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:

Multiscale 3D genome organization underlies ILC2 ontogenesis and allergic airway inflammation

Abstract

Innate lymphoid cells (ILCs) are well-characterized immune cells that play key roles in host defense and tissue homeostasis. Yet, how the three-dimensional (3D) genome organization underlies the development and functions of ILCs is unknown. Herein, we carried out an integrative analysis of the 3D genome structure, chromatin accessibility and gene expression in mature ILCs. Our results revealed that the local 3D configuration of the genome is rewired specifically at loci associated with ILC biology to promote their development and functional differentiation. Importantly, we demonstrated that the ontogenesis of ILC2s and the progression of allergic airway inflammation are determined by a unique local 3D configuration of the region containing the ILC-lineage-defining factor Id2, which is characterized by multiple interactions between the Id2 promoter and distal regulatory elements bound by the transcription factors GATA-3 and RORα, unveiling the mechanism whereby the Id2 expression is specifically controlled in group 2 ILCs.

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: Large-scale 3D genome organization in ILCs.
Fig. 2: Differential local 3D structure at key loci correlates with the gene expression programs of each ILC subset.
Fig. 3: Multiple long-distance interactions between the Id2 promoter and LCR1 specifically control ILC2 homeostasis or development.
Fig. 4: Multiple long-distance interactions between the Id2 promoter and LCR1 determine early ILC2 development.
Fig. 5: LCR1 specifically controls group 2 ILC development through Id2 expression regulation.
Fig. 6: LCR1 determines the progression of AAI.

Similar content being viewed by others

Data availability

All bulk RNA-seq, ATAC-seq, Hi-C and scRNA-seq datasets have been uploaded to the GEO repository (accession no. GSE191312). The following published datasets were used: GEO GSE11187123, GEO GSE776954, GEO GSE1307759, GEO GSE16954241, GEO GSE14674522 and GEO GSE7942212. Source data are provided with this paper.

References

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

    Article  CAS  Google Scholar 

  2. Robinette, M. L. et al. Transcriptional programs define molecular characteristics of innate lymphoid cell classes and subsets. Nat. Immunol. 16, 306–317 (2015).

    Article  CAS  Google Scholar 

  3. Gury-BenAri, M. et al. The spectrum and regulatory landscape of intestinal innate lymphoid cells are shaped by the microbiome. Cell 166, 1231–1246.e13 (2016).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  5. Dixon, J. R. et al. Chromatin architecture reorganization during stem cell differentiation. Nature 518, 331–336 (2015).

    Article  CAS  Google Scholar 

  6. Fortin, J.-P. & Hansen, K. D. Reconstructing A/B compartments as revealed by Hi-C using long-range correlations in epigenetic data. Genome Biol. 16, 180 (2015).

    Article  Google Scholar 

  7. Dixon, J. R. et al. Topological domains in mammalian genomes identified by analysis of chromatin interactions. Nature 485, 376–380 (2012).

    Article  CAS  Google Scholar 

  8. Allan, D. S. J. et al. An in vitro model of innate lymphoid cell function and differentiation. Mucosal Immunol. 8, 340–351 (2015).

    Article  CAS  Google Scholar 

  9. 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  Google Scholar 

  10. Parker, M. E. et al. c-Maf regulates the plasticity of group 3 innate lymphoid cells by restraining the type 1 program. J. Exp. Med. 217, e20191030 (2020).

    Article  Google Scholar 

  11. Lieberman-Aiden, E. et al. Comprehensive mapping of long-range interactions reveals folding principles of the human genome. Science 326, 289–293 (2009).

    Article  CAS  Google Scholar 

  12. Hu, G. et al. Transformation of accessible chromatin and 3D nucleome underlies lineage commitment of early T cells. Immunity 48, 227–242.e8 (2018).

    Article  CAS  Google Scholar 

  13. Gibcus, J. H. & Dekker, J. The hierarchy of the 3D genome. Mol. Cell 49, 773–782 (2013).

    Article  CAS  Google Scholar 

  14. Fudenberg, G. et al. Formation of chromosomal domains by loop extrusion. Cell Rep. 15, 2038–2049 (2016).

    Article  CAS  Google Scholar 

  15. Rowley, M. J. & Corces, V. G. Organizational principles of 3D genome architecture. Nat. Rev. Genet. 19, 789–800 (2018).

    Article  CAS  Google Scholar 

  16. Chandra, T. et al. Global reorganization of the nuclear landscape in senescent cells. Cell Rep. 10, 471–483 (2015).

    Article  CAS  Google Scholar 

  17. Vian, L. et al. The energetics and physiological impact of cohesin extrusion. Cell 173, 1165–1178.e20 (2018).

    Article  CAS  Google Scholar 

  18. Yoon, S., Chandra, A. & Vahedi, G. Stripenn detects architectural stripes from chromatin conformation data using computer vision. Nat. Commun. 13, 1602 (2022).

    Article  CAS  Google Scholar 

  19. Engel, I. & Murre, C. The function of E- and Id proteins in lymphocyte development. Nat. Rev. Immunol. 1, 193–199 (2001).

