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:

Treg cells require the phosphatase PTEN to restrain TH1 and TFH cell responses

Abstract

The interplay between effector T cells and regulatory T cells (Treg cells) is crucial for adaptive immunity, but how Treg cells control diverse effector responses is elusive. We found that the phosphatase PTEN links Treg cell stability to repression of type 1 helper T cell (TH1 cell) and follicular helper T cell (TFH cell) responses. Depletion of PTEN in Treg cells resulted in excessive TFH cell and germinal center responses and spontaneous inflammatory disease. These defects were considerably blocked by deletion of interferon-γ, indicating coordinated control of TH1 and TFH responses. Mechanistically, PTEN maintained Treg cell stability and metabolic balance between glycolysis and mitochondrial fitness. Moreover, PTEN deficiency upregulates activity of the metabolic checkpoint kinase complex mTORC2 and the serine-threonine kinase Akt, and loss of this activity restores functioning of PTEN-deficient Treg cells. Our studies establish a PTEN-mTORC2 axis that maintains Treg cell stability and coordinates Treg cell–mediated control of effector responses.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Figure 1: Ptenfl/flFoxp3-Cre mice develop age-related autoimmune and lymphoproliferative disease.
Figure 2: Increased T cell activation and altered immune homeostasis in Ptenfl/flFoxp3-Cre mice.
Figure 3: Aberrant induction of TFH cell and GC responses in Ptenfl/flFoxp3-Cre mice.
Figure 4: Analysis of bone marrow–derived chimeras and Ptenfl/flFoxp3-Cre Ifng−/− mice.
Figure 5: PTEN deficiency impairs Treg stability.
Figure 6: PTEN-dependent gene expression and metabolic programs in Treg cells.
Figure 7: Dysregulation of mTORC2 activity in PTEN-deficient Treg cells is responsible for immune tolerance breakdown.
Figure 8: Heterozygous loss of Pten in Treg cells is sufficient to disrupt immune homeostasis.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Josefowicz, S.Z., Lu, L.F. & Rudensky, A.Y. Regulatory T cells: mechanisms of differentiation and function. Annu. Rev. Immunol. 30, 531–564 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Sakaguchi, S., Vignali, D.A., Rudensky, A.Y., Niec, R.E. & Waldmann, H. The plasticity and stability of regulatory T cells. Nat. Rev. Immunol. 13, 461–467 (2013).

    CAS  PubMed  Google Scholar 

  3. Koch, M.A. et al. The transcription factor T-bet controls regulatory T cell homeostasis and function during type 1 inflammation. Nat. Immunol. 10, 595–602 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Zheng, Y. et al. Regulatory T-cell suppressor program co-opts transcription factor IRF4 to control TH2 responses. Nature 458, 351–356 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Chaudhry, A. et al. CD4+ regulatory t cells control TH17 responses in a Stat3-dependent manner. Science 326, 986–991 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  6. Rubtsov, Y.P. et al. Stability of the regulatory T cell lineage in vivo. Science 329, 1667–1671 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Ouyang, W. et al. Novel Foxo1-dependent transcriptional programs control Treg cell function. Nature 491, 554–559 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Zhou, X. et al. Instability of the transcription factor Foxp3 leads to the generation of pathogenic memory T cells in vivo. Nat. Immunol. 10, 1000–1007 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Tsuji, M. et al. Preferential generation of follicular B helper T cells from Foxp3+ T cells in gut Peyer's patches. Science 323, 1488–1492 (2009).

    CAS  PubMed  Google Scholar 

  10. Bailey-Bucktrout, S.L. et al. Self-antigen-driven activation induces instability of regulatory T cells during an inflammatory autoimmune response. Immunity 39, 949–962 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Komatsu, N. et al. Pathogenic conversion of Foxp3+ T cells into TH17 cells in autoimmune arthritis. Nat. Med. 20, 62–68 (2014).

    CAS  PubMed  Google Scholar 

  12. Komatsu, N. et al. Heterogeneity of natural Foxp3+ T cells: a committed regulatory T-cell lineage and an uncommitted minor population retaining plasticity. Proc. Natl. Acad. Sci. USA 106, 1903–1908 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Miyao, T. et al. Plasticity of Foxp3+ T cells reflects promiscuous Foxp3 expression in conventional T cells but not reprogramming of regulatory T cells. Immunity 36, 262–275 (2012).

