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:

Epigenetic imprinting in innate lymphoid cell precursors directs the lineage segregation of innate lymphoid cells

Abstract

Innate lymphoid cells (ILCs) are essential for mucosal homeostasis, but the epigenetic regulation of their lineage segregation remains elusive. Here we simultaneously profiled the single-cell DNA methylome, chromatin accessibility and transcriptome of ILC subsets and ILC precursors (ILCPs) and found that ILCPs could be divided into two subgroups (ILCP1 and ILCP2). ILCP2s had highly heterogeneous DNA methylation profiles and could be divided into three groups according to their DNA methylation characteristics, which matched those of ILC subsets. We identified the signature methylation regions (SMRs) of each ILC subset and traced the DNA methylation imprinting during ILCP differentiation. ILCP2s with hypomethylated SMRs characteristic of ILC subsets differentiated into those subsets. DNA methylation editing of SMRs suppressed ILC lineage segregation, while deletion of Dnmt1 in ILCPs abrogated the heterogeneous distribution of SMRs and resulted in ILC differentiation defects. These findings provide evidence that epigenetic imprinting determines lineage segregation during immune cell development.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Single-cell multi-omics sequencing defines two stages of ILCPs.
Fig. 2: Features of DNA methylation and DMRs in ILCP1s and ILCP2s.
Fig. 3: Correlations among DNA methylation, chromatin accessibility and gene expression in ILCP subsets.
Fig. 4: DNA methylation heterogeneity and identification of SMRs in ILCP2s.
Fig. 5: DNA methylation imprints from ILCP2s to ILC subsets.
Fig. 6: DNA methylation contributes to maintaining the homeostasis of ILCPs.

Similar content being viewed by others

Data availability

All data, including raw sequencing files, have been deposited in the Genome Sequence Archive in the BIG Data Center, Chinese Academy of Sciences, under BioProject accession number PRJCA032162, and accession code CRA020302 at https://ngdc.cncb.ac.cn/gsa/. Source data are provided with this paper.

Code availability

The code used in analyzing the dataset is available on GitHub at https://github.com/lz269141050/single-cell-multiomics-sequencing-data-analyses.

References

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

    PubMed  CAS  Google Scholar 

  2. Cherrier, D. E., Serafini, N. & Di Santo, J. P. Innate lymphoid cell development: a T cell perspective. Immunity 48, 1091–1103 (2018).

    PubMed  CAS  Google Scholar 

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

    PubMed  CAS  Google Scholar 

  4. Ishizuka, I. E., Constantinides, M. G., Gudjonson, H. & Bendelac, A. The innate lymphoid cell precursor. Annu. Rev. Immunol. 34, 299–316 (2016).

    PubMed  CAS  Google Scholar 

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

    PubMed  PubMed Central  CAS  Google Scholar 

  6. Klose, C. S. N. et al. Differentiation of type 1 ILCs from a common progenitor to all helper-like innate lymphoid cell lineages. Cell 157, 340–356 (2014).

    PubMed  CAS  Google Scholar 

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

    PubMed  PubMed Central  CAS  Google Scholar 

  8. Li, E., Beard, C. & Jaenisch, R. Role for DNA methylation in genomic imprinting. Nature 366, 362–365 (1993).

    PubMed  CAS  Google Scholar 

  9. Greenberg, M. V. C. & Bourc’his, D. The diverse roles of DNA methylation in mammalian development and disease. Nat. Rev. Mol. Cell Biol. 20, 590–607 (2019).

    PubMed  CAS  Google Scholar 

  10. Smith, Z. D., Hetzel, S. & Meissner, A. DNAmethylation in mammalian development and disease. Nat. Rev. Genet. 26, 7–30 (2025).

    PubMed  CAS  Google Scholar 

  11. Guo, H. et al. The DNA methylation landscape of human early embryos. Nature 511, 606–610 (2014).

    PubMed  CAS  Google Scholar 

  12. Suarez-Alvarez, B. & Rodriguez, R. M. DNA methylation: a promising landscape for immune system-related diseases. Trends Genet. 28, 506–514 (2012).

    PubMed  CAS  Google Scholar 

  13. Trowbridge, J. J., Snow, J. W., Kim, J. & Orkin, S. H. DNA methyltransferase 1 is essential for and uniquely regulates hematopoietic stem and progenitor cells. Cell Stem Cell 5, 442–449 (2009).

