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:

Intrathymic dendritic cell-biased precursors promote human T cell lineage specification through IRF8-driven transmembrane TNF

Abstract

The cross-talk between thymocytes and thymic stromal cells is fundamental for T cell development. In humans, intrathymic development of dendritic cells (DCs) is evident but its physiological significance is unknown. Here we showed that DC-biased precursors depended on the expression of the transcription factor IRF8 to express the membrane-bound precursor form of the cytokine TNF (tmTNF) to promote differentiation of thymus seeding hematopoietic progenitors into T-lineage specified precursors through activation of the TNF receptor (TNFR)-2 instead of TNFR1. In vitro recapitulation of TNFR2 signaling by providing low-density tmTNF or a selective TNFR2 agonist enhanced the generation of human T cell precursors. Our study shows that, in addition to mediating thymocyte selection and maturation, DCs function as hematopoietic stromal support for the early stages of human T cell development and provide proof of concept that selective targeting of TNFR2 can enhance the in vitro generation of T cell precursors for clinical application.

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: TSP2- and HPC-annotated CD34+ human thymocytes express low levels of IRF8 and are bi-phenotypic and potent for T- and DC-lineages.
Fig. 2: IRF8 expression is induced during human T-lineage specification but is silenced in the subsequent commitment stage.
Fig. 3: Low dose of active IRF8 promotes generation of tmTNF-expressing CD7+CD123+ progenitors and T cell precursors.
Fig. 4: IRF8-dependent DC-biased CD123+CD127+ progenitors express tmTNF and augment generation of T cell precursors via cellular cross-talk.
Fig. 5: TNFR2 expression precedes induction of CD7 during early human T cell development.
Fig. 6: Selective targeting of TNFR2 enhances in vitro generation of T cell precursors.
Fig. 7: TNF-activated lymphoid progenitors downregulate expression of IFN-related genes and are competent in T-lineage development.

Similar content being viewed by others

Data availability

The datasets generated and/or analyzed in this study are available in the Gene Expression Omnibus with the following accession numbers: ex vivo human CD34+ thymocytes for scRNA-seq (GSE144870), 4-OHT-treated OP9-DLL4 cocultures for ATAC-seq (GSE179534) and RNA-seq (GSE179381) and TNF-activated ATO cultures for scRNA-seq (GSE211400). GRCh38 reference genome is publicly accessible at the NCBI Datasets Genomes. Source data are provided with this paper.

References

  1. Nunez, S. et al. The human thymus perivascular space is a functional niche for viral-specific plasma cells. Sci. Immunol. 1, eaah4447 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  2. 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 

  3. Watanabe, N. et al. Hassall’s corpuscles instruct dendritic cells to induce CD4+CD25+ regulatory T cells in human thymus. Nature 436, 1181–1185 (2005).

    Article  CAS  PubMed  Google Scholar 

  4. 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 

  5. Le, J. et al. Single-cell RNA-seq mapping of human thymopoiesis reveals lineage specification trajectories and a commitment spectrum in T cell development. Immunity 52, 1105–1118 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Martin-Gayo, E. et al. Spatially restricted JAG1-Notch signaling in human thymus provides suitable DC developmental niches. J. Exp. Med. 214, 3361–3379 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Han, J. & Zuniga-Pflucker, J. C. A 2020 view of thymus stromal cells in T cell development. J. Immunol. 206, 249–256 (2021).

    Article  CAS  PubMed  Google Scholar 

  8. Schmitt, T. M. & Zuniga-Pflucker, J. C. Induction of T cell development from hematopoietic progenitor cells by δ-like-1 in vitro. Immunity 17, 749–756 (2002).

    Article  CAS  PubMed  Google Scholar 

  9. Seet, C. S. et al. Generation of mature T cells from human hematopoietic stem and progenitor cells in artificial thymic organoids. Nat. Methods 14, 521–530 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Edgar, J. M., Michaels, Y. S. & Zandstra, P. W. Multi-objective optimization reveals time- and dose-dependent inflammatory cytokine-mediated regulation of human stem cell derived T-cell development. NPJ Regen. Med 7, 11 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Moirangthem, R.D. et al. A DL-4 and TNFα-based culture system to generate high numbers of nonmodified or genetically modified immunotherapeutic human T-lymphoid progenitors. Cell Mol. Immunol. 18, 1662–1676 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Smits, K. et al. Tumor necrosis factor promotes T-cell at the expense of B-cell lymphoid development from cultured human CD34+ cord blood cells. Exp. Hematol. 35, 1272–1278 (2007).

