Cancer Res 72: 917–927 Available: Accessed 2013 March 5..

Source: PubMed

3
Abonnieren