    PubMed  PubMed Central  CAS  Google Scholar 

  14. Broske, A. M. et al. DNA methylation protects hematopoietic stem cell multipotency from myeloerythroid restriction. Nat. Genet. 41, 1207–1215 (2009).

    PubMed  Google Scholar 

  15. Challen, G. A. et al. Dnmt3a is essential for hematopoietic stem cell differentiation. Nat. Genet. 44, 23–31 (2011).

    PubMed  PubMed Central  Google Scholar 

  16. Zhang, X. et al. Microbiota regulates the TET1-mediated DNA hydroxymethylation program in innate lymphoid cell differentiation. Nat. Commun. 15, 4792 (2024).

    PubMed  PubMed Central  CAS  Google Scholar 

  17. Huang, Y. et al. Single-cell multi-omics sequencing of human spermatogenesis reveals a DNA demethylation event associated with male meiotic recombination. Nat. Cell Biol. 25, 1520–1534 (2023).

    PubMed  CAS  Google Scholar 

  18. Yang, Q. et al. TCF-1 upregulation identifies early innate lymphoid progenitors in the bone marrow. Nat. Immunol. 16, 1044–1050 (2015).

    PubMed  PubMed Central  CAS  Google Scholar 

  19. Seehus, C. R. et al. The development of innate lymphoid cells requires TOX-dependent generation of a common innate lymphoid cell progenitor. Nat. Immunol. 16, 599–608 (2015).

    PubMed  PubMed Central  CAS  Google Scholar 

  20. Yan, R. et al. Decoding dynamic epigenetic landscapes in human oocytes using single-cell multi-omics sequencing. Cell Stem Cell 28, 1641–1656.e7 (2021).

    PubMed  CAS  Google Scholar 

  21. Liu, Y. et al. Deciphering transcriptome alterations in bone marrow hematopoiesis at single-cell resolution in immune thrombocytopenia. Signal Transduct. Target. Ther. 7, 347 (2022).

    PubMed  PubMed Central  CAS  Google Scholar 

  22. Murase, Y. et al. In vitro reconstitution of epigenetic reprogramming in the human germ line. Nature 631, 170–178 (2024).

    PubMed  PubMed Central  CAS  Google Scholar 

  23. Zhang, Z. et al. Integrated analysis of single-cell and bulk RNA sequencing data reveals a pan-cancer stemness signature predicting immunotherapy response. Genome Med. 14, 45 (2022).

    PubMed  PubMed Central  CAS  Google Scholar 

  24. Morabito, S., Reese, F., Rahimzadeh, N., Miyoshi, E. & Swarup, V. hdWGCNA identifies co-expression networks in high-dimensional transcriptomics data. Cell Rep. Methods 3, 100498 (2023).

    PubMed  PubMed Central  CAS  Google Scholar 

  25. Taberlay, P. C., Statham, A. L., Kelly, T. K., Clark, S. J. & Jones, P. A. Reconfiguration of nucleosome-depleted regions at distal regulatory elements accompanies DNA methylation of enhancers and insulators in cancer. Genome Res. 24, 1421–1432 (2014).

    PubMed  PubMed Central  CAS  Google Scholar 

  26. Mao, Y. et al. Genome-wide methylation and expression analyses reveal the epigenetic landscape of immune-related diseases for tobacco smoking. Clin. Epigenetics 13, 215 (2021).

    PubMed  PubMed Central  CAS  Google Scholar 

  27. Kohli, R. M. & Zhang, Y. TET enzymes, TDG and the dynamics of DNA demethylation. Nature 502, 472–479 (2013).

    PubMed  PubMed Central  CAS  Google Scholar 

  28. Ravichandran, M. et al. Pronounced sequence specificity of the TET enzyme catalytic domain guides its cellular function. Sci. Adv. 8, eabm2427 (2022).

    PubMed  PubMed Central  CAS  Google Scholar 

  29. Liu, X. et al. UHRF1 targets DNMT1 for DNA methylation through cooperative binding of hemi-methylated DNA and methylated H3K9. Nat. Commun. 4, 1563 (2013).

    PubMed  Google Scholar 

  30. Kong, X. et al. Defining UHRF1 domains that support maintenance of human colon cancer DNA methylation and oncogenic properties. Cancer Cell 35, 633–648.e7 (2019).

