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:

Srebp-controlled glucose metabolism is essential for NK cell functional responses

Abstract

Activated natural killer (NK) cells engage in a robust metabolic response that is required for normal effector function. Using genetic, pharmacological and metabolic analyses, we demonstrated an essential role for Srebp transcription factors in cytokine-induced metabolic reprogramming of NK cells that was independent of their conventional role in the control of lipid synthesis. Srebp was required for elevated glycolysis and oxidative phosphorylation and promoted a distinct metabolic pathway configuration in which glucose was metabolized to cytosolic citrate via the citrate–malate shuttle. Preventing the activation of Srebp or direct inhibition of the citrate–malate shuttle inhibited production of interferon-γ and NK cell cytotoxicity. Thus, Srebp controls glucose metabolism in NK cells, and this Srebp-dependent regulation is critical for NK cell effector function.

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

Figure 1: Srebp transcription factors are required for cytokine-induced NK cell growth and proliferation.
Figure 2: Srebp transcription factors are required for cytokine-induced glycolysis and OxPhos.
Figure 3: Inhibition of Srebp does not prevent mTORC1 signaling.
Figure 4: Glucose is metabolized to cytosolic citrate in cytokine activated NK cells.
Figure 5: Cytosolic citrate metabolism is needed to sustain elevated OxPhos and glycolysis.
Figure 6: The Srebp-controlled citrate–malate shuttle is required for NK cell effector function.
Figure 7: Srebp activity is important for therapeutic NK cell anti-tumor responses in vivo.

Similar content being viewed by others

References

  1. Pearce, E.L., Poffenberger, M.C., Chang, C.H. & Jones, R.G. Fueling immunity: insights into metabolism and lymphocyte function. Science 342, 1242454 (2013).

    PubMed  PubMed Central  Google Scholar 

  2. Loftus, R.M. & Finlay, D.K. Immunometabolism: cellular metabolism turns immune regulator. J. Biol. Chem. 291, 1–10 (2016).

    CAS  PubMed  Google Scholar 

  3. Assmann, N. & Finlay, D.K. Metabolic regulation of immune responses: therapeutic opportunities. J. Clin. Invest. 126, 2031–2039 (2016).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  5. Lynch, L.A. et al. Are natural killer cells protecting the metabolically healthy obese patient? Obesity 17, 601–605 (2009).

    CAS  PubMed  Google Scholar 

  6. O'Shea, D., Cawood, T.J., O'Farrelly, C. & Lynch, L. Natural killer cells in obesity: impaired function and increased susceptibility to the effects of cigarette smoke. PLoS One 5, e8660 (2010).

    PubMed  PubMed Central  Google Scholar 

  7. Laue, T. et al. Altered NK cell function in obese healthy humans. BMC Obes. 2, 1 (2015).

    PubMed  PubMed Central  Google Scholar 

  8. Gardiner, C.M. NK cell function and receptor diversity in the context of HCV infection. Front. Microbiol. 6, 1061 (2015).

    PubMed  PubMed Central  Google Scholar 

  9. Oliviero, B. et al. Natural killer cell functional dichotomy in chronic hepatitis B and chronic hepatitis C virus infections. Gastroenterology. 137, 1151–1160 (2009).

    CAS  PubMed  Google Scholar 

  10. Arnold, M. et al. Global burden of cancer attributable to high body-mass index in 2012: a population-based study. Lancet Oncol. 16, 36–46 (2015).

    PubMed  Google Scholar 

  11. Sun, Y. et al. Weight and prognosis for influenza A(H1N1)pdm09 infection during the pandemic period between 2009 and 2011: a systematic review of observational studies with meta-analysis. Infect. Dis. 48, 813–822 (2016).

