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.

  • Resource
  • Published:

Deconvoluting global cytokine signaling networks in natural killer cells

Abstract

Cytokine signaling via signal transducer and activator of transcription (STAT) proteins is crucial for optimal antiviral responses of natural killer (NK) cells. However, the pleiotropic effects of both cytokine and STAT signaling preclude the ability to precisely attribute molecular changes to specific cytokine–STAT modules. Here, we employed a multi-omics approach to deconstruct and rebuild the complex interaction of multiple cytokine signaling pathways in NK cells. Proinflammatory cytokines and homeostatic cytokines formed a cooperative axis to commonly regulate global gene expression and to further repress expression induced by type I interferon signaling. These cytokines mediated distinct modes of epigenetic regulation via STAT proteins, and collective signaling best recapitulated global antiviral responses. The most dynamically responsive genes were conserved across humans and mice, which included a cytokine–STAT-induced cross-regulatory program. Thus, an intricate crosstalk exists between cytokine signaling pathways, which governs NK cell responses.

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

Access options

Rent or buy this article

Prices vary by article type

from$1.95

to$39.95

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

Fig. 1: Cytokine levels and phosphorylation of STATs in NK cells in spleen and blood peak early during MCMV infection.
Fig. 2: Transcriptional cooperation and antagonism among proinflammatory and homeostatic cytokines.
Fig. 3: IL-12/IL-18 and IL-2/IL-15 are the predominant mediators of changes in chromatin accessibility.
Fig. 4: IFN-α promotes changes in H3K4me3 enrichment at promoter regions.
Fig. 5: Integration of cytokine-induced transcriptional and STAT-binding profiles reveal cross-regulatory interactions.
Fig. 6: Combination of proinflammatory and homeostatic cytokines best correlates with early MCMV infection in vivo.
Fig. 7: Highly modulated genes induced by cytokines are conserved between mice and humans.

Similar content being viewed by others

Data availability

Data generated in this study have been deposited in the Gene Expression Omnibus (Super-Series accession numbers GSE140044 and GSE164116).

References

  1. Jost, S. & Altfeld, M. Control of human viral infections by natural killer cells. Annu. Rev. Immunol. 31, 163–194 (2013).

    Article  CAS  PubMed  Google Scholar 

  2. Long, E. O., Kim, H. S., Liu, D., Peterson, M. E. & Rajagopalan, S. Controlling natural killer cell responses: integration of signals for activation and inhibition. Annu. Rev. Immunol. 31, 227–258 (2013).

    Article  CAS  PubMed  Google Scholar 

  3. Lin, J. X. & Leonard, W. J. Fine-tuning cytokine signals. Annu. Rev. Immunol. 37, 295–324 (2019).

    Article  CAS  PubMed  Google Scholar 

  4. Meraz, M. A. et al. Targeted disruption of the Stat1 gene in mice reveals unexpected physiologic specificity in the JAK–STAT signaling pathway. Cell 84, 431–442 (1996).

    Article  CAS  PubMed  Google Scholar 

  5. Nguyen, K. B. et al. Critical role for STAT4 activation by type 1 interferons in the interferon-γ response to viral infection. Science 297, 2063–2066 (2002).

    Article  CAS  PubMed  Google Scholar 

  6. Pfeffer, L. M. et al. STAT3 as an adapter to couple phosphatidylinositol 3-kinase to the IFNAR1 chain of the type I interferon receptor. Science 276, 1418–1420 (1997).

    Article  CAS  PubMed  Google Scholar 

  7. Meinke, A., Barahmand-Pour, F., Wohrl, S., Stoiber, D. & Decker, T. Activation of different Stat5 isoforms contributes to cell-type-restricted signaling in response to interferons. Mol. Cell. Biol. 16, 6937–6944 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Gollob, J. A. et al. Altered interleukin-12 responsiveness in Th1 and Th2 cells is associated with the differential activation of STAT5 and STAT1. Blood 91, 1341–1354 (1998).

    Article  CAS  PubMed  Google Scholar 

  9. Bukowski, J. F., Woda, B. A., Habu, S., Okumura, K. & Welsh, R. M. Natural killer cell depletion enhances virus synthesis and virus-induced hepatitis in vivo. J. Immunol. 131, 1531–1538 (1983).

