Abstract
Viral infection outcomes are sex biased, with males generally more susceptible than females. Paradoxically, the numbers of antiviral natural killer (NK) cells are increased in males. We demonstrate that while numbers of NK cells are increased in male mice, they display decreased effector function compared to females in mice and humans. These differences were not solely dependent on gonadal hormones, because they persisted in gonadectomized mice. Kdm6a (which encodes the protein UTX), an epigenetic regulator that escapes X inactivation, was lower in male NK cells, while NK cell-intrinsic UTX deficiency in female mice increased NK cell numbers and reduced effector responses. Furthermore, mice with NK cell-intrinsic UTX deficiency showed increased lethality to mouse cytomegalovirus. Integrative multi-omics analysis revealed a critical role for UTX in regulating chromatin accessibility and gene expression critical for NK cell homeostasis and effector function. Collectively, these data implicate UTX as a critical molecular determinant of sex differences in NK cells.
This is a preview of subscription content, access via your institution
Access options
Access Nature and 54 other Nature Portfolio journals
Get Nature+, our best-value online-access subscription
$32.99 / 30 days
cancel any time
Subscribe to this journal
Receive 12 print issues and online access
$259.00 per year
only $21.58 per issue
Buy this article
- Purchase on SpringerLink
- Instant access to full article PDF
Prices may be subject to local taxes which are calculated during checkout






Similar content being viewed by others
Data availability
Sequencing datasets are accessible from the Gene Expression Omnibus under accession number GSE185065. Source data are provided with this paper. Further information and requests for resources and reagents should be directed and will be fulfilled by the corresponding authors.
References
Wilkinson, N. M., Chen, H. C., Lechner, M. G. & Su, M. A. Sex differences in immunity. Annu Rev. Immunol. 40, 75–94 (2022).
Klein, S. L. & Flanagan, K. L. Sex differences in immune responses. Nat. Rev. Immunol. 16, 626–638 (2016).
Pardue, M.-L. & Wizemann, T. M. Exploring the biological contributions to human health: does sex matter? The National Academies Press https://doi.org/10.17226/10028 (2001).
Gianella, S. et al. Sex differences in CMV replication and HIV persistence during suppressive ART. Open Forum Infect. Dis. 7, ofaa289 (2020).
Takahashi, T. & Iwasaki, A. Sex differences in immune responses. Science 371, 347–348 (2021).
Patin, E. et al. Natural variation in the parameters of innate immune cells is preferentially driven by genetic factors. Nat. Immunol. 19, 302–314 (2018).
Huang, Z. et al. Effects of sex and aging on the immune cell landscape as assessed by single-cell transcriptomic analysis. Proc. Natl Acad. Sci. USA 118, e2023216118 (2021).
Talebizadeh, Z., Simon, S. D. & Butler, M. G. X chromosome gene expression in human tissues: male and female comparisons. Genomics 88, 675–681 (2006).
Fang, H., Disteche, C. M. & Berletch, J. B. X inactivation and escape: epigenetic and structural features. Front. Cell Dev. Biol. 7, 219 (2019).
Chen, X. et al. Sex difference in neural tube defects in p53-null mice is caused by differences in the complement of X not Y genes. Dev. Neurobiol. 68, 265–273 (2008).
Smith-Bouvier, D. L. et al. A role for sex chromosome complement in the female bias in autoimmune disease. J. Exp. Med. 205, 1099–1108 (2008).
Souyris, M. et al. TLR7 escapes X chromosome inactivation in immune cells. Sci. Immunol. 3, eaap8855 (2018).
Hammer, Q., Ruckert, T. & Romagnani, C. Natural killer cell specificity for viral infections. Nat. Immunol. 19, 800–808 (2018).
Orange, J. S. Natural killer cell deficiency. J. Allergy Clin. Immunol. 132, 515–525 (2013).
Bukowski, J. F., Warner, J. F., Dennert, G. & Welsh, R. M. Adoptive transfer studies demonstrating the antiviral effect of natural killer cells in vivo. J. Exp. Med. 161, 40–52 (1985).
Brown, M. G. et al. Vital involvement of a natural killer cell activation receptor in resistance to viral infection. Science 292, 934–937 (2001).
