Skip to main content

Advertisement

Log in

Multiplexed bioluminescence microscopy via phasor analysis

  • Article
  • Published:

From Nature Methods

View current issue Submit your manuscript

Abstract

Bioluminescence imaging with luciferase–luciferin pairs is a well-established technique for visualizing biological processes across tissues and whole organisms. Applications at the microscale, by contrast, have been hindered by a lack of detection platforms and easily resolved probes. We addressed this limitation by combining bioluminescence with phasor analysis, a method commonly used to distinguish spectrally similar fluorophores. We built a camera-based microscope equipped with special optical filters to directly assign phasor locations to unique luciferase–luciferin pairs. Six bioluminescent reporters were easily resolved in live cells, and the readouts were quantitative and instantaneous. Multiplexed imaging was also performed over extended time periods. Bioluminescent phasor further provided direct measures of resonance energy transfer in single cells, setting the stage for dynamic measures of cellular and molecular features. The merger of bioluminescence with phasor analysis fills a long-standing void in imaging capabilities, and will bolster future efforts to visualize biological events in real time and over multiple length scales.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1: Bioluminescent phasor imaging.
Fig. 2: Facile multicomponent imaging with bioluminescent phasor.
Fig. 3: Continuous, excitation-free imaging of tumor spheroids.

Similar content being viewed by others

Data availability

Source images with unprocessed data are available on Figshare: https://doi.org/10.6084/m9.figshare.19607862.

Code availability

The algorithm that supports the findings of this study is available on GitHub: https://github.com/LorenzoScipioni/Phasor–Bioluminescence.

References

  1. Love, A. C. & Prescher, J. A. Seeing (and using) the light: recent developments in bioluminescence technology. Cell Chem. Biol. 27, 904–920 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  2. Syed, A. J. & Anderson, J. C. Applications of bioluminescence in biotechnology and beyond. Chem. Soc. Rev. 50, 5668–5705 (2021).

    Article  CAS  PubMed  Google Scholar 

  3. Mezzanotte, L., van ‘t Root, M., Karatas, H., Goun, E. A. & Löwik, C. In vivo molecular bioluminescence imaging: new tools and applications. Trends Biotechnol. 35, 640–652 (2017).

    Article  CAS  PubMed  Google Scholar 

  4. Contag, C. H. & Bachmann, M. H. Advances in in vivo bioluminescence imaging of gene expression. Annu. Rev. Biomed. Eng. 4, 235–260 (2002).

    Article  CAS  PubMed  Google Scholar 

  5. Yeh, H.-W. & Ai, H.-W. Development and applications of bioluminescent and chemiluminescent reporters and biosensors. Annu. Rev. Anal. Chem. 12, 129–150 (2019).

    Article  CAS  Google Scholar 

  6. Yao, Z., Zhang, B. S. & Prescher, J. A. Advances in bioluminescence imaging: new probes from old recipes. Curr. Opin. Chem. Biol. 45, 148–156 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Rathbun, C. M. & Prescher, J. A. Bioluminescent probes for imaging biology beyond the culture dish. Biochemistry 56, 5178–5184 (2017).

    Article  CAS  PubMed  Google Scholar 

  8. Ogoh, K. et al. Bioluminescence microscopy using a short focal-length imaging lens. J. Microsc. 253, 191–197 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Rodriguez, E. A. et al. The growing and glowing toolbox of fluorescent and photoactive proteins. Trends Biochem. Sci. 42, 111–129 (2017).

    Article  CAS  PubMed  Google Scholar 

  10. Rathbun, C. M. et al. Rapid multicomponent bioluminescence imaging via substrate unmixing. ACS Chem. Biol. 16, 682––690 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Kobayashi, H., Picard, L.-P., Schönegge, A.-M. & Bouvier, M. Bioluminescence resonance energy transfer–based imaging of protein–protein interactions in living cells. Nat. Protoc. 14, 1084–1107 (2019).