    CAS  PubMed  Google Scholar 

  10. Biron, C. A., Byron, K. S. & Sullivan, J. L. Severe herpesvirus infections in an adolescent without natural killer cells. N. Engl. J. Med. 320, 1731–1735 (1989).

    Article  CAS  PubMed  Google Scholar 

  11. Orange, J. S. & Biron, C. A. Characterization of early IL-12, IFN-αβ, and TNF effects on antiviral state and NK cell responses during murine cytomegalovirus infection. J. Immunol. 156, 4746–4756 (1996).

    CAS  PubMed  Google Scholar 

  12. Schneider, K. et al. Lymphotoxin-mediated crosstalk between B cells and splenic stroma promotes the initial type I interferon response to cytomegalovirus. Cell Host Microbe 3, 67–76 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Pien, G. C., Satoskar, A. R., Takeda, K., Akira, S. & Biron, C. A. Cutting edge: selective IL-18 requirements for induction of compartmental IFN-γ responses during viral infection. J. Immunol. 165, 4787–4791 (2000).

    Article  CAS  PubMed  Google Scholar 

  14. Orange, J. S., Wang, B., Terhorst, C. & Biron, C. A. Requirement for natural killer cell–produced interferon-γ in defense against murine cytomegalovirus infection and enhancement of this defense pathway by interleukin 12 administration. J. Exp. Med. 182, 1045–1056 (1995).

    Article  CAS  PubMed  Google Scholar 

  15. Chaix, J. et al. Cutting edge: priming of NK cells by IL-18. J. Immunol. 181, 1627–1631 (2008).

    Article  CAS  PubMed  Google Scholar 

  16. Madera, S. & Sun, J. C. Cutting edge: stage-specific requirement of IL-18 for antiviral NK cell expansion. J. Immunol. 194, 1408–1412 (2015).

    Article  CAS  PubMed  Google Scholar 

  17. Biron, C. A., Young, H. A. & Kasaian, M. T. Interleukin 2-induced proliferation of murine natural killer cells in vivo. J. Exp. Med. 171, 173–188 (1990).

    Article  CAS  PubMed  Google Scholar 

  18. Nguyen, K. B. et al. Coordinated and distinct roles for IFN-αβ, IL-12, and IL-15 regulation of NK cell responses to viral infection. J. Immunol. 169, 4279–4287 (2002).

    Article  CAS  PubMed  Google Scholar 

  19. Wiedemann, G. M. et al. Divergent role for STAT5 in the adaptive responses of natural killer cells. Cell Rep. 33, 108498 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Sun, J. C., Beilke, J. N. & Lanier, L. L. Adaptive immune features of natural killer cells. Nature 457, 557–561 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Daniels, K. A. et al. Murine cytomegalovirus is regulated by a discrete subset of natural killer cells reactive with monoclonal antibody to Ly49H. J. Exp. Med. 194, 29–44 (2001).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Arase, H., Mocarski, E. S., Campbell, A. E., Hill, A. B. & Lanier, L. L. Direct recognition of cytomegalovirus by activating and inhibitory NK cell receptors. Science 296, 1323–1326 (2002).

    Article  CAS  PubMed  Google Scholar 

  23. Smith, H. R. et al. Recognition of a virus-encoded ligand by a natural killer cell activation receptor. Proc. Natl Acad. Sci. USA 99, 8826–8831 (2002).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Cooper, M. A. et al. Cytokine-induced memory-like natural killer cells. Proc. Natl Acad. Sci. USA 106, 1915–1919 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Sun, J. C. et al. Proinflammatory cytokine signaling required for the generation of natural killer cell memory. J. Exp. Med. 209, 947–954 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Madera, S. et al. Type I IFN promotes NK cell expansion during viral infection by protecting NK cells against fratricide. J. Exp. Med. 213, 225–233 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Ali, A. K., Oh, J. S., Vivier, E., Busslinger, M. & Lee, S. H. NK cell-specific Gata3 ablation identifies the maturation program required for bone marrow exit and control of proliferation. J. Immunol. 196, 1753–1767 (2016).

