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.

  • Technical Report
  • Published:

Single-fluorophore biosensors for sensitive and multiplexed detection of signalling activities

Abstract

Unravelling the dynamic molecular interplay behind complex physiological processes such as neuronal plasticity requires the ability to both detect minute changes in biochemical states in response to physiological signals and track multiple signalling activities simultaneously. Fluorescent protein-based biosensors have enabled the real-time monitoring of dynamic signalling processes within the native context of living cells, yet most commonly used biosensors exhibit poor sensitivity (for example, due to low dynamic range) and are limited to imaging signalling activities in isolation. Here, we address this challenge by developing a suite of excitation ratiometric kinase activity biosensors that offer the highest reported dynamic range and enable the detection of subtle changes in signalling activity that could not be reliably detected previously, as well as a suite of single-fluorophore biosensors that enable the simultaneous tracking of as many as six distinct signalling activities in single living cells.

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: Design and characterization of ExRai-AKAR.
Fig. 2: ExRai-AKAR shows improved performance over previous-generation AKARs.
Fig. 3: ExRai-AKAR amplifies minute activity changes and reveals compartmentalized PKA signalling in growth factor-stimulated PC12 cells.
Fig. 4: Construction of ExRai-CKAR and ExRai-AktAR based on a generalized design.
Fig. 5: AKAR and CKAR colour variants based on cp-T-sapphire and cpBFP.
Fig. 6: Generating redshifted single-fluorophore sensors using ddRFP.
Fig. 7: Multiplexed activity imaging using single-fluorophore biosensors.

Similar content being viewed by others

Data availability

Source data for bar graphs shown in Figs. 16 and Supplementary Fig. 6 have been provided as Supplementary Table 1. All other data supporting the findings of this study are available upon reasonable request.

References

  1. Depry, C., Mehta, S. & Zhang, J. Multiplexed visualization of dynamic signaling networks using genetically encoded fluorescent protein-based biosensors. Pflugers Arch. 465, 373–381 (2013).

    Article  CAS  Google Scholar 

  2. Piljic, A. & Schultz, C. Simultaneous recording of multiple cellular events by FRET. ACS Chem. Biol. 3, 156–160 (2008).

    Article  CAS  Google Scholar 

  3. Nagai, T., Sawano, A., Park, E. S. & Miyawaki, A. Circularly permuted green fluorescent proteins engineered to sense Ca2+. Proc. Natl Acad. Sci. USA 98, 3197–3202 (2001).

    Article  CAS  Google Scholar 

  4. Nakai, J., Ohkura, M. & Imoto, K. A high signal-to-noise Ca2+ probe composed of a single green fluorescent protein. Nat. Biotechnol. 19, 137–141 (2001).

    Article  CAS  Google Scholar 

  5. Yang, H. H. et al. Subcellular imaging of voltage and calcium signals reveals neural processing in vivo. Cell 166, 245–257 (2016).

    Article  CAS  Google Scholar 

  6. Berg, J., Hung, Y. P. & Yellen, G. A genetically encoded fluorescent reporter of ATP:ADP ratio. Nat. Methods 6, 161–166 (2009).

    Article  CAS  Google Scholar 

  7. Marvin, J. S. et al. An optimized fluorescent probe for visualizing glutamate neurotransmission. Nat. Methods 10, 162–170 (2013).

    Article  CAS  Google Scholar 

  8. Odaka, H., Arai, S., Inoue, T. & Kitaguchi, T. Genetically-encoded yellow fluorescent cAMP indicator with an expanded dynamic range for dual-color imaging. PLoS ONE 9, e100252 (2014).

    Article  Google Scholar 

  9. Zhang, J., Hupfeld, C. J., Taylor, S. S., Olefsky, J. M. & Tsien, R. Y. Insulin disrupts beta-adrenergic signalling to protein kinase A in adipocytes. Nature 437, 569–573 (2005).

    Article  CAS  Google Scholar 

  10. 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  Google Scholar 

  11. Tsien, R. Y. The green fluorescent protein. Annu. Rev. Biochem. 67, 509–544 (1998).

    Article  CAS  Google Scholar 

  12. Zhao, Y. et al. An expanded palette of genetically encoded Ca2+ indicators. Science 333, 1888–1891 (2011).

    Article  CAS  Google Scholar 

  13. Depry, C., Allen, M. D. & Zhang, J. Visualization of PKA activity in plasma membrane microdomains. Mol. Biosyst. 7, 52–58 (2011).