Welsh, R. M., Brubaker, J. O., Vargas-Cortes, M. & O’Donnell, C. L. Natural killer (NK) cell response to virus infections in mice with severe combined immunodeficiency. The stimulation of NK cells and the NK cell-dependent control of virus infections occur independently of T and B cell function. J. Exp. Med. 173, 1053–1063 (1991).
Bancroft, G. J., Shellam, G. R. & Chalmer, J. E. Genetic influences on the augmentation of natural killer cells during murine cytomegalovirus infection: correlation with patterns of resistance. J. Immunol. 126, 988–994 (1981).
Menees, K. B. et al. Sex- and age-dependent alterations of splenic immune cell profile and NK cell phenotypes and function in C57BL/6J mice. Immun. Ageing 18, 3 (2021).
Mujal, A. M., Delconte, R. B. & Sun, J. C. Natural killer cells: from innate to adaptive features. Annu. Rev. Immunol. 39, 417–447 (2021).
Loh, J., Chu, D. T., O’Guin, A. K., Yokoyama, W. M. & Virgin, H. W. T. Natural killer cells utilize both perforin and gamma interferon to regulate murine cytomegalovirus infection in the spleen and liver. J. Virol. 79, 661–667 (2005).
Orange, J. S., Wang, B., Terhorst, C. & Biron, C. A. Requirement for natural killer cell-produced interferon gamma in defense against murine cytomegalovirus infection and enhancement of this defense pathway by interleukin-12 administration. J. Exp. Med. 182, 1045–1056 (1995).
Nakaya, M., Tachibana, H. & Yamada, K. Effect of estrogens on the interferon-gamma producing cell population of mouse splenocytes. Biosci. Biotechnol. Biochem. 70, 47–53 (2006).
Chiossone, L. et al. Maturation of mouse NK cells is a 4-stage developmental program. Blood 113, 5488–5496 (2009).
Wainer Katsir, K. & Linial, M. Human genes escaping X-inactivation revealed by single-cell expression data. BMC Genomics 20, 201 (2019).
Yang, F., Babak, T., Shendure, J. & Disteche, C. M. Global survey of escape from X inactivation by RNA-sequencing in mouse. Genome Res. 20, 614–622 (2010).
Berletch, J. B. et al. Escape from X inactivation varies in mouse tissues. PLoS Genet. 11, e1005079 (2015).
Arnold, A. P. Four core genotypes and XY* mouse models: update on impact on SABV research. Neurosci. Biobehav. Rev. 119, 1–8 (2020).
Hasegawa, H. et al. Activation of p53 by Nutlin-3a, an antagonist of MDM2, induces apoptosis and cellular senescence in adult T-cell leukemia cells. Leukemia 23, 2090–2101 (2009).
Riggan, L. et al. The transcription factor Fli1 restricts the formation of memory precursor NK cells during viral infection. Nat. Immunol. 23, 556–567 (2022).
Min-Oo, G., Bezman, N. A., Madera, S., Sun, J. C. & Lanier, L. L. Proapoptotic Bim regulates antigen-specific NK cell contraction and the generation of the memory NK cell pool after cytomegalovirus infection. J. Exp. Med. 211, 1289–1296 (2014).
Louis, C. et al. NK cell-derived GM-CSF potentiates inflammatory arthritis and is negatively regulated by CIS. J. Exp. Med. 217, e20191421 (2020).
Bjorkstrom, N. K., Strunz, B. & Ljunggren, H. G. Natural killer cells in antiviral immunity. Nat. Rev. Immunol. 22, 112–123 (2022).
Smyth, M. J. et al. Perforin is a major contributor to NK cell control of tumor metastasis. J. Immunol. 162, 6658–6662 (1999).
Van der Meulen, J., Speleman, F. & Van Vlierberghe, P. The H3K27me3 demethylase UTX in normal development and disease. Epigenetics 9, 658–668 (2014).
Wang, S. P. et al. A UTX-MLL4-p300 transcriptional regulatory network coordinately shapes active enhancer landscapes for eliciting transcription. Mol. Cell 67, 308–321 (2017).
