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:

Analysis of redox landscapes and dynamics in living cells and in vivo using genetically encoded fluorescent sensors

Abstract

Cellular oxidation–reduction reactions are mainly regulated by pyridine nucleotides (NADPH/NADP+ and NADH/NAD+), thiols, and reactive oxygen species (ROS) and play central roles in cell metabolism, cellular signaling, and cell-fate decisions. A comprehensive evaluation or multiplex analysis of redox landscapes and dynamics in intact living cells is important for interrogating cell functions in both healthy and disease states; however, until recently, this goal has been limited by the lack of a complete set of redox sensors. We recently reported the development of a series of highly responsive, genetically encoded fluorescent sensors for NADPH that substantially strengthen the existing toolset of genetically encoded sensors for thiols, H2O2, and NADH redox states. By combining sensors with unique spectral properties and specific subcellular targeting domains, our approach allows simultaneous imaging of up to four different sensors. In this protocol, we first describe strategies for multiplex fluorescence imaging of these sensors in single cells; then we demonstrate how to apply these sensors to study changes in redox landscapes during the cell cycle, after macrophage activation, and in living zebrafish. This approach can be adapted to different genetically encoded fluorescent sensors and various analytical platforms such as fluorescence microscopy, high-content imaging systems, flow cytometry, and microplate readers. A typical preparation of cells or zebrafish expressing different sensors takes 2–3 d; microscopy imaging or flow-cytometry analysis can be performed within 5–60 min.

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: Simplified schematic of intracellular redox-buffering systems.
Fig. 2: Properties of iNap1–4 sensors.
Fig. 3: Three-parametric imaging of NADPH, NADH, and thiol redox state in single cells.
Fig. 4: Four-parametric imaging of NADPH, NADH, thiol redox, and H2O2 in single cells.
Fig. 5: Real-time monitoring of NADPH, NADH, thiol redox, and H2O2 dynamics during the cell cycle.
Fig. 6: Overview of flow-cytometry analysis.
Fig. 7: Overview of zebrafish imaging.
Fig. 8: Comprehensive analysis of cytosolic and mitochondrial redox state in resting and activated mouse macrophages by flow cytometry.
Fig. 9: Spatiotemporal imaging of NADPH, NADH, and H2O2 in living zebrafish.

Similar content being viewed by others

References

  1. Zhao, Y., Zhang, Z., Zou, Y. & Yang, Y. Visualization of nicotine adenine dinucleotide redox homeostasis with genetically encoded fluorescent sensors. Antioxid. Redox Signal. 28, 213–229 (2018).

    Article  CAS  Google Scholar 

  2. Fan, J. et al. Quantitative flux analysis reveals folate-dependent NADPH production. Nature 510, 298–302 (2014).

    Article  CAS  Google Scholar 

  3. Cairns, R. A., Harris, I. S. & Mak, T. W. Regulation of cancer cell metabolism. Nat. Rev. Cancer 11, 85–95 (2011).

    Article  CAS  Google Scholar 

  4. Gorrini, C., Harris, I. S. & Mak, T. W. Modulation of oxidative stress as an anticancer strategy. Nat. Rev. Drug Discov. 12, 931–947 (2013).

    Article  CAS  Google Scholar 

  5. Ying, W. NAD+/NADH and NADP+/NADPH in cellular functions and cell death: regulation and biological consequences. Antioxid. Redox Signal. 10, 179–206 (2008).

    Article  CAS  Google Scholar 

  6. Blacker, T. S. et al. Separating NADH and NADPH fluorescence in live cells and tissues using FLIM. Nat. Commun. 5, 3936 (2014).

    Article  CAS  Google Scholar 

  7. Maddocks, O. D., Labuschagne, C. F. & Vousden, K. H. Localization of NADPH production: a wheel within a wheel. Mol. Cell 55, 158–160 (2014).

    Article  CAS  Google Scholar 

  8. Tao, R. et al. Genetically encoded fluorescent sensors reveal dynamic regulation of NADPH metabolism. Nat. Methods 14, 720–728 (2017).