    Article  CAS  PubMed  Google Scholar 

  13. Weekx, S. F. et al. Generation of T cells from adult human hematopoietic stem cells and progenitors in a fetal thymic organ culture system: stimulation by tumor necrosis factor-α. Blood 95, 2806–2812 (2000).

    Article  CAS  PubMed  Google Scholar 

  14. Black, R. A. et al. A metalloproteinase disintegrin that releases tumour-necrosis factor-α from cells. Nature 385, 729–733 (1997).

    Article  CAS  PubMed  Google Scholar 

  15. Azzawi, M. & Hasleton, P. Tumour necrosis factor α and the cardiovascular system: its role in cardiac allograft rejection and heart disease. Cardiovasc. Res. 43, 850–859 (1999).

    Article  CAS  PubMed  Google Scholar 

  16. Parry, S. L., Sebbag, M., Feldmann, M. & Brennan, F. M. Contact with T cells modulates monocyte IL-10 production: role of T cell membrane TNF-α. J. Immunol. 158, 3673–3681 (1997).

    Article  CAS  PubMed  Google Scholar 

  17. Lee, J. et al. Lineage specification of human dendritic cells is marked by IRF8 expression in hematopoietic stem cells and multipotent progenitors. Nat. Immunol. 18, 877–888 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Taghon, T., Waegemans, E. & Van de Walle, I. Notch signaling during human T cell development. Curr. Top. Microbiol. Immunol. 360, 75–97 (2012).

    CAS  PubMed  Google Scholar 

  19. Marquez, C., Trigueros, C., Fernandez, E. & Toribio, M. L. The development of T and non-T cell lineages from CD34+ human thymic precursors can be traced by the differential expression of CD44. J. Exp. Med. 181, 475–483 (1995).

    Article  CAS  PubMed  Google Scholar 

  20. 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  Google Scholar 

  21. Cytlak, U. et al. Differential IRF8 transcription factor requirement defines two pathways of dendritic cell development in humans. Immunity 53, 353–370 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Zeng, Y. et al. Single-Cell RNA sequencing resolves spatiotemporal development of Pre-thymic lymphoid progenitors and thymus organogenesis in human embryos. Immunity 51, 930–948 (2019).

    Article  CAS  PubMed  Google Scholar 

  23. 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  Google Scholar 

  24. Cieslak, A. et al. RUNX1-dependent RAG1 deposition instigates human TCR-δ locus rearrangement. J. Exp. Med. 211, 1821–1832 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Van de Walle, I. et al. Jagged2 acts as a δ-like Notch ligand during early hematopoietic cell fate decisions. Blood 117, 4449–4459 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  26. Xu, H. et al. Notch-RBP-J signaling regulates the transcription factor IRF8 to promote inflammatory macrophage polarization. Nat. Immunol. 13, 642–650 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Shyamsunder, P. et al. Identification of a novel enhancer of CEBPE essential for granulocytic differentiation. Blood 133, 2507–2517 (2019).

    Article  CAS  PubMed  Google Scholar 

  28. Fischer, R., Kontermann, R. E. & Pfizenmaier, K. Selective targeting of TNF receptors as a novel therapeutic approach. Front. Cell Dev. Biol. 8, 401 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  29. Sontag, S. et al. Modelling IRF8 deficient human hematopoiesis and dendritic cell development with engineered iPS Cells. Stem Cells 35, 898–908 (2017).

    Article  CAS  PubMed  Google Scholar 

  30. Montel-Hagen, A. et al. Organoid-Induced differentiation of conventional t cells from human pluripotent stem cells. Cell Stem Cell 24, 376–389 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Aggarwal, S., Gollapudi, S. & Gupta, S. Increased TNF-α-induced apoptosis in lymphocytes from aged humans: changes in TNF-α receptor expression and activation of caspases. J. Immunol. 162, 2154–2161 (1999).