    Article  CAS  PubMed  Google Scholar 

  28. Nguyen, K. B. et al. Interferon-αβ-mediated inhibition and promotion of interferon-γ: STAT1 resolves a paradox. Nat. Immunol. 1, 70–76 (2000).

    Article  CAS  PubMed  Google Scholar 

  29. Miyagi, T. et al. High basal STAT4 balanced by STAT1 induction to control type 1 interferon effects in natural killer cells. J. Exp. Med. 204, 2383–2396 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Lau, C. M. et al. Epigenetic control of innate and adaptive immune memory. Nat. Immunol. 19, 963–972 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Putz, E. M. et al. CDK8-mediated STAT1-S727 phosphorylation restrains NK cell cytotoxicity and tumor surveillance. Cell Rep. 4, 437–444 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Zhao, X. D. et al. Whole-genome mapping of histone H3 Lys4 and 27 trimethylations reveals distinct genomic compartments in human embryonic stem cells. Cell Stem Cell 1, 286–298 (2007).

    Article  CAS  PubMed  Google Scholar 

  33. Geary, C. D. et al. Non-redundant ISGF3 components promote NK cell survival in an auto-regulatory manner during viral infection. Cell Rep. 24, 1949–1957 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Rapp, M. et al. Core-binding factor β and Runx transcription factors promote adaptive natural killer cell responses. Sci. Immunol. 2, eaan3796 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  35. Villarino, A. V. et al. Subset- and tissue-defined STAT5 thresholds control homeostasis and function of innate lymphoid cells. J. Exp. Med. 214, 2999–3014 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Barbulescu, K. et al. IL-12 and IL-18 differentially regulate the transcriptional activity of the human IFN-γ promoter in primary CD4+ T lymphocytes. J. Immunol. 160, 3642–3647 (1998).

    CAS  PubMed  Google Scholar 

  37. Pipkin, M. E. et al. Interleukin-2 and inflammation induce distinct transcriptional programs that promote the differentiation of effector cytolytic T cells. Immunity 32, 79–90 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  38. Min-Oo, G. & Lanier, L. L. Cytomegalovirus generates long-lived antigen-specific NK cells with diminished bystander activation to heterologous infection. J. Exp. Med. 211, 2669–2680 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Nabekura, T. & Lanier, L. L. Tracking the fate of antigen-specific versus cytokine-activated natural killer cells after cytomegalovirus infection. J. Exp. Med. 213, 2745–2758 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Mehrotra, P. T., Wu, D., Crim, J. A., Mostowski, H. S. & Siegel, J. P. Effects of IL-12 on the generation of cytotoxic activity in human CD8+ T lymphocytes. J. Immunol. 151, 2444–2452 (1993).

    CAS  PubMed  Google Scholar 

  41. Hennessy, R. J. et al. Quantifying NK cell growth and survival changes in response to cytokines and regulatory checkpoint blockade helps identify optimal culture and expansion conditions. J. Leukoc. Biol. 105, 1341–1354 (2019).

    Article  CAS  PubMed  Google Scholar 

  42. Romee, R. et al. Cytokine-induced memory-like natural killer cells exhibit enhanced responses against myeloid leukemia. Sci. Transl. Med. 8, 357ra123 (2016).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  43. Lee, S. H. et al. Suppressor of cytokine signaling 2 regulates IL-15-primed human NK cell function via control of phosphorylated Pyk2. J. Immunol. 185, 917–928 (2010).

    Article  CAS  PubMed  Google Scholar 

  44. Cousens, L. P., Orange, J. S., Su, H. C. & Biron, C. A. Interferon-αβ inhibition of interleukin 12 and interferon-γ production in vitro and endogenously during viral infection. Proc. Natl Acad. Sci. USA 94, 634–639 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Ni, J., Miller, M., Stojanovic, A., Garbi, N. & Cerwenka, A. Sustained effector function of IL-12/15/18-preactivated NK cells against established tumors. J. Exp. Med. 209, 2351–2365 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Romee, R. et al. Cytokine activation induces human memory-like NK cells. Blood 120, 4751–4760 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Bezman, N. A. et al. Molecular definition of the identity and activation of natural killer cells. Nat. Immunol. 13, 1000–1009 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Schlums, H. et al. Cytomegalovirus infection drives adaptive epigenetic diversification of NK cells with altered signaling and effector function. Immunity 42, 443–456 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  49. Sadelain, M. Chimeric antigen receptors: driving immunology towards synthetic biology. Curr. Opin. Immunol. 41, 68–76 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  50. Madisen, L. et al. A robust and high-throughput Cre reporting and characterization system for the whole mouse brain. Nat. Neurosci. 13, 133–140 (2010).

