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:

Control of nutrient uptake by IRF4 orchestrates innate immune memory

Abstract

Natural killer (NK) cells are innate cytotoxic lymphocytes with adaptive immune features, including antigen specificity, clonal expansion and memory. As such, NK cells share many transcriptional and epigenetic programs with their adaptive CD8+ T cell siblings. Various signals ranging from antigen, co-stimulation and proinflammatory cytokines are required for optimal NK cell responses in mice and humans during virus infection; however, the integration of these signals remains unclear. In this study, we identified that the transcription factor IRF4 integrates signals to coordinate the NK cell response during mouse cytomegalovirus infection. Loss of IRF4 was detrimental to the expansion and differentiation of virus-specific NK cells. This defect was partially attributed to the inability of IRF4-deficient NK cells to uptake nutrients required for survival and memory generation. Altogether, these data suggest that IRF4 is a signal integrator that acts as a secondary metabolic checkpoint to orchestrate the adaptive response of NK cells during viral infection.

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

Access options

Buy this article

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

Fig. 1: Dynamic regulation of IRF4 in NK cells in response to viral infection.
Fig. 2: IRF4 is required for NK cell-mediated protection against MCMV.
Fig. 3: IRF4 is required for adaptive NK cell differentiation during viral infection.
Fig. 4: IRF4 controls the transcriptional program of effector NK cells.
Fig. 5: IRF4 regulates nutrient uptake in antiviral NK cells.
Fig. 6: Disruption of iron homeostasis impairs the adaptive NK cell response to viral infection.
Fig. 7: IRF4 directly regulates adaptive features of NK cells.

Similar content being viewed by others

Data availability

All published data generated and supporting the findings of this study have been deposited to NCBI’s GEO or European Genome–Phenome Archive as described. scRNA-seq and ChIP-seq data generated in this study have been deposited at GEO under accession code GSE236556. Source data are provided with this paper.

References

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

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Mujal, A. M., Delconte, R. B. & Sun, J. C. Natural killer cells: from innate to adaptive features. Annu. Rev. Immunol. 39, 417–447 (2021).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  4. Adams, E. J. et al. Structural elucidation of the m157 mouse cytomegalovirus ligand for Ly49 natural killer cell receptors. Proc. Natl Acad. Sci. USA 104, 10128–10133 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  5. 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).

    CAS  PubMed  Google Scholar 

  6. Nabekura, T. et al. Costimulatory molecule DNAM-1 is essential for optimal differentiation of memory natural killer cells during mouse cytomegalovirus infection. Immunity 40, 225–234 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Wiedemann, G. M. et al. Deconvoluting global cytokine signaling networks in natural killer cells. Nat. Immunol. 22, 627–638 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Hammer, Q., Ruckert, T. & Romagnani, C. Natural killer cell specificity for viral infections. Nat. Immunol. 19, 800–808 (2018).

    CAS  PubMed  Google Scholar 

  13. Zhao, G. N., Jiang, D. S. & Li, H. Interferon regulatory factors: at the crossroads of immunity, metabolism, and disease. Biochim. Biophys. Acta 1852, 365–378 (2015).

    CAS  PubMed  Google Scholar 

  14. Huber, M. & Lohoff, M. IRF4 at the crossroads of effector T-cell fate decision. Eur. J. Immunol. 44, 1886–1895 (2014).

    CAS  PubMed  Google Scholar 

  15. Mittrucker, H. W. et al. Requirement for the transcription factor LSIRF/IRF4 for mature B and T lymphocyte function. Science 275, 540–543 (1997).

    CAS  PubMed  Google Scholar 

  16. Duncan, G. S., Mittrucker, H. W., Kagi, D., Matsuyama, T. & Mak, T. W. The transcription factor interferon regulatory factor-1 is essential for natural killer cell function in vivo. J. Exp. Med. 184, 2043–2048 (1996).

    CAS  PubMed  Google Scholar 

  17. Lohoff, M. et al. Deficiency in the transcription factor interferon regulatory factor (IRF)-2 leads to severely compromised development of natural killer and T helper type 1 cells. J. Exp. Med. 192, 325–336 (2000).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Ogasawara, K. et al. Requirement for IRF-1 in the microenvironment supporting development of natural killer cells. Nature 391, 700–703 (1998).

