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:

Long noncoding RNA lncKdm2b is required for ILC3 maintenance by initiation of Zfp292 expression

Abstract

Innate lymphoid cells (ILCs) communicate with other hematopoietic and nonhematopoietic cells to regulate immunity, inflammation and tissue homeostasis. How ILC lineages develop and are maintained remains largely unknown. In this study we observed that a divergent long noncoding RNA (lncRNA), lncKdm2b, was expressed at high levels in intestinal group 3 ILCs (ILC3s). LncKdm2b deficiency in the hematopoietic system led to reductions in the number and effector functions of ILC3s. LncKdm2b expression sustained the maintenance of ILC3s by promoting their proliferation through activation of the transcription factor Zfp292. Mechanistically, lncKdm2b recruited the chromatin organizer Satb1 and the nuclear remodeling factor (NURF) complex onto the Zfp292 promoter to initiate its transcription. Deletion of Zfp292 or Bptf also abrogated the maintenance of ILC3s, leading to susceptibility to bacterial infection. Therefore, our findings reveal that lncRNAs may represent an additional layer of regulation of ILC development and function.

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

Figure 1: LncKdm2b deficiency impairs the maintenance and effector functions of ILC3s.
Figure 2: LncKdm2b knockout suppresses ILC3 proliferation.
Figure 3: LncKdm2b associates with Satb1.
Figure 4: LncKdm2b activates Zfp292 expression through association with Satb1.
Figure 5: LncKdm2b recruits Satb1 and the NURF complex onto the Zfp292 promoter to trigger its expression.
Figure 6: Zfp292 deficiency impairs ILC3 maintenance and effector functions.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

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

    Article  CAS  PubMed  Google Scholar 

  2. Buonocore, S. et al. Innate lymphoid cells drive interleukin-23-dependent innate intestinal pathology. Nature 464, 1371–1375 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Ebihara, T. et al. Runx3 specifies lineage commitment of innate lymphoid cells. Nat. Immunol. 16, 1124–1133 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Bernink, J.H. et al. Interleukin-12 and -23 control plasticity of CD127+ group 1 and group 3 innate lymphoid cells in the intestinal lamina propria. Immunity 43, 146–160 (2015).

    CAS  PubMed  Google Scholar 

  5. Brestoff, J.R. et al. Group 2 innate lymphoid cells promote beiging of white adipose tissue and limit obesity. Nature 519, 242–246 (2015).

    CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  8. Guo, X. et al. Innate lymphoid cells control early colonization resistance against intestinal pathogens through ID2-dependent regulation of the microbiota. Immunity 42, 731–743 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Song, C. et al. Unique and redundant functions of NKp46+ ILC3s in models of intestinal inflammation. J. Exp. Med. 212, 1869–1882 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Diefenbach, A., Colonna, M. & Koyasu, S. Development, differentiation, and diversity of innate lymphoid cells. Immunity 41, 354–365 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Serafini, N. et al. Gata3 drives development of RORγt+ group 3 innate lymphoid cells. J. Exp. Med. 211, 199–208 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. De Obaldia, M.E. & Bhandoola, A. Transcriptional regulation of innate and adaptive lymphocyte lineages. Annu. Rev. Immunol. 33, 607–642 (2015).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  17. Kiss, E.A. et al. Natural aryl hydrocarbon receptor ligands control organogenesis of intestinal lymphoid follicles. Science 334, 1561–1565 (2011).

    CAS  PubMed  Google Scholar 

  18. Qiu, J. et al. The aryl hydrocarbon receptor regulates gut immunity through modulation of innate lymphoid cells. Immunity 36, 92–104 (2012).

    CAS  PubMed  Google Scholar 

  19. Zhong, C. et al. Group 3 innate lymphoid cells continuously require the transcription factor GATA-3 after commitment. Nat. Immunol. 17, 169–178 (2016).

    CAS  PubMed  Google Scholar 

  20. Batista, P.J. & Chang, H.Y. Long noncoding RNAs: cellular address codes in development and disease. Cell 152, 1298–1307 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Djebali, S. et al. Landscape of transcription in human cells. Nature 489, 101–108 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Kretz, M. et al. Control of somatic tissue differentiation by the long non-coding RNA TINCR. Nature 493, 231–235 (2013).

    CAS  PubMed  Google Scholar 

  23. Zhu, P. et al. lnc-β-Catm elicits EZH2-dependent β-catenin stabilization and sustains liver CSC self-renewal. Nat. Struct. Mol. Biol. 23, 631–639 (2016).

