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:

Metabolic reprogramming of natural killer cells in obesity limits antitumor responses

Abstract

Up to 49% of certain types of cancer are attributed to obesity, and potential mechanisms include overproduction of hormones, adipokines, and insulin. Cytotoxic immune cells, including natural killer (NK) cells and CD8+ T cells, are important in tumor surveillance, but little is known about the impact of obesity on immunosurveillance. Here, we show that obesity induces robust peroxisome proliferator-activated receptor (PPAR)-driven lipid accumulation in NK cells, causing complete ‘paralysis’ of their cellular metabolism and trafficking. Fatty acid administration, and PPARα and PPARδ (PPARα/δ) agonists, mimicked obesity and inhibited mechanistic target of rapamycin (mTOR)-mediated glycolysis. This prevented trafficking of the cytotoxic machinery to the NK cell–tumor synapse. Inhibiting PPARα/δ or blocking the transport of lipids into mitochondria reversed NK cell metabolic paralysis and restored cytotoxicity. In vivo, NK cells had blunted antitumor responses and failed to reduce tumor growth in obesity. Our results demonstrate that the lipotoxic obese environment impairs immunosurveillance and suggest that metabolic reprogramming of NK cells may improve cancer outcomes in obesity.

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: NK cells from HFD mice show upregulation of lipid metabolism-related genes and downregulation of the killing machinery.
Fig. 2: Obesity leads to NK cell loss of function and lipid accumulation.
Fig. 3: Lipid uptake and accumulation leads to NK cell loss of function.
Fig. 4: Lipid accumulation during obesity impairs mTOR pathway.
Fig. 5: Lipid accumulation during obesity negatively impacts NK cell metabolism.
Fig. 6: Activation of the PPARα/δ pathway induces functional and metabolic defects in NK cells.
Fig. 7: The decreased ability of NK cells to kill tumor cells in obesity is related to a defect of lytic granule polarization and can be reversed with metabolic reprograming.
Fig. 8: Lipid uptake reduces NK cell antitumor activity in vivo.

Similar content being viewed by others

Data availability

The data that support the findings of this study are available from the corresponding author upon reasonable request.

References

  1. Global status report on noncommunicable diseases 2014 (WHO, 2004).

  2. Calle, E. E. & Kaaks, R. Overweight, obesity and cancer: epidemiological evidence and proposed mechanisms. Nat. Rev. Cancer 4, 579–591 (2004).

    Article  CAS  Google Scholar 

  3. Renehan, A. G., Tyson, M., Egger, M., Heller, R. F. & Zwahlen, M. Body-mass index and incidence of cancer: a systematic review and meta-analysis of prospective observational studies. Lancet 371, 569–578 (2008).

    Article  Google Scholar 

  4. Falagas, M. E. & Kompoti, M. Obesity and infection. Lancet. Infect. Dis. 6, 438–446 (2006).

    Article  Google Scholar 

  5. Sjostrom, L. et al. Effects of bariatric surgery on cancer incidence in obese patients in Sweden (Swedish Obese Subjects Study): a prospective, controlled intervention trial. Lancet Oncol. 10, 653–662 (2009).

    Article  Google Scholar 

  6. Renehan, A. G., Zwahlen, M. & Egger, M. Adiposity and cancer risk: new mechanistic insights from epidemiology. Nat. Rev. Cancer 15, 484–498 (2015).

    Article  CAS  Google Scholar 

  7. Park, J., Morley, T. S., Kim, M., Clegg, D. J. & Scherer, P. E. Obesity and cancer mechanisms underlying tumour progression and recurrence. Nat. Rev. Endocrinol. 10, 455–465 (2014).

    Article  CAS  Google Scholar 

  8. Pascual, G. et al. Targeting metastasis-initiating cells through the fatty acid receptor CD36. Nature 541, 41–45 (2017).

    Article  CAS  Google Scholar 

  9. Beyaz, S. et al. High-fat diet enhances stemness and tumorigenicity of intestinal progenitors. Nature 531, 53–58 (2016).

    Article  CAS  Google Scholar 

  10. Vivier, E., Tomasello, E., Baratin, M., Walzer, T. & Ugolini, S. Functions of natural killer cells. Nat. Immunol. 9, 503–510 (2008).