    Article  CAS  Google Scholar 

  14. Komatsu, N. et al. Development of an optimized backbone of FRET biosensors for kinases and GTPases. Mol. Biol. Cell 22, 4647–4656 (2011).

    Article  CAS  Google Scholar 

  15. Herbst, K. J., Allen, M. D. & Zhang, J. Spatiotemporally regulated protein kinase A activity is a critical regulator of growth factor-stimulated extracellular signal-regulated kinase signaling in PC12 cells. Mol. Cell. Biol. 31, 4063–4075 (2011).

    Article  CAS  Google Scholar 

  16. Nam, H.-S. & Benezra, R. High levels of Id1 expression define B1 type adult neural stem cells. Cell Stem Cell 5, 515–526 (2009).

    Article  CAS  Google Scholar 

  17. Newman, R. H., Fosbrink, M. D. & Zhang, J. Genetically encodable fluorescent biosensors for tracking signaling dynamics in living cells. Chem. Rev. 111, 3614–3666 (2011).

    Article  CAS  Google Scholar 

  18. Herbst, K. J., Allen, M. D. & Zhang, J. Luminescent kinase activity biosensors based on a versatile bimolecular switch. J. Am. Chem. Soc. 133, 5676–5679 (2011).

    Article  CAS  Google Scholar 

  19. Gao, X. & Zhang, J. Spatiotemporal analysis of differential Akt regulation in plasma membrane microdomains. Mol. Biol. Cell 19, 4366–4373 (2008).

    Article  CAS  Google Scholar 

  20. Zapata-Hommer, O. & Griesbeck, O. Efficiently folding and circularly permuted variants of the Sapphire mutant of GFP. BMC Biotechnol. 3, 5 (2003).

    Article  Google Scholar 

  21. Hung, Y. P., Albeck, J. G., Tantama, M. & Yellen, G. Imaging cytosolic NADH-NAD(+) redox state with a genetically encoded fluorescent biosensor. Cell Metab. 14, 545–554 (2011).

    Article  CAS  Google Scholar 

  22. Akerboom, J. et al. Genetically encoded calcium indicators for multi-color neural activity imaging and combination with optogenetics. Front. Mol. Neurosci. 6, 2 (2013).

    Article  CAS  Google Scholar 

  23. Alford, S. C., Abdelfattah, A. S., Ding, Y. & Campbell, R. E. A fluorogenic red fluorescent protein heterodimer. Chem. Biol. 19, 353–360 (2012).

    Article  CAS  Google Scholar 

  24. Ding, Y. et al. Ratiometric biosensors based on dimerization-dependent fluorescent protein exchange. Nat. Methods 12, 195–198 (2015).

    Article  CAS  Google Scholar 

  25. Violin, J. D. et al. beta2-adrenergic receptor signaling and desensitization elucidated by quantitative modeling of real time cAMP dynamics. J. Biol. Chem. 283, 2949–2961 (2008).

    Article  CAS  Google Scholar 

  26. Huganir, R. L. & Nicoll, R. A. AMPARs and synaptic plasticity: the last 25 years. Neuron 80, 704–717 (2013).

    Article  CAS  Google Scholar 

  27. Diering, G. H., Gustina, A. S. & Huganir, R. L. PKA-GluA1 coupling via AKAP5 controls AMPA receptor phosphorylation and cell-surface targeting during bidirectional homeostatic plasticity. Neuron 84, 790–805 (2014).

    Article  CAS  Google Scholar 

  28. Sanderson, J. L., Gorski, J. A. & Dell’Acqua, M. L. NMDA receptor-dependent ltd requires transient synaptic incorporation of Ca2+-permeable AMPARs mediated by AKAP150-anchored PKA and calcineurin. Neuron 89, 1000–1015 (2016).

    Article  CAS  Google Scholar 

  29. Wang, J. et al. Multiple kinases involved in the nicotinic modulation of gamma oscillations in the rat hippocampal CA3 area. Front. Cell. Neurosci. 11, 57 (2017).

    PubMed  PubMed Central  Google Scholar 

  30. Rao, V. et al. PKA phosphorylation of cardiac troponin I modulates activation and relaxation kinetics of ventricular myofibrils. Biophys. J. 107, 1196–1204 (2014).

    Article  CAS  Google Scholar 

  31. Hu, Z. et al. Acute mechanical stretch promotes eNOS activation in venous endothelial cells mainly via PKA and Akt pathways. PLoS ONE 8, e71359 (2013).

