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:

Foxp3 and Toll-like receptor signaling balance Treg cell anabolic metabolism for suppression

Abstract

CD4+ effector T cells (Teff cells) and regulatory T cells (Treg cells) undergo metabolic reprogramming to support proliferation and immunological function. Although signaling via the lipid kinase PI(3)K (phosphatidylinositol-3-OH kinase), the serine-threonine kinase Akt and the metabolic checkpoint kinase complex mTORC1 induces both expression of the glucose transporter Glut1 and aerobic glycolysis for Teff cell proliferation and inflammatory function, the mechanisms that regulate Treg cell metabolism and function remain unclear. We found that Toll-like receptor (TLR) signals that promote Treg cell proliferation increased PI(3)K-Akt-mTORC1 signaling, glycolysis and expression of Glut1. However, TLR-induced mTORC1 signaling also impaired Treg cell suppressive capacity. Conversely, the transcription factor Foxp3 opposed PI(3)K-Akt-mTORC1 signaling to diminish glycolysis and anabolic metabolism while increasing oxidative and catabolic metabolism. Notably, Glut1 expression was sufficient to increase the number of Treg cells, but it reduced their suppressive capacity and Foxp3 expression. Thus, inflammatory signals and Foxp3 balance mTORC1 signaling and glucose metabolism to control the proliferation and suppressive function of Treg 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: Treg cell proliferation is associated with increased glycolysis and anabolic signaling.
Figure 2: Signaling via TLR1 and TLR2 drives Treg cell glycolysis and proliferation, yet reduces suppressive capacity.
Figure 3: Foxp3 reduces aerobic glycolysis and promotes mitochondrial oxidative metabolism.
Figure 4: Foxp3 expression is sufficient to suppress glycolysis and promote oxidative metabolism.
Figure 5: Glut1 expression increases the number and size of Treg cells but reduces their suppressive capacity.
Figure 6: Increased glucose uptake impairs the suppressive ability of Treg cells.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Buck, M.D., O'Sullivan, D. & Pearce, E.L. T cell metabolism drives immunity. J. Exp. Med. 212, 1345–1360 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. DeBerardinis, R.J. & Chandel, N.S. Fundamentals of cancer metabolism. Sci. Adv. 2, e1600200 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Chang, C.H. et al. Post-transcriptional control of T cell effector function by aerobic glycolysis. Cell 153, 1239–1251 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Macintyre, A.N. et al. The glucose transporter Glut1 is selectively essential for CD4 T cell activation and effector function. Cell Metab. 20, 61–72 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Beier, U.H. et al. Essential role of mitochondrial energy metabolism in Foxp3+ T-regulatory cell function and allograft survival. FASEB J. 29, 2315–2326 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Gerriets, V.A. et al. Metabolic programming and PDHK1 control CD4+ T cell subsets and inflammation. J. Clin. Invest. 125, 194–207 (2015).

    Article  PubMed  Google Scholar 

  7. Michalek, R.D. et al. Cutting edge: distinct glycolytic and lipid oxidative metabolic programs are essential for effector and regulatory CD4+ T cell subsets. J. Immunol. 186, 3299–3303 (2011).

    Article  CAS  PubMed  Google Scholar 

  8. Shi, L.Z. et al. HIF1alpha-dependent glycolytic pathway orchestrates a metabolic checkpoint for the differentiation of TH17 and Treg cells. J. Exp. Med. 208, 1367–1376 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Smigiel, K.S., Srivastava, S., Stolley, J.M. & Campbell, D.J. Regulatory T-cell homeostasis: steady-state maintenance and modulation during inflammation. Immunol. Rev. 259, 40–59 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Zeng, H. & Chi, H. Metabolic control of regulatory T cell development and function. Trends Immunol. 36, 3–12 (2015).

    Article  CAS  PubMed  Google Scholar 

  11. Laidlaw, B.J. et al. Production of IL-10 by CD4+ regulatory T cells during the resolution of infection promotes the maturation of memory CD8(+) T cells. Nat. Immunol. 16, 871–879 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Nosbaum, A. et al. Cutting edge: regulatory T cells facilitate cutaneous wound healing. J. Immunol. 196, 2010–2014 (2016).