    CAS  PubMed  Google Scholar 

  19. Mace, E. M. et al. Biallelic mutations in IRF8 impair human NK cell maturation and function. J. Clin. Invest. 127, 306–320 (2017).

    PubMed  Google Scholar 

  20. Adams, N. M. et al. Transcription factor IRF8 orchestrates the adaptive natural killer cell response. Immunity 48, 1172–1182 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  21. Beaulieu, A. M., Zawislak, C. L., Nakayama, T. & Sun, J. C. The transcription factor Zbtb32 controls the proliferative burst of virus-specific natural killer cells responding to infection. Nat. Immunol. 15, 546–553 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  22. Bravo Garcia-Morato, M. et al. New human combined immunodeficiency caused by interferon regulatory factor 4 (IRF4) deficiency inherited by uniparental isodisomy. J. Allergy Clin. Immunol. 141, 1924–1927 (2018).

    PubMed  Google Scholar 

  23. Man, K. et al. The transcription factor IRF4 is essential for TCR affinity-mediated metabolic programming and clonal expansion of T cells. Nat. Immunol. 14, 1155–1165 (2013).

    CAS  PubMed  Google Scholar 

  24. Geary, C. D. et al. Non-redundant ISGF3 components promote NK cell survival in an auto-regulatory manner during viral infection. Cell Rep. 24, 1949–19576 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Rückert, T., Lareau, C. A., Mashreghi, M. F., Ludwig, L. S. & Romagnani, C. Clonal expansion and epigenetic inheritance of long-lasting NK cell memory. Nat. Immunol. 23, 1551–1563 (2022).

    PubMed  PubMed Central  Google Scholar 

  27. Holmes, T. D. et al. The transcription factor Bcl11b promotes both canonical and adaptive NK cell differentiation. Sci. Immunol. 6, eabc9801 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  28. Kallies, A. et al. A role for Blimp1 in the transcriptional network controlling natural killer cell maturation. Blood 117, 1869–1879 (2011).

    CAS  PubMed  Google Scholar 

  29. Yao, S. et al. Interferon regulatory factor 4 sustains CD8(+) T cell expansion and effector differentiation. Immunity 39, 833–845 (2013).

    CAS  PubMed  Google Scholar 

  30. Flommersfeld, S. et al. Fate mapping of single NK cells identifies a type 1 innate lymphoid-like lineage that bridges innate and adaptive recognition of viral infection. Immunity 54, 2288–2304 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. Finak, G. et al. MAST: a flexible statistical framework for assessing transcriptional changes and characterizing heterogeneity in single-cell RNA sequencing data. Genome Biol. 16, 278 (2015).

    PubMed  PubMed Central  Google Scholar 

  32. Intlekofer, A. M. et al. Effector and memory CD8+ T cell fate coupled by T-bet and eomesodermin. Nat. Immunol. 6, 1236–1244 (2005).

    CAS  PubMed  Google Scholar 

  33. Omilusik, K. D. et al. Transcriptional repressor ZEB2 promotes terminal differentiation of CD8+ effector and memory T cell populations during infection. J. Exp. Med. 212, 2027–2039 (2015).

    PubMed  PubMed Central  Google Scholar 

  34. van Helden, M. J. et al. Terminal NK cell maturation is controlled by concerted actions of T-bet and Zeb2 and is essential for melanoma rejection. J. Exp. Med. 212, 2015–2025 (2015).

    PubMed  PubMed Central  Google Scholar 

  35. Zook, E. C. et al. Transcription factor ID2 prevents E proteins from enforcing a naive T lymphocyte gene program during NK cell development. Sci. Immunol. 3, eaao2139 (2018).

  36. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Milner, J. J. et al. Runx3 programs CD8(+) T cell residency in non-lymphoid tissues and tumours. Nature 552, 253–257 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Wang, D. et al. The transcription factor Runx3 establishes chromatin accessibility of cis-regulatory landscapes that drive memory cytotoxic T lymphocyte formation. Immunity 48, 659–674 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Delconte, R. B. et al. The helix-loop-helix protein ID2 governs NK cell fate by tuning their sensitivity to interleukin-15. Immunity 44, 103–115 (2016).

