Skip to main content

Thank you for visiting nature.com. You are using a browser version with limited support for CSS. To obtain the best experience, we recommend you use a more up to date browser (or turn off compatibility mode in Internet Explorer). In the meantime, to ensure continued support, we are displaying the site without styles and JavaScript.

  • Article
  • Published:

The IRE1 endoplasmic reticulum stress sensor activates natural killer cell immunity in part by regulating c-Myc

Abstract

Natural killer (NK) cells are critical mediators of host immunity to pathogens. Here, we demonstrate that the endoplasmic reticulum stress sensor inositol-requiring enzyme 1 (IRE1α) and its substrate transcription factor X-box-binding protein 1 (XBP1) drive NK cell responses against viral infection and tumors in vivo. IRE1α-XBP1 were essential for expansion of activated mouse and human NK cells and are situated downstream of the mammalian target of rapamycin signaling pathway. Transcriptome and chromatin immunoprecipitation analysis revealed c-Myc as a new and direct downstream target of XBP1 for regulation of NK cell proliferation. Genetic ablation or pharmaceutical blockade of IRE1α downregulated c-Myc, and NK cells with c-Myc haploinsufficency phenocopied IRE1α-XBP1 deficiency. c-Myc overexpression largely rescued the proliferation defect in IRE1α−/− NK cells. Like c-Myc, IRE1α-XBP1 also promotes oxidative phosphorylation in NK cells. Overall, our study identifies a IRE1α-XBP1-cMyc axis in NK cell immunity, providing insight into host protection against infection and cancer.

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: Induction of IRE1α-XBP1 UPR in mouse and human activated NK cells in vitro and in vivo.
Fig. 2: IRE1α is required for optimal protective antiviral NK cell responses.
Fig. 3: IRE1α-XBP1 controls infection-induced NK cell proliferation but not survival.
Fig. 4: IRE1α-XBP1 supports NK homeostatic proliferation.
Fig. 5: IRE1 supports NK cell OXPHOS and mitochondrial function.
Fig. 6: XBP1 promotes NK cell proliferation at least partially via direct regulation of c-Myc.
Fig. 7: Restoration of c-Myc in the absence of IRE1 rescues the NK cell proliferation defect.
Fig. 8: Intrinsic requirement of IRE1α-XBP1 for NK cell-mediated antitumor immunity.

Similar content being viewed by others

Data availability

The RNA-seq data were deposited in the Gene Expression Omnibus under the accession number GSE113214. The remaining data that support the findings of this study are available from the corresponding authors upon request. Materials will be provided with material transfer agreements in place as appropriate.

References

  1. Caligiuri, M. A. Human natural killer cells. Blood 112, 461–469 (2008).

    Article  CAS  Google Scholar 

  2. 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  Google Scholar 

  3. Zawislak, C. L. et al. Stage-specific regulation of natural killer cell homeostasis and response against viral infection by microRNA-155. Proc. Natl Acad. Sci. USA 110, 6967–6972 (2013).

    Article  CAS  Google Scholar 

  4. 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  Google Scholar 

  5. 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).

    Article  CAS  Google Scholar 

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

    Article  Google Scholar 

  7. Morvan, M. G. & Lanier, L. L. NK cells and cancer: you can teach innate cells new tricks. Nat. Rev. Cancer 16, 7–19 (2016).

    Article  CAS  Google Scholar 

  8. Yoshida, H., Matsui, T., Yamamoto, A., Okada, T. & Mori, K. XBP1 mRNA is induced by ATF6 and spliced by IRE1 in response to ER stress to produce a highly active transcription factor. Cell 107, 881–891 (2001).

    Article  CAS  Google Scholar 

  9. Lee, A. H., Iwakoshi, N. N. & Glimcher, L. H. XBP-1 regulates a subset of endoplasmic reticulum resident chaperone genes in the unfolded protein response. Mol. Cell. Biol. 23, 7448–7459 (2003).

    Article  CAS  Google Scholar 

  10. Lee, A. H., Heidtman, K., Hotamisligil, G. S. & Glimcher, L. H. Dual and opposing roles of the unfolded protein response regulated by IRE1alpha and XBP1 in proinsulin processing and insulin secretion. Proc. Natl Acad. Sci. USA 108, 8885–8890 (2011).

