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:

MEF2C regulates NK cell effector functions through control of lipid metabolism

An Author Correction to this article was published on 19 April 2024

This article has been updated

Abstract

Natural killer (NK) cells are a critical first line of defense against viral infection. Rare mutations in a small subset of transcription factors can result in decreased NK cell numbers and function in humans, with an associated increased susceptibility to viral infection. However, our understanding of the specific transcription factors governing mature human NK cell function is limited. Here we use a non-viral CRISPR–Cas9 knockout screen targeting genes encoding 31 transcription factors differentially expressed during human NK cell development. We identify myocyte enhancer factor 2C (MEF2C) as a master regulator of human NK cell functionality ex vivo. MEF2C-haploinsufficient patients and mice displayed defects in NK cell development and effector function, with an increased susceptibility to viral infection. Mechanistically, MEF2C was required for an interleukin (IL)-2- and IL-15-mediated increase in lipid content through regulation of sterol regulatory element-binding protein (SREBP) pathways. Supplementation with oleic acid restored MEF2C-deficient and MEF2C-haploinsufficient patient NK cell cytotoxic function. Therefore, MEF2C is a critical orchestrator of NK cell antiviral immunity by regulating SREBP-mediated lipid metabolism.

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: MEF2C is required for human NK cell proliferation and effector function.
Fig. 2: Patients with MCHS present with a functional NK cell deficiency.
Fig. 3: MEF2C haploinsufficiency disrupts antiviral immunity.
Fig. 4: MEF2C is required for IL-15- and mTORC1-induced metabolic reprogramming.
Fig. 5: MEF2C promotes cytokine-activated SREBP signaling and increased lipid content in NK cells.
Fig. 6: Fatty acid supplementation restores MEF2C-deficient NK cell cytotoxicity.

Similar content being viewed by others

Data availability

Sequence data have been deposited in the GEO database under the accession code GSE245463. Gene expression data in sorted human immune cells were provided by the DICE project. Publicly available RNA-seq datasets for human peripheral NK cells sorted by flow cytometry were accessed at GSE112813. Publicly available RNA-seq datasets for cytokine-stimulated NK cells were accessed at GSE140035. RNA-seq data were aligned using the reference mouse genome mm10 or the reference human genome hg38. All other data are available in the main text or Supplementary Information. Source data are provided with this paper.

Change history

References

  1. Bjorkstrom, N. K., Strunz, B. & Ljunggren, H. G. Natural killer cells in antiviral immunity. Nat. Rev. Immunol. 22, 112–123 (2022).

    Article  PubMed  Google Scholar 

  2. Mace, E. M. et al. Mutations in GATA2 cause human NK cell deficiency with specific loss of the CD56bright subset. Blood 121, 2669–2677 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. 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 

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

    Article  PubMed  Google Scholar 

  5. Cottineau, J. et al. Inherited GINS1 deficiency underlies growth retardation along with neutropenia and NK cell deficiency. J. Clin. Invest. 127, 1991–2006 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  6. Gineau, L. et al. Partial MCM4 deficiency in patients with growth retardation, adrenal insufficiency, and natural killer cell deficiency. J. Clin. Invest. 122, 821–832 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Grier, J. T. et al. Human immunodeficiency-causing mutation defines CD16 in spontaneous NK cell cytotoxicity. J. Clin. Invest. 122, 3769–3780 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Cooper, M. A. et al. Human natural killer cells: a unique innate immunoregulatory role for the CD56bright subset. Blood 97, 3146–3151 (2001).

    Article  CAS  PubMed  Google Scholar 

  9. Fehniger, T. A. et al. CD56bright natural killer cells are present in human lymph nodes and are activated by T cell-derived IL-2: a potential new link between adaptive and innate immunity. Blood 101, 3052–3057 (2003).

    Article  CAS  PubMed  Google Scholar 

  10. Michel, T. et al. Human CD56bright NK cells: an update. J. Immunol. 196, 2923–2931 (2016).

    Article  CAS  PubMed  Google Scholar 

  11. Lopez-Verges, S. et al. CD57 defines a functionally distinct population of mature NK cells in the human CD56dimCD16+ NK-cell subset. Blood 116, 3865–3874 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Kee, B. L., Morman, R. E. & Sun, M. Transcriptional regulation of natural killer cell development and maturation. Adv. Immunol. 146, 1–28 (2020).