    Article  CAS  PubMed  Google Scholar 

  32. 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 

  33. Hoebeke, I. et al. T-, B- and NK-lymphoid, but not myeloid cells arise from human CD34+CD38CD7+ common lymphoid progenitors expressing lymphoid-specific genes. Leukemia 21, 311–319 (2007).

    Article  CAS  PubMed  Google Scholar 

  34. Grell, M. et al. The transmembrane form of tumor necrosis factor is the prime activating ligand of the 80 kDa tumor necrosis factor receptor. Cell 83, 793–802 (1995).

    Article  CAS  PubMed  Google Scholar 

  35. Grell, M., Wajant, H., Zimmermann, G. & Scheurich, P. The type 1 receptor (CD120a) is the high-affinity receptor for soluble tumor necrosis factor. Proc. Natl Acad. Sci. USA 95, 570–575 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Maney, N. J., Reynolds, G., Krippner-Heidenreich, A. & Hilkens, C. M. U. Dendritic cell maturation and survival are differentially regulated by TNFR1 and TNFR2. J. Immunol. 193, 4914–4923 (2014).

    Article  CAS  PubMed  Google Scholar 

  37. Dong, Y. et al. Essential protective role of tumor necrosis factor receptor 2 in neurodegeneration. Proc. Natl Acad. Sci. USA 113, 12304–12309 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Dybedal, I., Bryder, D., Fossum, A., Rusten, L. S. & Jacobsen, S. E. Tumor necrosis factor (TNF)-mediated activation of the p55 TNF receptor negatively regulates maintenance of cycling reconstituting human hematopoietic stem cells. Blood 98, 1782–1791 (2001).

    Article  CAS  PubMed  Google Scholar 

  39. Zielske, S. P. & Braun, S. E. Cytokines: value-added products in hematopoietic stem cell gene therapy. Mol. Ther. 10, 211–219 (2004).

    Article  CAS  PubMed  Google Scholar 

  40. Balan, S. et al. Large-scale human dendritic cell differentiation revealing notch-dependent lineage bifurcation and heterogeneity. Cell Rep. 24, 1902–1915 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Kirkling, M. E. et al. Notch signaling facilitates in vitro generation of cross-presenting classical dendritic cells. Cell Rep. 23, 3658–3672 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. 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 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Luche, H. et al. The earliest intrathymic precursors of CD8α+ thymic dendritic cells correspond to myeloid-type double-negative 1c cells. Eur. J. Immunol. 41, 2165–2175 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Moore, A. J. et al. Transcriptional priming of intrathymic precursors for dendritic cell development. Development 139, 373–384 (2012).

    Article  CAS  PubMed  Google Scholar 

  45. Porritt, H. E. et al. Heterogeneity among DN1 prothymocytes reveals multiple progenitors with different capacities to generate T cell and non-T cell lineages. Immunity 20, 735–745 (2004).

    Article  CAS  PubMed  Google Scholar 

  46. Rothenberg, E. V. Single-cell insights into the hematopoietic generation of T-lymphocyte precursors in mouse and human. Exp. Hematol. 95, 1–12 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Weber, B. N. et al. A critical role for TCF-1 in T-lineage specification and differentiation. Nature 476, 63–68 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. De Smedt, M. et al. T-lymphoid differentiation potential measured in vitro is higher in CD34+CD38/lo hematopoietic stem cells from umbilical cord blood than from bone marrow and is an intrinsic property of the cells. Haematologica 96, 646–654 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  49. Offner, F., Kerre, T., De Smedt, M. & Plum, J. Bone marrow CD34 cells generate fewer T cells in vitro with increasing age and following chemotherapy. Br. J. Haematol. 104, 801–808 (1999).

    Article  CAS  PubMed  Google Scholar 

  50. Patel, E. et al. Diverse T-cell differentiation potentials of human fetal thymus, fetal liver, cord blood and adult bone marrow CD34 cells on lentiviral Delta-like-1-modified mouse stromal cells. Immunology 128, e497–e505 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  51. Swat, W. et al. Essential role of PI3Kδ and PI3Kγ in thymocyte survival. Blood 107, 2415–2422 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Xue, L., Chiang, L., Kang, C. & Winoto, A. The role of the PI3K-AKT kinase pathway in T-cell development beyond the β checkpoint. Eur. J. Immunol. 38, 3200–3207 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Silva, A. et al. PTEN posttranslational inactivation and hyperactivation of the PI3K/Akt pathway sustain primary T cell leukemia viability. J. Clin. Invest. 118, 3762–3774 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Dolens, A. C., Van de Walle, I. & Taghon, T. Approaches to study human T cell development. Methods Mol. Biol. 1323, 239–251 (2016).