    Article  CAS  Google Scholar 

  11. Lee, A. H., Scapa, E. F., Cohen, D. E. & Glimcher, L. H. Regulation of hepatic lipogenesis by the transcription factor XBP1. Science 320, 1492–1496 (2008).

    Article  CAS  Google Scholar 

  12. Kaser, A. et al. XBP1 links ER stress to intestinal inflammation and confers genetic risk for human inflammatory bowel disease. Cell 134, 743–756 (2008).

    Article  CAS  Google Scholar 

  13. Chen, X. et al. XBP1 promotes triple-negative breast cancer by controlling the HIF1α pathway. Nature 508, 103–107 (2014).

    Article  CAS  Google Scholar 

  14. Condamine, T. et al. ER stress regulates myeloid-derived suppressor cell fate through TRAIL-R-mediated apoptosis. J. Clin. Invest. 124, 2626–2639 (2014).

    Article  CAS  Google Scholar 

  15. Yan, D., Wang, H. W., Bowman, R. L. & Joyce, J. A. STAT3 and STAT6 signaling pathways synergize to promote cathepsin secretion from macrophages via IRE1α activation. Cell Rep. 16, 2914–2927 (2016).

    Article  CAS  Google Scholar 

  16. Song, M. et al. IRE1alpha-XBP1 controls T cell function in ovarian cancer by regulating mitochondrial activity. Nature 562, 423–428 (2018).

    Article  CAS  Google Scholar 

  17. Cubillos-Ruiz, J. R. et al. ER stress sensor XBP1 controls anti-tumor immunity by disrupting dendritic cell homeostasis. Cell 161, 1527–1538 (2015).

    Article  CAS  Google Scholar 

  18. Beaulieu, A. M. & Sun, J. C. Tracking effector and memory NK cells during MCMV infection. Methods Mol. Biol. 1441, 1–12 (2016).

    Article  CAS  Google Scholar 

  19. Iwawaki, T., Akai, R., Kohno, K. & Miura, M. A transgenic mouse model for monitoring endoplasmic reticulum stress. Nat. Med. 10, 98–102 (2004).

    Article  CAS  Google Scholar 

  20. 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  Google Scholar 

  21. Orr, M. T. et al. Ly49H signaling through DAP10 is essential for optimal natural killer cell responses to mouse cytomegalovirus infection. J. Exp. Med. 206, 807–817 (2009).

    Article  CAS  Google Scholar 

  22. Brandt, C. et al. Food perception primes hepatic ER homeostasis via melanocortin-dependent control of mTOR activation. Cell 175, 1321–1335.e20 (2018).

    Article  CAS  Google Scholar 

  23. Hsu, H. S. et al. Involvement of ER stress, PI3K/AKT activation, and lung fibroblast proliferation in bleomycin-induced pulmonary fibrosis. Sci. Rep. 7, 14272 (2017).

    Article  Google Scholar 

  24. Zheng, H. et al. Leptin promotes allergic airway inflammation through targeting the unfolded protein response pathway. Sci. Rep. 8, 8905 (2018).

    Article  Google Scholar 

  25. Cerwenka, A. & Lanier, L. L. Natural killer cell memory in infection, inflammation and cancer. Nat. Rev. Immunol. 16, 112–123 (2016).

    Article  CAS  Google Scholar 

  26. Dokun, A. O. et al. Specific and nonspecific NK cell activation during virus infection. Nat. Immunol. 2, 951–956 (2001).

    Article  CAS  Google Scholar 

  27. Sun, J. C., Beilke, J. N., Bezman, N. A. & Lanier, L. L. Homeostatic proliferation generates long-lived natural killer cells that respond against viral infection. J. Exp. Med. 208, 357–368 (2011).

    Article  CAS  Google Scholar 

  28. Mao, Y. et al. IL-15 activates mTOR and primes stress-activated gene expression leading to prolonged antitumor capacity of NK cells. Blood 128, 1475–1489 (2016).

    Article  CAS  Google Scholar 

  29. Wagner, J. A. et al. CD56bright NK cells exhibit potent antitumor responses following IL-15 priming. J. Clin. Invest. 127, 4042–4058 (2017).

