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:

Single-cell gene expression reveals a landscape of regulatory T cell phenotypes shaped by the TCR

A Publisher Correction to this article was published on 03 May 2018

This article has been updated

Abstract

CD4+ T regulatory cells (Treg) are central to immune homeostasis, their phenotypic heterogeneity reflecting the diverse environments and target cells that they regulate. To understand this heterogeneity, we combined single-cell RNA-seq, activation reporter and T cell receptor (TCR) analysis to profile thousands of Treg or conventional CD4+FoxP3 T cells (Tconv) from mouse lymphoid organs and human blood. Treg and Tconv pools showed areas of overlap, as resting ‘furtive’ Tregs with overall similarity to Tconvs or as a convergence of activated states. All Tregs expressed a small core of FoxP3-dependent transcripts, onto which additional programs were added less uniformly. Among suppressive functions, Il2ra and Ctla4 were quasiconstant, inhibitory cytokines being more sparsely distributed. TCR signal intensity did not affect resting/activated Treg proportions but molded activated Treg programs. The main lines of Treg heterogeneity in mice were strikingly conserved in human blood. These results reveal unexpected TCR-shaped states of activation, providing a framework to synthesize previous observations of Treg heterogeneity.

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

Fig. 1: Treg and Tconv scRNA-seq datasets.
Fig. 2: Some Tregs and Tconvs show closely related transcriptomes.
Fig. 3: Treg-signature expression in single Treg and Tconv cells.
Fig. 4: Resting and activated Treg states identified by scRNA-seq.
Fig. 5: Treg states associated with different TCR-signaling strength.
Fig. 6: Tregs that express the same TCRα/β sequence have related transcriptional programs.
Fig. 7: Similitudes in human and mouse Treg heterogeneity.

Similar content being viewed by others

Change history

  • 03 May 2018

    In the version of this article initially published, the Supplementary Note was missing. The Supplementary Note has now been provided online and is cited in the Methods section of the article. The error has been corrected in the HTML and PDF version of the article.

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).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Campbell, D. J. & Koch, M. A. Phenotypical and functional specialization of FOXP3+ regulatory T cells. Nat. Rev. Immunol. 11, 119–130 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Luo, C. T. & Li, M. O. Transcriptional control of regulatory T cell development and function. Trends. Immunol. 34, 531–539 (2013).

    Article  CAS  PubMed  Google Scholar 

  5. Vignali, D. A., Collison, L. W. & Workman, C. J. How regulatory T cells work. Nat. Rev. Immunol. 8, 523–532 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Miyara, M. et al. Functional delineation and differentiation dynamics of human CD4+ T cells expressing the FoxP3 transcription factor. Immunity 30, 899–911 (2009).

  9. Kleinewietfeld, M. et al. CCR6 expression defines regulatory effector/memory-like cells within the CD25+CD4+ T-cell subset. Blood 105, 2877–2886 (2005).

  10. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. 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 

  12. Hill, J. A. et al. Foxp3 transcription-factor-dependent and -independent regulation of the regulatory T cell transcriptional signature. Immunity 27, 786–800 (2007).

  13. Gavin, M. A. et al Single-cell analysis of normal and FOXP3-mutant human T cells: FOXP3 expression without regulatory T cell development. Proc. Natl Acad. Sci. USA 103, 6659–6664 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. 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).

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Zemmour, D., Pratama, A., Loughhead, S. M., Mathis, D. & Benoist, C. Flicr, a long noncoding RNA, modulates Foxp3 expression and autoimmunity. Proc. Natl Acad. Sci. USA 114, E3472–E3480 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Lathrop, S. K. et al. Peripheral education of the immune system by colonic commensal microbiota. Nature 478, 250–254 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Li, M. O. & Rudensky, A. Y. T cell receptor signalling in the control of regulatory T cell differentiation and function. Nat. Rev. Immunol. 16, 220–233 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Levine, A. G., Arvey, A., Jin, W. & Rudensky, A. Y. Continuous requirement for the TCR in regulatory T cell function. Nat. Immunol. 15, 1070–1078 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Hashimshony, T., Wagner, F., Sher, N. & Yanai, I. CEL-Seq: single-cell RNA-Seq by multiplexed linear amplification. Cell Rep. 2, 666–673 (2012).