    CAS  PubMed  Google Scholar 

  14. Smigiel, K.S. et al. CCR7 provides localized access to IL-2 and defines homeostatically distinct regulatory T cell subsets. J. Exp. Med. 211, 121–136 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Crotty, S. Follicular helper CD4 T cells (TFH). Annu. Rev. Immunol. 29, 621–663 (2011).

    CAS  PubMed  Google Scholar 

  16. Linterman, M.A. et al. Follicular helper T cells are required for systemic autoimmunity. J. Exp. Med. 206, 561–576 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Liu, X. et al. Transcription factor achaete-scute homologue 2 initiates follicular T-helper-cell development. Nature 507, 513–518 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Chung, Y. et al. Follicular regulatory T cells expressing Foxp3 and Bcl-6 suppress germinal center reactions. Nat. Med. 17, 983–988 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Linterman, M.A. et al. Foxp3+ follicular regulatory T cells control the germinal center response. Nat. Med. 17, 975–982 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Chi, H. Regulation and function of mTOR signalling in T cell fate decisions. Nat. Rev. Immunol. 12, 325–338 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Powell, J.D., Pollizzi, K.N., Heikamp, E.B. & Horton, M.R. Regulation of immune responses by mTOR. Annu. Rev. Immunol. 30, 39–68 (2012).

    CAS  PubMed  Google Scholar 

  22. Lee, K. et al. Mammalian target of rapamycin protein complex 2 regulates differentiation of Th1 and Th2 cell subsets via distinct signaling pathways. Immunity 32, 743–753 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  23. Delgoffe, G.M. et al. The kinase mTOR regulates the differentiation of helper T cells through the selective activation of signaling by mTORC1 and mTORC2. Nat. Immunol. 12, 295–303 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Yang, K. et al. T cell exit from quiescence and differentiation into Th2 cells depend on Raptor-mTORC1-mediated metabolic reprogramming. Immunity 39, 1043–1056 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Yang, K., Neale, G., Green, D.R., He, W. & Chi, H. The tumor suppressor Tsc1 enforces quiescence of naive T cells to promote immune homeostasis and function. Nat. Immunol. 12, 888–897 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Suzuki, A. et al. T cell–specific loss of Pten leads to defects in central and peripheral tolerance. Immunity 14, 523–534 (2001).

    CAS  PubMed  Google Scholar 

  28. Liu, X. et al. Distinct roles for PTEN in prevention of T cell lymphoma and autoimmunity in mice. J. Clin. Invest. 120, 2497–2507 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Song, M.S. et al. Nuclear PTEN regulates the APC-CDH1 tumor-suppressive complex in a phosphatase-independent manner. Cell 144, 187–199 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Bensinger, S.J. et al. Distinct IL-2 receptor signaling pattern in CD4+CD25+ regulatory T cells. J. Immunol. 172, 5287–5296 (2004).

    CAS  PubMed  Google Scholar 

  31. Zanin-Zhorov, A. et al. Scaffold protein Disc large homolog 1 is required for T-cell receptor-induced activation of regulatory T-cell function. Proc. Natl. Acad. Sci. USA 109, 1625–1630 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Delgoffe, G.M. et al. Stability and function of regulatory T cells is maintained by a neuropilin-1-semaphorin-4a axis. Nature 501, 252–256 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Walsh, P.T. et al. PTEN inhibits IL-2 receptor-mediated expansion of CD4+ CD25+ Tregs. J. Clin. Invest. 116, 2521–2531 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Rubtsov, Y.P. et al. Regulatory T cell-derived interleukin-10 limits inflammation at environmental interfaces. Immunity 28, 546–558 (2008).

    CAS  PubMed  Google Scholar 

  35. Lee, S.K. et al. Interferon-γ excess leads to pathogenic accumulation of follicular helper T cells and germinal centers. Immunity 37, 880–892 (2012).

    CAS  PubMed  Google Scholar 

  36. Nakayamada, S. et al. Early Th1 cell differentiation is marked by a Tfh cell-like transition. Immunity 35, 919–931 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Ray, J.P. et al. Transcription factor STAT3 and type I interferons are corepressive insulators for differentiation of follicular helper and T helper 1 cells. Immunity 40, 367–377 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Shin, H.M. et al. Epigenetic modifications induced by Blimp-1 Regulate CD8+ T cell memory progression during acute virus infection. Immunity 39, 661–675 (2013).

