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.

  • Registered Report
  • Published:

Quantitative assessment of near-infrared fluorescent proteins

Abstract

Recent progress in fluorescent protein development has generated a large diversity of near-infrared fluorescent proteins (NIR FPs), which are rapidly becoming popular probes for a variety of imaging applications. However, the diversity of NIR FPs poses a challenge for end-users in choosing the optimal one for a given application. Here we conducted a systematic and quantitative assessment of intracellular brightness, photostability, oligomeric state, chemical stability and cytotoxicity of 22 NIR FPs in cultured mammalian cells and primary mouse neurons and identified a set of top-performing FPs including emiRFP670, miRFP680, miRFP713 and miRFP720, which can cover a majority of imaging applications. The top-performing proteins were further validated for in vivo imaging of neurons in Caenorhabditis elegans, zebrafish, and mice as well as in mice liver. We also assessed the applicability of the selected NIR FPs for multicolor imaging of fusions, expansion microscopy and two-photon imaging.

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: Quantitative characterization of NIR FPs in cultured mammalian cells.
Fig. 2: Quantitative characterization of the selected NIR FPs expressed in L2/3 cortical neurons in mouse brain tissue.
Fig. 3: Characterization of the selected NIR FPs in neurons in live larval zebrafish at 4 dpf.
Fig. 4: Characterization of the selected NIR FPs in neurons in live C.elegans.
Fig. 5: Multicolor imaging with NIR FPs.

Similar content being viewed by others

Data availability

The mass spectrometry proteomics data generated in this study have been deposited in the iProX database with the dataset identifier IPX0005767000 (https://www.iprox.cn//page/SCV017.html?query=IPX0005767000). The total size of the files acquired for this study was about 2 TB, which exceeds the limit of the FigShare repository, therefore only the most essential raw datasets, that is, the raw images with metadata supporting the results in Figs. 15, Extended Data Figs. 17 and Supplementary Figs. 2, 3, 5, 7, 9, 11 are available at FigShare (https://figshare.com/authors/Hanbin_Zhang/14524646 and https://doi.org/10.6084/m9.figshare.21975770). The rest of the files are available from the corresponding author upon request. Source data files are provided with this paper. All plasmids used in this study are available from Addgene and WeKwikGene (https://wekwikgene.wllsb.edu.cn/). Source data are provided with this paper.

Code availability

The custom MatLab code for analysis photostability data is available at Zenodo (https://doi.org/10.5281/zenodo.7992722).

References

  1. Prasher, D. C., Eckenrode, V. K., Ward, W. W., Prendergast, F. G. & Cormier, M. J. Primary structure of the Aequorea victoria green-fluorescent protein. Gene 111, 229–233 (1992).

    Article  CAS  PubMed  Google Scholar 

  2. Shcherbakova, D. M., Stepanenko, O. V., Turoverov, K. K. & Verkhusha, V. V. Near-infrared fluorescent proteins: multiplexing and optogenetics across scales. Trends Biotechnol. 36, 1230–1243 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  3. Filonov, G. S. et al. Bright and stable near-infrared fluorescent protein for in vivo imaging. Nat. Biotechnol. 29, 757–761 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  4. Chu, J. et al. Non-invasive intravital imaging of cellular differentiation with a bright red-excitable fluorescent protein. Nat. Methods 11, 572–578 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  5. Shcherbakova, D. M. & Verkhusha, V. V. Near-infrared fluorescent proteins for multicolor in vivo imaging. Nat. Methods 10, 751–754 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  6. Babakhanova, S. et al. Rapid directed molecular evolution of fluorescent proteins in mammalian cells. Protein Sci. 31, 728–751 (2022).

