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:

Redox lipid reprogramming commands susceptibility of macrophages and microglia to ferroptotic death

Abstract

Ferroptotic death is the penalty for losing control over three processes—iron metabolism, lipid peroxidation and thiol regulation—that are common in the pro-inflammatory environment where professional phagocytes fulfill their functions and yet survive. We hypothesized that redox reprogramming of 15-lipoxygenase (15-LOX) during the generation of pro-ferroptotic signal 15-hydroperoxy-eicosa-tetra-enoyl-phosphatidylethanolamine (15-HpETE-PE) modulates ferroptotic endurance. Here, we have discovered that inducible nitric oxide synthase (iNOS)/NO-enrichment of activated M1 (but not alternatively activated M2) macrophages/microglia modulates susceptibility to ferroptosis. Genetic or pharmacologic depletion/inactivation of iNOS confers sensitivity on M1 cells, whereas NO donors empower resistance of M2 cells to ferroptosis. In vivo, M1 phagocytes, in comparison to M2 phagocytes, exert higher resistance to pharmacologically induced ferroptosis. This resistance is diminished in iNOS-deficient cells in the pro-inflammatory conditions of brain trauma or the tumour microenvironment. The nitroxygenation of eicosatetraenoyl (ETE)-PE intermediates and oxidatively truncated species by NO donors and/or suppression of NO production by iNOS inhibitors represent a novel redox mechanism of regulation of ferroptosis in pro-inflammatory conditions.

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: Differential sensitivity of activated M1 and alternatively activated M2 macrophages and microglia to RSL3-induced ferroptosis.
Fig. 2: Sensitivity of activated (M1) macrophages and microglial cells to RSL3-induced ferroptosis depends on the levels of iNOS expression.
Fig. 3: NO protects cells against RSL3-induced ferroptosis.
Fig. 4: NO suppresses RSL3-induced accumulation of oxidatively modified PE species in alternatively activated (M2) RAW 264.7 macrophages.
Fig. 5: iNOS/NO-driven mechanisms of ferroptosis regulation in vivo.
Fig. 6: NO/O2 interactions with 15-LOX-2 and entry/exit pathways observed in MD simulations of 15-LOX-2 and the 15-LOX-2–PEBP1 complex.

Similar content being viewed by others

Data availability

The raw data are available at the following link https://data.mendeley.com/datasets/t4jyhhf3gw/draft?a=76c6ac56-4305-463e-bebc-b004c2000edb.

Code used for the analysis of the MD simulations of NO interactions with 15-LOX has been made available in two formats: (1) Jupyter Notebook and (2) html (https://onedrive.live.com/?authkey=%21AFEVmP5sOP1km0s&id=4960CA1B5C7F3FD%219568&cid=04960CA1B5C7F3FD).

References

  1. Stockwell, B. R. et al. Ferroptosis: a regulated cell death nexus linking metabolism, redox biology, and disease. Cell 171, 273–285 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  2. Dixon, S. J. et al. Ferroptosis: an iron-dependent form of nonapoptotic cell death. Cell 149, 1060–1072 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  3. Friedmann Angeli, J. P. et al. Inactivation of the ferroptosis regulator Gpx4 triggers acute renal failure in mice. Nat. Cell Biol. 16, 1180–1191 (2014).

    CAS  PubMed  Google Scholar 

  4. Stoyanovsky, D. A. et al. Iron catalysis of lipid peroxidation in ferroptosis: regulated enzymatic or random free radical reaction? Free Radic. Biol. Med. 133, 153–161 (2018).

    PubMed  PubMed Central  Google Scholar 

  5. Gugiu, B. G. et al. Identification of oxidatively truncated ethanolamine phospholipids in retina and their generation from polyunsaturated phosphatidylethanolamines. Chem. Res. Toxicol. 19, 262–271 (2006).

    CAS  PubMed  Google Scholar 

  6. Pizzimenti, S. et al. Interaction of aldehydes derived from lipid peroxidation and membrane proteins. Front. Physiol. 4, 242 (2013).

    PubMed  PubMed Central  Google Scholar 

  7. Hoff, H. F., O’Neil, J., Wu, Z., Hoppe, G. & Salomon, R. L. Phospholipid hydroxyalkenals: biological and chemical properties of specific oxidized lipids present in atherosclerotic lesions. Arterioscler. Thromb. Vasc. Biol. 23, 275–282 (2003).