    PubMed  PubMed Central  CAS  Google Scholar 

  31. 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).

    PubMed  PubMed Central  CAS  Google Scholar 

  32. Liu, Q., Lee, J. H., Kang, H. M. & Kim, C. H. Identification of the niche and mobilization mechanism for tissue-protective multipotential bone marrow ILC progenitors. Sci. Adv. 8, eabq1551 (2022).

    PubMed  PubMed Central  CAS  Google Scholar 

  33. Stelzer, Y., Shivalila, C. S., Soldner, F., Markoulaki, S. & Jaenisch, R. Tracing dynamic changes of DNA methylation at single-cell resolution. Cell 163, 218–229 (2015).

    PubMed  PubMed Central  CAS  Google Scholar 

  34. Hackett, J. A. et al. Germline DNA demethylation dynamics and imprint erasure through 5-hydroxymethylcytosine. Science 339, 448–452 (2013).

    PubMed  CAS  Google Scholar 

  35. Liu, X. S. et al. Editing DNA methylation in the mammalian genome. Cell 167, 233–247e.17 (2016).

    PubMed  PubMed Central  CAS  Google Scholar 

  36. Gamble, N. et al. PU.1 and BCL11B sequentially cooperate with RUNX1 to anchor mSWI/SNF to poise the T cell effector landscape. Nat. Immunol. 25, 860–872 (2024).

    PubMed  PubMed Central  CAS  Google Scholar 

  37. Takahashi, Y. et al. Transgenerational inheritance of acquired epigenetic signatures at CpG islands in mice. Cell 186, 715–731.e9 (2023).

    PubMed  CAS  Google Scholar 

  38. Yu, V. W. C. et al. Epigenetic memory underlies cell-autonomous heterogeneous behavior of hematopoietic stem cells. Cell 167, 1310–1322.e7 (2016).

    PubMed  CAS  Google Scholar 

  39. Yu, Y. et al. Single-cell RNA-seq identifies a PD-1hi ILC progenitor and defines its development pathway. Nature 539, 102–106 (2016).

    PubMed  CAS  Google Scholar 

  40. Peng, V. et al. Whole-genome profiling of DNA methylation and hydroxymethylation identifies distinct regulatory programs among innate lymphocytes.Nat. Immunol. 23, 619–631 (2022).

    PubMed  PubMed Central  CAS  Google Scholar 

  41. Schneider, C. et al. Tissue-resident group 2 innate lymphoid cells differentiate by layered ontogeny and in situ perinatal priming. Immunity 50, 1425–1438.e5 (2019).

    PubMed  PubMed Central  CAS  Google Scholar 

  42. Sun, X. et al. Transcriptional switch of hepatocytes initiates macrophage recruitment and T-cell suppression in endotoxemia. J. Hepatol. 77, 436–452 (2022).

    PubMed  CAS  Google Scholar 

  43. Morabito, S. et al. Single-nucleus chromatin accessibility and transcriptomic characterization of Alzheimer’s disease. Nat. Genet. 53, 1143–1155 (2021).

    PubMed  PubMed Central  CAS  Google Scholar 

  44. Shao, F. et al. FOXO1 orchestrates the intestinal homeostasis via neuronal signaling in group 3 innate lymphoid cells. J. Exp. Med. 220, e20230133 (2023).

    PubMed  PubMed Central  CAS  Google Scholar 

  45. Zhao, M. et al. Maturation and specialization of group 2 innate lymphoid cells through the lung–gut axis. Nat. Commun. 13, 7600 (2022).

    PubMed  PubMed Central  CAS  Google Scholar 

  46. Zhao, M. et al. Spatially resolved transcriptomics reveals distinct pulmonary immune responses during influenza virus and SARS-CoV-2 infections. Sci. Bull. (Beijing) 68, 3137–3141 (2023).

    PubMed  Google Scholar 

  47. Yu, D. et al. Antigen-presenting innate lymphoid cells induced by BCG vaccination promote a respiratory antiviral immune response through the skin‒lung axis. Cell. Mol. Immunol. 22, 390–402 (2025).

    PubMed  CAS  Google Scholar 

  48. Guo, X. et al. Induction of innate lymphoid cell-derived interleukin-22 by the transcription factor STAT3 mediates protection against intestinal infection. Immunity 40, 25–39 (2014).

