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:

Reciprocal transcription factor networks govern tissue-resident ILC3 subset function and identity

Abstract

Innate lymphoid cells (ILCs) are guardians of mucosal immunity, yet the transcriptional networks that support their function remain poorly understood. We used inducible combinatorial deletion of key transcription factors (TFs) required for ILC development (RORγt, RORα and T-bet) to determine their necessity in maintaining ILC3 identity and function. Both RORγt and RORα were required to preserve optimum effector functions; however, RORα was sufficient to support robust interleukin-22 production among the lymphoid tissue inducer (LTi)-like ILC3 subset, but not natural cytotoxicity receptor (NCR)+ ILC3s. Lymphoid tissue inducer-like ILC3s persisted with only selective loss of phenotype and effector functions even after the loss of both TFs. In contrast, continued RORγt expression was essential to restrain transcriptional networks associated with type 1 immunity within NCR+ ILC3s, which coexpress T-bet. Full differentiation to an ILC1-like population required the additional loss of RORα. Together, these data demonstrate how TF networks integrate within mature ILCs after development to sustain effector functions, imprint phenotype and restrict alternative differentiation programs.

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: RORα and RORγt combine to preserve functions in NCR+, but not LTi-like, ILC3 subsets.
Fig. 2: NCR+ ILCs undergo extensive transcriptomic changes upon TF deletion.
Fig. 3: Loss of both RORγt and RORα expression within NCR+ ILCs drives full differentiation to a mature ILC1-like phenotype.
Fig. 4: Ex-ILC3s and ILC1s exhibit a comparable spectrum of phenotypes within the SILP.
Fig. 5: RORα expression is sufficient to preserve IL-23-mediated expression of IL-22 by non-LTi-like ILC3s in the absence of RORγt and T-bet.

Similar content being viewed by others

Data availability

scRNA-seq data on ‘understanding transcriptional identity in ILCs’ are available in the ArrayExpress database under accession no. E-MTAB-9795. ATAC–seq data are available in NCBI’s Gene Expression Omnibus60 under accession number GSE163149. Further information and requests for resources should be directed to and will be fulfilled by the corresponding authors. Source data are provided with this paper.

Code availability

The custom code used did not differ significantly from the intended implementation that we referenced nor did it contain any new math. The custom R code was used only for visualization in this study.

References

  1. Artis, D. & Spits, H. The biology of innate lymphoid cells. Nature 517, 293–301 (2015).

    Article  CAS  PubMed  Google Scholar 

  2. Withers, D. R. & Hepworth, M. R. Group 3 innate lymphoid cells: communications hubs of the intestinal immune system. Front Immunol. 8, 1298 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Walker, J. A., Barlow, J. L. & McKenzie, A. N. Innate lymphoid cells—how did we miss them? Nat. Rev. Immunol. 13, 75–87 (2013).

    Article  CAS  PubMed  Google Scholar 

  4. Spits, H. et al. Innate lymphoid cells—a proposal for uniform nomenclature. Nat. Rev. Immunol. 13, 145–149 (2013).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Melo-Gonzalez, F. & Hepworth, M. R. Functional and phenotypic heterogeneity of group 3 innate lymphoid cells. Immunology 150, 265–275 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Colonna, M. Innate lymphoid cells: diversity, plasticity, and unique functions in immunity. Immunity 48, 1104–1117 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Withers, D. R. et al. Transient inhibition of ROR-γt therapeutically limits intestinal inflammation by reducing TH17 cells and preserving group 3 innate lymphoid cells. Nat. Med. 22, 319–323 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Mjosberg, J. M. et al. Human IL-25- and IL-33-responsive type 2 innate lymphoid cells are defined by expression of CRTH2 and CD161. Nat. Immunol. 12, 1055–1062 (2011).

