Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

Identification of lineage-specifying cytokines that signal all CD8+-cytotoxic-lineage-fate 'decisions' in the thymus

Abstract

T cell antigen receptor (TCR) signaling in the thymus initiates positive selection, but the CD8+-lineage fate is thought to be induced by cytokines after TCR signaling has ceased, although this remains controversial and unproven. We have identified four cytokines (IL-6, IFN-γ, TSLP and TGF-β) that did not signal via the common γ-chain (γc) receptor but that, like IL-7 and IL-15, induced expression of the lineage-specifying transcription factor Runx3d and signaled the generation of CD8+ T cells. Elimination of in vivo signaling by all six of these 'lineage-specifying cytokines' during positive selection eliminated Runx3d expression and completely abolished the generation of CD8+ single-positive thymocytes. Thus, this study proves that signaling during positive selection by lineage-specifying cytokines is responsible for all CD8+-lineage-fate 'decisions' in the thymus.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Figure 1: The induction of Runx3d by non-γc cytokine signals.
Figure 2: Non-γc cytokines generate SP8 cells during FTOC.
Figure 3: In vivo signaling by non-γc cytokines is required for the generation of γc-independent CD8+ T cells.
Figure 4: Non-γc cytokines do not generate a unique subset of CD8+ T cells.
Figure 5: TGF-β contributes to generation of γc-independent CD8+ T cells.
Figure 6: Elimination of in vivo signaling by all lineage-specifying cytokines.
Figure 7: In vivo cytokine signaling is always required for the generation of SP8 cells.

Similar content being viewed by others

References

  1. Starr, T.K., Jameson, S.C. & Hogquist, K.A. Positive and negative selection of T cells. Annu. Rev. Immunol. 21, 139–176 (2003).

    CAS  PubMed  Google Scholar 

  2. Carpenter, A.C. & Bosselut, R. Decision checkpoints in the thymus. Nat. Immunol. 11, 666–673 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Teh, H.S. et al. Thymic major histocompatibility complex antigens and the αβ T-cell receptor determine the CD4/CD8 phenotype of T cells. Nature 335, 229–233 (1988).

    CAS  PubMed  Google Scholar 

  4. Brugnera, E. et al. Coreceptor reversal in the thymus: signaled CD4+8+ thymocytes initially terminate CD8 transcription even when differentiating into CD8+ T cells. Immunity 13, 59–71 (2000).

    CAS  PubMed  Google Scholar 

  5. Singer, A. New perspectives on a developmental dilemma: the kinetic signaling model and the importance of signal duration for the CD4/CD8 lineage decision. Curr. Opin. Immunol. 14, 207–215 (2002).

    CAS  PubMed  Google Scholar 

  6. Singer, A., Adoro, S. & Park, J.H. Lineage fate and intense debate: myths, models and mechanisms of CD4- versus CD8-lineage choice. Nat. Rev. Immunol. 8, 788–801 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Littman, D.R. How Thymocytes Achieve Their Fate. J. Immunol. 196, 1983–1984 (2016).

    CAS  PubMed  Google Scholar 

  8. He, X. et al. The zinc finger transcription factor Th-POK regulates CD4 versus CD8 T-cell lineage commitment. Nature 433, 826–833 (2005).

    CAS  PubMed  Google Scholar 

  9. Kappes, D.J., He, X. & He, X. Role of the transcription factor Th-POK in CD4:CD8 lineage commitment. Immunol. Rev. 209, 237–252 (2006).

    CAS  PubMed  Google Scholar 

  10. Xiong, Y. & Bosselut, R. The enigma of CD4-lineage specification. Eur. J. Immunol. 41, 568–574 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Collins, A., Littman, D.R. & Taniuchi, I. RUNX proteins in transcription factor networks that regulate T-cell lineage choice. Nat. Rev. Immunol. 9, 106–115 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Taniuchi, I. et al. Differential requirements for Runx proteins in CD4 repression and epigenetic silencing during T lymphocyte development. Cell 111, 621–633 (2002).

