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:

The histone demethylase UTX regulates the lineage-specific epigenetic program of invariant natural killer T cells

Abstract

Invariant natural killer T cells (iNKT cells) are innate-like lymphocytes that protect against infection, autoimmune disease and cancer. However, little is known about the epigenetic regulation of iNKT cell development. Here we found that the H3K27me3 histone demethylase UTX was an essential cell-intrinsic factor that controlled an iNKT-cell lineage-specific gene-expression program and epigenetic landscape in a demethylase-activity-dependent manner. UTX-deficient iNKT cells exhibited impaired expression of iNKT cell signature genes due to a decrease in activation-associated H3K4me3 marks and an increase in repressive H3K27me3 marks within the promoters occupied by UTX. We found that JunB regulated iNKT cell development and that the expression of genes that were targets of both JunB and the iNKT cell master transcription factor PLZF was UTX dependent. We identified iNKT cell super-enhancers and demonstrated that UTX-mediated regulation of super-enhancer accessibility was a key mechanism for commitment to the iNKT cell lineage. Our findings reveal how UTX regulates the development of iNKT cells through multiple epigenetic mechanisms.

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: H3K27 demethylases are essential for the maturation and development of iNKT cells.
Figure 2: UTX is required for the lineage-specific expression of signature genes in iNKT cell development.
Figure 3: UTX regulates the chromatin landscape of iNKT cells.
Figure 4: UTX occupies the promoters of iNKT cell signature genes that exhibit UTX-dependent chromatin regulation.
Figure 5: The demethylase activity of UTX is required for the generation of iNKT cells.
Figure 6: JunB partners with UTX to regulate the iNKT cell signature and development.
Figure 7: UTX deficiency impairs the activation of PLZF target genes in iNKT cells.
Figure 8: UTX facilitates the accessibility of super-enhancers in iNKT cells.

Similar content being viewed by others

Accession codes

Primary accessions

Gene Expression Omnibus

References

  1. Bendelac, A., Savage, P.B. & Teyton, L. The biology of NKT cells. Annu. Rev. Immunol. 25, 297–336 (2007).

    CAS  PubMed  Google Scholar 

  2. Rossjohn, J., Pellicci, D.G., Patel, O., Gapin, L. & Godfrey, D.I. Recognition of CD1d-restricted antigens by natural killer T cells. Nat. Rev. Immunol. 12, 845–857 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Kawano, T. et al. CD1d-restricted and TCR-mediated activation of Vα14 NKT cells by glycosylceramides. Science 278, 1626–1629 (1997).

    CAS  PubMed  Google Scholar 

  4. Cui, J. et al. Requirement for Vα14 NKT cells in IL-12-mediated rejection of tumors. Science 278, 1623–1626 (1997).

    CAS  PubMed  Google Scholar 

  5. Bendelac, A. Positive selection of mouse NK1+ T cells by CD1-expressing cortical thymocytes. J. Exp. Med. 182, 2091–2096 (1995).

    CAS  PubMed  Google Scholar 

  6. Lazarevic, V. et al. The gene encoding early growth response 2, a target of the transcription factor NFAT, is required for the development and maturation of natural killer T cells. Nat. Immunol. 10, 306–313 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Kovalovsky, D. et al. The BTB-zinc finger transcriptional regulator PLZF controls the development of invariant natural killer T cell effector functions. Nat. Immunol. 9, 1055–1064 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Savage, A.K. et al. The transcription factor PLZF directs the effector program of the NKT cell lineage. Immunity 29, 391–403 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Seiler, M.P. et al. Elevated and sustained expression of the transcription factors Egr1 and Egr2 controls NKT cell lineage differentiation in response to TCR signaling. Nat. Immunol. 13, 264–271 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Matsuda, J.L. et al. Homeostasis of Vα14i NKT cells. Nat. Immunol. 3, 966–974 (2002).

    CAS  PubMed  Google Scholar 

  11. Townsend, M.J. et al. T-bet regulates the terminal maturation and homeostasis of NK and Vα14i NKT cells. Immunity 20, 477–494 (2004).

    CAS  PubMed  Google Scholar 

  12. Engel, I. & Kronenberg, M. Transcriptional control of the development and function of Vα14i NKT cells. Curr. Top. Microbiol. Immunol. 381, 51–81 (2014).

    PubMed  Google Scholar 

  13. Cohen, N.R. et al. Shared and distinct transcriptional programs underlie the hybrid nature of iNKT cells. Nat. Immunol. 14, 90–99 (2013).