    Article  CAS  Google Scholar 

  9. Veech, R. L., Eggleston, L. V. & Krebs, H. A. The redox state of free nicotinamide-adenine dinucleotide phosphate in the cytoplasm of rat liver. Biochem. J. 115, 609–619 (1969).

    Article  CAS  Google Scholar 

  10. Canto, C. et al. AMPK regulates energy expenditure by modulating NAD+ metabolism and SIRT1 activity. Nature 458, 1056–1060 (2009).

    Article  CAS  Google Scholar 

  11. Wise, D. D. & Shear, J. B. Tracking variations in nicotinamide cofactors extracted from cultured cells using capillary electrophoresis with multiphoton excitation of fluorescence. Anal. Biochem. 326, 225–233 (2004).

    Article  CAS  Google Scholar 

  12. Lewis, C. A. et al. Tracing compartmentalized NADPH metabolism in the cytosol and mitochondria of mammalian cells. Mol. Cell 55, 253–263 (2014).

    Article  CAS  Google Scholar 

  13. Quinn, K. P. et al. Quantitative metabolic imaging using endogenous fluorescence to detect stem cell differentiation. Sci. Rep. 3, 3432 (2013).

    Article  Google Scholar 

  14. Hanson, G. T. et al. Investigating mitochondrial redox potential with redox-sensitive green fluorescent protein indicators. J. Biol. Chem. 279, 13044–13053 (2004).

    Article  CAS  Google Scholar 

  15. Dooley, C. T. et al. Imaging dynamic redox changes in mammalian cells with green fluorescent protein indicators. J. Biol. Chem. 279, 22284–22293 (2004).

    Article  CAS  Google Scholar 

  16. Gutscher, M. et al. Real-time imaging of the intracellular glutathione redox potential. Nat. Methods 5, 553–559 (2008).

    Article  CAS  Google Scholar 

  17. Fan, Y., Chen, Z. & Ai, H. W. Monitoring redox dynamics in living cells with a redox-sensitive red fluorescent protein. Anal. Chem. 87, 2802–2810 (2015).

    Article  CAS  Google Scholar 

  18. Belousov, V. V. et al. Genetically encoded fluorescent indicator for intracellular hydrogen peroxide. Nat. Methods 3, 281–286 (2006).

    Article  CAS  Google Scholar 

  19. Ermakova, Y. G. et al. Red fluorescent genetically encoded indicator for intracellular hydrogen peroxide. Nat. Commun. 5, 5222 (2014).

    Article  CAS  Google Scholar 

  20. Albrecht, S. C., Barata, A. G., Grosshans, J., Teleman, A. A. & Dick, T. P. In vivo mapping of hydrogen peroxide and oxidized glutathione reveals chemical and regional specificity of redox homeostasis. Cell Metab. 14, 819–829 (2011).

    Article  CAS  Google Scholar 

  21. Morgan, B. et al. Real-time monitoring of basal H2O2 levels with peroxiredoxin-based probes. Nat. Chem. Biol. 12, 437–443 (2016).

    Article  CAS  Google Scholar 

  22. Zhao, Y. et al. SoNar, a highly responsive NAD+/NADH sensor, allows high-throughput metabolic screening of anti-tumor agents. Cell Metab. 21, 777–789 (2015).

    Article  CAS  Google Scholar 

  23. Zhao, Y. et al. In vivo monitoring of cellular energy metabolism using SoNar, a highly responsive sensor for NAD(+)/NADH redox state. Nat. Protoc. 11, 1345–1359 (2016).

    Article  CAS  Google Scholar 

  24. Zhao, Y. et al. Genetically encoded fluorescent sensors for intracellular NADH detection. Cell Metab. 14, 555–566 (2011).

    Article  CAS  Google Scholar 

  25. Wiederkehr, A. & Demaurex, N. Illuminating redox biology using NADH- and NADPH-specific sensors. Nat. Methods 14, 671–672 (2017).

    Article  CAS  Google Scholar 

  26. Zhao, Y. & Yang, Y. Profiling metabolic states with genetically encoded fluorescent biosensors for NADH. Curr. Opin. Biotechnol. 31C, 86–92 (2015).