    Article  CAS  PubMed  Google Scholar 

  13. Arpaia, N. et al. A distinct function of regulatory T cells in tissue protection. Cell 162, 1078–1089 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Dang, E.V. et al. Control of T(H)17/T(reg) balance by hypoxia-inducible factor 1. Cell 146, 772–784 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Oestreich, K.J. et al. Bcl-6 directly represses the gene program of the glycolysis pathway. Nat. Immunol. 15, 957–964 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Delgoffe, G.M. et al. The mTOR kinase differentially regulates effector and regulatory T cell lineage commitment. Immunity 30, 832–844 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Procaccini, C. et al. An oscillatory switch in mTOR kinase activity sets regulatory T cell responsiveness. Immunity 33, 929–941 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Zeng, H. et al. mTORC1 couples immune signals and metabolic programming to establish T(reg)-cell function. Nature 499, 485–490 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Huynh, A. et al. Control of PI(3) kinase in Treg cells maintains homeostasis and lineage stability. Nat. Immunol. 16, 188–196 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Shrestha, S. et al. Treg cells require the phosphatase PTEN to restrain TH1 and TFH cell responses. Nat. Immunol. 16, 178–187 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Park, Y. et al. TSC1 regulates the balance between effector and regulatory T cells. J. Clin. Invest. 123, 5165–5178 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Heng, T.S. & Painter, M.W. The Immunological Genome Project: networks of gene expression in immune cells. Nat. Immunol. 9, 1091–1094 (2008).

    Article  CAS  PubMed  Google Scholar 

  23. Peng, G. et al. Toll-like receptor 8-mediated reversal of CD4+ regulatory T cell function. Science 309, 1380–1384 (2005).

    Article  CAS  PubMed  Google Scholar 

  24. Sutmuller, R.P. et al. Toll-like receptor 2 controls expansion and function of regulatory T cells. J. Clin. Invest. 116, 485–494 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Voo, K.S. et al. Targeting of TLRs inhibits CD4+ regulatory T cell function and activates lymphocytes in human peripheral blood mononuclear cells. J. Immunol. 193, 627–634 (2014).

    Article  CAS  PubMed  Google Scholar 

  26. Nyirenda, M.H. et al. TLR2 stimulation drives human naive and effector regulatory T cells into a Th17-like phenotype with reduced suppressive function. J. Immunol. 187, 2278–2290 (2011).

    Article  CAS  PubMed  Google Scholar 

  27. Sujino, T. et al. Tissue adaptation of regulatory and intraepithelial CD4+ T cells controls gut inflammation. Science 352, 1581–1586 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Wang, S. et al. MyD88 adaptor-dependent microbial sensing by regulatory T cells promotes mucosal tolerance and enforces commensalism. Immunity 43, 289–303 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Scharschmidt, T.C. et al. A wave of regulatory T cells into neonatal skin mediates tolerance to commensal microbes. Immunity 43, 1011–1021 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Luo, L. et al. Rab8a interacts directly with PI(3)Kγ to modulate TLR4-driven PI(3)K and mTOR signalling. Nat. Commun. 5, 4407 (2014).

    Article  CAS  PubMed  Google Scholar 

  31. Caro-Maldonado, A. et al. Metabolic reprogramming is required for antibody production that is suppressed in anergic but exaggerated in chronically BAFF-exposed B cells. J. Immunol. 192, 3626–3636 (2014).

    CAS  PubMed  Google Scholar 

  32. Everts, B. et al. TLR-driven early glycolytic reprogramming via the kinases TBK1-IKKɛ supports the anabolic demands of dendritic cell activation. Nat. Immunol. 15, 323–332 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Jacobs, S.R. et al. Glucose uptake is limiting in T cell activation and requires CD28-mediated Akt-dependent and independent pathways. J. Immunol. 180, 4476–4486 (2008).

    Article  CAS  PubMed  Google Scholar 

  34. Fisson, S. et al. Continuous activation of autoreactive CD4+ CD25+ regulatory T cells in the steady state. J. Exp. Med. 198, 737–746 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Williams, L.M. & Rudensky, A.Y. Maintenance of the Foxp3-dependent developmental program in mature regulatory T cells requires continued expression of Foxp3. Nat. Immunol. 8, 277–284 (2007).

