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.

  • Resource
  • Published:

Molecular mechanisms of lineage decisions in metabolite-specific T cells

An Author Correction to this article was published on 24 January 2023

This article has been updated

Abstract

Mucosal-associated invariant T cells (MAIT cells) recognize the microbial metabolite 5-(2-oxopropylideneamino)-6-d-ribitylaminouracil (5-OP-RU) presented by the MHC class Ib molecule, MR1. MAIT cells acquire effector functions during thymic development, but the mechanisms involved are unclear. Here we used single-cell RNA-sequencing to characterize the developmental path of 5-OP-RU-specific thymocytes. In addition to the known MAIT1 and MAIT17 effector subsets selected on bone-marrow-derived hematopoietic cells, we identified 5-OP-RU-specific thymocytes that were selected on thymic epithelial cells and differentiated into CD44 naive T cells. MAIT cell positive selection required signaling through the adapter, SAP, that controlled the expression of the transcription factor, ZBTB16. Pseudotemporal ordering of single cells revealed transcriptional trajectories of 5-OP-RU-specific thymocytes selected on either thymic epithelial cells or hematopoietic cells. The resulting model illustrates T cell lineage decisions.

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: ScRNA-seq identifies distinct MAIT subsets in the thymus.
Fig. 2: ScRNA-seq identifies naive MR1:5-OP-RU+ cells.
Fig. 3: Distinct cells select naive or effector MR1:5-OP-RU+ T cells.
Fig. 4: SAP controls positive selection of MAIT cells.
Fig. 5: Modeling MAIT cell development on positive selection.
Fig. 6: Network inference identifies regulators of MAIT cell ontogeny.
Fig. 7: SAP signaling enhances EGR2 expression and induces ZBTB16.

Similar content being viewed by others

Data availability

Microarray data, sequencing data, raw and processed expression matrices produced by the different pipelines have been deposited at the European Bioinformatics Institute (EMBL-EBI) under the accession numbers E-MTAB-7702 (microarray) and E-MTAB-7704 (single-cell RNA-seq). The data that support the findings of this study are available from the corresponding author upon request.

Change history

References

  1. Boudinot, P. et al. Restricting nonclassical MHC genes coevolve with TRAV genes used by innate-like T cells in mammals. Proc. Natl Acad. Sci. USA 113, E2983–E2992 (2016).

    Article  CAS  Google Scholar 

  2. Treiner, E. et al. Selection of evolutionarily conserved mucosal-associated invariant T cells by MR1. Nature 422, 164–169 (2003).

    Article  CAS  Google Scholar 

  3. Franciszkiewicz, K. et al. MHC class I-related molecule, MR1, and mucosal-associated invariant T cells. Immunol. Rev. 272, 120–138 (2016).

    Article  CAS  Google Scholar 

  4. Koay, H. F. et al. A three-stage intrathymic development pathway for the mucosal-associated invariant T cell lineage. Nat. Immunol. 17, 1300–1311 (2016).

    Article  CAS  Google Scholar 

  5. Martin, E. et al. Stepwise development of MAIT cells in mouse and human. PLoS Biol. 7, e54 (2009).

    Article  Google Scholar 

  6. Salou, M. et al. A common transcriptomic program acquired in the thymus defines tissue residency of MAIT and NKT subsets. J. Exp. Med. 216, 133–151 (2019).

    Article  CAS  Google Scholar 

  7. Legoux, F., Salou, M. & Lantz, O. Unconventional or preset alphabeta T cells: evolutionarily conserved tissue-resident T cells recognizing nonpeptidic ligands. Ann. Rev. Cell Dev. Biol. 33, 511–535 (2017).

    Article  CAS  Google Scholar 

  8. Bendelac, A. Positive selection of mouse NK1+ T cells by CD1-expressing cortical thymocytes. J. Exp. Med. 182, 2091–2096 (1995).

    Article  CAS  Google Scholar 

  9. Seach, N. et al. Double-positive thymocytes select mucosal-associated invariant T cells. J. Immunol. 191, 6002–6009 (2013).