    PubMed  PubMed Central  CAS  Google Scholar 

  49. Ghoneim, H. E. et al. De novo epigenetic programs inhibit PD-1 blockade-mediated T cell rejuvenation. Cell 170, 142–157.e19 (2017).

    PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank T. Zhao (Institute of Microbiology, Chinese Academy of Sciences) and K. Zhang (Department of Laboratory Animal Science, Health Science Center, Peking University) for their technical support. We thank Z. Zhang (Institute of Biophysics, Chinese Academy of Sciences) for valuable advice. S.W. received support from the National Key R&D Program of China (2022YFC2302900 and 2021YFA1300202), the CAS Project for Young Scientists in Basic Research (YSBR-010),Key Research Program of Frontier Sciences of Chinese Academy of Sciences (ZDBS-LY-SM025) and the Beijing Natural Science Foundation (JQ24043).

Author information

Authors and Affiliations

Contributions

Z.L., F.S. and Q.Z. performed the experiments and analyzed data. M.Z., S.G., X.Z., D.Y. and J.Z. carried out the experiments. P.X. performed data analysis. S.W. initiated the study, organized, designed and wrote the paper.

Corresponding author

Correspondence to Shuo Wang.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks Yi Zhang and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Ioana Staicu, 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 Single-cell multi-omics analyses reveal two developmental stages of ILCPs.

a, Schematic representation showing the isolation of Lin(CD3/CD19/CD11b/Gr-1/TER-119/CD45R)CD45+CD127+NK1.1+NKp46+ ILC1s and Lin(CD3/CD19/CD11b/Gr-1/TER-119/CD45R)CD45+CD127+KLRG1+ ILC2s from the lamina propria of small intestine of wild-type (WT) C57BL/6 mice and Lin(CD3/CD19/CD11b/Gr-1/TER-119/CD45R/NK1.1/KLRG1)CD45+CD127+RORγt-GFP+ ILC3s from RORγt-GFP+ mice, Lin(CD3/CD19/CD45R/CD11b/TER-119/Gr-1)c-Kit+CD127+α4β7+PLZF-GFP+ ILCPs from the bone marrow of Zbtb16 (encodes PLZF)-GFP mice, followed by single-cell multi-omics sequencing and analyses of the transcriptome, chromatin accessibility and DNA methylome. b, Flow cytometry gating strategy showing the isolation of Lin(CD3/CD19/CD45R/CD11b/TER-119/Gr-1)CD127+c-Kit+α4β7+PLZF-GFP+ ILCPs from bone marrow of Zbtb16-GFP mice. c, Heatmap showing expression levels of the marker genes in ILCP1s and ILCP2s. Statistical analyses were performed using Wilcoxon Rank Sum test. d, UMAP plots showing the distribution of CytoTRACE scores in ILCPs. e, RNA velocity analysis showing potential developmental trajectories of ILCPs. f, Dot plot showing positive regulation of lymphoid differentiation scores along pseudotime in ILCPs, calculated using the AddModuleScore function. g, Two-dimensional plots showing dynamic expression of Dnmt1 and Uhrf1 genes in ILCPs along the pseudotime. b-g, Data are representative of at least two independent experiments. Schematic in a was created using BioRender.com.

Extended Data Fig. 2 Chromatin accessibility and DNA methylation properties of ILCP1s and ILCP2s.