    Article  CAS  PubMed  Google Scholar 

  12. Hall, M. P. et al. Engineered luciferase reporter from a deep sea shrimp utilizing a novel imidazopyrazinone xubstrate. ACS Chem. Biol. 7, 1848–1857 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  13. Dixon, A. S. et al. NanoLuc complementation reporter optimized for accurate measurement of protein interactions in cells. ACS Chem. Biol. 11, 400–408 (2016).

    Article  CAS  PubMed  Google Scholar 

  14. Azad, T. et al. SARS-CoV-2 S1 NanoBiT: a nanoluciferase complementation-based biosensor to rapidly probe SARS-CoV-2 receptor recognition. Biosens. Bioelectron. 180, 113122 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. England, C. G., Ehlerding, E. B. & Cai, W. NanoLuc: a small luciferase is brightening up the field of bioluminescence. Bioconjug. Chem. 27, 1175–1187 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Chu, J. et al. A bright cyan-excitable orange fluorescent protein facilitates dual-emission microscopy and enhances bioluminescence imaging in vivo. Nat. Biotechnol. 34, 760–767 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Takai, A. et al. Expanded palette of Nano-lanterns for real-time multicolor luminescence imaging. Proc. Natl Acad. Sci. USA 112, 4352 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  18. Schaub, F. X. et al. Fluorophore-NanoLuc BRET reporters enable sensitive in vivo optical imaging and flow cytometry for monitoring tumorigenesis. Cancer Res. 75, 5023–5033 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Saito, K. et al. Luminescent proteins for high-speed single-cell and whole-body imaging. Nat. Commun. 3, 1262 (2012).

    Article  PubMed  CAS  Google Scholar 

  21. Su, Y. et al. Novel NanoLuc substrates enable bright two-population bioluminescence imaging in animals. Nat. Methods 17, 852–860 (2020).

    Article  CAS  PubMed  Google Scholar 

  22. Fereidouni, F., Bader, A. N. & Gerritsen, H. C. Spectral phasor analysis allows rapid and reliable unmixing of fluorescence microscopy spectral images. Opt. Express 20, 12729–12741 (2012).

    Article  CAS  PubMed  Google Scholar 

  23. Malacrida, L., Ranjit, S., Jameson, D. M. & Gratton, E. The phasor plot: a universal circle to advance fluorescence lifetime analysis and interpretation. Annu. Rev. Biophys. 50, 575–593 (2021).

    Article  CAS  PubMed  Google Scholar 

  24. Cutrale, F. et al. Hyperspectral phasor analysis enables multiplexed 5D in vivo imaging. Nat. Methods 14, 149–152 (2017).

    Article  CAS  PubMed  Google Scholar 

  25. Gammon, S. T., Leevy, W. M., Gross, S., Gokel, G. W. & Piwnica-Worms, D. Spectral unmixing of multicolored bioluminescence emitted from heterogeneous biological sources. Anal. Chem. 78, 1520–1527 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Hedde, P. N., Cinco, R., Malacrida, L., Kamaid, A. & Gratton, E. Phasor-based hyperspectral snapshot microscopy allows fast imaging of live, three-dimensional tissues for biomedical applications. Commun. Biol. 4, 721 (2021).

    Article  PubMed  PubMed Central  Google Scholar 

  27. Dvornikov, A. & Gratton, E. Hyperspectral imaging in highly scattering media by the spectral phasor approach using two filters. Biomed. Opt. Express 9, 3503–3511 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Iwano, S. et al. Single-cell bioluminescence imaging of deep tissue in freely moving animals. Science 359, 935 (2018).

    Article  CAS  PubMed  Google Scholar 

  29. Reddy, G. R., Thompson, W. C. & Miller, S. C. Robust light emission from cyclic alkylaminoluciferin substrates for firefly luciferase. J. Am. Chem. Soc. 132, 13586–13587 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  30. Rathbun, C. M. et al. Parallel screening for rapid identification of orthogonal bioluminescent tools. ACS Cent. Sci. 3, 1254–1261 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Jones, K. A. et al. Orthogonal luciferase–luciferin pairs for bioluminescence imaging. J. Am. Chem. Soc. 139, 2351–2358 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Harwood, K. R., Mofford, D. M., Reddy, G. R. & Miller, S. C. Identification of mutant firefly luciferases that efficiently utilize aminoluciferins. Chem. Biol. 18, 1649–1657 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Branchini, B. R., Magyar, R. A., Murtiashaw, M. H. & Portier, N. C. The role of active site residue arginine 218 in firefly luciferase bioluminescence. Biochemistry 40, 2410–2418 (2001).