    CAS  PubMed  Google Scholar 

  13. Park, J.H. et al. Signaling by intrathymic cytokines, not T cell antigen receptors, specifies CD8 lineage choice and promotes the differentiation of cytotoxic-lineage T cells. Nat. Immunol. 11, 257–264 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  14. Yu, Q., Erman, B., Bhandoola, A., Sharrow, S.O. & Singer, A. In vitro evidence that cytokine receptor signals are required for differentiation of double positive thymocytes into functionally mature CD8+ T cells. J. Exp. Med. 197, 475–487 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Kimura, M.Y. et al. Timing and duration of MHC I positive selection signals are adjusted in the thymus to prevent lineage errors. Nat. Immunol. 17, 1415–1423 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Xiong, Y. & Bosselut, R. CD4-CD8 differentiation in the thymus: connecting circuits and building memories. Curr. Opin. Immunol. 24, 139–145 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Kurd, N. & Robey, E.A. T-cell selection in the thymus: a spatial and temporal perspective. Immunol. Rev. 271, 114–126 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. McCaughtry, T.M. et al. Conditional deletion of cytokine receptor chains reveals that IL-7 and IL-15 specify CD8 cytotoxic lineage fate in the thymus. J. Exp. Med. 209, 2263–2276 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Decaluwe, H. et al. γc deficiency precludes CD8+ T cell memory despite formation of potent T cell effectors. Proc. Natl. Acad. Sci. USA 107, 9311–9316 (2010).

    CAS  PubMed  Google Scholar 

  20. Hanada, T. et al. A mutant form of JAB/SOCS1 augments the cytokine-induced JAK/STAT pathway by accelerating degradation of wild-type JAB/CIS family proteins through the SOCS-box. J. Biol. Chem. 276, 40746–40754 (2001).

    CAS  PubMed  Google Scholar 

  21. Voon, D.C., Hor, Y.T. & Ito, Y. The RUNX complex: reaching beyond haematopoiesis into immunity. Immunology 146, 523–536 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Vaillant, F. et al. A full-length Cbfa1 gene product perturbs T-cell development and promotes lymphomagenesis in synergy with myc. Oncogene 18, 7124–7134 (1999).

    CAS  PubMed  Google Scholar 

  23. Yoshimura, A., Naka, T. & Kubo, M. SOCS proteins, cytokine signalling and immune regulation. Nat. Rev. Immunol. 7, 454–465 (2007).

    CAS  PubMed  Google Scholar 

  24. Lee, K.S. et al. Runx2 is a common target of transforming growth factor β1 and bone morphogenetic protein 2, and cooperation between Runx2 and Smad5 induces osteoblast-specific gene expression in the pluripotent mesenchymal precursor cell line C2C12. Mol. Cell. Biol. 20, 8783–8792 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. El-Asady, R. et al. TGF-beta-dependent CD103 expression by CD8+ T cells promotes selective destruction of the host intestinal epithelium during graft-versus-host disease. J. Exp. Med. 201, 1647–1657 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Konkel, J.E. et al. Control of the development of CD8αα+ intestinal intraepithelial lymphocytes by TGF-β. Nat. Immunol. 12, 312–319 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Ouyang, W. et al. TGF-β cytokine signaling promotes CD8+ T cell development and low-affinity CD4+ T cell homeostasis by regulation of interleukin-7 receptor α expression. Immunity 39, 335–346 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Shi, M.J. & Stavnezer, J. CBFα3 (AML2) is induced by TGF-β1 to bind and activate the mouse germline Ig alpha promoter. J. Immunol. 161, 6751–6760 (1998).

    CAS  PubMed  Google Scholar 

  29. Kulkarni, A.B. & Karlsson, S. Transforming growth factor-β1 knockout mice. A mutation in one cytokine gene causes a dramatic inflammatory disease. Am. J. Pathol. 143, 3–9 (1993).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Shull, M.M. et al. Targeted disruption of the mouse transforming growth factor-β1 gene results in multifocal inflammatory disease. Nature 359, 693–699 (1992).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Egawa, T., Tillman, R.E., Naoe, Y., Taniuchi, I. & Littman, D.R. The role of the Runx transcription factors in thymocyte differentiation and in homeostasis of naive T cells. J. Exp. Med. 204, 1945–1957 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Kohu, K. et al. Overexpression of the Runx3 transcription factor increases the proportion of mature thymocytes of the CD8 single-positive lineage. J. Immunol. 174, 2627–2636 (2005).

    CAS  PubMed  Google Scholar 

  33. Grueter, B. et al. Runx3 regulates integrin alpha E/CD103 and CD4 expression during development of CD4-/CD8+ T cells. J. Immunol. 175, 1694–1705 (2005).