    CAS  PubMed  Google Scholar 

  40. Loftus, R. M. et al. Amino acid-dependent cMyc expression is essential for NK cell metabolic and functional responses in mice. Nat. Commun. 9, 2341 (2018).

    PubMed  PubMed Central  Google Scholar 

  41. Frost, J. N. et al. Evaluation of perturbed iron-homeostasis in a prospective cohort of patients with COVID-19. Wellcome Open Res. 7, 173 (2022).

    PubMed  PubMed Central  Google Scholar 

  42. Frost, J. N. et al. Plasma iron controls neutrophil production and function. Sci. Adv. 8, eabq5384 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  43. Bonadonna, M. et al. Iron regulatory protein (IRP)-mediated iron homeostasis is critical for neutrophil development and differentiation in the bone marrow. Sci. Adv. 8, eabq4469 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Li, P. et al. BATF-JUN is critical for IRF4-mediated transcription in T cells. Nature 490, 543–546 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Kaech, S. M. & Cui, W. Transcriptional control of effector and memory CD8+ T cell differentiation. Nat. Rev. Immunol. 12, 749–761 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  46. Laidlaw, B. J. & Cyster, J. G. Transcriptional regulation of memory B cell differentiation. Nat. Rev. Immunol. 21, 209–220 (2021).

    CAS  PubMed  Google Scholar 

  47. Adams, N. M. et al. Cytomegalovirus infection drives avidity selection of natural killer cells. Immunity 50, 1381–1390 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Grassmann, S. et al. Distinct surface expression of activating receptor Ly49H drives differential expansion of NK cell clones upon murine cytomegalovirus infection. Immunity 50, 1391–1400 (2019).

    CAS  PubMed  Google Scholar 

  49. Gordon, S. M. et al. The transcription factors T-bet and Eomes control key checkpoints of natural killer cell maturation. Immunity 36, 55–67 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Yang, C. Y. et al. The transcriptional regulators Id2 and Id3 control the formation of distinct memory CD8+ T cell subsets. Nat. Immunol. 12, 1221–1229 (2011).

    CAS  PubMed  Google Scholar 

  51. Pearce, E. L. et al. Control of effector CD8+ T cell function by the transcription factor eomesodermin. Science 302, 1041–1043 (2003).

    CAS  PubMed  Google Scholar 

  52. Sheppard, S. et al. Lactate dehydrogenase A-dependent aerobic glycolysis promotes natural killer cell anti-viral and anti-tumor function. Cell Rep. 35, 109210 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  53. Mah-Som, A. Y. et al. Reliance on Cox10 and oxidative metabolism for antigen-specific NK cell expansion. Cell Rep. 35, 109209 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Littwitz-Salomon, E. et al. Metabolic requirements of NK cells during the acute response against retroviral infection. Nat. Commun. 12, 5376 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  55. Ganz, T. & Nemeth, E. Iron homeostasis in host defence and inflammation. Nat. Rev. Immunol. 15, 500–510 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Lane, D. J. et al. Cellular iron uptake, trafficking and metabolism: key molecules and mechanisms and their roles in disease. Biochim. Biophys. Acta 1853, 1130–1144 (2015).

    CAS  PubMed  Google Scholar 

  57. Tussiwand, R. et al. Compensatory dendritic cell development mediated by BATF–IRF interactions. Nature 490, 502–507 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Ciofani, M. et al. A validated regulatory network for Th17 cell specification. Cell 151, 289–303 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Glasmacher, E. et al. A genomic regulatory element that directs assembly and function of immune-specific AP-1-IRF complexes. Science 338, 975–980 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  60. Kim, S. et al. High amount of transcription factor IRF8 engages AP1-IRF composite elements in enhancers to direct type 1 conventional dendritic cell identity. Immunity 53, 759–774 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  61. Consortium, I. R. F. I. et al. A multimorphic mutation in IRF4 causes human autosomal dominant combined immunodeficiency. Sci. Immunol. 8, eade7953 (2023).

    Google Scholar 

  62. Thouenon, R. et al. A neomorphic mutation in the interferon activation domain of IRF4 causes a dominant primary immunodeficiency. J. Exp. Med. 220, e20221292 (2023).