    Article  CAS  Google Scholar 

  32. Dunn, T. A. et al. Imaging of cAMP levels and protein kinase A activity reveals that retinal waves drive oscillations in second-messenger cascades. J. Neurosci. 26, 12807–12815 (2006).

    Article  CAS  Google Scholar 

  33. Smith, F. D. et al. Local protein kinase A action proceeds through intact holoenzymes. Science 356, 1288–1293 (2017).

    Article  CAS  Google Scholar 

  34. Sample, V. et al. Regulation of nuclear PKA revealed by spatiotemporal manipulation of cyclic AMP. Nat. Chem. Biol. 8, 375–382 (2012).

    Article  CAS  Google Scholar 

  35. Niino, Y., Hotta, K. & Oka, K. Blue fluorescent cGMP sensor for multiparameter fluorescence imaging. PLoS ONE 5, e9164 (2010).

    Article  Google Scholar 

  36. Aye-Han, N.-N. & Zhang, J. A multiparameter live cell imaging approach to monitor cyclic AMP and protein kinase A dynamics in parallel. Methods Mol. Biol. 1071, 207–215 (2014).

    Article  CAS  Google Scholar 

  37. Harada, K. et al. Red fluorescent protein-based cAMP indicator applicable to optogenetics and in vivo imaging. Sci. Rep. 7, 7351 (2017).

    Article  Google Scholar 

  38. Tewson, P. et al. Simultaneous detection of Ca2+ and diacylglycerol signaling in living cells. PLoS ONE 7, e42791 (2012).

    Article  CAS  Google Scholar 

  39. Suzuki, K. et al. Five colour variants of bright luminescent protein for real-time multicolour bioimaging. Nat. Commun. 7, 13718 (2016).

    Article  CAS  Google Scholar 

  40. Mo, G. C. H. et al. Genetically encoded biosensors for visualizing live-cell biochemical activity at super-resolution. Nat. Methods 14, 427–434 (2017).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

The authors are grateful to L. Looger, G. Yellen, M. Matsuda, T. Kitaguchi and R. Campbell for generously providing plasmids and to S. Taylor for providing purified PKA catalytic subunit. We also wish to thank E. Greenwald for helping with the bleed-through analysis and correction, as well as E. Lopez Ortega for helping with subcloning and neuronal imaging experiments. This work was supported by Brain Initiative Grant R01 MH111516 (to R.L.H. and J.Z.) and by R35 CA197622, R01 DK073368 and R01 GM111665 (to J.Z.). Y.Z. is supported by the National Natural Science Foundation of China (grant 31771125), the Ministry of Science and Technology of the People’s Republic of China (grant 2017YFE0103400) and The One Thousand Talents Plan-Recruitment Program for Young Professionals.

Author information

Authors and Affiliations

Authors

Contributions

S.M. and J.Z. conceived of the project. S.M. designed ExRai-AKAR and generated all of the biosensors used in the study, with assistance from B.T.; A.M. performed in vitro characterization of purified biosensors; S.M. and J.-f.Z. performed live-cell imaging in HeLa, HEK293T, NIH3T3 and PC12 cells; Y.Z., R.H.R. and R.L.H. devised the neuronal experiments; Y.Z. and R.H.R. performed live-cell imaging in primary cortical neurons; R.L.H. and J.Z. supervised the project and coordinated the experiments; S.M., Y.Z., R.H.R., J.-f.Z. and A.M. analysed the data; and S.M., Y.Z., R.H.R., R.L.H. and J.Z. wrote the manuscript.

Corresponding authors

Correspondence to Sohum Mehta, Richard L. Huganir or Jin Zhang.

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.

Integrated supplementary information

Supplementary Figure 1 Additional characterization of ExRai-AKAR.

(a) Normalized excitation spectra of unphosphorylated ExRai-AKAR (1 μM) measured in buffer with the indicated pH values (see Methods). (b) Plot of the maximum intensity of the 380 nm excitation peak (teal) and the 488 nm excitation peak at each pH. Non-linear regression was performed to determine the pKa value for each excitation peak. (c) Plot of the ratio of the 480 nm and 380 nm excitation peaks obtained at each pH. Data points in b and c represent the mean ± SEM of three independent determinations. (d) The ExRai-AKAR response is reversible. Average time-course showing the 480/380 excitation ratio change in HeLa cells expressing ExRai-AKAR and treated with 50 μM Fsk and 100 μM IBMX (Fsk/IBMX) followed by 20 μM H89. The curve is plotted as the 480/380 excitation ratio normalized with respect to time 0 (R/R0). The solid line represents the mean, and the shaded area represents SEM. n=6 cells from a single experiment; experiment was repeated 10 times with similar results. (e-g) Individual single-cell traces showing the non-normalized PKA-stimulated changes in (e) 480 nm-excited fluorescence intensity, (f) 380 nm-excited fluorescence intensity, and (g) 480/380 excitation ratio in HeLa cells expressing ExRai-AKAR and treated with Fsk/IBMX. n = 8 cells from the same representative experiment shown in Fig. 1; experiment was repeated 11 times with similar results

Supplementary Figure 2 Investigating compartmentalized PKA signaling in growth factor-stimulated PC12 cells.

