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:

CaMKII is a RIP3 substrate mediating ischemia- and oxidative stress–induced myocardial necroptosis

Abstract

Regulated necrosis (necroptosis) and apoptosis are crucially involved in severe cardiac pathological conditions, including myocardial infarction, ischemia-reperfusion injury and heart failure. Whereas apoptotic signaling is well defined, the mechanisms that underlie cardiomyocyte necroptosis remain elusive. Here we show that receptor-interacting protein 3 (RIP3) triggers myocardial necroptosis, in addition to apoptosis and inflammation, through activation of Ca2+-calmodulin–dependent protein kinase (CaMKII) rather than through the well-established RIP3 partners RIP1 and MLKL. In mice, RIP3 deficiency or CaMKII inhibition ameliorates myocardial necroptosis and heart failure induced by ischemia-reperfusion or by doxorubicin treatment. RIP3-induced activation of CaMKII, via phosphorylation or oxidation or both, triggers opening of the mitochondrial permeability transition pore and myocardial necroptosis. These findings identify CaMKII as a new RIP3 substrate and delineate a RIP3-CaMKII-mPTP myocardial necroptosis pathway, a promising target for the treatment of ischemia- and oxidative stress–induced myocardial damage and heart failure.

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

Figure 1: RIP3 deficiency protects mouse hearts from I/R-induced necrosis and heart failure.
Figure 2: Dox-induced myocardial necrosis, cardiomyopathy and mortality are alleviated in Ripk3−/− mice.
Figure 3: RIP3-induced cardiomyocyte necroptosis is independent of RIP1 and MLKL.
Figure 4: RIP3-induced CaMKII activation is essential for cardiomyocyte necroptosis.
Figure 5: CaMKII is activated by RIP3 via both phosphorylation and oxidation.
Figure 6: RIP3-mediated cardiomyocyte apoptosis and necroptosis contribute to I/R-induced myocardial injury.

Similar content being viewed by others

References

  1. Kung, G., Konstantinidis, K. & Kitsis, R.N. Programmed necrosis, not apoptosis, in the heart. Circ. Res. 108, 1017–1036 (2011).

    CAS  PubMed  Google Scholar 

  2. Whelan, R.S., Kaplinskiy, V. & Kitsis, R.N. Cell death in the pathogenesis of heart disease: mechanisms and significance. Annu. Rev. Physiol. 72, 19–44 (2010).

    CAS  PubMed  Google Scholar 

  3. Cho, Y.S. et al. Phosphorylation-driven assembly of the RIP1-RIP3 complex regulates programmed necrosis and virus-induced inflammation. Cell 137, 1112–1123 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  4. He, S. et al. Receptor interacting protein kinase-3 determines cellular necrotic response to TNF-α. Cell 137, 1100–1111 (2009).

    CAS  PubMed  Google Scholar 

  5. Zhang, D.W. et al. RIP3, an energy metabolism regulator that switches TNF-induced cell death from apoptosis to necrosis. Science 325, 332–336 (2009).

    CAS  PubMed  Google Scholar 

  6. Kaiser, W.J. et al. RIP3 mediates the embryonic lethality of caspase-8-deficient mice. Nature 471, 368–372 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  7. Oberst, A. et al. Catalytic activity of the caspase-8-FLIP(L) complex inhibits RIPK3-dependent necrosis. Nature 471, 363–367 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  8. Upton, J.W., Kaiser, W.J. & Mocarski, E.S. Virus inhibition of RIP3-dependent necrosis. Cell Host Microbe 7, 302–313 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  9. Upton, J.W., Kaiser, W.J. & Mocarski, E.S. DAI/ZBP1/DLM-1 complexes with RIP3 to mediate virus-induced programmed necrosis that is targeted by murine cytomegalovirus vIRA. Cell Host Microbe 11, 290–297 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  10. Roychowdhury, S., McMullen, M.R., Pisano, S.G., Liu, X. & Nagy, L.E. Absence of receptor interacting protein kinase 3 prevents ethanol-induced liver injury. Hepatology 57, 1773–1783 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  11. Sun, L. et al. Mixed lineage kinase domain-like protein mediates necrosis signaling downstream of RIP3 kinase. Cell 148, 213–227 (2012).