    Article  CAS  PubMed  Google Scholar 

  7. Qian, Y. et al. A genetically encoded near-infrared fluorescent calcium ion indicator. Nat. Methods 16, 171–174 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  8. Qian, Y. et al. Improved genetically encoded near-infrared fluorescent calcium ion indicators for in vivo imaging. PLoS Biol. 18, e3000965 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Shcherbakova, D. M., Cox Cammer, N., Huisman, T. M., Verkhusha, V. V. & Hodgson, L. Direct multiplex imaging and optogenetics of Rho GTPases enabled by near-infrared FRET. Nat. Chem. Biol. 14, 591–600 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Shcherbakova, D. M. et al. Bright monomeric near-infrared fluorescent proteins as tags and biosensors for multiscale imaging. Nat. Commun. 7, 12405 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Kamper, M., Ta, H., Jensen, N. A., Hell, S. W. & Jakobs, S. Near-infrared STED nanoscopy with an engineered bacterial phytochrome. Nat. Commun. 9, 4762 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  12. Wegner, W. et al. In vivo mouse and live cell STED microscopy of neuronal actin plasticity using far-red emitting fluorescent proteins. Sci. Rep. 7, 11781 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  13. Matlashov, M. E. et al. A set of monomeric near-infrared fluorescent proteins for multicolor imaging across scales. Nat. Commun. 11, 239 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  14. Piatkevich, K. D., Subach, F. V. & Verkhusha, V. V. Engineering of bacterial phytochromes for near-infrared imaging, sensing, and light-control in mammals. Chem. Soc. Rev. 42, 3441–3452 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Shcherbakova, D. M., Baloban, M. & Verkhusha, V. V. Near-infrared fluorescent proteins engineered from bacterial phytochromes. Curr. Opin. Chem. Biol. 27, 52–63 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  16. Piatkevich, K. D. et al. Near-infrared fluorescent proteins engineered from bacterial phytochromes in neuroimaging. Biophys. J. 113, 2299–2309 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Shemetov, A. A. et al. A near-infrared genetically encoded calcium indicator for in vivo imaging. Nat. Biotechnol. 39, 368–377 (2021).

    Article  CAS  PubMed  Google Scholar 

  18. Yu, D. et al. A naturally monomeric infrared fluorescent protein for protein labeling in vivo. Nat. Methods 12, 763–765 (2015).

    Article  PubMed  PubMed Central  Google Scholar 

  19. Paez-Segala, M. G. et al. Fixation-resistant photoactivatable fluorescent proteins for CLEM. Nat. Methods 12, 215–218 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Campbell, B. C., Paez-Segala, M. G., Looger, L. L., Petsko, G. A. & Liu, C. F. Chemically stable fluorescent proteins for advanced microscopy. Nat. Methods 19, 1612–1621 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  21. Tillberg, P. W. et al. Protein-retention expansion microscopy of cells and tissues labeled using standard fluorescent proteins and antibodies. Nat. Biotechnol. 34, 987–992 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Cranfill, P. J. et al. Quantitative assessment of fluorescent proteins. Nat. Methods 13, 557–562 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  23. Costantini, L. M., Fossati, M., Francolini, M. & Snapp, E. L. Assessing the tendency of fluorescent proteins to oligomerize under physiologic conditions. Traffic 13, 643–649 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  24. Li, X. D. et al. Design of small monomeric and highly bright near-infrared fluorescent proteins. Biochim. Biophys. Acta Mol. Cell Res. 1866, 1608–1617 (2019).

    Article  CAS  PubMed  Google Scholar 

  25. Shaner, N. C. et al. A bright monomeric green fluorescent protein derived from Branchiostoma lanceolatum. Nat. Methods 10, 407–409 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  26. Day, R. N. & Davidson, M. W. The fluorescent protein palette: tools for cellular imaging. Chem. Soc. Rev. 38, 2887–2921 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  27. Drobizhev, M., Makarov, N. S., Tillo, S. E., Hughes, T. E. & Rebane, A. Two-photon absorption properties of fluorescent proteins. Nat. Methods 8, 393–399 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  28. Piatkevich, K. D. et al. A robotic multidimensional directed evolution approach applied to fluorescent voltage reporters. Nat. Chem. Biol. 14, 352–360 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Mikhaylov, A. et al. Hot-band absorption can mimic entangled two-photon absorption. J. Phys. Chem. Lett. 13, 1489–1493 (2022).