    Article  Google Scholar 

  30. Cross, B. C. et al. The molecular basis for selective inhibition of unconventional mRNA splicing by an IRE1-binding small molecule. Proc. Natl Acad. Sci. USA 109, E869–E878 (2012).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  32. Buck, M. D., Sowell, R. T., Kaech, S. M. & Pearce, E. L. Metabolic instruction of immunity. Cell 169, 570–586 (2017).

    Article  CAS  Google Scholar 

  33. 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).

    Article  Google Scholar 

  34. O’Sullivan, T. E. & Sun, J. C. Innate lymphoid cell immunometabolism. J. Mol. Biol. 429, 3577–3586 (2017).

    Article  Google Scholar 

  35. Morrish, F. & Hockenbery, D. MYC and mitochondrial biogenesis. Cold Spring Harb. Perspect. Med. 4, a014225 (2014).

    Article  Google Scholar 

  36. van Riggelen, J., Yetil, A. & Felsher, D. W. MYC as a regulator of ribosome biogenesis and protein synthesis. Nat. Rev. Cancer 10, 301–309 (2010).

    Article  Google Scholar 

  37. Farrell, A. S. & Sears, R. C. MYC degradation. Cold Spring Harb. Perspect. Med. 4, a014365 (2014).

    Article  Google Scholar 

  38. Lu, Y. et al. MYC targeted long noncoding RNA DANCR promotes cancer in part by reducing p21 levels. Cancer Res. 78, 64–74 (2018).

    Article  CAS  Google Scholar 

  39. Clauss, I. M., Chu, M., Zhao, J. L. & Glimcher, L. H. The basic domain/leucine zipper protein hXBP-1 preferentially binds to and transactivates CRE-like sequences containing an ACGT core. Nucleic Acids Res. 24, 1855–1864 (1996).

    Article  CAS  Google Scholar 

  40. Allan, D. S. et al. An in vitro model of innate lymphoid cell function and differentiation. Mucosal Immunol. 8, 340–351 (2015).

    Article  CAS  Google Scholar 

  41. Robertson, M. J. et al. Characterization of a cell line, NKL, derived from an aggressive human natural killer cell leukemia. Exp. Hematol. 24, 406–415 (1996).

    CAS  Google Scholar 

  42. Frazier, W. R., Steiner, N., Hou, L., Dakshanamurthy, S. & Hurley, C. K. Allelic variation in KIR2DL3 generates a KIR2DL2-like receptor with increased binding to its HLA-C ligand. J. Immunol. 190, 6198–6208 (2013).

    Article  CAS  Google Scholar 

  43. Foley, B. et al. The biology of NK cells and their receptors affects clinical outcomes after hematopoietic cell transplantation (HCT). Immunol. Rev. 258, 45–63 (2014).

    Article  CAS  Google Scholar 

  44. Ruggeri, L. et al. Effectiveness of donor natural killer cell alloreactivity in mismatched hematopoietic transplants. Science 295, 2097–2100 (2002).

    Article  CAS  Google Scholar 

  45. Molgora, M. et al. IL-1R8 is a checkpoint in NK cells regulating anti-tumour and anti-viral activity. Nature 551, 110–114 (2017).

    Article  Google Scholar 

  46. Bottcher, J. P. et al. NK cells stimulate recruitment of cDC1 into the tumor microenvironment promoting cancer immune control. Cell 172, 1022–1037.e14 (2018).

    Article  CAS  Google Scholar 

  47. Siddiquey, M. N. A., Zhang, H., Nguyen, C. C., Domma, A. J. & Kamil, J. P. The human cytomegalovirus endoplasmic reticulum-resident glycoprotein UL148 activates the unfolded protein response. J. Virol. 92, e00896-18 (2018).

    Article  Google Scholar 

  48. Sun, J. C. & Lanier, L. L. Is there natural killer cell memory and can it be harnessed by vaccination? NK cell memory and immunization strategies against infectious diseases and cancer. Cold Spring Harb. Perspect. Biol. 10, a029538 (2018).

