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:

Live-cell measurements of kinase activity in single cells using translocation reporters

Abstract

Although kinases are important regulators of many cellular processes, measuring their activity in live cells remains challenging. We have developed kinase translocation reporters (KTRs), which enable multiplexed measurements of the dynamics of kinase activity at a single-cell level. These KTRs are composed of an engineered construct in which a kinase substrate is fused to a bipartite nuclear localization signal (bNLS) and nuclear export signal (NES), as well as to a fluorescent protein for microscopy-based detection of its localization. The negative charge introduced by phosphorylation of the substrate is used to directly modulate nuclear import and export, thereby regulating the reporter's distribution between the cytoplasm and nucleus. The relative cytoplasmic versus nuclear fluorescence of the KTR construct (the C/N ratio) is used as a proxy for the kinase activity in living, single cells. Multiple KTRs can be studied in the same cell by fusing them to different fluorescent proteins. Here, we present a protocol to execute and analyze live-cell microscopy experiments using KTRs. We describe strategies for development of new KTRs and procedures for lentiviral expression of KTRs in a cell line of choice. Cells are then plated in a 96-well plate, from which multichannel fluorescent images are acquired with automated time-lapse microscopy. We provide detailed guidance for a computational analysis and parameterization pipeline. The entire procedure, from virus production to data analysis, can be completed in 10 d.

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: Schematic representation of KTRs.
Figure 2: General schematic showing how live-cell measurements of kinase activity are performed using kinase translocation reporters (KTRs).
Figure 3: NES and bNLS consensus sequences.
Figure 4: Schematic outlining image and downstream analysis workflow.

Similar content being viewed by others

References

  1. Manning, G., Whyte, D.B., Martinez, R., Hunter, T. & Sudarsanam, S. The protein kinase complement of the human genome. Science 298, 1912–1934 (2002).

    Article  CAS  Google Scholar 

  2. Cohen, P. & Philip, C. The role of protein phosphorylation in human health and disease. Eur. J. Biochem. 268, 5001–5010 (2001).

    Article  CAS  Google Scholar 

  3. Purvis, J.E. & Lahav, G. Encoding and decoding cellular information through signaling dynamics. Cell 152, 945–956 (2013).

    Article  CAS  Google Scholar 

  4. Selimkhanov, J. et al. Accurate information transmission through dynamic biochemical signaling networks. Science 346, 1370–1373 (2014).

    Article  CAS  Google Scholar 

  5. Shalek, A.K. et al. Single-cell RNA-seq reveals dynamic paracrine control of cellular variation. Nature 510, 363–369 (2014).

    Article  CAS  Google Scholar 

  6. Zernicka-Goetz, M., Morris, S.A. & Bruce, A.W. Making a firm decision: multifaceted regulation of cell fate in the early mouse embryo. Nat. Rev. Genet. 10, 467–477 (2009).

    Article  CAS  Google Scholar 

  7. Vlamakis, H., Aguilar, C., Losick, R. & Kolter, R. Control of cell fate by the formation of an architecturally complex bacterial community. Genes Dev. 22, 945–953 (2008).

    Article  CAS  Google Scholar 

  8. Spencer, S.L. et al. The proliferation-quiescence decision is controlled by a bifurcation in CDK2 activity at mitotic exit. Cell 155, 369–383 (2013).

    Article  CAS  Google Scholar 

  9. Albeck, J.G., Mills, G.B. & Brugge, J.S. Frequency-modulated pulses of ERK activity transmit quantitative proliferation signals. Mol. Cell 49, 249–261 (2013).

    Article  CAS  Google Scholar 

  10. Regot, S., Hughey, J.J., Bajar, B.T., Carrasco, S. & Covert, M.W. High-sensitivity measurements of multiple kinase activities in live single cells. Cell 157, 1724–1734 (2014).

    Article  CAS  Google Scholar 

  11. Kondoh, K., Terasawa, K., Morimoto, H. & Nishida, E. Regulation of nuclear translocation of extracellular signal-regulated kinase 5 by active nuclear import and export mechanisms. Mol. Cell. Biol. 26, 1679–1690 (2006).