    CAS  PubMed  Google Scholar 

  8. Doll, S. et al. ACSL4 dictates ferroptosis sensitivity by shaping cellular lipid composition. Nat. Chem. Biol. 13, 91–98 (2017).

    CAS  PubMed  Google Scholar 

  9. Wenzel, S. E. et al. PEBP1 wardens ferroptosis by enabling lipoxygenase generation of lipid death signals. Cell 171, 628–641 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Kagan, V. E. et al. Oxidized arachidonic and adrenic PEs navigate cells to ferroptosis. Nat. Chem. Biol. 13, 81–90 (2017).

    PubMed  Google Scholar 

  11. Conrad, M. et al. Regulation of lipid peroxidation and ferroptosis in diverse species. Genes Dev. 32, 602–619 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  12. Matsushita, M. et al. T cell lipid peroxidation induces ferroptosis and prevents immunity to infection. J. Exp. Med. 212, 555–568 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  13. Feng, H. & Stockwell, B. R. Unsolved mysteries: how does lipid peroxidation cause ferroptosis? PLoS Biol. 16, e2006203 (2018).

    PubMed  PubMed Central  Google Scholar 

  14. Murray, P. J. Macrophage polarization. Annu. Rev. Physiol. 79, 541–566 (2017).

    CAS  PubMed  Google Scholar 

  15. O’Donnell, V. B. et al. 15-Lipoxygenase catalytically consumes nitric oxide and impairs activation of guanylate cyclase. J. Biol. Chem. 274, 20083–20091 (1999).

    PubMed  Google Scholar 

  16. Lorsbach, R. B., Murphy, W. J., Lowenstein, C. J., Snyder, S. H. & Russell, S. W. Expression of the nitric oxide synthase gene in mouse macrophages activated for tumor cell killing. Molecular basis for the synergy between interferon-gamma and lipopolysaccharide. J. Biol. Chem. 268, 1908–1913 (1993).

    CAS  PubMed  Google Scholar 

  17. Gao, M., Monian, P., Quadri, N., Ramasamy, R. & Jiang, X. Glutaminolysis and transferrin regulate ferroptosis. Mol. Cell 59, 298–308 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Corna, G. et al. Polarization dictates iron handling by inflammatory and alternatively activated macrophages. Haematologica 95, 1814–1822 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  19. Zhang, Y. et al. Imidazole ketone erastin induces ferroptosis and slows tumor growth in a mouse lymphoma model. Cell Chem. Biol. 26, 623–633 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  20. Borland, C. et al. Permeability and diffusivity of nitric oxide in human plasma and red cells. Nitric Oxide 78, 51–59 (2018).

    CAS  PubMed  Google Scholar 

  21. Dar, H. H. et al. Pseudomonas aeruginosa utilizes host polyunsaturated phosphatidylethanolamines to trigger theft-ferroptosis in bronchial epithelium. J. Clin. Invest. 128, 4639–4653 (2018).

    PubMed  PubMed Central  Google Scholar 

  22. Yamanaka, K. et al. A novel fluorescent probe with high sensitivity and selective detection of lipid hydroperoxides in cells. RSC Adv. 2, 7894–7900 (2012).

    CAS  Google Scholar 

  23. Shah, R., Shchepinov, M. S. & Pratt, D. A. Resolving the role of lipoxygenases in the initiation and execution of ferroptosis. ACS Cent. Sci. 4, 387–396 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Monroe, L. L. et al. Zymosan-induced peritonitis: effects on cardiac function, temperature regulation, translocation of bacteria, and role of dectin-1. Shock 46, 723–730 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  25. Kenny, E. M. et al. Ferroptosis contributes to neuronal death and functional outcome after traumatic brain injury. Crit. Care Med. 47, 410–418 (2018).

    Google Scholar 

  26. Bayir, H. et al. Enhanced oxidative stress in iNOS-deficient mice after traumatic brain injury: support for a neuroprotective role of iNOS. J. Cereb. Blood Flow Metab. 25, 673–684 (2005).

    CAS  PubMed  Google Scholar 

  27. Loane, D. J. & Kumar, A. Microglia in the TBI brain: the good, the bad, and the dysregulated. Exp. Neurol. 275, 316–327 (2016).

    CAS  PubMed  Google Scholar 

  28. Bayir, H. et al. Neuronal NOS-mediated nitration and inactivation of manganese superoxide dismutase in brain after experimental and human brain injury. J. Neurochem. 101, 168–181 (2007).