    Article  CAS  Google Scholar 

  20. Mowel, W. K. et al. Group 1 innate lymphoid cell lineage identity is determined by a cis-regulatory element marked by a long non-coding RNA. Immunity 47, 435–449.e8 (2017).

    Article  CAS  Google Scholar 

  21. 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  Google Scholar 

  22. Ferreira, A. C. F. et al. RORα is a critical checkpoint for T cell and ILC2 commitment in the embryonic thymus. Nat. Immunol. 22, 166–178 (2021).

    Article  CAS  Google Scholar 

  23. Miyamoto, C. et al. Runx/Cbfβ complexes protect group 2 innate lymphoid cells from exhausted-like hyporesponsiveness during allergic airway inflammation. Nat. Commun. 10, 447 (2019).

    Article  CAS  Google Scholar 

  24. Wong, S. H. et al. Transcription factor RORα is critical for nuocyte development. Nat. Immunol. 13, 229–236 (2012).

    Article  CAS  Google Scholar 

  25. Mjösberg, J. et al. The transcription factor GATA3 is essential for the function of human type 2 innate lymphoid cells. Immunity 37, 649–659 (2012).

    Article  Google Scholar 

  26. KleinJan, A. et al. Enforced expression of Gata3 in T cells and group 2 innate lymphoid cells increases susceptibility to allergic airway inflammation in mice. J. Immunol. 192, 1385–1394 (2014).

    Article  CAS  Google Scholar 

  27. Bando, J. K., Nussbaum, J. C., Liang, H.-E. & Locksley, R. M. Type 2 innate lymphoid cells constitutively express arginase-I in the naïve and inflamed lung. J. Leukoc. Biol. 94, 877–884 (2013).

    Article  CAS  Google Scholar 

  28. Bando, J. K., Liang, H.-E. & Locksley, R. M. Identification and distribution of developing innate lymphoid cells in the fetal mouse intestine. Nat. Immunol. 16, 153–160 (2015).

    Article  CAS  Google Scholar 

  29. Nussbaum, J. C. et al. Type 2 innate lymphoid cells control eosinophil homeostasis. Nature 502, 245–248 (2013).

    Article  CAS  Google Scholar 

  30. Carlens, J. et al. Common γ-chain-dependent signals confer selective survival of eosinophils in the murine small intestine. J. Immunol. 183, 5600–5607 (2009).

    Article  CAS  Google Scholar 

  31. Zook, E. C. & Kee, B. L. Development of innate lymphoid cells. Nat. Immunol. 17, 775–782 (2016).

    Article  CAS  Google Scholar 

  32. Michieletto, M. F. & Henao-Mejia, J. Ontogeny and heterogeneity of innate lymphoid cells and the noncoding genome. Immunol. Rev. 300, 152–166 (2021).

    Article  CAS  Google Scholar 

  33. Martinez-Gonzalez, I., Steer, C. A. & Takei, F. Lung ILC2s link innate and adaptive responses in allergic inflammation. Trends Immunol. 36, 189–195 (2015).

    Article  CAS  Google Scholar 

  34. Seillet, C. et al. Deciphering the innate lymphoid cell transcriptional program. Cell Rep. 17, 436–447 (2016).

    Article  CAS  Google Scholar 

  35. Xu, W. et al. An Id2RFP-reporter mouse redefines innate lymphoid cell precursor potentials. Immunity 50, 1054–1068.e3 (2019).

    Article  CAS  Google Scholar 

  36. 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.e7 (2019).

    Article  CAS  Google Scholar 

  37. Constantinides, M. G., McDonald, B. D., Verhoef, P. A. & Bendelac, A. A committed precursor to innate lymphoid cells. Nature 508, 397–401 (2014).

    Article  CAS  Google Scholar 

  38. Hammad, H. & Lambrecht, B. N. The basic immunology of asthma. Cell 184, 1469–1485 (2021).

    Article  CAS  Google Scholar 

  39. Barlow, J. L. et al. IL-33 is more potent than IL-25 in provoking IL-13-producing nuocytes (type 2 innate lymphoid cells) and airway contraction. J. Allergy Clin. Immunol. 132, 933–941 (2013).

    Article  CAS  Google Scholar 

  40. Barlow, J. L. et al. Innate IL-13-producing nuocytes arise during allergic lung inflammation and contribute to airways hyperreactivity. J. Allergy Clin. Immunol. 129, 191–198.e4 (2012).

    Article  CAS  Google Scholar 

  41. Kasal, D. N. et al. A Gata3 enhancer necessary for ILC2 development and function. Proc. Natl Acad. Sci. USA 118, e2106311118 (2021).

    Article  CAS  Google Scholar 

  42. Halim, T. Y. F. et al. Group 2 innate lymphoid cells are critical for the initiation of adaptive T helper 2 cell-mediated allergic lung inflammation. Immunity 40, 425–435 (2014).

