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:

TCF1–LEF1 co-expression identifies a multipotent progenitor cell (TH2-MPP) across human allergic diseases

Abstract

Repetitive exposure to antigen in chronic infection and cancer drives T cell exhaustion, limiting adaptive immunity. In contrast, aberrant, sustained T cell responses can persist over decades in human allergic disease. To understand these divergent outcomes, we employed bioinformatic, immunophenotyping and functional approaches with human diseased tissues, identifying an abundant population of type 2 helper T (TH2) cells with co-expression of TCF7 and LEF1, and features of chronic activation. These cells, which we termed TH2-multipotent progenitors (TH2-MPP) could self-renew and differentiate into cytokine-producing effector cells, regulatory T (Treg) cells and follicular helper T (TFH) cells. Single-cell T-cell-receptor lineage tracing confirmed lineage relationships between TH2-MPP, TH2 effectors, Treg cells and TFH cells. TH2-MPP persisted despite in vivo IL-4 receptor blockade, while thymic stromal lymphopoietin (TSLP) drove selective expansion of progenitor cells and rendered them insensitive to glucocorticoid-induced apoptosis in vitro. Together, our data identify TH2-MPP as an aberrant T cell population with the potential to sustain type 2 inflammation and support the paradigm that chronic T cell responses can be coordinated over time by progenitor cells.

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: Single-cell atlas of GATA3+ lymphocytes across human allergic diseases.
Fig. 2: Aberrant maintenance of TCF1 and LEF1 expression by TH2 cells infiltrating human nasal polyps.
Fig. 3: Pseudobulk analysis identifies transcriptional signatures of TH2-MPP and circulating blood memory cells.
Fig. 4: Metabolic heterogeneity of type 2 lymphocytes.
Fig. 5: Single TCR clones exhibit clonal expansion and cross-cluster overlap along the TH2-MPP differentiation trajectory.
Fig. 6: TH2-MPP differentiation modeled in vitro.
Fig. 7: TH2-MPP cells couple self-renewal with effector differentiation to maintain the TH2 lineage.

Similar content being viewed by others

Data availability

The newly generated scRNA-seq data have been uploaded to the GEO repository (GSE255544)80. Other data sets used in this study include: asthma (GSE164015 and GSE193816), atopic dermatitis (GSE153760 and GSE158432), allergic conjunctivitis (GSE203217), bullous pemphigoid (https://zenodo.org/deposit/5228495), CRSwNP (GSE179292), EoE (GSE175930), lichen planus (https://zenodo.org/deposit/5228495), lymphedema (GSA HRA000901) and UC (Broad Single Cell Portal, SCP259).

Code availability

All analysis and figures were generated using publicly available software packages. No custom code was used in this study.

References

  1. Paley, M. A. et al. Progenitor and terminal subsets of CD8+ T cells cooperate to contain chronic viral infection. Science 338, 1220–1225 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Im, S. J. et al. Defining CD8+ T cells that provide the proliferative burst after PD-1 therapy. Nature 537, 417–421 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Utzschneider, D. T. et al. T cell factor 1-expressing memory-like CD8+ T cells sustain the immune response to chronic viral infections. Immunity 45, 415–427 (2016).

    Article  CAS  PubMed  Google Scholar 

  4. Wu, T. et al. The TCF1–Bcl6 axis counteracts type I interferon to repress exhaustion and maintain T cell stemness. Sci. Immunol. 1, eaai8593 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  5. Sade-Feldman, M. et al. Defining T cell states associated with response to checkpoint immunotherapy in melanoma. Cell 175, 998–1013 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Philip, M. & Schietinger, A. CD8+ T cell differentiation and dysfunction in cancer. Nat. Rev. Immunol. 22, 209–223 (2022).