    CAS  PubMed  Google Scholar 

  29. Foley, L. M. et al. Magnetic resonance imaging assessment of macrophage accumulation in mouse brain after experimental traumatic brain injury. J. Neurotrauma 26, 1509–1519 (2009).

    PubMed  PubMed Central  Google Scholar 

  30. Brune, B. et al. Redox control of inflammation in macrophages. Antioxid. Redox Signal. 19, 595–637 (2013).

    PubMed  PubMed Central  Google Scholar 

  31. Ivanov, I., Kuhn, H. & Heydeck, D. Structural and functional biology of arachidonic acid 15-lipoxygenase-1 (ALOX15). Gene 573, 1–32 (2015).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Lawrence, T. & Natoli, G. Transcriptional regulation of macrophage polarization: enabling diversity with identity. Nat. Rev. Immunol. 11, 750–761 (2011).

    CAS  PubMed  Google Scholar 

  33. Mantovani, A., Marchesi, F., Malesci, A., Laghi, L. & Allavena, P. Tumour-associated macrophages as treatment targets in oncology. Nat. Rev. Clin. Oncol. 14, 399–416 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Murray, P. J. et al. Macrophage activation and polarization: nomenclature and experimental guidelines. Immunity 41, 14–20 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Kanazawa, M., Ninomiya, I., Hatakeyama, M., Takahashi, T. & Shimohata, T. Microglia and monocytes/macrophages polarization reveal novel therapeutic mechanism against stroke. Int. J. Mol. Sci. 18, 2135 (2017).

    PubMed Central  Google Scholar 

  36. Anthonymuthu, T. S. et al. Empowerment of 15-lipoxygenase catalytic competence in selective oxidation of membrane ETE-PE to ferroptotic death signals, HpETE-PE. J. Am. Chem. Soc. 140, 17835–17839 (2018).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Zilka, O. et al. On the mechanism of cytoprotection by ferrostatin-1 and liproxstatin-1 and the role of lipid peroxidation in ferroptotic cell death. ACS Cent. Sci. 3, 232–243 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Cao, J. Y. & Dixon, S. J. Mechanisms of ferroptosis. Cell. Mol. Life Sci. 73, 2195–2209 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  39. Rubbo, H. et al. Nitric oxide inhibition of lipoxygenase-dependent liposome and low-density lipoprotein oxidation: termination of radical chain propagation reactions and formation of nitrogen-containing oxidized lipid derivatives. Arch. Biochem. Biophys. 324, 15–25 (1995).

    CAS  PubMed  Google Scholar 

  40. Saam, J., Ivanov, I., Walther, M., Holzhutter, H. G. & Kuhn, H. Molecular dioxygen enters the active site of 12/15-lipoxygenase via dynamic oxygen access channels. Proc. Natl Acad. Sci. USA 104, 13319–13324 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  41. Rubbo, H. et al. Nitric oxide regulation of superoxide and peroxynitrite-dependent lipid peroxidation. Formation of novel nitrogen-containing oxidized lipid derivatives. J. Biol. Chem. 269, 26066–26075 (1994).

    CAS  PubMed  Google Scholar 

  42. O’Donnell, V. B. et al. Nitric oxide inhibition of lipid peroxidation: kinetics of reaction with lipid peroxyl radicals and comparison with ɑ-tocopherol. Biochemistry 36, 15216–15223 (1997).

    PubMed  Google Scholar 

  43. Napoli, C. et al. Effects of nitric oxide on cell proliferation: novel insights. J. Am. Coll. Cardiol. 62, 89–95 (2013).

    CAS  PubMed  Google Scholar 

  44. Thomas, D. D. et al. The chemical biology of nitric oxide: implications in cellular signaling. Free Radic. Biol. Med. 45, 18–31 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  45. Perrotta, C. et al. Nitric oxide generated by tumor-associated macrophages is responsible for cancer resistance to cisplatin and correlated with syntaxin 4 and acid sphingomyelinase inhibition. Front. Immunol. 9, 1186 (2018).

    PubMed  PubMed Central  Google Scholar 

  46. Xie, B. S. et al. Inhibition of ferroptosis attenuates tissue damage and improves long-term outcomes after traumatic brain injury in mice. CNS Neurosci. Ther. 25, 465–475 (2019).

    CAS  PubMed  Google Scholar 

  47. Amaral, E. P. et al. A major role for ferroptosis in Mycobacterium tuberculosis-induced cell death and tissue necrosis. J. Exp. Med. 216, 556–570 (2019).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Martin-Sanchez, D. et al. Ferroptosis, but not necroptosis, is important in nephrotoxic folic acid-induced AKI. J. Am. Soc. Nephrol. 28, 218–229 (2017).

