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:

Distinct and temporary-restricted epigenetic mechanisms regulate human αβ and γδ T cell development

Abstract

The development of TCRαβ and TCRγδ T cells comprises a step-wise process in which regulatory events control differentiation and lineage outcome. To clarify these mechanisms, we employed RNA-sequencing, ATAC–sequencing and ChIPmentation on well-defined thymocyte subsets that represent the continuum of human T cell development. The chromatin accessibility dynamics show clear stage specificity and reveal that human T cell-lineage commitment is marked by GATA3- and BCL11B-dependent closing of PU.1 sites. A temporary increase in H3K27me3 without open chromatin modifications is unique for β-selection, whereas emerging γδ T cells, which originate from common precursors of β-selected cells, show large chromatin accessibility changes due to strong T cell receptor (TCR) signaling. Furthermore, we unravel distinct chromatin landscapes between CD4+ and CD8+ αβ-lineage cells that support their effector functions and reveal gene-specific mechanisms that define mature T cells. This resource provides a framework for studying gene regulatory mechanisms that drive normal and malignant human T cell development.

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: The transcriptome and accessible chromatin landscape of human T cell development.
Fig. 2: Discontinuous gene expression and chromatin accessibility dynamics during human T cell development.
Fig. 3: Transcriptional regulation of differentiating thymocytes by activating and repressing histone modifications.
Fig. 4: Decreased accessibility of PU.1 binding sites marks T cell commitment.
Fig. 5: GATA3 and BCL11B close PU.1 binding sites in early thymocytes.
Fig. 6: Differential regulation of the αβ–γδ bifurcation at the transcriptome and chromatin level.
Fig. 7: Early αβ T cell development is marked by a temporary increase in H3K27me3.
Fig. 8: Transcriptional and epigenetic characterization of mature naive αβ and γδ T cells.

Similar content being viewed by others

Data availability

The sequencing data have been deposited in the National Center for Biotechnology Information Gene Expression Omnibus (GEO) and are available through the GSE151081 accession ID. Publicly available data were retrieved from the ChIP Atlas or the GEO with the following GEO accession codes: GSM2893605, GSM3686939, GSM1557623, GSM2592810, GSM1816090, GSM1013125, GSM1059446 and GSM956030. Any other data that support the findings of the present study are available from the corresponding author upon request.

References

  1. Willcox, B. E. & Willcox, C. R. γδ TCR ligands: the quest to solve a 500-million-year-old mystery. Nat. Immunol. 20, 121–128 (2019).

    Article  CAS  PubMed  Google Scholar 

  2. Melandri, D. et al. The γδTCR combines innate immunity with adaptive immunity by utilizing spatially distinct regions for agonist selection and antigen responsiveness. Nat. Immunol. 19, 1352–1365 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Silva-Santos, B., Mensurado, S. & Coffelt, S. B. γδ T cells: pleiotropic immune effectors with therapeutic potential in cancer. Nat. Rev. Cancer 19, 392–404 (2019).

    Article  CAS  PubMed  Google Scholar 

  4. Hayday, A. C. γδ T cell update: adaptate orchestrators of immune surveillance. J. Immunol. 203, 311–320 (2019).

    Article  CAS  PubMed  Google Scholar 

  5. Wu, Y. et al. An innate-like Vδ1+ γδ T cell compartment in the human breast is associated with remission in triple-negative breast cancer. Sci. Transl. Med. 11, eaax9364 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Ravens, S. et al. Human γδ T cells are quickly reconstituted after stem-cell transplantation and show adaptive clonal expansion in response to viral infection. Nat. Immunol. 18, 393–401 (2017).

    Article  CAS  PubMed  Google Scholar 

  7. Ribeiro, M. et al. Meningeal γδ T cell-derived IL-17 controls synaptic plasticity and short-term memory. Sci. Immunol. 4, eaay5199 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Ciofani, M. & Zuniga-Pflucker, J. C. Determining γδ versus αβ T cell development. Nat. Rev. Immunol. 10, 657–663 (2010).

