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:

Remodeling of Il4-Il13-Il5 locus underlies selective gene expression

Abstract

The type 2 cytokines, interleukin (IL)-4, IL-13 and IL-5 reside within a multigene cluster. Both innate (ILC2) and adaptive T helper 2 (TH2) lymphocytes secrete type 2 cytokines with diverse production spectra. Using transcription factor footprint and chromatin accessibility, we systemically cataloged regulatory elements (REs) denoted as SHS-I/II, KHS-I/II, +6.5kbIl13, 5HS-I(a, b, c, d, e), 5HS-II and 5HS-III(a, b, c) across the extended Il4-Il13-Il5 locus in mice. Physical proximities among REs were coordinately remodeled in three-dimensional space after cell activation, leading to divergent compartmentalization of Il4, Il13 and Il5 with varied combinations of REs. Deletions of REs revealed no single RE solely accounted for selective regulation of a given cytokine in vivo. Instead, individual RE differentially contribute to proper genomic positioning of REs and target genes. RE deletions resulted in context-dependent dysregulation of cytokine expression and immune response in tissue. Thus, signal-dependent remodeling of three-dimensional configuration underlies divergent cytokine outputs from the type 2 loci.

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: Genome-wide transcription, TF loading and 3D conformation in ILC2s and TH2 cells before and after activation.
Fig. 2: Activation induced TF loading and inferred CTCF loops across the type 2 cytokine locus.
Fig. 3: Activation induced 3D remodeling of type 2 cytokine loci in ILC2 and TH2 cells.
Fig. 4: Contribution of enhancers and structural REs during allergic lung inflammation.
Fig. 5: REs downstream of Il5 differently contribute to chromatin architecture.
Fig. 6: Deletion of +6.5kbIl13 alters TAD structure, enhancer accessibility and IL-13 and IL-5 expression of ILC2s.
Fig. 7: Conserved REs and organization of the IL4-IL13-IL5 locus in human TH2 cells.

Similar content being viewed by others

Data availability

Raw and processed data for ATAC-seq, mRNA-seq, ChIP-seq and Hi-C generated in this study have been deposited in the National Center for Biotechnology Information Gene Expression Omnibus (GSE266349). Raw and processed data for ATAC-seq and mRNA-seq of mouse ILC2s were obtained from our previously published study (GSE131996)14. Source data are provided with this paper.

Code availability

No custom-made code was used in the analysis except for ATAC-seq analysis pipeline (available on request). The pipelines for analysis can be obtained by e-mailing hiroyuki.nagashima@nih.gov.

References

  1. Moro, K. et al. Innate production of T(H)2 cytokines by adipose tissue-associated c-Kit(+)Sca-1(+) lymphoid cells. Nature 463, 540–544 (2010).

    Article  PubMed  CAS  Google Scholar 

  2. Neill, D. R. et al. Nuocytes represent a new innate effector leukocyte that mediates type-2 immunity. Nature 464, 1367–1370 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  3. Price, A. E. et al. Systemically dispersed innate IL-13-expressing cells in type 2 immunity. Proc. Natl Acad. Sci. USA 107, 11489–11494 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  6. Spits, H. & Mjosberg, J. Heterogeneity of type 2 innate lymphoid cells. Nat. Rev. Immunol. https://doi.org/10.1038/s41577-022-00704-5 (2022).

    Article  PubMed  PubMed Central  Google Scholar 

  7. Klose, C. S. N. & Artis, D. Innate lymphoid cells control signaling circuits to regulate tissue-specific immunity. Cell Res. 30, 475–491 (2020).

    Article  PubMed  PubMed Central  Google Scholar 

  8. Kabata, H., Moro, K. & Koyasu, S. The group 2 innate lymphoid cell (ILC2) regulatory network and its underlying mechanisms. Immunol. Rev. 286, 37–52 (2018).

    Article  PubMed  CAS  Google Scholar 

  9. Nakayama, T. et al. Th2 cells in health and disease. Annu Rev. Immunol. 35, 53–84 (2017).

    Article  PubMed  CAS  Google Scholar 

  10. Ansel, K. M., Djuretic, I., Tanasa, B. & Rao, A. Regulation of Th2 differentiation and Il4 locus accessibility. Annu Rev. Immunol. 24, 607–656 (2006).