    CAS  PubMed  Google Scholar 

  14. Whyte, W.A. et al. Master transcription factors and mediator establish super-enhancers at key cell identity genes. Cell 153, 307–319 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Hnisz, D. et al. Super-enhancers in the control of cell identity and disease. Cell 155, 934–947 (2013).

    CAS  PubMed  Google Scholar 

  16. Kouzarides, T. Chromatin modifications and their function. Cell 128, 693–705 (2007).

    CAS  PubMed  Google Scholar 

  17. Rada-Iglesias, A. et al. A unique chromatin signature uncovers early developmental enhancers in humans. Nature 470, 279–283 (2011).

    CAS  PubMed  Google Scholar 

  18. Creyghton, M.P. et al. Histone H3K27ac separates active from poised enhancers and predicts developmental state. Proc. Natl. Acad. Sci. USA 107, 21931–21936 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Simon, J.A. & Kingston, R.E. Mechanisms of polycomb gene silencing: knowns and unknowns. Nat. Rev. Mol. Cell Biol. 10, 697–708 (2009).

    CAS  PubMed  Google Scholar 

  20. Bernstein, B.E. et al. A bivalent chromatin structure marks key developmental genes in embryonic stem cells. Cell 125, 315–326 (2006).

    CAS  PubMed  Google Scholar 

  21. Agger, K. et al. UTX and JMJD3 are histone H3K27 demethylases involved in HOX gene regulation and development. Nature 449, 731–734 (2007).

    CAS  PubMed  Google Scholar 

  22. Lan, F. et al. A histone H3 lysine 27 demethylase regulates animal posterior development. Nature 449, 689–694 (2007).

    CAS  PubMed  Google Scholar 

  23. Lee, M.G. et al. Demethylation of H3K27 regulates polycomb recruitment and H2A ubiquitination. Science 318, 447–450 (2007).

    CAS  PubMed  Google Scholar 

  24. De Santa, F. et al. The histone H3 lysine-27 demethylase Jmjd3 links inflammation to inhibition of polycomb-mediated gene silencing. Cell 130, 1083–1094 (2007).

    CAS  PubMed  Google Scholar 

  25. Heintzman, N.D. et al. Histone modifications at human enhancers reflect global cell-type-specific gene expression. Nature 459, 108–112 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Welstead, G.G. et al. X-linked H3K27me3 demethylase Utx is required for embryonic development in a sex-specific manner. Proc. Natl. Acad. Sci. USA 109, 13004–13009 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  27. Mansour, A.A. et al. The H3K27 demethylase Utx regulates somatic and germ cell epigenetic reprogramming. Nature 488, 409–413 (2012).

    CAS  PubMed  Google Scholar 

  28. Li, Q. et al. Critical role of histone demethylase Jmjd3 in the regulation of CD4+ T-cell differentiation. Nat. Commun. 5, 5780 (2014).

    CAS  PubMed  Google Scholar 

  29. Lee, S., Lee, J.W. & Lee, S.K. UTX, a histone H3-lysine 27 demethylase, acts as a critical switch to activate the cardiac developmental program. Dev. Cell 22, 25–37 (2012).

    CAS  PubMed  Google Scholar 

  30. Thieme, S. et al. The histone demethylase UTX regulates stem cell migration and hematopoiesis. Blood 121, 2462–2473 (2013).

    CAS  PubMed  Google Scholar 

  31. Ntziachristos, P. et al. Contrasting roles of histone 3 lysine 27 demethylases in acute lymphoblastic leukaemia. Nature 514, 513–517 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. van Haaften, G. et al. Somatic mutations of the histone H3K27 demethylase gene UTX in human cancer. Nat. Genet. 41, 521–523 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. Cook, K.D. et al. T follicular helper cell-dependent clearance of a persistent virus infection requires T cell expression of the histone demethylase UTX. Immunity 43, 703–714 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Manna, S. et al. Histone H3 lysine 27 demethylases Jmjd3 and Utx are required for T-cell differentiation. Nat. Commun. 6, 8152 (2015).

    PubMed  Google Scholar 

  35. Dobenecker, M.W. et al. Coupling of T cell receptor specificity to natural killer T cell development by bivalent histone H3 methylation. J. Exp. Med. 212, 297–306 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. Benlagha, K., Wei, D.G., Veiga, J., Teyton, L. & Bendelac, A. Characterization of the early stages of thymic NKT cell development. J. Exp. Med. 202, 485–492 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Lee, Y.J., Holzapfel, K.L., Zhu, J., Jameson, S.C. & Hogquist, K.A. Steady-state production of IL-4 modulates immunity in mouse strains and is determined by lineage diversity of iNKT cells. Nat. Immunol. 14, 1146–1154 (2013).