(a) Domain structures of ExRai-AKAR-NES and ExRai-AKAR-Kras. (b, c) Representative average excitation ratio time-courses of (b) ExRai-AKAR-NES (n = 5 cells) and (c) ExRai-AKAR-Kras (n = 4 cells) in HeLa cells treated with Fsk/IBMX. Inset: representative fluorescence images showing the (b) cytosolic or (c) plasma membrane distribution of probe fluorescence in the individual excitation channels. Curves are plotted as 480/380 excitation ratio normalized with respect to time 0 (R/R0). Solid lines represent the mean, and the shaded areas represent SEM. Scale bar, 30 μm. (d-i) Growth factor-induced PKA responses in PC12 cells monitored using ExRai-AKAR. (d, e) Excitation ratio time-courses of the ExRai-AKAR-Kras response in PC12 cells stimulated with (d) 200 ng/mL NGF (n = 8 cells pooled from 4 experiments) or (e) 100 ng/mL EGF (n = 7 cells pooled from 4 experiments). Inset: Representative images showing the plasma membrane distribution of probe fluorescence in the individual excitation channels. Scale bars, 10 μm. (f-i) Excitation ratio time-courses of the ExRai-AKAR responses in PC12 cells stimulated with (f, h) 200 ng/mL NGF (n = 17 cells pooled from 8 experiments) or (g, i) 100 ng/mL EGF (n = 16 cells pooled from 6 experiments) and recorded in the (f, g) nucleus or (h, i) cytosol. All curves are plotted as 480/380 excitation ratio normalized with respect to time 0 (R/R0). Thick lines indicate the average responses, and thin lines depict individual single-cell traces. Experiments in b and c were each repeated 3 times with similar results

Supplementary Figure 3 Non-normalized response curves for additional cpFP-based sensors.

(a, b) Individual single-cell traces showing the non-normalized changes in 480/380 excitation ratio in (a) ExRai-CKAR-expressing HeLa cells treated with 100 ng/mL PMA (n = 5 cells from the same representative experiment shown in Fig. 3b) and (b) in ExRai-AktAR-expressing NIH3T3 cells treated with 50 ng/mL PDGF (n = 15 cells pooled from 12 experiments). (c, d) Individual single-cell traces showing non-normalized changes in fluorescence intensity in (c) sapphireAKAR-expressing HeLa cells treated with 50 μM Fsk and 100 μM IBMX (Fsk/IBMX; n = 5 cells from the same representative experiment shown in Fig. 4c) and (d) in sapphireCKAR-expressing HeLa cells treated with 100 ng/mL PMA (n = 4 cells from the same representative experiment shown in Fig. 4d). (e, f) Individual single-cell traces showing non-normalized changes in fluorescence intensity in (e) blueAKAR-expressing HeLa cells treated with Fsk/IBMX (n = 4 cells from the same representative experiment shown in Fig. 4g) and (f) in blueCKAR-expressing HeLa cells treated with PMA (n = 5 cells from the same representative experiment shown in Fig 4h). Experiments were repeated (a) 5, (c) 6, (d) 5, (e) 5, and (f) 6 times with similar results

Supplementary Figure 4 Characterization of red-shifted single-fluorophore activity sensors.