    Article  CAS  PubMed  Google Scholar 

  7. Walker, J. A. & McKenzie, A. N. J. TH2 cell development and function. Nat. Rev. Immunol. 18, 121–133 (2018).

    Article  CAS  PubMed  Google Scholar 

  8. Nakayama, T. et al. TH2 cells in health and disease. Annu. Rev. Immunol. 35, 53–84 (2016).

    Article  PubMed  Google Scholar 

  9. Gause, W. C., Rothlin, C. & Loke, P. Heterogeneity in the initiation, development and function of type 2 immunity. Nat. Rev. Immunol. 20, 603–614 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Nelson, R. W. et al. T cell receptor cross-reactivity between similar foreign and self peptides influences naive cell population size and autoimmunity. Immunity 42, 95–107 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Bangert, C. et al. Persistence of mature dendritic cells, TH2A, and Tc2 cells characterize clinically resolved atopic dermatitis under IL-4Rα blockade. Sci. Immunol. 6, eabe2749 (2021).

    Article  CAS  PubMed  Google Scholar 

  12. Smillie, C. S. et al. Intra- and inter-cellular rewiring of the human colon during ulcerative colitis. Cell 178, 714–730 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Ma, J. et al. Single-cell analysis pinpoints distinct populations of cytotoxic CD4+ T cells and an IL-10+CD109+ TH2 cell population in nasal polyps. Sci. Immunol. 6, eabg6356 (2021).

    Article  CAS  PubMed  Google Scholar 

  14. Liu, X. et al. Single-cell RNA sequencing of subcutaneous adipose tissues identifies therapeutic targets for cancer-associated lymphedema. Cell Discov. 8, 58 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Wen, T. et al. Single-cell RNA sequencing identifies inflammatory tissue T cells in eosinophilic esophagitis. J. Clin. Invest. 129, 2014–2028 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Morgan, D. M. et al. Clonally expanded, GPR15-expressing pathogenic effector TH2 cells are associated with eosinophilic esophagitis. Sci. Immunol. 6, eabi5586 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Alladina, J. et al. A human model of asthma exacerbation reveals transcriptional programs and cell circuits specific to allergic asthma. Sci. Immunol. 8, eabq6352 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Siddiqui, S. et al. Epithelial miR-141 regulates IL-13-induced airway mucus production. JCI Insight 6, e139019 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  20. Okano, M. et al. Interleukin-33-activated neuropeptide CGRP-producing memory TH2 cells cooperate with somatosensory neurons to induce conjunctival itch. Immunity 55, 2352–2368 (2022).

    Article  CAS  PubMed  Google Scholar 

  21. Liu, Y. et al. Classification of human chronic inflammatory skin disease based on single-cell immune profiling. Sci. Immunol. 7, eabl9165 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Mitson-Salazar, A. et al. Hematopoietic prostaglandin D synthase defines a proeosinophilic pathogenic effector human TH2 cell subpopulation with enhanced function. J. Allergy Clin. Immunol. 137, 907–918 (2016).

    Article  CAS  PubMed  Google Scholar 

  23. Wambre, E. et al. A phenotypically and functionally distinct human TH2 cell subpopulation is associated with allergic disorders. Sci. Transl. Med. 9, eaam9171 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  24. Faustino, L. D. et al. Interleukin-33 activates regulatory T cells to suppress innate γδ T cell responses in the lung. Nat. Immunol. 21, 1371–1383 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Morimoto, Y. et al. Amphiregulin-producing pathogenic memory T helper 2 cells instruct eosinophils to secrete osteopontin and facilitate airway fibrosis. Immunity 49, 134–150 (2018). .

  26. Zhao, X., Shan, Q. & Xue, H.-H. TCF1 in T cell immunity: a broadened frontier. Nat. Rev. Immunol. 22, 147–157 (2022).

    Article  CAS  PubMed  Google Scholar 

  27. Kumar, B. V. et al. Human tissue-resident memory T cells are defined by core transcriptional and functional signatures in lymphoid and mucosal sites. Cell Rep. 20, 2921–2934 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Skon, C. N. et al. Transcriptional downregulation of S1pr1 is required for the establishment of resident memory CD8+ T cells. Nat. Immunol. 14, 1285–1293 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Hombrink, P. et al. Programs for the persistence, vigilance and control of human CD8+ lung-resident memory T cells. Nat. Immunol. 17, 1467–1478 (2016).