    Article  CAS  PubMed  Google Scholar 

  21. Klein, A. M. et al. Droplet barcoding for single-cell transcriptomics applied to embryonic stem cells. Cell 161, 1187–1201 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Wagner, A., Regev, A. & Yosef, N. Revealing the vectors of cellular identity with single-cell genomics. Nat. Biotechnol. 34, 1145–1160 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Zilionis, R. et al. Single-cell barcoding and sequencing using droplet microfluidics. Nat. Protoc. 12, 44–73 (2017).

    Article  CAS  PubMed  Google Scholar 

  24. Sawant, D. V. & Vignali, D. A. Once a Treg, always a Treg? Immunol. Rev. 259, 173–191 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. van der Maaten, L. & Hinton, G. Visualizing high-dimensional data using t-SNE. J. Mach. Learn. Res. 9, 2579–2605 (2008).

    Google Scholar 

  26. Kim, H. J. et al. Stable inhibitory activity of regulatory T cells requires the transcription factor Helios. Science 350, 334–339 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Sage, P. T. & Sharpe, A. H. T follicular regulatory cells. Immunol. Rev. 271, 246–259 (2016).

    Article  CAS  PubMed  Google Scholar 

  28. Wakamatsu, E., Mathis, D. & Benoist, C. Convergent and divergent effects of costimulatory molecules in conventional and regulatory CD4+ T cells. Proc. Natl. Acad. Sci. USA 110, 1023–1028 (2013).

    Article  CAS  PubMed  Google Scholar 

  29. Samstein, R. M. et al. Foxp3 exploits a pre-existent enhancer landscape for regulatory T cell lineage specification. Cell 151, 153–166 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Kitagawa, Y. et al. Guidance of regulatory T cell development by Satb1-dependent super-enhancer establishment. Nat. Immunol. 18, 173–183 (2017).

    Article  CAS  PubMed  Google Scholar 

  31. 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).

    Article  CAS  PubMed  Google Scholar 

  32. Meredith, M., Zemmour, D., Mathis, D. & Benoist, C. Aire controls gene expression in the thymic epithelium with ordered stochasticity. Nat. Immunol. 16, 942–949 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Hennig, C. Cluster-wise assessment of cluster stability. Comput. Stat. Data. Anal. 52, 258–271 (2007).

    Article  Google Scholar 

  34. Cretney, E. et al. The transcription factors Blimp-1 and IRF4 jointly control the differentiation and function of effector regulatory T cells. Nat. Immunol. 12, 304–311 (2011).

    Article  CAS  PubMed  Google Scholar 

  35. Feng, Y. et al. A mechanism for expansion of regulatory T-cell repertoire and its role in self-tolerance. Nature 528, 132–136 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Liu, Y. et al. Inhibition of p300 impairs Foxp3+ T regulatory cell function and promotes antitumor immunity. Nat. Med. 19, 1173–1177 (2013).

  37. Roychoudhuri, R. et al. BACH2 represses effector programs to stabilize T(reg)-mediated immune homeostasis. Nature 498, 506–510 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Jaitin, D. A. et al. Massively parallel single-cell RNA-seq for marker-free decomposition of tissues into cell types. Science 343, 776–779 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Shalek, A. K. et al. Single-cell RNA-seq reveals dynamic paracrine control of cellular variation. Nature 510, 363–369 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Moran, A. E. et al. T cell receptor signal strength in Treg and iNKT cell development demonstrated by a novel fluorescent reporter mouse. J. Exp. Med. 208, 1279–1289 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Pesenacker, A. M. et al. A regulatory T-cell gene signature is a specific and sensitive biomarker to identify children with new-onset type 1 diabetes. Diabetes 65, 1031–1039 (2016).