  63. Guerin, A. et al. IRF4 haploinsufficiency in a family with Whipple’s disease. eLife 7, e32340 (2018).

  64. Huntington, N. D., Cursons, J. & Rautela, J. The cancer-natural killer cell immunity cycle. Nat. Rev. Cancer 20, 437–454 (2020).

    CAS  PubMed  Google Scholar 

  65. Bald, T., Krummel, M. F., Smyth, M. J. & Barry, K. C. The NK cell-cancer cycle: advances and new challenges in NK cell-based immunotherapies. Nat. Immunol. 21, 835–847 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  66. Berrien-Elliott, M. M. et al. Hematopoietic cell transplantation donor-derived memory-like NK cells functionally persist after transfer into patients with leukemia. Sci. Transl. Med. 14, eabm1375 (2022).

    CAS  PubMed  PubMed Central  Google Scholar 

  67. Marin, N. D. et al. Memory-like differentiation enhances NK cell responses to melanoma. Clin. Cancer Res. 27, 4859–4869 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  68. Gauthier, L. et al. Multifunctional natural killer cell engagers targeting NKp46 trigger protective tumor immunity. Cell 177, 1701–1713 (2019).

    CAS  PubMed  Google Scholar 

  69. Andre, P. et al. Anti-NKG2A mAb is a checkpoint inhibitor that promotes anti-tumor immunity by unleashing both T and NK cells. Cell 175, 1731–1743 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  70. Zhang, Q. et al. Blockade of the checkpoint receptor TIGIT prevents NK cell exhaustion and elicits potent anti-tumor immunity. Nat. Immunol. 19, 723–732 (2018).

    CAS  PubMed  Google Scholar 

  71. Liu, E. et al. Use of CAR-transduced natural killer cells in CD19-positive lymphoid tumors. N. Engl. J. Med. 382, 545–553 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  72. Knorr, D. A., Bachanova, V., Verneris, M. R. & Miller, J. S. Clinical utility of natural killer cells in cancer therapy and transplantation. Semin. Immunol. 26, 161–172 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  73. Seo, H. et al. BATF and IRF4 cooperate to counter exhaustion in tumor-infiltrating CAR T cells. Nat. Immunol. 22, 983–995 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  74. Mercier, F. E., Sykes, D. B. & Scadden, D. T. Single targeted exon mutation creates a true congenic mouse for competitive hematopoietic stem cell transplantation: the C57BL/6-CD45.1(STEM) mouse. Stem Cell Rep. 6, 985–992 (2016).

    CAS  Google Scholar 

  75. Fodil-Cornu, N. et al. Ly49h-deficient C57BL/6 mice: a new mouse cytomegalovirus-susceptible model remains resistant to unrelated pathogens controlled by the NK gene complex. J. Immunol. 181, 6394–6405 (2008).

    CAS  PubMed  Google Scholar 

  76. Stoeckius, M. et al. Cell hashing with barcoded antibodies enables multiplexing and doublet detection for single cell genomics. Genome Biol. 19, 224 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  77. Azizi, E. et al. Single-cell map of diverse immune phenotypes in the breast tumor microenvironment. Cell 174, 1293–1308 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  78. Shekhar, K. et al. Comprehensive classification of retinal bipolar neurons by single-cell transcriptomics. Cell 166, 1308–1323 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  79. Wolf, F. A. et al. PAGA: graph abstraction reconciles clustering with trajectory inference through a topology preserving map of single cells. Genome Biol. 20, 59 (2019).

    PubMed  PubMed Central  Google Scholar 

  80. Wolf, F. A., Angerer, P. & Theis, F. J. SCANPY: large-scale single-cell gene expression data analysis. Genome Biol. 19, 15 (2018).

    PubMed  PubMed Central  Google Scholar 

  81. Subramanian, A. et al. Gene set enrichment analysis: a knowledge-based approach for interpreting genome-wide expression profiles. Proc. Natl Acad. Sci. USA 102, 15545–15550 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  82. van Dijk, D. et al. Recovering gene interactions from single-cell data using data diffusion. Cell 174, 716–729 (2018).