    Article  CAS  PubMed  Google Scholar 

  30. Schnell, A. et al. Stem-like intestinal TH17 cells give rise to pathogenic effector T cells during autoimmunity. Cell 184, 6281–6298 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Karmaus, P. W. F. et al. Metabolic heterogeneity underlies reciprocal fates of TH17 cell stemness and plasticity. Nature 565, 101–105 (2019).

    Article  CAS  PubMed  Google Scholar 

  32. Gearty, S. V. et al. An autoimmune stem-like CD8 T cell population drives type 1 diabetes. Nature 602, 156–161 (2022).

    Article  CAS  PubMed  Google Scholar 

  33. Wang, W. et al. Single-cell profiling identifies mechanisms of inflammatory heterogeneity in chronic rhinosinusitis. Nat. Immunol. 23, 1484–1494 (2022).

    Article  CAS  PubMed  Google Scholar 

  34. Utzschneider, D. T. et al. Active maintenance of T cell memory in acute and chronic viral infection depends on continuous expression of FOXO1. Cell Rep. 22, 3454–3467 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Delpoux, A. et al. Continuous activity of Foxo1 is required to prevent anergy and maintain the memory state of CD8+ T cells. J. Exp. Med. 215, 575–594 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Staron, M. M. et al. The transcription factor FoxO1 sustains expression of the inhibitory receptor PD-1 and survival of antiviral CD8+ T cells during chronic infection. Immunity 41, 802–814 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Tibbitt, C. A. et al. Single-cell RNA sequencing of the T helper cell response to house dust mites defines a distinct gene expression signature in airway TH2 cells. Immunity 51, 169–184 (2018).

    Article  Google Scholar 

  38. Ulrich, B. J. et al. Allergic airway recall responses require IL-9 from resident memory CD4+ T cells. Sci. Immunol. 7, eabg9296 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Brown, I. K. et al. Single cell analysis of host response to helminth infection reveals the clonal breadth, heterogeneity, and tissue-specific programming of the responding CD4+ T cell repertoire. PLoS Pathog. 17, e1009602 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Radtke, D. et al. TH2 single-cell heterogeneity and clonal distribution at distant sites in helminth-infected mice. eLife 11, e74183 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Hara-Chikuma, M. et al. Chemokine-dependent T cell migration requires aquaporin-3–mediated hydrogen peroxide uptake. J. Exp. Med. 209, 1743–1752 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Kurtulus, S. et al. TIGIT predominantly regulates the immune response via regulatory T cells. J. Clin. Invest. 125, 4053–4062 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Mata, S. H.-D. L. et al. Cytotoxic CD4+ tissue-resident memory T cells are associated with asthma severity. Med 4, 875–897 (2023).

    Article  Google Scholar 

  44. Wang, Y.-H. et al. Maintenance and polarization of human TH2 central memory T cells by thymic stromal lymphopoietin-activated dendritic cells. Immunity 24, 827–838 (2006).

    Article  CAS  PubMed  Google Scholar 

  45. Chen, J. et al. NR4A transcription factors limit CAR T cell function in solid tumours. Nature 567, 530–534 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Liu, X. et al. Genome-wide analysis identifies NR4A1 as a key mediator of T cell dysfunction. Nature 567, 525–529 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Lohoff, M. et al. Early growth response protein-1 (Egr-1) Is preferentially expressed in T helper type 2 (TH2) cells and is involved in acute transcription of the TH2 cytokine interleukin-4*. J. Biol. Chem. 285, 1643–1652 (2010).

    Article  CAS  PubMed  Google Scholar 

  48. Wagle, M. V. et al. Antigen-driven EGR2 expression is required for exhausted CD8+ T cell stability and maintenance. Nat. Commun. 12, 2782 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Martinez, G. J. et al. The transcription factor NFAT promotes exhaustion of activated CD8+ T cells. Immunity 42, 265–278 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Geltink, R. I. K., Kyle, R. L. & Pearce, E. L. Unraveling the complex interplay between T cell metabolism and function. Annu. Rev. Immunol. 36, 461–488 (2018).