    CAS  PubMed  Google Scholar 

  39. Wang, H. et al. The transcription factor Foxp1 is a critical negative regulator of the differentiation of follicular helper T cells. Nat. Immunol. 15, 667–675 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl. Acad. Sci. USA 102, 15545–15550 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. 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).

    CAS  PubMed  Google Scholar 

  42. Shi, L.Z. et al. HIF1α-dependent glycolytic pathway orchestrates a metabolic checkpoint for the differentiation of TH17 and Treg cells. J. Exp. Med. 208, 1367–1376 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Huynh, A. et al. Phosphatase PTEN–mediated control of the kinase PI(3)K in Treg cells maintains homeostasis and lineage stability. Nat. Immunol. http://dx.doi.org/10.1038/ni.3077 (5 January 2015).

  44. Gavin, M.A., Clarke, S.R., Negrou, E., Gallegos, A. & Rudensky, A. Homeostasis and anergy of CD4+CD25+ suppressor T cells in vivo. Nat. Immunol. 3, 33–41 (2002).

    CAS  PubMed  Google Scholar 

  45. León, B., Bradley, J.E., Lund, F.E., Randall, T.D. & Ballesteros-Tato, A. FoxP3+ regulatory T cells promote influenza-specific Tfh responses by controlling IL-2 availability. Nat. Commun. 5, 3495 (2014).

    PubMed  Google Scholar 

  46. Soond, D.R. et al. Pten loss in CD4 T cells enhances their helper function but does not lead to autoimmunity or lymphoma. J. Immunol. 188, 5935–5943 (2012).

    CAS  PubMed  Google Scholar 

  47. Kang, S.G. et al. MicroRNAs of the miR-1792 family are critical regulators of TFH differentiation. Nat. Immunol. 14, 849–857 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Garcia-Cao, I. et al. Systemic elevation of PTEN induces a tumor-suppressive metabolic state. Cell 149, 49–62 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Wei, J., Yang, K. & Chi, H. Cutting edge: discrete functions of mTOR signaling in invariant NKT cell development and NKT17 fate decision. J. Immunol. 193, 4297–4301 (2014).

    CAS  PubMed  Google Scholar 

  50. Liu, G. et al. The receptor S1P1 overrides regulatory T cell-mediated immune suppression through Akt-mTOR. Nat. Immunol. 10, 769–777 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Liu, G., Yang, K., Burns, S., Shrestha, S. & Chi, H. The S1P(1)-mTOR axis directs the reciprocal differentiation of TH1 and Treg cells. Nat. Immunol. 11, 1047–1056 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank J. Wei, D. Bastardo Blanco and S. Brown for help with immunological assays; C. Cloer and B. Rhode for animal colony management; A. Rudensky for Foxp3YFP-Cre mice and the St. Jude Immunology FACS core facility for cell sorting. Supported by US National Institutes of Health (AI105887, AI101407, CA176624 and NS064599 to H.C.), the American Cancer Society (to H.C.) and the Crohn's and Colitis Foundation of America (to H.C.), and the Arthritis Foundation (to K.Y.).

Author information

Authors and Affiliations

Authors

Contributions

S.S. and K.Y. designed and performed cellular, molecular and biochemical experiments and contributed to writing the manuscript; C.G. did imaging assays; P.V. did histopathology analysis; G.N. did bioinformatic analyses; and H.C. designed experiments, wrote the manuscript and provided overall direction.

Corresponding author

Correspondence to Hongbo Chi.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Deletion of PTEN and dysregulation of immunoglobulins in Ptenfl/flFoxp3-Cre mice.

(a) Analysis of Pten mRNA expression in Treg and naive CD4+ T cells (left) and PTEN protein expression in CD4+YFP+, CD4+YFP and CD8+ T cells from the spleen of WT and Ptenfl/flFoxp3-Cre mice (right). (b-d) Quantification of IgG subclasses (b), IgM (c) and IgA (d) in the serum of WT and Ptenfl/flFoxp3-Cre mice (the IgG2a binding antibody shows cross-reactivity with IgG2c). Data are representative of at least two independent experiments (a) and one experiment (b-d; n = 22 WT and 24 Ptenfl/flFoxp3-Cre mice). NS, not significant; *P < 0.05 and **P < 0.01. Data are mean ± s.e.m.