    Article  PubMed  CAS  Google Scholar 

  10. Geremia, A. et al. IL-23-responsive innate lymphoid cells are increased in inflammatory bowel disease. J. Exp. Med. 208, 1127–1133 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Vivier, E. et al. Innate lymphoid cells: 10 years on. Cell 174, 1054–1066 (2018).

    Article  CAS  PubMed  Google Scholar 

  12. Gao, Y. et al. Tumor immunoevasion by the conversion of effector NK cells into type 1 innate lymphoid cells. Nat. Immunol. 18, 1004–1015 (2017).

    Article  CAS  PubMed  Google Scholar 

  13. Eberl, G. et al. An essential function for the nuclear receptor RORγt in the generation of fetal lymphoid tissue inducer cells. Nat. Immunol. 5, 64–73 (2004).

    Article  CAS  PubMed  Google Scholar 

  14. Walker, J. A. et al. Polychromic reporter mice reveal unappreciated innate lymphoid cell progenitor heterogeneity and elusive ILC3 progenitors in bone marrow. Immunity 51, 104–118 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Lo, B. C. et al. The orphan nuclear receptor RORα and group 3 innate lymphoid cells drive fibrosis in a mouse model of Crohn’s disease. Sci. Immunol. 1, eaaf8864 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  16. Lo, B. C. et al. The transcription factor RORα preserves ILC3 lineage identity and function during chronic intestinal infection. J. Immunol. 203, 3209–3215 (2019).

    Article  CAS  PubMed  Google Scholar 

  17. Halim, T. Y. et al. Retinoic-acid-receptor-related orphan nuclear receptor alpha is required for natural helper cell development and allergic inflammation. Immunity 37, 463–474 (2012).

    Article  CAS  PubMed  Google Scholar 

  18. Luche, H., Weber, O., Nageswara Rao, T., Blum, C. & Fehling, H. J. Faithful activation of an extra-bright red fluorescent protein in ‘knock-in’ Cre-reporter mice ideally suited for lineage tracing studies. Eur. J. Immunol. 37, 43–53 (2007).

    Article  CAS  PubMed  Google Scholar 

  19. Satoh-Takayama, N. et al. Microbial flora drives interleukin 22 production in intestinal NKp46+ cells that provide innate mucosal immune defense. Immunity 29, 958–970 (2008).

    Article  CAS  PubMed  Google Scholar 

  20. Sawa, S. et al. Lineage relationship analysis of RORγt+ innate lymphoid cells. Science 330, 665–669 (2010).

    Article  CAS  PubMed  Google Scholar 

  21. Hirota, K. et al. Fate mapping of IL-17-producing T cells in inflammatory responses. Nat. Immunol. 12, 255–263 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Lazarevic, V. et al. T-bet represses TH17 differentiation by preventing Runx1-mediated activation of the gene encoding RORγt. Nat. Immunol. 12, 96–104 (2011).

    Article  CAS  PubMed  Google Scholar 

  23. Scott, C. L. et al. The transcription factor ZEB2 is required to maintain the tissue-specific identities of macrophages. Immunity 49, 312–325 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Van de Sande, B. et al. A scalable SCENIC workflow for single-cell gene regulatory network analysis. Nat. Protoc. 15, 2247–2276 (2020).

    Article  PubMed  CAS  Google Scholar 

  26. Yagi, R. et al. The transcription factor GATA3 Is critical for the development of all IL-7Rα-expressing innate lymphoid cells. Immunity 40, 378–388 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Koues, O. I. et al. Distinct gene regulatory pathways for human innate versus adaptive lymphoid cells. Cell 165, 1134–1146 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Veldhoen, M. et al. The aryl hydrocarbon receptor links TH17-cell-mediated autoimmunity to environmental toxins. Nature 453, 106–109 (2008).

    Article  CAS  PubMed  Google Scholar 

  29. Quintana, F. J. et al. Control of Treg and TH17 cell differentiation by the aryl hydrocarbon receptor. Nature 453, 65–71 (2008).