Gozdecka, M. et al. UTX-mediated enhancer and chromatin remodeling suppresses myeloid leukemogenesis through noncatalytic inverse regulation of ETS and GATA programs. Nat. Genet. 50, 883–894 (2018).
Wang, C. et al. UTX regulates mesoderm differentiation of embryonic stem cells independent of H3K27 demethylase activity. Proc. Natl Acad. Sci. USA 109, 15324–15329 (2012).
Shih, H. Y. et al. Developmental acquisition of regulomes underlies innate lymphoid cell functionality. Cell 165, 1120–1133 (2016).
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).
Kaya-Okur, H. S. et al. CUT&Tag for efficient epigenomic profiling of small samples and single cells. Nat. Commun. 10, 1930 (2019).
Kupz, A. et al. Contribution of Thy1+ NK cells to protective IFN-gamma production during Salmonella typhimurium infections. Proc. Natl Acad. Sci. USA 110, 2252–2257 (2013).
Langmead, B. & Salzberg, S. L. Fast gapped-read alignment with Bowtie 2. Nat. Methods 9, 357–359 (2012).
Chen, E. Y. et al. Enrichr: interactive and collaborative HTML5 gene list enrichment analysis tool. BMC Bioinformatics 14, 128 (2013).
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).
Rapp, M. et al. Core-binding factor beta and Runx transcription factors promote adaptive natural killer cell responses. Sci. Immunol. 2, eaan3796 (2017).
Simonetta, F., Pradier, A. & Roosnek, E. T-bet and eomesodermin in NK cell development, maturation and function. Front. Immunol. 7, 241 (2016).
Presnell, J. S., Schnitzler, C. E. & Browne, W. E. KLF/SP transcription factor family evolution: expansion, diversification and innovation in eukaryotes. Genome Biol. Evol. 7, 2289–2309 (2015).
Kramer, B. et al. Early IFN-alpha signatures and persistent dysfunction are distinguishing features of NK cells in severe COVID-19. Immunity 54, 2650–2669 (2021).
D’Agostino, P. et al. Sex hormones modulate inflammatory mediators produced by macrophages. Ann. N. Y. Acad. Sci. 876, 426–429 (1999).
Lu, F. X. et al. The strength of B cell immunity in female rhesus macaques is controlled by CD8+ T cells under the influence of ovarian steroid hormones. Clin. Exp. Immunol. 128, 10–20 (2002).
Singh, R. P. & Bischoff, D. S. Sex hormones and gender influence the expression of markers of regulatory T cells in SLE patients. Front. Immunol. 12, 619268 (2021).
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).
Beyaz, S. et al. The histone demethylase UTX regulates the lineage-specific epigenetic program of invariant natural killer T cells. Nat. Immunol. 18, 184–195 (2017).
Mitchell, J. E. et al. UTX promotes CD8+ T cell-mediated antiviral defenses but reduces T cell durability. Cell Rep. 35, 108966 (2021).
Schmiedel, B. J. et al. Impact of genetic polymorphisms on human immune cell gene expression. Cell 175, 1701–1715 (2018).
Bosselut, R. Pleiotropic functions of H3K27Me3 demethylases in immune cell differentiation. Trends Immunol. 37, 102–113 (2016).
Kechin, A., Boyarskikh, U., Kel, A. & Filipenko, M. cutPrimers: a new tool for accurate cutting of primers from reads of targeted next generation sequencing. J. Comput. Biol. 24, 1138–1143 (2017).
Hong, S. et al. Identification of JmjC domain-containing UTX and JMJD3 as histone H3 lysine 27 demethylases. Proc. Natl Acad. Sci. USA 104, 18439–18444 (2007).
Van Laarhoven, P. M. et al. Kabuki syndrome genes KMT2D and KDM6A: functional analyses demonstrate critical roles in craniofacial, heart and brain development. Hum. Mol. Genet. 24, 4443–4453 (2015).
Rezvani, K. Adoptive cell therapy using engineered natural killer cells. Bone Marrow Transpl. 54, 785–788 (2019).
Eckelhart, E. et al. A novel Ncr1-Cre mouse reveals the essential role of STAT5 for NK-cell survival and development. Blood 117, 1565–1573 (2011).