    Article  CAS  PubMed  Google Scholar 

  36. Basu, S., Hubbard, B. & Shevach, E.M. Foxp3-mediated inhibition of Akt inhibits Glut1 (glucose transporter 1) expression in human T regulatory cells. J. Leukoc. Biol. 97, 279–283 (2015).

    Article  CAS  PubMed  Google Scholar 

  37. Arvey, A. et al. Inflammation-induced repression of chromatin bound by the transcription factor Foxp3 in regulatory T cells. Nat. Immunol. 15, 580–587 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Wilhelm, K. et al. FOXO1 couples metabolic activity and growth state in the vascular endothelium. Nature 529, 216–220 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Panduro, M., Benoist, C. & Mathis, D. Tissue Tregs. Annu. Rev. Immunol. 34, 609–633 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Cretney, E., Kallies, A. & Nutt, S.L. Differentiation and function of Foxp3+ effector regulatory T cells. Trends Immunol. 34, 74–80 (2013).

    Article  CAS  PubMed  Google Scholar 

  41. Ostroukhova, M. et al. The role of low-level lactate production in airway inflammation in asthma. Am. J. Physiol. Lung Cell. Mol. Physiol. 302, L300–L307 (2012).

    Article  CAS  PubMed  Google Scholar 

  42. Leifer, C.A. & Medvedev, A.E. Molecular mechanisms of regulation of Toll-like receptor signaling. J. Leukoc. Biol. http://dx.doi.org/10.1189/jlb.2MR0316-117RR (published online 24 June 2016).

  43. Xie, X. et al. IkappaB kinase epsilon and TANK-binding kinase 1 activate AKT by direct phosphorylation. Proc. Natl. Acad. Sci. USA 108, 6474–6479 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  44. Ohl, K. & Tenbrock, K. Regulatory T cells in systemic lupus erythematosus. Eur. J. Immunol. 45, 344–355 (2015).

    Article  CAS  PubMed  Google Scholar 

  45. Byng-Maddick, R. & Ehrenstein, M.R. The impact of biological therapy on regulatory T cells in rheumatoid arthritis. Rheumatology (Oxford) 54, 768–775 (2015).

    Article  CAS  Google Scholar 

  46. Rathmell, J.C., Elstrom, R.L., Cinalli, R.M. & Thompson, C.B. Activated Akt promotes increased resting T cell size, CD28-independent T cell growth, and development of autoimmunity and lymphoma. Eur. J. Immunol. 33, 2223–2232 (2003).

    Article  CAS  PubMed  Google Scholar 

  47. Allan, S.E., Song-Zhao, G.X., Abraham, T., McMurchy, A.N. & Levings, M.K. Inducible reprogramming of human T cells into Treg cells by a conditionally active form of FOXP3. Eur. J. Immunol. 38, 3282–3289 (2008).

    Article  CAS  PubMed  Google Scholar 

  48. Huang, W., Sherman, B.T. & Lempicki, R.A. Systematic and integrative analysis of large gene lists using DAVID bioinformatics resources. Nat. Protoc. 4, 44–57 (2009).

    Article  CAS  Google Scholar 

  49. Thomas, P.D. et al. PANTHER: a library of protein families and subfamilies indexed by function. Genome Res. 13, 2129–2141 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Chen, C., Rowell, E.A., Thomas, R.M., Hancock, W.W. & Wells, A.D. Transcriptional regulation by Foxp3 is associated with direct promoter occupancy and modulation of histone acetylation. J. Biol. Chem. 281, 36828–36834 (2006).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank members of the Rathmell and Wells laboratories for discussions, and the Immunological Genome Project. Supported by the Crohn's and Colitis Foundation of America (Senior Research Grant to J.C.R.), the Alliance for Lupus Research (J.C.R.), the US National Institutes of Health (R01HL 108006 and R01105550DK to J.C.R.; P01HL018646 to L.A.T. and J.C.R.; F31CA183529 to R.J.K.; R00CA168997 to J.W.L.; and R01AI070807 and P01AI073489 to A.D.W.), and the German Research Foundation (Deutsche Forschungsgemeinschaft; P.J.S.).