    Article  CAS  PubMed  Google Scholar 

  30. Hepworth, M. R. et al. Innate lymphoid cells regulate CD4+ T cell responses to intestinal commensal bacteria. Nature 498, 113–117 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Monticelli, L. A. et al. Arginase 1 is an innate lymphoid-cell-intrinsic metabolic checkpoint controlling type 2 inflammation. Nat. Immunol. 17, 656–665 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Karagiannis, F. et al. Lipid-droplet formation drives pathogenic group 2 innate lymphoid cells in airway inflammation. Immunity 52, 885 (2020).

    Article  CAS  PubMed  Google Scholar 

  33. Sanos, S. L. et al. RORγt and commensal microflora are required for the differentiation of mucosal interleukin-22-producing NKp46+ cells. Nat. Immunol. 10, 83–91 (2009).

    Article  CAS  PubMed  Google Scholar 

  34. Cella, M. et al. Subsets of ILC3-ILC1-like cells generate a diversity spectrum of innate lymphoid cells in human mucosal tissues. Nat. Immunol. 20, 980–991 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Peng, H. et al. Liver-resident NK cells confer adaptive immunity in skin-contact inflammation. J. Clin. Invest. 123, 1444–1456 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Sojka, D. K. et al. Tissue-resident natural killer (NK) cells are cell lineages distinct from thymic and conventional splenic NK cells. Elife 3, e01659 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Bai, L. et al. Liver type 1 innate lymphoid cells develop locally via an interferon-gamma-dependent loop. Science https://doi.org/10.1126/science.aba4177 (2021).

  38. Klose, C. S. et al. A T-bet gradient controls the fate and function of CCR6-RORγt+ innate lymphoid cells. Nature 494, 261–265 (2013).

    Article  CAS  PubMed  Google Scholar 

  39. Vonarbourg, C. et al. Regulated expression of nuclear receptor RORγt confers distinct functional fates to NK cell receptor-expressing RORγt+ innate lymphocytes. Immunity 33, 736–751 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Bottcher, J. P. et al. NK cells stimulate recruitment of cDC1 into the tumor microenvironment promoting cancer immune control. Cell 172, 1022–1037 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Brewitz, A. et al. CD8+ T cells orchestrate pDC-XCR1+ dendritic cell spatial and functional cooperativity to optimize priming. Immunity 46, 205–219 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Stehle, C. et al. T-bet and RORα control lymph node formation by regulating embryonic innate lymphoid cell differentiation. Nat. Immunol. https://doi.org/10.1038/s41590-021-01029-6 (in the press).

  43. Taylor, J. S. & Raes, J. Duplication and divergence: the evolution of new genes and old ideas. Annu. Rev. Genet. 38, 615–643 (2004).

    Article  CAS  PubMed  Google Scholar 

  44. Ciofani, M. et al. A validated regulatory network for Th17 cell specification. Cell 151, 289–303 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Gokmen, M. R. et al. Genome-wide regulatory analysis reveals that T-bet controls Th17 lineage differentiation through direct suppression of IRF4. J. Immunol. 191, 5925–5932 (2013).

    Article  PubMed  CAS  Google Scholar 

  46. Rawlins, E. L., Clark, C. P., Xue, Y. & Hogan, B. L. The Id2+ distal tip lung epithelium contains individual multipotent embryonic progenitor cells. Development 136, 3741–3745 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Choi, G. B. et al. The maternal interleukin-17a pathway in mice promotes autism-like phenotypes in offspring. Science 351, 933–939 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Oliphant, C. J. et al. MHCII-mediated dialog between group 2 innate lymphoid cells and CD4+ T cells potentiates type 2 immunity and promotes parasitic helminth expulsion. Immunity 41, 283–295 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Intlekofer, A. M. et al. Anomalous type 17 response to viral infection by CD8+ T cells lacking T-bet and eomesodermin. Science 321, 408–411 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Eberl, G. & Littman, D. R. Thymic origin of intestinal alphabeta T cells revealed by fate mapping of RORγt+ cells. Science 305, 248–251 (2004).