    Article  PubMed  CAS  Google Scholar 

  11. Lee, G. R., Kim, S. T., Spilianakis, C. G., Fields, P. E. & Flavell, R. A. T helper cell differentiation: regulation by cis elements and epigenetics. Immunity 24, 369–379 (2006).

    Article  PubMed  CAS  Google Scholar 

  12. Lee, G. R. Transcriptional regulation of T helper type 2 differentiation. Immunology 141, 498–505 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  13. Buniello, A. et al. The NHGRI-EBI GWAS Catalog of published genome-wide association studies, targeted arrays and summary statistics 2019. Nucleic Acids Res. 47, D1005–D1012 (2019).

    Article  PubMed  CAS  Google Scholar 

  14. Nagashima, H. et al. Neuropeptide CGRP limits group 2 innate lymphoid cell responses and constrains type 2 inflammation. Immunity 51, 682–695 e686 (2019).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  15. Pelly, V. S. et al. IL-4-producing ILC2s are required for the differentiation of T(H)2 cells following Heligmosomoides polygyrus infection. Mucosal Immunol. 9, 1407–1417 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  16. Noval Rivas, M., Burton, O. T., Oettgen, H. C. & Chatila, T. IL-4 production by group 2 innate lymphoid cells promotes food allergy by blocking regulatory T-cell function. J. Allergy Clin. Immunol. 138, 801–811 e809 (2016).

    Article  PubMed  CAS  Google Scholar 

  17. Kubo, M. Innate and adaptive type 2 immunity in lung allergic inflammation. Immunol. Rev. 278, 162–172 (2017).

    Article  PubMed  CAS  Google Scholar 

  18. Lee, G. R., Fields, P. E., Griffin, T. J. & Flavell, R. A. Regulation of the Th2 cytokine locus by a locus control region. Immunity 19, 145–153 (2003).

    Article  PubMed  CAS  Google Scholar 

  19. Loots, G. G. et al. Identification of a coordinate regulator of interleukins 4, 13, and 5 by cross-species sequence comparisons. Science 288, 136–140 (2000).

    Article  PubMed  CAS  Google Scholar 

  20. Ansel, K. M., Lee, D. U. & Rao, A. An epigenetic view of helper T cell differentiation. Nat. Immunol. 4, 616–623 (2003).

    Article  PubMed  CAS  Google Scholar 

  21. Fields, P. E., Lee, G. R., Kim, S. T., Bartsevich, V. V. & Flavell, R. A. Th2-specific chromatin remodeling and enhancer activity in the Th2 cytokine locus control region. Immunity 21, 865–876 (2004).

    Article  PubMed  CAS  Google Scholar 

  22. Lee, D. U. & Rao, A. Molecular analysis of a locus control region in the T helper 2 cytokine gene cluster: a target for STAT6 but not GATA3. Proc. Natl Acad. Sci. USA 101, 16010–16015 (2004).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  23. Koh, B. H. et al. Th2 LCR is essential for regulation of Th2 cytokine genes and for pathogenesis of allergic asthma. Proc. Natl Acad. Sci. USA 107, 10614–10619 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  24. Williams, A. et al. Hypersensitive site 6 of the Th2 locus control region is essential for Th2 cytokine expression. Proc. Natl Acad. Sci. USA 110, 6955–6960 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  25. Hwang, S. S., Jang, S. W. & Lee, G. R. RHS6-mediated chromosomal looping and nuclear substructure binding is required for Th2 cytokine gene expression. Biochim. Biophys. Acta Gene Regul. Mech. 1860, 383–391 (2017).

    Article  PubMed  CAS  Google Scholar 

  26. Mohrs, M. et al. Deletion of a coordinate regulator of type 2 cytokine expression in mice. Nat. Immunol. 2, 842–847 (2001).

    Article  PubMed  CAS  Google Scholar 

  27. Lee, G. R., Fields, P. E. & Flavell, R. A. Regulation of IL-4 gene expression by distal regulatory elements and GATA-3 at the chromatin level. Immunity 14, 447–459 (2001).