    CAS  PubMed  Google Scholar 

  24. Zhu, P. et al. LncBRM initiates YAP1 signalling activation to drive self-renewal of liver cancer stem cells. Nat. Commun. 7, 13608 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Luo, S. et al. Divergent lncRNAs regulate gene expression and lineage differentiation in pluripotent cells. Cell Stem Cell 18, 637–652 (2016).

    CAS  PubMed  Google Scholar 

  26. Anderson, K.M. et al. Transcription of the non-coding RNA upperhand controls Hand2 expression and heart development. Nature 539, 433–436 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  29. Katayama, S. et al. Antisense transcription in the mammalian transcriptome. Science 309, 1564–1566 (2005).

    PubMed  Google Scholar 

  30. Yasui, D., Miyano, M., Cai, S., Varga-Weisz, P. & Kohwi-Shigematsu, T. SATB1 targets chromatin remodelling to regulate genes over long distances. Nature 419, 641–645 (2002).

    CAS  PubMed  Google Scholar 

  31. McKenzie, A.N., Spits, H. & Eberl, G. Innate lymphoid cells in inflammation and immunity. Immunity 41, 366–374 (2014).

    CAS  PubMed  Google Scholar 

  32. Bouskra, D. et al. Lymphoid tissue genesis induced by commensals through NOD1 regulates intestinal homeostasis. Nature 456, 507–510 (2008).

    CAS  PubMed  Google Scholar 

  33. De Togni, P. et al. Abnormal development of peripheral lymphoid organs in mice deficient in lymphotoxin. Science 264, 703–707 (1994).

    CAS  PubMed  Google Scholar 

  34. Cella, M. et al. A human natural killer cell subset provides an innate source of IL-22 for mucosal immunity. Nature 457, 722–725 (2009).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  36. Sonnenberg, G.F., Fouser, L.A. & Artis, D. Border patrol: regulation of immunity, inflammation and tissue homeostasis at barrier surfaces by IL-22. Nat. Immunol. 12, 383–390 (2011).

    CAS  PubMed  Google Scholar 

  37. Guttman, M. et al. Ab initio reconstruction of cell type–specific transcriptomes in mouse reveals the conserved multi-exonic structure of lincRNAs. Nat. Biotechnol. 28, 503–510 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Cai, S., Han, H.J. & Kohwi-Shigematsu, T. Tissue-specific nuclear architecture and gene expression regulated by SATB1. Nat. Genet. 34, 42–51 (2003).

    CAS  PubMed  Google Scholar 

  39. Savarese, F. et al. Satb1 and Satb2 regulate embryonic stem cell differentiation and Nanog expression. Genes Dev. 23, 2625–2638 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. Beyer, M. et al. Repression of the genome organizer SATB1 in regulatory T cells is required for suppressive function and inhibition of effector differentiation. Nat. Immunol. 12, 898–907 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Will, B. et al. Satb1 regulates the self-renewal of hematopoietic stem cells by promoting quiescence and repressing differentiation commitment. Nat. Immunol. 14, 437–445 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Satoh, Y. et al. The Satb1 protein directs hematopoietic stem cell differentiation toward lymphoid lineages. Immunity 38, 1105–1115 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Steidl, U. et al. A distal single nucleotide polymorphism alters long-range regulation of the PU.1 gene in acute myeloid leukemia. J. Clin. Invest. 117, 2611–2620 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Hartley, P.D. & Madhani, H.D. Mechanisms that specify promoter nucleosome location and identity. Cell 137, 445–458 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Clapier, C.R. & Cairns, B.R. Regulation of ISWI involves inhibitory modules antagonized by nucleosomal epitopes. Nature 492, 280–284 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Tosi, A. et al. Structure and subunit topology of the INO80 chromatin remodeler and its nucleosome complex. Cell 154, 1207–1219 (2013).

    CAS  PubMed  Google Scholar 

  47. Ruthenburg, A.J. et al. Recognition of a mononucleosomal histone modification pattern by BPTF via multivalent interactions. Cell 145, 692–706 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Hamiche, A., Sandaltzopoulos, R., Gdula, D.A. & Wu, C. ATP-dependent histone octamer sliding mediated by the chromatin remodeling complex NURF. Cell 97, 833–842 (1999).

    CAS  PubMed  Google Scholar 

  49. Landry, J. et al. Essential role of chromatin remodeling protein Bptf in early mouse embryos and embryonic stem cells. PLoS Genet. 4, e1000241 (2008).