  43. Fontenot, J. D., Rasmussen, J. P., Gavin, M. A. & Rudensky, A. Y. A function for interleukin 2 in Foxp3-expressing regulatory T cells. Nat. Immunol. 6, 1142–1151 (2005).

    Article  CAS  PubMed  Google Scholar 

  44. Mahmud, S. A. et al. Costimulation via the tumor-necrosis factor receptor superfamily couples TCR signal strength to the thymic differentiation of regulatory T cells. Nat. Immunol. 15, 473–481 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Liu, Z. et al. Immune homeostasis enforced by co-localized effector and regulatory T cells. Nature 528, 225–230 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Beyer, M. et al. Repression of the genome organizer SATB1 in regulatory T cells is required for suppressive function and inhibition of effector differentiation. Nat. Immunol. 12, 898–907 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Sefik, E. et al. Individual intestinal symbionts induce a distinct population of RORγ+ regulatory T cells. Science 349, 993–997 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Malissen, B. & Bongrand, P. Early T cell activation: integrating biochemical, structural, and biophysical cues. Annu. Rev. Immunol. 33, 539–561 (2015).

    Article  CAS  PubMed  Google Scholar 

  50. Ashouri, J. F. & Weiss, A. Endogenous Nur77 is a specific indicator of antigen receptor signaling in human T and B cells. J. Immunol. 198, 657–668 (2017).

    Article  CAS  PubMed  Google Scholar 

  51. Bettelli, E. et al. Reciprocal developmental pathways for the generation of pathogenic effector TH17 and regulatory T cells. Nature 441, 235–238 (2006).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Ferraro, A. et al Interindividual variation in human T regulatory cells. Proc. Natl Acad. Sci. USA 111, E1111–E1120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Kim, D. et al. TopHat2: accurate alignment of transcriptomes in the presence of insertions, deletions and gene fusions. Genome. Biol. 14, R36 (2013).

    Article  PubMed  PubMed Central  Google Scholar 

  55. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome. Biol. 15, 550 (2014).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Grün, D. et al. Single-cell messenger RNA sequencing reveals rare intestinal cell types. Nature 525, 251–255 (2015).

    Article  PubMed  Google Scholar 

  57. Krijthe, J. H. Rtsne: T-Distributed Stochastic Neighbor Embedding using a Barnes–Hut Implementation. GitHub https://github.com/jkrijthe/Rtsne/ (2015).

  58. Hennig, C. fpc: Flexible Procedures for Clustering. R package version 2.1-10. Comprehensive R Archive Network https://CRAN.R-project.org/package=fpc (2015).

  59. Godec, J. et al. Compendium of immune signatures identifies conserved and species-specific biology in response to inflammation. Immunity 44, 194–206 (2016).

  60. Baker, F. J., Lee, M., Chien, Y. H. & Davis, M. M. Restricted islet-cell reactive T cell repertoire of early pancreatic islet infiltrates in NOD mice. Proc. Natl. Acad. Sci. USA 99, 9374–9379 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Burzyn, D. et al. A special population of regulatory T cells potentiates muscle repair. Cell 155, 1282–1295 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Bolotin, D. A. et al. MiXCR: software for comprehensive adaptive immunity profiling. Nat. Methods 12, 380–381 (2015).

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank E. Sefik for help with colon profiling and K. Hattori, C. Araneo K. Waraska, M. Thorsen and R. Steen for help with mice, profiling, sorting and sequencing. This work was supported by NIH grants AI051530, AI116834 and AI125603 to C.B./D.M. D.Z. and E.K. were supported by a PhD fellowship from the Boehringer Ingelheim Fonds.