    Article  CAS  PubMed  Google Scholar 

  34. Branchini, B. R., Southworth, T. L., Murtiashaw, M. H., Boije, H. & Fleet, S. E. A mutagenesis study of the putative luciferin binding site residues of firefly luciferase. Biochemistry 42, 10429–10436 (2003).

    Article  CAS  PubMed  Google Scholar 

  35. Frei, M. S. et al. Engineered HaloTag variants for fluorescence lifetime multiplexing. Nat. Methods 19, 65–70 (2022).

    Article  CAS  PubMed  Google Scholar 

  36. Frei, M. S., Koch, B., Hiblot, J. & Johnsson, K. Live-cell fluorescence lifetime multiplexing using synthetic fluorescent probes. ACS Chem. Biol., https://pubs.acs.org/doi/10.1021/acschembio.2c00041(2022).

  37. Mehl, B. P. et al. Live cell biosensors based on the fluorescence lifetime of environment-sensing dyes. Preprint at bioRxiv https://doi.org/10.1101/2022.02.08.479035 (2022).

  38. Zhang, B. S., Jones, K. A., McCutcheon, D. C. & Prescher, J. A. Pyridone luciferins and mutant luciferases for bioluminescence imaging. ChemBioChem 19, 470–477 (2018).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  39. Yeh, H.-w et al. ATP-independent bioluminescent reporter variants to improve in vivo imaging. ACS Chem. Biol. 14, 959–965 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Bajar, B. T., Wang, E. S., Zhang, S., Lin, M. Z. & Chu, J. A guide to fluorescent protein FRET pairs. Sensors 6, 1488 (2016).

    Article  CAS  Google Scholar 

  41. Lay, C. S. et al. Probing the binding of interleukin-23 to individual receptor components and the IL-23 heteromeric receptor complex in living cells using NanoBRET. Cell Chem. Biol. 29, 19–29.e16 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Machleidt, T. et al. NanoBRET—a novel BRET platform for the analysis of protein–protein Interactions. ACS Chem. Biol. 10, 1797–1804 (2015).

    Article  CAS  PubMed  Google Scholar 

  43. Hiblot, J. et al. Luciferases with tunable emission wavelengths. Angew. Chem. Int. Ed. 56, 14556–14560 (2017).

    Article  CAS  Google Scholar 

  44. Aper, S. J. A., Dierickx, P. & Merkx, M. Dual readout BRET/FRET sensors for measuring intracellular zinc. ACS Chem. Biol. 11, 2854–2864 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Yang, J. et al. Coupling optogenetic stimulation with NanoLuc-based luminescence (BRET) Ca2+ sensing. Nat. Commun. 7, 13268 (2016).

    Article  PubMed  PubMed Central  Google Scholar 

  46. Inagaki, S. et al. Genetically encoded bioluminescent voltage indicator for multi-purpose use in wide range of bioimaging. Sci. Rep. 7, 42398–42398 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  47. Yevtodiyenko, A. et al. Portable bioluminescent platform for in vivo monitoring of biological processes in non-transgenic animals. Nat. Commun. 12, 2680 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Celinskis, D. et al. Miniaturized devices for bioluminescence imaging in freely behaving animals. Preprint at bioRxiv https://doi.org/10.1101/2020.06.15.152546 (2020).

  49. Steinhardt, R. C. et al. Brominated luciferins are versatile bioluminescent probes. ChemBioChem 18, 96–100 (2017).

    Article  CAS  PubMed  Google Scholar 

  50. Brennan, C. K., Ornelas, M. Y., Yao, Z. W. & Prescher, J. A. Multicomponent bioluminescence imaging with naphthylamino luciferins. ChemBioChem 22, 2650–2654 (2021).

