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:

Plasma membrane translocation of trimerized MLKL protein is required for TNF-induced necroptosis

An Erratum to this article was published on 31 January 2014

This article has been updated

Abstract

The mixed lineage kinase domain-like protein (MLKL) has recently been identified as a key RIP3 (receptor interacting protein 3) downstream component of tumour necrosis factor (TNF)-induced necroptosis. MLKL is phosphorylated by RIP3 and is recruited to the necrosome through its interaction with RIP3. However, it is still unknown how MLKL mediates TNF-induced necroptosis. Here, we report that MLKL forms a homotrimer through its amino-terminal coiled-coil domain and locates to the cell plasma membrane during TNF-induced necroptosis. By generating different MLKL mutants, we demonstrated that the plasma membrane localization of trimerized MLKL is critical for mediating necroptosis. Importantly, we found that the membrane localization of MLKL is essential for Ca2+ influx, which is an early event of TNF-induced necroptosis. Furthermore, we identified that TRPM7 (transient receptor potential melastatin related 7) is a MLKL downstream target for the mediation of Ca2+ influx and TNF-induced necroptosis. Hence, our study reveals a crucial mechanism of MLKL-mediated TNF-induced necroptosis.

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: MLKL forms homotrimeric protein on necrosis induction.
Figure 2: RIP3 kinase activity is critical for MLKL trimerization
Figure 3: The coiled-coil domain 2 of MLKL is responsible for its trimerization.
Figure 4: MLKL locates to plasma membrane following necroptosis induction.
Figure 5: Plasma membrane translocation of trimerized MLKL in response to necroptosis induction.
Figure 6: MLKL-mediated calcium influx is involved in plasma membrane rupture during necroptosis.
Figure 7: Calcium channels are involved in necroptosis.
Figure 8: TRPM7 is involved in MLKL-mediated necroptosis.

Similar content being viewed by others

Change history

  • 16 December 2013

    In the version of this article originally published the 'Anti-FLAG' label at the top-right of the right panel in Fig. 1b should have read 'Anti-GFP'. This error has now been corrected in the online versions of the Article.

References

  1. Chen, G. & Goeddel, D. V. TNF-R1 signaling: a beautiful pathway. Science 296, 1634–1635 (2002).

    Article  CAS  PubMed  Google Scholar 

  2. Ashkenazi, A. & Dixit, V. M. Apoptosis control by death and decoy receptors. Curr. Opin. Cell Biol. 11, 255–260 (1999).

    Article  CAS  PubMed  Google Scholar 

  3. Grivennikov, S. I., Greten, F. R. & Karin, M. Immunity, inflammation, and cancer. Cell 140, 883–899 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Vandenabeele, P., Galluzzi, L., Vanden Berghe, T. & Kroemer, G. Molecular mechanisms of necroptosis: an ordered cellular explosion. Nat. Rev. Mol. Cell Biol. 11, 700–714 (2010).

    Article  CAS  PubMed  Google Scholar 

  5. Christofferson, D. E. & Yuan, J. Necroptosis as an alternative form of programmed cell death. Curr. Opin. Cell Biol. 22, 263–268 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Green, D. R., Oberst, A., Dillon, C. P., Weinlich, R. & Salvesen, G.S. RIPK-dependent necrosis and its regulation by caspases: a mystery in five acts. Mol. Cell 44, 9–16 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  9. 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).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  11. 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).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. O’Donnell, M. A. et al. Caspase 8 inhibits programmed necrosis by processing CYLD. Nat. Cell Biol. 13, 1437–1442 (2011).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Narayan, N. et al. The NAD-dependent deacetylase SIRT2 is required for programmed necrosis. Nature 492, 199–204 (2012).

    Article  CAS  PubMed  Google Scholar 

  14. Mocarski, E. S., Upton, J. W. & Kaiser, W. J. Viral infection and the evolution of caspase 8-regulated apoptotic and necrotic death pathways. Nat. Rev. Immunol. 12, 79–88 (2012).

    Article  CAS  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  16. Zhang, H. et al. Functional complementation between FADD and RIP1 in embryos and lymphocytes. Nature 471, 373–376 (2011).

    CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Lin, Y. et al. Tumor necrosis factor-induced nonapoptotic cell death requires receptor-interacting protein-mediated cellular reactive oxygen species accumulation. J. Biol. Chem. 279, 10822–10828 (2004).

    Article  CAS  PubMed  Google Scholar 

  19. Zong, W. X. & Thompson, C. B. Necrotic death as a cell fate. Genes Dev. 20, 1–15 (2006).

    Article  CAS  PubMed  Google Scholar 

  20. Wang, Z., Jiang, H., Chen, S., Du, F. & Wang, X. The mitochondrial phosphatase PGAM5 functions at the convergence point of multiple necrotic death pathways. Cell 148, 228–243 (2012).

    Article  CAS  PubMed  Google Scholar 

  21. Wolf, E., Kim, P. S. & Berger, B. MultiCoil: a program for predicting two- and three-stranded coiled coils. Protein Sci. 6, 1179–1189 (1997).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Degterev, A. et al. Chemical inhibitor of nonapoptotic cell death with therapeutic potential for ischemic brain injury. Nat. Chem. Biol. 1, 112–119 (2005).