    Article  CAS  PubMed  Google Scholar 

  51. Chapman, N. M. & Chi, H. Metabolic adaptation of lymphocytes in immunity and disease. Immunity 55, 14–30 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Chen, Y.-H. et al. Asymmetric PI3K activity in lymphocytes organized by a PI3K-mediated polarity pathway. Cell Rep. 22, 860–868 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Cosset, É. et al. Glut3 addiction is a druggable vulnerability for a molecularly defined subpopulation of glioblastoma. Cancer Cell 32, 856–868 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Peng, M. et al. Aerobic glycolysis promotes T helper 1 cell differentiation through an epigenetic mechanism. Science 354, 481–484 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Chang, C.-H. et al. Posttranscriptional control of T cell effector function by aerobic glycolysis. Cell 153, 1239–1251 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Wang, Y. et al. The kinase complex mTORC2 promotes the longevity of virus-specific memory CD4+ T cells by preventing ferroptosis. Nat. Immunol. 23, 303–317 (2022).

    Article  PubMed  Google Scholar 

  57. Voss, K. et al. Elevated transferrin receptor impairs T cell metabolism and function in systemic lupus erythematosus. Sci. Immunol. 8, eabq0178 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Argüello, R. J. et al. SCENITH: a flow cytometry-based method to functionally profile energy metabolism with single-cell resolution. Cell Metab. 32, 1063–1075 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  59. Lopes, N. et al. Distinct metabolic programs established in the thymus control effector functions of γδ T cell subsets in tumor microenvironments. Nat. Immunol. 22, 179–192 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Riggan, L. et al. The transcription factor Fli1 restricts the formation of memory precursor NK cells during viral infection. Nat. Immunol. 23, 556–567 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Johnson, M. O. et al. Distinct regulation of TH17 and TH1 cell differentiation by glutaminase-dependent metabolism. Cell 175, 1780–1795 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Healey, D. C. C. et al. Targeting in vivo metabolic vulnerabilities of TH2 and TH17 cells reduces airway inflammation. J. Immunol. 206, 1127–1139 (2021).

    Article  CAS  PubMed  Google Scholar 

  63. Reynolds, G. et al. Developmental cell programs are co-opted in inflammatory skin disease. Science 371, eaba6500 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Alkon, N. et al. Single-cell RNA sequencing defines disease-specific differences between chronic nodular prurigo and atopic dermatitis. J. Allergy Clin. Immunol. 152, 420–435 (2023).

    Article  CAS  PubMed  Google Scholar 

  65. Upadhyaya, B., Yin, Y., Hill, B. J., Douek, D. C. & Prussin, C. Hierarchical IL-5 expression defines a subpopulation of highly differentiated human TH2 cells. J. Immunol. 187, 3111–3120 (2011).

    Article  CAS  PubMed  Google Scholar 

  66. Bachert, C. et al. Efficacy and safety of dupilumab in patients with severe chronic rhinosinusitis with nasal polyps (LIBERTY NP SINUS-24 and LIBERTY NP SINUS-52): results from two multicentre, randomised, double-blind, placebo-controlled, parallel-group phase 3 trials. Lancet 394, 1638–1650 (2019).

    Article  CAS  PubMed  Google Scholar 

  67. Simpson, E. L. et al. Two phase 3 trials of dupilumab versus placebo in atopic dermatitis. N. Engl. J. Med. 375, 2335–2348 (2016).

    Article  CAS  PubMed  Google Scholar 

  68. Castro, M. et al. Dupilumab efficacy and safety in moderate-to-severe uncontrolled asthma. N. Engl. J. Med. 378, 2486–2496 (2018).

    Article  CAS  PubMed  Google Scholar 

  69. Ebina-Shibuya, R. & Leonard, W. J. Role of thymic stromal lymphopoietin in allergy and beyond. Nat. Rev. Immunol. 23, 24–37 (2023).