    Article  CAS  Google Scholar 

  43. Huang, Y. et al. IL-25-responsive, lineage-negative KLRG1hi cells are multipotential ‘inflammatory’ type 2 innate lymphoid cells. Nat. Immunol. 16, 161–169 (2015).

    Article  CAS  Google Scholar 

  44. Xiao, Q. et al. PPARγ enhances ILC2 function during allergic airway inflammation via transcription regulation of ST2. Mucosal Immunol. 14, 468–478 (2021).

    Article  CAS  Google Scholar 

  45. Yang, Q. et al. T cell factor 1 is required for group 2 innate lymphoid cell generation. Immunity 38, 694–704 (2013).

    Article  CAS  Google Scholar 

  46. Toki, S. et al. TSLP and IL-33 reciprocally promote each other’s lung protein expression and ILC2 receptor expression to enhance innate type-2 airway inflammation. Allergy 75, 1606–1617 (2020).

    Article  CAS  Google Scholar 

  47. Lin, Y. C. et al. Global changes in the nuclear positioning of genes and intra- and interdomain genomic interactions that orchestrate B cell fate. Nat. Immunol. 13, 1196–1204 (2012).

    Article  CAS  Google Scholar 

  48. van Schoonhoven, A., Huylebroeck, D., Hendriks, R. W. & Stadhouders, R. 3D genome organization during lymphocyte development and activation. Briefings Funct. Genomics 19, 71–82 (2020).

    Article  Google Scholar 

  49. Phillips-Cremins, J. E. et al. Architectural protein subclasses shape 3D organization of genomes during lineage commitment. Cell 153, 1281–1295 (2013).

    Article  CAS  Google Scholar 

  50. Claringbould, A. & Zaugg, J. B. Enhancers in disease: molecular basis and emerging treatment strategies. Trends Mol. Med. 27, 1060–1073 (2021).

    Article  CAS  Google Scholar 

  51. Laffont, S. et al. Androgen signaling negatively controls group 2 innate lymphoid cells. J. Exp. Med. 214, 1581–1592 (2017).

    Article  CAS  Google Scholar 

  52. Servant, N. et al. HiC-Pro: an optimized and flexible pipeline for Hi-C data processing. Genome Biol. 16, 259 (2015).

    Article  Google Scholar 

  53. Abdennur, N. & Mirny, L. A. Cooler: scalable storage for Hi-C data and other genomically labeled arrays. Bioinformatics 36, 311–316 (2020).

    Article  CAS  Google Scholar 

  54. Wolff, J. et al. Galaxy HiCExplorer 3: a web server for reproducible Hi-C, capture Hi-C and single-cell Hi-C data analysis, quality control and visualization. Nucleic Acids Res. 48, W177–W184 (2020).

    Article  CAS  Google Scholar 

  55. Wolff, J. et al. Galaxy HiCExplorer: a web server for reproducible Hi-C data analysis, quality control and visualization. Nucleic Acids Res. 46, W11–W16 (2018).

    Article  CAS  Google Scholar 

  56. Ramírez, F. et al. High-resolution TADs reveal DNA sequences underlying genome organization in flies. Nat. Commun. 9, 189 (2018).

    Article  Google Scholar 

  57. Roayaei Ardakany, A., Gezer, H. T., Lonardi, S. & Ay, F. Mustache: multi-scale detection of chromatin loops from Hi-C and Micro-C maps using scale-space representation. Genome Biol. 21, 256 (2020).

    Article  Google Scholar 

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

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  60. 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).

    Article  CAS  Google Scholar 

  61. Fasolino, M. et al. Genetic variation in type 1 diabetes reconfigures the 3D chromatin organization of T cells and alters gene expression. Immunity 52, 257–274.e11 (2020).

    Article  CAS  Google Scholar 

  62. Zhang, Y. et al. Model-based analysis of ChIP-Seq (MACS). Genome Biol. 9, R137 (2008).

    Article  Google Scholar 

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

    Article  Google Scholar 

  64. Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).

    Article  Google Scholar 

  65. Hafemeister, C. & Satija, R. Normalization and variance stabilization of single-cell RNA-seq data using regularized negative binomial regression. Genome Biol. 20, 296 (2019).

    Article  CAS  Google Scholar 

  66. Satija, R., Farrell, J. A., Gennert, D., Schier, A. F. & Regev, A. Spatial reconstruction of single-cell gene expression data. Nat. Biotechnol. 33, 495–502 (2015).

    Article  CAS  Google Scholar 

  67. Trapnell, C. et al. The dynamics and regulators of cell fate decisions are revealed by pseudotemporal ordering of single cells. Nat. Biotechnol. 32, 381–386 (2014).

    Article  CAS  Google Scholar 

  68. Mi, H. et al. Protocol Update for large-scale genome and gene function analysis with the PANTHER classification system (v.14.0). Nat. Protoc. 14, 703–721 (2019).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The work in this manuscript was supported by funds from CHOP and the NIH grant R01HL136572 (J. H-M.); the Burroughs Welcome Fund, the Chang Zuckerberg Initiative Award, and the NIH grant R01AI168240 (J. H-M. and G.V.); the PEW Charitable Trust (J. H-M. and J.J. T-C.); the NIH grants UC4 DK112217, U01DK11221702A1, R01HL145754, U01DK127768, U01DA052715 (G.V); and F31DK122677 (M.T.J.).