Lau, C. M. et al. Epigenetic control of innate and adaptive immune memory. Nat. Immunol. 19, 963–972 (2018).
Smith, L. M., McWhorter, A. R., Masters, L. L., Shellam, G. R. & Redwood, A. J. Laboratory strains of murine cytomegalovirus are genetically similar to but phenotypically distinct from wild strains of virus. J. Virol. 82, 6689–6696 (2008).
Weizman, O. E. et al. ILC1 confer early host protection at initial sites of viral infection. Cell 171, 795–808 (2017).
Zhang, Y. et al. Model-based analysis of ChIP–seq (MACS). Genome Biol. 9, R137 (2008).
Anders, S., Pyl, P. T. & Huber, W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).
Love, M. I., Huber, W. & Anders, S. Moderated estimation of fold change and dispersion for RNA-seq data with DESeq2. Genome Biol. 15, 550 (2014).
Dembele, D. & Kastner, P. Fuzzy C-means method for clustering microarray data. Bioinformatics 19, 973–980 (2003).
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).
Raudvere, U. et al. g:Profiler: a web server for functional enrichment analysis and conversions of gene lists (2019 update). Nucleic Acids Res. 47, W191–W198 (2019).
Acknowledgements
We thank members of the T.E.O. and M.A.S. laboratories for helpful discussion. We thank the UCLA Technology Center for Genomics and Bioinformatics for RNA-seq library preparation and the Cedars Sinai Applied Genomics, Computation, and Translational Core Facility for ATAC-seq library preparation. T.E.O. is supported by the National Institutes of Health (NIH; AI145997) and UC CRCC (CRN-20-637105). We thank the blood donors and UCLA/CFAR Virology Core Laboratory for providing human PBMCs for study, funded by UCLA CFAR grant 5P30 AI028697. We thank M. Lechner for use of a BioRender license to produce the schematic in Fig. 6. M.A.S. is supported by the NIH (NS107851, AI143894, DK119445), Department of Defense (USAMRAA PR200530) and National Organization of Rare Diseases. M.I.C. is supported by Ruth L. Kirschstein National Research Service Awards (GM007185 and AI007323), and a Whitcome Fellowship from the Molecular Biology Institute at UCLA. L.R. is supported by the Warsaw fellowship from the MIMG department at UCLA. J.H.L. is supported by the NIH NIAMS (T32AR071307) and the UCLA Medical Scientist Training Program (NIH NIGMS T32GM008042). A.P.A. is supported by the NIH (HD100298).
Author information
Authors and Affiliations
Contributions
M.I.C., L.R., J.H.L., R.Y.T., H.H., A.P.A., T.E.O. and M.A.S. designed the study; M.I.C., J.H.L., R.Y.T., L.R. and S.C. performed the experiments; M.I.C., F.M., B.C. and M.P. performed bioinformatics analysis; M.I.C., M.A.S., J.H.L. and T.E.O. wrote the paper.
Corresponding authors
Ethics declarations
Competing interests
T.E.O. is a scientific advisor for Modulus Therapeutics and Xyphos companies that have financial interest in human NK cell-based therapeutics. The other authors declare no competing interests.
Peer review
Peer review information
Nature Immunology thanks Mihalis Verykokakis, Edith Heard, and the other, anonymous, reviewers for their contribution to the peer review of this work. Primary Handling Editor: L. A. Dempsey, in collaboration with the rest of the Nature Immunology team. Peer reviewer reports are available.
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 Sex differences in IFN-γ production in response to IL-12/18.