    Google Scholar 

  12. Donnelly, R.P. et al. mTORC1-dependent metabolic reprogramming is a prerequisite for NK cell effector function. J. Immunol. 193, 4477–4484 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

  14. Kidani, Y. et al. Sterol regulatory element-binding proteins are essential for the metabolic programming of effector T cells and adaptive immunity. Nat. Immunol. 14, 489–499 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  15. Berod, L. et al. De novo fatty acid synthesis controls the fate between regulatory T and T helper 17 cells. Nat. Med. 20, 1327–1333 (2014).

    CAS  PubMed  Google Scholar 

  16. Shimomura, I., Shimano, H., Korn, B.S., Bashmakov, Y. & Horton, J.D. Nuclear sterol regulatory element-binding proteins activate genes responsible for the entire program of unsaturated fatty acid biosynthesis in transgenic mouse liver. J. Biol. Chem. 273, 35299–35306 (1998).

    CAS  PubMed  Google Scholar 

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

    PubMed  PubMed Central  Google Scholar 

  18. Horton, J.D., Goldstein, J.L. & Brown, M.S. SREBPs: activators of the complete program of cholesterol and fatty acid synthesis in the liver. J. Clin. Invest. 109, 1125–1131 (2002).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Radhakrishnan, A., Ikeda, Y., Kwon, H.J., Brown, M.S. & Goldstein, J.L. Sterol-regulated transport of SREBPs from endoplasmic reticulum to Golgi: oxysterols block transport by binding to Insig. Proc. Natl. Acad. Sci. USA 104, 6511–6518 (2007).

    CAS  PubMed  Google Scholar 

  20. Hawkins, J.L. et al. Pharmacologic inhibition of site 1 protease activity inhibits sterol regulatory element-binding protein processing and reduces lipogenic enzyme gene expression and lipid synthesis in cultured cells and experimental animals. J. Pharmacol. Exp. Ther. 326, 801–808 (2008).

    CAS  PubMed  Google Scholar 

  21. Bakan, I. & Laplante, M. Connecting mTORC1 signaling to SREBP-1 activation. Curr. Opin. Lipidol. 23, 226–234 (2012).

    CAS  PubMed  Google Scholar 

  22. Hameyer, D. et al. Toxicity of ligand-dependent Cre recombinases and generation of a conditional Cre deleter mouse allowing mosaic recombination in peripheral tissues. Physiol. Genomics 31, 32–41 (2007).

    CAS  PubMed  Google Scholar 

  23. Sun, T., Hayakawa, K., Bateman, K.S. & Fraser, M.E. Identification of the citrate-binding site of human ATP-citrate lyase using X-ray crystallography. J. Biol. Chem. 285, 27418–27428 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Gnoni, G.V., Priore, P., Geelen, M.J. & Siculella, L. The mitochondrial citrate carrier: metabolic role and regulation of its activity and expression. IUBMB Life 61, 987–994 (2009).

    CAS  PubMed  Google Scholar 

  25. Sato, R. et al. Transcriptional regulation of the ATP citrate-lyase gene by sterol regulatory element-binding proteins. J. Biol. Chem. 275, 12497–12502 (2000).

    CAS  PubMed  Google Scholar 

  26. Shimano, H. et al. Sterol regulatory element-binding protein-1 as a key transcription factor for nutritional induction of lipogenic enzyme genes. J. Biol. Chem. 274, 35832–35839 (1999).

    CAS  PubMed  Google Scholar 

  27. Amemiya-Kudo, M. et al. Transcriptional activities of nuclear SREBP-1a, -1c, and -2 to different target promoters of lipogenic and cholesterogenic genes. J. Lipid Res. 43, 1220–1235 (2002).

    CAS  PubMed  Google Scholar 

  28. Moon, Y.A., Lee, J.J., Park, S.W., Ahn, Y.H. & Kim, K.S. The roles of sterol regulatory element-binding proteins in the transactivation of the rat ATP citrate-lyase promoter. J. Biol. Chem. 275, 30280–30286 (2000).