    Article  CAS  PubMed  Google Scholar 

  9. Lee, S. Y., Stadanlick, J., Kappes, D. J. & Wiest, D. L. Towards a molecular understanding of the differential signals regulating αβ/γδ T lineage choice. Semin. Immunol. 22, 237–246 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Taghon, T., Yui, M. A., Pant, R., Diamond, R. A. & Rothenberg, E. V. Developmental and molecular characterization of emerging β- and γδ-selected pre-T cells in the adult mouse thymus. Immunity 24, 53–64 (2006).

    Article  CAS  PubMed  Google Scholar 

  11. Dik, W. A. et al. New insights on human T cell development by quantitative T cell receptor gene rearrangement studies and gene expression profiling. J. Exp. Med. 201, 1715–1723 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Van de Walle, I. et al. An early decrease in Notch activation is required for human TCR-αβ lineage differentiation at the expense of TCR-γδ T cells. Blood 113, 2988–2998 (2009).

    Article  CAS  PubMed  Google Scholar 

  13. Boehm, T. & Bleul, C. C. Thymus-homing precursors and the thymic microenvironment. Trends Immunol. 27, 477–484 (2006).

    Article  CAS  PubMed  Google Scholar 

  14. Bhandoola, A., von Boehmer, H., Petrie, H. T. & Zuniga-Pflucker, J. C. Commitment and developmental potential of extrathymic and intrathymic T cell precursors: plenty to choose from. Immunity 26, 678–689 (2007).

    Article  CAS  PubMed  Google Scholar 

  15. Hao, Q. L. et al. Human intrathymic lineage commitment is marked by differential CD7 expression: identification of CD7 lympho-myeloid thymic progenitors. Blood 111, 1318–1326 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Weerkamp, F. et al. Human thymus contains multipotent progenitors with T/B lymphoid, myeloid, and erythroid lineage potential. Blood 107, 3131–3137 (2006).

    Article  CAS  PubMed  Google Scholar 

  17. Lavaert, M. et al. Integrated scRNA-seq identifies human postnatal thymus seeding progenitors and regulatory dynamics of differentiating immature thymocytes. Immunity 52, 1088–1104 (2020).

    Article  CAS  PubMed  Google Scholar 

  18. Zhou, W. et al. Single-cell analysis reveals regulatory gene expression dynamics leading to lineage commitment in early T cell development. Cell Syst. 9, 321–337.e9 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Yui, M. A. & Rothenberg, E. V. Developmental gene networks: a triathlon on the course to T cell identity. Nat. Rev. Immunol. 14, 529–545 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Hayes, S. M., Li, L. & Love, P. E. TCR signal strength influences αβ/γδ lineage fate. Immunity 22, 583–593 (2005).

    Article  CAS  PubMed  Google Scholar 

  21. Haks, M. C. et al. Attenuation of γδTCR signaling efficiently diverts thymocytes to the αβ lineage. Immunity 22, 595–606 (2005).

    Article  CAS  PubMed  Google Scholar 

  22. Lauritsen, J. P. et al. Marked induction of the helix–loop–helix protein Id3 promotes the γδ T cell fate and renders their functional maturation Notch independent. Immunity 31, 565–575 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Taghon, T. & Rothenberg, E. V. Molecular mechanisms that control mouse and human TCR-αβ and TCR-γδ T cell development. Semin. Immunopathol. 30, 383–398 (2008).

    Article  CAS  PubMed  Google Scholar 

  24. Blom, B. et al. Disruption of αβ but not of γδ T cell development by overexpression of the helix–loop–helix protein Id3 in committed T cell progenitors. EMBO J. 18, 2793–2802 (1999).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Hu, G. et al. Expression and regulation of intergenic long noncoding RNAs during T cell development and differentiation. Nat. Immunol. 14, 1190–1198 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Mingueneau, M. et al. The transcriptional landscape of αβ T cell differentiation. Nat. Immunol. 14, 619–632 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Casero, D. et al. Long non-coding RNA profiling of human lymphoid progenitor cells reveals transcriptional divergence of B cell and T cell lineages. Nat. Immunol. 16, 1282–1291 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Cante-Barrett, K. et al. Loss of CD44dim expression from early progenitor cells marks T-cell lineage commitment in the human thymus. Front. Immunol. 8, 32 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  29. Buenrostro, J. D., Wu, B., Chang, H. Y. & Greenleaf, W. J. ATAC-seq: a method for assaying chromatin accessibility genome-wide. Curr. Protoc. Mol. Biol. 109, 21.29.1–21.29.9 (2015).