    CAS  PubMed  Google Scholar 

  13. Ramirez, K. et al. Gene deregulation and chronic activation in natural killer cells deficient in the transcription factor ETS1. Immunity 36, 921–932 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Daussy, C. et al. T-bet and Eomes instruct the development of two distinct natural killer cell lineages in the liver and in the bone marrow. J. Exp. Med. 211, 563–577 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Jeevan-Raj, B. et al. The transcription factor Tcf1 contributes to normal NK cell development and function by limiting the expression of granzymes. Cell Rep. 20, 613–626 (2017).

    Article  CAS  PubMed  Google Scholar 

  16. Boos, M. D., Yokota, Y., Eberl, G. & Kee, B. L. Mature natural killer cell and lymphoid tissue-inducing cell development requires Id2-mediated suppression of E protein activity. J. Exp. Med. 204, 1119–1130 (2007).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Gascoyne, D. M. et al. The basic leucine zipper transcription factor E4BP4 is essential for natural killer cell development. Nat. Immunol. 10, 1118–1124 (2009).

    Article  CAS  PubMed  Google Scholar 

  18. Kamizono, S. et al. Nfil3/E4bp4 is required for the development and maturation of NK cells in vivo. J. Exp. Med. 206, 2977–2986 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Rautela, J., Surgenor, E. & Huntington, N. D. Drug target validation in primary human natural killer cells using CRISPR RNP. J. Leukoc. Biol. 108, 1397–1408 (2020).

    Article  CAS  PubMed  Google Scholar 

  22. Riggan, L. et al. CRISPR–Cas9 ribonucleoprotein-mediated genomic editing in mature primary innate immune cells. Cell Rep. 31, 107651 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Collins, P. L. et al. Gene regulatory programs conferring phenotypic identities to human NK cells. Cell 176, 348–360 (2019).

    Article  CAS  PubMed  Google Scholar 

  24. Khameneh, H. J. et al. Myc controls NK cell development, IL-15-driven expansion, and translational machinery. Life Sci. Alliance 6, e202302069 (2023).

  25. Li, Z. Y. et al. The transcriptional repressor ID2 supports natural killer cell maturation by controlling TCF1 amplitude. J. Exp. Med. 218, e20202032 (2021).

  26. 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).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Okada, S. et al. Impairment of immunity to Candida and Mycobacterium in humans with bi-allelic RORC mutations. Science 349, 606–613 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Cooley Coleman, J. A. et al. Comprehensive investigation of the phenotype of MEF2C-related disorders in human patients: a systematic review. Am. J. Med. Genet. A 185, 3884–3894 (2021).

    Article  PubMed  Google Scholar 

  29. Harrington, A. J. et al. MEF2C hypofunction in neuronal and neuroimmune populations produces MEF2C haploinsufficiency syndrome-like behaviors in mice. Biol. Psychiatry 88, 488–499 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Lin, Q., Schwarz, J., Bucana, C. & Olson, E. N. Control of mouse cardiac morphogenesis and myogenesis by transcription factor MEF2C. Science 276, 1404–1407 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Mitrovic, M. et al. The NK cell response to mouse cytomegalovirus infection affects the level and kinetics of the early CD8+ T-cell response. J. Virol. 86, 2165–2175 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Weizman, O. E. et al. ILC1 confer early host protection at initial sites of viral infection. Cell 171, 795–808 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Mah, A. Y. et al. Glycolytic requirement for NK cell cytotoxicity and cytomegalovirus control. JCI Insight 2, e95128 (2017).

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Bonacina, F. et al. The low-density lipoprotein receptor–mTORC1 axis coordinates CD8+ T cell activation. J. Cell Biol. 221, e202202011 (2022).

  36. Shimano, H. & Sato, R. SREBP-regulated lipid metabolism: convergent physiology—divergent pathophysiology. Nat. Rev. Endocrinol. 13, 710–730 (2017).

    Article  CAS  PubMed  Google Scholar 

  37. Martin-Rufino, J. D. et al. Massively parallel base editing to map variant effects in human hematopoiesis. Cell 186, 2456–2474 (2023).