    Article  CAS  PubMed  Google Scholar 

  70. Taves, M. D. & Ashwell, J. D. Glucocorticoids in T cell development, differentiation and function. Nat. Rev. Immunol. 21, 233–243 (2021).

    Article  CAS  PubMed  Google Scholar 

  71. Kabata, H. et al. Thymic stromal lymphopoietin induces corticosteroid resistance in natural helper cells during airway inflammation. Nat. Commun. 4, 2675 (2013).

    Article  PubMed  Google Scholar 

  72. Gowthaman, U. et al. Identification of a T follicular helper cell subset that drives anaphylactic IgE. Science 365, eaaw6433 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  73. Rivas, M. N. & Chatila, T. A. Regulatory T cells in allergic diseases. J. Allergy Clin. Immunol. 138, 639–652 (2016).

    Article  PubMed Central  Google Scholar 

  74. Worm, M. et al. Efficacy and safety of multiple dupilumab dose regimens after initial successful treatment in patients with atopic dermatitis. JAMA Dermatol. 156, 131–143 (2020).

    Article  PubMed  Google Scholar 

  75. Blank, C. U. et al. Defining ‘T cell exhaustion’. Nat. Rev. Immunol. 19, 665–674 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  76. Borcherding, N., Bormann, N. L. & Kraus, G. scRepertoire: an R-based toolkit for single-cell immune receptor analysis. F1000Res. 9, 47 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  77. Street, K. et al. Slingshot: cell lineage and pseudotime inference for single-cell transcriptomics. BMC Genomics 19, 477 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  78. Marsh, S., Salmon, M. & Hoffman, P. scCustomize: custom visualizations & functions for streamlined analyses of single cell sequencing (The Comprehensive R Archive Network, 2024).

  79. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Kratchmarov, R. & Brennan, P. Single cell RNA sequencing and single cell TCR sequencing of type 2 lymphocytes in human nasal polyps. GEO https://www.ncbi.nlm.nih.gov/geo/query/acc.cgi?acc=GSE255544 (2024).

Download references

Acknowledgements

This work was supported by National Institutes of Health grants U19AI095219, T32AI007306 and K23AI139352, the Lupus Research Alliance, the Food Allergy Science Initiative (FASI) at the Broad Institute, the Arthritis National Science Foundation Vic Braden Family Fellowship and generous support from the Vinik and Karol Families. We are grateful to the people who provided the biological samples used in this study. We thank M. Brenner (Brigham & Women’s Hospital) for providing the DN32 cell line. We thank A. Chicoine, J. Case and the Brigham and Women’s Hospital Center for Cellular Profiling Flow Cytometry Core for assistance with cell sorting, as well as G. Watts, Z. Zhu and the BWH Center for Cellular Profiling Single Cell Genomics Core, and the Dana–Farber Cancer Institute Center for Cancer Genomics Services Core for assistance with single-cell transcriptomics. We appreciate insightful comments on the paper provided by D. Schaefer-Babajew and M. ElTanbouly, Rockefeller University, and thank G. Manoim for assistance with streamlining import of public sequencing data sets.

Author information

Authors and Affiliations

Authors

Contributions

R.K. performed the majority of experiments and analyzed data. R.K. and P.J.B. designed the project, supervised experiments and analysis and wrote the paper. S.D., G.D., D.F.D., D.A.R. and M.G.-A. analyzed computational data. X.J., W.H, T.R. and A.M. performed experiments and analyzed data. C.M.B., J.R.A., R.P.K., B.J.M., E.M.T., S.A., N.B., R.W.B., A.Z.M., S.L., R.R., W.G.S., T.M.L., K.M.B. and J.A.B. managed and contributed critical resources and aided in experimental design. All authors edited the paper.

Corresponding author

Correspondence to Patrick J. Brennan.