    Google Scholar 

  30. Park, J. E. et al. A cell atlas of human thymic development defines T cell repertoire formation. Science 367, eaay3224 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Joachims, M. L., Chain, J. L., Hooker, S. W., Knott-Craig, C. J. & Thompson, L. F. Human αβ and γδ thymocyte development: TCR gene rearrangements, intracellular TCRβ expression, and γδ developmental potential—differences between men and mice. J. Immunol. 176, 1543–1552 (2006).

    Article  CAS  PubMed  Google Scholar 

  32. Schmidl, C., Rendeiro, A. F., Sheffield, N. C. & Bock, C. ChIPmentation: fast, robust, low-input ChIP-seq for histones and transcription factors. Nat. Methods 12, 963–965 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Rothenberg, E. V., Ungerback, J. & Champhekar, A. Forging T-lymphocyte identity: intersecting networks of transcriptional control. Adv. Immunol. 129, 109–174 (2016).

    Article  CAS  PubMed  Google Scholar 

  34. Van de Walle, I. et al. GATA3 induces human T-cell commitment by restraining Notch activity and repressing NK-cell fate. Nat. Commun. 7, 11171 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Ha, V. L. et al. The T-ALL related gene BCL11B regulates the initial stages of human T-cell differentiation. Leukemia 31, 2503–2514 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Barski, A. AP-1 transcription factor reprograms T cell epigenome during activation. J. Immunol. 198(Suppl. 1), 124.6 (2017).

    Google Scholar 

  37. Coffey, F. et al. The TCR ligand-inducible expression of CD73 marks γδ lineage commitment and a metastable intermediate in effector specification. J. Exp. Med. 211, 329–343 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Taniuchi, I. CD4 helper and CD8 cytotoxic T cell differentiation. Annu Rev. Immunol. 36, 579–601 (2018).

    Article  CAS  PubMed  Google Scholar 

  39. Hosokawa, H. et al. Transcription factor PU.1 represses and activates gene expression in early T cells by redirecting partner transcription factor binding. Immunity 48, 1119–1134.e7 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Yukawa, M. et al. AP-1 activity induced by co-stimulation is required for chromatin opening during T cell activation. J. Exp. Med. 217, e20182009 (2020).

    Article  PubMed  CAS  Google Scholar 

  42. Dobin, A. et al. STAR: ultrafast universal RNA-seq aligner. Bioinformatics 29, 15–21 (2013).

    Article  CAS  PubMed  Google Scholar 

  43. Abdel-Wahab, O. et al. ASXL1 mutations promote myeloid transformation through loss of PRC2-mediated gene repression. Cancer Cell 22, 180–193 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Velten, L. et al. Human haematopoietic stem cell lineage commitment is a continuous process. Nat. Cell Biol. 19, 271–281 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Corces, M. R. et al. An improved ATAC-seq protocol reduces background and enables interrogation of frozen tissues. Nat. Methods 14, 959–962 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Taghon, T. et al. HOX-A10 regulates hematopoietic lineage commitment: evidence for a monocyte-specific transcription factor. Blood 99, 1197–1204 (2002).

    Article  CAS  PubMed  Google Scholar 

  47. Zhu, L. J. Integrative analysis of ChIP-chip and ChIP-seq dataset. Methods Mol. Biol. 1067, 105–124 (2013).

    Article  CAS  PubMed  Google Scholar 

  48. Zhu, L. J. et al. ChIPpeakAnno: a bioconductor package to annotate ChIP-seq and ChIP-chip data. BMC Bioinform. 11, 237 (2010).

    Article  CAS  Google Scholar 

  49. 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).

    PubMed  PubMed Central  Google Scholar 

  50. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  51. Chen, E. Y. et al. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinform. 14, 128 (2013).