Author information

Authors and Affiliations

Authors

Contributions

These studies were conceptualized by M.F.M., W.K.M., G.V. and J.H.-M. All experiments were designed, performed and analyzed by M.F.M. or W.K.M. with some help from M.L.C., M.T.J. and J.M.W. J.J.T.-C performed all of the bioinformatic analysis with the help of S.Y. for the stripe analysis and of P.L., A.W. and C.A.T. for scRNA-seq analysis. A.C. generated Hi-C, RNA-seq and ATAC-seq libraries and quality controls. L.J. generated the novel mouse strains. J.H.-M. and G.V. supervised the biological and computational parts of this project, respectively. M.F.M., J.J.T.-C. and J.H.-M. wrote the manuscript. G.V. edited the manuscript.

Corresponding authors

Correspondence to Golnaz Vahedi or Jorge Henao-Mejia.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks Shigeo Koyasu and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Peer reviewer reports are available. 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 Comparison of accessibility between the MNK3 cell line and primary ILC3.

a, Venn diagram representation of the differential chromatin accessibility between ATAC-seq peaks of MNK3 and small intestine (SI) CD4+ ILC3, and SI NCR+ ILC3 cells. Absolute numbers of peaks differentially open (red) or closed (blue) in MNK3, or with similar accessibility (gray) in all samples, are represented. b, View of the chromatin accessibility (ATAC-seq signal) at the Rorc, Il17a, Il17f, Il22 and Il23r loci in small intestine (SI) CD4+ ILC3, SI NCR+ ILC3, and MNK3 cells. c, Venn diagram representation of the differential accessibility analysis results in the region displayed in the right panel. d, View of the chromatin accessibility (ATAC-seq signal) at the Id2 locus in SI CD4+ ILC3, SI NCR+ ILC3, and MNK3 cells. Absolute numbers of peaks differentially open (red) or closed (blue) in MNK3, or with similar accessibility (gray) in all samples, are shown. e, Representative flow cytometry plots showing cell viability and the expression of ILC3-specific markers (IL-22, GM-CSF, CD90.2, RORγt, IL-17 and ID2) by MNK3 cells after stimulation with IL-2, IL-23, and IL-1β. The red line represents activated MNK3 cells and the black line represents unstained MNK3 cells.

Extended Data Fig. 2 Changes in compartment distribution in ILCs are accompanied by changes in expression patterns concordant with euchromatin/heterochromatin state.

a, Fraction of regions in the A or B compartments in common lymphoid progenitors (CLP), group 1 (splenic NK cells), group 2 (bone marrow ILC2s), and group 3 (MNK3) ILC. b, Proportion of regions in CLPs that flipped compartment status in at least one ILC group (brown) or that remained unchanged in all ILC groups (gray). Sidebar indicates the proportion of flipped regions that were in the A (red) or B (blue) compartment at the CLP stage. c, Examples of compartment state distribution in the vicinity of genes with known roles in ILC biology such as Id2, Tcf7, Nfil3, Gata3, Lifr and Tox. Regions in the A and B compartment correspond to positive (red) and negative (blue) values in the PC1 bar, respectively. Each square represents 25kb. d, Percentage of expressed genes among genes within compartments that underwent similar flipping from CLP. The Y-axis represents the different combinations of final compartment states in ILC for flipping regions originally in the status indicated on the left. Parenthetical numbers indicate the total number of genes overlapping the corresponding group of regions. Expressed genes correspond to genes with average RPKM > 1. RPKM: Reads per kilobase of transcripts per million mapped reads. Letter triplets represent the compartment status in group 1, group 2 and group 3 ILC, respectively. e, (left) Heatmap of the compartment status in ILCs of flipping regions that were in the B compartment in CLPs. Regions in the A and B compartment correspond to positive (red) and negative (blue) values in the PC1 bar. (right) Comparison of gene expression levels between group 1 (blue boxes), group 2 (green boxes) and group 3 (red boxes) ILCs of groups of genes undergoing similar compartment flipping from CLP. The Y-axis represents the different combinations of final compartment states in ILC. RPKM: Reads per kilobase of transcripts per million mapped reads. Letter triplets represent the compartment status in group 1, group 2 and group 3 ILC, respectively. Box shows dataset quartiles and whiskers the distribution range. Dots represent outliers as determined by the inter-quartile range. f, (left) Heatmap of the compartment status in ILCs of flipping regions that were in the A compartment in CLP. Regions in the A and B compartment correspond to positive (red) and negative (blue) values in the PC1 bar. (right) Comparison of gene expression levels between group 1 (blue boxes), group 2 (green boxes) and group 3 (red boxes) ILCs of groups of genes undergoing similar compartment flipping from CLP. The Y-axis represents the different combinations of final compartment states in ILC. RPKM: Reads per kilobase of transcripts per million mapped reads. Letter triplets represent the compartment status in group 1, group 2 and group 3 ILC, respectively. Box shows dataset quartiles and whiskers the distribution range. Dots represent outliers as determined by the inter-quartile range. g, Table with the p-values (Mann-Whitney U two-tailed test) for Fig. 1e,f of the pairwise comparison between ILC of expression levels of genes that underwent similar compartment flippings from the A or B compartment in CLP. Letter triplets represent the compartment status in group 1, group 2 and group 3 ILC, respectively.

