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:

A puromycin-dependent activity-based sensing probe for histochemical staining of hydrogen peroxide in cells and animal tissues

Abstract

Hydrogen peroxide (H2O2) is a key member of the reactive oxygen species family of transient small molecules that has broad contributions to oxidative stress and redox signaling. The development of selective and sensitive chemical probes can enable the study of H2O2 biology in cell, tissue and animal models. Peroxymycin-1 is a histochemical activity–based sensing probe that responds to H2O2 via chemoselective boronate oxidation to release puromycin, which is then covalently incorporated into nascent proteins by the ribosome and can be detected by antibody staining. Here, we describe an optimized two-step, one-pot protocol for synthesizing Peroxymycin-1 with improved yields over our originally reported procedure. We also present detailed procedures for applying Peroxymycin-1 to a broad range of biological samples spanning cells to animal tissues for profiling H2O2 levels through histochemical detection by using commercially available anti-puromycin antibodies. The preparation of Peroxymycin-1 takes 9 h, the confocal imaging experiments of endogenous H2O2 levels across different cancer cell lines take 1 d, the dot blot analysis of mouse liver tissues takes 1 d and the confocal imaging of mouse liver tissues takes 3–4 d.

This is a preview of subscription content, access via your institution

Access options

Buy this article

Prices may be subject to local taxes which are calculated during checkout

Fig. 1: Overview of hydrogen peroxide detection with Peroxymycin-1 in cells and mice.
Fig. 2: Synthetic and mechanistic scheme of Peroxymycin-1.
Fig. 3: In vitro analysis of Peroxymycin-1 reactivity and selectivity.
Fig. 4: Confocal microscopy images of endogenous H2O2 detection by using Peroxymycin-1.
Fig. 5: Profiling H2O2 levels with Peroxymycin-1 in liver tissue of mice fed a normal diet and a high-fat diet.

Similar content being viewed by others

Data availability

Data are available through figshare: Fig. 2c, https://doi.org/10.6084/m9.figshare.16639975; Fig. 3a, https://doi.org/10.6084/m9.figshare.16639570; Fig. 4, https://doi.org/10.6084/m9.figshare.16640434; Fig. 5, https://doi.org/10.6084/m9.figshare.16639537; Supplementary Fig. 1, https://doi.org/10.6084/m9.figshare.18480879; Supplementary Fig. 2, https://doi.org/10.6084/m9.figshare.18480885; Supplementary Fig. 3, https://doi.org/10.6084/m9.figshare.18480888; Supplementary Fig. 4, https://doi.org/10.6084/m9.figshare.18480891; Supplementary Fig. 5, https://doi.org/10.6084/m9.figshare.18480894; Supplementary Fig. 6, https://doi.org/10.6084/m9.figshare.18480897; Supplementary Fig. 7, https://doi.org/10.6084/m9.figshare.18480900; Supplementary Fig. 8, https://doi.org/10.6084/m9.figshare.18480903. Source data are provided with this paper.

References

  1. Baynes, J. W. Role of oxidative stress in development of complications in diabetes. Diabetes 40, 405–412 (1991).

    Article  CAS  PubMed  Google Scholar 

  2. Multhaup, G. et al. Reactive oxygen species and Alzheimer’s disease. Biochem. Pharmacol. 54, 533–539 (1997).

    Article  CAS  PubMed  Google Scholar 

  3. Stone, J. R. & Yang, S. Hydrogen peroxide: a signaling messenger. Antioxid. Redox Signal. 8, 243–270 (2006).

    Article  CAS  PubMed  Google Scholar 

  4. Rhee, S. G. H2O2, a necessary evil for cell signaling. Science 312, 1882–1883 (2006).

    Article  PubMed  Google Scholar 

  5. D’Autréaux, B. & Toledano, M. B. ROS as signaling molecules: mechanisms that generate specificity in ROS homeostasis. Nat. Rev. Mol. Cell Biol. 8, 813–824 (2007).

    Article  PubMed  CAS  Google Scholar 

  6. Finkel, T., Serrano, M. & Blasco, M. A. The common biology of cancer and ageing. Nature 448, 767–774 (2007).

    Article  CAS  PubMed  Google Scholar 

  7. Winterbourn, C. C. Reconciling the chemistry and biology of reactive oxygen species. Nat. Chem. Biol. 4, 278–286 (2008).