    Article  CAS  Google Scholar 

  10. Griewank, K. et al. Homotypic interactions mediated by Slamf1 and Slamf6 receptors control NKT cell lineage development. Immunity 27, 751–762 (2007).

    Article  CAS  Google Scholar 

  11. Savage, A. K. et al. The transcription factor PLZF directs the effector program of the NKT cell lineage. Immunity 29, 391–403 (2008).

    Article  CAS  Google Scholar 

  12. Mao, A. P. et al. Multiple layers of transcriptional regulation by PLZF in NKT-cell development. Proc. Natl Acad. Sci. USA 113, 7602–7607 (2016).

    Article  CAS  Google Scholar 

  13. Dutta, M. et al. A role for Ly108 in the induction of promyelocytic zinc finger transcription factor in developing thymocytes. J. Immunol. 190, 2121–2128 (2013).

    Article  CAS  Google Scholar 

  14. Tuttle, K. D. et al. TCR signal strength controls thymic differentiation of iNKT cell subsets. Nat. Commun. 9, 2650 (2018).

    Article  Google Scholar 

  15. Seiler, M. P. et al. Elevated and sustained expression of the transcription factors Egr1 and Egr2 controls NKT lineage differentiation in response to TCR signaling. Nat. Immunol. 13, 264–271 (2012).

    Article  CAS  Google Scholar 

  16. Cui, Y. et al. Mucosal-associated invariant T cell-rich congenic mouse strain allows functional evaluation. J. Clin. Invest. 125, 4171–4185 (2015).

    Article  Google Scholar 

  17. Levine, J. H. et al. Data-driven phenotypic dissection of AML reveals progenitor-like cells that correlate with prognosis. Cell 162, 184–197 (2015).

    Article  CAS  Google Scholar 

  18. Kirchner, J. & Bevan, M. J. ITM2A is induced during thymocyte selection and T cell activation and causes downregulation of CD8 when overexpressed in CD4(+)CD8(+) double positive thymocytes. J. Exp. Med. 190, 217–228 (1999).

    Article  CAS  Google Scholar 

  19. Haghverdi, L., Lun, A. T. L., Morgan, M. D. & Marioni, J. C. Batch effects in single-cell RNA-sequencing data are corrected by matching mutual nearest neighbors. Nat. Biotechnol. 36, 421–427 (2018).

    Article  CAS  Google Scholar 

  20. Uehara, S., Grinberg, A., Farber, J. M. & Love, P. E. A role for CCR9 in T lymphocyte development and migration. J. Immunol. 168, 2811–2819 (2002).

    Article  CAS  Google Scholar 

  21. Weinreich, M. A. & Hogquist, K. A. Thymic emigration: when and how T cells leave home. J. Immunol. 181, 2265–2270 (2008).

    Article  CAS  Google Scholar 

  22. Wang, H. & Hogquist, K.A. CCR7 defines a precursor for murine iNKT cells in thymus and periphery. eLife 7, e34793 (2018).

    Article  Google Scholar 

  23. Constantinides, M. G., McDonald, B. D., Verhoef, P. A. & Bendelac, A. A committed precursor to innate lymphoid cells. Nature 508, 397–401 (2014).

    Article  CAS  Google Scholar 

  24. Trapnell, C. et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 32, 381–386 (2014).

    Article  CAS  Google Scholar 

  25. Davie, K. et al. A single-cell transcriptome atlas of the aging drosophila brain. Cell 174, 982–998 (2018).

    Article  CAS  Google Scholar 

  26. Huynh-Thu, V.A., Irrthum, A., Geurts, P. & Wehenkel, L. Inferring regulatory networks from expression data using tree-based methods. PLoS ONE 5, e12776 (2010).

    Article  Google Scholar 

  27. Longabaugh, W. J. R. et al. Bcl11b and combinatorial resolution of cell fate in the T-cell gene regulatory network. Proc. Natl Acad. Sci. USA 114, 5800–5807 (2017).

    Article  CAS  Google Scholar 

  28. Aibar, S. et al. SCENIC: single-cell regulatory network inference and clustering. Nat. Methods 14, 1083–1086 (2017).

    Article  CAS  Google Scholar 

  29. Clauset, A., Newman, M. E. & Moore, C. Finding community structure in very large networks. Phys. Rev. E 70, 066111 (2004).