    CAS  PubMed  Google Scholar 

  12. Welz, P.S. et al. FADD prevents RIP3-mediated epithelial cell necrosis and chronic intestinal inflammation. Nature 477, 330–334 (2011).

    CAS  PubMed  Google Scholar 

  13. Luedde, M. et al. RIP3, a kinase promoting necroptotic cell death, mediates adverse remodelling after myocardial infarction. Cardiovasc. Res. 103, 206–216 (2014).

    CAS  PubMed  Google Scholar 

  14. Zhao, J. et al. Mixed lineage kinase domain-like is a key receptor interacting protein 3 downstream component of TNF-induced necrosis. Proc. Natl. Acad. Sci. USA 109, 5322–5327 (2012).

    CAS  PubMed  Google Scholar 

  15. Humphries, F., Yang, S., Wang, B. & Moynagh, P.N. RIP kinases: key decision makers in cell death and innate immunity. Cell Death Differ. 22, 225–236 (2015).

    CAS  PubMed  Google Scholar 

  16. Wu, J. et al. Mlkl knockout mice demonstrate the indispensable role of Mlkl in necroptosis. Cell Res. 23, 994–1006 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  17. Nakayama, H. et al. Ca2+- and mitochondrial-dependent cardiomyocyte necrosis as a primary mediator of heart failure. J. Clin. Invest. 117, 2431–2444 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  18. Ferreira, A.L., Matsubara, L.S. & Matsubara, B.B. Anthracycline-induced cardiotoxicity. Cardiovasc. Hematol. Agents Med. Chem. 6, 278–281 (2008).

    CAS  PubMed  Google Scholar 

  19. Singal, P.K. & Iliskovic, N. Doxorubicin-induced cardiomyopathy. N. Engl. J. Med. 339, 900–905 (1998).

    CAS  PubMed  Google Scholar 

  20. Yeh, E.T. & Bickford, C.L. Cardiovascular complications of cancer therapy: incidence, pathogenesis, diagnosis, and management. J. Am. Coll. Cardiol. 53, 2231–2247 (2009).

    CAS  PubMed  Google Scholar 

  21. Gewirtz, D.A. A critical evaluation of the mechanisms of action proposed for the antitumor effects of the anthracycline antibiotics adriamycin and daunorubicin. Biochem. Pharmacol. 57, 727–741 (1999).

    CAS  PubMed  Google Scholar 

  22. Minotti, G., Menna, P., Salvatorelli, E., Cairo, G. & Gianni, L. Anthracyclines: molecular advances and pharmacologic developments in antitumor activity and cardiotoxicity. Pharmacol. Rev. 56, 185–229 (2004).

    CAS  PubMed  Google Scholar 

  23. Degterev, A. et al. Identification of RIP1 kinase as a specific cellular target of necrostatins. Nat. Chem. Biol. 4, 313–321 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  24. Smith, C.C. et al. Necrostatin: a potentially novel cardioprotective agent? Cardiovasc. Drugs Ther. 21, 227–233 (2007).

    CAS  PubMed  Google Scholar 

  25. Lim, S.Y., Davidson, S.M., Mocanu, M.M., Yellon, D.M. & Smith, C.C. The cardioprotective effect of necrostatin requires the cyclophilin-D component of the mitochondrial permeability transition pore. Cardiovasc. Drugs Ther. 21, 467–469 (2007).

    CAS  PubMed  PubMed Central  Google Scholar 

  26. Oerlemans, M.I. et al. Inhibition of RIP1-dependent necrosis prevents adverse cardiac remodeling after myocardial ischemia-reperfusion in vivo. Basic Res. Cardiol. 107, 270 (2012).

    PubMed  Google Scholar 

  27. Wang, H. et al. Mixed lineage kinase domain-like protein MLKL causes necrotic membrane disruption upon phosphorylation by RIP3. Mol. Cell 54, 133–146 (2014).

    CAS  PubMed  Google Scholar 

  28. Vila-Petroff, M. et al. CaMKII inhibition protects against necrosis and apoptosis in irreversible ischemia-reperfusion injury. Cardiovasc. Res. 73, 689–698 (2007).