    CAS  PubMed  Google Scholar 

  49. Uderhardt, S. et al. 12/15-lipoxygenase orchestrates the clearance of apoptotic cells and maintains immunologic tolerance. Immunity 36, 834–846 (2012).

    CAS  PubMed  Google Scholar 

  50. Wu, Y. L. et al. In situ labeling of immune cells with iron oxide particles: an approach to detect organ rejection by cellular MRI. Proc. Natl Acad. Sci. USA 103, 1852–1857 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  51. RayA. & DittelB. N. Isolation of mouse peritoneal cavity cells. J. Vis. Exp. 35, e1488 (2010).

    Google Scholar 

  52. Lian, H., Roy, E. & Zheng, H. Protocol for primary microglial culture preparation. Bio Protoc 6, e1989 (2016).

    PubMed  Google Scholar 

  53. Elmore, M. R. et al. Colony-stimulating factor 1 receptor signaling is necessary for microglia viability, unmasking a microglia progenitor cell in the adult brain. Neuron 82, 380–397 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Weischenfeldt, J. & Porse, B. Bone marrow-derived macrophages (BMM): isolation and applications. CSH Protoc. 2008, pdb.prot5080 (2008).

    PubMed  Google Scholar 

  55. Harris, L. A. et al. BioNetGen 2.2: advances in rule-based modeling. Bioinformatics 32, 3366–3368 (2016).

    CAS  PubMed  PubMed Central  Google Scholar 

  56. Liu, B., Gyori, B. & Thiagarajan, P. In Automated Reasoning for Systems Biology and Medicine (eds. Liò, P. & Zuliani, P.) 63–92 (Springer International Publishing, Cham; 2019).

  57. Phillips, J. C. et al. Scalable molecular dynamics with NAMD. J. Comput. Chem. 26, 1781–1802 (2005).

    CAS  PubMed  PubMed Central  Google Scholar 

  58. Tovchigrechko, A. & Vakser, I. A. GRAMM-X public web server for protein–protein docking. Nucleic Acids Res. 34, W310–W314 (2006).

    CAS  PubMed  PubMed Central  Google Scholar 

  59. Frisch, M. et al. Gaussian 03, Revision B.05 (Gaussian, 2003).

  60. Bakan, A. et al. Evol and ProDy for bridging protein sequence evolution and structural dynamics. Bioinformatics 30, 2681–2683 (2014).

    CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This work was supported by NIH (HL114453-06, U19AI068021, CA165065-06, NS076511, NS061817, P41GM103712) and by Russian academic excellence project ‘5-100’.

Author information

Authors and Affiliations

Authors

Contributions

V.E.K. and H.B. conceived the study. A.A.K., Q.Y., H.H.D., G.V.S., H.-C.T., M.A.A. and L.A.P. performed experiments with cells. V.E.K., H.B. and D.I.G. designed in vivo experiments. Q.Y., Y.L.W., R.K. and Y.G. performed in vivo experiments. A.A.K., Q.Y. and H.H.D. analyzed the data. Y.Y.T., T.S.A. and V.A.T. performed MS measurements and analyzed data. Y.Y.T., T.S.A. and R.M.D. discussed and interpreted MS results. K.M.-R., B.L. and I.H.S. performed computational modelling. I.B. supervised computational studies. C.M.S.C. performed imaging experiments and participated in interpreting them. H.B., D.A.S., R.K.M. and D.I.G. participated in formulating the idea and interpreting the data. P.S.T. and J.S.G. participated in the discussion and helped in writing the manuscript. Y.Y.T., I.B. and H.B. participated in writing the manuscript. H.B. and V.E.K. wrote the manuscript.

Corresponding authors

Correspondence to Dmitry I. Gabrilovich, Hülya Bayır or Valerian E. Kagan.

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

Supplementary Information

Supplementary Figs. 1–11.

Reporting Summary

Supplementary Tables 1 and 2

Supplementary Tables 1 and 2.

Supplementary Video 1

Fragment of MD simulations of competitive binding of NO and O2 molecules to 15-LOX-2.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kapralov, A.A., Yang, Q., Dar, H.H. et al. Redox lipid reprogramming commands susceptibility of macrophages and microglia to ferroptotic death. Nat Chem Biol 16, 278–290 (2020). https://doi.org/10.1038/s41589-019-0462-8

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41589-019-0462-8

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