    Article  PubMed  CAS  Google Scholar 

  28. Yamashita, M. et al. Identification of a conserved GATA3 response element upstream proximal from the interleukin-13 gene locus. J. Biol. Chem. 277, 42399–42408 (2002).

    Article  PubMed  CAS  Google Scholar 

  29. Tanaka, S. et al. The enhancer HS2 critically regulates GATA-3-mediated Il4 transcription in T(H)2 cells. Nat. Immunol. 12, 77–85 (2011).

    Article  PubMed  CAS  Google Scholar 

  30. Agarwal, S. & Rao, A. Modulation of chromatin structure regulates cytokine gene expression during T cell differentiation. Immunity 9, 765–775 (1998).

    Article  PubMed  CAS  Google Scholar 

  31. Ansel, K. M. et al. Deletion of a conserved Il4 silencer impairs T helper type 1-mediated immunity. Nat. Immunol. 5, 1251–1259 (2004).

    Article  PubMed  CAS  Google Scholar 

  32. Djuretic, I. M. et al. Transcription factors T-bet and Runx3 cooperate to activate IFNg and silence Il4 in T helper type 1 cells. Nat. Immunol. 8, 145–153 (2007).

    Article  PubMed  CAS  Google Scholar 

  33. Agarwal, S., Avni, O. & Rao, A. Cell-type-restricted binding of the transcription factor NFAT to a distal IL-4 enhancer in vivo. Immunity 12, 643–652 (2000).

    Article  PubMed  CAS  Google Scholar 

  34. Solymar, D. C., Agarwal, S., Bassing, C. H., Alt, F. W. & Rao, A. A 3′ enhancer in the IL-4 gene regulates cytokine production by Th2 cells and mast cells. Immunity 17, 41–50 (2002).

    Article  PubMed  CAS  Google Scholar 

  35. Vijayanand, P. et al. Interleukin-4 production by follicular helper T cells requires the conserved Il4 enhancer hypersensitivity site V. Immunity 36, 175–187 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  36. Wallrapp, A. et al. The neuropeptide NMU amplifies ILC2-driven allergic lung inflammation. Nature 549, 351–356 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Klose, C. S. N. et al. The neuropeptide neuromedin U stimulates innate lymphoid cells and type 2 inflammation. Nature 549, 282–286 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  40. Bonev, B. et al. Multiscale 3D genome rewiring during mouse neural development. Cell 171, 557–572.e524 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  41. Pekowska, A. et al. Gain of CTCF-anchored chromatin loops marks the exit from naive pluripotency. Cell Syst. 7, 482–495.e410 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Phanstiel, D. H. et al. Static and dynamic DNA loops form AP-1-bound activation hubs during macrophage development. Mol. Cell 67, 1037–1048.e1036 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  44. Rao, S. S. P. et al. Cohesin loss eliminates all loop domains. Cell 171, 305–320.e24 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  45. Nora, E. P. et al. Targeted degradation of CTCF decouples local insulation of chromosome domains from genomic compartmentalization. Cell 169, 930–944.e922 (2017).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  46. Cuartero, S. et al. Control of inducible gene expression links cohesin to hematopoietic progenitor self-renewal and differentiation. Nat. Immunol. 19, 932–941 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  47. Wendt, K. S. et al. Cohesin mediates transcriptional insulation by CCCTC-binding factor. Nature 451, 796–U793 (2008).

    Article  PubMed  CAS  Google Scholar 

  48. Ribeiro de Almeida, C. et al. Critical role for the transcription regulator CCCTC-binding factor in the control of Th2 cytokine expression. J. Immunol. 182, 999–1010 (2009).

    Article  PubMed  CAS  Google Scholar 

  49. Takemoto, N. et al. Th2-specific DNase I-hypersensitive sites in the murine IL-13 and IL-4 intergenic region. Int. Immunol. 10, 1981–1985 (1998).