    CAS  PubMed  Google Scholar 

  29. Ling, H. et al. Ca2+/calmodulin-dependent protein kinase II δ mediates myocardial ischemia/reperfusion injury through nuclear factor-κB. Circ. Res. 112, 935–944 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  30. Joiner, M.L. et al. CaMKII determines mitochondrial stress responses in heart. Nature 491, 269–273 (2012).

    CAS  PubMed  PubMed Central  Google Scholar 

  31. He, B.J. et al. Oxidation of CaMKII determines the cardiotoxic effects of aldosterone. Nat. Med. 17, 1610–1618 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  32. Zhang, R. et al. Calmodulin kinase II inhibition protects against structural heart disease. Nat. Med. 11, 409–417 (2005).

    CAS  PubMed  Google Scholar 

  33. Zhu, W. et al. Activation of CaMKIIδC is a common intermediate of diverse death stimuli-induced heart muscle cell apoptosis. J. Biol. Chem. 282, 10833–10839 (2007).

    CAS  PubMed  Google Scholar 

  34. Zhu, W.Z. et al. Linkage of β1-adrenergic stimulation to apoptotic heart cell death through protein kinase A-independent activation of Ca2+/calmodulin kinase II. J. Clin. Invest. 111, 617–625 (2003).

    CAS  PubMed  PubMed Central  Google Scholar 

  35. Zhang, T. et al. The δC isoform of CaMKII is activated in cardiac hypertrophy and induces dilated cardiomyopathy and heart failure. Circ. Res. 92, 912–919 (2003).

    CAS  PubMed  Google Scholar 

  36. Ling, H. et al. Requirement for Ca2+/calmodulin-dependent kinase II in the transition from pressure overload-induced cardiac hypertrophy to heart failure in mice. J. Clin. Invest. 119, 1230–1240 (2009).

    CAS  PubMed  PubMed Central  Google Scholar 

  37. Erickson, J.R., He, B.J., Grumbach, I.M. & Anderson, M.E. CaMKII in the cardiovascular system: sensing redox states. Physiol. Rev. 91, 889–915 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

  38. Baines, C.P. et al. Loss of cyclophilin D reveals a critical role for mitochondrial permeability transition in cell death. Nature 434, 658–662 (2005).

    CAS  PubMed  Google Scholar 

  39. Nakagawa, T. et al. Cyclophilin D-dependent mitochondrial permeability transition regulates some necrotic but not apoptotic cell death. Nature 434, 652–658 (2005).

    CAS  PubMed  Google Scholar 

  40. Brenner, C. & Moulin, M. Physiological roles of the permeability transition pore. Circ. Res. 111, 1237–1247 (2012).

    CAS  PubMed  Google Scholar 

  41. Kroemer, G., Galluzzi, L. & Brenner, C. Mitochondrial membrane permeabilization in cell death. Physiol. Rev. 87, 99–163 (2007).

    CAS  PubMed  Google Scholar 

  42. He, S., Liang, Y., Shao, F. & Wang, X. Toll-like receptors activate programmed necrosis in macrophages through a receptor-interacting kinase-3-mediated pathway. Proc. Natl. Acad. Sci. USA 108, 20054–20059 (2011).

    CAS  PubMed  Google Scholar 

  43. Erickson, J.R. et al. A dynamic pathway for calcium-independent activation of CaMKII by methionine oxidation. Cell 133, 462–474 (2008).

    CAS  PubMed  PubMed Central  Google Scholar 

  44. Li, Q. et al. Receptor interacting protein 3 suppresses vascular smooth muscle cell growth by inhibition of the phosphoinositide 3-kinase-Akt axis. J. Biol. Chem. 285, 9535–9544 (2010).

    CAS  PubMed  Google Scholar 

  45. Sun, X. et al. RIP3, a novel apoptosis-inducing kinase. J. Biol. Chem. 274, 16871–16875 (1999).

    CAS  PubMed  Google Scholar 

  46. Coupienne, I., Fettweis, G. & Piette, J. RIP3 expression induces a death profile change in U2OS osteosarcoma cells after 5-ALA-PDT. Lasers Surg. Med. 43, 557–564 (2011).