    Article  CAS  PubMed  Google Scholar 

  55. Dolens, A. C. et al. Distinct Notch1 and BCL11B requirements mediate human γδ/αβ T cell development. EMBO Rep. 21, e49006 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Taghon, T. et al. Enforced expression of GATA-3 severely reduces human thymic cellularity. J. Immunol. 167, 4468–4475 (2001).

    Article  CAS  PubMed  Google Scholar 

  57. Chen, E. L. Y. et al. Cutting edge: TCR-β selection is required at the CD4+CD8+ stage of human T cell development. J. Immunol. 206, 2271–2276 (2021).

    Article  CAS  PubMed  Google Scholar 

  58. Itoh, K. et al. Reproducible establishment of hemopoietic supportive stromal cell lines from murine bone marrow. Exp. Hematol. 17, 145–153 (1989).

    CAS  PubMed  Google Scholar 

  59. 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 

  60. Van de Walle, I., Davids, K. & Taghon, T. Characterization and isolation of human T cell progenitors. Methods Mol. Biol. 1323, 221–237 (2016).

    Article  PubMed  Google Scholar 

  61. McCarthy, D. J., Campbell, K. R., Lun, A. T. & Wills, Q. F. Scater: pre-processing, quality control, normalization and visualization of single-cell RNA-seq data in R. Bioinformatics 33, 1179–1186 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. 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 

  63. Andreatta, M. & Carmona, S. J. UCell: robust and scalable single-cell gene signature scoring. Comput Struct. Biotechnol. J. 19, 3796–3798 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Hao, Y. et al. Integrated analysis of multimodal single-cell data. Cell 184, 3573–3587 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Yui, M.A., Feng, N. & Rothenberg, E.V. Fine-scale staging of T cell lineage commitment in adult mouse thymus. J Immunol 185, 284–293 (2010).

    Article  CAS  PubMed  Google Scholar 

  66. De Smedt, M. et al. Active form of Notch imposes T cell fate in human progenitor cells. J. Immunol. 169, 3021–3029 (2002).

    Article  PubMed  Google Scholar 

  67. 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  PubMed  Google Scholar 

  68. Mace, E. M. et al. Biallelic mutations in IRF8 impair human NK cell maturation and function. J. Clin. Invest. 127, 306–320 (2017).

    Article  PubMed  Google Scholar 

  69. Feil, R., Wagner, J., Metzger, D. & Chambon, P. Regulation of Cre recombinase activity by mutated estrogen receptor ligand-binding domains. Biochem. Biophys. Res. Commun. 237, 752–757 (1997).

    Article  CAS  PubMed  Google Scholar 

  70. De Decker, M. et al. HES1 and HES4 have non-redundant roles downstream of Notch during early human T-cell development. Haematologica 106, 130–141 (2021).

    Article  PubMed  Google Scholar 

  71. 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 

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

    Article  CAS  PubMed  Google Scholar 

  73. Martin, M. Cutadapt removes adapter sequences from high-throughput sequencing reads. EMBnet J. 17, 3 (2011).

    Article  Google Scholar 

  74. Li, B. & Dewey, C. N. RSEM: accurate transcript quantification from RNA-seq data with or without a reference genome. BMC Bioinf. 12, 323 (2011).

    Article  CAS  Google Scholar 

  75. 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  PubMed  Google Scholar 

  76. Roels, J. et al. Distinct and temporary-restricted epigenetic mechanisms regulate human αβ and γδ T cell development. Nat. Immunol. 21, 1280–1292 (2020).

    Article  CAS  PubMed  Google Scholar 

  77. Gaspar, J. M. NGmerge: merging paired-end reads via novel empirically-derived models of sequencing errors. BMC Bioinf. 19, 536 (2018).