Extended Data Fig. 3 Local structures of the 3D genome architecture are comparable at the sub-megabase scale and correlate with gene expression in each ILC group.

a, Comparison of the pileup plots computed over conserved vs ILC-specific (left) boundary elements and (right) Topological Associated Domains (TADs) in each group of ILCs. Numbers in the corner indicate average signal strength. b, Boxplot of cross-boundary ratios of TADs for group 1 (blue), group 2 (green) and group 3 (red) ILCs. Box shows dataset quartiles and whiskers the distribution range. Dots represent outliers as determined by the inter-quartile range. c, Boxplot of the size distribution of TADs defined by boundaries detected in each ILC (blue; group 1 ILC, green; group 2 ILC, red; group 3 ILC) and by boundaries conserved across ILCs (white). Box shows dataset quartiles and whiskers the distribution range. Dots represent outliers as determined by the inter-quartile range. d, Expression levels of genes with critical roles in ILC biology in group 1 (blue bars), group 2 (green bars) and group 3 (red bars) ILCs obtained by RNA-sequencing. RPKM: Reads per kilobase of transcripts per million mapped reads. e, Pileup plots of stripes detected in group 1 (left), group 2 (middle) and group 3 ILC (right). Axes in units of mean feature (stripes) size as computed for each group ILC.

Extended Data Fig. 4 3D architecture, chromatin accessibility, and enhancer activity patterns at the Id2 locus in each ILC group.

a, Visualization of the contact heatmap at the Id2 locus in (top) group 1, (middle) group 2, and (bottom) group 3 ILCs. Chromatin accessible regions determined by ATAC-seq (gray track), and H3K27ac deposition (blue track) determined by ChIP-seq (group1 and group 2) or Cut & Run (group 3) in each group of ILCs. Colored arcs represent loops detected in the corresponding contact map (blue; group 1 ILC, green; group 2 ILC, red; group 3 ILC). The location of the Id2 promoter is indicated with an extended vertical dashed line. Dotted and solid lines represent sub-TAD and TAD boundaries, respectively. Arrowheads indicate the position of boundaries conserved across all ILCs. Scale bar in heatmap represents normalized contact frequency.

Extended Data Fig. 5 The locus control region 1 (LCR1), but not LCR2, is specifically required for the development of all tissue resident ILC2s.

a, Representative transcription factor binding motifs enriched in ATAC-seq peaks (peaks from all ILCs pooled) within LCR1 (red section) and LCR2 (blue section). Numbers represent log odds detection scores as computed by HOMER. b, (top) Quantification of absolute numbers of splenic CD4+ T cells, CD8+ T cells, and NK cells in their different maturation states in LCR1+/+ and LCR1/ mice. (bottom) Quantification of absolute numbers of splenic naive (CD62L+, CD44) and effector (CD62L, CD44+) CD4+ and CD8+ T cell populations, as well as CD8+ and CD11b+ dendritic cells. Splenic T cell numbers are one representative experiment that was repeated six times (n = 8 LCR1+/+ and 8 LCR1−/−). Splenic NK cell numbers are a pool of two independent experiments. This experiment was repeated four (n = 9 LCR1+/+ and 9 LCR1−/−). Tregs represents a pool of two independent experiments (n = 9 LCR1+/+ and 9 LCR1−/−) and was repeated three times. Myeloid cell numbers represent one experiment (n = 3 LCR1+/+ and 3 LCR1−/−). Error bars = SEM; and p-values: ns = not significant (Treg panel: Mann-Whitney U two-tailed test; Splenic T cells NK cell and Myeloid cell: Two-way ANOVA with multiple comparison and Bonferroni correction). c, Quantification of absolute numbers of splenic CD4+ T cells, CD8+ T cells, and NK cells in their different maturation state in LCR2+/+ and LCR2/ mice (n = 4 LCR2+/+ and 4 LCR2−/−). Data represents one experiment that was repeated two times. Error bars = SEM; and p-values: ns = not significant, * = p ≤ 0.05, ** = p ≤ 0.01, *** = p ≤ 0.0005 (Two-way ANOVA with multiple comparison and Bonferroni correction). d, Representative flow cytometry gating strategy used for the identification of group 1 (NK cell), group 2 (ILC2s), and group 3 (ILC3s) ILC in small intestine lamina propria (siLP) of LCR1+/+ and LCR1/ animals. e, Quantification of lungs and bone marrow (BM) ILC2s numbers of Arginase1 reporter mouse (Arg1YFP) and Arg1YFP; LCR1−/− mice (Live, CD45+, Lineage, CD127+, CD90.2+, ST2+, YFP+) at steady state. Data is a pool of two independent experiments and were repeated two times. Each dot represents an individual mouse (n = 7 Arg1YFP; LCR1+/+ and 7 Arg1YFP; LCR1/). Error bars = SEM; and p-values: ns = not significant, * = p ≤ 0.05, ** = p ≤ 0.01, *** = p ≤ 0.0005 (Mann-Whitney U two-tailed test). f, Quantification of IL-5 in the serum of LCR1+/+ and LCR1/ mice at steady state by cytometry bead assay (CBA). Data is representative of two experiments. Each dot represents an individual mouse, (n = 7 LCR1+/+ and 6 LCR1/). Error bars = SEM; and p-values: ns = not significant, * = p ≤ 0.05 (Mann-Whitney U two-tailed test). g, (top) Representative flow cytometry gating strategy used for the identification eosinophils in the blood, spleen, and lungs of LCR1+/+ and LCR1/ mice at steady state and (bottom) their subsequent quantification. Data represents one experiment. Each dot represents an individual mouse, (n = 4 LCR1+/+ and 4 LCR1/). Error bars = SEM; and p-values: ns = not significant, * = p ≤ 0.05 (Mann-Whitney U two-tailed test).