a) Representative dot plots showing gating strategy to identify CD3− TCRβ- NK1.1+ mouse NK cells. b) Representative contour plots, (c) percentage IFN-γ+, and (d) normalized IFN-γ MFI of female and male WT NK cells with cultured no treatment (NT) or IL-12 (20 ng/mL) and IL-18 (10 ng/mL) for 4 hours (n = 14 per group). e) Representative contour plots of CD3−CD56+ female (n = 6) and male (n = 7) human NK cells cultured and stimulated with 10 ng/mL of IL-12 for 16 hours in the presence of K562 cells. f) Representative dot plots of splenic NK cells (CD3−TCRβ- NK1.1+) in gonadectomized female and male mice. g) Representative contour plots of total splenic NK cells isolated from gonadectomized female and male mice and cultured with no treatment (NT) or IL-15 (50 ng/mL) and IL-12 (20 ng/mL) for 4 hours. h) Representative contour plots and i) percentage CD27−CD11b− (DN), CD27+CD11b− (CD27 SP), CD27+CD11b+ (DP), and CD27−CD11b+ (CD11b SP) of total splenic NK cells from female and male mice (n = 7 per group). j) Representative contour plots and k) percentage CD27−CD11b− (DN), CD27+CD11b− (CD27 SP), CD27+CD11b+ (DP), and CD27−CD11b+ (CD11b SP) of total splenic NK cells from gonadectomized mice (n = 6 per group). Data are representative of 2-3 independent experiments. Samples were compared using unpaired two-tailed Student’s t test and data points are presented as individual mice with the mean ± SEM (N.S., Not Significant; **, p < 0.01; ****, p < 0.0001). Specific p-values are as follows: c:[NT = 0.987; IL-12+IL-18 = 0.00125]; d:[NT = 0.4144; IL-12+IL-18 < 0.0001]; i:[DN > 0.999; CD27 SP = 0.9514; DP = 0.8995; CD11b SP = 0.6517]; k:[DN = 0.0951; CD27 SP = 0.225; DP = 0.161; CD11b SP = 0.789].
Extended Data Fig. 2 UTX expression in Four Core Genotypes mice and maturation in UTX mouse models.
a) Relative expression of Kdm6a (encodes the protein UTX) by RT-qPCR in Four Core Genotypes mice, in which male or female gonads present are independent of XX or XY chromosome composition, normalized to female WT expression (XX with ovaries) (XX ovaries: n = 4; XY ovaries: n = 5; XX testes: n = 3; XY testes: n = 5). b) Relative UTX MFI in splenic NK cells isolated from female WT (n = 19), male WT (n = 19), female UTXHet (n = 7), and female UTXNKD (n = 4) mice. c) CD11b and CD27 expression within NK cells isolated from female WT (n = 13), male WT (n = 7), and female UTXHet (n = 8) mice. d) Representative western blot showing protein expression of UTX in NK cells isolated from female WT and UTXNKD mice compared to β-actin loading control. Data are representative of 2-3 independent experiments. Samples were compared using a) unpaired two-tailed Student’s t test, b,c) one-way ANOVA with Tukey’s correction for multiple comparisons. Data points are presented as individual mice with the mean ± SEM (N.S., Not Significant; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001). Specific p-values are as follows: a:[Ovaries – XX vs. XY < 0.0001; Testes – XX vs. XY = 0.0016]; b:[F WT vs. M WT = 0.003; F UTXNKD vs. F UTXHet and F WT vs. F UTXNKD < 0.0001; M WT vs. F UTXHet = 0.7435; F UTXHet vs. F WT = 0.0004]; c > 0.999.
Extended Data Fig. 3 UTX regulates NK cell fitness.