    CAS  PubMed  Google Scholar 

  29. Infantino, V., Iacobazzi, V., De Santis, F., Mastrapasqua, M. & Palmieri, F. Transcription of the mitochondrial citrate carrier gene: role of SREBP-1, upregulation by insulin and downregulation by PUFA. Biochem. Biophys. Res. Commun. 356, 249–254 (2007).

    CAS  PubMed  Google Scholar 

  30. Guillerey, C., Huntington, N.D. & Smyth, M.J. Targeting natural killer cells in cancer immunotherapy. Nat. Immunol. 17, 1025–1036 (2016).

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Cui, G. et al. Liver X receptor (LXR) mediates negative regulation of mouse and human Th17 differentiation. J. Clin. Invest. 121, 658–670 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  33. York, A.G. et al. Limiting cholesterol biosynthetic flux spontaneously engages type I IFN signaling. Cell 163, 1716–1729 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Im, S.S. et al. Linking lipid metabolism to the innate immune response in macrophages through sterol regulatory element binding protein-1a. Cell Metab. 13, 540–549 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Reboldi, A. et al. Inflammation. 25-Hydroxycholesterol suppresses interleukin-1-driven inflammation downstream of type I interferon. Science 345, 679–684 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  36. O'Sullivan, D. et al. Memory CD8+ T cells use cell-intrinsic lipolysis to support the metabolic programming necessary for development. Immunity 41, 75–88 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Huang, S.C. et al. Cell-intrinsic lysosomal lipolysis is essential for alternative activation of macrophages. Nat. Immunol. 15, 846–855 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Fernández-Sánchez, A. et al. DNA demethylation and histone H3K9 acetylation determine the active transcription of the NKG2D gene in human CD8+ T and NK cells. Epigenetics 8, 66–78 (2013).

    PubMed  PubMed Central  Google Scholar 

  39. Chang, S. & Aune, T.M. Histone hyperacetylated domains across the Ifng gene region in natural killer cells and T cells. Proc. Natl. Acad. Sci. USA 102, 17095–17100 (2005).

    CAS  PubMed  Google Scholar 

  40. Miyake, T. et al. IκBζ is essential for natural killer cell activation in response to IL-12 and IL-18. Proc. Natl. Acad. Sci. USA 107, 17680–17685 (2010).

    CAS  PubMed  Google Scholar 

  41. Cheng, S.C. et al. mTOR- and HIF-1α-mediated aerobic glycolysis as metabolic basis for trained immunity. Science 345, 1250684 (2014).

    PubMed  PubMed Central  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  Google Scholar 

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

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Balmer, M.L. et al. Memory CD8+ T Cells require increased concentrations of acetate induced by stress for optimal function. Immunity 44, 1312–1324 (2016).

    CAS  PubMed  Google Scholar 

  46. Murakami, H., Tamasawa, N., Matsui, J., Yasujima, M. & Suda, T. Plasma oxysterols and tocopherol in patients with diabetes mellitus and hyperlipidemia. Lipids 35, 333–338 (2000).

    CAS  PubMed  Google Scholar 

  47. Bertolotti, M. et al. Increased appearance rate of 27-hydroxycholesterol in vivo in hypercholesterolemia: a possible compensatory mechanism. Nutr. Metab. Cardiovasc. Dis. 22, 823–830 (2012).

    CAS  PubMed  Google Scholar 

  48. Eibinger, G. et al. On the role of 25-hydroxycholesterol synthesis by glioblastoma cell lines. Implications for chemotactic monocyte recruitment. Exp. Cell Res. 319, 1828–1838 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. York, A.G. & Bensinger, S.J. Subverting sterols: rerouting an oxysterol-signaling pathway to promote tumor growth. J. Exp. Med. 210, 1653–1656 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  50. Gold, E.S. et al. 25-Hydroxycholesterol acts as an amplifier of inflammatory signaling. Proc. Natl. Acad. Sci. USA 111, 10666–10671 (2014).