    Article  CAS  PubMed  Google Scholar 

  23. Li, J. et al. The RIP1/RIP3 necrosome forms a functional amyloid signaling complex required for programmed necrosis. Cell 150, 339–350 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Oancea, E., Teruel, M. N., Quest, A. F. & Meyer, T. Green fluorescent protein (GFP)-tagged cysteine-rich domains from protein kinase C as fluorescentindicators for diacylglycerol signaling in living cells. J. Cell Biol. 140, 485–498 (1998).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Aarts, M. et al. A key role for TRPM7 channels in anoxic neuronal death. Cell 115, 863–877 (2003).

    Article  CAS  PubMed  Google Scholar 

  26. McNulty, S. & Fonfria, E. The role of TRPM channels in cell death. Pflugers Arch. 451, 235–242 (2005).

    Article  CAS  PubMed  Google Scholar 

  27. Bates-Withers, C., Sah, R. & Clapham, D.E. TRPM7, the Mg(2+) inhibited channel and kinase. Adv. Exp. Med. Biol. 704, 173–183 (2011).

    Article  CAS  PubMed  Google Scholar 

  28. Tian, L. et al. Imaging neural activity in worms, flies and mice with improved GCaMP calcium indicators. Nat. Methods 6, 875–881 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Murphy, J. M. et al. The pseudokinase MLKL mediates necroptosis via a molecular switch mechanism. Immunity 39, 443–453 (2013).

    Article  CAS  PubMed  Google Scholar 

  30. Sciacca, M. F. et al. Cations as switches of amyloid-mediatedmembrane disruption mechanisms: calcium and IAPP. Biophys. J. 104, 173–184 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Desai, B. N. et al. Cleavage of TRPM7 releases the kinase domain from the ion channel and regulates its participation in Fas-induced apoptosis. Dev. Cell 22, 1149–1162 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Newton, K., Sun, X. & Dixit, V. M. Kinase RIP3 is dispensable for normal NF-kappa Bs, signaling by the B-cell and T-cell receptors, tumor necrosis factor receptor 1, and Toll-like receptors 2 and 4. Mol. Cell Biol. 24, 1464–1469 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

We thank J. Han for MLKL-deficient MEF cells and MLKL antibody, and L. Looger for GCaMP3 plasmid. This research was supported by the Intramural Research Program of the Center for Cancer Research, National Cancer Institute, National Institutes of Health.

Author information

Authors and Affiliations

Authors

Contributions

Z.C. and Z-G.L. conceived the study. Z.C. and S.J. did most of the experimental work. J.Z. did some confocal imaging experiments and cell death assays using MEF cells and TRPM7 shRNA JurkatFADD−/− cells. L-g.W. and H-C.C. designed and performed whole-cell patch clamp recording. Y.W. helped with confocal microscopy. J.L. generated MEF cells from RIP3-deficient mice and performed analysis. Z.C., S.C. and Z-G.L. wrote the paper. Z-G.L. supervised the study. All authors discussed the results and implications and commented on the manuscript at all stages.

Corresponding author

Correspondence to Zheng-Gang Liu.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Structure determination and assembly pattern prediction of coiled-coil domain, related to Fig. 1.

(a) Sequence alignment of the N-terminal coiled-coil domain of MLKL from vertebrate species. Two distinctive heptad repeat pattern (abcdefg)n characteristic of a coiled-coil architecture are indicated above the alignment. Coiled-coil residues occupying hydrophobic ‘a’ and ‘d’ position are denoted by yellow and red. These coiled-coil motifs are commonly shared among the MLKL orthologs in different vertebrate species. (b) Prediction of oligomerization states of coiled-coil domain of MLKL by Multicoil program (http://groups.csail.mit.edu/cb/multicoil/cgi-bin/multicoil.cgi). The score for potential coiled-coils with a dimeric probability is indicated by blue line and a trimeric probability is indicated by red line. The sum of these two probabilities is the total probability for forming a coiled-coil, which is indicated by black line.

Supplementary Figure 2 Quantification of plasma membrane translocation of DsRed-MLKL in response to TSZ treatment, related to Fig. 4d.

(a-c) MLKL shRNA HT29 cells transfected with DsRed tagged WT MLKL (a), CC1 (b) or CC2 (c) mutants and then treated with TSZ as indicated. A line intensity profile across the cell was obtained in a given image. Representative intensity profiles are shown in the right. Y-axis, Gray level; X-axis, Line distance. (d) The image shown in (a) was used for a representative example to show how to calculate a relative ratio of flurorescence intensity between plasma membrane adjacent area and cytosolic area. The ratio [R] was calculated by dividing the plasma membrane intensity [Ipm] by the average cytosolic fluorescence intensity [Icyt]. Scale bar, 5 μm.

Supplementary Figure 3 Subcellular localization of endogenous MLKL and RIP3 in HT29 cells.