    Article  CAS  PubMed  Google Scholar 

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

    Article  Google Scholar 

  52. Zheng, G. X. et al. Massively parallel digital transcriptional profiling of single cells. Nat. Commun. 8, 14049 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Bourgon, R., Gentleman, R. & Huber, W. Independent filtering increases detection power for high-throughput experiments. Proc. Natl Acad. Sci. USA 107, 9546–9551 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Lun, A. T., Bach, K. & Marioni, J. C. Pooling across cells to normalize single-cell RNA-sequencing data with many zero counts. Genome Biol. 17, 75 (2016).

    Article  PubMed  CAS  Google Scholar 

  55. 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).

    CAS  PubMed  Google Scholar 

  56. Huber, D. & Vitek, W. sSeq: Shrinkage estimation of dispersion in Negative Binomial models for RNA-seq experiments with small sample size. R package version 1.24.0 (2019).

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Suo, S. et al. Revealing the critical regulators of cell identity in the mouse cell atlas. Cell Rep. 25, 1436–1445 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Ghaem-Maghami, M. et al. Intimin-specific immune responses prevent bacterial colonization by the attaching-effacing pathogen Citrobacter rodentium. Infect. Immun. 69, 5597–5605 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  60. Edgar, R., Domrachev, M. & Lash, A. E. Gene Expression Omnibus: NCBI gene expression and hybridization array data repository. Nucleic Acids Res. 30, 207–210 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank A. Ptasinska (Institute of Cancer and Genomic Sciences, University of Birmingham), for advice and help with ATAC–seq experiments, Genomics Birmingham, the genomic and sequencing facility of the University of Birmingham, and the University of Birmingham Flow Cytometry Platform. We thank G. Howell and the University of Manchester flow cytometry core, and A. Hayes and C. Morrisroe in the University of Manchester Genomic Technologies core facility for their help with scRNA-seq. We thank G. Frankel (Imperial College, London), for the provision of C. rodentium ICC169. We thank the following for their kind provision of mice: D. Littman (Howard Hughes Medical Institute, New York; Rorccre mice), A. McKenzie (MRC-LMB, Cambridge; Rorafl/fl mice), B. Stockinger (The Francis Crick Institute, London; Il17acre mice), S. Reiner (Columbia University, New York; Tbx21fl/fl mice) and J. Fehling (Ulm University, Germany; Rosa26tdRFP mice). We thank C. Stehle and C. Romagnani (Charité University Medicine Berlin, Germany) for critical discussion of data and the manuscript. This work was supported by a Senior Research Fellowship from the Wellcome Trust to D.R.W. (110199/Z/15/Z) and by a Royal Society and Wellcome Trust Sir Henry Dale Fellowship (105644/Z/14/Z) and a Lister Institute of Preventative Medicine Prize to M.R.H. This work was also supported by a Wellcome Trust ISSF funding award (105610/Z/14/Z) to M.R.H. C.M.F. was supported by a Wellcome Investigator award (106898/A/15/Z) to J. E. Allen.

Author information

Authors and Affiliations

Authors

Contributions

R.F. designed and performed experiments, analyzed data and contributed to writing the manuscript. C.M.F. analyzed sequencing data and performed in silico experiments. C.W., S.L.B. and J.S. performed and assisted with experiments. S.T.H.N., S.M.B. and S.A. analyzed sequencing data and provided extensive bioinformatics support. M.R.H. designed and performed experiments, analyzed data and wrote the manuscript. D.R.W. designed experiments, analyzed data and wrote the manuscript.

Corresponding authors

Correspondence to Matthew R. Hepworth or David R. Withers.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Immunology thanks the anonymous reviewers for their contribution to the peer review of this work. Peer reviewer reports are available. L. A. Dempsey 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.