    CAS  PubMed  Google Scholar 

  51. Matsuda, M. et al. SREBP cleavage-activating protein (SCAP) is required for increased lipid synthesis in liver induced by cholesterol deprivation and insulin elevation. Genes Dev. 15, 1206–1216 (2001).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. van der Goot, A.T. et al. Delaying aging and the aging-associated decline in protein homeostasis by inhibition of tryptophan degradation. Proc. Natl. Acad. Sci. USA 109, 14912–14917 (2012).

    CAS  PubMed  Google Scholar 

  53. Dettmer, K. et al. Metabolite extraction from adherently growing mammalian cells for metabolomics studies: optimization of harvesting and extraction protocols. Anal. Bioanal. Chem. 399, 1127–1139 (2011).

    CAS  PubMed  Google Scholar 

  54. Masood, A., Stark, K.D. & Salem, N. Jr. A simplified and efficient method for the analysis of fatty acid methyl esters suitable for large clinical studies. J. Lipid Res. 46, 2299–2305 (2005).

    CAS  PubMed  Google Scholar 

  55. Dettmer, K. Assessment of ionic liquid stationary phases for the GC analysis of fatty acid methyl esters. Anal. Bioanal. Chem. 406, 4931–4939 (2014).

    CAS  PubMed  Google Scholar 

  56. Millard, P., Letisse, F., Sokol, S. & Portais, J.C. IsoCor: correcting MS data in isotope labeling experiments. Bioinformatics 28, 1294–1296 (2012).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank the Comparative Medicine Unit, Trinity College Dublin for use of their facilities. Supported by Science Foundation Ireland (12/IP/1286 and 13/CDA/2161 for the D.K.F. laboratory); Irish Cancer Society (research scholarship CRS15OBR to K.L.O'B. which includes support from the Children's Leukaemia Research Project), Coordenadoria de Aperfeiçoamento de Pessoal de Nível Superior (CAPES), Brazil (research scholarship BEX 13446134 to V.Z.B.) and Deutsche Forschungsgemeinschaft (KFO-262 for K.D.).

Author information

Authors and Affiliations

Authors

Contributions

Conceptualization, N.A., R.P.D. and D.K.F.; methodology, N.A., R.P.D., L.D., V.Z.-B., C.M.G., K.D. and D.K.F.; investigation, N.A., K.L.O'B. R.P.D., L.D., V.Z.-B., R.M.L., K.D. and P.H.; writing (original draft), N.A. and D.K.F.; writing (review and editing), N.A., K.L.O'B., P.J.O., C.M.G. and D.K.F.; and supervision, P.J.O., L.L., C.M.G. and D.K.F.

Corresponding author

Correspondence to David K Finlay.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Srebp transcription factors are activated in NK cells stimulated with IL-2 plus IL-12.

(a) Schematic figure of Srebp activation and processing. Srebp is synthesized as an integral membrane protein within the Endoplasmic Reticulum (ER). Following translocation of Srebp along with Scap to the Golgi (←) two protease cleavage events release the active Srebp transcription factor, which then enters the nucleus (↑) and binds to Sterol regulatory element (SRE) containing promoters. 25HC inhibits Srebp activation by promoting the interaction of Scap with Insig, an ER anchor protein, thus preventing Srebp translocation to the Golgi. PF429242 inhibits site 1 protease (S1P) preventing Srebp processing. Deletion of Scap inhibits Srebp activation by preventing Srebp translocation to the Golgi. (b) Quantitative RT-PCR analysis of Fasn, Scd1, Hmgcs1 and Acat2 mRNA in NK cells left unstimulated or stimulated for 18 h with IL-2 + IL-12 +/- PF429242. (c) Analysis of the expression of Fasn, Hmgcs1 and Acat2 mRNA in NK cells stimulated for 18 h with IL-2 + IL-12 +/- rapamycin by qRT-PCR. (d) Gating strategy for the analysis of murine NK cells. Data shown is mean +/- S.E.M of 3-6 experiments. mRNA values shown are relative to IL-2 + IL-12 stimulated NK cells. * P<0.05, ** P <0.01, *** P <0.001, **** P <0.0001. (1-sample t-test).