    Article  CAS  Google Scholar 

  78. Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  79. Langmead, B., Wilks, C., Antonescu, V. & Charles, R. Scaling read aligners to hundreds of threads on general-purpose processors. Bioinformatics 35, 421–432 (2019).

    Article  CAS  PubMed  Google Scholar 

  80. Lawrence, M. et al. Software for computing and annotating genomic ranges. PLoS Comput. Biol. 9, e1003118 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  81. 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 

  82. Hu, Y. & Smyth, G. K. ELDA: extreme limiting dilution analysis for comparing depleted and enriched populations in stem cell and other assays. J. Immunol. Methods 347, 70–78 (2009).

    Article  CAS  PubMed  Google Scholar 

  83. Lehmann, J. S., Zhao, A., Sun, B., Jiang, W. & Ji., S. Multiplex cytokine profiling of stimulated mouse splenocytes using a cytometric bead-based immunoassay platform. J. Vis. Exp. 129, 56440 (2017).

    Google Scholar 

  84. 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  PubMed  PubMed Central  Google Scholar 

  85. Mclnnes, L., Healy, J. & Melville, J. UMAP: uniform manifold approximation and projection for dimension reduction. Preprint at arXiv https://doi.org/10.48550/arXiv.1802.03426 (2018).

Download references

Acknowledgements

We thank C. de Bock (KU Leuven) for the ATAC-seq protocol, J.C. Zúñiga-Pflücker (University of Toronto) for OP9-DLL4-7FS stromal cell line, K. Francois and G. Van Nooten (Department of Human Structure and Repair, Ghent University Hospital) for thymus tissue, the Red Cross Flanders and the Ghent University Hospital Hematopoietic Biobank for cord blood and buffy coat, M. Guilliams (VIB, Ghent University) for an aliquot of IRF8 antibody, K. Weening and A. Kuchmiy (Ghent University) for assistance with molecular cloning, S. Vermaut and K. Reynvoet (Ghent University) for assistance with flow cytometry and cell sorting, F. Branco Madeira (Ghent University) for C57/BL6 mice, M. De Smedt and Jean Plum (Ghent University) for assistance in processing and collection of human tissue, E. De Meester (NXTGNT, Ghent University) for assistance in the preparation of samples for scRNA-seq and R. Colman (Ghent University) for assistance with statistical analyses. This work was supported by the Fund for Scientific Research Flanders (FWO, grants G053816N and G053916N to T.T.), The Concerted Research Action from the Ghent University Research Fund (GOA, BOF18-GOA-024 to T.T.), The Foundation against Cancer (Stichting Tegen Kanker, 2016-094 and 2020-114 to T.T.) and the Chan Zuckerberg Initiative (CZF2019-002445 to T.T.). 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. Research reported in this publication was performed at the CORE Flow Cytometry and NXTGNT sequencing facilities of Ghent University, Belgium.

Author information

Authors and Affiliations

Authors

Contributions

K.L.L. conceived the study, designed and performed experiments, analyzed data and wrote the manuscript. J.R. analyzed bulk ATAC-seq data. M.L. and T.P. analyzed the previously published scRNA-seq data. T.P. and L.B. analyzed the scRNA-seq data generated in this study. L.T. analyzed bulk RNA-seq data. I.V. assisted to set up experiments. I.V.W. performed an experiment related to regulation of IRF8 expression. J.V., B.V., G.L., P.V.V. and C.L. provided reagents. F.V.N provided expertise in ATAC- and RNA-seq. V.P., R.F., R.E.K. and K.P. provided TNFR2-selective TNF mutein (EHD2-scTNFR2). G.D. provided IRF8-related constructs. S.S. and M.Z. provided IRF8+/+ and IRF8−/− iPSCs. T.T. supervised the study, designed experiments and wrote the manuscript. All authors have seen, reviewed and approved the final version of the manuscript.

Corresponding author

Correspondence to Tom Taghon.