a) Representative flow cytometry plots of splenic NK cells from female WT, male WT, and female UTXHet, and female UTXNKD mice. b) Representative contour plots and c) percent of WT(CD45.1+) or UTXNKD(CD45.2+) cells of total splenic T (left) and NK (right) cells in 1:1 WT:UTXNKD mBMC mice (n = 3). d) Percentage of Ki67+ cells in blood of 4:1 WT:UTXNKD mBMCs (n = 28), injection ratio used to normalize WT and UTXNKD NK cell numbers. e) Representative histogram (left) and quantification (right) of CFSE expansion, division and proliferation indices calculated using FlowJo’s Proliferation tool of CFSE-labeled splenic NK cells isolated from WT and UTXNKD mice stimulated ex vivo with IL-15 (50 ng/mL) for 4 days (n = 4). f) Schematic showing adoptive transfer of CTV-labeled congenically distinct WT (CD451x2) and UTXNKD (CD45.2+) NK cells transferred into WT (CD45.1+) recipients at a 1:1 ratio with analysis of CTV dilution and WT:UTXNKD ratio on D7 by flow cytometry. g) Representative histograms showing CTV dilution of congenically distinct WT (CD451x2) and UTXNKD (CD45.2+) NK cells transferred into WT (CD45.1+) recipients before transfer (left) and on day 7 post-transfer (right). h) Representative histograms and i) percentage of cleaved caspase 3+ splenic NK cells from female WT and male WT treated ex vivo with IL-15 (5 ng/mL) and DMSO (Female WT: n = 7; Male WT: n = 8) or 2.5 uM Nutlin-3a (Female WT: n = 3; Male WT: n = 4) for 24 hours. j) Representative histograms and k) percentage of cleaved caspase 3+ splenic NK cells from gonadectomized female and male mice (n = 6) treated ex vivo with IL-15 (5 ng/mL) and DMSO or 2.5uM Nutlin-3a for 24 h. l) Representative histograms of Bcl-2 (left) and Bim (right) of flow cytometry in splenic NK cells from female WT and UTXNKD mice (n = 5). “Ctrl”:unstained flow cytometry controls. Data are representative of 2-3 independent experiments. Samples were compared using unpaired two-tailed Student’s t test with Welch’s correction and data points are presented as individual mice with the mean ± SEM (N.S., Not Significant; *, p < 0.05; **, p < 0.01; ****, p < 0.0001). Specific p-values are as follows: c:[T cells=0.705; NK Cells=0.0202]; d < 0.0001; e:[Expansion=0.0192; Division=0.0253; Proliferation=0.0032]; i:[DMSO = 0.0096; Nutlin-3a = 0.0473]; k:[DMSO = 0.001; Nutlin-3a = 0.0013].
Extended Data Fig. 4 UTX enhances effector function independent of gonadal hormone and maturation.
a) Representative contour plots of total NK cells from female WT, male WT, female UTXHet, and female UTXNKD mice cultured with IL-15 (50 ng/mL) and IL-12 (20 ng/mL) for 4 h. b) Absolute number of IFN-γ+ NK cells from female WT, male WT, and female UTXHet mice stimulated with no treatment (NT) or IL-15 (50 ng/mL) and IL-12 (20 ng/mL) for 4 h (n = 8). c) Specific lysis of MHC Class I deficient MC38 (Target) cells by female WT (n = 4), male WT (n = 5), and gonadectomized female (n = 3) and male (n = 6) NK cells for 16 h at 4:1 effector:target ratio, normalized to lysis by female WT. d) Representative histograms and e) MFI of CD107a (n = 5 per group), granzyme b (GzmB) (Female:n = 5; Male:n = 4), and perforin (n = 5) of female WT and male WT NK cells incubated with IL-15 (50 ng/mL) only or additionally stimulated with plate-bound anti-NK1.1 antibody (PK136). “Ctrl” refers to matched unstained control for flow cytometry. Representative f) contour plots of IFN-γ and g) histogram of GzmB expressing total splenic NK cells on D1.5 post MCMV infection of 4:1 WT:UTXNKD mixed bone marrow chimeras (mBMCs), ratio used to normalize cell numbers between genotypes. h) IFN-γ protein production in UTXNKD compared to WT NK cells within maturation subsets: CD27−CD11b− (DN), CD27+CD11b− (CD27 SP), CD27+CD11b+ (DP), and CD27−CD11b+ (CD11b SP) isolated from 4:1 WT:UTXNKD mBMC 1.5 days post-MCMV infection (n = 6). i) Representative contour plots of total NK cells derived from 1:1 WT:iUTX-/- mBMC mice on day 1.5 post MCMV infection, normalized to WT (n = 6). Data are representative of 2-3 independent experiments. Samples were compared using paired two-tailed Student’s t test and data points are presented as individual mice with the mean ± SEM (N.S., Not Significant; *, p < 0.05; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001). Specific p-values are as follows: b:[F WT vs. M WT = 0.0162; M WT vs. UTXHet = 0.968; F WT vs. F UTXHet = 0.0288]; c:[F WT vs. F Gonadectomy=0.245; F WT vs. M WT = 0.0086; M WT vs. M Gonadectomy=0.998; F Gonadectomy vs. M Gonadectomy=0.0247]; e:[CD107a – NT = 0.54 and NK1.1 = 0.772; GzmB – NT = 0.0004 and NK1.1 = 0.00129; Perforin – NT = 0.0101 and NK1.1 < .0001]; h:[DN = 0.03281; CD27 SP = 0.0186; DP = 0.0231; CD11b SP = 0.0114].