a, GCH methylation analysis showing chromatin accessibility 1 kb upstream and 1 kb downstream of the transcription start sites (TSSs) in ILCP1s and ILCP2s. b, GCH methylation analysis showing chromatin accessibility around 2 kb upstream and 2 kb downstream of the gene body in ILCP1s and ILCP2s. c, Bar plot showing relative enrichment scores of NDRs in genomic elements in ILCP subsets. Enrichment scores were calculated as the ratio of NDRs in each genomic element compared to genomic background. SINE (short interspersed nuclear element), LINE (long interspersed nuclear element), LTR (long terminal repeat). d, Heatmap showing chromatin accessibility of common NDRs (C1, n = 16,898), ILCP1-specific NDRs (C2, n = 23,021), and ILCP2-specific NDRs (C3, n = 56,986) in ILCP1s and ILCP2s. e, Pie charts showing distribution of common NDRs (C1), ILCP1-specific NDRs (C2), and ILCP2-specific NDRs (C3) across genomic elements in ILCP1s and ILCP2s. f, Alluvial plots showing dynamic changes of promoter states (homogeneously open, divergent, closed) from ILCP1s to ILCP2s. g, Bar graph showing the distribution of DNA methylation levels in all WCG sites in ILCP1s and ILCP2s. All DNA methylation values were retained to one decimal place. h, Boxplot showing the expression levels of Dnmt1 and Uhrf1 genes in ILCP1s (n = 106 cells) and ILCP2s (n = 206 cells). Each box represented the median and the 25% and 75% quartiles, and the whiskers indicate 1.5 times the interquartile range. Statistical analyses were performed using two-tailed unpaired Student’s t test. i, Two-dimensional plot showing the dynamic expression of scores for DNA methylation maintenance genes along the pseudotime. Data are representative of at least two independent experiments.

Extended Data Fig. 3 Correlation among DNA methylation, chromatin accessibility and gene expression in ILCPs.

a, Volcano plot showing DMRs and genes related to DMRs in their enhancers in ILCP2s compared to ILCP1s. The genes with hypomethylated DMRs in ILCP2s were shown in blue (methylation difference of DMR < -0.1, P < 0.05). The genes with hypermethylated DMRs of ILCP2s were shown in red (methylation difference of DMR > 0.1, P < 0.05). Statistical analyses were performed using Fisher’s exact test. b, Bar graph representing enrichment scores of genes with differentially hypomethylated WCG in enhancers (Diff-hypomethyl-WCG), genes with differentially hypermethylated GCH in enhancers (Diff-hypermethyl-GCH) and upregulated genes in TH1 + TH2 or TH17 cell differentiation KEGG pathways in ILCP2s compared to ILCP1s. Statistical analyses were performed using hypergeometric test. Data are representative of at least two independent experiments.

Extended Data Fig. 4 DNA methylation heterogeneity in ILCP2s.

a-c, Representative flow cytometry plots showing expression of UHRF1-mCherry in Lin(CD3/CD19/CD11b/Gr-1/TER-119/CD45R)CD45+CD127+NK1.1+NKp46+ ILC1s (a), Lin(CD3/CD19/CD11b/Gr-1/TER-119/CD45R)CD45+CD127+KLRG1+ ILC2s (b) and Lin(CD3/CD19/CD11b/Gr-1/TER-119/CD45R/NK1.1/KLRG1)CD45loCD127+CD90hi ILC3s (c) from the small intestine of UHRF1-mCherry reporter mice and WT mice. d, tSNE plot showing gene expression analysis of ILCP1s, ILCP2s, mCherry+ ILCPs, mCherry ILCPs, and mCherry+ ILCPs cultured in vitro for two days (Day 2 in vitro). e, Heatmap showing the expression of marker genes of ILCP1s and ILCP2s in ILCP1s, ILCP2s, mCherry+ ILCPs, mCherry ILCPs, and mCherry+ ILCPs cultured in vitro for two days. f, Heatmap showing WCG methylation levels of selected SMRs in ILCP1s (n = 106 cells), ILCP2_ILC1s (n = 67 cells), ILCP2_ILC2s (n = 69 cells), and ILCP2_ILC3s (n = 70 cells). g, Dot plot showing RNA expression levels of SMR-related genes, including ILCP signature genes (Zbtb16 and Tox), ILC1 signature genes (Tbx21 and Ifng), ILC2 signature genes (Bcl11b and Il4), and ILC3 signature genes (Rorc and Il17a) in ILCP2_ILC1, ILCP2_ILC2, ILCP2_ILC3, ILC1, ILC2 and ILC3 subsets. h, Dot plot displaying GCH methylation levels at SMRs in ILCP and ILC signature genes in ILCP2_ILC1, ILCP2_ILC2, ILCP2_ILC3, ILC1, ILC2 and ILC3 subsets. i, UMAP analysis showing cell type clustering based on transcriptome data of ILCP2_ILC1s (n = 67 cells), ILCP2_ILC2s (n = 69 cells), ILCP2_ILC3s (n = 70 cells), ILC1Ps (n = 46 cells), ILC2Ps (n = 43 cells), and ILC3Ps (n = 35 cells). (ILC1Ps, ILC2Ps, and ILC3Ps from dataset GSE131038 and GSE193835). j, Heatmap showing expression levels of representative ILC marker genes in ILCP2 subsets and precursor cells of ILCs. Lineage-determining transcription factors are highlighted in red. Statistical analyses were performed using two-sided Wilcoxon rank sum test. Data represented at least two (d-j) or three (a-c) independent experiments.