    Article  CAS  PubMed  Google Scholar 

  51. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Bolger, A. M., Lohse, M. & Usadel, B. Trimmomatic: a flexible trimmer for Illumina sequence data. Bioinformatics 30, 2114–2120 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Speir, M. L. et al. The UCSC Genome Browser database: 2016 update. Nucleic Acids Res. 44, D717–D725 (2016).

    Article  CAS  PubMed  Google Scholar 

  54. Patro, R., Duggal, G., Love, M. I., Irizarry, R. A. & Kingsford, C. Salmon provides fast and bias-aware quantification of transcript expression. Nat. Methods 14, 417–419 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Soneson, C., Love, M. I. & Robinson, M. D. Differential analyses for RNA-seq: transcript-level estimates improve gene-level inferences. F1000Res 4, 1521 (2015).

    Article  PubMed  CAS  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Lawrence, M. et al. Software for computing and annotating genomic ranges. PLoS Comput. Biol. 9, e1003118 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  59. Zhu, L. J. et al. ChIPpeakAnno: a Bioconductor package to annotate ChIP-seq and ChIP-chip data. BMC Bioinf. 11, 237 (2010).

    Article  CAS  Google Scholar 

  60. 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).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  62. Wright, M. W., Eyre, T. A., Lush, M. J., Povey, S. & Bruford, E. A. HCOP: the HGNC comparison of orthology predictions search tool. Mamm. Genome 16, 827–828 (2005).

    Article  PubMed  Google Scholar 

  63. Seal, R. L., Gordon, S. M., Lush, M. J., Wright, M. W. & Bruford, E. A. genenames.org: the HGNC resources in 2011. Nucleic Acids Res. 39, D514–D519 (2011).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Ramirez, F. et al. deepTools2: a next generation web server for deep-sequencing data analysis. Nucleic Acids Res. 44, W160–W165 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  66. Gu, Z., Eils, R. & Schlesner, M. Complex heatmaps reveal patterns and correlations in multidimensional genomic data. Bioinformatics 32, 2847–2849 (2016).

    Article  CAS  PubMed  Google Scholar 

  67. Hahne, F. & Ivanek, R. Visualizing genomic data using Gviz and bioconductor. Methods Mol. Biol. 1418, 335–351 (2016).

    Article  PubMed  Google Scholar 

Download references

Acknowledgements

We thank members of the Sun laboratory for comments, discussions, technical support and experimental assistance. We thank V. Pascual (Weill Cornell Medicine) for sharing reagents. We thank L. Lanier (University of California, San Francisco) for providing mice and critical feedback of our manuscript. We acknowledge the use of the Integrated Genomics Operation Core, funded by the National Cancer Institute Cancer Center Support Grant (P30CA08748); Cycle for Survival; and the Marie-Josée and Henry R. Kravis Center for Molecular Oncology. G.M.W. and S.G. were supported through research fellowships by the Deutsche Forschungsgemeinschaft (DFG Forschungsstipendium WI-4927/1-2 and GR 5503/1-1). N.M.A. was supported by a Medical Scientist Training Program grant from the National Institute of General Medical Sciences (T32GM007739 to the Weill Cornell–Rockefeller–Sloan Kettering Tri-Institutional MD–PhD Program) and by an F30 Predoctoral Fellowship from the National Institute of Allergy and Infectious Diseases (F30 AI136239). J.B.L. was supported by the François Wallace Monahan Fellowship. K.C.H. was supported by the National Institutes of Health (AI125651, HL129472 and AI069197) and the Leukemia Lymphoma Society Scholar Award. J.C.S. was supported by the Ludwig Center for Cancer Immunotherapy, the American Cancer Society, the Burroughs Wellcome Fund and the National Institutes of Health (AI100874, AI130043 and P30CA008748). C.M.L. was supported by the Cancer Research Institute as a Cancer Research Institute–Carson Family Fellow.