    PubMed  PubMed Central  Google Scholar 

  50. Song, H., Spichiger-Haeusermann, C. & Basler, K. The ISWI-containing NURF complex regulates the output of the canonical Wingless pathway. EMBO Rep. 10, 1140–1146 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Zhu, X. et al. An efficient genotyping method for genome-modified animals and human cells generated with CRISPR/Cas9 system. Sci. Rep. 4, 6420 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Lee, J.S. et al. AHR drives the development of gut ILC22 cells and postnatal lymphoid tissues via pathways dependent on and independent of Notch. Nat. Immunol. 13, 144–151 (2011).

    PubMed  PubMed Central  Google Scholar 

  53. Ye, B. et al. Cytosolic carboxypeptidase CCP6 is required for megakaryopoiesis by modulating Mad2 polyglutamylation. J. Exp. Med. 211, 2439–2454 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Chu, C., Qu, K., Zhong, F.L., Artandi, S.E. & Chang, H.Y. Genomic maps of long noncoding RNA occupancy reveal principles of RNA-chromatin interactions. Mol. Cell 44, 667–678 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank D.R. Littman (New York University, New York, New York, USA) for providing pMIGR plasmid, and P. Zhu (Institute of Biophysics, CAS, Beijing, China) for pcDNA4/TO-mSatb1 plasmid. C. rodentium was a gift from B. Ge (Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China). We thank J. Li (Cnkingbio Company Ltd., Beijing, China) for technical support. We also thank S. Meng, D. Fan, X. Wu, L. Zhou, Y. Teng and J. Jia for technical support. This work was supported by the National Natural Science Foundation of China (grants 91640203, 31530093, and 91419308 to Z.F.; 31470864 and 31670886 to B.Y.; and 31601189 to X.Z.) and the Strategic Priority Research Programs of the Chinese Academy of Sciences (grants XDB19030203 and XDA12020219 to Z.F.).

Author information

Authors and Affiliations

Authors

Contributions

B.L. designed and conducted experiments, analyzed data and wrote the paper; B.Y. carried out experiments and analyzed data; L.Y. and X.Z. constructed plasmids and established mouse tools; G.H., P. Z., J.W., Y.D. and X.Q. analyzed data; R.C. initiated the study and analyzed data; Y.T. designed animal experiments and analyzed data; and Z.F. initiated the study and organized, designed, and wrote the paper.

Corresponding authors

Correspondence to Runsheng Chen, Yong Tian or Zusen Fan.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Construction of lncKdm2b–RFP reporter and lncKdm2b-knockout mice.

(a) ILC3s (LinCD45loCD90hi) were isolated from small intestines, followed by immunofluorescence staining. LncKdm2b was detected by RNA fluorescence in situ hybridization (RNA-FISH). Green: lncKdm2b probe or scramble probe; red: CD127 or rat IgG2a-κ; purple: CD45 or rat IgG2b-κ. Nuclei were counterstained by DAPI. Scale bar, 5 μm. (b) Strategies for construction of lncKdm2bflox/flox and lncKdm2bRFP mice. (c) Total RNA from BM cells of lncKdm2bflox/flox or lncKdm2bflox/flox;Vav-Cre mice were extracted, followed by RNA hybridization. LncKdm2b specific probe was labeled with biotin. 18S RNA served as a loading control. (d,e) Analysis of ILC1s (d) and ILC2s (e) in small intestines from lncKdm2bflox/flox or lncKdm2bflox/flox;Vav-Cre mice by FACS. ILC1s were gated on CD3ɛCD45+NK1.1+NKp46+ and ILC2s were gated on LinThy1.2+CD25+KLRG1+ST2+. n=6 for each group. (f,g) Analysis of NK cells in spleen (f) and ILC1s in liver (g) from lncKdm2bflox/flox;Vav-Cre or lncKdm2bflox/flox;Vav-Cre+ mice. NK cells were gated on NKp46+NK1.1+. ILC1s were gated on CD3ɛDX5TCRβNK1.1+CD49a+. n=6 for each group. (h) Total RNAs were extracted from ILC3s derived from lncKdm2bflox/flox or lncKdm2bflox/floxRorc-Cre mice, followed by analysis of lncKdm2b expression with RT-qPCR. (i) Numbers of NKp46+ ILC3s, CD4+ ILC3s and CD4 ILC3s in lncKdm2b+/+ and lncKdm2b-/- mice were calculated and shown as means±SD. n=6 per group. (j,k) LncKdm2bflox/flox and lncKdm2bflox/floxRorc-Cre mice were infected with C. rodentium. 6 days later, bacterial counts (CFUs) in feces (j) and colon length (k) were measured. n=6 for each group. (l) LPLs were isolated from lncKdm2b+/+ and lncKdm2b-/- mice post C. rodentium infection, and stimulated with IL-23 for 4 h. Then IL-22+ ILC3s were analyzed by FACS. (m) Secreted IL-22 protein by LPLs as in l was tested by ELISA after stimulated with IL-23 for 24h. n=6 each group. *p<0.01 and **p<0.001 by two-tailed Student’s t test. All data are representative of three independent experiments.