    Article  Google Scholar 

  30. Mao, A. P., Ishizuka, I. E., Kasal, D. N., Mandal, M. & Bendelac, A. A shared Runx1-bound Zbtb16 enhancer directs innate and innate-like lymphoid lineage development. Nat. Commun. 8, 863 (2017).

    Article  Google Scholar 

  31. Lazarevic, V. et al. The gene encoding early growth response 2, a target of the transcription factor NFAT, is required for the development and maturation of natural killer T cells. Nat. Immunol. 10, 306–313 (2009).

    Article  CAS  Google Scholar 

  32. Lu, Y. et al. SLAM receptors foster iNKT cell development by reducing TCR signal strength after positive selection. Nat. Immunol. 20, 447–457 (2019).

    Article  CAS  Google Scholar 

  33. Zhao, F., Cannons, J. L., Dutta, M., Griffiths, G. M. & Schwartzberg, P. L. Positive and negative signaling through SLAM receptors regulate synapse organization and thresholds of cytolysis. Immunity 36, 1003–1016 (2012).

    Article  CAS  Google Scholar 

  34. Rahimpour, A. et al. Identification of phenotypically and functionally heterogeneous mouse mucosal-associated invariant T cells using MR1 tetramers. J. Exp. Med. 212, 1095–1108 (2015).

    Article  CAS  Google Scholar 

  35. Legoux, F. P. & Moon, J. J. Peptide:MHC tetramer-based enrichment of epitope-specific T cells. J. Vis. Exp. 22, 4420 (2012).

    Google Scholar 

  36. Picelli, S. et al. Full-length RNA-seq from single cells using Smart-seq2. Nat. Protoc. 9, 171–181 (2014).

    Article  CAS  Google Scholar 

  37. Dash, P. et al. Paired analysis of TCRalpha and TCRbeta chains at the single-cell level in mice. J. Clin. Invest. 121, 288–295 (2011).

    Article  CAS  Google Scholar 

  38. Patro, R., Duggal, G., Love, M. I., Irizarry, R. A. & Kingsford, C. Salmon provides fast and bias-aware quantification of transcript expression. Nat. Methods 14, 417–419 (2017).

    Article  CAS  Google Scholar 

  39. Zemmour, D. et al. Single-cell gene expression reveals a landscape of regulatory T cell phenotypes shaped by the TCR. Nat. Immunol. 19, 291–301 (2018).

    Article  CAS  Google Scholar 

  40. Soneson, C. & Robinson, M. D. Bias, robustness and scalability in single-cell differential expression analysis. Nat. Methods 15, 255–261 (2018).

    Article  CAS  Google Scholar 

  41. Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).

    Article  Google Scholar 

  42. Butler, A., Hoffman, P., Smibert, P., Papalexi, E. & Satija, R. Integrating single-cell transcriptomic data across different conditions, technologies, and species. Nat. Biotechnol. 36, 411–420 (2018).

    Article  CAS  Google Scholar 

  43. Robinson, M. D., McCarthy, D. J. & Smyth, G. K. edgeR: a Bioconductor package for differential expression analysis of digital gene expression data. Bioinformatics 26, 139–140 (2010).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the ICGex NGS platform of the Institut Curie (S. Lameiras, S. Baulande, M. Bohec) for technical help with single-cell RNA-seq experiments. ICGex is supported by the grants ANR10EQPX03 (Equipex) and ANR10INBS0908 (France Génomique Consortium) from the Agence Nationale de la Recherche (‘Investissements d’Avenir’ program), by the Canceropole Ile-de-France and by the SiRICCurie program. For the affymetrix data, we thank A. Vieillefon, A. Rapinat and D. Gentien from the Genomics Platform of the Translational Research Department at Institut Curie. We thank V. Dangles-Marie, M. Garcia, C. Pauchard, the mouse facility technicians and the flow cytometry core at Institut Curie. We thank N. Manel for reviewing the manuscript. F.L. was supported by a Marie-Skłodowska Curie individual fellowship (706353) from the European Commission. This work was supported by the Institut National de la Santé et de la Recherche Médicale, Institut Curie, Agence Nationale de la Recherche (Blanc (MAIT, NEOMAIT and DIABMAIT) and Labex DCBIOL). O.L.’s group was supported by the ‘Equipe labellisée de la Ligue Contre le Cancer’.