    Article  CAS  Google Scholar 

  12. Nardozzi, J.D., Lott, K. & Cingolani, G. Phosphorylation meets nuclear import: a review. Cell Commun. Signal. 8, 32 (2010).

    Article  CAS  Google Scholar 

  13. Noursadeghi, M. et al. Quantitative imaging assay for NF-κB nuclear translocation in primary human macrophages. J. Immunol. Methods 329, 194–200 (2008).

    Article  CAS  Google Scholar 

  14. Maryu, G., Matsuda, M. & Aoki, K. Multiplexed fluorescence imaging of ERK and Akt activities and cell-cycle progression. Cell Struct. Funct. 41, 81–92 (2016).

    Article  CAS  Google Scholar 

  15. Oldach, L. & Zhang, J. Genetically encoded fluorescent biosensors for live-cell visualization of protein phosphorylation. Chem. Biol. 21, 186–197 (2014).

    Article  CAS  Google Scholar 

  16. Lam, A.J. et al. Improving FRET dynamic range with bright green and red fluorescent proteins. Nat. Methods 9, 1005–1012 (2012).

    Article  CAS  Google Scholar 

  17. Kosugi, S., Hasebe, M., Tomita, M. & Yanagawa, H. Nuclear export signal consensus sequences defined using a localization-based yeast selection system. Traffic 9, 2053–2062 (2008).

    Article  CAS  Google Scholar 

  18. Kosugi, S., Hasebe, M., Tomita, M. & Yanagawa, H. Systematic identification of cell cycle-dependent yeast nucleocytoplasmic shuttling proteins by prediction of composite motifs. Proc. Natl. Acad. Sci. USA 106, 10171–10176 (2009).

    Article  CAS  Google Scholar 

  19. Sieracki, N.A. & Komarova, Y.A. Studying cell signal transduction with biomimetic point mutations. In Manipulation of DNA and Protein –Examples from Current Research, 381–392. (In Tech, 2013).

  20. Miyamoto, T. et al. Compartmentalized AMPK signaling illuminated by genetically encoded molecular sensors and actuators. Cell Rep. 11, 657–670 (2015).

    Article  CAS  Google Scholar 

  21. Miura, H., Haruko, M., Michiyuki, M. & Kazuhiro, A. Development of a FRET biosensor with high specificity for Akt. Cell Struct. Funct. 39, 9–20 (2014).

    Article  CAS  Google Scholar 

  22. Seong, J. et al. Detection of focal adhesion kinase activation at membrane microdomains by fluorescence resonance energy transfer. Nat. Commun. 2, 406 (2011).

    Article  Google Scholar 

  23. Ubersax, J.A. & Ferrell, J.E. Jr. Mechanisms of specificity in protein phosphorylation. Nat. Rev. Mol. Cell Biol. 8, 530–541 (2007).

    Article  CAS  Google Scholar 

  24. Pearce, L.R., Komander, D. & Alessi, D.R. The nuts and bolts of AGC protein kinases. Nat. Rev. Mol. Cell Biol. 11, 9–22 (2010).

    Article  CAS  Google Scholar 

  25. Moritz, A. et al. Akt-RSK-S6 kinase signaling networks activated by oncogenic receptor tyrosine kinases. Sci. Signal. 3, ra64 (2010).

    Article  Google Scholar 

  26. Hornbeck, P.V. et al. PhosphoSitePlus, 2014: mutations, PTMs and recalibrations. Nucleic Acids Res. 43, D512–D520 (2015).

    Article  CAS  Google Scholar 

  27. McNulty, D.E., Sikorski, T.W. & Annan, R.S. Identification and analysis of protein phosphorylation by mass spectrometry. in Analysis of Protein Post-Translational Modifications by Mass Spectrometry 17–87 (2016).