    Article  CAS  Google Scholar 

  11. Mace, E. M. et al. Cell biological steps and checkpoints in accessing NK cell cytotoxicity. Immunol. Cell Biol. 92, 245–255 (2014).

    Article  CAS  Google Scholar 

  12. Orange, J. S. Formation and function of the lytic NK-cell immunological synapse. Nat. Rev. Immunol. 8, 713–725 (2008).

    Article  CAS  Google Scholar 

  13. Chowdhury, D. & Lieberman, J. Death by a thousand cuts: granzyme pathways of programmed cell death. Annu. Rev. Immunol. 26, 389–420 (2008).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  15. 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  Google Scholar 

  16. Bjorntorp, P., Bergman, H. & Varnauskas, E. Plasma free fatty acid turnover rate in obesity. Acta Med. Scand. 185, 351–356 (1969).

    Article  CAS  Google Scholar 

  17. Jensen, M. D., Haymond, M. W., Rizza, R. A., Cryer, P. E. & Miles, J. M. Influence of body fat distribution on free fatty acid metabolism in obesity. J. Clin. Invest. 83, 1168–1173 (1989).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  19. 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).

    Article  Google Scholar 

  20. Jahn, J. et al. Decreased NK cell functions in obesity can be reactivated by fat mass reduction. Obesity (Silver Spring) 23, 2233–2241 (2015).

    Article  CAS  Google Scholar 

  21. Perdu, S. et al. Maternal obesity drives functional alterations in uterine NK cells. JCI Insight 1, e85560 (2016).

    Article  Google Scholar 

  22. Laplante, M. & Sabatini, D. M. mTOR signaling in growth control and disease. Cell 149, 274–293 (2012).

    Article  CAS  Google Scholar 

  23. Finlay, D. K. et al. PDK1 regulation of mTOR and hypoxia-inducible factor 1 integrate metabolism and migration of CD8+ T cells. J. Exp. Med. 209, 2441–2453 (2012).

    Article  CAS  Google Scholar 

  24. Wang, R. & Green, D. R. Metabolic checkpoints in activated T cells. Nat. Immunol. 13, 907–915 (2012).

    Article  CAS  Google Scholar 

  25. Sengupta, S., Peterson, T. R., Laplante, M., Oh, S. & Sabatini, D. M. mTORC1 controls fasting-induced ketogenesis and its modulation by ageing. Nature 468, 1100–1104 (2010).

    Article  CAS  Google Scholar 

  26. Lefebvre, P., Chinetti, G., Fruchart, J. C. & Staels, B. Sorting out the roles of PPAR alpha in energy metabolism and vascular homeostasis. J. Clin. Invest. 116, 571–580 (2006).

    Article  CAS  Google Scholar 

  27. Barber, D. F., Faure, M. & Long, E. O. LFA-1 contributes an early signal for NK cell cytotoxicity. J. Immunol. 173, 3653–3659 (2004).

    Article  CAS  Google Scholar 

  28. Mace, E. M., Monkley, S. J., Critchley, D. R. & Takei, F. A dual role for talin in NK cell cytotoxicity: activation of LFA-1-mediated cell adhesion and polarization of NK cells. J. Immunol. 182, 948–956 (2009).

    Article  CAS  Google Scholar 

  29. James, A. M. et al. Rapid activation receptor- or IL-2-induced lytic granule convergence in human natural killer cells requires Src, but not downstream signaling. Blood 121, 2627–2637 (2013).

    Article  CAS  Google Scholar 

  30. Mentlik, A. N., Sanborn, K. B., Holzbaur, E. L. & Orange, J. S. Rapid lytic granule convergence to the MTOC in natural killer cells is dependent on dynein but not cytolytic commitment. Mol. Biol. Cell 21, 2241–2256 (2010).

    Article  CAS  Google Scholar 

  31. Liu, D., Martina, J. A., Wu, X. S., Hammer, J. A. 3rd & Long, E. O. Two modes of lytic granule fusion during degranulation by natural killer cells. Immunol. Cell Biol. 89, 728–738 (2011).

    Article  CAS  Google Scholar 

  32. Menager, M. M. et al. Secretory cytotoxic granule maturation and exocytosis require the effector protein hMunc13-4. Nat. Immunol. 8, 257–267 (2007).

