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:

A regulatory role for TGF-β signaling in the establishment and function of the thymic medulla

Abstract

Medullary thymic epithelial cells (mTECs) are critical in establishing and maintaining the appropriate microenvironment for negative selection and maturation of immunocompetent T cells with a self-tolerant T cell antigen receptor repertoire. Cues that direct proliferation and maturation of mTECs are provided by members of the tumor necrosis factor (TNF) superfamily expressed on developing thymocytes. Here we demonstrate a negative role of the morphogen TGF-β in tempering these signals under physiological conditions, limiting both growth and function of the thymic medulla. Eliminating TGF-β signaling specifically in TECs or by pharmacological means increased the size of the mTEC compartment, enhanced negative selection and functional maturation of medullary thymocytes as well as the production of regulatory T cells, thus reducing the autoreactive potential of peripheral T cells.

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: Enhanced mTEC expansion in the absence of TEC-specific TGF-β signaling.
Figure 2: Altered TGF-β signaling in TEC affects the maturation of mTECs.
Figure 3: TGF-β dampens the thymocyte-derived signals required for mTEC differentiation.
Figure 4: TGF-β and its target gene Myc affect proliferation of mTECs.
Figure 5: Consequences of the altered medulla in Tgfbr2fl/flFN1-Cre mice on thymocyte development and export.
Figure 6: Increased generation of Treg cells in the thymus of Tgfbr2fl/flFN1-Cre mice.
Figure 7: Reduced autoimmunity in Tgfbr2fl/flFN1-Cre mice upon chronic in vivo ablation of Foxp3+ regulatory T cells.

Similar content being viewed by others

References

  1. Anderson, G. & Takahama, Y. Thymic epithelial cells: working class heroes for T cell development and repertoire selection. Trends Immunol. 33, 256–263 (2012).

    CAS  PubMed  Google Scholar 

  2. Rode, I. & Boehm, T. Regenerative capacity of adult cortical thymic epithelial cells. Proc. Natl. Acad. Sci. USA 109, 3463–3468 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Stritesky, G.L., Jameson, S.C. & Hogquist, K.A. Selection of self-reactive T cells in the thymus. Annu. Rev. Immunol. 30, 95–114 (2012).

    CAS  PubMed  Google Scholar 

  4. Wirnsberger, G., Hinterberger, M. & Klein, L. Regulatory T-cell differentiation versus clonal deletion of autoreactivethymocytes. Immunol. Cell Biol. 89, 45–53 (2011).

    PubMed  Google Scholar 

  5. van Ewijk, W. The thymus: “Interactive teaching during lymphopoiesis”. Immunol. Lett. 138, 7–8 (2011).

    CAS  PubMed  Google Scholar 

  6. Kajiura, F. et al. NF-kappa B-inducing kinase establishes self-tolerance in a thymicstroma-dependent manner. J. Immunol. 172, 2067–2075 (2004).

    CAS  PubMed  Google Scholar 

  7. Akiyama, T. et al. The tumor necrosis factor family receptors RANK and CD40 cooperatively establish the thymicmedullary microenvironment and self-tolerance. Immunity 29, 423–437 (2008).

    CAS  PubMed  Google Scholar 

  8. Hikosaka, Y. et al. The cytokine RANKL produced by positively selected thymocytes fosters medullary thymic epithelial cells that express autoimmune regulator. Immunity 29, 438–450 (2008).

    CAS  PubMed  Google Scholar 

  9. Irla, M. et al. Autoantigen-specific interactions with CD4(+) thymocytes control mature medullary thymic epithelial cell cellularity. Immunity 29, 451–463 (2008).

    CAS  PubMed  Google Scholar 

  10. Nitta, T., Ohigashi, I., Nakagawa, Y. & Takahama, Y. Cytokine crosstalk for thymic medulla formation. Curr. Opin. Immunol. 23, 190–197 (2011).

    CAS  PubMed  Google Scholar 

  11. Venanzi, E.S., Gray, D.H.D., Benoist, C. & Mathis, D. Lymphotoxin pathway and Aire influences on thymic medullary epithelial cells are unconnected. J. Immunol. 179, 5693–5700 (2007).

    CAS  PubMed  Google Scholar 

  12. Derbinski, J. & Kyewski, B. How thymic antigen presenting cells sample the body's self-antigens. Curr. Opin. Immunol. 22, 592–600 (2010).

    CAS  PubMed  Google Scholar 

  13. Zuklys, S. et al. Normal thymic architecture and negative selection are associated with Aire expression, the gene defective in the autoimmune-polyendocrinopathy-candidiasis-ectodermal dystrophy (APECED). J. Immunol. 165, 1976–1983 (2000).

    CAS  PubMed  Google Scholar 

  14. Anderson, M.S. et al. Projection of an immunological self shadow within the thymus by the aire protein. Science 298, 1395–1401 (2002).