Supplementary Figure 2 Analysis of immune cells in Ptenfl/flFoxp3-Cre mice.

(a-c) Analysis and quantification of CD4+, CD8+ (a), B cells (b), conventional dendritic cells (MCHII+CD11c+) and neutrophils (Ly6G+CD11b+) (c) in the spleen of WT and Ptenfl/flFoxp3-Cre mice. (d) Expression of CXCR3 on CD4+ T cells from the spleen of WT and Ptenfl/flFoxp3-Cre mice; numbers above graphs indicate MFI. (e) Expression of IL-4 and IL-17 in CD4+ and CD8+ T cells from WT and Ptenfl/flFoxp3-Cre mice after in vitro stimulation for 4 h. (f) Flow cytometry analysis of Treg cells in CD4+ T cells from peripheral lymph nodes (PLNs) and mesenteric lymph nodes (MLNs) of WT and Ptenfl/flFoxp3-Cre mice. Data are representative of three or more independent experiments (a,b,d-f) and two independent experiments (c). NS, not significant. Data are mean ± s.e.m. Numbers indicate percentage of cells in quadrants or gates.

Supplementary Figure 3 Analysis of TFH and GC B cells.

(a) Flow cytometry of CXCR5 and PD-1 expression (gated on CD4+TCRβ+ cells) in the MLNs of WT and Ptenfl/flFoxp3-Cre mice. (b,c) Flow cytometry of CXCR5 and ICOS (b), and CXCR5 and Bcl6 (c) expression (gated on CD4+TCRβ+ cells) in the spleen and MLNs of WT and Ptenfl/flFoxp3-Cre mice. (d) Flow cytometry of GL7 and CD95 expression (gated on CD19+ cells) in the MLNs of WT and Ptenfl/flFoxp3-Cre mice. (e,f) Analysis of TFH (e) and GC B cells (f) in the spleen of mice immunized with NP-OVA 7 d previously. Data are representative of three independent experiments (a-d) and one experiment (e,f; n = 4 WT, 4 Ptenfl/flFoxp3-Cre mice). *P < 0.05. Data are mean ± s.e.m. Numbers indicate percentage of cells in gates.

Supplementary Figure 4 Regulation of Treg cells in mixed chimeras and Ptenfl/flFoxp3-Cre Ifng−/− mice.

(a) Sublethally irradiated Rag1−/− mice were reconstituted with a 1:1 mix of CD45.1+ BM and either CD45.2+ WT or Ptenfl/flFoxp3-Cre BM cells. Following reconstitution, the mixed chimeras were analyzed for intracellular staining of IFN-γ in CD8+ T cells. (b) Cytokine production of IFN-γ, IL-17 and IL-4 by CD4+ T cells from WT, Ptenfl/flFoxp3-Cre, Ifng−/− and Ptenfl/flFoxp3-Cre Ifng−/− mice after in vitro stimulation for 4 h. (c) PNA staining of spleen sections (magnification, x2; scale bars, 2 mm). (d) Immunofluorescence of MLN sections for the staining of CD3 (red), PNA (green) and IgD (white) (scale 500 μm). Right, quantification of germinal center area. Data are representative of three independent experiments (a,b) and one experiment (c,d; n = 3 WT, 6 Ptenfl/flFoxp3-Cre, 2 Ifng−/− and 3 Ptenfl/flFoxp3-Cre Ifng−/− mice). Data are mean ± s.e.m. Numbers indicate percentage of cells in quadrants or gates.

Supplementary Figure 5 Analysis of Treg cells in Ptenfl/flFoxp3-Cre Ifng−/− mice and Ptenfl/flFoxp3-Cre Treg cell phenotypes under steady state and after adoptive transfer.

(a) Flow cytometry analysis of Treg cells in WT, Ptenfl/flFoxp3-Cre, Ifng−/− and Ptenfl/flFoxp3-Cre Ifng−/− mice. (b) Expression of CXCR3, CXCR5, T-bet, Bcl6, IRF4 and p-STAT3 in Treg cells from the spleen of WT and Ptenfl/flFoxp3-Cre mice; numbers above graphs indicate MFI. (c) Analysis of CXCR3 and T-bet expression in Treg cells in the spleen of WT, Ptenfl/flFoxp3-Cre, Ifng−/− and Ptenfl/flFoxp3-Cre Ifng−/− mice. (d) Analysis of Foxp3 (left) and CD25 (right) expression in PLNs and MLNs of CD45.1+ recipients after transferring WT and Ptenfl/flFoxp3-Cre Treg (CD45.2+) cells; numbers above graphs indicate MFI. Data are representative of at two independent experiments (a-d). Numbers indicate percentage of cells in gates.