Author information

Authors and Affiliations

Authors

Contributions

G.M.W., C.M.L. and J.C.S. designed the study. G.M.W., C.M.L., E.K.S., S.G., S.S., J.B.L., N.M.A. and C.D. performed the experiments. C.M.L. performed the bioinformatic analyses. J.B.L. and K.C.H. consulted on the experimental procedures for the human data. C.M.L. and J.C.S. wrote the manuscript.

Corresponding authors

Correspondence to Joseph C. Sun or Colleen M. Lau.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

Peer review information Nature Immunology thanks Lisa Forbes, Dagmar Gotthardt and Chiara Romagnani for their contribution to the peer review of this work. L. A. Dempsey was the primary editor on this article and managed its editorial process and peer review in collaboration with the rest of the editorial team.

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

Extended data

Extended Data Fig. 1 Cytokines peak early during MCMV infection.

Livers and spleen were harvested at days 0, 0.5, 1, 2, 3, 4, 5, 7 and 14 after MCMV infection. Organs were lysed for protein extracts, and then measured for cytokine levels. Plots show cytokine levels of IFN-α, IL-12p70, IL-18, IL-2, and IL-15 over time. For (a), n = 5, and for (b-c), n = 3-5. a, Plots show cytokine levels in the liver of IFN-α, IL-12p70, IL-18, IL-2, and IL-15 over time, with log10 scale on y-axis. Lines connect at medians of each time point. * p = 0.016-0.032 from two-sided Mann-Whitney-Wilcoxon test comparing to day 0. b, Graphs show mean ± SD of phosphorylated STAT MFI values (p-STAT1 [Ser], p-STAT4, [Tyr] and p-STAT5 [Tyr]) proteins from liver CD3ε TCRβ NK1.1+ NK cells. P-values are calculated by two-sided Welch’s t-test. c, Graphs show mean ± SD of p-STAT1 (Tyr) MFI values from spleen CD3ε TCRβ NK1.1+ NK cells. P-values are calculated by two-sided Welch’s t-test. d, Schematic of global profiling strategy. MFI = mean fluorescence intensity.

Extended Data Fig. 2 IL-2/IL-15 synergizes with IL-12/18.

RNA-seq was performed on sorted NK cells (CD3ε TCRβ CD19 F4/80 NK1.1+) cultured for 3 h with indicated cytokine conditions. n = 3. a, Flow plots show gating strategy for sorted murine NK cells before stimulation. b, Heatmap shows row-scaled log-transformed expression values from RNA-seq data of the all DE genes that show an FDR-adjusted p-value < 0.05, |log2 FC | > 1, and TPM > 5, as grouped in Fig. 2c. c, Heatmap shows row-scaled log-transformed expression values from top 100 synergistic genes, defined as DE (FDR-adjusted p-value < 0.05; TPM > 5) in IL-12/IL-18 plus IL-2/IL-15 versus unstimulated conditions, but not DE (FDR-adjusted p-value > 0.05; TPM > 5) in either IL-12/IL-18 or IL-2/IL-15 compared to unstimulated conditions. Genes were ranked by p-value from IL-12/IL-18 plus IL-2/IL-15 versus unstimulated comparison. d, Bar plot depicts number of cooperative IL-12/18:IL-2/IL-15 genes that show the same or different directional log2FC (versus unstimulated conditions) among indicated DE (FDR-adjusted p-value < 0.05) genes compared to combined stimulation by IL-12/IL-18. Genes that are designated as “different” display at least one condition where the FC does not modulate in the same direction as combined IL-12/18 conditions. DE = differentially expressed; FDR = false discovery rate; FC = fold change.

Extended Data Fig. 3 IL-12/18 antagonizes IFN-α signaling.