    Article  PubMed  CAS  Google Scholar 

  50. Rao, S. S. P. et al. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping. Cell 159, 1665–1680 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  51. Nassar, L. R. et al. The UCSC Genome Browser database: 2023 update. Nucleic Acids Res. 51, D1188–D1195 (2023).

    Article  PubMed  CAS  Google Scholar 

  52. Trieu, T., Oluwadare, O., Wopata, J. & Cheng, J. L. GenomeFlow: a comprehensive graphical tool for modeling and analyzing 3D genome structure. Bioinformatics 35, 1416–1418 (2019).

    Article  PubMed  CAS  Google Scholar 

  53. Lee, G. R., Spilianakis, C. G. & Flavell, R. A. Hypersensitive site 7 of the TH2 locus control region is essential for expressing TH2 cytokine genes and for long-range intrachromosomal interactions. Nat. Immunol. 6, 42–48 (2005).

    Article  PubMed  CAS  Google Scholar 

  54. Spilianakis, C. G. & Flavell, R. A. Long-range intrachromosomal interactions in the T helper type 2 cytokine locus. Nat. Immunol. 5, 1017–1027 (2004).

    Article  PubMed  CAS  Google Scholar 

  55. Dekker, J. & Mirny, L. The 3D genome as moderator of chromosomal communication. Cell 164, 1110–1121 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Sanborn, A. L. et al. Chromatin extrusion explains key features of loop and domain formation in wild-type and engineered genomes. Proc. Natl Acad. Sci. USA 112, E6456–E6465 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  58. Marszalek, J. R., Ruiz-Lozano, P., Roberts, E., Chien, K. R. & Goldstein, L. S. Situs inversus and embryonic ciliary morphogenesis defects in mouse mutants lacking the KIF3A subunit of kinesin-II. Proc. Natl Acad. Sci. USA 96, 5043–5048 (1999).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Liu, S. et al. Regulation of T helper cell differentiation by the interplay between histone modification and chromatin interaction. Immunity https://doi.org/10.1016/j.immuni.2024.03.018 (2024).

    Article  PubMed  PubMed Central  Google Scholar 

  60. Ishigaki, K. et al. Large-scale genome-wide association study in a Japanese population identifies novel susceptibility loci across different diseases. Nat. Genet. 52, 669–679 (2020).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  61. Liang, H. E. et al. Divergent expression patterns of IL-4 and IL-13 define unique functions in allergic immunity. Nat. Immunol. 13, 58–66 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  62. 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.e191-194 (2012).

  63. Huang, Y. et al. IL-25-responsive, lineage-negative KLRG1(hi) cells are multipotential ‘inflammatory’ type 2 innate lymphoid cells. Nat. Immunol. 16, 161–169 (2015).

    Article  PubMed  CAS  Google Scholar 

  64. Gabriele, M. et al. Dynamics of CTCF- and cohesin-mediated chromatin looping revealed by live-cell imaging. Science 376, 496–501 (2022).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Wang, H. et al. One-step generation of mice carrying mutations in multiple genes by CRISPR/Cas-mediated genome engineering. Cell 153, 910–918 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  66. Corces, M. R. et al. Lineage-specific and single-cell chromatin accessibility charts human hematopoiesis and leukemia evolution. Nat. Genet. 48, 1193–1203 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  67. Rao, S. S. P. et al. A 3D map of the human genome at kilobase resolution reveals principles of chromatin looping [Erratum in 159, 1665, 2014]. Cell 162, 687–688 (2015).

    Article  CAS  Google Scholar 

  68. Bentley, D. R. et al. Accurate whole human genome sequencing using reversible terminator chemistry. Nature 456, 53–59 (2008).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  69. Trapnell, C. et al. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat. Protoc. 7, 562–578 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  71. Robinson, J. T. et al. Integrative genomics viewer. Nat. Biotechnol. 29, 24–26 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  72. R Core Team. R: A Language and Environment for Statistical Computing (R Foundation for Statistical Computing, 2021).