    Article  CAS  PubMed  Google Scholar 

  30. Babakhanova, S. et al. Rapid directed molecular evolution of fluorescent proteins in mammalian cells. Protein Sci. 31, 728–751 (2022).

    Article  CAS  PubMed  Google Scholar 

  31. Holowiecki, A., O’Shields, B. & Jenny, M. J. Characterization of heme oxygenase and biliverdin reductase gene expression in zebrafish (Danio rerio): basal expression and response to pro-oxidant exposures. Toxicol. Appl. Pharmacol. 311, 74–87 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Qian, Y. et al. Improved genetically encoded near-infrared fluorescent calcium ion indicators for in vivo imaging. PLOS Biol. 18, e3000965 (2020).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. Seo, J. et al. PICASSO allows ultra-multiplexed fluorescence imaging of spatially overlapping proteins without reference spectra measurements. Nat. Commun. 13, 2475 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  34. Chiang, H. J. et al. HyU: Hybrid Unmixing for longitudinal in vivo imaging of low signal-to-noise fluorescence. Nat. Methods 20, 248–258 (2023).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Stepanova, T. et al. Visualization of microtubule growth in cultured neurons via the use of EB3-GFP (end-binding protein 3-green fluorescent protein). J. Neurosci. 23, 2655–2664 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  36. Riedl, J. et al. Lifeact: a versatile marker to visualize F-actin. Nat. Methods 5, 605–607 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  37. Kimura, S., Noda, T. & Yoshimori, T. Dissection of the autophagosome maturation process by a novel reporter protein, tandem fluorescent-tagged LC3. Autophagy 3, 452–460 (2007).

    Article  CAS  PubMed  Google Scholar 

  38. Namikawa, K. et al. Modeling neurodegenerative spinocerebellar ataxia type 13 in zebrafish using a Purkinje neuron specific tunable coexpression system. J. Neurosci. 39, 3948–3969 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Rodrik-Outmezguine, V. S. et al. Overcoming mTOR resistance mutations with a new-generation mTOR inhibitor. Nature 534, 272–276 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Meyer, M. P. & Smith, S. J. Evidence from in vivo imaging that synaptogenesis guides the growth and branching of axonal arbors by two distinct mechanisms. J. Neurosci. 26, 3604–3614 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  41. Niell, C. M., Meyer, M. P. & Smith, S. J. In vivo imaging of synapse formation on a growing dendritic arbor. Nat. Neurosci. 7, 254–260 (2004).

    Article  CAS  PubMed  Google Scholar 

  42. Li, L. et al. Overexpression of heme oxygenase 1 impairs cognitive ability and changes the plasticity of the synapse. J. Alzheimers Dis. 47, 595–608 (2015).

    Article  CAS  PubMed  Google Scholar 

  43. Papadaki, S. et al. Dual-expression system for blue fluorescent protein optimization. Sci. Rep. 12, 10190 (2022).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Drobizhev, M. et al. Local electric field controls fluorescence quantum yield of red and far-red fluorescent proteins. Front. Mol. Biosci. 8, 633217 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  45. Luchowski, R. et al. Instrument response standard in time-resolved fluorescence. Rev. Sci. Instrum. 80, 033109 (2009).

    Article  PubMed  Google Scholar 

  46. Canty, L., Hariharan, S., Liu, Q., Haney, S. A. & Andrews, D. W. Peak emission wavelength and fluorescence lifetime are coupled in far-red, GFP-like fluorescent proteins. PLoS One 13, e0208075 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  47. Drobizhev, M., Molina, R. S. & Hughes, T. E. Characterizing the two-photon absorption properties of fluorescent molecules in the 680–1300 nm spectral range. Bio Protoc. 10(2), e3498 (2020).

    CAS  PubMed  PubMed Central  Google Scholar 

  48. Kim, J.-Y., Grunke, S. D., Levites, Y., Golde, T. E. & Jankowsky, J. L. Intracerebroventricular viral injection of the neonatal mouse brain for persistent and widespread neuronal transduction. J. Vis. Exp. 15(91), 51863 (2014).