    Article  Google Scholar 

  52. Kuleshov, M. V. et al. Enrichr: a comprehensive gene set enrichment analysis web server 2016 update. Nucleic Acids Res. 44, W90–W97 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Oki, S. et al. ChIP-Atlas: a data-mining suite powered by full integration of public ChIP-seq data. EMBO Rep. 19, e46255 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  54. Ramírez, F., Dündar, F., Diehl, S., Grüning, B. A. & Manke, T. deepTools: a flexible platform for exploring deep-sequencing data. Nucleic Acids Res. 42, W187–W191 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank C. de Bock for help on the ATAC–seq protocol; S. Vermaut and K. Reynvoet for flow cytometry help; K. Weening for assistance in molecular cloning; K. Francois and G. Van Nooten (Department of Cardiac Surgery, Ghent University Hospital) for thymus tissue; the Red Cross Flanders for CB; and the Ghent University Hospital Hematopoietic Biobank. This work was supported by the Fund for Scientific Research Flanders (FWO, grant nos. G037514N, G053816N and G053916N to T.T., fellowship grant to S.S.); the Concerted Research Action from the Ghent University Research Fund (GOA, grant no. BOF18-GOA-024 to T.T. and P.V.V.); the European Research Council (ERC, grant no. StG 639784 to P.V.V.); the Foundation against Cancer (Stichting Tegen Kanker, grant no. FAF-F/2016/824 to T.T.); and the Cancer Research Institute Ghent (CRIG, YIPOC to A.K.). The computational resources and services used in this work were provided by the VSC (Flemish Supercomputer Center), funded by the Research Foundation—Flanders (FWO) and the Flemish Government, Department EWI.

Author information

Authors and Affiliations

Authors

Contributions

J.R., A.K., M.D.D., S.S., K.L.L. and F.V.N. performed the experiments. J.R., A.K. and M.L. performed the analyses. G.L., B.V. and P.V.V. provided critical reagents and J.R., A.K. and T.T. designed the research and wrote the paper. All authors have seen, reviewed and approved the final version.

Corresponding author

Correspondence to Tom Taghon.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Peer reviewer reports are available. Ioana Visan and Laurie A. Dempsey were the primary editors on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Schematic overview of human T cell development.

Schematic overview of the sorted populations that represent all major stages of human T cell development. During T cell development, a CD34+ hematopoietic precursor cell (HPC) enters the thymus. Immature CD34+ thymocytes undergo a T cell specification (CD34+CD1) and T cell commitment (CD34+CD1+) phase. Further differentiation leads to the upregulation of CD4. These early CD34+CD4+ immature single positive thymocytes (ISP) are the precursors of both αβ- and γδ-lineage cells. The combination of an in-frame rearranged TCRβ chain with the invariant preTCRα induces β-selection and the generation of ISP CD28+ T cells that further mature towards double positive and eventually single positive αβ-lineage T cells. Alternatively, when a TCRγδ is successfully rearranged and expressed in CD34+CD4+ precursor cells, differentiation towards the γδ T-cell lineage is predominantly initiated. Immature γδ CD1+ T cells then further differentiate towards mature naïve γδ CD1 T cells.

Extended Data Fig. 2 Feature distribution of ATAC peaks.

a, Mean read count frequency of ATAC peaks at the transcription start site. b, Mean frequency (two donors) of genomic feature distribution of all significant ATAC peaks in all stages of T cell development.

Extended Data Fig. 3 Integration of bulk RNAseq with single cell RNAseq of human postnatal thymus.

a, Uniform manifold approximation and projection (UMAP) of the single cell RNAseq dataset available from30 with integration of our 11 bulk RNAseq subsets (red diamonds). DN: CD4 CD8 double negative; DP: CD4+ CD8+; P: proliferative; Q: quiescent; T(agonist): agonist selected T cells; DP (late): Positively selected DP T cells. b, Classification of single cells to each of the 11 bulk populations by logistic regression. c, Probability that single cells are classified according to any of the bulk RNAseq samples by logistic regression.