Ethics declarations

Competing interests

K.L.L. and T.T. filed a PCT application (PCT/EP2022/063712: Generating T cell precursors via agonizing tumor necrosis factor receptor 2) with the European Patent Office on 20th May 2022. K.P., R.F. and R.E.K. are named inventors on a patent covering the TNFR2-specific agonist (EZH2-scTNFR2). R.F. and R.E.K. received funding from Resano, a company that has licensed the technology to generate TNFR2-specific agonist. R.E.K. is a consultant for Immatics, Oncomatryx, Roche and SunRock. K.P. is a consultant for Oncomatryx, Resano and SunRock. The remaining authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks Matthew Collin, Ivan Maillard and Christopher Seet for their contribution to the peer review of this work. Peer reviewer reports are available. Primary Handling Editor: Ioana Visan, in collaboration with the Nature Immunology team.

Additional information

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

Extended data

Extended Data Fig. 1 Immunophenotyping of ex vivo murine and human immature thymocytes.

a, Scheme summarizes the developmental trajectories of the previously annotated CD34+ postnatal thymocytes for T- and DC-lineages. b,c, Scheme illustrates the development of murine thymocytes during T lineage specification and commitment (top), and their identification by flow cytometric gating (bottom) (b). Expression of IRF8 and GATA3 (n = 2) for the thymocyte subsets (top) shown in (b) and for the lin and lin+ fractions of thymocytes (bottom) (c). d, Four cellular subsets of human immature thymocytes (n = 2) that express different levels of IRF8 and CD1a were overlaid in order to show their expression for PU.1 and CD123. e-g, Flow cytometry gating of ex vivo linCD4CD34+ thymocytes (n = 9) based on their expression levels of CD123 and CD1a (e). Frequencies of the cellular subsets (n = 9) identified in (e) (f). These subsets were sorted (top) and their expression for CD123, CD34 and IRF8 were subsequently examined by flow cytometry (bottom) (n = 2) (g). Data are representatives of one (b-d), nine (e) and two (g) independent experiments. Data are presented as mean±s.d. of nine (f) independent experiments. DN, double negative; n, biological replicates (f).

Source data

Extended Data Fig. 2 Impact of low dose IRF8 activity on early human T cell development.

a, Scheme illustrates the downstream independent analyses of empty vector (control)- or IRF8-ERT2-transduced HSPCs that were cocultured with OP9-DLL4 stromal cells. b, Flow cytometric staining of IRF8 expression in empty vector (control)- and IRF8-ERT2-transduced cord blood HSPCs at day 2 post transduction. The transduced cells were not treated with 4-OHT. c,d, Volcano plots depict genes that were differentially expressed in 0 nM (c) and 50 nM (d) of 4-OHT treated IRF8-ERT2-expressing cells compared to the untreated control. Genes with a FDR of <0.05 and a log2 fold change of differential expression of >1 or < -1 were considered significant. e, RNA-seq revealed changes in expression of Notch target genes in empty vector (control)- and IRF8-ERT2 (treated with 50 nM 4-OHT)-expressing CD7+ cells from matched donors (n = 3). The normalized counts are CPM. f, The enrichment scores of IFN-related gene signature for the previously annotated populations of human CD34+ thymocytes. g,h, Transcription factor motif analyses for the chromatin regions that are significantly and uniquely opened (g) or closed (h) in IRF8-ERT2-expressing CD7+ cells that were treated with 50 nM of 4-OHT. i,j, Flow cytometric analysis to examine the expression of tmTNF and TNFR1 during in vitro human early T cell development (i) and their expression patterns (n = 3) were quantified proportionally (j). Data are representatives of two (b) and one (i) independent experiments. Data are presented as mean±s.d. of one experiment (j). Motif enrichment was determined by HOMER algorithm93 that uses ZOOPS scoring coupled with the hypergeometric or binomial enrichment calculations (g,h). Data are analyzed by two-way ANOVA with Šídák’s multiple comparisons test (j). FDR, false discovery rate; CPM, counts per million; TF, transcription factor; n, biological replicates (e,j); exact P values are provided in the Source data.