    Article  Google Scholar 

  27. 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 

  28. Bilan, D. S. et al. Genetically encoded fluorescent indicator for imaging NAD(+)/NADH ratio changes in different cellular compartments. Biochim. Biophys. Acta 1840, 951–957 (2014).

    Article  CAS  Google Scholar 

  29. Zhao, Y. & Yang, Y. Real-time and high-throughput analysis of mitochondrial metabolic states in living cells using genetically encoded NAD+/NADH sensors. Free Radic. Biol. Med. 100, 43–52 (2016).

    Article  CAS  Google Scholar 

  30. Zhang, J. et al. Determination of the cytosolic NADPH/NADP ratio in Saccharomyces cerevisiae using shikimate dehydrogenase as sensor reaction. Sci. Rep. 5, 12846 (2015).

    Article  CAS  Google Scholar 

  31. Hedeskov, C. J., Capito, K. & Thams, P. Cytosolic ratios of free [NADPH]/[NADP+] and [NADH]/[NAD+] in mouse pancreatic islets, and nutrient-induced insulin secretion. Biochem. J. 241, 161–167 (1987).

    Article  CAS  Google Scholar 

  32. Zhang, Q., Piston, D. W. & Goodman, R. H. Regulation of corepressor function by nuclear NADH. Science 295, 1895–1897 (2002).

    CAS  PubMed  Google Scholar 

  33. Mayevsky, A. & Rogatsky, G. G. Mitochondrial function in vivo evaluated by NADH fluorescence: from animal models to human studies. Am. J. Physiol. Cell Physiol. 292, C615–C640 (2007).

    Article  CAS  Google Scholar 

  34. Murphy, M. P. et al. Unraveling the biological roles of reactive oxygen species. Cell Metab. 13, 361–366 (2011).

    Article  CAS  Google Scholar 

  35. Pollak, N., Niere, M. & Ziegler, M. NAD kinase levels control the NADPH concentration in human cells. J. Biol. Chem. 282, 33562–33571 (2007).

    Article  CAS  Google Scholar 

  36. Ohashi, K., Kawai, S. & Murata, K. Identification and characterization of a human mitochondrial NAD kinase. Nat. Commun. 3, 1248 (2012).

    Article  Google Scholar 

  37. Cambronne, X. A. et al. Biosensor reveals multiple sources for mitochondrial NAD(+). Science 352, 1474–1477 (2016).

    Article  CAS  Google Scholar 

  38. Cameron, W. D. et al. Apollo-NADP(+): a spectrally tunable family of genetically encoded sensors for NADP(+). Nat. Methods 13, 352–358 (2016).

    Article  Google Scholar 

  39. Hoek, J. B. & Rydstrom, J. Physiological roles of nicotinamide nucleotide transhydrogenase. Biochem. J. 254, 1–10 (1988).

    Article  CAS  Google Scholar 

  40. Zhang, X., Edwards, J. P. & Mosser, D. M. The expression of exogenous genes in macrophages: obstacles and opportunities. Methods Mol. Biol. 531, 123–143 (2009).

    Article  CAS  Google Scholar 

  41. Ghesquiere, B., Wong, B. W., Kuchnio, A. & Carmeliet, P. Metabolism of stromal and immune cells in health and disease. Nature 511, 167–176 (2014).

    Article  CAS  Google Scholar 

  42. Karlsson, J., von Hofsten, J. & Olsson, P.E. Generating transparent zebrafish: a refined method to improve detection of gene expression during embryonic development. Mar. Biotechnol. (NY) 3, 522–527 (2001).

    Article  CAS  Google Scholar 

  43. Villalobos, A., Ness, J. E., Gustafsson, C., Minshull, J. & Govindarajan, S. Gene Designer: a synthetic biology tool for constructing artificial DNA segments. BMC Bioinformatics 7, 285 (2006).

    Article  Google Scholar 

  44. Schwartz, M. A., Schaller, M. D. & Ginsberg, M. H. Integrins: emerging paradigms of signal transduction. Annu. Rev. Cell Dev. Biol. 11, 549–599 (1995).