Extended data

Extended Data Fig. 1 ILC3 subset-specific deletion of TFs reveals that LTi-like cells express normal levels of IL-22 in the absence of both RORγt and RORα.

Il-17acre mice were used to enable ILC3 subset specific targeting of TF-deletion. a Gating strategy for identification of tdRFP+ ILCs from SILP of Il17acre mice. b Expression of RORγt versus NKp46 by tdRFP and tdRFP+ ILCs from SILP of Il17acre mice. c Proportion of ILCs in the SILP of Il17acre mice that express tdRFP at different ages (wks) post birth. d Representative CCR6, CD4, c-Kit, NK1.1 and NKp46 expression by tdRFP+ ILCs from SILP of Il17acre mice. e Proportion of tdRFP+ ILCs from SILP of Il17acre mice expressing molecules assessed in ‘d’. f Representative expression of RORγt versus CD127 by tdRFP and tdRFP+ ILCs from SILP of Il17acre, Il17aΔRORγt and Il17aΔRORγt/RORα mice. g Percentage of tdRFP and tdRFP+ ILCs expressing RORγt (Il17acre: n = 12; Il17aΔRORγt: n = 11; from 4 independent experiments). h Representative expression of RORγt versus IL-17A and IL-22 by tdRFP+ ILCs from SILP of Il17acre, Il17aΔRORγt and Il17aΔRORγt/RORα mice. i Total number of tdRFP+ ILCs from SILP of Il17acre, Il17aΔRORγt and Il17aΔRORγt/RORα mice expressing IL-17A (top) and IL-22 (bottom) (Il17acre: n = 19; Il17aΔRORγt: n = 14; Il17aΔRORγt/RORα: n = 19; from 6 independent experiments). Each data point on graphs is a mouse, horizontal bars denote the median. Statistical significance in ‘g’ was tested using unpaired two-tailed Student’s t tests, and in ‘i’ an ordinary one-way ANOVA with Tukey’s test for multiple comparisons. ns = not significant, **P ≤ 0.01, ****P ≤ 0.0001. Exact P values are provided in the source data.

Source data

Extended Data Fig. 2 Efficient inducible deletion of targeted transcription factors in Id2creERT2, Id2iΔRORγt, Id2iΔRORγt/RORα and Id2iΔRORγt/Tbet mice.

a Representative histograms showing expression of RORγt (upper row) and T-bet (lower row) by tdRFP+KLRG1- ILCs from SILP of Id2creERT2, Id2iΔRORγt, Id2iΔRORγt/RORα and Id2iΔRORγt/Tbet mice, dotted line denotes positive/negative staining. b Proportion of tdRFP+KLRG1- ILCs expressing RORγt and T-bet in Id2creERT2 (n=20), Id2iΔRORγt (n=13), Id2iΔRORγt/RORα (n=8) and Id2iΔRORγt/Tbet (n=17) mice (Data are representative of 7 independent experiments). c Schematics of Rorc and Rora cDNA showing exons, the location of LoxP sites and the qPCR primer annealing sites, floxed exons removed by cre-mediated deletion are in light grey. Relative expression of transcripts for the exon3-exon4, exon7-exon8 and exon8-exon9 junctions of the Rorc locus (d) and for the exon3-exon4, exon7-exon8 and exon9-exon10 junctions of the Rora locus (e) as depicted in ‘c’, determined by qPCR analysis of cDNA prepared from tdRFP+ ILCs sorted from SILP of Id2creERT2 (n=5), Id2iΔRORγt (n=7), Id2iΔRORγt/RORα (n=4) and Id2iΔRORγt/Tbet (n=2) mice (Data are representative of 3 independent experiments). f Mapping of the reads detected by scRNA-seq along the Rorc locus in tdRFP+ ILCs from SILP of Id2creERT2, Id2iΔRORγt, Id2iΔRORγt/RORα and Id2iΔRORγt/Tbet mice. The localisation of LoxP-flanked exons is depicted by a grey box. Each data point on graphs is a mouse, horizontal bars in ‘b’ denote the median, bars in ‘d’ and ‘e’ denote the mean, with error bars denoting the SEM. Statistical significance in ‘b’ was tested using an ordinary one-way ANOVA with Dunnett’s test for multiple comparisons. ****P ≤ 0.0001.