Source data

Extended Data Fig. 6 The LCR1 impairs ILC2 development through reduction of Id2 expression and skewing toward ILC1- and ILC3-like ILC progenitors.

a, (left) Expression levels of Il7r and Id2 over the Uniform Manifold Approximation and Projection of the scRNA-seq data set described in Fig. 4a. (right), Violin plot representation of the expression levels of Id2 and Il7r across all identified clusters in Fig.4a. For each cluster (horizontal axis), the expression levels in LCR1+/+ (gray) and LCR1/ (red) mice is shown. b, Hierarchical representation of similarity between the clusters identified in Fig. 4a. Similarity was measured using complete linkage over the Pearson correlation matrix between clusters. c, Gating strategy used to identify putative ILC3p in the bone marrow of LCR1+/+ and LCR1/ animals. Lin, CD127+ cells were pre-gated on Singlets, Live, CD45+ cells. d, Gating strategy for the identification of CLP: Common Lymphoid Progenitors (Live, CD45+, Lin, CD127+, CD25, ICOS, CD135+, α4β7); α-LP: alpha-Lymphoid Progenitors (Live, CD45+, Lin, CD127+, ICOS, CD135+, α4β7+, Id2+, CD25, CD117+); ChILP: Common helper Innate Lymphoid Progenitors (Live, CD45+, Lin, CD127+, CD135, α4β7+, CD25); ILCp: Innate Lymphoid Cell precursors (Live, CD45+, Lin, CD127+, ICOS, CD25, CD117+, Sca1, α4β7+, ID2+); ILC2p: group 2 Innate Lymphoid Cell precursors (Live, CD45+, Lin, CD127+, CD90.2+, ST2+ or CD25+, ICOS+). e, Quantification of ILC2 progenitors in the fetal liver (FL), fetal intestine (FI) or fetal lungs (FLu)of LCR1+/+ and LCR1/ E15.5 embryos. Data represents the pool of two independent experiments. Each dot represents an individual embryo (numbers: FL: = 21 LCR1+/+ and 20 LCR1/ ; FI and FLu: 11 LCR1+/+ and 10 LCR1/). Error bars = SEM; and p-values: ns = not significant, * = p ≤ 0.05, ** = p ≤ 0.01, *** = p ≤ 0.0005, **** = p ≤ 0.0001 (Mann-Whitney U two-tailed test). f, Volcano plot displaying upregulated (red rectangle) and downregulated (blue rectangle) genes in LCR1+/+ when compared to LCR1/ cells for clusters in Fig. 5a representing ILCp populations. g, Savitzky-Golay smoothing curve of the normalized expression levels of Tox, Bcl11b, Ets1, Tcf7, and Zbtb16 along the pseudotime axis determined in Fig. 5e. P-value corresponds to Mann-Whitney U test between smoothed signals from LCR1+/+ (black curve) and LCR1/ (red curve) mice. h, (left) Quantification of ID2 mean fluorescence intensity (MFI) in LCR2+/+ and LCR2−/− bone marrow (BM) CLP, a4b7+ CLP, aLP, ChILP, ILCp, ILC2p gated as shown in Extended Data Fig. 6d. (right) Representative histogram of ID2 expression in LCR2+/+ and LCR2−/− BM. Each dot represents an individual mouse (n = 3 LCR2+/+ and 4 LCR2/) and was observed in 2 independent experiments. Error bars = SEM; and p-values: ns = not significant, * = p ≤ 0.05, ** = p ≤ 0.01 (Two-way ANOVA with multiple comparison and Bonferroni correction). i, Representative FACS plots and histograms of GFP expression in wild-type (CD45.1+) or LCR1/ CD45.2 BM previously transduced with retroviral particles encoding an empty vector expressing the GFP (empty_RV - green) or retroviral particles encoding Id2 and the GFP (Id2_RV – blue), at least eight weeks post engraftment in CD45.1+ hosts. Each dot represents an individual mouse (n = 8 LCR1/ empty_RV; 12 LCR1/ Id2_RV). Error bars = SEM; and p-values: ns = not significant, * = p ≤ 0.05, ** = p ≤ 0.01 (Two-way ANOVA with multiple comparison and Bonferroni correction).