Supplementary Figure 6 The transcriptional profiles controlled by PTEN in Treg cells.

(a) PCA mapping of WT and Ptenfl/flFoxp3-Cre Treg cells. (b) The list of top 10 gene sets upregulated in Ptenfl/flFoxp3-Cre Treg cells by GSEA. NES, normalized enrichment score.

Supplementary Figure 7 PTEN represses mTORC2 signaling in Treg cells to maintain immune homeostasis.

(a,b) PTEN expression in resting and short-term stimulated Treg cells (a) and phosphorylation of 4E-BP1 and PTEN in resting and long-term stimulated Treg cells (b) isolated from WT and Ptenfl/flFoxp3-Cre mice. (c) Expression of CD69, CD62L, CXCR5, ICOS and PD-1, and measurements of ROS, mitochondrial mass, and mitochondrial membrane potential (TMRM) in Treg cells from the spleen of WT, Ptenfl/flFoxp3-Cre, Rictorfl/flFoxp3-Cre and Ptenfl/flRictorfl/flFoxp3-Cre mice; numbers above graphs indicate MFI. (d,e) Flow cytometry analysis of WT, Ptenfl/flFoxp3-Cre, Rictorfl/flFoxp3-Cre and Ptenfl/flRictorfl/flFoxp3-Cre mice for CD62L and CD44 expression on CD4+ and CD8+ T cells (d), cytokine production of CD4+ T cells (e). (f) PNA staining of spleen sections (magnification, x2; scale bars, 2 mm). (g) Immunofluorescence of MLN sections for the staining of CD3 (red), PNA (green) and IgD (white) (scale 500 μm) in WT, Ptenfl/flFoxp3-Cre, Rictorfl/flFoxp3-Cre and Ptenfl/flRictorfl/flFoxp3-Cre mice (quantitative results of WT and Ptenfl/flFoxp3-Cre mice in Supplementary Fig. 4d are shown here for comparison). Data are representative of two independent experiments (a,b), three independent experiments (c-e) and one experiment (f,g; n = 2 Rictorfl/flFoxp3-Cre and 3 Ptenfl/flRictorfl/flFoxp3-Cre mice). Data are mean ± s.e.m. Numbers indicate percentage of cells in quadrants.

Supplementary Figure 8 PTEN expression and immune dysregulation in Ptenfl/+Foxp3-Cre mice and the model describing PTEN signaling in Treg cells.

(a,b) Analysis of Pten mRNA (a) and protein (b) expression in Treg, naive CD4+ (CD62L+CD44), and CD8+ T cells from WT and Ptenfl/+Foxp3-Cre mice. Numbers below the PTEN lanes indicate band intensity relative to that of β-actin (b). (c) Immunofluorescence of spleen sections of WT and Ptenfl/+Foxp3-Cre mice for the staining of CD3 (red) and PNA (green) (scale bars, 60 μm). Right, quantification of germinal center area. (d) Images of peripheral lymph nodes from WT and Ptenfl/+Foxp3-Cre mice (~5 months old) with lymphoproliferative disease. Data are representative of two independent experiments (a-d). *P < 0.001. Data are mean ± s.e.m. (e) Schematics of PTEN signaling in Treg cell functions and immune tolerance. Loss of PTEN in Treg cells dysregulates mTORC2 signaling and metabolic and transcriptional programs, leading to the disrupted stability of Treg cells (as evidenced by the downregulation of CD25 expression, as well as other abnormalities not depicted here). Associated with loss of Treg cell stability and the ensuing TH1 responses and IFN-γ production is the aberrant induction of TFH responses, spontaneous formation of germinal center responses, and development of autoimmune and lymphoproliferative disease.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 (PDF 4079 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Shrestha, S., Yang, K., Guy, C. et al. Treg cells require the phosphatase PTEN to restrain TH1 and TFH cell responses. Nat Immunol 16, 178–187 (2015). https://doi.org/10.1038/ni.3076

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3076

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