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:

CD103hi Treg cells constrain lung fibrosis induced by CD103lo tissue-resident pathogenic CD4 T cells

Abstract

Tissue-resident memory T cells (TRM cells) are crucial mediators of adaptive immunity in nonlymphoid tissues. However, the functional heterogeneity and pathogenic roles of CD4+ TRM cells that reside within chronic inflammatory lesions remain unknown. We found that CD69hiCD103lo CD4+ TRM cells produced effector cytokines and promoted the inflammation and fibrotic responses induced by chronic exposure to Aspergillus fumigatus. Simultaneously, immunosuppressive CD69hiCD103hiFoxp3+ CD4+ regulatory T cells were induced and constrained the ability of pathogenic CD103lo TRM cells to cause fibrosis. Thus, lung tissue-resident CD4+ T cells play crucial roles in the pathology of chronic lung inflammation, and CD103 expression defines pathogenic effector and immunosuppressive tissue-resident cell subpopulations in the inflamed lung.

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: Repeated exposure to Aspergillus antigen induced tissue-resident CD4+ T cells in the inflamed lung.
Fig. 2: Tissue-resident CD69hi T cells showed unique profibrotic features.
Fig. 3: CD69hiCD103lo CD4+ T cells were the effector cytokine producers in the inflamed lung.
Fig. 4: Depletion of Foxp3+ CD4+ Treg cells resulted in exacerbation of chronic lung inflammation.
Fig. 5: CD4+ TRM cells were induced by chronic Aspergillus antigen exposure.
Fig. 6: CD44hiCD69hi CD4+ TRM regulomes show fibrotic features.
Fig. 7: CD4+ TRM cells are sufficient to induce lung inflammation accompanied by fibrotic responses.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding author upon reasonable request. The RNA-Seq and ATAC-Seq data reported in this paper are available under Gene Expression Omnibus accession number GSE133399.

Code availability

The code that supports the findings of this study is available from the corresponding author upon reasonable request.

References

  1. Manz, R. A., Hauser, A. E., Hiepe, F. & Radbruch, A. Maintenance of serum antibody levels. Annu. Rev. Immunol. 23, 367–386 (2005).

    CAS  PubMed  Google Scholar 

  2. Pulendran, B. & Ahmed, R. Immunological mechanisms of vaccination. Nat. Immunol. 12, 509–517 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Sallusto, F., Geginat, J. & Lanzavecchia, A. Central memory and effector memory T cell subsets: function, generation, and maintenance. Annu. Rev. Immunol. 22, 745–763 (2004).

    CAS  PubMed  Google Scholar 

  4. Masopust, D. & Soerens, A. G. Tissue-resident T cells and other resident leukocytes. Annu. Rev. Immunol. 37, 521–546 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Mueller, S. N. & Mackay, L. K. Tissue-resident memory T cells: local specialists in immune defence. Nat. Rev. Immunol. 16, 79–89 (2016).

    CAS  PubMed  Google Scholar 

  6. Gebhardt, T. et al. Memory T cells in nonlymphoid tissue that provide enhanced local immunity during infection with herpes simplex virus. Nat. Immunol. 10, 524–530 (2009).

    CAS  PubMed  Google Scholar 

  7. Kimura, M. Y. et al. Crucial role for CD69 in allergic inflammatory responses: CD69-Myl9 system in the pathogenesis of airway inflammation. Immunol. Rev. 278, 87–100 (2017).

    CAS  PubMed  Google Scholar 

  8. Sathaliyawala, T. et al. Distribution and compartmentalization of human circulating and tissue-resident memory T cell subsets. Immunity 38, 187–197 (2013).

    CAS  PubMed  Google Scholar 

  9. Watanabe, R. et al. Human skin is protected by four functionally and phenotypically discrete populations of resident and recirculating memory T cells. Sci. Transl. Med. 7, 279ra39 (2015).

    PubMed  PubMed Central  Google Scholar 

  10. Park, C. O. & Kupper, T. S. The emerging role of resident memory T cells in protective immunity and inflammatory disease. Nat. Med. 21, 688–697 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. O’Shea, J. J. & Paul, W. E. Mechanisms underlying lineage commitment and plasticity of helper CD4+ T cells. Science 327, 1098–1102 (2010).

