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.

  • Protocol
  • Published:

A real-time fluorometric method for the simultaneous detection of cell death type and rate

Abstract

Several cell death assays have been developed based on a single biochemical parameter such as caspase activation or plasma membrane permeabilization. Our fluorescent apoptosis/necrosis (FAN) assay directly measures cell death and distinguishes between caspase-dependent apoptosis and caspase-independent necrosis of cells grown in any multiwell plate. Cell death is monitored in standard growth medium as an increase in fluorescence intensity of a cell-impermeable dye (SYTOX Green) after plasma membrane disintegration, whereas apoptosis is detected through caspase-mediated release of a fluorophore from its quencher (DEVD-amc). The assay determines the normalized percentage of dead cells and caspase activation per condition as an end-point measurement or in real time (automated). The protocol can be applied to screen drugs, proteins or siRNAs for interference with cell death while simultaneously detecting cell death modality switching between apoptosis and necrosis. Initial preparation may take up to 5 d, but the typical hands-on time is 2 h.

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: Principle and validation of the fluorescent apoptosis–necrosis assay.
Figure 2: Detection of apoptosis and necrosis by the FAN assay in different model systems.

Similar content being viewed by others

References

  1. Jouan-Lanhouet, S. et al. Necroptosis, in vivo detection in experimental disease models. Semin. Cell Dev. Biol. 35, 2–13 (2014).

    Article  CAS  Google Scholar 

  2. Linkermann, A. et al. The potential role of necroptosis in diseases. Necrotic Cell Death Vol. 2 (eds. Shen, H. & Vandenabeele, P.) 1–21 (Humana Press, 2014).

  3. Murphy, J.M. & Silke, J. Ars Moriendi; the art of dying well – new insights into the molecular pathways of necroptotic cell death. EMBO Rep. 15, 155–164 (2014).

    Article  CAS  Google Scholar 

  4. Vanden Berghe, T., Linkermann, A., Jouan-Lanhouet, S., Walczak, H. & Vandenabeele, P. Regulated necrosis: the expanding network of non-apoptotic cell death pathways. Nat. Rev. Mol. Cell Biol. 15, 135–147 (2014).

    Article  CAS  Google Scholar 

  5. Vanden Berghe, T., Kaiser, W.J., Bertrand, M.J.M. & Vandenabeele, P. Molecular crosstalk between apoptosis, necroptosis and survival signaling. Mol. Cell. Oncol. 2, e975093 (2015).

    Article  Google Scholar 

  6. Vercammen, D., Vandenabeele, P., Beyaert, R., Declercq, W. & Fiers, W. Tumour necrosis factor-induced necrosis versus anti-Fas-induced apoptosis in L929 cells. Cytokine 9, 801–808 (1997).

    Article  CAS  Google Scholar 

  7. Dondelinger, Y. et al. RIPK3 contributes to TNFR1-mediated RIPK1 kinase-dependent apoptosis in conditions of cIAP1/2 depletion or TAK1 kinase inhibition. Cell Death Differ. 20, 1381–1392 (2013).

    Article  CAS  Google Scholar 

  8. Kalai, M. et al. Tipping the balance between necrosis and apoptosis in human and murine cells treated with interferon and dsRNA. Cell Death Differ. 9, 981–994 (2002).

    Article  CAS  Google Scholar 

  9. Lawlor, K.H. et al. RIPK3 promotes cell death and NLRP3 inflammasome activation in the absence of MLKL. Nat. Commun. 6, 6282 (2015).

    Article  CAS  Google Scholar 

  10. Remijsen, Q. et al. Depletion of RIPK3 or MLKL blocks TNF-driven necroptosis and switches towards a delayed RIPK1 kinase-dependent apoptosis. Cell Death Dis. 5, e1004 (2014).

    Article  CAS  Google Scholar 

  11. Vanden Berghe, T., Kalai, M., van Loo, G., Declercq, W. & Vandenabeele, P. Disruption of HSP90 function reverts tumor necrosis factor-induced necrosis to apoptosis. J. Biol. Chem. 278, 5622–5629 (2003).

    Article  CAS  Google Scholar 

  12. Vanden Berghe, T. et al. Differential signaling to apoptotic and necrotic cell death by Fas-associated death domain protein FADD. J. Biol. Chem. 279, 7925–7933 (2004).

    Article  CAS  Google Scholar 

  13. 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  Google Scholar 

  14. Lindhagen, E., Nygren, P. & Larsson, R. The fluorometric microculture cytotoxicity assay. Nat. Protoc. 3, 1364–1369 (2008).