Source data

Extended Data Fig. 7 Disruption of the 3D architecture of the Id2 locus reduces ILC2 numbers in the lung but does not drastically change other immune cell populations at steady state.

a, Representative flow cytometry gating strategy used for the identification of immune cell populations in digested lungs or in bronchoalveolar lavage fluid (BALF) at steady state or after 16 days of house dust mite (HDM) challenge in LCR1+/+ and LCR1/ animals. b, Quantification of frequencies among CD45+ cells and absolute numbers of lung parenchyma ILC2s in LCR1+/+ and LCR1/ mice at steady state, (n = 8 LCR1+/+ and 8 LCR1/). Data are representative of one experiment that was repeated five times. Error bars = SEM; and p-values: ns = not significant, * = p ≤ 0.05, ** = p ≤ 0.01, *** = p ≤ 0.0005 (Mann-Whitney U two-tailed test). c, Quantification of total number of cells of the indicated immune cell populations in Extended Data Fig. 7a in the lung parenchyma of LCR1+/+ and LCR1/ mice at steady state, (n = 12 LCR1+/+ and 12 LCR1/). Data are a pool of two independent experiments, except CD4+ and CD8+ TRM which is a representative experiment. (n = 8 LCR1+/+ and 8 LCR1−/−). Error bars = SEM; and p-values: ns = not significant, * = p ≤ 0.05 (Mann-Whitney U two-tailed test or Two-way ANOVA with multiple comparison and Bonferroni correction).

Source data

Extended Data Fig. 8 Acute intranasal papain and IL-33 challenges in LCR1-deficient mice.

a, Schematic representation of (left) the experimental acute papain or (right) recombinant IL-33 (rIL-33) challenges. Mice were challenged intranasally with either 30mg of papain (in 20mL of PBS) or with 200ng of rIL-33 (in 20ml of PBS) for four consecutive days. Euthanasia was carried out 24 hours after the last challenge. Lung parenchyma ILC2s and eosinophil numbers were assessed by flow cytometry. b, Flow cytometry gating strategy used for the identification of lung parenchyma ILC2s and eosinophils in LCR1+/+ and LCR1−/− mice challenged with papain or LCR1+/+ mice treated with PBS (20mL). Cells were pre-gated on SSC/FSC and Singlets. c, Quantification of ILC2 frequency among CD45+ cells, ILC2 and eosinophil numbers in LCR1+/+ and LCR1−/− mice challenged with papain, or LCR1+/+ mice treated with PBS (20mL). Data represents one experiment with males (green dots; n = 4 LCR1+/+ PBS, 4 LCR1+/+, and 4 LCR1−/− papain – 11.4-12.2-week-old). Error bars = SEM; and p-values: ns = not significant, * = p ≤ 0.05, ** = p ≤ 0.01, *** = p ≤ 0.0005, **** = p ≤ 0.0001 (One-way ANOVA with multiple comparisons and Bonferroni correction). d, Flow cytometry gating strategy used for the identification of lung parenchyma ILC2s and eosinophils in LCR1+/+ and LCR1−/− mice challenged with rIL-33 (200ng) or LCR1+/+ mice treated with PBS (20mL). Cells were pre-gated on SSC/FSC and Singlets. e, Quantification of ILC2 frequency among CD45+ cells, ILC2 and Eosinophil numbers in LCR1+/+ and LCR1−/− mice challenged with rIL-33 (200ng), or LCR1+/+ mice treated with PBS (20mL). Data is representative of two independent experiments (green dots; n = 3 LCR1+/+ PBS, 3 LCR1+/+ rIL-33, and 4 LCR1−/− rIL-33 – 9.4-9.9-week-old) and was repeated two times. Error bars = SEM; and p-values: ns = not significant, * = p ≤ 0.05, ** = p ≤ 0.01, *** = p ≤ 0.0005, **** = p ≤ 0.0001 (One-way ANOVA with multiple comparisons and Bonferroni correction).