Extended Data Fig. 4 Branching of αβ and γδ T cells at the single cell level.

a, STEMNET analysis to predict the branching of αβ and γδ T cell lineages based on similarities in the transcriptome. For this analysis, the single cell annotation corresponding to the bulk RNAseq populations from panel Extended Data Fig. 3b was used (denoted as ‘scthymic_population’, for example scCD34+CD1 are all single cells classified as CD34+CD1 by using our sorted population as a reference). Similarity of the transcriptome of scγδ CD1+ or scDP CD3+ T cells to more immature cell populations is shown (n = 5 donors, individual datapoints and standard deviation are shown). Cells were classified to either scγδ CD1+ or scDP CD3+ by probability of similarity. This can be interpreted as the relative potential that a certain subset would directly give rise to the corresponding reference subset. This revealed the branching point of αβ and γδ T cells; at this point the potential to directly give rise to γδ CD1+ dropped below the potential to directly form DP CD3+ T cells, thus marking the bifurcation point. In this analysis, CD34+CD4+ cells have mixed lineage potential, but also ISP CD28+ could still give rise to γδ CD1+ cells whereas the probability for DP CD3 cells to form γδ CD1+ cells is very low. b, Expression of RAG1 and RAG2 in pseudo-bulk counts of single cell populations from Extended Data Fig. 3b. The expression of RAG genes indicates the capacity to rearrange the TCR chain; only from the CD34+CD4+ subsets RAG expression appears and can thus support the formation of functional αβ and γδ T cells.

Extended Data Fig. 5 K-means clustering of significantly variably expressed genes.

Scaled, log transformed gene expression of genes inside each of the 25 k-means clusters of the significantly variably expressed genes as determined by Chi-squared test (p adj.< 0.05). Indication of a selection of genes inside each cluster.

Extended Data Fig. 6 Changes in RNAseq and ATACseq during early T cell development.

Volcano plots of a, gene expression changes and b, chromatin accessibility changes during early T cell development (CD34+CD1 compared to HPC).

Extended Data Fig. 7 H3K27ac and H3K27me3 peaks that overlap with open chromatin sites closed upon T cell commitment.

H3K27ac and H3K27me3 peaks that overlap with open chromatin sites that are more accessible in CD34+CD1 compared to CD34+CD1+ thymocytes (Fig. 4a). Numbers of overlapping regions are indicated on the left of each heatmap.

Extended Data Fig. 8 Genome browser view of ATACseq and histone marks at the SOX13, ZBTB16, RORC and RAG gene loci.

Tracks show open chromatin regions and ChIPmentation data for indicated histone marks for genomic loci at selected stages of thymocyte development as indicated. Overlay for two donors is shown.

Extended Data Fig. 9 TCR stimulation of ISP CD28+ T cells diverts development from the DP αβ T cell fate.

a, Flow cytometric analysis of CD73 expression in β-selected ISP CD28+, total DP, total γδ, SP CD4+ and SP CD8+ T cells. b, NT5E (coding for CD73) gene expression in αβ and γδ T cells. CD34+CD4+ are shown as reference values from which γδ and αβ T cells develop and are representative for 2 donors. c,d, Flow cytometric analysis of CD4 and CD73 (c) and CD4 and CD8 (d) staining in unstimulated β-selected ISP CD28+ thymocytes and ISP CD28+ thymocytes stimulated 24 hours with 2.5 μL (‘Low stimulated’) or 10 μL (‘High stimulated’) CD3/CD28 ImmunoCult per 100 000 cells after 6 days of culture, representative for 3 donors. (a,c,d) Numbers indicate the frequencies for each corresponding quadrant.

Extended Data Fig. 10 TCR stimulation has a minimal impact on the transcriptome.

a, Number of differentially expressed genes in pairwise comparisons between different conditions. The upper right part of the diagonal indicates upregulated genes in the conditions indicated on the x-axis compared to conditions on the y-axis. The lower left part of the diagonal indicates genes downregulated in conditions on the x-axis as compared to the y-axis. The comparison with the largest number of differentially expressed genes is marked (for example 4645 genes are significantly upregulated in unstimulated (‘No stim’) compared to ISP CD28+ thymocytes). b, Volcano plot of gene expression in stimulated (“Low”+”High”) compared to unstimulated cells. Triangles indicate datapoints outside of axis range.

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Roels, J., Kuchmiy, A., De Decker, M. et al. Distinct and temporary-restricted epigenetic mechanisms regulate human αβ and γδ T cell development. Nat Immunol 21, 1280–1292 (2020). https://doi.org/10.1038/s41590-020-0747-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-020-0747-9

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