    CAS  PubMed  Google Scholar 

  15. Anderson, M.S. & Su, M.A. Aire and T cell development. Curr. Opin. Immunol. 23, 198–206 (2011).

    CAS  PubMed  Google Scholar 

  16. Massagué, J. TGFβ signalling in context. Nat. Rev. Mol. Cell Biol. 13, 616–630 (2012).

    PubMed  PubMed Central  Google Scholar 

  17. 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 beta. J. Exp. Med. 179, 1495–1506 (1994).

    CAS  PubMed  Google Scholar 

  18. Chen, W., Frank, M.E., Jin, W. & Wahl, S.M. TGF-beta released by apoptotic T cells contributes to an immunosuppressive milieu. Immunity 14, 715–725 (2001).

    CAS  PubMed  Google Scholar 

  19. Li, M.O. & Flavell, R.A. TGF-beta: a master of all T cell trades. Cell 134, 392–404 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Hauri-Hohl, M.M. et al. TGF-beta signaling in thymic epithelial cells regulates thymic involution and postirradiation reconstitution. Blood 112, 626–634 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Zuklys, S. et al. Stabilized beta-catenin in thymic epithelial cells blocks thymus development and function. J. Immunol. 182, 2997–3007 (2009).

    CAS  PubMed  Google Scholar 

  22. Bonnon, C. & Atanasoski, S. c-Ski in health and disease. Cell Tissue Res. 347, 51–64 (2012).

    CAS  PubMed  Google Scholar 

  23. Rossi, S.W. et al. RANK signals from CD4+3- inducer cells regulate development of Aire-expressing epithelial cells in the thymic medulla. J. Exp. Med. 204, 1267–1272 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Meilin, A., Shoham, J., Schreiber, L. & Sharabi, Y. The role of thymocytes in regulating thymic epithelial cell growth and function. Scand. J. Immunol. 42, 185–190 (1995).

    CAS  PubMed  Google Scholar 

  25. Trumpp, A. et al. c-Myc regulates mammalian body size by controlling cell number but not cell size. Nature 414, 768–773 (2001).

    CAS  PubMed  Google Scholar 

  26. Gomis, R.R., Alarcón, C., Nadal, C., Van Poznak, C. & Massagué, J. C/EBPbeta at the core of the TGFbeta cytostatic response and its evasion in metastatic breast cancer cells. Cancer Cell 10, 203–214 (2006).

    CAS  PubMed  Google Scholar 

  27. Kurobe, H. et al. CCR7-dependent cortex-to-medulla migration of positively selected thymocytes is essential for establishing central tolerance. Immunity 24, 165–177 (2006).

    CAS  PubMed  Google Scholar 

  28. Nitta, T., Nitta, S., Lei, Y., Lipp, M. & Takahama, Y. CCR7-mediated migration of developing thymocytes to the medulla is essential for negative selection to tissue-restricted antigens. Proc. Natl. Acad. Sci. USA 106, 17129–17133 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Hsu, F.-C., Pajerowski, A.G., Nelson-Holte, M., Sundsbak, R. & Shapiro, V.S. NKAP is required for T cell maturation and acquisition of functional competency. J. Exp. Med. 208, 1291–1304 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Eck, S.C. et al. Developmental alterations in thymocyte sensitivity are actively regulated by MHC class II expression in the thymic medulla. J. Immunol. 176, 2229–2237 (2006).

    CAS  PubMed  Google Scholar 

  31. Li, J. et al. Developmental pathway of CD4+CD8− medullary thymocytes during mouse ontogeny and its defect in Aire−/− mice. Proc. Natl. Acad. Sci. USA 104, 18175–18180 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Stephen, T.L., Tikhonova, A., Riberdy, J.M. & Laufer, T.M. The activation threshold of CD4+ T cells is defined by TCR/peptide-MHC class II interactions in the thymic medulla. J. Immunol. 183, 5554–5562 (2009).

    CAS  PubMed  Google Scholar 

  33. Stephen, T.L., Wilson, B.S. & Laufer, T.M. Subcellular distribution of Lck during CD4 T-cell maturation in the thymic medulla regulates the T-cell activation threshold. Proc. Natl. Acad. Sci. USA 109, 7415–7420 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Leung, M.W.L., Shen, S. & Lafaille, J.J. TCR-dependent differentiation of thymic Foxp3+ cells is limited to small clonal sizes. J. Exp. Med. 206, 2121–2130 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Bautista, J.L. et al. Intraclonal competition limits the fate determination of regulatory T cells in the thymus. Nat. Immunol. 10, 610–617 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Tang, Q. et al. Cutting edge: CD28 controls peripheral homeostasis of CD4+CD25+ regulatory T cells. J. Immunol. 171, 3348–3352 (2003).