Extended Data Fig. 5 DNA methylation tracing and editing of SMRs in ILCPs.

a, Representative flow cytometry plots showing GFP expression in LinCD127+c-Kit+α4β7+PD-1+ ILCPs transduced with lentivirus carrying Dazl-CGI-Snrpn-GFP methylation reporter systems. The GFP expression in mCherry+ ILCPs was measured two days after transduction. b, Barplots showing proportions of GFP+ ILCPs among mCherry+ ILCPs expressing Tbx21-SMR-Snrpn-GFP (Tbx21), Bcl11b-SMR-Snrpn-GFP (Bcl11b), and Rorc-SMR-Snrpn-GFP (Rorc) reporter systems two days after lentiviral transduction. Data are shown as mean ± s.d. (n = 4 cultures per group). c, Boxplot showing DNA methylation levels of SMRs after DNA methylation editing in ILCPs. LinCD127+c-Kit+α4β7+PD-1+ ILCPs were transduced with lentiviruses encoding either active dCas9-DNMT1 (dC-DNMT1) or a catalytically inactive mutant (dC-dDNMT1), along with SMR-targeting gRNAs. The DNA methylation levels of SMRs were measured by bisulfite sequencing and shown. Each box represents the median and the 25% and 75% quartiles, and the whiskers indicate 1.5 times the interquartile range. Statistical analyses were performed using two-tailed unpaired Student’s t test. (n = 10 per group). Data are representative of at least three independent experiments.

Source data

Extended Data Fig. 6 Hypomethylation of SMRs guides SWI/SNF complex binding.

a, Volcano plot showing DEGs between ILCs and ILCP2s. Statistical analyses were performed using two-tailed unpaired Student’s t test. The blue dots represent genes which were downregulated in ILCs compared with ILCP2s (log2(fold change) < -0.4, P < 0.05). The red dots represent genes which were upregulated in ILCs compared with ILCP2s (log2(fold change) > 0.4, P < 0.05). b, Boxplots showing Smarcc1 (coding BAF155) expression levels in ILCP1s, ILCP2s and ILCP2s in differentiation stages. UHRF1-mCherry+ ILCP (ILCP1s) and UHRF1-mCherry ILCPs (ILCP2s) were isolated and ILCP2s were cultured with OP9-DL1 cells in media with 25 ng/ml IL-7 and 25 ng/ml SCF for the indicated days in vitro. The expression levels of Smarcc1 gene were analyzed by qPCR and shown. Each box represents the median and the 25% and 75% quartiles, and the whiskers indicate 1.5 times the interquartile range. Statistical analyses were performed using one-way ANOVA. (n = 3 cultures per group). c, Bar graph showing BAF155 occupancy at SMRs in ILCPs assessed by CUT&Tag-qPCR. ILCPs were transduced with dC-DNMT1 or catalytically inactive dC-dDNMT1, and enrichment of BAF155 was quantified. Data are presented as mean ± s.d. Statistical analyses were performed using two-tailed unpaired Student’s t-test. (n = 3 cultures per group). d, ATAC-seq analysis showing chromatin accessibility of SMRs in ILCPs transduced with dC-DNMT1 or dC-dDNMT1 and SMR-targeting gRNAs after 7 days in vitro. e, Boxplots showing the expression levels of Tbx21, Bcl11b, and Rorc genes in ILCPs transduced with dC-DNMT1 or dC-dDNMT1 and SMR-targeting gRNAs at day 7 of culture of ILCPs. Each box represents the median and the 25% and 75% quartiles, and the whiskers indicate 1.5 times the interquartile range. Statistical analyses were performed using two-tailed unpaired Student’s t test. (n = 3 cultures per group). Data are representative of at least two (a, d) or three (b-c, e) independent experiments.