(a) Initial unsuccessful attempt to generate a red-shifted kinase activity sensor based on cpRFP. Top: Domain structure of redAKAR prototype containing cp-mRuby sandwiched between a PKA substrate sequence and FHA1 domain. Bottom: Average time-course of RFP fluorescence intensity in HEK293T cells transfected with redAKAR and treated with 50 μM Fsk. The solid line indicates the mean, and the shaded area indicates SEM. Curve is plotted as RFP fluorescence intensity normalized with respect to time 0 (F/F0). n = 14 cells from a representative experiment. Inset: Representative epifluorescence image showing very dim RFP fluorescence in HEK293T cells transfected with redAKAR. Scale bar, 30 μm. (b) The RAB-EKARev response is reversible. Average time-course showing the change in RFP fluorescence intensity in HEK293T cells expressing RAB-EKARev and treated with 100 ng/mL EGF followed by 20 μM U0126 to reverse the response. The curve is plotted as RFP fluorescence intensity normalized with respect to time 0 (F/F0). The solid line represents the mean, and the shaded area represents SEM. n = 18 cells from a representative experiment. (g) Individual single-cell traces showing non-normalized RFP fluorescence intensity changes in (left) RAB-EKARev-expressing HEK293T cells treated with 100 ng/mL EGF (n = 10 cells from the same representative experiment shown in Fig. 5c), (middle) in RAB-AKARev-expressing HeLa cells treated with Fsk/IBMX (n = 6 cells from the same representative experiment shown in Fig. 5d), and (right) in RAB-ICUE-expressing HEK293T cells treated with Fsk/IBMX (n = 6 cells from the same representative experiment shown in Fig. 5e). Experiments were repeated (b) 9, (c) 7, (d) 3, and (e) 4 times with similar results; experiment in a was repeated twice with similar results

Supplementary Figure 5 Additional representative curves and bleed-through analysis for three-parameter imaging in HeLa cells.

(a) Representative single-cell time-courses showing the changes in BFP, RFP, and T-sapphire (TsR) fluorescence intensity in HeLa cells co-expressing blueAKAR (blue), RAB-EKARev (red), and sapphireCKAR (teal) upon sequential treatment with 50 μM Fsk and 100 μM IBMX (Fsk/IBMX), 100 ng/mL EGF, and 100 ng/mL PMA. (b) Representative single-cell time-courses showing the changes in BFP, RFP, and T-sapphire fluorescence intensity in HeLa cells co-expressing blueAKAR (blue), RAB-ICUE (red), and sapphireCKAR (teal) upon sequential treatment with Fsk/IBMX and PMA. (c) Alternative three-parameter imaging of PKA, cAMP, and PKC. Representative single-cell traces of the change in TsR, RFP, and BFP fluorescence intensity in HeLa cells co-expressing sapphireAKAR (teal), RAB-ICUE (red), and blueCKAR (blue) upon sequential treatment with Fsk/IBMX and PMA (n = 14 cells total across 6 independent experiments). All curves are plotted as fluorescence intensity normalized with respect to time 0 (F/F0). (d) To analyze fluorescence bleed-through, HeLa cells were individually transfected with sapphireCKAR, blueAKAR, RAB-EKARev, or RAB-ICUE, and images were acquired in the TsR, BFP, and RFP channels for cells expressing each construct. Representative scatter plots show the pixel intensity in each channel as a function of the pixel intensity in the probe channel, and line traces show the pixel intensity in the probe channel alone. Obvious bleed-through of fluorescence intensity was only detected in the TsR channel in cells expressing blueAKAR, and the bleed-through correction factor was calculated by determining the slope of the TsR vs BFP pixel intensity graph for blueAKAR-expressing cells. (e, f) Representative single-cell traces of TsR channel intensity in HeLa cells transfected only with blueAKAR before (dashed line) and after (solid line) BFP bleed-through correction. (g) Representative fluorescence images for each construct in each channel. Scale bars, 30 μm. Single-cell traces in a and b are derived from the same datasets as Figs. 6a (5 independent experiments) and 6b (3 independent experiments), respectively. Experiments in d-g were performed 5 times with similar results

Supplementary Figure 6 Characterization of single-fluorophore kinase sensors in cultured rat cortical neurons and HeLa cells.