    Article  CAS  PubMed  Google Scholar 

  38. McAuley, G. E. et al. Human T cell generation is restored in CD3δ severe combined immunodeficiency through adenine base editing. Cell 186, 1398–1416 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Cheng, J. et al. Accurate proteome-wide missense variant effect prediction with AlphaMissense. Science 381, eadg7492 (2023).

    Article  CAS  PubMed  Google Scholar 

  40. Keating, S. E. et al. Metabolic reprogramming supports IFN-γ production by CD56bright NK cells. J. Immunol. 196, 2552–2560 (2016).

    Article  CAS  PubMed  Google Scholar 

  41. Assmann, N. et al. Srebp-controlled glucose metabolism is essential for NK cell functional responses. Nat. Immunol. 18, 1197–1206 (2017).

    Article  CAS  PubMed  Google Scholar 

  42. Duvel, K. et al. Activation of a metabolic gene regulatory network downstream of mTOR complex 1. Mol. Cell 39, 171–183 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  43. Lewis, C. A., Griffiths, B., Santos, C. R., Pende, M. & Schulze, A. Genetic ablation of S6-kinase does not prevent processing of SREBP1. Adv. Enzym. Regul. 51, 280–290 (2011).

    Article  CAS  Google Scholar 

  44. Saleiro, D. & Platanias, L. C. Intersection of mTOR and STAT signaling in immunity. Trends Immunol. 36, 21–29 (2015).

    Article  CAS  PubMed  Google Scholar 

  45. Molkentin, J. D., Black, B. L., Martin, J. F. & Olson, E. N. Mutational analysis of the DNA binding, dimerization, and transcriptional activation domains of MEF2C. Mol. Cell. Biol. 16, 2627–2636 (1996).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Michelet, X. et al. Metabolic reprogramming of natural killer cells in obesity limits antitumor responses. Nat. Immunol. 19, 1330–1340 (2018).

    Article  CAS  PubMed  Google Scholar 

  47. Kobayashi, T. et al. Increased lipid metabolism impairs NK cell function and mediates adaptation to the lymphoma environment. Blood 136, 3004–3017 (2020).

    Article  CAS  PubMed  Google Scholar 

  48. Qin, W. H. et al. High serum levels of cholesterol increase antitumor functions of nature killer cells and reduce growth of liver tumors in mice. Gastroenterology 158, 1713–1727 (2020).

    Article  CAS  PubMed  Google Scholar 

  49. Tanaka, T., Porter, C. M., Horvath-Arcidiacono, J. A. & Bloom, E. T. Lipophilic statins suppress cytotoxicity by freshly isolated natural killer cells through modulation of granule exocytosis. Int. Immunol. 19, 163–173 (2007).

    Article  CAS  PubMed  Google Scholar 

  50. Hillyard, D. Z. et al. Statins inhibit NK cell cytotoxicity by membrane raft depletion rather than inhibition of isoprenylation. Atherosclerosis 191, 319–325 (2007).

    Article  CAS  PubMed  Google Scholar 

  51. Cheng, M. I. et al. The X-linked epigenetic regulator UTX controls NK cell-intrinsic sex differences. Nat. Immunol. 24, 780–791 (2023).

  52. 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).

  53. Wang, T. et al. Identification and characterization of essential genes in the human genome. Science 350, 1096–1101 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Kluesner, M. G. et al. CRISPR–Cas9 cytidine and adenosine base editing of splice-sites mediates highly-efficient disruption of proteins in primary and immortalized cells. Nat. Commun. 12, 2437 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Divakaruni, A. S., Paradyse, A., Ferrick, D. A., Murphy, A. N. & Jastroch, M. Analysis and interpretation of microplate-based oxygen consumption and pH data. Methods Enzymol. 547, 309–354 (2014).