    Google Scholar 

  49. Shu, X. et al. Mammalian expression of infrared fluorescent proteins engineered from a bacterial phytochrome. Science 324, 804–807 (2009).

    Article  PubMed  PubMed Central  Google Scholar 

  50. Westerfield, M. The Zebrafish Book. A Guide for the Laboratory Use of Zebrafish (Danio rerio), 5th edn (Univ. Oregon Press, 2007).

  51. Urasaki, A., Morvan, G. & Kawakami, K. Functional dissection of the Tol2 transposable element identified the minimal cis-sequence and a highly repetitive sequence in the subterminal region essential for transposition. Genetics 174, 639–649 (2006).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Mishima, Y. et al. Zebrafish miR-1 and miR-133 shape muscle gene expression and regulate sarcomeric actin organization. Genes Dev. 23, 619–632 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Ishikawa, K., Sato, M., Ito, M. & Yoshida, T. Importance of histidine residue 25 of rat heme oxygenase for its catalytic activity. Biochem. Biophys. Res. Commun. 182, 981–986 (1992).

    Article  CAS  PubMed  Google Scholar 

  54. Green, R. A. et al. Expression and imaging of fluorescent proteins in the C. elegans gonad and early embryo. Methods Cell Biol. 85, 179–218 (2008).

    Article  CAS  PubMed  Google Scholar 

  55. Baraban, M., Anselme, I., Schneider-Maunoury, S. & Giudicelli, F. Zebrafish embryonic neurons transport messenger RNA to axons and growth cones in vivo. J. Neurosci. 33, 15726–15734 (2013).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  56. Strack, R. L. et al. A rapidly maturing far-red derivative of DsRed-Express2 for whole-cell labeling. Biochemistry 48, 8279–8281 (2009).

    Article  CAS  PubMed  Google Scholar 

  57. Rodriguez, E. A. et al. A far-red fluorescent protein evolved from a cyanobacterial phycobiliprotein. Nat. Methods 13, 763–769 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Ding, W. L. et al. Far-red acclimating cyanobacterium as versatile source for bright fluorescent biomarkers. Biochim. Biophys. Acta Mol. Cell Res. 1865, 1649–1656 (2018).

    Article  CAS  PubMed  Google Scholar 

  59. Yu, D. et al. An improved monomeric infrared fluorescent protein for neuronal and tumour brain imaging. Nat. Commun. 5, 3626 (2014).

    Article  CAS  PubMed  Google Scholar 

  60. Rogers, O. C., Johnson, D. M. & Firnberg, E. mRhubarb: engineering of monomeric, red-shifted, and brighter variants of iRFP using structure-guided multi-site mutagenesis. Sci. Rep. 9, 15653 (2019).

    Article  PubMed  PubMed Central  Google Scholar 

Download references

Acknowledgements

The authors thank J. Pan, J. Hu, X. Bai and the Mass Spectrometry and Metabolomics Core Facility of Westlake University for protein sample mass spectrometry analysis. The authors also thank Z. Dong from Westlake University for help with the interpretation of the mass spectrometry results; the Laboratory Animal Resources Center, the Flow Cytometry Core Facility and the Microscopy Core Facility in Westlake University. Henrietta Lacks, and the HeLa cell line that was established from her tumor cells without her knowledge or consent in 1951, have made significant contributions to scientific progress and advances in human health. We are grateful to Henrietta Lacks, now deceased, and to her surviving family members for their contributions to biomedical research. This work is supported by start-up funding from the Foundation of Westlake University, National Natural Science Foundation of China grant 32050410298 and 32171093, 2020 BBRF Young Investigator Grant 28961, and MRIC Funding 103536022023 to K.D.P., the German Research Foundation (DFG, K1949/7-2) project 241961032 and VW-Vorab des Landes Niedersachsen (project HomeoHirn(ZN3673)) to R.W.K., the Foundation of Westlake University and the National Natural Science Foundation of China grant 32071151 to L.W., and the NIH BRAIN program U24NS109107 grant to M.D.