    PubMed  PubMed Central  Google Scholar 

  12. Sallusto, F., Cassotta, A., Hoces, D., Foglierini, M. & Lanzavecchia, A. Do memory CD4 T cells keep their cell-type programming: plasticity versus fate commitment? T-cell heterogeneity, plasticity, and selection in humans. Cold Spring Harb. Perspect. Biol. 10, a029421 (2018).

    PubMed  PubMed Central  Google Scholar 

  13. Nakayama, T. et al. TH2 cells in health and disease. Annu. Rev. Immunol. 35, 53–84 (2017).

    CAS  PubMed  Google Scholar 

  14. Arens, R. & Schoenberger, S. P. Plasticity in programming of effector and memory CD8 T-cell formation. Immunol. Rev. 235, 190–205 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Cheuk, S. et al. CD49a expression defines tissue-resident CD8+ T cells poised for cytotoxic function in human skin. Immunity 46, 287–300 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  16. Ganesan, A. P. et al. Tissue-resident memory features are linked to the magnitude of cytotoxic T cell responses in human lung cancer. Nat. Immunol. 18, 940–950 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Laidlaw, B. J. et al. CD4+ T cell help guides formation of CD103+ lung-resident memory CD8+ T cells during influenza viral infection. Immunity 41, 633–645 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Iijima, N. & Iwasaki, A. T cell memory. A local macrophage chemokine network sustains protective tissue-resident memory CD4 T cells. Science 346, 93–98 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Ohnmacht, C. et al. The microbiota regulates type 2 immunity through RORγt+ T cells. Science 349, 989–993 (2015).

    CAS  PubMed  Google Scholar 

  20. Arpaia, N. et al. A distinct function of regulatory T cells in tissue protection. Cell 162, 1078–1089 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Endo, Y., Hirahara, K., Yagi, R., Tumes, D. J. & Nakayama, T. Pathogenic memory type TH2 cells in allergic inflammation. Trends Immunol. 35, 69–78 (2014).

    CAS  PubMed  Google Scholar 

  23. Islam, S. A. et al. Mouse CCL8, a CCR8 agonist, promotes atopic dermatitis by recruiting IL-5+ TH2 cells. Nat. Immunol. 12, 167–177 (2011).

    CAS  PubMed  Google Scholar 

  24. 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 e9 (2016).

    CAS  PubMed  Google Scholar 

  25. Seumois, G. et al. Transcriptional profiling of TH2 cells identifies pathogenic features associated with asthma. J. Immunol. 197, 655–664 (2016).

    CAS  PubMed  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  27. 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.e6 (2018).

    CAS  PubMed  Google Scholar 

  28. Wuthrich, M., Deepe, G. S. Jr. & Klein, B. Adaptive immunity to fungi. Annu. Rev. Immunol. 30, 115–148 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  29. Knutsen, A. P. et al. Fungi and allergic lower respiratory tract diseases. J. Allergy Clin. Immunol. 129, 280–291 (2012)

    PubMed  Google Scholar 

  30. Jaakkola, M. S., Ieromnimon, A. & Jaakkola, J. J. Are atopy and specific IgE to mites and molds important for adult asthma? J. Allergy Clin. Immunol. 117, 642–648 (2006).

    CAS  PubMed  Google Scholar 

  31. Eming, S. A., Wynn, T. A. & Martin, P. Inflammation and metabolism in tissue repair and regeneration. Science 356, 1026–1030 (2017).

    CAS  PubMed  Google Scholar 

  32. Anderson, K. G. et al. Cutting edge: intravascular staining redefines lung CD8 T cell responses. J. Immunol. 189, 2702–2706 (2012).

    CAS  PubMed  Google Scholar 

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

    Google Scholar 

  34. Sakaguchi, S., Miyara, M., Costantino, C. M. & Hafler, D. A. FOXP3+ regulatory T cells in the human immune system. Nat. Rev. Immunol. 10, 490–500 (2010).

    CAS  PubMed  Google Scholar 

  35. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Bromley, S. K., Thomas, S. Y. & Luster, A. D. Chemokine receptor CCR7 guides T cell exit from peripheral tissues and entry into afferent lymphatics. Nat. Immunol. 6, 895–901 (2005).

    CAS  PubMed  Google Scholar 

  37. Durek, P. et al. Epigenomic profiling of human CD4+ T cells supports a linear differentiation model and highlights molecular regulators of memory development. Immunity 45, 1148–1161 (2016).