Source data

Extended Data Fig. 3 Expression of key genes associated with ILC superclusters.

a t-SNE plot showing ILC superclusters within all samples (Id2creERT2, Id2iΔRORγt, Id2iΔRORγt/RORα and Id2iΔRORγt/Tbet mice). b Gene expression (ALRA-imputed values) of core ILC genes unique and shared amongst ILC superclusters.

Extended Data Fig. 4 Binary regulon activity within ILC clusters.

a t-SNE plots generated from transcriptome (normalised and log-transformed counts) and b binary regulon activity (On/Off) generated via SCENIC analysis of cells within the 4 samples. c Heatmap of the mean binary regulon activity for all cells within each of the 11 transcriptomically-defined clusters. Regulons were filtered to exclude those which were rarely (< 4%) or constitutively (> 60%) active across all cells in the dataset. d Regulon specificity score for each of the 5 superclusters generated from regulon enrichment scores in each cell calculated using AUCell step of the SCENIC pipeline. Top 8 most specific regulons in each supercluster, relative to the entire dataset, are highlighted in red. e Gata3, Rorc and Tbx21 mRNA expression (ALRA-imputed values, top) and binary regulon activity (bottom).

Extended Data Fig. 5 RORγt and RORα combine to regulate the chromatin landscape of ILC3s.

To further investigate how transcriptomic changes after TF deletion might be influenced by alterations to the chromatin landscape, FACS-isolated tdRFP+ KLRG1- ILCs from Id2creERT2, Id2iΔRORγt, Id2iΔRORγt/RORα and Id2iΔRORγt/Tbet mice were assessed by ATAC-seq. a Principal component analysis of all samples using merged MACS2 broad peaks quantitated with size factor normalized reads per million (RPM). b Heat map showing all initial comparison of 10,910 significant changes in areas of open chromatin. c Primary genomic location of peaks and relative peak abundance around transcription start sites. d Heatmap of differentially enriched peaks between all knockouts (n = 2306) based on median centered size factor normalized log2RPM values, also showing the abundance of ROR response elements (RORE) and TBX21 motifs. Groups A to E refer to the distinct patterns of changes to open chromatin across the mouse models. e UCSC genome browser display of mouse Id2, Il17a/Il17f and Il22 loci with average traces from ILCs from Id2creERT2 (blue), Id2iΔRORγt (yellow), Id2iΔRORγt/RORα (red) and Id2iΔRORγt/Tbet (green) mice, alongside RORγt (49) and T-bet (50) ChIP-seq data, with RORE and TBX21 motifs marked.

Extended Data Fig. 6 Altered metabolism profile in LTi-like ILC3s lacking RORγt and RORα.