Ethics declarations

Competing interests

S.L. reports clinical trial funding from Sanofi Aventis Regeneron, GlaxoSmithKline, AstraZeneca and OptiNose and participated on advisory boards for GlaxoSmithKline, AstraZeneca, Sanofi Aventis, Regeneron, Genentech and Novartis. R.W.B. reports unrelated clinical trial funding from I-Mab Biopharma (CTA: HLW-ALZ-NAS-001). N.B. reports consultant fees for unrelated epidemiology research for GlaxoSmithKline. D.A.R. reports consultant fees for unrelated work from GlaxoSmithKline, AstraZeneca, Pfizer, HiFiBio Therapeutics, Scipher Medicine and Bristol-Myers Squibb and grant support unrelated to this work from Janssen, Merck and Bristol-Myers Squibb. D.F.D. reports unrelated consultant fees from Celldex Therapeutics. The other authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editor: L. A. Dempsey, 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 Transcriptomic characterization of key GATA3-expressing lymphocytes.

a, Violin plots showing expression level of key genes used to annotate UMAP clusters. b, Volcano plot of differentially expressed genes between peTh2/Th2A and ILC2 (left) and between γδ T cell clusters (right) from pseudobulk analysis and linear mixed model. Highlighted transcripts have absolute β > 1 and adjusted p-value <0.05 by False Discovery Rate (FDR). c, Violin plots showing expression level of key TRM-associated transcripts. d, Volcano plot of differentially expressed genes between TFH and Memory-like (left) and peTh2/Th2A and Memory-like clusters (right). Highlighted transcripts have absolute β > 1 and adjusted p-value <0.05 by FDR from pseudobulk analysis and linear mixed model.

Extended Data Fig. 2 Gene Set Enrichment Analysis of GATA3-expressing αβ T cell clusters.

Pseudobulk analysis was performed to identify marker transcripts differentiating each cluster from the integrated scRNAseq atlas dataset using a linear mixed model and controlling for dataset as random effect. Gene set enrichment analysis was then performed for each cluster based on biological process annotations. The top 10 pathways based on p-value and GeneRatio (count of genes in the core enrichment / total count of genes in this pathway) with a positive enrichment score and an adjusted p < 0.05 are displayed.

Extended Data Fig. 3 Pseudotime differentiation trajectories of memory-like cluster.

a, UMAP of αβ T cells from the tissue atlas (Fig. 1) after re-clustering. b, Violin plots of key transcripts used to annotate clusters. c, Left: Trajectories inferred by Slingshot algorithm superimposed on UMAP. Right: Three distinct lineages with cells from each lineage colored by pseudotime.

Extended Data Fig. 4 Transcription factor, cytokine, and surface marker expression profiles of GATA3+ T cell subsets in nasal polyps.

a, Quantification of GATA3+ T cells in healthy nasal mucosa, chronic rhinosinusitis without nasal polyps (CRSsNP), and chronic rhinosinusitis with nasal polyps (CRSwNP). For healthy mucosa and CRSsNP, N = 7 donors, for CRSwNP N = 14 donors. b, Identification of Th2 by CRTh2, GATA3, IL-4, IL-5, and IL-13 expression. c, Representative staining and quantification of IL-4, IFN-γ, IL-17, and CD38 for identification of T helper subsets. N = 15. d, Quantification of TCF1+/LEF1+ cell frequency in T helper subsets from indicated diseases. For EoE, N = 10 donors, for UC N = 6 donors, for Lymphedema, N = 3 donors. e, Left: Representative histogram of FOXO1 expression by CD8+ T cells (grey), GATA3-CD4+ T cells (blue), and GATA3+CD4+ T cells (black) in nasal polyps. Right: quantification of FOXO1 MFI for indicated subsets. N = 6 donors. f, Correlation of blood absolute eosinophil count (left, N = 15 donors), total serum IgE (middle, N = 10 donors), and nasal eosinophil cationic protein (ECP) (right, N = 21 donors) with TCF1/LEF1 co-expression by nasal polyp Th2 cells. p < 0.05. Pearson correlation coefficient. For all plots, ****p < 0.0001. ***p < 0.001. **p < 0.01. *p < 0.05. One-way ANOVA with Holm-Sidak correction for multiple comparisons. For all panels with box and whisker plots, box range denotes 25th to 75th percentiles, whiskers show minimum and maximum values.