    CAS  PubMed  Google Scholar 

  37. Aschenbrenner, K. et al. Selection of Foxp3+ regulatory T cells specific for self antigen expressed and presented by Aire+ medullary thymic epithelial cells. Nat. Immunol. 8, 351–358 (2007).

    CAS  PubMed  Google Scholar 

  38. Burchill, M.A et al. Linked T cell receptor and cytokine signaling govern the development of the regulatory T cell repertoire. Immunity 28, 112–121 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Hinterberger, M., Wirnsberger, G. & Klein, L. B7/CD28 in central tolerance: costimulation promotes maturation of regulatory T cell precursors and prevents their clonal deletion. Front. Immunol. 2, 30 (2011).

    PubMed  PubMed Central  Google Scholar 

  40. Klein, L. & Jovanovic, K. Regulatory T cell lineage commitment in the thymus. Semin. Immunol. 23, 401–409 (2011).

    CAS  PubMed  Google Scholar 

  41. Spence, P.J. & Green, E.A. Foxp3+ regulatory T cells promiscuously accept thymic signals critical for their development. Proc. Natl. Acad. Sci. USA 105, 973–978 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Coquet, J.M. et al. Epithelial and dendritic cells in the thymic medulla promote CD4+Foxp3+ regulatory T cell development via the CD27–CD70 pathway. J. Exp. Med. 210, 715–728 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Liston, A. et al. Differentiation of regulatory Foxp3+ T cells in the thymic cortex. Proc. Natl. Acad. Sci. USA 105, 11903–11908 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Kim, J.M., Rasmussen, J.P. & Rudensky, A.Y. Regulatory T cells prevent catastrophic autoimmunity throughout the lifespan of mice. Nat. Immunol. 8, 191–197 (2007).

    CAS  PubMed  Google Scholar 

  45. Marks, B.R. et al. Thymic self-reactivity selects natural interleukin 17-producing T cells that can regulate peripheral inflammation. Nat. Immunol. 10, 1125–1132 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Kim, J.S., Smith-Garvin, J.E., Koretzky, G.A. & Jordan, M.S. The requirements for natural Th17 cell development are distinct from those of conventional Th17 cells. J. Exp. Med. 208, 2201–2207 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  47. Villunger, A. et al. Negative selection of semimatureCD4(+)8(-)HSA+ thymocytes requires the BH3-only protein Bim but is independent of death receptor signaling. Proc. Natl. Acad. Sci. USA 101, 7052–7057 (2004).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. von Boehmer, H. & Kisielow, P. Negative selection of the T-cell repertoire: where and when does it occur? Immunol. Rev. 209, 284–289 (2006).

    PubMed  Google Scholar 

  49. Le Borgne, M. et al. The impact of negative selection on thymocyte migration in the medulla. Nat. Immunol. 10, 823–830 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Hirota, K. et al. T cell self-reactivity forms a cytokine milieu for spontaneous development of IL-17+ Th cells that cause autoimmune arthritis. J. Exp. Med. 204, 41–47 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank D.J. Campbell and J.A. Hamerman for helpful discussions about these data, and S. McCarty for help with preparation of the manuscript. This work was partially supported by grants from the Swiss National Science Foundation (M.H.-H. and G.A.H.).

Author information

Authors and Affiliations

Authors

Contributions

M.H.-H. performed all of the experiments in the manuscript, with help from S.Z.; M.H.-H., G.A.H. and S.F.Z. designed the experiments, analyzed data and contributed to writing the manuscript.

Corresponding author

Correspondence to Steven F Ziegler.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 FN1-Cre–mediated deletion of TGFβRII does not affect the phenotype of cTECs.

(a) Assessment of FN1-Cre-mediated genomic deletion of the floxed sequence in total TEC sorted from E14 thymic stroma, derived from either TβRIIlox/lox/FN1-Cre (denoted ‘Crepos’) or TβRIIlox/lox (‘Creneg’) embryos. (b) Gating strategy to identify TEC subpopulations from total thymic tissue, digested with Collagenase and DNAse. (c) Characterization of the cTEC compartment in 4-week-old TβRIIlox/lox/FN1-Cre mice or littermate controls for the expression of MHCII, β5t and DEC205.

Supplementary Figure 2 Altered expression of TRA with preserved thymic architecture in thymi with altered TGF-β signaling.

(a) Thymic section derived from 4 week old TβRIIlox/lox/FN1Cre or littermates stained for H&E, cytokeratin-5, cytokeratin-14 or UEA1 respectively. (b) Expression of indicated TRAs within total thymus of TβRIIlox/lox/FN1Cre (white bars, N=4), relative to expression found in littermate controls (black bars, N=5). (c) Absolute numbers of thymocytes and (d) relative distribution of thymocyte subpopulations in 6-8 week-old C57Bl/6 mice treated daily with SB431542 (white bars, N=8) or vehicle only (black bars, N=8) for 6 consecutive days, analysed on day 12 after the first injection. For statistical analysis, a two-tailed, unpaired t-test was used; NS denotes not statistically different whereas asterisks indicate a p-value of ≤0.05 (*),≤0.01 (**) or ≤0.001(***).