Author information

Authors and Affiliations

Authors

Contributions

F.L. designed and performed experiments, analyzed data and wrote the paper. J.G. designed and performed the bioinformatic analysis, analyzed data and wrote the paper. E.P. performed experiments. K.E. and K.B. provided key reagents. O.L. supervised the project and wrote the paper.

Corresponding authors

Correspondence to François Legoux or Olivier Lantz.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information: Ioana Visan was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team

Integrated supplementary information

Supplementary Figure 1 Single-cell RNA-seq identifies distinct immature and mature MAIT cell subsets in the thymus.

(a) Schematic representation of the experimental design used for generation of scRNA-seq data from tetramer+ thymocytes. (b) Gating strategy used throughout the study for the identification of tetramer+ cells. (c) Heatmap showing expression of the DE genes between different thymocyte subsets (log2, microarray), defining the T cell signatures used in Fig. 1. (d) Density histogram showing the numbers of UMIs detected per cell in 2 independent scRNA-seq analyses of WT tetramer+ thymocytes. Dashed blue lines represent the threshold used to define droplets containing true cells. Replicate wt02 has a higher fraction of droplets with low-transcriptional content, presumably cellular debris (arrow). (f) Scatter plots showing quality control metrics in the 2 independent scRNA-seq experiments. The violin represents the probability density at each value. Each dot represents one cell (wt01 = 3,319 cells, wt02 = 2,827 cells). (f) Scatter plot showing the number of cells per gene in the 2 independent scRNA-seq experiments. (g) Left: PCA, tSNE and UMAP representation of cell clusters identified in the 2 independent scRNA-seq experiments. Right: Expression levels of Cd24a, Rorc and Tbx21 on a UMAP embedding of both independent experiments. (h) UMAP visualization of the mutual nearest neighbor (MNN) correction across both scRNA-seq datasets.

Supplementary Figure 2 scRNA-seq identifies a small subset of naïve MR1:5-OP-RU-specific T cells in thymus and spleen.

(a) Histogram showing Trbv gene segment expression in scRNA-seq data. (b) Flow cytometry showing CD4 and CD8α expression in tetramer+CD24-CD62L+CD44- thymocytes from WT and Mr1−/− B6-MAITCast mice following tetramer-based magnetic enrichment. Representative of 2 independent experiments.

Supplementary Figure 3 SAP controls effector differentiation of MAIT cells.

(a) Left: tetramer staining and TCR-β expression in splenocytes from the indicated B6-MAITCast strains following tetramer-based enrichment. Representative of 2 independent experiments. Right: Frequency of tetramer+ splenocytes from WT (n=5), Sh2d1ay/− (n=7) and Mr1−/− (n=5) mice before magnetic enrichment. *p<0.01 (two-tailed Mann-Whitney tests). (b). TCR-β mfi in tetramer+ thymocytes relative to tetramerCD24 SP8 thymocytes from the same sample. Data from WT (n=6), Sh2d1ay/− (n=5) and Mr1−/− (n=6) mice are shown. *p<0.01, **p<0.001 (two-tailed t tests) (c) CD4 and CD8α expression in CD24tetramer+ thymocytes from the indicated strains. Representative of 2 independent experiments. (d). MR1 expression on thymocytes from the indicated mice after in vitro incubation with synthetic 5-OP-RU. Representative of two independent experiments. (e) Percentage of TRBV13+ cells in the indicated tetramer+ populations from WT (n=6) or Sh2d1ay/− (n=3) mice, pooled from two independent experiments. (f) Left: Zbtb16-eGFP reporter expression in tetramer+CD24CD44 thymocytes from the indicated mice. Representative of 2 independent experiments. Right: Zbtb16-eGFP mfi in the indicated tetramer+ thymocytes subsets relative to tetramerCD24 SP8 thymocytes from the same sample (n=3, mean with SEM is indicated). (g) Left: intranuclear Ki67 expression in CD24CD44tetramer+ thymocytes from the indicated mice. Representative of 2 independent experiments. Right: Percentage of Ki67+tetramer+ thymocytes in the indicated subsets in WT (n=9), Sh2d1ay/− (n=3) and Mr1−/− (n=3) mice (mean with SEM is indicated). (h). Left: tetramer staining and CD44 expression in lung T cells of the indicated mice. Representative of 3 independent experiments. Right: Frequency of tetramer+ cells in lungs of WT (n=8), Sh2d1ay/− (n=5) and Mr1−/− (n=5) mice. *p<0.01 (two-tailed Mann-Whitney tests).