    Article  Google Scholar 

  49. Orange, J. S. Natural killer cell deficiency. J. Allergy Clin. Immunol. 132, 515–525 (2013).

    Article  CAS  Google Scholar 

  50. Vivier, E., Ugolini, S., Blaise, D., Chabannon, C. & Brossay, L. Targeting natural killer cells and natural killer T cells in cancer. Nat. Rev. Immunol. 12, 239–252 (2012).

    Article  CAS  Google Scholar 

  51. Barrow, A. D. et al. Natural killer cells control tumor growth by sensing a growth factor. Cell 172, 534–548 e519 (2018).

    Article  CAS  Google Scholar 

  52. Ferrari de Andrade, L. et al. Antibody-mediated inhibition of MICA and MICB shedding promotes NK cell-driven tumor immunity. Science 359, 1537–1542 (2018).

    Article  CAS  Google Scholar 

  53. Adams, N. M. et al. Transcription factor IRF8 orchestrates the adaptive natural killer cell response. Immunity 48, 1172–1182 e1176 (2018).

    Article  CAS  Google Scholar 

  54. 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.e13 (2018).

    Article  CAS  Google Scholar 

  55. van Montfoort, N. et al. NKG2A blockade potentiates CD8 T cell immunity induced by cancer vaccines. Cell 175, 1744–1755.e15 (2018).

    Article  Google Scholar 

  56. Barry, K. C. et al. A natural killer-dendritic cell axis defines checkpoint therapy-responsive tumor microenvironments. Nat. Med. 24, 1178–1191 (2018).

    Article  CAS  Google Scholar 

  57. Rabacal, W. et al. Transcription factor KLF2 regulates homeostatic NK cell proliferation and survival. Proc. Natl Acad. Sci. USA 113, 5370–5375 (2016).

    Article  CAS  Google Scholar 

  58. Wang, Y. et al. The IL-15-AKT-XBP1s signaling pathway contributes to effector functions and survival in human NK cells. Nat. Immunol. 20, 10–17 (2019).

    Article  CAS  Google Scholar 

  59. Marcais, A. et al. The metabolic checkpoint kinase mTOR is essential for IL-15 signaling during the development and activation of NK cells. Nat. Immun. 15, 749–757 (2014).

    Article  CAS  Google Scholar 

  60. Yang, C. et al. mTORC1 and mTORC2 differentially promote natural killer cell development. eLife 7, e35619 (2018).

    Article  Google Scholar 

  61. Zhao, N. et al. Pharmacological targeting of MYC-regulated IRE1/XBP1 pathway suppresses MYC-driven breast cancer. J. Clin. Invest. 128, 1283–1299 (2018).

    Article  Google Scholar 

  62. Kortlever, R. M. et al. Myc cooperates with ras by programming inflammation and immune suppression. Cell 171, 1301–1315.e14 (2017).

    Article  CAS  Google Scholar 

  63. Chou, C. et al. c-Myc-induced transcription factor AP4 is required for host protection mediated by CD8 + T cells. Nat. Immunol. 15, 884–893 (2014).

    Article  CAS  Google Scholar 

  64. Cichocki, F. et al. The transcription factor c-Myc enhances KIR gene transcription through direct binding to an upstream distal promoter element. Blood 113, 3245–3253 (2009).

    Article  CAS  Google Scholar 

  65. Zakiryanova, G. K. et al. Alterations of oncogenes expression in NK cells in patients with cancer. Immun. Inflamm. Dis. 5, 493–502 (2017).

    Article  CAS  Google Scholar 

  66. 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).

    Article  CAS  Google Scholar 

  67. Iwawaki, T., Akai, R., Yamanaka, S. & Kohno, K. Function of IRE1 alpha in the placenta is essential for placental development and embryonic viability. Proc. Natl Acad. Sci. USA 106, 16657–16662 (2009).

    Article  CAS  Google Scholar 

  68. Trapnell, C. et al. Differential gene and transcript expression analysis of RNA-seq experiments with TopHat and Cufflinks. Nat. Protoc. 7, 562–578 (2012).

    Article  CAS  Google Scholar 

  69. Anders, S., Pyl, P. T. & Huber, W. HTSeq—a Python framework to work with high-throughput sequencing data. Bioinformatics 31, 166–169 (2015).

    Article  CAS  Google Scholar 

  70. Anders, S. & Huber, W. Differential expression analysis for sequence count data. Genome Biol. 11, R106 (2010).

    Article  CAS  Google Scholar 

  71. 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).