    Article  CAS  PubMed  Google Scholar 

  56. Arguello, R. J. et al. SCENITH: a flow cytometry-based method to functionally profile energy metabolism with single-cell resolution. Cell Metab. 32, 1063–1075 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  57. Schmiedel, B. J. et al. Impact of genetic polymorphisms on human immune cell gene expression. Cell 175, 1701–1715 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Weizman, O. E. et al. Mouse cytomegalovirus-experienced ILC1s acquire a memory response dependent on the viral glycoprotein m12. Nat. Immunol. 20, 1004–1011 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We deeply thank the patients with MCHS and their families who participated in this study. We thank the nurses, staff and physicians at Greenwood Genetic Center for their outstanding evaluation and care for children with genetic disorders and inborn errors of immunity. We thank the blood donors, E. Faure, A. Catapang and the UCLA CFAR Virology Core for providing healthy donor peripheral blood samples. We thank S. Feng and the UCLA Broad Stem Cell Research Center High-Throughput Sequencing Core for assistance with RNA-seq. We thank the B. Moriarity laboratory at the University of Minnesota for providing the plasmid encoding ABE8e and associated protocols. We thank M. Lechner at UCLA for sharing the β2M−/− MC38 cell line. We thank the O’Sullivan, Bensinger, Cowan, Covarrubias, Su, Divakaruni, Hoffman and Christofk laboratories for helpful discussion. We acknowledge the rich literature of inborn errors of immunity and NK cell deficiency and regret that not all studies could be discussed. T.E.O. is supported by the National Institutes of Health (R01AI145997, R01AI174519) and the Hypothesis Fund. J.H.L. is supported by the National Institutes of Health (T32GM008042, T32AR071307, T32AI007323, 1F30AI181449-01) and a UCLA Molecular Biology Institute Whitcome Fellowship. C.D.L. is supported by the National Institutes of Health (T32GM008042). C.G.B. is supported by a UCLA Eugene Cota V. Robles Fellowship. C.W.C. is supported by the National Institutes of Health (R01MH111464) and a SFARI Pilot Award (649452). A.S.D. is supported by the National Institutes of Health (R35GM138003), the W.M. Keck Foundation (995337) and an Agilent Early Career Professor Award. A.B.B. is supported by the National Institutes of Health (T32GM136614). The UCLA CFAR Virology Core is supported by the National Institutes of Health (5P30AI028697).

Author information

Authors and Affiliations

Authors

Contributions

J.H.L. designed the project, performed and analyzed all experiments and wrote the paper with input from all authors. A.Z., C.D.L., S.N.S. and Q.F. performed and analyzed experiments. J.H.J. analyzed sequencing data. V.S., S.N.S., E.T.P. and C.G.B. tested sgRNA species and performed the CRISPR cRNP screen. A.B.B. and A.S.D. performed and analyzed Seahorse metabolic experiments. L.R. developed the initial human NK cell culture and CRISPR cRNP system. A.G. and C.W.C. provided Mef2c+/− bone marrow. F.A., J.C.-C. and S.A.S. performed clinical evaluations of patients with MCHS and coordinated and provided patient samples. T.E.O. conceived and designed the project, supervised experiments and wrote the paper.

Corresponding author

Correspondence to Timothy E. O’Sullivan.

Ethics declarations

Competing interests

T.E.O. is a scientific advisor for Modulus Therapeutics and Xyphos Biosciences, companies that have a financial interest in human NK cell-based therapeutics. The Regents of the University of California have filed a provisional patent application with the United States Patent and Trademark Office for using oleate supplementation as a method of augmenting adoptive NK cell therapy. J.H.L. and T.E.O. are listed as inventors on this patent application. The other authors declare no competing interests.

Peer review

Peer review information

Nature Immunology thanks the anonymous reviewers for their contribution to the peer review of this work. Primary Handling Editor: N. Bernard, in collaboration with the Nature Immunology team.

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 Functional CRISPR cRNP screen in primary human NK cells identifies positive regulators of human NK cell function.