Source data

Extended Data Fig. 9 Disruption of the 3D architecture of the Id2 locus in ILC2 reduces HDM-induced allergic airway inflammation progression.

a, Quantification of NK and NKT cells numbers in the lungs of LCR1+/+ and LCR1/ mice 16 days after the initial HDM challenge. Data is a pool of two age and sex-matched independent experiments containing males (green dots; n = 5 LCR1+/+ and 5 LCR1/ – age 12.9-week-old) and females (orange dots, n = 8 LCR1+/+ and 8 LCR1/ – age 6.9-week-old). The experiments were repeated three times. Error bars = SEM; and p-values: ns = not significant, * = p ≤ 0.05, ** = p ≤ 0.01 (Mann-Whitney U two-tailed test). b, Representative histological sections of lungs stained with hematoxylin and eosin (H&E - top) or Periodic Shift Acid (PAS - bottom) of 1 LCR1+/+ mice treated with PBS. The scale represents a length of 500mM and the magnification used is 40x. c, Representative flow cytometry plot (left) and quantification (right) of IL-5+ and IL-13+ producing CD4+ Th2 cells (CD4+, GATA3+) from lung parenchyma. Cells were ex vivo stimulated with PMA and Ionomycin for 4 hours in presence of Golgi inhibitor and identified as Live, CD45+, TCRβ+, CD4+, GATA-3+ cells Data is a pool of two age and sex-matched independent experiments containing males (green dots; n = 5 LCR1+/+ and 5 LCR1/ – age 12.9-week-old) and females (orange dots, n = 8 LCR1+/+ and 8 LCR1/ – age 6.9 week-old). The experiment was repeated three times. Error bars = SEM; and p-values: ns = not significant (Mann-Whitney U two-tailed test). d, (top left) Representative flow cytometry plots of IL-13+ and IL-5+ producing naive CD4+ T cells from LCR1+/+ and LCR1/ mice polarized under Th2 conditions for 6 days. (Bottom left) Proportion of IL-13+, IL-5+ and IL-13+/IL-5+ producing Th2 cells. (Top right), Quantification of the geometric mean fluorescence intensity (gMFI) of GATA-3, and frequency of activated (CD44+) CD4+ Th2 cells at day 6 after polarization. Data is representative of three independent experiments. Each dot represents an individual mouse, (Females; n = 4 LCR1+/+ and 4 LCR1/). Error bars = SEM; and p-values: ns = not significant (Mann-Whitney U two-tailed test).

Source data

Supplementary information

Reporting Summary

Peer Review File

Supplementary Tables

Supplementary Table 1. Compartment scores of common lymphoid progenitor, group 1, 2 and 3 ILCs. Each region is annotated with the corresponding set of overlapping genes. Related to Fig. 1 and Extended Data Fig. 1. Supplementary Table 2. RPKM values of group 1, 2 and 3 ILCs bulk RNA-seq data. Related to Figs. 1 and 2 and Extended Data Figs. 1–3. Supplementary Table 3. Cross-boundary ratio of conserved TADs in group 1, 2 and 3 ILCs. Related to Fig. 2. Supplementary Table 4. Coordinates of loops called in group 1, 2 and 3 ILCs. Loops pooled together. Seventh column contains unique identifier for further reference in Supplementary Tables 5–7 and Extended Data Fig. 7. Related to Fig. 3 and Extended Data Fig. 3. Supplementary Tables 5–7. Unique identifiers of loops detected in group 1 (Supplementary Table 5), 2 (Supplementary Table 6) and 3 (Supplementary Table 7) ILCs. Related to Fig. 3 and Extended Data Fig. 3. Supplementary Tables 8–10. Output of the Stripenn algorithm for group 1 (Supplementary Table 8), 2 (Supplementary Table 9) and 3 (Supplementary Table 10) ILCs. Related to Extended Data Fig. 3. Supplementary Table 11. List of motifs identified in LCR1, LCR2 and Rroid regions found by HOMER (knownMotifs.txt file). Related to Extended Data Fig. 5. Supplementary Table 12. List of marker genes of the clusters identified by scRNA-seq. Supplementary Table 13. sgRNA used to generate LCR1/, LCR2/, Gata3_BS/, Rora_BS/ and Rora_Gata3dKO mice.

Source data

Source Data Fig. 1

Source data for the histograms present in Fig. 1.

Source Data Fig. 3

Source data for the histograms present in Fig. 3.

Source Data Fig. 4

Source data for the histograms present in Fig. 4.

Source Data Fig. 5

Source data for the histograms present in Fig. 5.

Source Data Fig. 6

Source data for the histograms present in Fig. 6.

Source Data Fig. 6

Source data for the histology images in Fig. 6.

Source Data Extended Data Fig. 5

Source data for the histograms present in Extended Data Fig. 5.

Source Data Extended Data Fig. 6

Source data for the histograms present in Extended Data Fig. 6.

Source Data Extended Data Fig. 7

Source data for the histograms present in Extended Data Fig. 7.

Source Data Extended Data Fig. 8

Source data for the histograms present in Extended Data Fig. 8.

Source Data Extended Data Fig. 9

Source data for the histograms present in Extended Data Fig. 9.

Source Data Extended Data Fig. 9

Source data for the histology images in Extended Data Fig. 9.

Rights and permissions

Springer Nature or its licensor 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

Michieletto, M.F., Tello-Cajiao, J.J., Mowel, W.K. et al. Multiscale 3D genome organization underlies ILC2 ontogenesis and allergic airway inflammation. Nat Immunol 24, 42–54 (2023). https://doi.org/10.1038/s41590-022-01295-y

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-022-01295-y

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