    Article  CAS  Google Scholar 

  72. Chen, X. et al. Integration of external signaling pathways with the core transcriptional network in embryonic stem cells. Cell 133, 1106–1117 (2008).

    Article  CAS  Google Scholar 

  73. Peinado, H. et al. Melanoma exosomes educate bone marrow progenitor cells toward a pro-metastatic phenotype through MET. Nat. Med. 18, 883–891 (2012).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank E. Vivier (Aix Marseille University) for Ncr1iCre mice; R. Wang (Nationwide Children’s Hospital) for Mycfl/fl mice; R. Sears (Oregon Health & Science University) for Mycfsf/fsf mice; J. Ritz (Dana-Farber Cancer Institute) for the NKL cell line; C. Hurley (Georgetown University) for the KHYG-1 cell line; D. Lyden (Weill Cornell Medicine) for the B16F10 cell line; members of the Sun laboratory for sharing viral reagents, cytokines and flow cytometry antibodies and for providing experimental assistance; members of the Glimcher laboratory—M. Raundhal, C. Lentucci, R. Xu, S. Ghosh and A. Gonzalez for technical support; S. Adoro (Case Western Reserve University) and J. Cubillos-Ruiz (Weill Cornell Medicine) for critical reading of the manuscript and helpful discussion; M. Song (Weill Cornell Medicine) and the Wucherpfennig laboratory (Dana-Farber Cancer Institute) for insightful comments; J. Xiang (Weill Cornell Genomics and Epigenomics Core) for RNA-seq; J. McCormick (Weill Cornell Medicine) and DFCI Jimmy Fund Flow Cytometry Core for cell sorting; Specialized Histopathology Core at Brigham & Women’s Hospital for histology; L. Cohen-Gould and J. Jimenez (Microscopy and Image Analysis Core Facility at Weill Cornell Medicine) for electron microscopy; X. Liu (Metabolism and Mitochondrial Research Core, Beth Israel Deaconess Medical Center) for metabolic flux assays; D. Neuberg (Dana-Farber Cancer Institute) for advising on statistical analysis; and S. Schneider (Dana-Farber Cancer Institute) for manuscript editing. L.H.G. and H.D. were supported by Institutional Funding from The Dana-Farber Cancer Institute. H.D. was also supported by a Helen Gurley Brown Presidential Initiative Award (Dana-Farber Cancer Institute). J.C.S. was supported by the Ludwig Center for Cancer Immunotherapy, the American Cancer Society, the Burroughs Wellcome Fund and the NIH (grant nos. AI100874, AI130043 and P30CA008748). N.M.A. was supported by an MSTP Grant from the National Institute of General Medical Sciences of the NIH to the Weill Cornell/Rockefeller/Sloan Kettering Tri-Institutional MD-PhD Program (grant no. T32GM007739) and by an F30 Predoctoral Fellowship from National Institue of Allergy and Infectious Diseases of the NIH (no. F30 AI136239). X.C. was supported by a NIH grant (no. 1 R37 CA228304-01).

Author information

Authors and Affiliations

Authors

Contributions

L.H.G. conceptualized the project. L.H.G and J.C.S provided supervision. L.H.G., J.C.S, H.D. and N.M.A. designed the experiments. H.D. carried out the experiments. H.D., L.H.G. and J.C.S. analyzed the data. Y.X. performed the bioinformatics analysis. J.C. did the ChIP experiments and analysis under the supervision of X.C. D.S.J.A. and J.R.C. provided critical reagents. H.D., N.M.A. and Y.X. made the figures. H.D. and N.M.A. wrote the original draft. X.C. proposed key experiments and provided critical feedback on the manuscript. J.C.S. and L.H.G. reviewed and edited the manuscript and figures.

Corresponding author

Correspondence to Laurie H. Glimcher.

Ethics declarations

Competing interests

L.H.G. is a former Director of Bristol-Myers Squibb and is currently on the board of directors of and holds equity in GlaxoSmithKline Pharmaceuticals and the Waters Corporation. She chairs the scientific advisory board, is a co-founder of and holds equity in Quentis Therapeutics. She also serves on the scientific advisory boards of Repare Therapeutics, Abpro and Kaleido Therapeutics.