RNA-seq or flow cytometry was performed on sorted NK cells (CD3ε TCRβ CD19 F4/80 NK1.1+) cultured for 3 h with indicated cytokine conditions. n = 3. a, Bar plots show mean of normalized counts of Stat1 transcript across various cytokine conditions assayed by RNA-seq. b, Graphs depict percent of NK cells positive for intracellular IFN-γ staining (left), mean fluorescence intensity (MFI) of IFN-γ+ cells (middle), and MFI of p-STAT4 (Tyr; right). Bar indicates mean ± SD. P-values are calculated by unpaired two-sided Student’s t-test comparing WT to Stat1–/–. c, Bar plot depicts number of antagonistic IL-12/18:IFN-α genes, as shown in Extended Data Fig. 2d. d, Left panel shows heatmap of row-scaled log-transformed expression values of the 86 common differentially expressed genes between IFN-α, IL-12/IL-18 and IL-2/IL-15, hierarchically clustered into 4 groups. Box plots of z-scores from each cluster are depicted in the right panel. Centre, median; box limits, first and third percentiles; whiskers, 1.5 × interquartile range. Dotted lines show median z-score of IFN-α plus IL-12/IL-18 plus IL-2/15 condition. Tiles to the left of gene names indicate whether the genes are bound by STAT1 (green), STAT4 (blue), or STAT5 (orange) by ChIP-seq.

Extended Data Fig. 4 Epigenetic contribution of IL-12 versus IL-18.

ATAC-seq was performed on sorted NK cells (CD3ε TCRβ CD19 F4/80 NK1.1+) cultured for 3 h with IL-12, IL-18, or IL-12 + L-18. n = 3. a, Tables show results for de novo motif analysis by HOMER software on top 25% DA regions (see Methods). From left to right, columns show discovered motif sequence (top) with the best matched known motif (bottom), best matched known transcription factor family, p-value, similarity score, source of data used to derive known motif, and DA group. For all groups, top 3 ranked on p-value are shown. b, Bar plot depicts number of DA regions (FDR-adjusted p-value < 0.05) that overlap with STAT4-bound ChIP-seq regions, broken down by DA group. Colors indicate FC direction of DA regions.FDR = false discovery rate; DA = differentially accessible.

Extended Data Fig. 5 Presence of all cytokine conditions best correlates with early in vivo MCMV infection.

For in vitro studies, RNA-seq and ATAC-seq was performed as described in Figs. 2 and 3, respectively. H3K4me3 ChIP-seq was performed on sorted NK cells (CD3ε TCRβ CD19 F4/80 NK1.1+) cultured for 3 h with IFN-α, IL-12/IL-18, IL-2/IL-15, or a combination of all three. For in vivo studies, RNA-seq and ATAC-seq was performed on sorted Ly49H+ NK cells (TCRβCD19CD3εF4/80NK1.1+Ly49H+) at indicated timepoints after MCMV infection. H3K4me3 ChIP-seq was performed on sorted Ly49H+ NK cells (TCRβCD19CD3εF4/80NK1.1+Ly49H+) at indicated time points after MCMV infection from either WT mice or NKp46-CreERT2 Rosa26-tdTomato reporter mice (gated additionally on TdTom+). Heatmaps show matrices of Spearman correlation coefficients using log2 FC comparing cytokine-stimulated versus unstimulated conditions or d2 versus d0 post-infection using (a) RNA-seq data, (b) ATAC-seq, or (c) H3K4me3 ChIP-seq data. Circle sizes are proportional to coefficients. X-ed out values indicate p > 0.05. For in vitro studies, n = 2-8, and for in vivo studies, n = 2-4. (d) Scatter plots of ChIP-seq log2 FC comparing cytokine-stimulated versus unstimulated (x-axis) conditions to d2 versus d0 post-infection (y-axis). (e) Center dots indicate Spearman correlations calculated from ChIP-seq values depicted in (c). Ranges show 95% confidence interval calculated by Fisher’s z-transformation.