(a) Average time-course of ExRai-AKAR (Ex488) and dsRed responses in the cell soma following Fsk stimulation. Insets: Representative ExRai-AKAR fluorescence images before (-Fsk) and after (+Fsk) stimulation and maximal responses (F/F0) (****p<0.0001, two-tailed paired Student’s t-test; n = 19 neurons pooled from 3 experiments]). Data are shown as dot plots; bars represent mean ± SEM. (b) Average time-course of RAB-EKARev and GFP responses in the cell soma following DHPG stimulation. Insets: Representative RAB-EKARev fluorescence images before (-DHPG) and after (+DHPG) stimulation and maximal responses (F/F0) (P = 0.0117, paired Student’s t-test; n = 6 neurons pooled from 3 experiments). Data are shown as dot plots; bars represent mean ± s.e.m. (c) Average time-course of ExRai-AKAR (Ex488), RAB-EKARev, and BCaMP responses in the cell soma following Fsk stimulation. Inset: maximal responses (F/F0) of ExRai-AKAR, RAB-EKARev, and BCaMP (p = 0.0005, ****p<0.0001, Kruskal-Wallis test; n = 68 neurons pooled from 6 experiments). (d) Average time-course of ExRai-AKAR (488 nm excitation), RAB-EKARev, and BCaMP responses in the cell soma following DHPG stimulation. Inset: Maximal responses (F/F0) of ExRai-AKAR, RAB-EKARev, and BCaMP (****p<0.0001, Kruskal-Wallis test; n = 104 neurons pooled from 10 experiments). Box-and-whisker plots depict the median, interquartile range, min, max, and mean (+). All data points are shown. Maximum responses are calculated with respect to the initial intensity at time 0 (F/F0). (e) Domain structures of Lyn-sapphireAKAR and sapphireAKAR-NLS. (f, g) Average time-courses of (f) Lyn-sapphireAKAR (n = 5 cells) and (g) sapphireAKAR-NLS (n = 6 cells) responses in HeLa cells treated with 50 μM Fsk and 100 μM IBMX (Fsk/IBMX). (h) Domain structure of Lyn-RAB-EKARev and RAB-EKARev-NLS. (i, j) Average time-courses of (i) Lyn-RAB-EKARev (n = 9 cells) and RAB-EKARev-NLS (n = 18 cells) in HeLa cells treated with 100 ng/mL EGF. Data in f, g, i, and j are pooled from two independent experiments; representative images show probe localization. Curves are normalized with respect to the average baseline intensity (a-d) or to time 0 (F/F0) (f, g, i, and j). Solid lines represent the mean; shaded areas, SEM. Scale bars, 10 μm. See Supplementary Table 1 for bar graph source data

Supplementary Figure 7 Additional representative curves and bleed-through analysis for four- and six-parameter imaging in HeLa cells.

(a) Representative single-cell time-courses showing the changes in T-sapphire (TsR), YFP, BFP, and RFP fluorescence intensity in HeLa cells co-expressing sapphireAKAR (teal), Flamindo2 (yellow), blueCKAR (blue), and RCaMP (red) upon sequential treatment with 50 μM Fsk and 100 μM IBMX (Fsk/IBMX), 100 ng/mL PMA, and 1 μM ionomycin (iono). (b, c) Representative single-cell time-courses of the changes in TsR, YFP, BFP, and RFP fluorescence intensity in HeLa cells co-expressing Lyn-sapphireAKAR (teal), sapphireAKAR-NLS (light blue), Flamindo2 (yellow), RAB-EKARev-NLS (red), Lyn-RAB-EKARev (pink), and B-GECO1 (blue) upon sequential treatment with Fsk/IBMX, EGF, and 100 μM histamine, showing cells both without (b) and with (c) EGF-induced calcium transients. All curves are plotted as fluorescence intensity normalized with respect to time 0 (F/F0). (d) To analyze fluorescence bleed-through, HeLa cells were individually transfected with sapphireAKAR, blueCKAR, Flamindo2, or RCaMP, and images were acquired in the TsR, BFP, YFP, and RFP channels for cells expressing each construct. Representative scatter plots show the pixel intensity in each channel as a function of the pixel intensity in the probe channel, and line traces show the pixel intensity in the probe channel alone. Obvious bleed-through of fluorescence intensity was only detected in the TsR channel in cells expressing blueCKAR, and the bleed-through correction factor was calculated by determining the slope of the TsR vs BFP pixel intensity graph for blueCKAR-expressing cells. (e) Representative single-cell traces of TsR channel intensity in a HeLa cell transfected only with blueCKAR before (dashed line) and after (solid line) BFP bleed-through correction. (f) Representative fluorescence images for each construct in each channel. Scale bars, 30 μm. Single-cell traces in a and b, c are derived from the same datasets as Figures 6e (12 independent experiments) and 6f (10 independent experiments), respectively. Experiments in d-f were repeated 5 times with similar results

Supplementary information

Supplementary Information

Supplementary Figures 1–7 and Supplementary Table 1 legend.

Reporting Summary

Supplementary Table 1

Statistics source data.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Mehta, S., Zhang, Y., Roth, R.H. et al. Single-fluorophore biosensors for sensitive and multiplexed detection of signalling activities. Nat Cell Biol 20, 1215–1225 (2018). https://doi.org/10.1038/s41556-018-0200-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41556-018-0200-6

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