Author information

Authors and Affiliations

Authors

Contributions

H.Z. and S.P. performed characterization in cultured mammalian cells. X.S. characterized proteins in the expansion microscopy protocols. M.D. and H.Z. characterized proteins in vitro. H.Z. and X.S. performed the mouse experiments, X.W., M.R., R.W.K. and K.N. performed the zebrafish experiments, S.P., L.Y. and L.W. performed the C.elegans experiments. H.Z., S.P. and K.D.P. analyzed the data and wrote the manuscript with input from all of the authors. K.D.P. designed and oversaw all aspects of the project.

Corresponding author

Correspondence to Kiryl D. Piatkevich.

Ethics declarations

Competing interests

K.D.P. is the co-founder of a company that pursues commercial applications of expansion microscopy and is listed as an inventor on several patent applications concerning development of new expansion microscopy methods. All other authors have no competing interests.

Peer review

Peer review information

Nature Methods thanks Erik Snapp, Lin Tian, and the other, anonymous, reviewer 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 Quantitative assessment of intracellular brightness and photostability of NIR FPs in live HEK cells.

(a) Intracellular normalized brightness of NIR FPs imaged in three channels: (i) Cy5-LP, (ii) Cy5-BP and (iii) Cy5.5 under 55 mW/mm2 excitation power (n > 2000 cells for each NIR-FP from three independent transfections in each channel). Brightness for each FP was normalized to the EGFP signal (here and in panel e). Asterisks (*) indicate signal-to-background ratio <2.0 throughout; in panels a-c box plots highlighted with brown correspond to top 5 NIR FPs for each panel. (b) Photobleaching half-times of NIR-FPs under Cy5 (i and ii, excitation power 58 mW/mm2) and Cy5.5 (iii, excitation power 56–58 mW/mm2; n > 93 cells for each NIR-FP from four independent transfections). (c) Product of mean brightness and mean half-time fluorescence presented as bar graph (mean ± SEM) (due to small value of SEM the error bars are not visible for some bars). (d) Normalized photobleaching curves of NIR FPs under (i) Cy5 (excitation power 58 mW/mm2) and (ii) Cy5.5 (excitation power 56–58 mW/mm2) illumination for the results shown in panel b (n > 93 cells for each NIR-FP from four independent transfections in each channel). Fluorescence was normalized to the intensity value of corresponding FP at t = 0 s. NIR FPs that exhibited signal-to-background ratio lower than 2.0 in Cy5.5 channel are not shown in the graph, however the corresponding curves are available in the source datasets except for mNeptune2.5, which was not visible in Cy5.5 channel at all and thus was not measured. (e) Side-by-side normalized brightness comparison without and with BV administration imaged in (i) Cy5-LP, (ii) Cy5-BP and (iii) Cy5.5 under 55 mW/mm2 excitation power (n > 2000 cells for each NIR-FP from three independent transfections in each channel). Outliers not shown but included in all calculations and available in the source datasets. See Supplementary Table 2 and Supplementary Dataset 1 for the detailed descriptive statistics and exact p-values.

Source data

Extended Data Fig. 2 Quantitative assessment of intracellular brightness and photostability NIR FPs in live primary cultured mouse neurons.

(a, b) Intracellular normalized brightness of NIR FPs imaged after (a) Calcium Phosphate transfection and (b) viral transduction in Cy5-LP, (ii) Cy5-BP and (iii) Cy5.5 under 55 mW/mm2 excitation power (n > 40 neurons for each NIR FP from two independent cultures). Brightness for each FP was normalized to the EGFP signal (here and in panel f). In panels a-d box plots highlighted with brown correspond to top 5 NIR FPs for each panel. (c) Photobleaching half-times of NIR FPs under Cy5 (i and ii, excitation power 55 mW/mm2) and Cy5.5 (iii, excitation power 55 mW/mm2; n > 42 neurons for each NIR FP from two independent cultures). NA – not applicable due to low fluorescence signal. (d) Product of mean brightness and mean half-time fluorescence presented in bar graph (mean ± SEM) (due to small value of SEM the error bars are not visible for some bars). (e) Normalized photobleaching curves of NIR-FPs under (i) Cy5 (excitation power 55 mW/mm2) and (ii) Cy5.5 (excitation power 55 mW/mm2) illumination for the results shown in c. Fluorescence was normalized to the intensity value of corresponding FP at t = 0 s. (f) Side-by-side normalized brightness comparison before and after BV administration imaged in (i) Cy5-LP, (ii) Cy5-BP and (iii) Cy5.5 under 55 mW/mm2 power (n > 20 neurons for each NIR FP from two independent cultures). see Supplementary Dataset 1 for the detailed descriptive statistics and exact p-values.