    CAS  PubMed  Google Scholar 

  38. Hondowicz, B. D. et al. Interleukin-2-dependent allergen-specific tissue-resident memory cells drive asthma. Immunity 44, 155–166 (2016).

    CAS  PubMed  Google Scholar 

  39. Ito, M. et al. Brain regulatory T cells suppress astrogliosis and potentiate neurological recovery. Nature 565, 246–250 (2019).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  41. Garg, G. et al. Blimp1 prevents methylation of Foxp3 and loss of regulatory T cell identity at sites of inflammation. Cell Rep. 26, 1854–1868.e5 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Cepek, K. L. et al. Adhesion between epithelial cells and T lymphocytes mediated by E-cadherin and the alpha E beta 7 integrin. Nature 372, 190–193 (1994).

    CAS  PubMed  Google Scholar 

  44. Lee, Y. T. et al. Environmental and antigen receptor-derived signals support sustained surveillance of the lungs by pathogen-specific cytotoxic T lymphocytes. J. Virol. 85, 4085–4094 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Makinde, T., Murphy, R. F. & Agrawal, D. K. The regulatory role of TGF-β in airway remodeling in asthma. Immunol. Cell Biol. 85, 348–356 (2007).

    CAS  PubMed  Google Scholar 

  46. Van den Bossche, J., Malissen, B., Mantovani, A., De Baetselier, P. & Van Ginderachter, J. A. Regulation and function of the E-cadherin/catenin complex in cells of the monocyte-macrophage lineage and DCs. Blood 119, 1623–1633 (2012).

    CAS  PubMed  Google Scholar 

  47. Lambrecht, B. N. & Hammad, H. The airway epithelium in asthma. Nat. Med. 18, 684–692 (2012).

    CAS  PubMed  Google Scholar 

  48. Foster, P. S. et al. Modeling TH2 responses and airway inflammation to understand fundamental mechanisms regulating the pathogenesis of asthma. Immunol. Rev. 278, 20–40 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Greenberger, P. A. et al. Allergic bronchopulmonary aspergillosis. J. Allergy Clin. Immunol. Pract. 2, 703–708 (2014).

    PubMed  PubMed Central  Google Scholar 

  50. Higashiyama, R. et al. Negligible contribution of bone marrow-derived cells to collagen production during hepatic fibrogenesis in mice. Gastroenterology 137, 1459–1466.e1 (2009).

    CAS  PubMed  Google Scholar 

  51. Rubtsov, Y. P. et al. Regulatory T cell-derived interleukin-10 limits inflammation at environmental interfaces. Immunity 28, 546–558 (2008).

    CAS  PubMed  Google Scholar 

  52. Kerzerho, J. et al. Programmed cell death ligand 2 regulates TH9 differentiation and induction of chronic airway hyperreactivity. J. Allergy Clin. Immunol. 131, 1048–1057.e2 (2013).

    CAS  PubMed  Google Scholar 

  53. Hirahara, K. et al. Repressor of GATA regulates TH2-driven allergic airway inflammation and airway hyperresponsiveness. J. Allergy Clin. Immunol. 122, 512–520.e11 (2008).

    CAS  PubMed  Google Scholar 

  54. Shinoda, K. et al. Thy1+IL-7+ lymphatic endothelial cells in iBALT provide a survival niche for memory T-helper cells in allergic airway inflammation. Proc. Natl Acad. Sci. USA 113, E2842–E2851 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Mato, N. et al. Memory-type ST2+CD4+ T cells participate in the steroid-resistant pathology of eosinophilic pneumonia. Sci. Rep. 7, 6805 (2017).

    PubMed  PubMed Central  Google Scholar 

  56. Onodera, A. et al. Menin controls the memory TH2 cell function by maintaining the epigenetic integrity of TH2 cells. J. Immunol. 199, 1153–1162 (2017).

    CAS  PubMed  Google Scholar 

  57. Onodera, A. et al. Spatial interplay between polycomb and trithorax complexes controls transcriptional activity in T lymphocytes. Mol. Cell Biol. 35, 3841–3853 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Buenrostro, J. D., Giresi, P. G., Zaba, L. C., Chang, H. Y. & Greenleaf, W. J. Transposition of native chromatin for fast and sensitive epigenomic profiling of open chromatin, DNA-binding proteins and nucleosome position. Nat. Methods 10, 1213–1218 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Shih, H. Y. et al. Developmental acquisition of regulomes underlies innate lymphoid cell functionality. Cell 165, 1120–1133 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Li, H. et al. The Sequence Alignment/Map format and SAMtools. Bioinformatics 25, 2078–2079 (2009).