Additional information

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

Integrated supplementary information

Supplementary Figure 1 qPCR validation of upregulation of the IRE1α/XBP1 pathway in activated NK cells, and requirement of STAT4 and mTOR signaling pathways in driving IRE1α /XBP1.

(a) Quantitative real time PCR analysis of indicated UPR genes in sorting, purified splenic NK cells from wild-type mice at day 1 post MCMV infection. β-ACTIN was used as reference, and data were normalized to the uninfected control expression levels. (b) Quantitative real time PCR analysis of indicated UPR genes in sorting-purified splenic NK cells from wild-type mice after 16 hr culture in the presence or absence of IL-12 and IL-18. (c) Heat map of RNA-seq analysis (GSE106138) showing the expression of canonical IRE1α/XBP1 target genes in WT and STAT4-deficient Ly49H+ NK cells sorted from the spleens of mixed BM chimeric mice at day 2 PI. (d) Flow cytometric analysis of IRE1α activation in cytokine-activated NK cells after pharmaceutical inhibition of mTOR. NK cells from ERAI reporter mice were stimulated with mouse recombinant IL-12 (20 ng/ml) and IL-18 (10 ng/ml) for 6 hrs in the presence of mTOR inhibitor rapamycin (Rapa, 5 nM and 10 nM for low and high dose, respectively), Ku-0063794 (Ku, 1.5 μM and 3 μM for low and high dose, respectively), PP242 (PP, 0.5 μM and 1 μM for low and high dose, respectively), or IRE1 inhibitor 4μ8C (2.5 μM and 5 μM for low and high dose, respectively), or DMSO control. Representative flow cytometric plot (upper) and quantifications of relative inhibition efficiency (bottom) are shown. ns, not significant; * p < 0.05, ** p < 0.01, *** p < 0.0001 and **** p < 0.0001. Two-way analysis of variance (ANOVA) with the Sidak post-test performed on a. Two tailed unpaired Student’s t-test performed on b. All error bars, mean with s.d. from biological replicates. n = 3 mice/group in a. n = 3 mice in b; with technical triplicates for ex vivo culture. Each column is a different mouse in c. n = 3 ERAI mice in d. Experiments were independently repeated three (a, b) and two (d) times.

Supplementary Figure 2 IRE1α/XBP1 is dispensable in NK cell development & maturation.

(a) Validation of IRE1NK KO efficiency: XBP1 splicing assay (upper panel) and quantitative real time PCR analysis (bottom panel) of indicated UPR genes in sorting-purified splenic NK cells from IRE1NK and IRE1f/f littermate mice after 4 hr ex vivo incubation in the presence or absence of tunicamycin, a pharmacologic inducer of ER stress. The quantitative real time PCR data are presented as relative expression to β-Actin. (b) Flow cytometric analysis of NK cell percentages and absolute numbers in BM, spleen and lung of IRE1NK and XBP1NK naïve mice, in comparison to their Cre-littermate controls. BM NK cells are identified by LinCD122 and splenic NK cells are identified by LinNK1.1+. (c) Flow cytometric analysis of NK cell development in BM of IRE1NK and XBP1NK mice: percentages of NK cells that are NK progenitors (NKP, DX5 NK1.1), immature NK cells (iNK, DX5 NK1.1+) or mature NK (mNK, DX5 NK1.1+) are shown. (d) Flow cytometric analysis of NK cell maturation in spleen of IRE1NK and XBP1NK mice: percentages of NK cells that are immature (CD27+ CD11b), mature (CD27 CD11b+) or intermediate stage (CD27+ CD11b+) are shown. (e) Flow cytometric analysis of NK cell repopulation in mixed BM chimera mice: WT (CD45.1): IRE1NK (CD45.2), and WT (CD45.1): IRE1vav1 (CD45.2) BM chimeras were generated as in the schematic (left); NK cell repopulation in the irradiated recipient mice was assessed in the peripheral blood at week 8 after BM transfer (right). (f) As in d, representative histogram plots showing surface expression of cytokine receptors and activating receptors in splenic NK cells from naïve IRE1NK and XBP1NK in comparison to their Cre- littermate control mice. ns: not significant, * p < 0.05, ** p < 0.01, *** p < 0.001 and ****p < 0.0001. Two tailed unpaired Student’s t-test performed on a. All error bars, mean with s.d.. n = 3 mice/group in a, f. n = 5 mice/group in b-d. n = 10 mice/group in e. Experiments were independently repeated three (a-e) and two (a, f) times.