  73. Langmead, B., Trapnell, C., Pop, M. & Salzberg, S. L. Ultrafast and memory-efficient alignment of short DNA sequences to the human genome. Genome Biol. 10, R25 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  74. Xu, H. et al. FastUniq: a fast de novo duplicates removal tool for paired short reads. PLoS ONE 7, e52249 (2012).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  76. Heinz, S. et al. Simple combinations of lineage-determining transcription factors prime cis-regulatory elements required for macrophage and B cell identities. Mol. Cell 38, 576–589 (2010).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  77. Durand, N. C. et al. Juicer provides a one-click system for analyzing loop-resolution Hi-C experiments. Cell Syst. 3, 95–98 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  PubMed  PubMed Central  Google Scholar 

  79. 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  PubMed  PubMed Central  CAS  Google Scholar 

  80. 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  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank H. Y. Shih for critical input in interpreting and analyzing the data. We thank S. Dell’Orso and F. Naz (Genome Analysis Core Facility, NIAMS); J. Simone, J. Lay and K. Tinsley (Flow Cytometry Section, NIAMS); S. Jung, H. W. Sun, A. Uhlman, V. Chaitankar and S. R. Brooks (Biodata Mining and Discovery Section, NIAMS); and C. Liu (Transgenic Core, NHLBI) for their technical support. This study used the high-performance computational capabilities of the Biowulf Linux cluster at the NIH. This work was supported by the Intramural Research Programs of NIAMS (grant no. ZIA AR041159 to J.J.O’S.). H.N. was supported by the JSPS Research Fellowship for Japanese Biomedical and Behavioural Researchers at NIH (grant no. 71703).

Author information

Authors and Affiliations

Authors

Contributions

H.N. and J.J.O’S. initiated and designed the project. H.N. performed experiments, analyzed, visualized the data and drafted the manuscript. J.S. performed and analyzed experiments. K.J. contributed the computational analysis. F.P. assisted with the in situ Hi-C. A.P. contributed the design of focused Hi-C. Y.K. and J.J.O’S. supervised the project and edited the manuscript.

Corresponding authors

Correspondence to Hiroyuki Nagashima or John J. O’Shea.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editor: L. A. Dempsey, in collaboration with the Nature Immunology team.

Additional information

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

Extended data

Extended Data Fig. 1 Gene expression, TF loading, and in situ Hi-C of ILC2s and Th2 cells, related to Fig. 1.

a and b, Gene expression of transcription factors in mouse Th2 and ILC2s by mRNA-seq (GSE131996). c, Aggregated NFAT1 load near HIGs (+/-50kb from TSS) were calculated and binned into 3 categories (common (n = 45), ILC2 specific (n = 70) or Th2 specific (n = 49)) and graphed (Y-axis) for 4 sample conditions (X-axis), as in Fig. 1d. The lower and the upper edge of the box represent the first percentile and the third percentile of the data, respectively. The line inside the box represents the median. The whiskers represent the minimum, maximum and its variability in comparison to the interquartile range. A two-sided t test was used to statistically evaluate the difference of the NFAT1 load between ILC2 specific HIGs and Th2 specific HIGs. d, Interaction frequency (Y axis) vs distance (X axis) of chromatin, measured by HiC for 8 samples. e, Percentage of local chromatin interactions. f, Percentage of interchromosomal interactions. g, Number of TADs detected in individual samples. All TADs are merged by merge2Dbed.pl function by HOMER for downstream analysis.

Source data

Extended Data Fig. 2 Genomic mapping of REs controlling type 2 cytokine locus, related to Fig. 2.

a and b, Five select regions for SHS-I/II, KHS-I/II, +6.5kbIl13, 5HS-II/5HS-III (a + b + c), and 5HS-I(a+b+c+d+e) identified in Fig. 2 were further magnified with phyloP Scores (Mammal Basewise Conservation of 59 vertebrate) showing DNA conservation across species (red; highly conserved, blue; less conserved) and CTCF, GATA3, and ATAC peaks in stimulated ILC2s. The location and directionality of CTCF motifs are depicted with red arrows indicating forward and blue indicating reverse in a. GATA3 motifs are depicted in purple rectangles in a (1 motif) and b (5 motifs). Data from two biological replicates were used for presentation.

Extended Data Fig. 3 Remodeling of Il13/Rad50/Il5 loci induced by SDTFs.