    PubMed  PubMed Central  Google Scholar 

  83. Joshi, N. S. et al. Inflammation directs memory precursor and short-lived effector CD8(+) T cell fates via the graded expression of T-bet transcription factor. Immunity 27, 281–295 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  84. 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).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  88. Hammer, Q. et al. Peptide-specific recognition of human cytomegalovirus strains controls adaptive natural killer cells. Nat. Immunol. 19, 453–463 (2018).

    CAS  PubMed  Google Scholar 

  89. 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).

    PubMed  PubMed Central  Google Scholar 

  90. Lareau, C. A. et al. Massively parallel single-cell mitochondrial DNA genotyping and chromatin profiling. Nat. Biotechnol. 39, 451–461 (2021).

    CAS  PubMed  Google Scholar 

  91. Stuart, T., Srivastava, A., Madad, S., Lareau, C. A. & Satija, R. Single-cell chromatin state analysis with Signac. Nat. Methods 18, 1333–1341 (2021).

    CAS  PubMed  PubMed Central  Google Scholar 

  92. Korsunsky, I. et al. Fast, sensitive and accurate integration of single-cell data with Harmony. Nat. Methods 16, 1289–1296 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  93. Stuart, T. et al. Comprehensive integration of single-cell data. Cell 177, 1888–1902 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank members of the Sun laboratory, K. Lupo and X. Chen, as well as the Single-Cell Analytics Innovation Lab at the Sloan Kettering Institute for technical support and experimental assistance, especially R. Chaligné. We also thank L. Lanier for helpful discussions. K. Murphy and T. Murphy provided mice critical to this study. T. Holmes and Y. Bryceson shared sequencing data from their study. We acknowledge the use of the Integrated Genomics Operation Core, funded by the National Cancer Institute Cancer Center Support Grant (P30CA008748), Cycle for Survival and the Marie-Josée and Henry R. Kravis Center for Molecular Oncology. J.N.F. is a Cancer Research Institute Irvington Fellow supported by the Cancer Research Institute (award CRI4186). K.C.H. and J.-B.L.L were supported by funding from the National Institutes of Health (R01 AI150999, R01HL155741, U01AI069197). 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, AI155558 and P30CA008748).

Author information

Authors and Affiliations

Authors

Contributions

E.K.S. and J.C.S. designed the study. E.K.S., H.K., J.-B.L.L., A.J.A., C.K.W., L.P. and J.N.F. performed experiments. E.K.S. and T.R. performed bioinformatics analyses. K.C.H., C.R. and J.C.S. provided critical resources and supervision for the study. E.K.S. and J.C.S. wrote the manuscript.

Corresponding author

Correspondence to Joseph C. Sun.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks Dagmar Gotthardt and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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 Evidence of IRF4-mediated transcriptional control in NK cells.

a. Heatmap of IRF family members expression in NK cells throughout the course of MCMV infection represented as z-score of log2 normalized counts based on RNA-seq. b. Representative histogram of IRF4 expression in WT and Irf4/ NK cells on day 2 PI (n = 4 biological replicates). c. Tracks (top) show chromatin accessibility dynamics of the Irf4 locus in Ly49H+ NK cells at days 0, 2, 4, and 7 PI as assessed by ATAC-seq. Graphs (bottom) show normalized counts for each peak indicated. d. Tables of enriched known motifs of highly accessible regions (log2FC > 1 & padj < 0.05) on day 4 PI versus day 2 PI from ATAC-seq data of Ly49H+ NK cells after MCMV infection. e. Heatmap of motif score from de novo motif analysis on highly accessible regions (log2FC > 1 & padj < 0.05) during day 0, 2, 4, and 7 transition based on ATAC-seq data of Ly49H+ NK cells after MCMV infection. f. IRF4 expression of sorted naïve splenic NK cells after an overnight stimulation with the indicated stimuli. IRF4 induction is displayed as fold change of IRF4 MFI over unstimulated condition (n = 6 biological replicates per condition). Two-way ANOVA test adjusted for multiple comparisons was used for statistical analysis. g. Representative histogram of IRF4 expression upon indicated stimulation gated on human CD56bright NK cells. Data are presented as paired fold change of IRF4 MFI compared to unstimulated condition (n = 8 donors per condition). Two-way ANOVA test adjusted for multiple comparisons was used for statistical analysis. h. UMAP embedding of scATAC-seq data from in vitro stimulated human NK cells (sorted on CD3CD14CD19CD7+NKG2C+) from HCMV donors. i. Coverage plot of the IRF4 locus from in vitro stimulated human NK cells as in (H).