Supplementary Figure 4 T cell development modeling reveals major transcriptional changes upon positive selection by hematopoietic cells.

(a) Zbtb16-eGFP reporter expression in tetramer+CD24CD44+ thymocytes expressing CD138 (MAIT17) or CD122 (MAIT1). Representative of 5 independent experiments. (b) Partition-based Approximate Graph Abstraction based on scRNA-seq data from WT tetramer+ thymocytes. Size of the nodes (dots) reflects the number of cells per cluster, thickness of the edges (lines) is proportional to the transition probability between cell types (minimum threshold=0.1). (c) Diffusion map based on scRNA-seq data from merged WT and Sh2d1ay/− tetramer+ thymocytes. (d) Raw counts of spliced and unspliced variants, intron retention, and signature of spliceosome inhibitors (see Supplementary Table 1a) visualized on a UMAP embedding. (e) Inference of transcriptional dynamics with velocyto. Vectors of RNA velocities (arrows) are projected on a UMAP embedding (left) or diffusion map embedding (right). Intron retention is defined as the level of unspliced transcripts over all forms of mRNA. Spliceosome inhibitor signature is derived from the list provided in Supplementary Table 1a-h.

Supplementary Figure 5 Network analysis identifies Egr2 as a key gene during early differentiation of MAIT cells.

(a) Gene regulatory networks inferred from WT and Sh2d1ay/− samples (representation layout: Kuwai-Kamai algorithm). (b) Top: Global inferred network showing gene communities identified by a greedy optimization of modularity. Bottom: Representation of the global network provided as supplementary material (cytoscape file), blue and orange edges represent expression linkages identified in WT and Sh2d1ay/− samples, respectively (representation layouts: Kuwai-Kamai algorithm). (c) Number of genes present in each community identified in (b). (d) List of the top 25 genes per community identified in (b), sorted by decreased normalized betweenness centrality. (e) Functional enrichment test of gene communities identified in (b) (see Methods). (f) Graph showing the start of expression, inferred from the pseudotime defined in Fig. 5, of the top 25 (by decreased betweenness centrality) TF genes from the communities identified in (b).

Supplementary Figure 6 Graphical summary of the mechanisms of lineage decisions in MR1:5-OP-RU-specific T cells.

Left: generation of naive metabolite-specific T cells upon positive selection by MR1 on thymic epithelial cells. Right: differentiation of effector MAIT cells selected by MR1 on hematopoietic cells. The color code refers to the clusters defined in Fig. 1b and Fig. 7c.

Supplementary information

Supplementary Information

Supplementary Figs. 1–6, Supplementary Table 2 and Supplementary Note

Reporting Summary

Supplementary Table 1a

Lists of gene signatures.

Supplementary Table 1b

Differentially expressed genes per cluster.

Supplementary Table 1c

Gene communities identified by network inference.

Supplementary Table 1d

Functional enrichment tests per gene community.

Cytoscape

Gene regulatory networks inferred from WT and Sh2d1ay/− samples

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Legoux, F., Gilet, J., Procopio, E. et al. Molecular mechanisms of lineage decisions in metabolite-specific T cells. Nat Immunol 20, 1244–1255 (2019). https://doi.org/10.1038/s41590-019-0465-3

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-019-0465-3

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