    PubMed  Google Scholar 

  47. Pazdernik, N.J., Donner, D.B., Goebl, M.G. & Harrington, M.A. Mouse receptor interacting protein 3 does not contain a caspase-recruiting or a death domain but induces apoptosis and activates NF-κB. Mol. Cell. Biol. 19, 6500–6508 (1999).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Moriwaki, K. & Chan, F.K. RIP3: a molecular switch for necrosis and inflammation. Genes Dev. 27, 1640–1649 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  49. Montaigne, D. et al. Doxorubicin induces mitochondrial permeability transition and contractile dysfunction in the human myocardium. Mitochondrion 11, 22–26 (2011).

    CAS  PubMed  Google Scholar 

  50. Montaigne, D. et al. Stabilization of mitochondrial membrane potential prevents doxorubicin-induced cardiotoxicity in isolated rat heart. Toxicol. Appl. Pharmacol. 244, 300–307 (2010).

    CAS  PubMed  Google Scholar 

  51. Salas, M.A. et al. The signalling pathway of CaMKII-mediated apoptosis and necrosis in the ischemia/reperfusion injury. J. Mol. Cell. Cardiol. 48, 1298–1306 (2010).

    CAS  PubMed  PubMed Central  Google Scholar 

  52. Buja, L.M. Myocardial ischemia and reperfusion injury. Cardiovasc. Pathol. 14, 170–175 (2005).

    CAS  PubMed  Google Scholar 

  53. Hausenloy, D.J. & Yellon, D.M. Myocardial ischemia-reperfusion injury: a neglected therapeutic target. J. Clin. Invest. 123, 92–100 (2013).

    CAS  PubMed  PubMed Central  Google Scholar 

  54. Cao, C.M. et al. MG53 constitutes a primary determinant of cardiac ischemic preconditioning. Circulation 121, 2565–2574 (2010).

    CAS  PubMed  Google Scholar 

  55. Peng, W. et al. Cardioprotection by CaMKII-δB is mediated by phosphorylation of heat shock factor 1 and subsequent expression of inducible heat shock protein 70. Circ. Res. 106, 102–110 (2010).

    CAS  PubMed  Google Scholar 

Download references

Acknowledgements

We thank X.-D. Wang for Ripk3−/− mice, M. Anderson for CaMKII plasmids, and H. Cheng and I.C. Bruce for comments on the manuscript. This work was supported by the National Basic Research Program of China (2013CB531200 (Y.Z.), 2012CB518000 (R.-P.X.) and 2012CB944500 (C.-M.C.)), the National Natural Science Foundation of China (81170100 (Y.Z.), 81370234 (Y.Z.), 81130073 (R.-P.X.), 81370233 (C.-M.C.), 91439107 (C.-M.C.), 81270190 (F.L.) and 31221002 (R.-P.X.)), National Science and Technology Major Projects for Major New Drugs Innovation and Development (2013ZX09508104) (R.-P.X.) and the Specialized Research Fund for the Doctoral Program of Higher Education (20110001120119) (Y.Z.).

Author information

Authors and Affiliations

Authors

Contributions

T.Z. and Y.Z. generated the initial idea and conducted key experiments. Y.Z. and R.-P.X. proposed the hypothesis, designed the study, supervised the experiments and data analysis, wrote the manuscript and interpreted results. T.Z., Y.Z., M.C., L.J., Y.W., F.L., Y.L., W.Z., H.S., J.Z., M.Z., H.W., J.G., X.Z., X.H. and C.-M.C. performed the experiments.

Corresponding authors

Correspondence to Yan Zhang or Rui-Ping Xiao.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1–14 and Supplementary Table 1 (PDF 1431 kb)

Dataset 1

Source Data for Supplementary Figures (XLS 156 kb)

Source data

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, T., Zhang, Y., Cui, M. et al. CaMKII is a RIP3 substrate mediating ischemia- and oxidative stress–induced myocardial necroptosis. Nat Med 22, 175–182 (2016). https://doi.org/10.1038/nm.4017

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nm.4017

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