(a) Differentially expressed transcription factors between human NK cell developmental subsets. (b) Primary human NK cells are isolated from fresh human PBMCs via negative magnetic bead selection. Following expansion in IL-2/15 for 9 days, cells are electroporated with CRISPR-Cas9 RNP complexes. CRISPR-edited NK cells are further expanded for 6 days before functional and flow cytometric analyses. (c) Left, quantification of percent IFN-γ+ in TRACcRNP or TCF7cRNP NK cells after 16 h stimulation with IL-2, IL-15, K562 cells, and IL-12 and/or IL-18. Right, specific lysis of K562 cells after 16 h coculture with IL-2 and IL-15 at indicated effector:target ratios. (d) Left, density of viable TRACcRNP or MYCcRNP NK cells 6 days after cRNP editing expanded with IL-2 and IL-15. Right, quantification of percent IFN-γ+ in TRACcRNP or MYCcRNP NK cells after 16 h stimulation with IL-2, IL-15, K562 cells, and IL-12 and/or IL-18. (e) Density of viable TRACcRNP or ZEB2cRNP NK cells 6 days after cRNP editing expanded with IL-2 and IL-15. (f) Quantification of percent IFN-γ+ in TRACcRNP or RORCcRNP NK cells after 16 h stimulation with IL-2, IL-15, K562 cells, and IL-12 and/or IL-18. (g) Representative gating strategy for peripheral human NK cells. Data are representative of n = 6-8 independent donors presented as individual paired donors. *p < 0.05, **p < 0.01 by two-sided paired t test. Specific p-values are as follows: c percent IFN-γ+ NT = 0.1686, IL-18 = 0.0200, IL-12 = 0.0136, IL-12/18 = 0.8445, % specific lysis 1:8 = 0.0399, 1:4 = 0.0366; d cells/mL = 0.0040, percent IFN-γ+ NT = 0.0769, IL-18 = 0.0128, IL-12 = 0.0244, IL-12/18 = 0.4241; e = 0.0216, f = 0.0456.

Extended Data Fig. 2 MEF2C is required for human NK cell function without impacting fitness.

(a) Immunoblot showing MEF2C protein expression in TRACcRNP or MEF2CcRNP NK cells 6 days after CRISPR cRNP editing. (b) Indel percentage by CRISPR cRNP editing in TRACcRNP or MEF2CcRNP NK cells 6 days after editing. Sanger sequencing results were analyzed using SYNTHEGO ICE analysis software. (c) Quantification of annexin+ early apoptotic or annexin+PI+ late apoptotic TRACcRNP or MEF2CcRNP NK cells 6 days after editing. (d) MFI of BCL2, BIM, or ratio of BCL2/BIM MFI in TRACcRNP or MEF2CcRNP NK cells 6 days after editing. (e) Specific lysis of A375 human melanoma cells after 16 h coculture with TRACcRNP or MEF2CcRNP NK cells in the presence of IL-2/15. (f) MFI of perforin in TRACcRNP or MEF2CcRNP NK cells 6 days after editing. Data are representative of n = 47 independent donors presented as individual paired donors. *p < 0.05, **p < 0.01 by two-sided paired t test. Specific p-values are as follows: c = 0.0361, 0.0204; d BCL2 = 0.0047, BIM = 0.2848, BCL2/BIM = 0.1384; e = 0.0405; f = 0.9390.

Source data

Extended Data Fig. 3 MEF2C haploinsufficiency disrupts CD56dim NK cells without impacting CD56bri or other circulating immune populations.

(a) Schematic of pathogenic point mutations in MCHS patients. (b) Expression of MEF2C in FACS-sorted peripheral human immune cells based on DICE database data. (c) Frequency of non-NK cell immune populations in peripheral blood of healthy donor control or MCHS patients. (d) Percent IFN-γ+ (above) and IFN-γ MFI of cytokine-producing cells (below) of total (left) or CD56bri (right) healthy donor control or MCHS patient NK cells stimulated for 16 h with IL-2, IL-15, K562 cells, and IL-12 after 5 d expansion in IL-2/15. (e) MEF2C transcript expression in NK cell maturation subsets. (f-g) MFI of perforin (f) or GzmB (g) in healthy donor or MCHS patient NK cells by maturation subset. (h) MFI of GzmB in healthy donor or MCHS patient CD56dim NK cells. (i) Schematic showing adenine base editor (ABE8e) mediated generation of MEF2C point mutation. (j) Assessment of point mutation frequency at targeted base using MEF2C-targeting sgRNA in conjunction with electroporation of ABE8e mRNA in healthy primary human NK cells. Point mutation rate was evaluated by Sanger sequencing and analysis using the EditR package on day 6 post ABE8e electroporation in culture with IL-2/15. (c) Left, gated on CD3+CD14- T cells, CD3CD14+ monocytes, and CD3CD19+ B cells. Center, gated on CD3+CD14CD4+ or CD3+CD14CD8+ cells. Right, gated on CD3CD14+CD16hi classical, CD3CD14+CD16int intermediate, or CD3CD14+CD16lo non-classical monocytes. (d,f-h) Gated on CD3CD56+ cells or CD3CD56dimCD16+ cells. Data represent mean ± SEM. Data are representative of (c,d,f-h) n = 5-8 independent healthy donors alongside n = 2 MCHS patients each sampled two independent times, (e) n = 34 independent donors, or (j) n = 6 independent donors. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001 by two-sided Student’s t test. Specific p-values are as follows: d CD56bri percent IFN-γ+ NT = 0.0010, IL-18 = 0.0029; g = 0.0198; h = 0.0039; j < 0.0001.