A compiled view of Il13/Rad50/Il5 loci showing chromatin accessibility, TFs binding and HiC interaction in ILC2s stimulated with IL-33 plus NMU. Data represents chr11: 53,623,000-53,745,000 (122kB). Red circles illustrate interactions between enhancers proximity to Il13 and Il5 and the locus control region (LCR).

Extended Data Fig. 4 Gating strategy for lung cells, related to Fig. 4.

a and b, A schematic gating strategy for flow cytometry analyses for mouse lung cells.

Extended Data Fig. 5 Humoral immune response of 5HS-Ie, 5HS-II and 5HS-III(a+b+c) KO mice, related to Fig. 4.

Quantification of papainspecific IgM, IgG1 and IgE in WT (naïve; n = 8, papain; n = 24), 5HS-Ie KO (naïve; n = 4, papain; n = 8), 5HS-II KO (naïve; n = 4, papain; n = 11) and 5HS-III(a+b+c) KO (naïve; n = 5, papain; n = 16) mice before and after administration of Papain, as in Fig. 4. Graphs show mean ± SEM.

Source data

Extended Data Fig. 6 Chromatin architecture of 5HS-II and 5HS-III(a+b+c) KO ILC2s, related to Fig. 5.

A split view of HiC heatmap of WT, 5HS-II KO and 5HS-III(a+b+c) KO ILC2s with or without stimulation with IL-33 + NMU for 1 h, as in Fig. 5c.

Extended Data Fig. 7 Contribution of +6.5kbIl13 during chronic lung inflammation, related to Fig. 6.

a, mRNA expression of Il4, Il13 and Il5 in WT and +6.5kbIl13 KO ILC2s stimulated with IL-33 + NMU measured by qPCR. The data were normalized to Actb, then further normalized to non-stimulated WT cells ( = 1). Graphs are representative plots from three independent experiments, and triplicates summary are shown. b-d, WT (n = 13) and +6.5kbIl13KO (Δ+6.5kbIl13, n = 10) mice were challenged with papain to induce lung inflammation. c, Flow cytometry for infiltrating cells recovered from the lungs. Cells evaluated were total live cells, eosinophils (CD11b+ Siglec-F+), neutrophils (CD11b+ Gr1+), Th2 (CD3ε+ TCRβ+ CD4+ CD44+ Foxp3- GATA3+) and ILC2s (Lin Thy1+ CD127+ GATA3+). d, Cytokine production from Th2 cells was evaluated by flow cytometry measuring IL-4, IL-13 and IL-5. Graphs in c and d show mean ± SEM. Statistical significance is depicted as p < 0.05, p < 0.01 and p < 0.001 (Two-sided t test).

Source data

Supplementary information

Reporting Summary

Supplementary Table 1

List of HIGs and WIGs, related to Fig. 1b.

Supplementary Table 2

List of common and cell-type-specific HIGs and WIGs, related to Fig. 1c.

Supplementary Table 3

List of total and differentially regulated TADs, related to Fig. 1e.

Supplementary Table 4

Guide RNAs and primers for the generation of mice.

Supplementary Table 5

Custom oligo pool for focused Hi-C.

Source data

Source Data Fig. 1

Statistical source data.

Source Data Fig. 3

Statistical source data.

Source Data Fig. 4

Statistical source data.

Source Data Fig. 5

Statistical source data.

Source Data Fig. 6

Statistical source data.

Source Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 1

Statistical source data.

Source Data Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Rights and permissions

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

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Nagashima, H., Shayne, J., Jiang, K. et al. Remodeling of Il4-Il13-Il5 locus underlies selective gene expression. Nat Immunol 25, 2220–2233 (2024). https://doi.org/10.1038/s41590-024-02007-4

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-024-02007-4

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
pFad - Phonifier reborn

Pfad - The Proxy pFad of © 2024 Garber Painting. All rights reserved.

Note: This service is not intended for secure transactions such as banking, social media, email, or purchasing. Use at your own risk. We assume no liability whatsoever for broken pages.


Alternative Proxies:

Alternative Proxy

pFad Proxy

pFad v3 Proxy

pFad v4 Proxy