Author information

Authors and Affiliations

Authors

Contributions

V.A.G., R.J.K., A.D.W. and J.C.R. designed the study, interpreted data and wrote the manuscript. V.A.G. and R.J.K. performed most of the experiments. M.O.J., S.C. and P.J.S. performed experiments to analyze Foxp3 regulation of metabolism. A.G.N. assisted V.A.G. and R.J.K. and maintained animals that were essential for the study. M.O.W. performed mass spectrometry. A.A.d.C. analyzed RNAseq data. J.W.L. assisted in metabolomics analysis. N.J.M. and L.A.T. assisted with data analysis and interpretation. A.D.W. performed microarray analysis.

Corresponding author

Correspondence to Jeffrey C Rathmell.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Treg cell metabolism is regulated by Foxp3 and inflammatory signals.

(a) CD4+Foxp3+ T cells and CD4+Foxp3- T cells were analyzed by flow cytometry for Ki67 expression levels. (b-c) CD4+CD25- T cells were isolated from the spleens of WT mice, polarized under Treg skewing conditions for 5 days and treated with vehicle (H2O) or 5 μg/mL Pam3CSK4 for the final 24 hrs. Cells were re-isolated by magnetic separation and analyzed for (b) forward scatter (FSC) and (c) glycolytic capacity using the Seahorse Extracellular Flux Analyzer. (d) Gene ontology analysis using PANTHER of pathways altered by Foxp3 deletion using gene expression data published by Williams and Rudensky (Nat. Immunol 8:277). (e-f) Primary murine CD4+CD25- T cells were activated and transduced with control or Foxp3 expressing retrovirus and (e) analyzed by chromatin immunoprecipitation-sequencing showing Foxp3 associated sites in the pyruvate dehydrogenase kinase 3 (PDK3) and PIK3cg loci or (f) analyzed by QPCR for expression of PDK3 mRNA. Data are representative of biological triplicate experiments (a-b, e-f), two independent experiments (c), or an analysis of previously published datasets with biological duplicates (d). Means and standard deviations are shown, * p<0.05.

Supplementary Figure 2 Foxp3 expression in non–T cell lineage inhibits anabolic growth signaling and gene expression.

a-e. Three individual clones of control and Foxp3-ER expressing FL5.12 cells were treated with 4OHT to activate Foxp3 and examined for (a-b) the expression of metabolic and related proteins, (c) select glycolytic gene expression by QPCR or (d-e) were extracted and analyzed using high-resolution LC-QE-MS. (d) A heat map with relative levels of metabolites using unsupervised hierarchical clustering or (e) select metabolite levels are shown. Data are representative of three independent experiments (a-b) or an analysis of three independent clones (c-e). (a-b) Gel bands are quantified, * p<0.05, **p<0.005.

Supplementary Figure 3 Constitutive Akt expression increases the number and frequency of Treg cells but diminishes suppressive function.

a-b. CD4+CD25- T cells were isolated from the spleens of control and mAkt-Tg mice and polarized under Treg skewing conditions. Cells were examined for (a) glucose uptake and (b) ROS levels as measured by DCFDA. c-i. Foxp3+ tTreg from the spleen of control and mAkt-Tg mice were examined for (c) tTreg number, (d) percentage and (e) cell size determined by forward scatter and were measured by flow cytometry. (f) CD25, (g) ICOS, (h) CD69 and (i) CD62L protein expression in CD4+Foxp3+ control and mAkt-Tg cells were measured by flow cytometry. (j) CD4+CD25- T cells were isolated from the spleens of control and mAkt-Tg mice and were polarized under Treg skewing conditions to measure inhibition of effector T cell (Teff) proliferation in an in vitro suppression assay. Data are representative of two independent experiments (a, b), three independent experiments (c, d, f-i), four independent experiments (e), or two experiments (f, j). Means and standard deviations are shown, * p<0.05.