    PubMed  PubMed Central  Google Scholar 

  62. Quinlan, A. R. & Hall, I. M. BEDTools: a flexible suite of utilities for comparing genomic features. Bioinformatics 26, 841–842 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We are grateful to Y. Kanno for critically reading and providing valuable suggestions on the manuscript. We are grateful to N. Iijima and K. J. Ishii for providing valuable suggestions on the parabiosis surgery. We are grateful to N. Takayama and K. Eto for providing valuable suggestions on the ATAC-Seq experiments. We thank T. Ito, K. Sugaya, M. Kato and T. Wada for excellent technical assistance. This work was supported by the following grants: Ministry of Education, Culture, Sports, Science and Technology Grants-in-Aid for Scientific Research (S) 26221305, JP19H05650, (C) 17K08876, 18K07164 and 19K16683; Practical Research Project for Allergic Diseases and Immunology (Research on Allergic Diseases and Immunology) from AMED (numbers JP19ek0410060 and JP19ek0410045); AMED-PRIME, AMED (number JP19gm6110005); AMED-CREST, AMED (number JP19gm1210003); the Mochida Memorial Foundation for Medical and Pharmaceutical Research, The Ichiro Kanehara Foundation for the Promotion of Medical Sciences and Medical Care; the Naito Foundation and the Takeda Science Foundation.

Author information

Authors and Affiliations

Authors

Contributions

T.I., K. Hirahara and T.N. conceived of and designed the experiments. T.I., K. Hirahara, K.K., M.K., A.A., Y.M., J.K., A.O. and N.M. performed the experiments. T.I., K. Hirahara, K.K., M.K., A.O., D.J.T., Y.G., K. Hagiwara, Y.I., T.S., K.T. and T.N. analyzed and interpreted the data. T.I., K. Hirahara, D.J.T. and T.N. wrote, reviewed and edited the paper.

Corresponding author

Correspondence to Toshinori Nakayama.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Zoltan Fehervari was the primary editor 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.

Integrated supplementary information

Supplementary Figure 1 Repeated exposure to Aspergillus antigen resulted in chronic lung inflammation.

(a) A schematic illustration of 7-week repeated Aspergillus antigen exposure. (b) The quantitative RT-PCR of Col1a1 in the lung from control BALB/c mice (control) and Aspergillus antigen-exposed mice (Asp-Ag) (n = 10 mice per group). (c) Representative confocal micrographs of the lung stained with anti-GFP (green), anti-type-I collagen (purple), anti-CD3 (yellow) and DAPI (blue) from wild-type C57BL/6 mice (WT) and Collagen 1a2-GFP transgenic mice with or without Aspergillus antigen exposure. Scale bars represent 20 μm. Representative findings of five independent experiments are shown. (d) The cytokine concentrations in BAL fluid from control mice (control) (n = 6) and Aspergillus antigen-exposed mice (Asp-Ag) (n = 8). Findings pooled from two independent experiments are shown. (e) The absolute number of total cells (total), macrophages (Mac), eosinophils (Eos), neutrophils (Neut) and lymphocytes (Lymph) in BAL fluid from control mice (control) (n = 6) and Aspergillus antigen-exposed mice (Asp-Ag) (n = 11). Findings pooled from two independent experiments are shown. (f) Airway hyperresponsiveness (AHR) was assessed by increased airway resistance with increasing doses of methacholine. Findings pooled from two independent experiments are shown (n = 5 mice per group). (g) The representative gating strategy for CD4+ T cells in the lung. (h, i) The cell surface profiles of CD44 and CD62L in the lung (h) and spleen (i) from control mice and Aspergillus antigen-exposed mice (Asp-Ag) (left), and pooled data from 5 mice per group (right). Data are the mean ± SD (1b, 1d, 1e, 1h and 1i) or the mean ± SEM (1f). P-values were calculated using the two-sided Mann-Whitney U test (1b, 1d, 1e, 1h and 1i) or two-way ANOVA test (1f). (j) Representative histological sections of the lung stained with Hematoxylin and Eosin from Aspergillus antigen-exposed wild-type BALB/c mice (left) and Foxn1nu mice (right) in lower magnification (upper) and higher magnification (lower) views. Scale bars represent 1 mm or 100 μm. Representative findings of two independent experiments are shown. (k) Representative histological sections of lungs stained with Sirius Red from Aspergillus antigen-exposed wild-type mice (left) and Foxn1nu mice (right). Scale bars represent 100 μm. More than three mice per group were analyzed.