Supplementary Figure 3 TGF-β limits RANKL-induced mTEC differentiation.

Lymphocyte-depleted fetal thymi were cultured in the presence of the indicated stimuli for 5 days and assessed for the up-regulation of MHCII and CD80 within the mTEC compartment. Note the reduction in MHCIIhigh and CD80pos cells in the presence of RANKL/TGFβ vs RANKL alone, and a corresponding increase in the MHCIIlow population.

Supplementary Figure 4 Reduced thymic cellularity but normal thymocyte development and maturation in mice with c-myc–deficient TECs.

(a) Thymocyte cellularity and (b) relative distribution of thymocyte subpopulations with respect to CD4 and CD8 (upper panel) or CD69 and TCRβ (lower panel) expression in 4 week old c-myclox/lox/FN1-Cre mice (white bar) and littermate controls (black bar).

Supplementary Figure 5 Thymocyte maturation, export and functional changes of peripheral T cells in Tgfbr2fl/flFN1-Cre mice.

(a) Absolute numbers of DP and DN thymocytes in 4 week-old TβRIIlox/lox/FN1-Cre mice (white bars, N=5) and controls (black bars, N=5). (b) Flow cytometry profiles (left panel) of thymocytes for CD69 and TCR□ expression derived from 4-week-old mice and (right panel) statistical summary of the indicated subpopulations (R3: TCRintCD69pos, R4: TCRhighCD69pos, R5: TCRhighCD69neg). White bars denote TβRIIlox/lox/FN1-Cre mice (N=5), black bars littermate controls (N=5). (c) Absolute numbers of immature (i.e. CD24highCD62Llow) CD4 or CD8 TCRhigh SP thymocytes from TβRIIlox/lox/FN1-Cre mice (white bars, N=5) and controls (black bars, N=7). (d) GFP expression in immature (upper panel) and mature (lower panel) TCRhigh SP4 thymocytes derived from either controls (shaded histogram) or TβRIIlox/lox/FN1-Cre mice (open histogram), which were lethally irradiated and reconstituted with T-cell-depleted bone marrow cells from congenically marked mice that express GFP under the control of the Rag2-promoter (representative of 3 (TβRIIlox/lox/FN1-Cre mice) and 4 (littermate controls) mice from two independent experiments are shown). (e) Percentage of splenic CD4 or CD8 T cells in TβRIIlox/lox/FN1-Cre mice and littermate controls at the age of 3 days (left panel) and 6 days (right panel). White bars denote TβRIIlox/lox/FN1-Cre mice, black bars littermate controls. Two individual litters per time point with a minimum of total 3 mice per genotype are shown. (f) Absolute numbers of FITCpos recent thymic emigrants in peripheral lymphoid organs recovered 24 hours after intrathymic injection of 10μl FITC. White bars denote TβRIIlox/lox/FN1-Cre mice (N=16), black bars littermate controls (N=14). (g) Frequency of CD69pos cells within sorted peripheral naïve CD4 T cells from TβRIIlox/lox/FN1-Cre mice (white bars) or littermate controls (black bars) 20 hours after stimulation with the indicated stimuli. (h) Proliferation assessed by CFSE-dilution in sorted peripheral naïve CD4 T cells derived from TβRIIlox/loxFN1-Cre mice (upper histogram) and littermate controls (lower histogram) after a 4-day stimulation culture with 1μl/ml α-CD3 and 5μg/ml α-CD28. For statistical analysis, a two-tailed, unpaired t-test was used; NS denotes not statistically different whereas asterisks indicate a p-value of ≤0.05 (*), ≤0.01 (**) or ≤0.001(***).

Supplementary Figure 6 Reduced frequency of natural TH17 cells in thymus and spleen of Tgfbr2fl/flFN1-Cre mice.

Flow cytometry profiles (left panel) and corresponding bar graphs and statistics (right panel) of CD4SP TCRhigh thymocytes (a) and splenic CD4pos T cells (b) derived from 4-week-old mice after a 4 hour culture with PMA, Ionomycin and Brefeledin A. Data shown from 2 experiments with a total of 9 mice (asterisks indicate a p-value of ≤0.05 (*) or ≤0.01 (**)). (a, b) White bars denote TβRIIlox/lox/FN1-Cre mice, black bars littermate controls.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 (PDF 5335 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hauri-Hohl, M., Zuklys, S., Holländer, G. et al. A regulatory role for TGF-β signaling in the establishment and function of the thymic medulla. Nat Immunol 15, 554–561 (2014). https://doi.org/10.1038/ni.2869

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.2869

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