  28. Peck, S.C. Analysis of protein phosphorylation: methods and strategies for studying kinases and substrates. Plant J. 45, 512–522 (2006).

    Article  CAS  Google Scholar 

  29. Van Valen, D.A. et al. Deep learning automates the quantitative analysis of individual cells in live-cell imaging experiments. PLoS Comput. Biol. 12, e1005177 (2016).

    Article  Google Scholar 

  30. Schindelin, J. et al. Fiji: an open-source platform for biological-image analysis. Nat. Methods 9, 676–682 (2012).

    Article  CAS  Google Scholar 

  31. Carpenter, A.E. et al. CellProfiler: image analysis software for identifying and quantifying cell phenotypes. Genome Biol. 7, R100 (2006).

    Article  Google Scholar 

  32. Edelstein, A.D. et al. Advanced methods of microscope control using μManager software. J. Biol. Methods 1, e10 (2014).

    Article  Google Scholar 

  33. Magidson, V. & Khodjakov, A. Circumventing photodamage in live-cell microscopy. Methods Cell Biol. 114, 545–560 (2013).

    Article  Google Scholar 

  34. Basu, S., Campbell, H.M., Dittel, B.N. & Ray, A. Purification of specific cell population by fluorescence activated cell sorting (FACS). J. Vis. Exp. http://dx.doi.org/10.3791/1546 (2010).

  35. Masip, M.E. et al. Reversible cryo-arrest for imaging molecules in living cells at high spatial resolution. Nat. Methods 13, 665–672 (2016).

    Article  CAS  Google Scholar 

  36. Tay, S. et al. Single-cell NF-kappaB dynamics reveal digital activation and analogue information processing. Nature 466, 267–271 (2010).

    Article  CAS  Google Scholar 

  37. Williams, R.G. et al. Generation and characterization of a stable MK2-EGFP cell line and subsequent development of a high-content imaging assay on the Cellomics ArrayScan platform to screen for p38 mitogen-activated protein kinase inhibitors. Methods Enzymol. 364–389 (2006).

  38. Anton, R. et al. A p38 substrate-specific MK2-EGFP translocation assay for identification and validation of new p38 inhibitors in living cells: a comprising alternative for acquisition of cellular p38 inhibition data. PLoS ONE 9, e95641 (2014).

    Article  Google Scholar 

  39. Reinhardt, H.C. et al. DNA damage activates a spatially distinct late cytoplasmic cell-cycle checkpoint network controlled by MK2-mediated RNA stabilization. Mol. Cell 40, 34–49 (2010).

    Article  CAS  Google Scholar 

  40. Samsonov, A. et al. Tagging of genomic STAT3 and STAT1 with fluorescent proteins and insertion of a luciferase reporter in the cyclin D1 gene provides a modified A549 cell line to screen for selective STAT3 inhibitors. PLoS ONE 8, e68391 (2013).

    Article  CAS  Google Scholar 

  41. Yissachar, N. et al. Dynamic response diversity of NFAT isoforms in individual living cells. Mol. Cell 49, 322–330 (2013).

    Article  CAS  Google Scholar 

  42. Gross, S.M. & Rotwein, P. Akt signaling dynamics in individual cells. J. Cell Sci. 128, 2509–2519 (2015).

    Article  CAS  Google Scholar 

  43. Katsura, Y. et al. An optogenetic system for interrogating the temporal dynamics of Akt. Sci. Rep. 5, 14589 (2015).

    Article  CAS  Google Scholar 

  44. Gross, S.M. & Peter, R. Mapping growth-factor-modulated Akt signaling dynamics. Development 143, e1.2 (2016).

    Article  Google Scholar 

  45. Schmierer, B. & Hill, C.S. Kinetic analysis of Smad nucleocytoplasmic shuttling reveals a mechanism for transforming growth factor-dependent nuclear accumulation of Smads. Mol. Cell. Biol. 25, 9845–9858 (2005).