    CAS  PubMed  Google Scholar 

  34. Wang, L. et al. Distinct functions for the transcription factors GATA-3 and ThPOK during intrathymic differentiation of CD4+ T cells. Nat. Immunol. 9, 1122–1130 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Hunter, C.A. & Jones, S.A. IL-6 as a keystone cytokine in health and disease. Nat. Immunol. 16, 448–457 (2015).

    CAS  PubMed  Google Scholar 

  36. Schroder, K., Hertzog, P.J., Ravasi, T. & Hume, D.A. Interferon-γ: an overview of signals, mechanisms and functions. J. Leukoc. Biol. 75, 163–189 (2004).

    CAS  PubMed  Google Scholar 

  37. Ziegler, S.F. et al. The biology of thymic stromal lymphopoietin (TSLP). Adv. Pharmacol. 66, 129–155 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Travis, M.A. & Sheppard, D. TGF-β activation and function in immunity. Annu. Rev. Immunol. 32, 51–82 (2014).

    CAS  PubMed  Google Scholar 

  39. Rochman, Y. et al. Thymic stromal lymphopoietin-mediated STAT5 phosphorylation via kinases JAK1 and JAK2 reveals a key difference from IL-7-induced signaling. Proc. Natl. Acad. Sci. USA 107, 19455–19460 (2010).

    CAS  PubMed  Google Scholar 

  40. Adoro, S. et al. Coreceptor gene imprinting governs thymocyte lineage fate. EMBO J. 31, 366–377 (2012).

    CAS  PubMed  Google Scholar 

  41. Chen, W. et al. Conversion of peripheral CD4+CD25 naive T cells to CD4+CD25+ regulatory T cells by TGF-beta induction of transcription factor Foxp3. J. Exp. Med. 198, 1875–1886 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Lee, Y.J., Holzapfel, K.L., Zhu, J., Jameson, S.C. & Hogquist, K.A. Steady-state production of IL-4 modulates immunity in mouse strains and is determined by lineage diversity of iNKT cells. Nat. Immunol. 14, 1146–1154 (2013).

    CAS  PubMed  Google Scholar 

  43. Pobezinsky, L.A. et al. Let-7 microRNAs target the lineage-specific transcription factor PLZF to regulate terminal NKT cell differentiation and effector function. Nat. Immunol. 16, 517–524 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Dewas, C. et al. TSLP expression: analysis with a ZsGreen TSLP reporter mouse. J. Immunol. 194, 1372–1380 (2015).

    CAS  PubMed  Google Scholar 

  45. Takahama, Y., Letterio, J.J., Suzuki, H., Farr, A.G. & Singer, A. Early progression of thymocytes along the CD4/CD8 developmental pathway is regulated by a subset of thymic epithelial cells expressing transforming growth factor β. J. Exp. Med. 179, 1495–1506 (1994).

    CAS  PubMed  Google Scholar 

  46. Fadok, V.A. et al. Macrophages that have ingested apoptotic cells in vitro inhibit proinflammatory cytokine production through autocrine/paracrine mechanisms involving TGF-β, PGE2, and PAF. J. Clin. Invest. 101, 890–898 (1998).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Catlett, I.M. & Hedrick, S.M. Suppressor of cytokine signaling 1 is required for the differentiation of CD4+ T cells. Nat. Immunol. 6, 715–721 (2005).

    CAS  PubMed  Google Scholar 

  48. Fry, T.J. & Mackall, C.L. Interleukin-7: master regulator of peripheral T-cell homeostasis? Trends Immunol. 22, 564–571 (2001).

    CAS  PubMed  Google Scholar 

  49. Chou, C. et al. The transcription factor AP4 mediates resolution of chronic viral infection through amplification of germinal center B cell responses. Immunity 45, 570–582 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Egawa, T. & Littman, D.R. Transcription factor AP4 modulates reversible and epigenetic silencing of the Cd4 gene. Proc. Natl. Acad. Sci. USA 108, 14873–14878 (2011).

    CAS  PubMed  Google Scholar 

  51. Al-Shami, A. et al. A role for thymic stromal lymphopoietin in CD4+ T cell development. J. Exp. Med. 200, 159–168 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Larsson, J. et al. TGF-β signaling-deficient hematopoietic stem cells have normal self-renewal and regenerative ability in vivo despite increased proliferative capacity in vitro. Blood 102, 3129–3135 (2003).