    Article  CAS  Google Scholar 

  15. Mosmann, T. Rapid colorimetric assay for cellular growth and survival: application to proliferation and cytotoxicity assays. J. Immunol. Methods 65, 55–63 (1983).

    Article  CAS  Google Scholar 

  16. O'Brien, J., Wilson, I., Orton, T. & Pognan, F. Investigation of the Alamar Blue (resazurin) fluorescent dye for the assessment of mammalian cell cytotoxicity. Eur. J. Biochem. 267, 5421–5426 (2000).

    Article  CAS  Google Scholar 

  17. Rossi, C. et al. Identifying druglike inhibitors of myelin-reactive T cells by phenotypic high-throughput screening of a small-molecule library. J. Biomol. Screen. 12, 481–489 (2007).

    Article  CAS  Google Scholar 

  18. Grootjans, S., Goossens, V., Vandenabeele, P. & Vanden Berghe, T. Methods to study and distinguish necroptosis. in Necrotic Cell Death Vol. 2 (eds. Shen, H. & Vandenabeele, P.) 335–361 (Humana Press, 2014).

  19. Vanden Berghe, T. et al. Determination of apoptotic and necrotic cell death in vitro and in vivo. Methods 61, 117–129 (2013).

    Article  CAS  Google Scholar 

  20. Riccardi, C. & Nicoletti, I. Analysis of apoptosis by propidium iodide staining and flow cytometry. Nat. Protoc. 1, 1458–1461 (2006).

    Article  CAS  Google Scholar 

  21. Vanden Berghe, T. et al. Necroptosis, necrosis and secondary necrosis converge on similar cellular disintegration features. Cell Death Differ. 17, 922–930 (2010).

    Article  CAS  Google Scholar 

  22. Demon, D. et al. Proteome-wide substrate analysis indicates substrate exclusion as a mechanism to generate caspase-7 versus caspase-3 specificity. Mol. Cell Proteomics 8, 2700–2714 (2009).

    Article  CAS  Google Scholar 

  23. Thornberry, N.A., Chapman, K.T. & Nicholson, D.W. Determination of caspase specificities using a peptide combinatorial library. Methods Enzymol. 322, 100–110 (2000).

    Article  CAS  Google Scholar 

  24. Denecker, G. et al. Death receptor-induced apoptotic and necrotic cell death: differential role of caspases and mitochondria. Cell Death Differ. 8, 829–840 (2001).

    Article  CAS  Google Scholar 

  25. Dondelinger, Y. et al. MLKL compromises plasma membrane integrity by binding to phosphatidylinositol phosphates. Cell Rep. 7, 971–981 (2014).

    Article  CAS  Google Scholar 

  26. Estornes, Y. et al. RIPK1 promotes death receptor-independent caspase-8-mediated apoptosis under unresolved ER stress conditions. Cell Death Dis. 5, e1555 (2014).

    Article  CAS  Google Scholar 

  27. Dondelinger, Y. et al. NF-κB-independent role of IKKα/IKKβ in preventing RIPK1 kinase-dependent apoptotic and necroptotic cell death during TNF signaling. Mol. Cell 60, 63–76 (2015).

    Article  CAS  Google Scholar 

  28. Aaes, T.L. et al. Vaccination with necroptotic cancer cells induces efficient anti-tumor immunity. Cell Rep. 15, 274–287 (2016).

    Article  CAS  Google Scholar 

  29. Krysko, O., De Ridder, L. & Cornelissen, M. Phosphatidylserine exposure during early primary necrosis (oncosis) in JB6 cells as evidenced by immunogold labeling technique. Apoptosis 9, 495–500 (2004).

    Article  CAS  Google Scholar 

  30. Thal, S.E., Zhu, C., Thal, S.C., Blomgren, K. & Plesnila, N. Role of apoptosis inducing factor (AIF) for hippocampal neuronal cell death following global cerebral ischemia in mice. Neurosci. Lett. 499, 1–3 (2011).

    Article  CAS  Google Scholar 

  31. Zhang, J. et al. EndoG links Bnip3-induced mitochondrial damage and caspase-independent DNA fragmentation in ischemic cardiomyocytes. PLoS ONE 6, e17998 (2011).

    Article  CAS  Google Scholar 

  32. Aguer, C. et al. Galactose enhances oxidative metabolism and reveals mitochondrial dysfunction in human primary muscle cells. PLoS ONE 6, e28536 (2011).

    Article  CAS  Google Scholar 

  33. Roth, B.L., Poot, M., Yue, S.T. & Millard, P.J. Bacterial viability and antibiotic susceptibility testing with SYTOX green nucleic acid stain. Appl. Environ. Microbiol. 63, 2421–2431 (1997).