Supplementary Figure 3 Dispensable function of IRE1α/XBP1 in NK cell priming, cytokine production or cytotoxicity.

(a) Gating strategy applied in analysis of co-transfer and mixed BM chimera experiments. (b-c) Flow cytometric analysis of splenic WT or IRE1NK Ly49H+ NK cells from mixed BM chimeric mice as indicated in Supplementary Figure 2e at day 2 PI: (b)IFN-γ and (c)CD69 and intracellular Granzyme B. (d) Flow cytometric analysis of co-transferred WT or IRE1NK Ly49H+ NK cells from the spleen of Ly49H-deficient recipients at day 2 PI: pan-Akt, p-Akt, and p-S6 with quantification normalized to WT expression levels as 100%. Two tailed unpaired Student’s t-test performed on b and c. All error bars, mean with s.d. n = 6 mixed BM chimera mice in b. n = 4 mice in c. n = 3 mice in d. Data were independently repeated three times.

Supplementary Figure 4 Minimal impact of IRE1α depletion on RIDD in infection-activated NK cells, and specific pharmaceutical blockade of IRE1α by 4μ8C.

(a) The volcano plot of RNA-seq analysis showing all genes in IRE1NK versus WT Ly49H+ splenic NK cells harvested from IRE1NK (CD45.2): WT (CD45.1) mixed BM chimera mice day 1.5 PI. RIDD target genes1 and XBP1 target genes1 are highlighted in red and blue, respectively. (b) IRE1NK and IRE1f/f NK cells from littermate animals were pre-labeled with CTV and cultured ex vivo with IL-2 and IL-15 as in Fig. 4f, g, but in the presence of 4μ8C (5 uM) or DMSO control treatment. Representative flow cytometric plots of CTV dilution and Ki-67 levels at day 3 are shown. Proliferating cells are defined as Ki-67+ CTVlo. n = 2 mice/group in b with technical duplicates per mouse. Experiments in b were independently repeated two times.

1. So, J.S. et al. Silencing of lipid metabolism genes through IRE1alpha-mediated mRNA decay lowers plasma lipids in mice. Cell Metab 16, 487-499 (2012).

Supplementary Figure 5 Disrupted mitochondrial morphology in IRE1NK cells during MCMV infection.

Representative electron microscopy of IRE1NK versus WT Ly49H+ splenic NK cells harvested from IRE1NK(CD45.2): WT (CD45.1) mixed BM chimera mice day 7 PI. The high-resolution plots at bottom show mitochondrial morphology. n = 3 mixed BM chimera mice.

Supplementary Figure 6 IRE1NK RNA-seq analysis highlights Myc as an XBP1 target gene.