Source data

Extended Data Fig. 3 Impact of IRF8 deficiency on early human hematopoiesis and T cell development.

a,b, Flow cytometric identification of CD326CD56+ EMPs that were generated from IRF8+/+ and IRF8-/- induced pluripotent stem cells (a), and their frequency (n = 6 per genotype) (b). c, Flow cytometric analysis to determine the impact of IRF8 loss on the generation of TNFR2+CD123+CD127+ hematopoietic progenitors. d-h, Scheme illustrates the collection of conditioned medium from EMO cultures of different conditions at the indicated time points (d). sTNF was measured using flow cytometric bead-based immunoassay in which a standard curve was generated (e). Concentration of sTNF detected in the conditioned medium (n = 6 per condition) (f). The fraction above the bars denotes the actual n from each condition that had detectable sTNF. Flow cytometric analysis of EMOs (n = 4 per condition), cultured with fresh or conditioned medium from day 7 onwards, to identify TNFR2+CD123+CD127+ hematopoietic progenitors (g) and their frequency (h). i,j, Frequency (i) and cell count (j) of CD7+CD5+ T cell precursors (n = 3) that were generated from tmTNF-expressing CD123+CD127+HLA-A2+ progenitors in the ATO-spiking experiment. k,l, Frequency (k) and cell count (l) of CD7+CD5+ T cell precursors (n = 3) that were generated from HLA-A2 HSPCs in the ATO-spiking experiment. Data are representatives of six (a), three (c) and two (g) independent experiments. Data are presented as mean±s.d. of six (b), two (h) and one (i-l) independent experiments. Data are presented as mean of two independent experiments (f: WT). Data are analyzed by two-tailed paired t test (b,k) and one-way ANOVA with Dunnett’s multiple comparisons test (h). WT, wild type; KO, knockout; MFI, median fluorescent intensity; n, EMP induction (b), pool of 2 EMOs (f,h), pool of 2 ATOs (i,j) and biological replicates (k-l); exact P values are provided in the Source data.

Source data

Extended Data Fig. 4 Immunophenotypic analysis of TNF-signaling components in hematopoietic precursors and during early T cell development.

a, Flow cytometric dot plots showing expression of TNFR2 and tmTNF (n = 4) on the 5 cellular subsets of ex vivo human immature thymocytes as shown in (Fig. 5a, b). b, ATOs were assembled using human HSPCs derived from fetal liver (CD34hiCD45+; n = 2), cord blood (linCD34+CD38; n = 2) or adult buffy coats (linCD34+; n = 4), and harvested for flow cytometry analyses at day 2, 4, 7 and 10 post culture. For each ontogenetic stage, expression of TNFR1 across different time points is compared to the FMO control and is shown in representative offsetting histograms. c,d, Flow cytometric analysis illustrates the TNFR2 expression profiles of human HSPCs from fetal liver, cord blood and adult buffy coat (n = 2 donors for each) (c). Median fluorescent intensity of TNFR2 for HSPCs from different sources (d). ATOs from each ontogenetic stage were assembled and analyzed independently (b). Data are representatives (a,c) and presented as mean (d) of one experiment; n, biological replicates (d).

Source data

Extended Data Fig. 5 Generation of a MS5 cell line that co-expresses human DLL4 and tmTNF (MS5-DLL4/tmTNF).

a, 6 weeks after sorting, based on co-expression of GFP (reporter for DLL4-encoding construct) and BFP (reporter for TNF-encoding construct), expression of human tmTNF and DLL4 on the transduced MS5 cells was determined by flow cytometry. The MS5 parental cell line served as a negative control. Previously generated MS5-DLL4 cell line served as a positive control for DLL4 expression. b, Expression of reporters, DLL4 and tmTNF on MS5-DLL4/tmTNF cells over time after sorting. Data are representative of one experiment.

Source data

Extended Data Fig. 6 Impact of specific TNFR2 activation on the generation of early T cell precursors.

a,b, Frequency (a) and cell count (b) of CD7+CD5+ T cell precursors that were generated from adult buffy coat linCD34+ HSPCs (n = 7) at day 10, without (control) and with TNFR2 activation (EHD2-scTNFR2 and tmTNF at 1% density), in the ATO system. Data are analyzed by two-tailed paired t test (a). n, biological replicates (a,b); exact P values are provided in the Source data.