    CAS  PubMed  Google Scholar 

  53. Egawa, T. & Littman, D.R. ThPOK acts late in specification of the helper T cell lineage and suppresses Runx-mediated commitment to the cytotoxic T cell lineage. Nat. Immunol. 9, 1131–1139 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank T. McCaughtry for initial contributions to the project; J.-H. Park, V. Lazarevic, D. Singer, X. Zhou and M. Kimura for critical reading of the manuscript; M. Kubo (Tokyo University of Science) for SOCSTg mice; W. Leonard (National Heart, Lung and Blood Institute) for TSLPRKO mice; W. Chen (National Institute of Dental and Craniofacial Research) for Tgfbr1fl mice; D. Littman (New York University) for Runx3dYFP mice; R. Bosselut (National Cancer Institute) for ThPOKKO mice; and S. Sharrow, A. Adams and L. Granger for flow cytometry. Supported by the Intramural Research Program of the US National Institutes of Health, the National Cancer Institute, the Center for Cancer Research and the US National Institutes of Health (R01AI097244-01A1 to T.E.).

Author information

Authors and Affiliations

Authors

Contributions

R.E. designed the study, performed experiments, analyzed data and contributed to the writing of the manuscript; T.K., X.T., T.I.G. and T.E. performed experiments, analyzed data and provided helpful discussions; A.A. and B.E. generated experimental mice; and A.S. designed and supervised the study, analyzed data and wrote the manuscript.

Corresponding author

Correspondence to Alfred Singer.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Cytokine receptors on developing thymocytes that can potentially signal Runx3d expression.

(a) Characterization of γc-independent SP8 cells. Stainings for maturation markers (top) and CD8 lineage markers (bottom) are shown for B6 (black) and γccKO (red) SP8 (CD8SP TCRβhi) cells, compared to DP (CD4+ CD8+) cells (shaded curves). (b) List of cytokines, cytokine receptor chains, and signaling molecules examined in this study. (c) Identification of intermediate stage (CD4+CD8loCD69+) thymocytes that have been signaled by MHC class I-dependent ligands to undergo positive selection and to differentiate into SP8 thymocytes. MHC class I-signaled thymocytes were obtained from MHCIIKOCD1dKO mice because MHCI-signaled thymocytes in these mice do not differentiate into CD4+ NKT cells. (d) Surface expression of non-γc cytokine receptor chains on intermediate stage thymocytes from MHCIIKOCD1dKO mice. Cytokine receptor stainings are shown in red, control stainings are in gray. One of two experiments is shown. (e) Schematic of the DP stimulation assay. Pre-selection DP thymocytes (CD69-CD4+CD8+) were electronically sorted and stimulated with PMA+Ionomycin for 16h; washed and rested in medium for 10h; and then cultured with cytokines for 16-20h. Cells were then harvested and their gene expression analyzed by quantitative PCR. (f) List of cytokines that we tested in the B6 DP stimulation assay and their upregulation of mRNAs encoding Runx3d, Runx1, Runx2, Cbfb and Bcl2 (−, no gene up-regulation; +, minimal gene up-regulation; ++, intermediate gene up-regulation; +++, high gene up-regulation).

Supplementary Figure 2 Effect of cytokines on the generation of SP8 cells in fetal thymic organ culture.

(a) Thymocyte profiles of E16.5 B6 thymic lobes before and after 5 days FTOC in medium (top and middle rows). Profile of TCRβhi thymocytes on d5 of FTOC (bottom row) and numbers in boxes within the profiles indicate frequency of cells within that box. (b) SP8 cell generation in γccKO FTOCs by non-γc cytokines. Bar graph of SP8 cell frequencies from γccKO FTOCs cultured with the indicated cytokines, with SP8 cell frequencies in each cytokine group compared to medium alone (horizontal red dashed line). As an additional comparison, SP8 cell frequencies from B6 FTOCs are also shown. Data are from 4-17 individual lobes combined from 2-6 experiments. (c) IL-13 fails to induce SP8 cell generation in FTOCs. Comparison of SP8 cell frequencies (left) and numbers (right) in γccKO FTOCs cultured for 5 days with or without IL-13. SP8 cells from B6 FTOC are shown for comparison (black bar). (d) Exogenous addition of any lineage-specifying cytokine increases SP8 cell generation in B6 FTOCs. Bar graph of SP8 cell frequencies (left) and numbers (right) in B6 FTOCs cultured for 5 days with the indicated cytokines compared to medium alone cultures. Mean and s.e.m. are shown. *P < 0.05, **P < 0.01, ***P < 0.001, ****P < 0.0001.