Extended Data Fig. 6 Combination of various cytokine conditions supports optimal NK cell effector responses in vitro.

a, Bar plots show intracellular IFN-γ after 3 h of indicated cytokine stimulation on sorted NK cells (CD3ε TCRβ CD19 F4/80 NK1.1+). Calculated p-values for all pairwise conditions are represented as a heatmap. n = 6. b, Bar plots show calculated percent lysis of Calcein AM-labeled YAC-1 cells incubated with sorted NK cells (CD3ε TCRβ CD19 F4/80 NK1.1+) in the indicated cytokine conditions. Calculated p-values for all pairwise conditions are represented as a heatmap. n = 3. c, Representative histograms of Cell Trace Violet staining on sorted NK cells from 3-day in vitro cultures under indicated conditions. Representative of 3 independent experiments. Dotted lines connect biological replicates, which are colored. * p < 0.05, ** p < 0.01, *** p < 0.001 by two-tailed paired Student’s t-test.

Extended Data Fig. 7 Transcriptional profiles of cytokine signaling in human NK cells.

RNA-seq was performed on sorted NK cells (CD3ε CD14 CD56+) derived from PBMCs of healthy donors and cultured for 3 h with IFN-α, IL-12/IL-18, or IL-2/IL-15. n = 6. a, Sorting strategy of human NK cells and NK cell percentages for each donor. b, Representative flow cytometric plot (left) and summarized proportions (right) of different NK cell subsets defined by cell surface phenotype. Bar plots show mean, and numbers indicate donor identifier. c, Principal component analysis of RNA-seq donor-corrected, log-transformed counts of all detectable genes in humans. d, Venn diagram of all commonly expressed, mappable DE (FDR-adjusted p-value < 0.05, |log2 FC|>1) human orthologs when comparing cytokine-stimulated versus unstimulated conditions. e, Heatmap matrix of Spearman correlation coefficients using log2 FC of each condition compared to unstimulated from all commonly expressed, mappable human DE genes. Circle sizes are proportional to coefficients. f, Volcano plots highlight top 15 DE genes (FDR-adjusted p-value < 0.05) ranked on p-value. x-axis depicts log2 FC and y-axis depicts –log10 (p-value) (Wald test). Shown are all expressed genes (TPM>5) comparing indicated cytokine stimulation versus unstimulated. Horizontal dashed line delineates p = 0.05, vertical dashed lines mark log2 FC of -1 and 1. Colored dots show DE genes. Triangles indicate capped p-values. DE = differentially expressed; FDR = false discovery rate; FC = fold change; TPM = transcripts-per-million.

Extended Data Fig. 8 Highly modulated genes induced by cytokines are conserved between mouse and human.

For human data, RNA-seq was performed on sorted NK cells (CD3ε CD14 CD56+) derived from PBMCs of healthy donors and cultured for 3 h with IFN-α, IL-12/IL-18, or IL-2/IL-15. Mouse data was performed as described in Fig. 2. For mouse, n = 3, and for human, n = 6. a, Venn diagram of all genes in the UCSC Known Gene reference annotation (white), all mappable orthologs annotated by NCBI and retrieved through HGNC (blue), and counted genes that showed an average TPM >5 across all conditions. Numbers show absolute numbers. b, Venn diagram of mappable and expressed orthologs from mouse and human. Numbers show absolute numbers. Intersection depicts commonly expressed mappable orthologs used for downstream DE analysis. c, Principal component analysis of RNA-seq uncorrected log-transformed counts on (left) and those corrected for replicate/donor variation (right) on commonly expressed mappable orthologs. d, Scatter plots of RNA-seq log2 FC comparing cytokine-stimulated versus unstimulated from human datasets (x-axis) and mouse datasets (y-axis). Shown are genes commonly DE in all three cytokine conditions across human and mouse. Plots highlight genes that show |log2 FC|> 1 in all three conditions (top) or two of the three conditions (bottom). DE = differentially expressed; FC = fold change; TPM = transcripts-per-million.

Supplementary information

Reporting Summary

Supplementary Tables

Consolidated Excel spreadsheetof Supplementary Tables 1–7.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Wiedemann, G.M., Santosa, E.K., Grassmann, S. et al. Deconvoluting global cytokine signaling networks in natural killer cells. Nat Immunol 22, 627–638 (2021). https://doi.org/10.1038/s41590-021-00909-1

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-021-00909-1

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