Source data

Extended Data Fig. 7 Dnmt1 deletion impairs ILCP differentiation.

a, Bar graph showing the proportions of UHRF1-mCherry+ and UHRF1-mCherry ILCPs in the bone marrow of Id2CreERT2 UHRF1-mCherry mice or Id2CreERT2Dnmt1fl/fl UHRF1-mCherry mice after TMX treatment. Data were shown as mean ± s.d. (n = 3 mice per group). b, Representative flow cytometry histograms (left) showing the expression levels of UHRF1-mCherry in Id2CreERT2Dnmt1fl/fl UHRF1-mCherry ILCPs in the cell culture with OP9-DL1 cells with 25 ng/ml IL-7 and 25 ng/ml SCF at the initial time point (green line), two days later with (yellow line) or without 4-OHT treatment (red line). ILCPs from WT mice served as a control (grey line). The MFI of UHRF1-mCherry was analyzed by flow cytometry and shown as mean ± s.d (right). Statistical analyses were performed using two-tailed unpaired Student’s t test. (n = 3 cultures per group). c, Principal component analysis (PCA) showing single-cell clustering based on DNA methylation levels in promoters, colored by the indicated cell type. UHRF1-mCherry+ ILCPs (ILCP1s) from Id2CreERT2Dnmt1fl/fl UHRF1-mCherry+ mice treated with TMX (n = 26 cells) were isolated for single-cell multiomics sequencing and clustered with ILCP1s (n = 106 cells) and ILCP2s (n = 206 cells). Data are representative of at least two (c) or three (a, b) independent experiments.

Source data

Extended Data Fig. 8 DNA methylation regulates the differentiation of ILCPs.

a,b, Representative flow cytometry plots showing the GFP expression in Id2CreERT2Dnmt1fl/fl ILCPs (CD127+c-Kit+α4β7+PD-1+) transduced with Tbx21-SMR-Snrpn-GFP reporter system in the media without (a) or with (b) 4-OHT. c, d, Bisulfite sequencing analysis showing SMR DNA methylation levels in Id2CreERT2Dnmt1fl/fl ILCPs transduced with SMR reporter systems in the media without (c) or with (d) 4-OHT. e, The proportion of the indicated lymphocytes from the thymus, the small intestine (SI), and the lung of Id2CreERT2 and Id2CreERT2Dnmt1fl/fl mice treated as in Fig. 6c. The frequencies of CD45+CD4CD8 double negative (DN) T, CD45+CD4+CD8+ double positive (DP) T, CD45+CD3+CD4+ T cells (CD4 T), CD45+CD3+CD8+ T cells (CD8 T), CD45+CD3+ T cells (T), CD45+CD19+ B cells (B) were shown as mean ± s.d. (n = 3 or 4 mice per group). f,g, Percentage of cleaved caspase3+ ILCPs among total ILCPs (f) and cell numbers of ILCPs (g) from the bone marrow of WT mice, cultured for 2 days with DNMT1 inhibitor (GSK-3484862) or not (vehicle) in vitro. Shown as mean ± s.d. (n = 4 cultures per group). h, i, Percentage of cleaved caspase3+ cells among ILC1s, ILC2s or ILC3s (h) and the number of ILC1, ILC2 and ILC3 (i) sorted from the small intestine of WT mice, and cultured for 2 days with DNMT1 inhibitor or not (vehicle) in vitro. Shown as mean ± s.d. (n = 3 cultures per group). Statistical analyses of e-i were performed using two-tailed unpaired Student’s t test. Data are representative of at least three independent experiments.

Source data

Supplementary information

Reporting Summary

Supplementary Table 1

Pearson correlation between the gene expression modules and DEGs.

Supplementary Table 2

Methylation enzyme-related gene sets.

Supplementary Table 3

Bisulfite sequencing PCR primer list.

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 Extended Data Fig. 5

Statistical source data.

Source Data Extended Data Fig. 6

Statistical source data.

Source Data Extended Data Fig. 7

Statistical source data.

Source Data Extended Data Fig. 8

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

Liu, Z., Shao, F., Zhang, Q. et al. Epigenetic imprinting in innate lymphoid cell precursors directs the lineage segregation of innate lymphoid cells. Nat Immunol 26, 1686–1698 (2025). https://doi.org/10.1038/s41590-025-02261-0

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue date:

  • DOI: https://doi.org/10.1038/s41590-025-02261-0

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