a Relative expression (z-score) of key genes associated with LTi-like ILC3 identification and function in clusters 2-4 (LTi-like supercluster) versus clusters 5 (NCR+ ILC3) and 9 (ILC1-like cells enriched in Id2iΔRORγt/RORα mice). b Representative expression of CCR6, c-Kit and NRP1 by NCR- ILC3s, identified as tdRFP+KLRG1-NKp46- cells from SILP of Id2creERT2, Id2iΔRORγt and Id2iΔRORγt/RORα mice, grey shaded histograms show expression by NCR+ ILC3s as a control. c Representative contour plots showing expression of MHCII by NCR- ILC3s from the SILP of Id2creERT2, Id2iΔRORγt and Id2iΔRORγt/RORα mice. d Heatmap (z-score) of differentially expressed genes identified via a three way comparison of LTi-like ILC3 cells (clusters 2-4 combined) derived from Id2creERT2, Id2iΔRORγt and Id2iΔRORγt/RORα mice and selected on the basis of a mean expression ≥0.1 fpkm, False Discovery Rate ≤0.001 and Fold Change in cluster average Z-score > +/− 0.4 . e Relative expression (z-score) of selected metabolism associated genes in total LTi-like ILC3 cells (cluster 2-4 combined) derived from Id2creERT2, Id2iΔRORγt and Id2iΔRORγt/RORα mice. f Representative protein expression of ARG1 in tdRFP+ LTi-like ILC3s from SILP of Id2creERT2, Id2iΔRORγt and Id2iΔRORγt/RORα mice. g Enumeration of % ARG1+ LTi-like ILCs and of the mean fluorescence intensity (MFI) of ARG1 expression assessed in ‘f’ (Id2cre: n=3; Id2iΔRORγt: n=3; Id2iΔRORγt/RORα: n=2. Data from one representative experiment out of two independent experiments). Each data point on the graph is a mouse, horizontal bars denote the median. Statistical significance was tested using an ordinary one-way ANOVA with Tukey’s test for multiple comparisons. ns = not significant, *P ≤ 0.05, **P ≤ 0.01, ***P ≤ 0.001, ****P ≤ 0.0001. Exact P values are provided in the source data.

Source data

Extended Data Fig. 7 Continued RORα expression maintains optimum Arg1 expression by ILC2s.

a Differentially expressed genes amongst ILC2 clusters in Id2creERT2 and Id2iΔRORγt/RORα. Log fold change ≥1.0, mean expression ≥0.1 fpkm, False Discovery Rate ≤0.02. Core ILC2 genes (no significant change) included for comparison. b Violin plots showing Arg1 expression (normalised and log-transformed counts) within the ILC2 cluster identified in Id2creERT2, Id2iΔRORγt, Id2iΔRORγt/RORα and Id2iΔRORγt/Tbet samples. c Representative protein expression of ARG1 in tdRFP+ ILC2s (KLRG1+ ILCs) from SILP of control Id2creERT2 and Id2iΔRORγt/RORα mice. d Percentage of ARG1+ ILC2s (tdRFP+ KLRG1+ ILCs) and MFI of ARG1 expression by ARG1+ ILC2s isolated from the SILP of control Id2creERT2 and Id2iΔRORγt/RORα mice (Id2creERT2: n = 5; Id2iΔRORγt/RORα: n = 3). Each data point on graphs is a mouse, horizontal bars denote the median. Statistical significance was tested using unpaired two-tailed Student’s t tests, *P ≤ 0.05. Exact P values are provided in the source data.

Source data

Extended Data Fig. 8 Validation of transcriptomic changes induced by deletion of RORγt and RORα.