    Article  CAS  Google Scholar 

  45. Hynes, R. O. Integrins: versatility, modulation, and signaling in cell adhesion. Cell 69, 11–25 (1992).

    Article  CAS  Google Scholar 

  46. Tiscornia, G., Singer, O. & Verma, I. M. Production and purification of lentiviral vectors. Nat. Protoc. 1, 241–245 (2006).

    Article  CAS  Google Scholar 

  47. Birsoy, K. et al. An essential role of the mitochondrial electron transport chain in cell proliferation is to enable aspartate synthesis. Cell 162, 540–551 (2015).

    Article  CAS  Google Scholar 

  48. Qiling, X. in Molecular Methods in Developmental Biology Vol. 127 (ed. Guille, M.) 125–132 (Humana Press, Totowa, NJ, 1999).

  49. Kelsh, R. N. et al. Zebrafish pigmentation mutations and the processes of neural crest development. Development 123, 369–389 (1996).

    CAS  PubMed  Google Scholar 

  50. Inaba, M., Yamanaka, H. & Kondo, S. Pigment pattern formation by contact-dependent depolarization. Science 335, 677 (2012).

    Article  CAS  Google Scholar 

  51. Ma, X. et al. Polo-like kinase 1 coordinates biosynthesis during cell cycle progression by directly activating pentose phosphate pathway. Nat. Commun. 8, 1506 (2017).

    Article  Google Scholar 

  52. Noda, T. & Amano, F. Differences in nitric oxide synthase activity in a macrophage-like cell line, RAW264.7 cells, treated with lipopolysaccharide (LPS) in the presence or absence of interferon-gamma (IFN-gamma): possible heterogeneity of iNOS activity. J. Biochem. 121, 38–46 (1997).

    Article  CAS  Google Scholar 

  53. Lohman, J. R. & Remington, S. J. Development of a family of redox-sensitive green fluorescent protein indicators for use in relatively oxidizing subcellular environments. Biochemistry 47, 8678–8688 (2008).

    Article  CAS  Google Scholar 

  54. Fan, Y. & Ai, H. W. Development of redox-sensitive red fluorescent proteins for imaging redox dynamics in cellular compartments. Anal. Bioanal. Chem. 408, 2901–2911 (2016).

    Article  CAS  Google Scholar 

  55. Ostergaard, H., Henriksen, A., Hansen, F. G. & Winther, J. R. Shedding light on disulfide bond formation: engineering a redox switch in green fluorescent protein. EMBO J. 20, 5853–5862 (2001).

    Article  CAS  Google Scholar 

  56. Markvicheva, K. N. et al. A genetically encoded sensor for H2O2 with expanded dynamic range. Bioorg. Med. Chem. 19, 1079–1084 (2011).

    Article  CAS  Google Scholar 

  57. Bilan, D. S. et al. HyPer-3: a genetically encoded H2O2 probe with improved performance for ratiometric and fluorescence lifetime imaging. ACS Chem. Biol. 8, 535–542 (2013).

    Article  CAS  Google Scholar 

  58. Williamson, D. H., Lund, P. & Krebs, H. A. The redox state of free nicotinamide-adenine dinucleotide in the cytoplasm and mitochondria of rat liver. Biochem. J. 103, 514–527 (1967).

    Article  CAS  Google Scholar 

Download references

Acknowledgements

We thank S.J. Remington for the roGFP1 vector; V.V. Belousov for the HyPer and HyPerRed vectors; J. Du for the pTol2 vector; J. Yi for the psPAX2 and pMD2.G vectors; N. Su, L. Huang, Q. Wang, P. Ni, and H. Zi for technical assistance; and S.C. Tribuna for secretarial assistance. This research was supported by the National Key Research and Development Program of China (2017YFC0906900, 2017YFA050400, 2016YFA0100602, and 2017YFA0103302), the NSFC (31722033, 91649123, 31671484, 31225008, 31470833, 91749203, 81525010, and 81420108017), the Shanghai Science and Technology Commission (14XD1401400, 16430723100, and 15YF1402600), the Young Elite Scientists Sponsorship Program by Cast, Shanghai Young Top-notch Talent, the State Key Laboratory of Bioreactor Engineering, the Fundamental Research Funds for the Central Universities, the US National Institutes of Health (HL061795, HG007690, and GM107618 to J.L.), and the American Heart Association (D700382 to J.L.).