Control shRNA (left panel) or MLKL shRNA (right panel) HT29 cells were treated with TSZ as indicated. Subcellular localization of (a) MLKL or (b) RIP3 was detected by confocal imaging of anti-MLKL antibody or anti-RIP3 antibody staining, respectively. Cell morphology was shown by F-actin probe phalloidin staining. DAPI was used for nuclear staining. White arrows indicate membrane localization. Scale bar, 10 μm.

Supplementary Figure 4 Cell surface protein isolation in MEF cells and J774A.1 cells.

MEF cells (a) or J774A.1 cells (b) were treated with TSZ as indicated. Total cellular lysate and the biotinylated, cell surface fraction were resolved on reducing gel and analyzed by immunoblot as indicated. Na+/K+ ATPase was used as a positive control for transmembrane/surface protein while Actin, COX IV and Calnexin were used as a positive control for cytosolic, mitochondrial and ER membrane proteins, respectively. Uncropped images of western blots are shown in Supplementary Fig. 8.

Supplementary Figure 5 MLKL-mediated Calcium influx is involved in plasma membrane rupture during necroptosis, related to Fig. 6.

(a) Flow cytometric analysis of intracellular Ca2+ of control shRNA, MLKL shRNA or RIP3 shRNA HT29 cells after treated with TSZ for 4 h with/out NSA or Nec-1 as indicated. Cells were harvested and the Fluo4 fluorescent cells were determined by FACS analysis using Fluo4. (b) HT29 cells were treated with TSZ either in normal DMEM or calcium free DMEM at different time points as indicated. The cell lysates were resolved on non-reducing gel and analyzed by immunoblot with anti-MLKL, anti RIP3, anti-RIP1 or anti-Actin antibodies. * indicates phosphorylated RIP3. (c) Representative image of live HT29 MLKL shRNA cells co-expressing DsRed-MLKL together with RIP3-EYFP treated with TSZ at time 0 and 4 hours in calcium free DMEM. Scale bar, 5 μm. Uncropped images of western blots are shown in Supplementary Fig. 8.

Supplementary Figure 6 Extracellular calcium is involved in necroptosis in JurkatFADD−/− and J774A.1 cells.

JurkatFADD−/− or J774A.1 cells were cultured with/out calcium and treated with DMSO control or TSZ as indicated. (a), (c) Cell survival was determined by PI staining. Results shown are averages ± s.e.m. from three independent experiments. *P<0.05, **P<0.01. Statistics source data for Supplementary Fig. 6 can be found in Supplementary Table 2. (b), (d) JurkatFADD−/− or J774A.1 cells expressing control shRNA, MLKL shRNA or cells cultured in calcium free medium were treated with TSZ as indicated. Cells were harvested and the Fluo4 fluorescent cells were determined by FACS analysis using Fluo4. (e), (f) Immunoblot analysis of JuarkatFADD−/− (e) or J774A.1 (f) clones expressing control-shRNA or MLKL-shRNA with anti-MLKL or anti-Actin antibodies. Uncropped images of western blots are shown in Supplementary Fig. 8.

Supplementary Figure 7 ROS and calcium are both required for necroptosis in U937 cells.

(a) U937 cells were either cultured in calcium free medium or cultured in calcium free medium and pre-treated with NAC (5 mM). Then, the cells were treated with TSZ. After 10 hours, cell survival was determined by PI staining. Results shown are averages of triplicates ± SEM. (b) U937 cells cultured in calcium free DMEM were treated with TSZ as indicated. Cells were harvested and the Fluo4 fluorescent cells were determined by FACS analysis using Fluo4. (c) Immunoblot analysis of U937 clones expressing control-shRNA or MLKL-shRNA with anti-MLKL or anti-Actin antibodies. * indicates non-specific band. **,P<0.01. Statistics source data for Supplementary Fig. 7 can be found in Supplementary Table 1. Uncropped images of western blots are shown in Supplementary Fig. 8.

Supplementary information

Supplementary Information

Supplementary Information (PDF 1708 kb)

Supplementary Table 1

Supplementary Information (XLSX 27 kb)

Representative time-lapse GCaMP3 fluorescence imaging of control-shRNA HT29 cells treated with TSZ for 8 hours.

Left, GFP channel; Middle, differential interference contrast channel (DIC); Right, Merged two channels. (MOV 2256 kb)

Representative time-lapse GCaMP3 fluorescence imaging of TRPM7-sh#1RNA HT29 cells treated with TSZ for 8 hours.

Left, GFP channel; Middle, differential interference contrast channel (DIC); Right, Merged two channels. (MOV 3095 kb)

Representative time-lapse GCaMP3 fluorescence imaging of control-shRNA HT29 cells cultured in calcium free DMEM and treated with TSZ for 8 hours.

Left, GFP channel; Middle, differential interference contrast channel (DIC); Right, Merged two channels. (MOV 3031 kb)

Rights and permissions

Reprints and permissions

About this article

Cite this article

Cai, Z., Jitkaew, S., Zhao, J. et al. Plasma membrane translocation of trimerized MLKL protein is required for TNF-induced necroptosis. Nat Cell Biol 16, 55–65 (2014). https://doi.org/10.1038/ncb2883

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/ncb2883

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