Supplementary Figure 4 Transgenic expression of Glut1 results in altered metabolic and immune phenotypes in Treg cells.

a-d. CD4+CD25- T cells were isolated from the spleens of control and Glut1-Tg mice and polarized under Treg skewing conditions. Cells were examined for (a) Glut1 expression levels by immunoblot, (b) glucose uptake, (c) ROS production as measured by DCFDA and (d) ECAR and OCR levels were measured using the Seahorse Extracellular Flux Analyzer before and after the addition of the specified inhibitors. e-f. CD4+Foxp3+ T cells from the spleens of control and Glut1-Tg mice were examined for expression of (e) CD25 and (f) Helios proteins. Data are representative of three independent experiments (a-b, d, e) or compiled data from three independent experiments (c, f). Means and standard deviations are shown, * p<0.05.

Supplementary Figure 5 Transgenic Glut1 expression alters the immunological phenotype of Treg cells.

a-f. CD4+CD25- T cells were isolated from the spleens of control and Glut1-Tg mice and polarized under Treg skewing conditions. Skewed cells were analyzed for (a) Foxp3 expression by flow cytometry, (b) cell size by FSC analysis, (c) CD25 and (d) ICOS protein expression by flow cytometry. (e) RNA expression levels of a panel of immunosuppressive related genes by QPCR and (f) percentage of IFNγ and IL-2 producing CD4+Foxp3+ cells by flow cytometry are shown. Data are representative of three independent experiments (a-d) or the average of six biological replicates (e) or four biological replicates (f). Means and standard deviations are shown, * p<0.05.

Supplementary Figure 6 Transgenic expression of Glut1 diminishes the suppressive ability of Treg cells in vitro and in vivo.

a. CD4+CD25- T cells were isolated from the spleens of control and Glut1-Tg mice and polarized under Treg skewing conditions. Control and Glut1-Tg Treg were functionally examined in an in vitro suppression assay to measure inhibition of effector T cells (Teff) proliferation and the Teff division index was calculated by Flowjo flow cytometry analysis software. b-d. RAG1-/- mice were injected with naïve effector (CD4+CD25-CD45RBhi) T cells to induce colitis. After weight loss indicated active disease was apparent, control or Glut1-Tg CD4+CD25+CD45RBlo Treg were sorted and analyzed by flow cytometry to assess sorted Treg. (b) The expression of CD25 and CD45Rb and (c) Foxp3 protein of sorted rescue Treg are shown. (d) At the termination of the experiment Foxp3 levels were assessed on CD4+ gated T cells in the spleens of recipient animals. (e) Thy1.1 naïve effector (CD4+CD25-CD45RBhi) T cells were adoptively transferred into RAG1-/- mice to initiate IBD. Thy1.2 control or Glut1-tg tTreg (CD4+CD25+CD45RBlo) T cells were sorted and injected after disease was apparent Foxp3 levels were then assessed by flow cytometry on adoptively transferred Thy1.1 effectors and Thy1.2 CD4 control and Glut1-tg Treg from mesenteric lymph nodes and spleens. Data are the result of three independent experiments (a), representative of three independent experiments (b) or is representative of two independent experiments with at least 5 mice per group (c, d). Means and standard deviations are shown, * p<0.05. (f) Model of our findings. Our findings show that Treg are metabolically heterogeneous and depend on activating and inflammatory signals as well as Foxp3 itself to coordinate metabolism. In the presence of inflammatory stimuli, such as TLR ligands, Treg increase mTORC1 signaling, Glut1, and glycolysis, which results in increased cell growth and proliferation. Suppressive capacity, however, can be impaired. As inflammatory signals are reduced, Foxp3 can tilt the balance away from mTORC1 signaling to favor oxidative metabolism that lowers proliferative ability but enhances suppression to promote inflammatory resolution. Metabolic transitions are critical in this process as increased Glut1 expression is sufficient to promote Treg growth while reducing suppression and stability.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–6 and Supplementary Tables 1 –6 (PDF 3206 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Gerriets, V., Kishton, R., Johnson, M. et al. Foxp3 and Toll-like receptor signaling balance Treg cell anabolic metabolism for suppression. Nat Immunol 17, 1459–1466 (2016). https://doi.org/10.1038/ni.3577

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3577

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