(a) IPA Upstream Analysis derived from RNA-seq (day 1.5 PI) as indicated in Fig. 6a: prediction of Myc regulation in IRE1NK cells. (b) IPA analysis of Functional overlap between IRE1- and c-Myc-regulated genes derived from RNA-seq analysis of Ly49H+ IRE1NK cells at day 1.5 PI. (c) Flow cytometric analysis of c-Myc induction at the level of transcription (upper row) and translation (middle row) in splenic NK cells from WT mice either naïve or at day 1 PI. (bottom row) Representative flow cytometric plots indicating the concomitant induction of c-Myc protein and Venus reporter expression in NK cells from indicated organs of ERAI transgenic mice at day 2 PI. (d) Quantitative real time PCR and flow cytometric analysis of the basal levels of c-Myc mRNA and protein in naïve NK cells from IRE1NK and IRE1f/f littermate control mice. (e) Quantitative real time PCR analysis of canonical c-Myc target genes in transferred Ly49H+ IRE1NK versus WT NK cells that were sorting-purified from the spleen of recipient Ly49H-deficient mice at day 1.5 PI. β-Actin was used as reference, and data are shown as the relative expression normalized to transferred WT NK cells. (f) Schematic of the putative XBP1 binding site in the c-Myc promoter region (based on SABiosciences’ proprietary database ENCODE). (g) Flow cytometric analysis of the kinetics of XBP1s, p-S6, p-Akt and pan-Akt expression in primary human NK cells after stimulation with IL-12 (20 ng/ml) and IL-18 (10 ng/ml) for the indicated time. (h) Purified splenic NK cells from IRE1NK and IRE1f/f littermate control mice were stimulated with mouse recombinant IL-12 (20 ng/ml) and IL-18 (10 ng/ml) for 1 hr and 16 hrs in the presence or absence of mTOR inhibitor rapamycin (10 nM). c-Myc levels were quantified by flow cytometry. (i) Flow cytometric analysis of c-Myc expression in transferred CD45.1 congenic WT and IRE1NK cells in the spleen of recipient Rag2−/−Il2rg−/− mice at specified time points after transfer. ns: not significant, * p < 0.05, ** p < 0.01, *** p < 0.001. One sample t-test performed on e. Two-way analysis of variance (ANOVA) with the Sidak post-test performed on h. Two tailed unpaired Student’s t-test performed on i. All error bars, mean with s.d. from biological replicates. n = 3~5 mice/group for all experiments and data were independently repeated two (c, g, h) or three times (d, e, i).

Supplementary Figure 7 Characterization of NK cell development and maturation in Myc+/− heterozygous mice (Het, referred to as ‘MycNK’ in Main Text) and Myc−/− (KO) mice.

(a) Quantitative real time PCR validation of c-Myc expression in Het and KO NK cells before and after IL-12 and IL-18 stimulation ex vivo. (b) Flow cytometric analysis of NK cell percentages and absolute numbers in BM and spleen of MycHet mice, in comparison to their Cre- littermate controls. BM NK cells are identified by LinCD122 and splenic NK cells are identified by LinNK1.1+. (c) as in b, except MycKO is shown. (d) Flow cytometric analysis of NK cell development in BM and NK cell maturation in spleen of MycHet mice. For BM (upper), percentages of NK cells that are NK progenitors (NKP, DX5 NK1.1), immature NK cells (iNK, DX5 NK1.1+) or mature NK (mNK, DX5 NK1.1+) are shown; for spleen (bottom), percentages of NK cells that are immature (CD27+ CD11b), mature (CD27 CD11b+) or intermediate stage (CD27+ CD11b+) are shown. (e) as in d, except MycKO is shown. (e) Representative histogram plots showing surface expression of cytokine receptors and activating receptors in splenic NK cells from MycHet mice in comparison to their Cre- littermate controls. (f) as in e, except MycKO is shown. p values as indicated. Two tailed unpaired Student’s t-test performed on a-e. All error bars, mean with s.e.m. Data are representative of three (a-e) and two (f, g) independent experiments. n = 3 mice/group for a, f, g. n = 4-5 mice/group for all experiments in b-e. Data were independently repeated three (b-e) or two (f, g) times.

Supplementary Figure 8 IRE1α-driven NK cell expansion is associated with the presence of immune cell types beneficial to tumor control.

(a) Representative flow cytometric plots and quantification of basal levels of NK cell numbers and relative percentage and Ki-67 and c-Myc expression in the lungs of naïve IRE1NK and IRE1f/f littermate control mice. (b) Gating strategy for c. (c) Percentage of conventional type 1 dendritic cells (cDC1) (left), CD8+ T cells (middle), and CD4+ T cells (right) in the lung and spleen of IRE1NK and IRE1f/f littermate control mice at day 20 following intravenous injection of B16F10 melanoma cells. * p < 0.05, ** p < 0.01. Two tailed unpaired Student’s t-test performed on c. All error bars, mean with s.e.m. n = 4 mice/group for a and 5 mice/group for c. Data were independently repeated three times.

Supplementary information

Supplementary Information

Supplementary Figures 1-8 and Supplementary Tables 1-3

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dong, H., Adams, N.M., Xu, Y. et al. The IRE1 endoplasmic reticulum stress sensor activates natural killer cell immunity in part by regulating c-Myc. Nat Immunol 20, 865–878 (2019). https://doi.org/10.1038/s41590-019-0388-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-019-0388-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