Source data

Extended Data Fig. 7 sTNF-mediated TNFR1-specific signaling is dose-dependent and accelerates differentiation of human HSPCs into T cell precursors at the expense of their maintenance.

a, Scheme of assembly for the ATOs that were treated without (control) or with sTNF for 10 days. b, Absolute counts of CD45+ cells harvested from an ATO that was aggregated with a normalized amount of 7,500 cord blood-derived linCD34+CD38 HSPCs (n = 10 donors for all conditions except n = 3 for treatment with 0.25 ng/mL of sTNF). c-e, Flow cytometric analysis to identify CD7+CD5+ T cell precursors that were generated from the ATOs (c), of which some remained undifferentiated and expressed CD34 (d). Quantification of the impact of sTNF treatment on the cell counts of T cell precursors and undifferentiated CD34+ HSPCs compared to the control (n = 10 for all conditions except n = 3 for treatment with 0.25 ng/mL of sTNF) (e). f,g, Flow cytometric analysis to determine HLA-DR expression on CD1a-expressing T-lineage committed precursors (f) and quantification of the cellular fractions (n = 10 for all conditions except n = 3 for treatment with 0.25 ng/mL of sTNF) that were positive (g). Data are presented as mean±s.d. (b,e,g) and representatives (c,d,f) of three independent experiments. Data are analyzed by two-tailed mixed-effects analysis with Dunnett’s post-hoc test (b,g) and two-tailed mixed-effects analysis with linear trend test (e). n, biological replicates (b,e,g); exact P values are provided in the Source data.

Source data

Extended Data Fig. 8 Differential impacts of sTNF and tmTNF on early T cell precursor generation.

a, Overlays of the UMAP for cells derived from specific (red: control, tmTNF or sTNF) and all conditions (gray). b, UMAP visualization of the cell cycle status for all 26 cellular clusters shown in Fig. 7a. c, The enrichment scores of IFN-related gene signature for all the annotated populations of hematopoietic cells shown in Fig. 7b. d, The relative distribution of cells, derived from ATOs without and with TNF stimulus, in cluster 4 and 15 that are annotated as lymphoid progenitors. e, Heatmap shows the average expression of IFN-related genes by the previously annotated populations of human CD34+ thymocytes. f-l, Scheme illustrates the experimental design where CD7+ T-specified progenitors, derived from control or TNF activated (sTNF at 0.25 ng/mL or tmTNF at 1% density)-ATOs assembled with cord blood linCD34+ HSPCs, were examined for their maturation potential towards later stages of T-lineage development (f). Frequency (g) and cell count (h) of CD4+CD8b+ thymocytes (n = 5) that were generated from the secondary ATO cultures. Frequency (i) and cell count (j) of CD3+TCRαβ+ T cells (n = 5) that were generated from the secondary ATO cultures. Frequency (k) and cell count (l) of CD3+TCRγδ+ T cells (n = 5) that were generated from the secondary ATO cultures. n, biological replicates (g-l).

Source data

Supplementary information

Supplementary Information

Supplementary Figs. 1–6.

Reporting Summary

Peer Review File

Supplementary Tables

Supplementary Tables 1–3.

Source data

Source Data Fig. 1

Graphic/statistical source data and exact P values.

Source Data Fig. 2

Graphic/statistical source data and exact P values.

Source Data Fig. 3

Graphic/statistical source data and exact P values.

Source Data Fig. 4

Graphic/statistical source data and exact P values.

Source Data Fig. 5

Graphic/statistical source data and exact P values.

Source Data Fig. 6

Graphic/statistical source data and exact P values.

Source Data Fig. 7

Graphic/statistical source data and exact P values.

Source Data Extended Data Fig. 1

Graphic/statistical source data and exact P values.

Source Data Extended Data Fig. 2

Graphic/statistical source data and exact P values.

Source Data Extended Data Fig. 3

Graphic/statistical source data and exact P values.

Source Data Extended Data Fig. 4

Graphic/statistical source data.

Source Data Extended Data Fig. 5

Graphic/statistical source data.

Source Data Extended Data Fig. 6

Graphic/statistical source data and exact P values.

Source Data Extended Data Fig. 7

Graphic/statistical source data and exact P values.

Source Data Extended Data Fig. 8

Graphic/statistical source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liang, K.L., Roels, J., Lavaert, M. et al. Intrathymic dendritic cell-biased precursors promote human T cell lineage specification through IRF8-driven transmembrane TNF. Nat Immunol 24, 474–486 (2023). https://doi.org/10.1038/s41590-022-01417-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-022-01417-6

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