Source data

Extended Data Fig. 3 Quantitative characterization of NIR FPs in fixed HEK cells.

(a) NIR-to-green fluorescence ratio in fixed HEK cells imaged in (i) Cy5-LP, (ii) Cy5-BP and (iii) Cy5.5 under 55 mW/mm2 excitation power (n > 1976 cells for each NIR FP from two independent transfections in each channel). Brightness for each FP was normalized to the EGFP signal. Asterisks (*) indicate signal-to-background ratio <2.0 throughout the figure based on brightness of fixed HEK cells; in panels a, c, d box plots highlighted with brown correspond to top 5 NIR FPs for each panel. (b) Normalized photobleaching curves of NIR FPs under (i) Cy5 (excitation power 55 mW/mm2) and (ii) Cy5.5 (excitation power 55 mW/mm2) illumination in fixed HEK cells (n = 40 cells for each NIR FP from two independent transfections in each channel). Fluorescence was normalized to the intensity value of corresponding FP at t = 0 s. NIR FPs that exhibited lower than 2.0 signal-to-background ratio in Cy5.5 channel are not shown in the graph; however, the corresponding curves are available in the source datasets. (c) The products of mean NIR-to-green fluorescence ratio and mean fluorescence half-time in (i) Cy5-LP and (ii) Cy5.5 are presented in the form of bar graph (mean ± SE). (d) The products of mean absolute NIR brightness to mean fluorescence half-time in (i) Cy5-LP and (ii) Cy5.5 are presented in the form of bar graph (mean ± SE). (e) Comparison of selected NIR FPs in proExM. (e) Representative images of NIR FPs in PFA-fixed and proExM treated HEK cells acquired in Cy5-LP (NIR FP) and FITC (GFP) channels (n > 61 cells for each NIR FP from one independent transfection). The dynamic brightness range was adjusted independently to facilitate visualization of transfected cells in fixed and proExM processes for each NIR FPs. Images were obtained through single-plane scanning and without projection processing. (f) Fluorescence retention of NIR FPs in HEK cells upon PFA fixation presented as bar graph (mean of NIR retention for each protein) (Cy5-LP, n > 1974 cells for each NIR FP from two independent transfections). Black dots indicate ratio of mean brightness before and after fixation for two transfections. (g) Absolute NIR fluorescence of fixed (brown box plots with notches) and proExM treated (orange box plots with notches) HEK cells (n > 61 cells for each NIR FP from one independent transfection). (h) Retention of GFP and NIR FPs’ fluorescence in fixed HEK cells after proExM treatment (n > 61 cells for each NIR FP from one independent transfection). See Supplementary Table 2 and Supplementary Dataset 1 for the detailed descriptive statistics and exact p-values.

Source data

Extended Data Fig. 4 Fluorescence imaging of NIR FPs fusions in live HeLa cells.

Representative images of (from top left) iRFP670-actin, miRFP670-2-actin, miRFP680-actin, miRFP713-actin, miRFP720-actin, H2B-iRFP670, H2B-miRFP670-2, H2B-miRFP680, H2B-miRFP713, H2B-miRFP720, Cx43-iRFP670, Cx43-miRFP670-2, Cx43-miRFP680, Cx43-miRFP713, Cx43-miRFP720, keratin-iRFP670, keratin-miRFP670-2, keratin-miRFP680, keratin-miRFP713, keratin-miRFP720, iRFP670-tubulin, miRFP670-2-tubulin, miRFP680-tubulin, miRFP713-tubulin, miRFP720-tubulin (n > 15 cells for each construct from two independent transfection). For each image the dynamic range was adjusted independently to facilitate visualization and images were generated through maximum projection.