Supplementary Figure 2 Srebp transcription factors are required for cytokine induced growth and proliferation of NK cells.

(a-c) CFSE-stained NK cells were left unstimulated or stimulated with IL-2 + IL-12 +/- PF429242 (a,b) or +/- C75 (b,c) for 24-48 h and analyzed by flow cytometry for FSC-A and CFSE-dilution, as indicated. (b) Quantitative analysis of proliferated NK cells after 48 h of NK cell stimulation with IL-2 + IL-12 +/- the inhibitors 25HC, PF429242, TOFA or C75. Statistical analysis relative to IL-2 + IL-12 stimulated NK cells is shown on the graph with other statistical comparisons summarized in the table below. (d) Viability analysis of NK cells stimulated with IL-2 + IL-12 for 20 h +/- 25HC, PF429242 or TOFA. (e,f) IL2-induced proliferation of cytotoxic T lymphocytes (CTL) was measured as the increase in cell number after 48 h +/- PF429242, 25HC (e) or TOFA (f). The dashed line indicates the density at which the cells were seeded. Data shown are representative of 5 experiments (a,c) or mean +/- S.E.M of 4 (e,f) or 5 (b,d) experiments. *P <0.05, ** P <0.01, **** P <0.0001. (One-way ANOVA with Tukey post-test (b,d,e); paired two-tailed t-test (f)).

Supplementary Figure 3 Calculations made from Seahorse traces.

(a) Calculation of (top) glycolytic reserve, glycolytic capacity and basal rate of glycolysis from extracellular acidification rate (ECAR) trace, and (bottom) spare respiratory capacity (SRC), maximal respiration and rate of basal oxidative phosphorylation (OxPhos) from oxygen consumption rate (OCR) trace. (b,c) Basal glycolysis (b) and glycolytic capacity (c) were calculated for NK cells stimulated for 18 h with IL-2 + IL-12 +/- TOFA or C75. Values are relative to NK cells stimulated in the absence of inhibitors. (d) ScapKO (Scapflox/flox x Tamox-Cre) or ScapWT (ScapWT/WT x Tamox-Cre) NK cells were analyzed by qRT-PCR for the expression of Scap, Fasn, Hmgcs1 and Acat2 mRNA. Values shown are relative to ScapWT NK cells. (e) Quantitative analysis of proliferated ScapKO and ScapWT NK cells stimulated for 48 h with IL-2 + IL-12. Data shown is mean +/- S.E.M of 3-4 experiments. *P <0.05, ** P <0.01. (Paired two-tailed t-test (e); one-sample t-test (b-d)).

Supplementary Figure 4 Srebp transcription factors are required for cytokine induced glycolysis and OxPhos.

(a) Quantitative RT-PCR analysis of G6pd mRNA in unstimulated NK cells or IL-2 + IL-12 stimulated NK cells treated for 18 h with or without the inhibitors 25HC or PF429242. Values shown are relative to IL-2 + IL-12 stimulated NK cells. (b) Schematic figure of 1,2-13C2-glucose flux through glycolysis and pentose phosphate pathway. Filled circles indicate 13C-labeled carbon. Data shown is mean +/- S.E.M of 3 experiments. (One-way ANOVA with Tukey post-test).

Supplementary Figure 5 Glucose is metabolized to cytosolic citrate in cytokine activated NK cells.