Source data

Extended Data Fig. 2 IRF4 is dispensable for NK cell development and homeostasis.

a–c. Number of NK cells in blood (A), liver (B), and spleen (C) of WT (n = 8), Irf4+/− (n = 4), or Irf4−/− (n = 6) mice at 8 weeks old. d–f. Maturation status based on the expression of CD27 and CD11b of WT (n = 8), Irf4+/− (n = 4), or Irf4−/− (n = 5 for blood, and n = 6 for liver and spleen) mice in the blood (D), liver (E), and spleen (F) at 8 weeks old. g. Percentage of Ly49H+ NK cells within NK cells (Lineage- NK1.1+ CD49b+) of WT (n = 8), Irf4+/− (n = 4), or Irf4−/− (n = 6) mice at 8 weeks old. h. Experimental schematic of WT:Irf4/ mixed bone-marrow chimera (mBMC) generation. i–k. Percentage of NK cells of WT or Irf4−/− mice from WT:Irf4/ mBMC in the blood (n = 60 biological replicates) (I) and spleen (n = 21 biological replicates) (J) after 8 weeks (I-J), 16 weeks (n = 3 biological replicates) or 6 months (n = 5 biological replicates) post-transplant (K). l. Representative of flow plots of CD62L versus CD27 of either WT or Irf4/ NK cells from WT:Irf4/ mBMC in the blood and spleen 8 weeks post-transplant. Data are represented as percentage of each subset within total NK cells of each genotype (n = 22 biological replicates in blood, and n = 13 biological replicates in spleen). m. Representative of flow plots of CD11b versus CD27 of either WT or Irf4/ NK cells from WT:Irf4/ mBMC in the blood and spleen 8 weeks post-transplant. Data are represented as percentage of each subset within total NK cells of each genotype (n = 22 biological replicates in blood, and n = 16 biological replicates in spleen). n. Histogram and percentage quantification of Ly49H+ NK cells within splenic NK cells of either WT or Irf4/ NK cells from WT:Irf4/ mBMC (n = 24 biological replicates). Data are represented as mean ± SEM and are representative of or pooled from at least two independent experiments. Unpaired (A-G) and paired (I-N) two-tailed t-tests were performed.

Source data

Extended Data Fig. 3 IRF4 is not required for lymphopenia-driven proliferation and differentiation.

a. Experimental schematic of adoptive transfer of WT and Irf4/ NK cells into Rag2/ Il2rg/ mice. b. Fold change chimerism between WT and Irf4/ NK cells on day 7 over day 0 (pre-transfer) in blood, liver, and spleen (n = 3 biological replicates in blood, and n = 6 biological replicates in liver and spleen). c. Representative histogram of IRF4 expression between transferred WT and Irf4−/− NK cells on day 7 post-transfer into Rag2/ Il2rg/. d. Representative flow plots of CD27 and CD11b expression between WT and Irf4/ NK cells on day 7 post-transfer. Data are represented as percentage of each subset within WT or Irf4/ NK cells in indicated tissues (n = 3 biological replicates). e. Histogram of CD122 and quantification of CD122 MFI between WT and Irf4−/− NK cells on day 7 post-transfer in the spleen (n = 9 biological replicates). f. Histogram of CD132 and quantification of CD132 MFI between WT and Irf4−/− NK cells on day 7 post-transfer in the spleen (n = 9 biological replicates). g. Histogram of pSTAT5 and quantification of pSTAT5 MFI between WT and Irf4−/− NK cells on day 7 post-transfer in the spleen (n = 9 biological replicates). H-J. h–j. Quantification of Ki-67 staining in the blood and spleen between WT and Irf4/ NK cells (H, n = 5 biological replicates per group), BIM and BCL2 MFI from splenic NK cells (I-J, n = 9 biological replicates). Data are represented as mean ± SEM and are representative of or pooled from at least two independent experiments. Paired two-tailed t-test was performed unless stated otherwise.