Extended Data Fig. 4 Mef2c haploinsufficiency impairs antiviral immunity.

(a) Schematic of WT:Mef2c+/- mixed bone marrow chimeric (mBMC) mice or WT and Mef2c+/- single bone marrow chimeric (sBMC) mice. (b) Percent WT or Mef2c+/- bone marrow-derived NK cells on D0 or D42 post bone marrow transplant of WT:Mef2c+/- mBMC mice. (c) Representative contour plots (left) and frequencies (right) of peripheral NK cell subsets of wild-type:Mef2c+/- mixed bone marrow chimeric (mBMC) mice after 4 weeks engraftment. (d-e) Percent IFN-γ+ (left) and IFN-γ MFI of cytokine-producing cells (right) of splenic NK cells from WT:Mef2c+/- mBMC mice stimulated ex vivo with IL-15 and IL-12 (d) or IL-18 (e) stratified by maturation subset. (f) Percent IFN-γ+ (left) and IFN-γ MFI of cytokine-producing cells (right) of splenic NK cells from WT:Mef2c+/- mBMC mice on D1.5 post MCMV. (g) Schematic of adoptive transfer of WT:Mef2c+/- mBMC splenocytes. (h) Schematic of adoptive transfer of CRISPR edited Rosa26cRNP or Mef2ccRNP NK cells. (i) Representative contour plots (left) and frequency (right) of Rosa26cRNP or Mef2ccRNP Ly49H+KLRG1+ mouse NK cells on D0 and D7 post MCMV. (j) Percent IFN-γ+ of Rosa26cRNP or Mef2ccRNP NK cells stimulated for 4 h with IL-12 or IL-18. (k) Percent specific lysis of MC38 β2 M-/- by Rosa26cRNP or Mef2ccRNP NK cells at 1:1 E:T. (l) Representative gating strategy for splenic mouse NK cells. (b-e,j) Gated on CD3TCRβ-NK1.1+ cells. (f,i) Gated on CD3TCRβ-NK1.1+Ly49H+KLRG1+ cells. Data shown as mean ± SEM, paired WT and Mef2c+/- NK cells from the same mBMC mouse, or paired Rosa26cRNP and Mef2ccRNP NK cells from the same mouse where applicable. Data representative of at least 2 independent experiments. Data are representative of (b,c) n = 23, (d,e) n = 11, (f) n = 10, (i,j) n = 4, and (k) n = 7 mice. *p < 0.05, **p < 0.01, ***p < 0.001, ****p < 0.0001 by two-sided paired t test. Specific p-values are as follows: b < 0.0001; c DN = 0.0020, DP < 0.0001, CD11b SP < 0.0001; d CD11b SP percent IFN-γ+ = 0.0144, CD11b SP IFN-γ MFI = 0.0469; e CD11b SP percent IFN-γ+ = 0.0447; f CD11b SP percent IFN-γ+ = 0.0032; DP IFN-γ MFI = 0.0409, CD11b SP IFN-γ MFI = 0.0016; i < 0.0001; j IL-12 percent IFN-γ+ = 0.0484, IL-18 percent IFN-γ+ = 0.0466; k = 0.0009.

Extended Data Fig. 5 MEF2C is required for cytokine-activated metabolic reprogramming.