    Article  CAS  Google Scholar 

  46. Wang, C.-C. et al. A time- and matrix-dependent TGFBR3–JUND–KRT5 regulatory circuit in single breast epithelial cells and basal-like premalignancies. Nat. Cell Biol. 16, 345–356 (2014).

    Article  CAS  Google Scholar 

  47. Hahn, A.T., Jones, J.T. & Meyer, T. Quantitative analysis of cell cycle phase durations and PC12 differentiation using fluorescent biosensors. Cell Cycle 8, 1044–1052 (2009).

    Article  CAS  Google Scholar 

  48. Zhao, B. et al. Inactivation of YAP oncoprotein by the Hippo pathway is involved in cell contact inhibition and tissue growth control. Genes Dev. 21, 2747–2761 (2007).

    Article  CAS  Google Scholar 

  49. Cohen-Saidon, C., Cohen, A.A., Sigal, A., Liron, Y. & Alon, U. Dynamics and variability of ERK2 response to EGF in individual living cells. Mol. Cell 36, 885–893 (2009).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank the members of Covert lab for advice and critical commentary on the manuscript, in particular K. Lane for supplying the images of primary macrophages in Supplementary Figure 1. We gratefully acknowledge funding from several sources, including a Distinguished Investigator award and a Discovery Center grant from the Paul Allen Family Foundation, as well as an NIH Pioneer Award (5DP1LM01150-05) to M.W.C., a Human Frontier Science Program (HFSP) postdoctoral fellowship (LT000529/ 2012-L) to S.R., the Nakajima Foundation Scholarship to T.K., a DOE CSGF grant (DE-FG02-97ER25308) to D.N.M., and a Systems Biology Center grant (P50 GM107615).

Author information

Authors and Affiliations

Authors

Contributions

S.R. developed the reporter strategy and designed the experiments. T.K. and D.N.M. developed the computational pipelines and prepared the protocols. J.J.H. and S.A. developed the general workflow for the modeling. T.K., S.J., and M.W.C. wrote the manuscript, with input from all authors.

Corresponding authors

Correspondence to Sergi Regot or Markus W Covert.

Ethics declarations

Competing interests

The authors declare no competing financial interests.

Integrated supplementary information

Supplementary Figure 1 KTR translocation in different cell lines.

Time relative to stimulation is shown on each images.

(A) HeLa cells expressing JNK KTR were stimulated with anisomycin 50 ng/ml.

(B) HEK293 cells expressing JNK KTR were stimulated with anisomycin 50 ng/ml.

(C) Raw 264.7 cells expressing JNK KTR were stimulated with LPS 100 ng/ml.

(D) PC-12 cells expressing ERK KTR were stimulated with NGF 25 ng/ml.

(E) Primary Bone Marrow-derived Macrophages (BMDMs) isolated from mice expressing JNK KTR were stimulated with LPS 100 ng/ml.

Supplementary Figure 2 KTR translocation and the estimated active JNK concentration.

(A) Heatmap illustrating the experimental C/N ratio, simulated C/N ratio and predicted active JNK concentration upon IL-1β stimulation. Each row denotes a color coded individual time course of an indicated property for a single cell. This is the final output from the jupyter notebook demonstration. (B) The population average traces of the C/N ratio (red), active JNK concentration (blue) and total phosphorylated KTR concentration (green). Note that the C/N ratio is acquired by direct analysis of the KTR image data, while the JNK and KTR concentrations are calculated by further analysis using a computational mechanistic model of KTR phosphorylation and dephosphorylation as well as nuclear or cytoplasmic translocation. The concentrations are normalized by minimum and maximum values.

Supplementary information

Supplementary Text and Figures

Supplementary Figures 1 and 2, Supplementary Methods, Supplementary Tables 1–3, Supplementary Notes 1 and 2, and Supplementary Data. (PDF 8160 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Kudo, T., Jeknić, S., Macklin, D. et al. Live-cell measurements of kinase activity in single cells using translocation reporters. Nat Protoc 13, 155–169 (2018). https://doi.org/10.1038/nprot.2017.128

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/nprot.2017.128

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