Source data

Extended Data Fig. 4 IRF4-deficient NK cells have intact effector functions.

a, b Representative histograms and quantification of IFN-γ production (A) and LAMP-1 (also known as CD107a) (B) from either naive WT (gray) or Irf4/ (orange) NK cells upon in vitro stimulation with nothing, IL-12 + IL-18, PMA + Ionomycin, or PMA + Ionomycin + IL-12 for 3 hours. c–f. Representative histograms (upper panels) and quantifications (bottom panels) of CD25, Gzmb, CD69, and IFN-γ expression gated on Ly49H+ NK cells from MCMV-infected WT:Irf4/ mBMC on day 2 PI. Data are represented as mean ± SEM and are representative of or pooled from at least two independent experiments. n = 3 biological replicates per group. Paired two-tailed t-test was performed unless stated otherwise.

Source data

Extended Data Fig. 5 scRNA-seq Analysis of wild-type versus IRF4-deficient NK cells.

a. UMAP embedding of WT (left) or Irf4/ (right) of Ly49H+ NK cells colored by genotypes and days post-infection. b. Proportion of each genotype from each time points per Louvain clusters. c. Volcano plot of scRNA-seq as determined by MAST between WT and Irf4/ Ly49H+ NK cells on day 0, 2, 4 and 7 PI. Blue and yellow points represent significant genes (FDR < 0.05) that are upregulated in WT or Irf4/ Ly49H+ NK cells, respectively. d. Number of differentially expressed genes between WT or Irf4/ Ly49H+ NK cells from each time point as identified by MAST. e. Violin plot quantification of normalized reads for MYC transcript by scRNA-seq between WT and Irf4/ Ly49H+ NK cells. f. Representative histogram of Myc protein by flow cytometry between WT (gray) and Irf4/ (orange) Ly49H+ NK cells on day 5 PI. g. Violin plot quantification of MAGIC-imputed reads for CD71 (Tfrc), IRP1 (Aco1), and IRP2 (Ireb2) on day 7 PI between WT (blue) and Irf4−/− (yellow) Ly49H+ NK cells by scRNA-seq.

Source data

Extended Data Fig. 6 Nutrient requirements for adaptive NK cell response.

a. Experimental schematic of PBS or BCH treatment upon MCMV infection in WT mice. b. Quantification of Ly49H+ NK cell numbers of MCMV-infected mice treated with PBS or BCH on day 5 PI from blood (n = 3 biological replicates per group). c. Percent of CD62L versus CD27 subsets on day 5 PI of PBS- (black) or BCH-treated (red) mice (n = 3 biological replicates per group). d. Representative histogram and gMFI of CD71 expression on Ly49H+ NK cells on day 5 PI in PBS or Deferiprone-treated mice. Upregulation of CD71 was used as a readout for successful iron chelation (n = 5 biological replicates per group). a–g. Percent of KLRG1+ Ly49H+ NK cells (E), and gMFIs of IL18Ra (F) and CD44 (G) on Ly49H+ NK cells on day 5 PI from PBS or Deferiprone-treated mice (n = 5 biological replicates per group). Data are represented as mean ± SEM and are representative of at least two independent experiments. Two-way ANOVA adjusted for multiple comparisons was performed unless stated otherwise.

Source data

Extended Data Fig. 7 Gating strategies to identify NK cells.

a. Gating strategy for sorting WT or Irf4/ Ly49H+ NK cells. b. Gating strategy for phenotypic analysis of Ly49H+ NK cells after lymphocytes, singlets and live cells cleanup (as depicted in Extended Data Fig. 7a). c. Gating strategy for human NK cells.

Source data

Supplementary information

Reporting Summary

Supplementary Table 1

A list of antibodies used for flow cytometric analysis and scRNA-seq HTO-tag.

Source data

Source Data main figures

Statistical source data.

Source Data extended data

Statistical source data.

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Santosa, E.K., Kim, H., Rückert, T. et al. Control of nutrient uptake by IRF4 orchestrates innate immune memory. Nat Immunol 24, 1685–1697 (2023). https://doi.org/10.1038/s41590-023-01620-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-023-01620-z

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