    CAS  PubMed  PubMed Central  Google Scholar 

  34. Fukunaga, M. & Yielding, L.W. Structure-function characterization of phenanthridinium compounds as mutagens in Salmonella. Mutat. Res. 121, 89–94 (1983).

    Article  CAS  Google Scholar 

  35. Kraman, M. et al. Suppression of antitumor immunity by stromal cells expressing fibroblast activation protein-α. Science 330, 827–830 (2010).

    Article  CAS  Google Scholar 

  36. Pasparakis, M. & Vandenabeele, P. Necroptosis and its role in inflammation. Nature 517, 311–320 (2015).

    Article  CAS  Google Scholar 

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

    Article  CAS  Google Scholar 

  38. 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  Google Scholar 

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

    Article  CAS  Google Scholar 

  40. Feng, S. et al. Cleavage of RIP3 inactivates its caspase-independent apoptosis pathway by removal of kinase domain. Cell. Signal. 19, 2056–2067 (2007).

    Article  CAS  Google Scholar 

  41. Yang, W.S. et al. Regulation of ferroptotic cancer cell death by GPX4. Cell 156, 317–331 (2014).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We acknowledge the following members of our laboratory: Q. Remijsen for his contribution to the initial development of the assay, and R. Roelandt, S. Martens, M. Aguileta and Y. Estornes for sharing data that were not included in the final manuscript. We acknowledge D. Fearon, University of Cambridge, for his kind gift of LL2-OVA cells. We are grateful to A. Bredan for editing the manuscript. Research in the Vandenabeele group is supported by Belgian grants (Interuniversity Attraction Poles, IAP 7/32), Flemish grants (Research Foundation Flanders: FWO G.0875.11, FWO G.0973.11, FWO G.0A45.12N, FWO G.0172.12, FWO G.0787.13N, FWO G.0607.13N, FWO KAN 31528711, FWO KAN 1504813N, FWO G0E04.16N), Methusalem grant (BOF09/01M00709 and BOF16/MET_V/007), Ghent University grants (MRP, GROUP-ID consortium, BOF14/GOA/019), a grant from the Foundation Against Cancer (2012-188) and grants from VIB. P.V. holds Methusalem grants (BOF09/01M00709 and BOF16/MET_V/007).

Author information

Authors and Affiliations

Authors

Contributions

S.G. and Y.D. developed the protocol. S.G., M.V., V.G., T.V.B. and P.V. designed the research, interpreted the data and wrote the paper. S.G., B.H., I.D. and B.W. carried out the experiments. M.V. performed the repeated-measurements data analysis. M.J.M.B. and D.V.K. provided critical remarks and discussion.

Corresponding authors

Correspondence to Sasker Grootjans or Tom Vanden Berghe.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 Validation of the FAN assay

a, The linear range for SYTOX Green decreases at lower concentrations (< 5μM) even if signal intensity is below the saturation level of the detector. b- c, Cell membrane permeabilization kinetics (b) and caspase activation (c) in IMR-32 cells treated with 2 μM Staurosporin (STS). d- e The effect of DEVD-amc on propidium iodide (PI) intensity and vice versa. Fas-overexpressing L929 cells were treated with 250 ng/ml anti-Fas antibody in the presence or absence of 20 μM DEVD-amc or 10μM PI, and amc intensity (d) and PI intensity (e) was measured 30 h later. f, The effect of phenol red on PI intensity is minimal for permeabilized L929 cells. g- h, Cell membrane permeabilization kinetics measured with 10μM PI (g) and caspase activation (h) in L929-Fas cells treated with anti-Fas Ab (250 ng/ml). The same plate was measured at 6 and 30 hours after stimulation, and returned to the tissue culture incubator in the interval time. Error bars = standard error of the mean, obtained from three independent experiments (n=3), each measured in triplicate. ***, p ≤ 0.001. Non-significant differences are not indicated. AV, arbitrary values.

Supplementary Figure 2 Example of a plate lay-out to determine to optimal combination of gain setting, SYTOX Green concentration and cell number.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1 and 2, Supplementary Table and Supplementary Note (PDF 747 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Grootjans, S., Hassannia, B., Delrue, I. et al. A real-time fluorometric method for the simultaneous detection of cell death type and rate. Nat Protoc 11, 1444–1454 (2016). https://doi.org/10.1038/nprot.2016.085

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nprot.2016.085

This article is cited by

Comments

By submitting a comment you agree to abide by our Terms and Community Guidelines. If you find something abusive or that does not comply with our terms or guidelines please flag it as inappropriate.

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