Supplementary Figure 2 Repeated exposure to Aspergillus antigen induced CD69hi CD4+ tissue-resident T cells in the lung.

(a) Cell surface markers of CD44hiCD62Llo CD4+ T cells in the spleen from control (blue lines) and Aspergillus antigen-exposed mice (red) with isotype-matched control antibody (gray). Representative findings from two independent experiments are shown. (b) The ratio of cells positive for the indicated cell surface marker in CD44hiCD62LloCD69hi CD4+ T cells in comparison to CD44hiCD62LloCD69lo CD4+ T cells from control mice (horizontal axis) and Aspergillus antigen-exposed mice (Asp-Ag, vertical axis). Upper and lower diagonal lines indicate the expression ratio in Aspergillus antigen-exposed mice in comparison to control mice is >2 (red) or <2 (blue), respectively. (c) The cell surface markers on CD44hiCD62LloCD69hi CD4+ T cells in the lung from control (blue lines) and Aspergillus antigen-exposed mice (red) with isotype-matched control antibody (gray). Representative findings from two independent experiments are shown. (d) Pooled data showing the absolute number of CD103hi T cells in CD44hiCD62LloCD69hi CD4+ T cells from control mice (n = 5) and Aspergillus antigen-exposed mice (n = 6). (e, f) Pooled data showing the proportion (e) and absolute number (f) of CD44hiCD62LloCD69hiCD103hi CD4+ T cells in the spleen from control mice (n = 5) and Aspergillus antigen-exposed mice (n = 6). (g) A schematic illustration of repeated Aspergillus antigen exposure followed by intravenous staining for CD4+ T cells. (h) Intravenous staining of anti-CD4 (CD4 (iv)) and anti-CD4 staining in CD4+ T cells in the peripheral blood from control mice (control) and Aspergillus antigen-exposed mice (Asp-Ag). Representative findings from two independent experiments are shown. Data are the mean ± SD. P-values were calculated using the two-sided Mann-Whitney U test (2d, 2e, and 2f). (i) Intravenous staining of anti-CD4 (CD4 (iv)) and anti-CD44 staining in CD4+ T cells in the lung from control mice (control) and Aspergillus antigen-exposed mice (Asp-Ag) (left), and the percentage of tissue-resident T cells in CD4+ T cell population from control mice (control) and Aspergillus antigen-exposed mice (Asp-Ag) (n = 9 mice per group). (j) Intravenous staining of anti-CD4 (CD4 (iv)) and anti-CD44 staining in CD4+ T cells in the spleen from control mice (control) and Aspergillus antigen-exposed mice (Asp-Ag) (left), and the percentage of tissue-resident T cells in CD4+ T cell population from control mice (control) and Aspergillus antigen-exposed mice (Asp-Ag) (n = 9 mice per group). (k) The absolute number of tissue-resident T cells in the CD44hiCD69hi CD4+ T cell population (red) and the CD44loCD69lo CD4+ T cell population (blue) from control mice (control; n = 9) and Aspergillus antigen-exposed mice (Asp-Ag; n = 8), which were investigated in Fig. 1h. (l) The t-distributed stochastic linear embedding (t-SNE) plot of CD4+ T cells in the lung from control (left) and Aspergillus antigen-exposed mice (right). CD4(iv) (-) CD4+ T cells and CD44hiCD69hi CD4+ T cells in the t-SNE space of the CD4+ T cells (green) are shown in red and blue, respectively. Representative findings from two independent experiments are shown. (m) Representative confocal micrograph of the lung in a control mouse (control) stained with anti-CD4 (red), anti-CD69 (green), and anti-type l collagen (blue). Scale bars represent 50 μm. Representative findings of three independent experiments are shown. Data are the mean ± SD. P-values were calculated using the two-sided Mann-Whitney U test (2i, 2j, and 2k).

Supplementary Figure 3 Tissue-resident CD69hi CD4+ T cells expressed high levels of fibrosis-related genes.