Author information

Authors and Affiliations

Authors

Contributions

D.Z., E.K. and R.Z. performed the experiments; D.Z., A.M.K., C.B. and D.M. designed the study, and analyzed and interpreted the data; D.Z., D.M. and C.B. wrote the manuscript.

Corresponding authors

Correspondence to Diane Mathis or Christophe Benoist.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Publisher’s note: Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Integrated supplementary information

Supplementary Figure 1 Quality-control plots and algorithm to identify contaminant cells in scRNA-seq.

a. Scatterplot matrix comparing the collapsed gene expression profiles of the Treg scRNAseq replicates, with Pearson correlation and P value (t test). b. Scatterplot matrix comparing the variability in gene expression (Fano Factor = variance/mean) within each of the different Treg scRNAseq replicates, with Pearson correlation and P value (t test). c. Illustration of the naïve Bayes algorithm used to find contaminant cell in the datasets. We calculated the likelihood for each single cell transcriptome to be drawn from any of the 249 immune cell types profiled by microarrays by the Immgen consortium (Heng, T. S. & Painter, M. W. The Immunological Genome Project: networks of gene expression in immune cells. Nat. Immunol. 9, 1091–1094 (2008)). Venn diagrams: Distribution of the most likely Immgen immune cell type assigned in each independent Tconv and Treg scRNAseq dataset. d. Treg vs Tconv changes in gene expression in pooled scRNAseq (x-axis) compared with the changes in a classic population RNAseq experiment (y-axis). Highlighted genes are genes are the canonical Treg signature genes12.

Supplementary Figure 2 Furtive Tregs can be found in multiple independent datasets and express FoxP3 by flow cytometry.

a. Distribution of the number of detected genes in Tconvs (in E), Tregs (outside E) and furtive Tregs (in E). E corresponds to the cell cluster delineated in Fig. 2a. Error bars correspond to the mean +- 1 standard deviation. b. Distribution of the number of uniquely identified mRNA reads per cell in Tconvs (in E), Tregs (outside E) and furtive Tregs (in E). E corresponds to the cell cluster delineated in Fig. 2a. Error bars correspond to the mean +- 1 standard deviation. c. Two-dimensional tSNE plot of TCRβ+ CD4+ T cells sequenced using the 10X Genomics platform. Tregs (red) and Tconvs (black) were identified using the naive Bayes algorithm that compares each single-cell transcriptome with each of the 249 immune-cell states profiled by the Immgen Consortium (see Methods). The Tconv area used to identify furtive Tregs is shown along with the proportion of Tregs and Tconvs relative to the total number of Tregs and Tconvs. d. Same tSNE plots as in c, but split to show Treg and Tconv cells separately (top row) and cells in which Foxp3 transcript was detected (bottom row). e. Two-dimensional tSNE plot of index-sorted TCRβ+ CD4+ FoxP3-GFP+ Tregs and FoxP3-GFP- Tconvs sequenced by CEL-Seq. The tSNE is split to show Tregs and Tconvs separately. The Tconv area used to identify furtive Tregs is shown along with the proportion of Tregs and Tconvs relative to the total number of Tregs and Tconvs. f. FoxP3-GFP protein expression (flow cytometry intensity) distribution of indexed-sorted Tconvs, Tregs and furtive Tregs shown in e.

Supplementary Figure 3 FoxP3 binds to the core Treg-signature genes.

a. Core Treg signature genes at the single cell level are the genes that were found to be consistently differentially expressed (>80%) when we compare a sample of closely related Treg and Tconv in the transcriptional space. For each single Treg cell (n = 708), we compared the expression profiles of its 50 closest Tregs and 50 closest Tconvs (by Pearson correlation distance) and deemed significant genes with a P value <0.05. We report in this plot the average of the gene expression fold change across the 708 comparisons (x axis) and the fraction of tests where this difference was significant (y axis). Canonical Treg signature genes are highlighted in red and blue12. b. Chromatin profiles for core Treg signature genes . Top to bottom: ATAC-seq in Tconvs, ATAC-seq in Tregs, FoxP3 Chip-Seq in Tregs (data from 29).