Author information

Authors and Affiliations

Authors

Contributions

Y. Zhao, Y.Y., Y. Zou, and M.S. conceived and designed the live-cell and zebrafish imaging experiments. Y. Zhao, Y.Y., and A.W. designed the flow-cytometry analysis experiment. Y. Zou, A.W., M.S., X.C., R.L., T.L., and C.Z. performed experiments. Z.Z., L.Z., Z.J., and J.L. gave technical support and conceptual advice. Y.Y., Y. Zhao, Y. Zou, A.W., M.S., and J.L. analyzed the data and wrote the manuscript.

Corresponding authors

Correspondence to Yi Yang or Yuzheng Zhao.

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.

Related links

Key references using this protocol

1. Tao, R. et al. Nat. Methods 14, 720–728 (2017): https://doi.org/10.1038/nmeth.4306

2. Zhao, Y. et al. Cell Metab. 21, 777–789 (2015): https://doi.org/10.1016/j.cmet.2015.04.009

3. Zhao, Y. et al. Nat. Protoc. 11, 1345–1359 (2016): https://doi.org/10.1038/nprot.2016.074

Integrated supplementary information

Supplementary Figure 1 Effect of diamide or oxamate treatment on subcellular pH.

(a and b) Fluorescence imaging (top) and fluorescence changes (bottom, n = 6 cells) in HeLa cells simultaneously expressing cytosol-localized iNapc and cytosol-localized pHRFP in response to 200 µM diamide (a) or 2 mM oxamate (b). (c and d) Fluorescence imaging (top) and fluorescence changes (bottom, n = 6 cells) in HeLa cells simultaneously expressing mitochondria-localized iNapc, nuclear-localized iNapc, and mitochondrial-localized pHRFP in response to 200 µM diamide (c) or 2 mM oxamate (d). Data are the mean ± s.d. All p values were obtained using unpaired two-tailed Student’s t test. *p < 0.05, ***p < 0.001. Scale bars, 10 µm.

Supplementary Figure 2 pH fluorescence imaging during the cell cycle.

(a and b) Fluorescence images (a) and quantification (b) of pH dynamics during cell division. Scale bars, 10 µm.

Supplementary Figure 3 Example gating strategy.

(a) Sample gating strategy for forward and side scatter (FSC/SSC). (b and c) Sample gating strategy for negative cells (b) and positive cells (sensor-expressing cells, c).

Supplementary Figure 4 Flow cytometry analysis of the pH state in resting and activated mouse macrophages.

(a) Cytosolic or mitochondrial pH detection in resting or activated RAW264.7 mouse macrophages by flow cytometry. (b) Quantitative data for cytosolic or mitochondrial pH sensor fluorescence were obtained from three or more independent detections by flow cytometry. Data are the mean ± s.e.m. All p values were obtained using unpaired two-tailed Student’s t test. *p < 0.05, **p < 0.01, ***p < 0.001.

Supplementary Figure 5 pH fluorescence imaging of zebrafish larvae.

(a and b) In vivo fluorescence imaging of zebrafish larvae expressing iNapc in response to 50 mM H2O2 (a) or 5 µM rotenone (b).

Supplementary information

Supplementary Figures 1–5

Supplementary Figures 1–5 and Supplementary Table 1: The numerical analysis of all cells and positive cells.

Reporting Summary

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zou, Y., Wang, A., Shi, M. et al. Analysis of redox landscapes and dynamics in living cells and in vivo using genetically encoded fluorescent sensors. Nat Protoc 13, 2362–2386 (2018). https://doi.org/10.1038/s41596-018-0042-5

Download citation

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-018-0042-5

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