    Article  CAS  PubMed  Google Scholar 

  51. Musicant, A. M. et al. CRTC1/MAML2 directs a PGC-1α-IGF-1 circuit that confers vulnerability to PPARγ inhibition. Cell Rep. 34, 108768 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

This research was supported by an Allen Distinguished Investigator Award, a Paul G. Allen Frontiers Group advised grant of the Paul G. Allen Family Foundation (to J.A.P. and M.A.D.), the US National Institutes of Health (grant no. R01 GM107630 to J.A.P., grant no. P41-GM103540 to L.S., E.G., and M.A.D., grant no. R01 DE030123 to A.L.A.) and the UNC University Cancer Research Fund (to A.L.A.). Z.Y. was supported by the National Science Foundation via the BEST IGERT (grant no. DGE-1144901) program and a Graduate Research Fellowship (grant no. DGE-1321846). C.K.B. was supported by the UCI Physical Sciences Machine Learning NEXUS program. We thank members of the Laboratory of Fluorescence Dynamics (LFD, UCI) for discussion. Some experiments were performed at the Laser Spectroscopy Laboratories (LSL) at UCI.

Author information

Authors and Affiliations

Authors

Contributions

Z.Y., C.K.B., L.S., M.A.D. and J.A.P. conceived the project idea. Z.Y., C.K.B., L.S. and H.C. performed the experiments. L.S. and H.C. designed and built the imaging setup. Z.Y., C.K.B. and G.T. prepared the biological samples. L.S. and E.G. wrote the codes and analyzed the raw imaging data. K.K.N., C.K.B., K.P.-S. and A.L.A. generated the reporter constructs. All authors analyzed data and contributed to the writing of the manuscript. All authors have given approval to the final version of the manuscript.

Corresponding authors

Correspondence to Michelle A. Digman or Jennifer A. Prescher.

Ethics declarations

Competing interests

The authors declare no competing interests.

Peer review

Peer review information

Nature Methods thanks Huiwang Ai, Francesco Cutrale and the other, anonymous, reviewer(s) for their contribution to the peer review of this work. Primary Handling Editor: Rita Strack, in collaboration with the Nature Methods team.

Additional information

Publisher’s note Springer Nature remains neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Extended data

Extended Data Fig. 1 Popular bioluminescence reporters produce distinct phasor signatures.

Solutions of recombinant (a) Fluc (500 nM), (b) Akaluc (10 μM), (c) yellow enhanced Nano-lantern (YeNL, 10 nM), and (d) yellow Nano-lantern (YNL, 100 nM) were mixed with their respective luciferin substrates (CycLuc129 or AkaLumine28 = 250 μM with 1 mM ATP; FRZ19 or CTZ17 = 50 μM). Images were acquired using the microscope setup pictured in Supplementary Fig. 1 using a 10 s/frame integration time. A total of 20 frames were collected for each sample and the phasor locations were computed. The emission maximum for each luciferase-luciferin pair is shown for reference.

Extended Data Fig. 2 Phasor signatures from BRET reporters.

HeLa cells were transiently transfected with plasmids encoding (a) Nluc, (b) CeNL, (c) GeNL, or (d) YeNL. The cells were then treated with FRZ (25–50 μM) ~36 h post transfection. Images were acquired using the microscope setup pictured in Supplementary Fig. 1 with a 20X air objective and 10 s/frame integration time. A total of 20 frames were collected for each sample and the phasor locations were computed. Each pixel in the resulting images was false colored according to the phasor signature. The expected phasor for each construct (based on its emission spectrum and BRET efficiency) is also shown for comparison. BRET efficiencies were determined by comparing the acceptor output to the total photon output. The images are color-coded according to the specific reporter, assigned after clustering. These experiments were repeated three times with similar results. Scale bars = 25 μm.

Extended Data Fig. 3 BRET reporters with similar emission maxima can be distinguished via phasor analysis.