    CAS  PubMed  Google Scholar 

  38. Gapin, L., Matsuda, J.L., Surh, C.D. & Kronenberg, M. NKT cells derive from double-positive thymocytes that are positively selected by CD1d. Nat. Immunol. 2, 971–978 (2001).

    CAS  PubMed  Google Scholar 

  39. Mao, A.P. et al. Multiple layers of transcriptional regulation by PLZF in NKT cell-cell development. Proc. Natl. Acad. Sci. USA 113, 7602–7607 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  40. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Pinello, L., Xu, J., Orkin, S.H. & Yuan, G.C. Analysis of chromatin-state plasticity identifies cell-type-specific regulators of H3K27me3 patterns. Proc. Natl. Acad. Sci. USA 111, E344–E353 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  42. Kanda, M. et al. Transcriptional regulator Bhlhe40 works as a cofactor of T-bet in the regulation of IFN-γ production in iNKT cells. Proc. Natl. Acad. Sci. USA 113, E3394–E3402 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Engel, I. et al. Innate-like functions of natural killer T cell subsets result from highly divergent gene programs. Nat. Immunol. 17, 728–739 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Williams, K.L. et al. BATF transgenic mice reveal a role for activator protein-1 in NKT cell development. J. Immunol. 170, 2417–2426 (2003).

    CAS  PubMed  Google Scholar 

  45. Jordan-Williams, K.L., Poston, S. & Taparowsky, E.J. BATF regulates the development and function of IL-17 producing iNKT cells. BMC Immunol. 14, 16 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Lawson, V.J., Maurice, D., Silk, J.D., Cerundolo, V. & Weston, K. Aberrant selection and function of invariant NKT cells in the absence of AP-1 transcription factor Fra-2. J. Immunol. 183, 2575–2584 (2009).

    CAS  PubMed  Google Scholar 

  47. Thomsen, M.K. et al. JUNB/AP-1 controls IFN-γ during inflammatory liver disease. J. Clin. Invest. 123, 5258–5268 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Miller, S.A., Mohn, S.E. & Weinmann, A.S. Jmjd3 and UTX play a demethylase-independent role in chromatin remodeling to regulate T-box family member-dependent gene expression. Mol. Cell 40, 594–605 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Pereira, R.M. et al. Jarid2 is induced by TCR signalling and controls iNKT cell maturation. Nat. Commun. 5, 4540 (2014).

    CAS  PubMed  Google Scholar 

  50. Vahedi, G. et al. Super-enhancers delineate disease-associated regulatory nodes in T cells. Nature 520, 558–562 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. Passegué, E., Wagner, E.F. & Weissman, I.L. JunB deficiency leads to a myeloproliferative disorder arising from hematopoietic stem cells. Cell 119, 431–443 (2004).

    PubMed  Google Scholar 

  52. Irizarry, R.A. et al. Exploration, normalization, and summaries of high density oligonucleotide array probe level data. Biostatistics 4, 249–264 (2003).

    PubMed  Google Scholar 

  53. Johnson, W.E., Li, C. & Rabinovic, A. Adjusting batch effects in microarray expression data using empirical Bayes methods. Biostatistics 8, 118–127 (2007).

    PubMed  Google Scholar 

  54. Smyth, G.K. Linear models and empirical bayes methods for assessing differential expression in microarray experiments. Stat. Appl. Genet. Mol. Biol. 3, Article3 (2004).

    PubMed  Google Scholar 

  55. McLean, C.Y. et al. GREAT improves functional interpretation of cis-regulatory regions. Nat. Biotechnol. 28, 495–501 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Kurachi, M. et al. The transcription factor BATF operates as an essential differentiation checkpoint in early effector CD8+ T cells. Nat. Immunol. 15, 373–383 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  57. Langmead, B. & Salzberg, S.L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  59. Rousseeuw, P.J. Silhouettes: A graphical aid to the interpretation and validation of cluster analysis. J. Comput. Appl. Math. 20, 53–65 (1987).