Relative expression of S100a4 (a) and Xcl1 (b) transcripts determined by qPCR analysis of cDNA prepared from tdRFP+ ILCs sorted from SILP of Id2creERT2 (n = 5), Id2iΔRORγt (n = 7), Id2iΔRORγt/RORα (n = 4) and Id2iΔRORγt/Tbet (n = 2) mice (Data are representative of 3 independent experiments). c Representative expression of CD94, NKG2, CD244 and NKp46, versus NK1.1, by tdRFP+ ILCs from SILP of Id2creERT2, Id2iΔRORγt, Id2iΔRORγt/RORα and Id2iΔRORγt/Tbet mice. d UCSC genome browser display of mouse Klrc1, Klrd1, Gzma and Ccl3/4 loci with average traces from ILCs from Id2creERT2 (blue), Id2iΔRORγt (yellow), Id2iΔRORγt/RORα (red) and Id2iΔRORγt/Tbet (green) mice, alongside RORγt (49) and T-bet (50) ChIP-seq data, with RORE and TBX21 motifs marked. e Violin plots of Ikzf1 and Ikzf3 expression across clusters 1-11. f Representative expression of Aiolos by ILC2s, NCR+ ILC3s, ILC1s/ex-ILC3s, DN ILC3s and LTi-like ILC3s using flow cytometry. g Percentage of ILC2s, NCR+ ILC3s, ILC1s/ex-ILC3s, DN ILC3s and LTi-like ILC3s expressing Aiolos (n = 4 for each ILC group/subset, data are representative of two independent experiments). Each data point on graphs is a mouse, horizontal bars in ‘a’ and ‘b’ denote the mean, with error bars denoting the SEM; horizontal bars in ‘g’ denote the median. Statistical significance in ‘g’ was tested using an ordinary one-way ANOVA with Tukey’s test for multiple comparisons. ns = not significant, **P ≤ 0.01, ****P ≤ 0.0001. Exact P values are provided in the source data.

Source data

Extended Data Fig. 9 Id2iΔRORγt/Tbet mice are able to control and clear Citrobacter rodentium infection.

To functionally test whether protective intestinal mechanisms were maintained in Id2iΔRORγt/Tbet mice, infection with Citrobacter rodentium was used. a C. rodentium counts expressed as CFU/g feces over the time course of infection in Id2iΔRORγt (n = 4) and Id2iΔRORγt/Tbet (n = 5 or 6 depending on the time point) mice versus littermate ‘no cre’ controls (n = 8; to ensure comparable microbiome). b Relative expression of RegIIIb and RegIIIg in Id2iΔRORγt and Id2iΔRORγt/Tbet mice versus littermate ‘no cre’ controls at 21 dpi with C. rodentium (no cre control: n = 8; Id2iΔRORγt: n = 4; Id2iΔRORγt/Tbet: n = 6). c Histological analysis of colon isolated from Id2creERT2, Id2iΔRORγt and Id2iΔRORγt/Tbet mice at 15 dpi with C. rodentium (n = 3 for each strain, one representative picture from one representative animal per strain). Each data point on graphs is a mouse, bars denote the mean with error bars denoting the SEM. Statistical significance was tested using a one-sided Kruskal-Wallis test with a Dunn’s test for multiple comparisons. *P ≤ 0.05. Exact P values are provided in the source data.

Source data

Supplementary information

Source data

Source Data Fig. 1

Detail of all individual raw data used in graphs in Fig. 1, and exact P values for statistical analyses.

Source Data Fig. 4

Detail of all individual raw data used in graphs in Fig. 4.

Source Data Fig. 5

Detail of all individual raw data used in graphs in Fig. 5, and exact P values for statistical analyses.

Source Data Extended Data Fig. 1

Detail of all individual raw data used in graphs in Extended Data Fig. 1, and exact P values for statistical analyses.

Source Data Extended Data Fig. 2

Detail of all individual raw data used in graphs in Extended Data Fig. 2, and exact P values for statistical analyses.

Source Data Extended Data Fig. 6

Detail of all individual raw data used in graphs in Extended Data Fig. 6, and exact P values for statistical analyses.

Source Data Extended Data Fig. 7

Detail of all individual raw data used in graphs in Extended Data Fig. 7, and exact P values for statistical analyses.

Source Data Extended Data Fig. 8

Detail of all individual raw data used in graphs in Extended Data Fig. 8, and exact P values for statistical analyses.

Source Data Extended Data Fig. 9

Detail of all individual raw data used in graphs in Extended Data Fig. 9, and exact P values for statistical analyses.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Fiancette, R., Finlay, C.M., Willis, C. et al. Reciprocal transcription factor networks govern tissue-resident ILC3 subset function and identity. Nat Immunol 22, 1245–1255 (2021). https://doi.org/10.1038/s41590-021-01024-x

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-021-01024-x

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