Supplementary Figure 2 LncKdm2b interacts with Sabt1.

(a) BrdU+ ILC3s (LinCD45+CD127+RORγt+) were analyzed by FACS. n=6 for each group. (b) Percentages of total ILC3s (LinCD45.2+CD127+RORγt+) in chimeras were checked by FACS. n=6 for each group. (c) Expression levels of lncKdm2b neighboring genes in lncKdm2b+/+ and lncKdm2b−/-ILC3s were detected by RT-qPCR. Data were normalized to endogenous Actb and shown as means ± SD. (d) Identification of Satb1 as an lncKdm2b binding protein. Satb1 protein sequences were identified by LTQ Orbitrap XL. (e) Relative mRNA levels of Zfp292 were examined by real-time qPCR. Fold changes were normalized to endogenous Actb. (f) Enrichment of RNA pol II on Zfp292 promoter in lncKdm2b+/+ and lncKdm2b-/- ILC3s. Nuclei were extracted from lncKdm2b+/+ or lncKdm2b-/-ILC3s, followed by ChIP-qPCR assays. (g) Generation of Satb1-/- mice. The schematic diagram of Satb1 KO construction by CRISPR/Cas9 technology (upper left). Frameshift of Satb1 gene was confirmed by DNA sequencing (lower left). Satb1 deletion was validated by immunoblotting (right panel). *p<0.01 by two-tailed Student’s t test. All data are representative of three independent experiments.

Supplementary Figure 3 Sabt1 interacts with the NURF complex.

(a) Identification of Bptf and Snf2l as Satb1 binding proteins. Bptf and Snf2l protein sequences were identified by LTQ Orbitrap XL. (b) Generation of Zfp292-/- mice. The schematic diagram of Satb1 KO construction by CRISPR/Cas9 technology (upper left). Frameshift of Zfp292 gene was confirmed by DNA sequencing (lower left). Zfp292 deletion was verified by immunoblotting (right panel). (c) LPLs from Zfp292+/+ and Zfp292-/- mice were isolated and stimulated with IL-23 for 4 h. Numbers of IL-22+ ILC3s were examined by FACS. *p<0.01 by two-tailed Student’s t test. All data are representative of three independent experiments.

Supplementary Figure 4 Deletion of Sabt1 or Bptf leads to reduced numbers of ILC3s.

(a,b) Percentages (a) and numbers (b) of ILC3s in small intestines derived from Satb1+/+ and Satb1-/- mice. Percentages of indicated cells were calculated and shown at right panel (a). n=6 per group. (c,d) Percentages (c) and numbers (d) of ILC3s in small intestines derived from Bptfflox/flox and Bptfflox/floxRorc-Cre mice. Percentages of indicated cells were calculated and shown at right panel (c). n=6 per group. (e) Bptfflox/flox and Bptfflox/floxRorc-Cre mice were infected with C. rodentium (5x109 CFUs per mouse). 6 days later, bacterial counts (CFUs) in feces were measured. n=6 for each group. (f) Bptfflox/floxRorc-Cre and Bptfflox/floxRorc-Cre+ mice were infection with C. rodentium (5x109 CFUs per mouse) for 6 days, followed by H&E staining for colons. Scale bar, 50 μm. (g) 1x106 CD45.1 BM cells with 1x106 CD45.2 LPLs from WT or TKO mice infected with Zfp292 expressing retroviral were transplanted into lethally irradiated CD45.1 recipients. Percentages of ILC3s derived from CD45.2+ donors were analyzed by FACS. n=6 for each group. (h) Numbers of ILC3s as in g were analyzed by FACS. n=6 for each group. (i) Percentages of Ki67+ ILC3s in Zfp292+/+, lncKdm2b overexpression in Zfp292+/+ or Zfp292-/- LPLs were examined by FACS. n=6 for each group. *p<0.01 and **p<0.001 by two-tailed Student’s t test. All data are representative of at least three independent experiments.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–4 and Supplementary Tables 1 and 2 (PDF 1037 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Liu, B., Ye, B., Yang, L. et al. Long noncoding RNA lncKdm2b is required for ILC3 maintenance by initiation of Zfp292 expression. Nat Immunol 18, 499–508 (2017). https://doi.org/10.1038/ni.3712

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3712

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