HeLa cells were transiently transfected with plasmids encoding (a) OeNL (b) ReNL, or (c) LumiScarlet. The cells were then treated with FRZ (25–50 μM) ~36 h post transfection. Images were acquired using the microscope setup pictured in Supplementary Fig. 1 with a 20X air objective and 10 s/frame integration time. A total of 20 frames were collected for each sample and the phasor locations were computed. Each pixel in the resulting images was false colored according to the phasor signature. The expected phasor for each construct (based on its emission spectrum and BRET efficiency) is also shown for comparison. The BRET efficiency was determined by comparing the acceptor output to the total photon output. The images are color-coded according to the specific reporter, assigned after clustering. These experiments were repeated three times with similar results. Scale bars = 25 μm.

Extended Data Fig. 4 Bioluminescent phasor reveals heterogeneous BRET efficiencies with single cell resolution.

(a) Pixels from images acquired with OeNL-expressing HeLa cells (Extended Data Fig. 3a) were re-assigned based on the computed BRET efficiency. The donor was assigned based on the phasor signature from Nluc-expressing cells (Extended Data Fig. 2a), and the acceptor location was assigned based on the fluorescent spectrum of mKOκ. (b) BRET heterogeneity observed with OeNL-expressing cells was validated on a confocal microscope (Zeiss LSM880) equipped with a 32-channel spectral detector. Multiple cells were analyzed per field of view, with similar results. (c) Pixels from images acquired with ReNL-expressing HeLa cells (Extended Data Fig. 3b) were re-assigned based on the computed BRET efficiency. The donor location was assigned based on the phasor signature from Nluc-expressing cells (Extended Data Fig. 2a), and the acceptor location was assigned based on the fluorescent spectrum of tdTomato. (d) BRET heterogeneity was validated on a confocal microscope (Zeiss LSM880) equipped with a 32-channel spectral detector. Multiple cells were analyzed per field of view, with similar results. Scale bars = 25 μm.

Extended Data Fig. 5 Multicomponent imaging with BRET reporters.

HeLa cells were transfected with either Nluc or a BRET reporter (CeNL, GeNL, YeNL, ReNL, OeNL, or LumiScarlet). The transfected cells were then randomly mixed and plated onto a glass slide 16 h post-transfection. The cells were treated with FRZ (25–50 μM). Images were acquired using the microscope setup pictured in Supplementary Fig. 1 with a 20X air objective and 10 s/frame integration time. A total of 20 frames were collected for each sample and the phasor locations were computed. Each pixel in the resulting image was false colored according to the phasor signature. Selected examples of (a, b) two-component and (c, d) four-component mixtures are shown. Each component in the mixture was assigned by referencing to the single populations shown in Extended Data Figs. 2 and 3. Images are representative of n > 3 independent experiments. Scale bars = 25 μm.

Extended Data Fig. 6 Phasor analysis is required to differentiate probes with similar acceptor fluorophores.

HeLa cells were transfected with plasmids encoding either ReNL or LumiScarlet. The transfected cells were then mixed and plated onto a glass slide 16 h post-transfection. The ‘TRITC filter’ image was obtained by summing the channels corresponding to the interval 578-633 nm, mimicking a 605/55 emission filter (left). The ‘fluorescence color’ image (center left) was obtained by transforming all channels of the spectral detector in RGB and combining them. The ‘phasor’ image (center right) was false-colored after phasor clustering (right). Multiple cells were analyzed per field of view, with similar results. Scale bars = 25 μm.

Extended Data Fig. 7 Spectral unmixing cannot resolve probes that can be separated via bioluminescent phasor.

HeLa cells were transfected with plasmids encoding either ReNL or LumiScarlet. (a) The transfected cells were plated across 96-well plates either as single populations (ReNL: 2.5×104 cells/well, LumiScarlet: 5×104 cells/well) or a mixed population (1.25×104 ReNL expressing cells and 2.5×104 LumiScarlet-expressing cells). (b) The samples were incubated with FRZ (50 µM) and photon outputs were measured on an IVIS imaging station with either no filter (open) or the conventional filters shown (GFP, DsRed, Cy5.5, ICG). Data are presented as mean values ± SEM of n = 3 biologically independent samples. (c) Images collected during the imaging session. (d) The cell mixtures were inseparable with a conventional bioluminescence spectral unmixing algorithm25. This experiment was repeated two times with similar results.

Extended Data Fig. 8 Single cell bioluminescence imaging.