(a) MFI of CD25/IL-2Rα (left) or CD122/IL-2Rβ (right) of peripheral blood NK cells from WT or Mef2c+/- sBMC mice on D0 or D1.5 of MCMV infection. (b) MFI of CD25/IL-2Rα (left) or CD122/IL-2Rβ (right) of TRACcRNP or MEF2CcRNP human NK cells 6 days post CRISPR edit. (c) MFI of pSTAT5 (left) or pSTAT1 (center) and representative histogram of pSTAT1 expression (right) in splenic NK cells from WT or Mef2c+/- sBMC mice on D1.5 of MCMV infection. FMO, fluorescence minus one control. (d) MFI of pSTAT5 (left) and representative histogram (right) of TRACcRNP or MEF2CcRNP human NK cells 6 days post CRISPR edit. FMO, fluorescence minus one control. (e) Maximal respiration of TRACcRNP or MEF2CcRNP human NK cells 6 days post CRISPR edit evaluated by Seahorse extracellular flux assay. (f) Metabolic dependencies (left) and translation rate (right) of TRACcRNP or MEF2CcRNP human NK cells 6 days post CRISPR edit measured by SCENITH. Gluc dep, glucose dependence; Mito dep, mitochondrial dependence; Glyc cap, glycolytic capacity; FAO/AAO cap, fatty acid oxidation/amino acid oxidation capacity; Co, control; DG, 2-deoxyglucose; O, oligomycin. (g) Metabolic dependencies of TRACcRNP or MEF2CcRNP human NK cells 6 days post CRISPR edit measured by SCENITH, stratified by maturation subset. (a) Gated on CD3TCRβ-NK1.1+ cells. (b,f,g) Gated on CD3-CD56+ cells. Data represent (a) n = 5-7 mice per group, (b,d) n = 3 paired independent donors, (c) n = 3-4 mice per group, (e) n = 5, or (f,g) n = 4-6 paired independent donors. *p < 0.05, **p < 0.01 by two-sided paired t test. Specific p-values are as follows: f mito dep = 0.0091, glyc cap = 0.0091, Co puro MFI = 0.0140, O puro MFI = 0.0040; g CD16− = 0.0492, 0.0492; CD16+CD57− = 0.0491, 0.0322, 0.0322, 0.0491.

Extended Data Fig. 6 MEF2C maintains SREBP activity in mouse and human NK cells.

(a) Heatmap showing differentially expressed genes from RNA-seq performed on human and mouse control and MEF2C knockout NK cells. (b) Gene Ontology pathway analysis of downregulated genes conserved between human MEF2CcRNP and mouse Mef2ccRNP NK cells compared to control edited cells ranked by FDR. (c) Gene Ontology pathway analysis of downregulated genes conserved between human MEF2CcRNP and mouse Mef2ccRNP NK cells compared to control edited cells ranked by FDR, excluding mitosis and cell division-related pathways. (d) Heat maps showing changes in gene expression of canonical SREBP pathway genes separated by biological replicate with hierarchical clustering of genes. (e) Transcript expression of SREBF1 and SCD1 in TRACcRNP or MEF2CcRNP human NK cells 6 days post CRISPR edit. (f) MFI of BODIPY 493/503 of total splenic NK cells from naive and D1.5 MCMV-infected mice. (g-h) MFI of LDLR (g) or BODIPY 493/503 (h) in TRACcRNP or LDLRcRNP human NK cells 6 days post CRISPR edit. (i) Proposed model of MEF2C-directed lipid metabolic reprogramming driving NK cell effector function in response to cytokine stimulation and mTORc1 activation. Data represent mean ± SEM or individual paired donors where applicable. (f) Gated on naïve CD3TCRβ-NK1.1+ cells or D1.5 CD3TCRβ-NK1.1+Ly49H+KLRG1+ cells. (g-h) Gated on CD3CD56+ cells. Data are representative of (a-d) n = 6 mice and n = 3 independent donors, (e) n = 67 independent donors, (f) n = 13 naive and 10 D1.5 mice, or (g-h) n = 6 independent donors. *p < 0.05, **p < 0.01 by two-sided paired t test or Student’s t test. Specific p-values are as follows: e = 0.0108, 0.0421; f = 0.0092; g = 0.0032, h = 0.0485.

Supplementary information

Source data

Source Data

Fig. 4 and Extended Data Fig. 2.

Unprocessed immunoblots for Fig. 4a–c and Extended Data Fig. 2a.

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

Li, J.H., Zhou, A., Lee, C.D. et al. MEF2C regulates NK cell effector functions through control of lipid metabolism. Nat Immunol (2024). https://doi.org/10.1038/s41590-024-01811-2

Download citation

  • Received:

  • Accepted:

  • Published:

  • DOI: https://doi.org/10.1038/s41590-024-01811-2

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