Supplementary Figure 4 Robustness of the splenic Treg clusters.

a. Scatterplot of the average gene expression in single Treg cells and variability of expression (Fano factor = variance/mean). Top 100 most variable genes expressed in >1% cells are in red and were used in Fig. 4 for clustering. b. Jaccard index of the Treg clusters after kmeans bootstrapping (measures how many times cells consistently clustered together). c. Silhouette plot of the Treg clusters after kmeans bootstrapping (measure of how close is each single cell to the cells in the same clusters vs to the cells in the neighbor cluster). d. Biclustering expression heatmap, as Fig. 4a, for 2 independent splenic Treg scRNAseq datasets (n = 2035 and n = 1,294 cells). Columns: Single Treg cells assigned to the same 6 clusters as Fig. 4 using a likelihood model (see Methods). Rows: most characteristic genes found in the 6 Treg clusters (per Fig. 4a). The proportion of Tregs in each cluster in indicated. e. Circular projection plot of the likelihood for each single Treg cell in the 2 independent datasets to belong to any of the 6 mouse clusters described in Fig. 4.

Supplementary Figure 5 Signature enrichment in single Treg cell clusters.

Single-cell Treg expression of various genesets computed or obtained from previous Treg studies (reference included). Differently expressed signatures between clusters are represented here, ordered by hierarchal clustering. Single Treg cells were ordered by clusters (columns). P values by ANOVA.

Supplementary Figure 6 Nr4a1-GFP expression correlates with an enrichment in cluster 3 and an impoverishment in cluster 1 and 2 cells.

Circular projection plot of the likelihood for each single Treg cells in the Nr4a1-GFP high, medium and low independent datasets to belong to any of the 6 mouse clusters described in Fig. 4 (n = 1000 cells in each plot). The proportion of cells in each cluster is indicated.

Supplementary Figure 7 Human Treg and Tconv single-cell datasets: Quality-control plots and definition of the core human Treg transcripts.

a. Scatterplot matrix comparing the collapsed gene expression profiles of the Treg and Tconv scRNAseq replicates, with Pearson correlation and P value (t test). b. Scatterplot matrix comparing the variability in gene expression (Fano Factor = variance/mean) within each of the different Treg and Tconv scRNAseq replicates, with Pearson correlation and P value (t test). c. Treg vs Tconv changes in the gene expression in pooled scRNAseq (x-axis) compared with the changes in an independent population microarrray experiment (y-axis)53. Highlighted genes are genes are the canonical Treg signature genes. d. Same tSNE plots of human Treg and Tconv as in Fig. 7b, but split to show the contribution of each donor. e. Core human Treg signature. The same analysis as described in Fig. S2 was performed. This scatterplot compares the average fold change in gene expression (x axis) with the fraction of tests (y axis) that were significantly differently expressed between human Tregs and Tconvs (similar to Supplementary Fig. 3a). f. Nr4a1-GFP expression distribution in Tregs isolated from the blood and spleen of three mice.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7

Life Sciences Reporting Summary

Supplementary Table 1

Single-cell datasets created in this study

Supplementary Table 2

Top 100 most variable genes amongst splenic Tregs

Supplementary Table 3

Average gene expression in each splenic Treg cluster

Supplementary Table 4

Differentially expressed genes between splenic Treg clusters

Supplementary Table 5

DNA primer for TCR sequencing

Supplementary Data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zemmour, D., Zilionis, R., Kiner, E. et al. Single-cell gene expression reveals a landscape of regulatory T cell phenotypes shaped by the TCR. Nat Immunol 19, 291–301 (2018). https://doi.org/10.1038/s41590-018-0051-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-018-0051-0

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