(a) HeLa cells stably expressing CeNL were treated with FRZ (50 μM). Images were acquired using the microscope setup pictured in Supplementary Fig. 1 with a 20X air objective and 10 s/frame integration time. A total of 20 frames were collected and the phasor location was computed. Analysis of a single CeNL-expressing HeLa cell was sufficient for a robust phasor output. (b) CeNL-expressing HeLa cells were serially diluted and seeded in a 96-well plate (1×104 – 1×100 cells/well). Samples were incubated with FRZ and light emission was recorded using an IVIS detector. Data are presented as mean values ± SEM of n = 3 biologically independent samples. (c) Photon output of individual CeNL-expressing cells. Cells were seeded a in 96 well plate (5 cells/well) and allowed to adhere. The next day, the cells were washed with PBS (100 µL) and fresh media (100 µL) was added. Samples were incubated with FRZ and light emission was recorded. Signal was normalized to the number of cells observed in the well. Data are presented as mean values ± SEM of n = 3 biologically independent samples.

Extended Data Fig. 9 Subcellular bioluminescent phasor imaging.

HeLa cells were transiently transfected with plasmids encoding YeNL localized to (from left to right): the membrane (CD8LS-YeNL-CD4), actin (YeNL-actin), mitochondria (Cox8×2-YeNL), ER (Carl-YeNL-KDEL) or nucleus (YeNL-H2B). The cells were treated with FRZ (25–50 μM) ~36 h post transfection. Images were acquired using the microscope setup pictured in Supplementary Fig. 1 with a 60x oil objective and 10 s/frame integration time. A total of 20 frames were collected for each sample and the raw intensity images (a) were subjected to image processing to remove out-of-focus signal. The resulting structural-enhanced images are shown in (b) and phasors computed for each construct are shown in (c). The phasor location of cytosolic YeNL is shown in black for comparison. These experiments were repeated three times with similar results. Scale bars = 25 μm.

Extended Data Fig. 10 Simultaneous tracking of two subcellular features.

HeLa cells were transiently transfected with plasmids encoding YeNL localized to the nucleus (YeNL-H2B) and GeNL localized to the membrane (CD8LS-GeNL-CD4). The cells were treated with FRZ (25 µM) ~36 h post transfection. Images were acquired using the microscope setup pictured in Supplementary Fig. 1 with a 60x oil objective and 10 s/frame integration time. A total of 360 frames were collected and the raw intensity images from (a) were subjected to image processing to remove out-of-focus signal. The resulting structural-enhanced images are shown in (b). Images are representative of n = 3 independent experiments. Scale bars = 25 µm.

Supplementary information

Supplementary Information

Supplementary Table 1 and Figs. 1–6.

Reporting Summary

Supplementary Video 1

Longitudinal tracking of tumor spheroid via bioluminescent phasor. MDA-MB231 spheroids were formed with three cell lines, stably expressing CeNL, YeNL or LumiScarlet and embedded in collagen. The spheroids were treated with FRZ (50 µM) and images were acquired using the microscope setup pictured in Supplementary Fig. 1 with a ×10 objective and 10-s frame integration time. The sample was continuously imaged for 2.5 h (900 frames total). Selected snapshots are shown in Fig. 2. Scale bars, 100 µm.

Supplementary Video 2

Longitudinal multi-organelle tracking via bioluminescent phasor. HeLa cells were transiently transfected with constructs comprising nuclear-localized YeNL and membraned-localized GeNL. The cells were treated with FRZ (25–50 μM) ~36 h post transfection. Images were acquired using the microscope setup pictured in Supplementary Fig. 1 with a ×60 oil objective and integration time of 10 s per frame. The sample was continuously imaged for 1 h (360 frames total). Selected snapshots at 0, 15, 30 and 45 min are shown in Fig. 2.

Supplementary Software

Required content for software submission.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Yao, Z., Brennan, C.K., Scipioni, L. et al. Multiplexed bioluminescence microscopy via phasor analysis. Nat Methods 19, 893–898 (2022). https://doi.org/10.1038/s41592-022-01529-9

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41592-022-01529-9

  • Springer Nature America, Inc.

This article is cited by

Navigation