    Article  CAS  Google Scholar 

  33. Tuli, A. et al. Arf-like GTPase Arl8b regulates lytic granule polarization and natural killer cell-mediated cytotoxicity. Mol. Biol. Cell 24, 3721–3735 (2013).

    Article  CAS  Google Scholar 

  34. Kupfer, A., Dennert, G. & Singer, S. J. Polarization of the Golgi apparatus and the microtubule-organizing center within cloned natural killer cells bound to their targets. Proc. Natl Acad. Sci. USA 80, 7224–7228 (1983).

    Article  CAS  Google Scholar 

  35. Boden, G. & Shulman, G. I. Free fatty acids in obesity and type 2 diabetes: defining their role in the development of insulin resistance and beta-cell dysfunction. Eur. J. Clin. Invest. 32, 14–23 (2002).

    Article  CAS  Google Scholar 

  36. Boulenouar, S. et al. Adipose type one innate lymphoid cells regulate macrophage homeostasis through targeted cytotoxicity. Immunity 46, 273–286 (2017).

    Article  CAS  Google Scholar 

  37. Wensveen, F. M. et al. NK cells link obesity-induced adipose stress to inflammation and insulin resistance. Nat. Immunol. 16, 376–385 (2015).

    Article  CAS  Google Scholar 

  38. O’Sullivan, T. E. et al. Adipose-resident group 1 innate lymphoid cells promote obesity-associated insulin resistance. Immunity 45, 428–441 (2016).

    Article  Google Scholar 

  39. Lee, B. C. et al. Adipose natural killer cells regulate adipose tissue macrophages to promote insulin resistance in obesity. Cell Metab. 23, 685–698 (2016).

    Article  CAS  Google Scholar 

  40. Muller, A. J. et al. Chronic inflammation that facilitates tumor progression creates local immune suppression by inducing indoleamine 2,3 dioxygenase. Proc. Natl Acad. Sci. USA 105, 17073–17078 (2008).

    Article  CAS  Google Scholar 

  41. Allison, D. J. & Ditor, D. S. Immune dysfunction and chronic inflammation following spinal cord injury. Spinal Cord 53, 14–18 (2015).

    Article  CAS  Google Scholar 

  42. Gentile, L. F. et al. Persistent inflammation and immunosuppression: a common syndrome and new horizon for surgical intensive care. J. Trauma Acute Care Surg. 72, 1491–1501 (2012).

    Article  CAS  Google Scholar 

  43. Cheng, S. C. et al. Broad defects in the energy metabolism of leukocytes underlie immunoparalysis in sepsis. Nat. Immunol. 17, 406–413 (2016).

    Article  CAS  Google Scholar 

  44. Gentles, A. J. et al. The prognostic landscape of genes and infiltrating immune cells across human cancers. Nat. Med. 21, 938–945 (2015).

    Article  CAS  Google Scholar 

  45. Cohen, N. R. et al. Shared and distinct transcriptional programs underlie the hybrid nature of iNKT cells. Nat. Immunol. 14, 90–99 (2013).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank M. Wilk and J. Barrett for assistance with experiments. This research was supported by National Institutes of Health (NIH) grant R01 AI11304603 (M.B.B.), European Research Council (ERC) Starting Grant 679173, a Cancer Research Institute CLIP grant and 16/FRL/3865 (L.L.).

Author information

Authors and Affiliations

Authors

Contributions

X.M., L.D., and L.L. conceived and designed the experiments, and wrote the manuscript. X.M., L.D., A.H., R.M.L., D.D., K.W., R.D., M.R., and L.L. performed the experiments. C.F. performed the RNA-seq analysis. A.T., A.V., W.P., D.O.’S., and B.S.N. obtained patient samples and coordinated the clinical investigations. S.B., C.O.F., K.H.G.M., M.B.B., and D.F. provided advice, reagents and critical insight.

Corresponding author

Correspondence to Lydia Lynch.

Ethics declarations

Competing interests

The authors declare no competing interests.

Additional information

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

Supplementary information

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Michelet, X., Dyck, L., Hogan, A. et al. Metabolic reprogramming of natural killer cells in obesity limits antitumor responses. Nat Immunol 19, 1330–1340 (2018). https://doi.org/10.1038/s41590-018-0251-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41590-018-0251-7

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