Extended Data Fig. 5 Expression of pathogenic effector markers, inhibitory receptors associated with exhaustion, and transcription factors by Th2 in nasal polyps.

a, TRM marker expression by nasal polyp T cells. Left: Representative plots of CD69 and CD103 expression by total CD4+ and CD8+ T cells. Middle: Quantification of indicated subsets by CD69 and CD103 expression. Right: Frequencies of CD103 expression by indicated subsets of GATA3+CD4+ T cells. N = 12 donors. b, Expression of surface activation and inhibitory receptors by GATA3+CD4+ T cell subsets. CD69: N = 15 donors, CD38: N = 6 donors, PD-1: N = 6 donors, CD39: N = 8 donors, TIGIT: N = 6 donors. c, Expression of peTh2/Th2A markers by GATA3+CD4+ T cell subsets. CD200R: N = 10 donors, CD161 N = 9 donors, CD49d: N = 7 donors. d, Expression of TCF1 and LEF1 by GATA3+CD4+ T cell subsets. TCF1: N = 8 donors. LEF1: N = 11 donors. All panels: ****p < 0.0001. ***p < 0.001. **p < 0.01. *p < 0.05. n.s. not significant. One-way ANOVA with Tukey’s correction for multiple comparisons. For all panels with box and whisker plots, box range denotes 25th to 75th percentiles, whiskers show minimum and maximum values. For all panels, red line = FOXP3+, black line = CRTh2+, and blue line = CRTh2- subsets respectively.

Extended Data Fig. 6 scRNAseq identifies distinct T cell clusters in human nasal polyps and paired peripheral blood.

a, Representative gating strategy for sorting indicated subsets for scRNAseq. P1: ILCs (CD45+CD127+ lin-), P2: CD8+ T cells, P3: CRTh2+ Th2, P4: CCR4-CRTh2-CD4+, P5: CCR4+CXCR3- Th2. b, Expression of GATA3 and CCR4 by CD4+ T cells in nasal polyps. Plot is representative of 3 independent tissue donors. c, UMAP of nasal polyp lymphocytes split by donor. d, Heatmap of top 20 differentially expressed genes for each cluster with >0.5 log2 fold change and padjusted <0.05 cutoffs, Right column: key genes used to annotate clusters highlighted. e, Violin plots of expression level of indicated genes used to annotate UMAP clusters. f, Violin plots of key subset-defining surface markers detected by DNA-barcoded antibody labeling. g, Violin plot of SLC2A3, SLC16A3, ACSL4, and FFAR3 transcript expression by nasal polyp lymphocytes.

Extended Data Fig. 7 Iterative expansion index analysis for nasal polyp T cell clusters.

Left: Iterative pseudotime lineage tracing for each specified origin node. Right: Average expansion indices +/− SEM for clones with overlap between each specified root node and the specified lineages. Positive values indicate clonal expansion in the putative effector cluster relative to root node; negative values indicate clonal expansion in the root cluster. Size of dots are scaled relative to the number of clones in that lineage. ** p < 0.01. n.s. not significant. One sample, two tailed Wilcoxon test.

Extended Data Fig. 8 scTCRseq analysis of lymphocytes in atopic dermatitis.

a, UMAP of integrated TCR-expressing T cells in atopic dermatitis. N = 5,622 clonotyped cells. b, Dot Plot of lineage-defining transcripts used to identify clusters. c, Left: FeaturePlot of GATA3 expression. Right: Joint Density function plot of indicated transcripts used to identify putative progenitor and effector clusters. d, Circos plot of clonal overlap between each cluster. Each overlapping clone is connected by a line, with heavier line weights indicative of more overlapping clones. e, Upper Left: Pseudotime lineages with Th2-MPP cluster as starting node projected onto UMAP space. Right: UMAP space split by lineage, with cells from each lineage colored by pseudotime. Lower left: Average expansion indices +/− SEM for clones with overlap between Th2-MPP and the specified lineages. Positive values indicate clonal expansion in the putative effector cluster relative to Th2-MPP; negative values indicate clonal expansion in the Th2-MPP cluster. Size of dots are scaled relative to the number of clones in that lineage. ****p < 0.0001. **p < 0.01. n.s. not significant. One sample, two tailed Wilcoxon test. N = 9 donors integrated across datasets.