Extended Data Fig. 5 UTY is expressed but not sufficient to compensate for loss of UTX in NK cell homeostasis and effector function.
a) Expression in transcripts per million of KDM6A (encodes the protein UTX) and KDM6C (encodes the protein UTY) using DICE database RNA-seq data on sorted NK cells from human females (n = 36) vs. males (n = 54). b) Relative expression of Kdm6a (encodes the protein UTX) and Kdm6c (encodes the protein UTY) by RT-qPCR in splenic NK cells isolated from female WT, male WT, and gonadectomized female and male mice (n = 6 per group). c) Representative flow cytometry dot plots and quantification of d) frequency and absolute numbers of NK cells in spleen of male WT and UTXNKD mice (n = 8 per group). e) Representative flow cytometry contour plots and quantification of f) percentage IFN-γ + and normalized IFN-γ MFI of total NK cells from male WT vs. male UTXNKD mice with no treatment (NT) or in response to IL-15 (50 ng/mL) and IL-12 (20 ng/mL) stimulation for 4 hours ex vivo, MFI normalized to male WT (n = 8 per group). g) Representative flow cytometry contour plots and quantification of h) percentage IFN-γ+ and normalized IFN-γ MFI of total NK cells from male WT vs. male UTXNKD mice with no treatment (NT) or in response to IL-12 (20 ng/mL) and IL-18 (10 ng/mL) stimulation for 4 hours ex vivo, MFI normalized to male WT (n = 8 per group). Data are representative of 2-3 independent experiments. Samples were compared using paired two-tailed Student’s t test and data points are presented as individual mice with the mean ± SEM (N.S., Not Significant; **, p < 0.01; ***, p < 0.001; ****, p < 0.0001). Specific p-values are as follows: a < 0.0001; b:[UTX - Female vs. Male=0.0008; rest<0.0001]; d:[%NK = 0.0003; No. NK = 0.002]; f:[%IFN-γ+- NT = 0.112; 15 + 12 = 0.00133; IFN-γMFI – NT = 0.145; 15 + 12 < 0.0001]; h[%IFN-γ+- NT = 0.112; 12 + 18 = 0.00135; IFN-γMFI – NT = 0.155; 12 + 18 < 0.0001].
Extended Data Fig. 6 Integrative ATAC, RNA, anti-UTX CUT&Tag sequencing analysis reveal concomitant changes in chromatin accessibility and transcription mediated by UTX.
a) Schematic of mBMC generated by 1:4 WT (CD45.1+) and UTXNKD (CD45.2+) bone marrow into a lymphodepleted host (CD451x2). After 6wks of reconstitution, splenic NK cells were sorted for ATAC-seq and RNA-seq library preparation. b) Principal component analysis (PCA) of (left) ATAC-seq and (right) RNA-seq changes in WT and UTXNKD NK cells. c) Volcano plot of differentially expressed genes by RNA-seq plotted by Log2FC of UTXNKD vs. WT (x-axis) and Log2 Mean Expression (y-axis). Dotted lines represent Log2FC cut offs (0.5 and 1). Red dots are genes in NK cell effector and developmental pathways. d) Scatter plot highlighting differentially accessible and expressed genes (FDR and p-value<0.05) colored by fuzzy c-means cluster (see Fig. 6). Mean log2FC of ATAC accessibility peaks (y-axis) and log2FC (x-axis) of RNA-seq transcript levels. Best fit regression line (red) with standard error (light red ribbon) (SEM). Positive correlation calculated by two-tailed Spearman correlation of dataset (R = 0.62, p < 2.2×1016). e) Heatmap displaying expression of cell death genes between WT and UTXNKD NK cells FDR < 0.05, adjusted p-value<0.05, and log2FC > 0.5. f) PCA analysis of anti-UTX CUT&Tag in sort-purified WT and UTXNKD NK cells (n = 3). g) Pathway analysis on UTX-bound genes that are decreased (red) or increased (blue) by expression by RNA-seq using Enrichr (p-value by Fisher’s exact test). h) Log2FC in UTXNKDvs.WT of ATAC accessibility (y-axis) plotted by either decreased (>-0.5Log2FC) (blue) or increased (>+0.5Log2FC) (purple) expression by RNA-seq (x-axis) of the UTX bound genes with significant accessibility and expression differences. i) Correlation plot of Log2FC of UTXNKDvs.WT RNA-seq values compared to corresponding ATAC-seq values for each UTX-bound gene. Linear regression was performed (black line) with the standard error 95% confidence intervals plotted (dotted red lines). Two-tailed Pearson’s correlation was performed (r = 0.5165; p-value<0.0001). j) HOMER motif analysis of ATAC-seq peaks grouped by transcription factor family (top) and transcription factor (bottom). Point size indicates percentage of target sequences featuring motif and red gradient indicates -log10(p-value) of enrichment. k) HOMER Motif analysis performed on UTX-bound peaks. % Target Sequences refers to percent of target motifs identified by the HOMER algorithm out of the background motifs. j,k) Cumulative binomial distribution statistical analysis was performed.