Extended Data Fig. 5 Two-photon cross-section spectra of selected NIR-FPs.

Two-photon cross-section spectra of mCardinal, emiRFP670, miRFP670-2, miRFP680, mRhubarb713, miRFP713, iRFP713, and miRFP720 presented versus laser wavelength used for excitation (GM, Goeppert-Mayer units).

Source data

Extended Data Fig. 6 Quantitative characterization of the selected NIR FPs expressed in L2/3 cortical neurons in mouse brain tissue.

(a, b, c) Intracellular normalized brightness of NIR FPs imaged in acute brain slices from (a) one-month, (b) two-month, (c) three-month old mice in (i) Cy5-LP, (ii) Cy5-BP, (iii) Cy5.5 channels (excitation power 55 mW/mm2; n ≥ 100 neurons from 3 mice for each protein in each channel). Brightness for each FP was normalized to the EGFP signal (here and in panel d). (d) Intracellular normalized brightness of NIR-FPs imaged in PFA-fixed brain slices from one-month old mice in (i) Cy5-LP, (ii) Cy5-BP, (ii) Cy5.5 (excitation power 55 mW/mm2; n > 90 neurons from 2 mice for each protein in each channel). (e, f) Normalized photobleaching curves of NIR-FPs measured in (e) acute brain slices and (f) PFA-fixed brain slices in (i) Cy5-LP and (ii) Cy5.5 channels (excitation power 55 mW/mm2; n > 40 neurons from 2 mice for each protein for each channel). Fluorescence was normalized to the intensity value of corresponding FP at t = 0 s. See Supplementary Dataset 1 for the detailed descriptive statistics and exact p-values.

Source data

Extended Data Fig. 7 Quantitative characterization of the selected NIR FPs expressed in mouse liver in vivo.

((a) Representative wide-field fluorescence images of NIR FPs expressed in mouse liver acquired in Cy5-LP, Cy5-BP, Cy5.5, and FITC channels (excitation power 55 mW/mm2 for Cy5 and Cy5.5 channels; n = 3 liver slices from 3-4 mice for each protein). (b,c,d) Intracellular normalized brightness of NIR FPs imaged in fresh liver tissue from three-month-old mice in (b) Cy5-LP, (c) Cy5-BP, (d) Cy5.5 channels (excitation power 55 mW/mm2; n > 154 cells from 3-4 mice for each protein in each channel). Brightness for each FP was normalized to the EGFP signal. See Supplementary Dataset 1 for the detailed descriptive statistics and exact p-values.

Source data

Extended Data Table 1 List of the NIR FPs selected for quantitative assessment in cultured cells56,57,58,59
Extended Data Table 2 Spectroscopic and biochemical properties of the selected NIR FPs measured in this study

Supplementary information

Supplementary Information

Supplementary Notes 1 and 2, Tables 1–11 and Figs. 1–21.

Reporting Summary

Supplementary Video 1

The dynamic protrusions in the growth cone are visualized using Lifeact-mClover3 marking F-actin, whereas EB3-emiRFP670 labels microtubule plus-ends moving towards the tip of the growth cone dominantly. Elapsed time in min : s. Scale bar, 5 µm.

Supplementary Dataset 1

Statistical analysis of the data in this paper

Supplementary Dataset 2

Oligonucleotide sequences

Source data

Rights and permissions

Springer Nature or its licensor (e.g. a society or other partner) holds exclusive rights to this article under a publishing agreement with the author(s) or other rightsholder(s); author self-archiving of the accepted manuscript version of this article is solely governed by the terms of such publishing agreement and applicable law.

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zhang, H., Papadaki, S., Sun, X. et al. Quantitative assessment of near-infrared fluorescent proteins. Nat Methods 20, 1605–1616 (2023). https://doi.org/10.1038/s41592-023-01975-z

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41592-023-01975-z

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