    Google Scholar 

  60. Mathelier, A. et al. JASPAR 2014: an extensively expanded and updated open-access database of transcription factor binding profiles. Nucleic Acids Res. 42, D142–D147 (2014).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the US National Institutes of Health Tetramer Facility for CD1d-tetramers, the Center for Cancer Computational Biology sequencing facility at the Dana-Farber Cancer Institute for Illumina HiSeq2000 sequencing, the Microarray core facility at the Dana-Farber Cancer Institute Molecular Biology Core Facilities for microarray analysis, and the Dana-Farber Cancer Institute Jimmy Fund FACS core facility for cell sorting. Supported by the Howard Hughes Medical Institute (S.H.O.), the US National Institutes of Health (P30DK0492 to S.H.O., and RO1 AI083426 to F.W.), the Cancer Research Institute Predoctoral Emphasis Pathway in Tumor Immunology (S.B.), the National Research Foundation of Korea (2012R1A6A3A03040248 to J.H.K.) and the National Human Genome Research Institute of the US National Institutes of Health (Career Development Award K99HG008399 to L.P.).

Author information

Authors and Affiliations

Authors

Contributions

S.B. designed, performed and interpreted experiments involving gene expression analysis, ChIP-Seq, ATAC-Seq, lentiviral transduction, qRT-PCR, ChIP-PCR and immunoprecipitation, with help from M.E.X., and generated UTX-KO and JMJD3-KO mice, with help from M.A.K.; J.H.K. designed, performed and interpreted experiments involving iNKT cell analysis by flow cytometry with help from Y.H.; L.P. designed, performed and interpreted bioinformatics analysis with help from J.H.; P.P.D., R.A.B. and R.D. assisted with ChIP-Seq analysis; A.H. and E.P. provided JunB-KO mice; W.N.H. and Ö.H.Y. participated in the design and interpretation of experiments; G.-C.Y. supervised bioinformatics analysis; S.H.O. and F.W. designed and supervised experiments; and S.B., J.H.K., S.H.O. and F.W. wrote the manuscript with support from L.P.

Corresponding authors

Correspondence to Stuart H Orkin or Florian Winau.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Generation of UTX-KO and JMJD3-KO mice.

(a) Utx locus in UTX-targeted embryonic stem cells (ESCs) that were provided from EUCOMM. (b) PCR analysis verifying loxP sites targeting exon 3 of Utx in ESC clones. (c) Targeting strategy for Jmjd3 locus to delete exons 17-19. (d) Southern blot analysis of Jmjd3-targeted ESC clones. (e) Representative genotyping of conditional UTX-or JMJD3-KO mice. (f) Analysis of Utx or Jmjd3 mRNA levels in blood, DP thymocytes, or iNKT cells by qRT-PCR using deletion-specific primers. Data are mean ± s.d. from three independent experiments.

Supplementary Figure 2 UTX is required for development of iNKT cells but not for their function.

(a) Conditional UTX-deficient (UTX KO) mice or wild-type (WT) littermates were injected intraperitoneally with 2 μg α-GalCer. Two hours later, lymphocytes from thymus and liver were collected for intracellular staining of IFN-γ and IL-4, prior to analysis by flow cytometry. Additionally, thymic iNKT cells were restimulated with PMA and Ionomycin for 4 hours before analysis. (b) Flow cytometry analysis of WT or UTX KO thymic iNKT cells by measurement of transcription factors T-bet, PLZF, and RORγt, gated on CD1d-tetramer+ cells. Shown are the absolute cell counts of NKT1, NKT2, or NKT17 cells among thymic iNKT cells from WT or UTX KO (n = 3). (c) Analysis of CD1d expression on DP thymocytes from WT or UTX KO mice using flow cytometry. (d) DP thymocytes were isolated from WT or UTX KO mice. Subsequently, mRNA transcripts of Vα14-Jα18 TCR were analyzed by qRT-PCR and normalized to constant alpha chain (Cα) amplification. Y-axis depicts relative expression (n = 5). Results in (b-d) are representative of three independent experiments. Data are mean ± s.e.m. **P < 0.01 as analyzed by unpaired t-test.

Supplementary Figure 3 Characterization of gene-expression alterations in control and UTX-KO thymic iNKT cells.

(a) Heat map of gene expression microarray (WT; n=5, KO; n=4). (b) GSEA of downregulated or (c) upregulated genes in UTX-deficient (KO) iNKT cells compared to wild-type (WT). ES, enrichment score; NES, normalized ES; FDR, false discovery rate. (d) iNKT cells in stage 0 and stage 1-3 have reduced signature gene expression in UTX KO as assessed by qRT-PCR. (e) Confirming the loss of UTX transcripts in stage 0 and stage 1-3 iNKT cells in UTX KO as assessed by qRT-PCR. Data are mean ± s.d. from three independent experiments.

Supplementary Figure 4 Distribution of H3K27me3 regions in control and UTX-KO iNKT cells.