Extended Data Fig. 9 Iterative expansion index analysis for atopic dermatitis T cell clusters.

Left: Iterative pseudotime lineage tracing for each specified origin node. Right: Average expansion indices +/− SEM for clones with overlap between each specified root node and the specified lineages. Positive values indicate clonal expansion in the putative effector cluster relative to root node; negative values indicate clonal expansion in the root cluster. Size of dots are scaled relative to the number of clones in that lineage. ** p < 0.01. n.s. not significant. One sample, two tailed Wilcoxon test.

Extended Data Fig. 10 Modeling Th2 heterogeneity, differentiation, and persistence.

a, Expression of IFN-γ and IL-13 in Th1 and Th2 cultures at day 7 of differentiation. Results are representative of 4 independent donors. b, Expression of IL-13 and IL-4 at day 7, day 14, and day 21 of culture. c, Expression of CD38 and CD69 versus cell division by Th2 cells at day 4 of differentiation. d, Left: Representative plots of GATA3 and PPARγ expression. Right: Representative histograms of CD49d expression by CD27+CRTh2- (red), CRTh2+CD161- (blue), and CRTh2+CD161+ (black) subsets. e, In vitro sorting strategy: Representative histogram of TCF1 expression by CD27+CRTh2- progenitor cells (black) and CD27- effector Th2 cells in vitro (red). f, Differentiation of TCF1-lo cells requires TCR stimulation. CD27+CRTh2- progenitor cells were sorted, labeled with CTV, and cultured for additional 7 days with (right) or without (left) TCR stimulation. Upper: Expression of TCF1 versus cell division (TCF1-lo cells denoted by gate). Lower: Expression of CD27 versus cell division (CD27-lo cells denoted by gate). g, Upper: Steady state expression of IL-13 in CD27-hi and CD27-lo subsets. Lower: Steady state expression of TCF1 and LEF1 in Th2-MPP. (a-g) Results are representative of 3 independent donors. h, Proliferative capacity of Th2-MPP and peTh2/Th2A cells. Representative plots of GATA3 versus cell division. I, Sorting strategy for isolation of Th2-MPP for Treg and TFH cell fate analysis. j, Left: Expression of TCF1 and LEF1 following Th2-MPP differentiation. Right: Expression of cell fate markers CRTh2, CD161, FOXP3, and PD-1 by TCF1 and LEF1-hi/lo subsets. k, Expression of TSLP and IL-33 by basal epithelial cells from patients with CRSwNP or CRSsNP. **p < 0.01 Unpaired, two tailed T test with Welch’s correction. N = 8 donors. l, Survival of CD27+ Th2 progenitors, sorted as in Fig. 6b, in culture for 10 days with IL-2, IL-33, or TSLP. Representative plots of LEF1 expression versus viability dye. Results are representative of 4 independent donors. m, Left: Representative plots of CRTh2 and TSLPR expression by GATA3+CD4+ T cells in nasal polyps. Right: Quantification of TSLPR expression by indicated GATA3+CD4+ T cell subsets **p < 0.01 *p < 0.05, 2-way ANOVA. N = 7 donors. n, Violin plot of IL1RL1, NR3C1, and FKBP5 expression by nasal polyp lymphocytes. For all panels with box and whisker plots, box range denotes 25th to 75th percentiles, whiskers show minimum and maximum values.

Supplementary information

Supplementary Information

Supplementary Fig. 1.

Reporting Summary

Supplementary Tables

Supplementary Tables 1–11.

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

Kratchmarov, R., Djeddi, S., Dunlap, G. et al. TCF1–LEF1 co-expression identifies a multipotent progenitor cell (TH2-MPP) across human allergic diseases. Nat Immunol (2024). https://doi.org/10.1038/s41590-024-01803-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41590-024-01803-2

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