Supplementary Figure 3 Lymph node SP8 cells in CytoQuad mice.

Numbers of CD8 TCRβ+ LN T cells in the indicated mice. Mean and s.e.m. are shown.

Supplementary Figure 4 γc cytokines and non-γ c cytokines signal overlapping CD8+ T cell populations.

Onion diagram for schematic representation of cytokine redundancy. We think that, in normal mice, all SP8 cell generation is signaled by the γc cytokine IL-7. In the absence of IL-7 signaling, fewer SP8 cells are generated and these are signaled by the γc cytokine IL-15. In the absence of γc signaling, still fewer SP8 cells are generated and these are signaled by four non-γc cytokines (IL-6, IFN-γ, TSLP, and TGF-β). It is only in the absence of in vivo signaling by the other three non-γc cytokines (IL-6, IFN-γ, and TSLP) that the contribution of TGF-β signaling to SP8 cell generation is appreciated. Most importantly, elimination of in vivo signaling by all six of these cytokines eliminates SP8 cell generation.

Supplementary Figure 5 Analysis of STAT- and SMAD-binding sites in the Runx3d locus.

(a) Vista conservation analysis of murine and human Runx3d genomic regions. Peaks represent degree of conservation. Colored circles indicate potential binding sites for STATs (red) and SMADs (blue). (b) Comparison of surface CD103 expression on SP8 thymocytes from the indicated mice.

Supplementary Figure 6 Lineage-specifying transcription factors and the generation of SP8 cells.

(a) The Runx3dYFP/YFP DP stimulation assay (schematized in supplementary Fig. 1e) was performed with pre-selection DP thymocytes from Runx3d-deficient (Runx3dYFP/YFP) mice. After PMA+ionomycin stimulation, the Runx3dYFP/YFP DP thymocytes were stimulated with various cytokines and assessed for expression of Runx1 mRNA. (b) Generation of MHC-II specific SP8 cells in ThPOK-deficient mice requires cytokine signaling. Thymocyte profiles from the indicated mice. Numbers in boxes in the profiles indicate CD8SP cell frequencies. Mean and s.e.m. are shown.

Supplementary Figure 7 Signaling events in the thymus that generate SP8 thymocytes: all generation of SP8 cells requires signaling by lineage-specifying cytokines.

TCR signaling of DP thymocytes selectively terminates Cd8 gene expression, causing surface CD8 protein expression to decline. (a) In wildtype mice, MHC-I-specific TCR signaling eventually ceases because of the loss of surface CD8 protein expression, which allows cells to then be signaled by lineage-specifying cytokines. Signaling of positively selected thymocytes by lineage-specifying cytokines induces high Runx3d expression which both inhibits Runx1 and mediates co-receptor reversal (i.e. Cd4 gene silencing and Cd8 gene re-expression), resulting in differentiation into SP8 cells. (b) In the absence of intra-thymic signaling by lineage-specifying cytokines, Runx3d expression is not induced and Runx1 is unable to generate SP8 cells. (c) In Runx3d-deficient mice, intra-thymic signaling by lineage-specifying cytokines augments the ability of Runx1 to mediate coreceptor reversal and promote SP8 cell generation. (d) In ThPOK-deficient mice, CD4-lineage specification cannot occur. Because MHC-II-specific TCR signaling in the thymus must eventually cease, perhaps because of limiting ligand expression, cessation of TCR signaling allows the cells to then be signaled by lineage-specifying cytokines that induce high Runx3d expression which then inhibits Runx1 and mediates co-receptor reversal with the result that MHC-II-specific thymocytes differentiate into SP8 cells.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Etzensperger, R., Kadakia, T., Tai, X. et al. Identification of lineage-specifying cytokines that signal all CD8+-cytotoxic-lineage-fate 'decisions' in the thymus. Nat Immunol 18, 1218–1227 (2017). https://doi.org/10.1038/ni.3847

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3847

This article is cited by

Search

Quick links

Nature Briefing

Sign up for the Nature Briefing newsletter — what matters in science, free to your inbox daily.

Get the most important science stories of the day, free in your inbox. Sign up for Nature Briefing