Supplementary information
Supplementary Information
Supplementary Data Table 1
Supplementary Data Tables
Supplementary Data Table 2: Table of the differentially accessible peaks between WT and UTX-NKD NK cells. Descriptive statistics are provided. DAG analysis was performed using a two-sided Wilcoxon rank sum test. Supplementary Data Table 3: Table of the differentially expressed genes between WT and UTX-NKD NK cells. Descriptive statistics are provided. DEG analysis was performed using a two-sided Wilcoxon rank sum test. Supplementary Data Table 4: Table of anti-UTX CUT&Tag peaks in WT and UTX-NKD NK cells.
Source data
Source Data Fig. 1
Statistical source data for all graphs in Fig. 1.
Source Data Fig. 2
Statistical source data for all graphs in Fig. 2.
Source Data Fig. 3
Statistical source data for all graphs in Fig. 3.
Source Data Fig. 4
Statistical source data for all graphs in Fig. 4.
Source Data Fig. 5
Statistical source data for all graphs in Fig. 5.
Source Data Fig. 6
Statistical source data for all graphs in Fig. 6.
Source Data Extended Data Fig. 1
Statistical source data for all graphs in Extended Data Fig. 1.
Source Data Extended Data Fig. 2
Statistical source data for all graphs in Extended Data Fig. 2.
Source Data Extended Data Fig. 2
Unprocessed blots for the protein ladder, UTX, and actin from Extended Data Fig. 2d.
Source Data Extended Data Fig. 3
Statistical source data for all graphs in Extended Data Fig. 3.
Source Data Extended Data Fig. 4
Statistical source data for all graphs in Extended Data Fig. 4.
Source Data Extended Data Fig. 5
Statistical source data for all graphs in Extended Data Fig. 5.
Source Data Extended Data Fig. 6
Statistical source data for all graphs in Extended Data Fig. 6.
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.
About this article
Cite this article
Cheng, M.I., Li, J.H., Riggan, L. et al. The X-linked epigenetic regulator UTX controls NK cell-intrinsic sex differences. Nat Immunol 24, 780–791 (2023). https://doi.org/10.1038/s41590-023-01463-8
Received:
Accepted:
Published:
Issue date:
DOI: https://doi.org/10.1038/s41590-023-01463-8
This article is cited by
-
Survey of gene, lncRNA and transposon transcription patterns in four mouse organs highlights shared and organ-specific sex-biased regulation
Genome Biology (2025)
-
TSC22 domain family member 3 links natural killer cells to CD8+ T cell-mediated drug hypersensitivity
Signal Transduction and Targeted Therapy (2025)
-
Species-specific serine metabolism differentially controls natural killer cell functions
Nature Metabolism (2025)
-
The Four Core Genotypes mouse model: evaluating the impact of a recently discovered translocation
Biology of Sex Differences (2024)
-
imply: improving cell-type deconvolution accuracy using personalized reference profiles
Genome Medicine (2024)