(a) A scatter plot showing the FKPM of a single gene in two individual sample sets in CD44lo T cells, CD44hiCD69lo T cells, CD44hiCD69hiCD103lo T cells, and CD44hiCD69hiCD103hi T cells. (b) A heatmap showing the similarity and clustering of CD44lo T cells, CD44hiCD69lo T cells, CD44hiCD69hiCD103lo T cells and CD44hiCD69hiCD103hi T cells using the gene expression profiles. (c) Venn diagram depicts the number of genes with expression levels that were increased >8-fold in CD44hiCD69hiCD103lo T cells (red circle) or CD44hiCD69hiCD103hi T cells (blue circle) in comparison to CD44lo T cells. (d) Genes for which a >8-fold expression change was observed in both CD44hiCD69hiCD103lo T cells and CD44hiCD69hiCD103hi T cells (n= 60) in comparison to CD44lo T cells are depicted by the heatmap. (e) The Venn diagram depicts the number of genes with expression levels that were decreased <0.25-fold in CD44hiCD69hiCD103lo T cells (red circle) or CD44hiCD69hiCD103hi T cells (blue circle) in comparison to CD44lo T cells. (f) Genes for which a <0.25-fold expression change was uniquely observed in CD44hiCD69hiCD103lo T cells (n= 7), CD44hiCD69hiCD103hi T cells (n= 23), or both CD44hiCD69hiCD103lo T cells and CD44hiCD69hiCD103hi T cells (n= 18) in comparison to CD44lo T cells are depicted by heatmap. (g) The relative gene expression profiles in the co-regulation network in naive (CD44lo) CD4+ T cells (left), CD69hiCD103lo CD4+ T cells (middle), and CD69hiCD103hi CD4+ T cells (right). (h) The list of GO terms in the indicated categories that are involved in fibrotic responses. Genes that are included in each GO term are listed on Supplementary Table 1.

Figure Supplementary 4 Tissue-resident CD69hiCD103lo CD4+ T cells exhibit high levels of cytokine production.

(a, b) The absolute numbers of Foxp3-, GATA3- and Rorγt-positive cells in CD44hiCD69hiCD103lo CD4+ T cells (a) and CD44hiCD69hiCD103hi CD4+ Treg cells (b) in the lung from Aspergillus antigen-exposed wild-type mice, which were investigated in Fig. 3b, c (samples stained with Foxp3 and GATA3; n=16, samples stained with Foxp3 and RORγt; n=11). (c) The absolute numbers of the indicated cytokines in CD44hiCD69hiCD103lo CD4+ T cells (red) and CD44hiCD69hiCD103hi CD4+ Treg cells (blue) in the lung from Aspergillus antigen-exposed mice (Asp-Ag), which were investigated in Fig. 3e. Findings of five pooled independent experiments are shown. Data are the mean ± SD. P-values were calculated using the two-sided Mann-Whitney U test.

Supplementary Figure 5 Foxp3+ Treg cells suppress the pathology of chronic lung inflammation.

(a) A schematic illustration of the repeated exposure to Aspergillus antigen accompanied by the intranasal administration of diphtheria toxin. (b, c) Intracellular-staining profiles of Foxp3 (right) in the lung (b) and spleen (c) from Aspergillus antigen-exposed (Asp-Ag) wild-type mice (n = 9) and Foxp3-DTR transgenic mice (n = 9) from three independent experiments (right). Findings of two pooled independent experiments are shown. (d) Percentages of CD69hiCD103hi CD4+ T cells (left) and CD69hiCD103lo CD4+ T cells (right) in the lung from Aspergillus antigen-exposed wild-type (n = 9) and Foxp3-DTR transgenic mice (n = 9). Findings of two pooled independent experiments are shown. (e) The absolute numbers of Foxp3-, GATA3- and Rorγt-positive cells in CD69hiCD103hi CD4+ T cells in the lung from Aspergillus antigen-exposed wild-type mice (n = 9) and Foxp3-DTR transgenic mice (n = 9), which were investigated in Fig. 4b. Findings of two pooled independent experiments are shown. (f) Intracellular-staining profiles of Foxp3, GATA3, and Rorγt in CD69hiCD103lo CD4+ T cells in the lung from Aspergillus antigen-exposed wild-type mice (n = 9) and Foxp3-DTR transgenic mice (n = 9). Findings of two pooled independent experiments are shown. (g) The absolute number of Foxp3-, GATA3- and Rorγt-positive cells in CD69hiCD103lo CD4+ T cells in the lung from Aspergillus antigen-exposed wild-type mice (n = 9) and Foxp3-DTR transgenic mice (n = 9), which were investigated in Supplementary Fig. 4f. Findings of two pooled independent experiments are shown. (h) The percentages of the indicated cytokines in CD69hiCD103hi CD4+ T cells in the lung from Aspergillus antigen-exposed mice (Asp-Ag). The findings of three pooled independent experiments are shown. Data are the mean ± SD. P-values were calculated using the two-sided Mann-Whitney U test (5b, 5c, and 5d), a two-way ANOVA (5e, 5f, and 5g) or an unpaired two-sided Student’s t-test (5h). (i) A schematic illustration of the repeated exposure of Rorcfl/fl mice and Rorcfl/flFoxp3-cre mice to Aspergillus antigen for 7 weeks. (j) The absolute numbers of CD69hiCD103hiFoxp3hiRorγthi CD4+ T cells in the lung from Rorcfl/fl mice and Rorcfl/flFoxp3-cre mice subjected to repeated exposure to Aspergillus antigen (n = 5 mice per group). Data are the mean ± SD. P-values were calculated using the two-sided Mann-Whitney U test in 5j.