(a) Analysis of isotopologue distribution of citrate in NK cells stimulated +/- IL-2 + IL-12 for 18 h in media containing 13C6-glucose. (b) Schematic figure of isotopologue distribution of citrate, succinate and fumarate in the case where the majority of citrate is exported to the cytosol (left) or where citrate progresses through the TCA cycle (right). (c) NK cells were stimulated for 18 h with IL-2 + IL-12 and then extracellular acidification rates (ECAR) were calculated at the second measurement after the addition of SB204990 or vehicle control. (d,e) IL2-maintained cytotoxic T lymphocytes (CTL) were analyzed for ECAR values at the second measurement following injection of BMS303141 (d), SB204990 (e) or vehicle control (DMSO). (f) Analysis of fold increase in ECAR values at the second measurement after injection of SB204990 to 18 h with IL-2 + IL-12-stimulated NK cells or IL2-cultured CTLs relative to DMSO control. Data shown is mean +/- S.E.M of 4 (d), 4-5 (f), 5 (c,e) or 6 (a) independent experiments. ns, non-significant, * P<0.05, ** P<0.01, *** P<0.001, **** P<0.0001. (Two-way ANOVA with Sidak post-test (a); paired (c,d,e) or unpaired (f) two-tailed t-test).

Supplementary Figure 6 The Srebp-controlled citrate–malate shuttle is required for NK cell effector function.

(a,b) NK cells were stimulated for 20 h with IL-2 + IL-12 in the presence or absence of TOFA or C75 and IFNγ production (a) and granzyme B (b) expression analyzed by flow cytometry. (c) Gating strategy for the analysis of human NK cells (d-g) Human NK cells were stimulated with IL-12 + IL-15 for 18 h +/- 25HC or PF249242 and IFNγ production (d,e) and granzyme B (f,g) expression analyzed by flow cytometry. Shown is MFI of IFN-γ positive NK cells (d-e). Data shown is mean +/- S.E.M of 4 (a,b), 7 (d), 8 (f) or 9 (e,g) experiments. * P<0.05, ns, non-significant. (One-way ANOVA with Tukey post-test (a,b); paired two-tailed t-test (d-g)).

Supplementary Figure 7 The Srebp dependent citrate–malate shuttle.

Glycolysis-derived pyruvate is typically considered to promote mitochondrial oxidative phosphorylation (OxPhos) through fuelling the tricarboxylic acid (TCA) cycle (left). The TCA cycle feeds electrons into the electron transport chain via NADH and FADH2 reducing equivalents. Herein, this study demonstrates that cytokine activated NK cells adopt a distinct metabolic configuration called the citrate-malate shuttle (CMS)(right). Pyruvate is metabolized to acetyl-CoA, which is used with oxaloacetate (OAA) to generate citrate. This mitochondrial citrate is then exported into the cytoplasm through the citrate-malate antiporter (Slc25a1). Slc25a1 is an obligate antiporter and so to sustain citrate export, malate must be imported into the mitochondria. In the cytoplasm, citrate is cleaved by ATP citrate lyase (ACLY) into acetyl-CoA and OAA. OAA is further metabolized to generate malate to be imported into the mitochondria, where it is converted back to OAA, thus completing the cycle. The net outputs of the CMS are (1) the import of electrons from NADH in the cytoplasm to generate mitochondrial NADH to fuel OxPhos, (2) the regeneration of NAD+ in the cytoplasm, an essential cofactor for GAPDH and therefore glycolysis, and (3) the generation of cytoplasmic acetyl-CoA, which is important for acetylation reactions and fatty acid synthesis. Cytokine activated NK cells primarily use the CMS, and not the TCA cycle, to drive OxPhos. Srebp transcription factors control the expression of the two key components of the CMS, Slc25a1 and ACLY.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–7 and Supplementary Table 1. (PDF 1312 kb)

Life Sciences Reporting Summary (PDF 200 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Assmann, N., O'Brien, K., Donnelly, R. et al. Srebp-controlled glucose metabolism is essential for NK cell functional responses. Nat Immunol 18, 1197–1206 (2017). https://doi.org/10.1038/ni.3838

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ni.3838

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