(a,b) Chromosomal distribution of WT-specific (a) or UTX-KO-specific (b) H3K27me3 peaks in iNKT cells. (c,d) Distribution of WT-specific (c) or UTX-KO-specific (d) H3K27me3 peaks within the genome of iNKT cells.

Supplementary Figure 5 Characterization of regions with H3K27me3 peaks in control and UTX-KO iNKT cells.

(a) GREAT analysis of WT-specific H3K27me3 peaks (n = 4,250). (b) GREAT analysis of UTX KO-specific H3K27me3 peaks (n = 9,645). (c,d) Assessing statistical significance of H3K27me3 distribution around the promoters of downregulated (c) and upregulated (d) genes between WT and UTX-deficient (KO) iNKT cells based on permutation test where 100,000 permutations were used to calculate the distribution of the difference between two average profiles. Black line depicts empirical null model, dotted red line indicates the observed value for the measured profiles. (e,f) GREAT analysis of all downregulated (e) or upregulated (f) genes in UTX KO iNKT cells compared to WT controls.

Supplementary Figure 6 Cluster analysis of the chromatin landscape around gene promoters in control or UTX-KO iNKT cells.

Heat maps represent the abundance of H3K4me3 and H3K27me3 marks around the promoters of downregulated (a) or upregulated genes (b) in UTX-deficient (KO) iNKT cells compared to wild-type (WT). Genes are subdivided into different clusters dependent on their histone mark pattern. C1-4 = clusters 1 to 4. (c) Integrating gene expression with chromatin state for upregulated genes. Average abundance of H3K27me3 and H3K4me3 around upregulated gene promoters is depicted as z-score in reads per million (rpm). Average profiles are grouped in three clusters of upregulated genes with WT in blue and KO in green. The upper panel shows expression of upregulated genes as logarithmic fold change (log2FC). (d) GREAT analysis of downregulated genes in UTX-deficient iNKT cells in cluster 3 and cluster 4.

Supplementary Figure 7 Intrinsic function of UTX is responsible for development of iNKT cells.

Mixed bone marrow-chimeric mice were generated using a 1:1 ratio of wild-type (WT) and conditional UTX-deficient (KO) bone marrow cells for transfer into Rag2-/- hosts (n = 5). Twelve weeks later, WT cells were detected by anti-CD45.1, and frequencies of thymic iNKT cells were analyzed by flow cytometry. (a) Depicted are the ratios of WT to KO iNKT cells gated on the different maturational stages. (b) Bar graph depicting the results from (a). (c) In the same experiment as described in (a), lymphocytes from liver were analyzed for iNKT cells (CD3+tetramer+) as well as conventional T cells (CD3+tetramer-). Depicted are the ratios of WT to KO T lymphocytes. Results are representative of two independent experiments.

Supplementary Figure 8 Characterization of iNKT cell super-enhancers.

(a,b) Epigenetic landscape of the super-enhancers for Tbx21 (a) and Il2rb (b) that exhibit UTX-dependent accessibility. Depicted are the ChIP-Seq tracks of H3K27ac and heat maps of ATAC-Seq, H3K27me3, and H3K4me3 in WT or UTX-deficient (KO) iNKT cells around the super-enhancer (SE) for Tbx21 (a) and Il2rb (b). Zoomed in tracks in Fig. 8f and Fig. 8g are shown in black rectangle and highlighted with a star. (c) GREAT analysis of the super-enhancer elements in iNKT cells. (d) GREAT analysis of the SE elements in iNKT cells that exhibit UTX-dependent accessibility. (e) Transcription factor target motif analysis using Haystack in iNKT cell SE elements that exhibit UTX-dependent accessibility. Depicted are the binding motifs of RELA and BHLHE40.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–8 (PDF 1767 kb)

Supplementary Table 1

List of genes that exhibit UTX-dependent chromatin regulation in cluster 3 and cluster 4 (XLS 47 kb)

Supplementary Table 2

List of JunB target genes in cluster 3 (XLSX 12 kb)

Supplementary Table 3

List of iNKT super enhancers (XLSX 59 kb)

Supplementary Table 4

List of super-enhancer regions that lost accessibility in UTX-KO (WT-specific) or gained accessibility in UTX-KO (KO-specific) (XLS 34 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Beyaz, S., Kim, J., Pinello, L. et al. The histone demethylase UTX regulates the lineage-specific epigenetic program of invariant natural killer T cells. Nat Immunol 18, 184–195 (2017). https://doi.org/10.1038/ni.3644

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3644

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