Figure Supplementary 6 CD103lo TRM and CD103hi tissue-resident Treg cells have distinct regulomes.

(a) A schematic illustration of repeated exposure of mice to Aspergillus antigen for 7 weeks with a 7-week rest period followed by an ATAC-Seq assay. (b) A heatmap showing the relative enrichment of TF motifs in the indicated CD4+ T cell subpopulation. (c, d, e, f, g) Comparison of ATAC-seq signals across tissue-resident CD4+ T cell signature genes, including (c) Il4, Il5 and Il13 loci, (d) Il17a, and Il17f loci, (e) Foxp3 locus, (f) Itgae locus, and (g) Areg locus. (h) The fibrosis-related genes expressed in CD44hiCD69hiCD103lo T cells unique (left, n= 15), CD44hiCD69hiCD103hi T cells unique (middle, n= 13), and both CD44hiCD69hiCD103lo T cells and CD44hiCD69hiCD103hi T cells (right, n= 27)are categorized into the indicated groups.

Figure Supplementary 7 CD4+ TRM cells in the lung induce the inflammation accompanied by fibrotic responses.

(a) A schematic illustration of the repeated exposure of mice to Aspergillus antigen for 7 weeks with a 7-week rest period followed by challenge with a single dose of Aspergillus antigen. (b, c) Representative histological sections of the lungs stained with Hematoxylin and Eosin (upper panel), PAS (middle panel), and Sirius red (lower panel) from four groups of mice (Sensitization (-) Rechallenge (-), Sensitization (-) Rechallenge (+), Sensitization (+) Rechallenge (-), Sensitization (+) Rechallenge (+) 24 hours (b) or 48 hours (c) after rechallenge of Aspergillus antigen are shown. Scale bars represent 100 μm. More than three mice per group were analyzed. (d) A schematic illustration of the repeated exposure of mice to Aspergillus antigen for 7 weeks with a 7-week rest period with FTY720 treatment followed by challenge with a single dose of Aspergillus antigen. (e) A schematic illustration of the repeated exposure of mice to Aspergillus antigen for 7 weeks with a 7-week rest period with FTY720 treatment and anti-CD103 treatment followed by challenge with a single dose of Aspergillus antigen. (f) A representative plot of intravenous staining of anti-CD4 (CD4 (iv)) and anti-CD103 staining in CD44loCD69lo CD4+ T cells (left), and CD44hiCD69hi CD4+ T cells (right) in the lung from control antibody-treated mice (control ab.) and anti-CD103 antibody-treated mice (anti-CD103 ab.). Representative findings from two independent experiments are shown. (g) The absolute numbers of CD44hiCD69hiCD103hi CD4+ T cells in the lung from anti-control antibody-treated mice (control ab.) and anti-CD103 antibody-treated mice (anti-CD103 ab.) (n = 3 mice per group). Data are the mean ± SD. P-values were calculated using an unpaired two-sided Student’s t-test (g).

Supplementary information

Supplementary Information

Supplementary Figs. 1–7 and Supplementary Table 1

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Ichikawa, T., Hirahara, K., Kokubo, K. et al. CD103hi Treg cells constrain lung fibrosis induced by CD103lo tissue-resident pathogenic CD4 T cells. Nat Immunol 20, 1469–1480 (2019). https://doi.org/10.1038/s41590-019-0494-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-019-0494-y

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