    Article  CAS  PubMed  Google Scholar 

  8. Dickinson, B. C. & Chang, C. J. Chemistry and biology of reactive oxygen species in signaling or stress responses. Nat. Chem. Biol. 7, 504–511 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  10. Schieber, M. & Chandel, N. S. ROS function in redox signaling and oxidative stress. Curr. Biol. 24, R453–R462 (2014).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  11. Reichmann, D., Voth, W. & Jakob, U. Maintaining a healthy proteome during oxidative stress. Mol. Cell 69, 203–213 (2018).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Sies, H. & Jones, D. P. Reactive oxygen species (ROS) as pleiotropic physiological signalling agents. Nat. Rev. Mol. Cell Biol. 21, 363–383 (2020).

    Article  CAS  PubMed  Google Scholar 

  13. Inoguchi, T. et al. High glucose level and free fatty acid stimulate reactive oxygen species production through protein kinase C–dependent activation of NAD(P)H oxidase in cultured vascular cells. Diabetes 49, 1939–1945 (2000).

    Article  CAS  PubMed  Google Scholar 

  14. Park, L. et al. Nox2-derived radicals contribute to neurovascular and behavioral dysfunction in mice overexpressing the amyloid precursor protein. Proc. Natl Acad. Sci. USA. 105, 1347–1352 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  15. Schmidt, K. N., Amstad, P., Cerutti, P. & Baeuerle, P. A. The roles of hydrogen peroxide and superoxide as messengers in the activation of transcription factor NF-κB. Chem. Biol. 2, 13–22 (1995).

    Article  CAS  PubMed  Google Scholar 

  16. Guyton, K. Z., Liu, Y., Gorospe, M., Xu, Q. & Holbrook, N. J. Activation of mitogen-activated protein kinase by H2O2: role in cell survival following oxidant injury. J. Biol. Chem. 271, 4138–4142 (1996).

    Article  CAS  PubMed  Google Scholar 

  17. Lee, S.-R., Kwon, K.-S., Kim, S.-R. & Rhee, S. G. Reversible inactivation of protein-tyrosine phosphatase 1B in A431 cells stimulated with epidermal growth factor. J. Biol. Chem. 273, 15366–15372 (1998).

    Article  CAS  PubMed  Google Scholar 

  18. Salmeen, A. et al. Redox regulation of protein tyrosine phosphatase 1B involves a sulphenyl-amide intermediate. Nature 423, 769–773 (2003).

    Article  CAS  PubMed  Google Scholar 

  19. Avshalumov, M. V. & Rice, M. E. Activation of ATP-sensitive K+ (KATP) channels by H2O2 underlies glutamate-dependent inhibition of striatal dopamine release. Proc. Natl Acad. Sci. USA. 100, 11729–11734 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Lambeth, J. D. NOX enzymes and the biology of reactive oxygen. Nat. Rev. Immunol. 4, 181–189 (2004).

    Article  CAS  PubMed  Google Scholar 

  21. Dinauer, M. C., Orkin, S. H., Brown, R., Jesaitis, A. J. & Parkos, C. A. The glycoprotein encoded by the X-linked chronic granulomatous disease locus is a component of the neutrophil cytochrome b complex. Nature 327, 717–720 (1987).

    Article  CAS  PubMed  Google Scholar 

  22. Volpp, B., Nauseef, W. & Clark, R. Two cytosolic neutrophil oxidase components absent in autosomal chronic granulomatous disease. Science 242, 1295–1297 (1988).

    Article  CAS  PubMed  Google Scholar 

  23. Clark, R. A. et al. Genetic variants of chronic granulomatous disease: prevalence of deficiencies of two cytosolic components of the NADPH oxidase system. N. Engl. J. Med. 321, 647–652 (1989).

    Article  CAS  PubMed  Google Scholar 

  24. Ohba, M., Shibanuma, M., Kuroki, T. & Nose, K. Production of hydrogen peroxide by transforming growth factor-beta 1 and its involvement in induction of egr-1 in mouse osteoblastic cells. J. Cell Biol. 126, 1079–1088 (1994).

    Article  CAS  PubMed  Google Scholar 

  25. Sundaresan, M., Yu, Z.-X., Ferrans, V. J., Irani, K. & Finkel, T. Requirement for generation of H2O2 for platelet-derived growth factor signal transduction. Science 270, 296–299 (1995).

    Article  CAS  PubMed  Google Scholar 

  26. Kimura, T., Okajima, F., Sho, K., Kobayashi, I. & Kondo, Y. Thyrotropin-induced hydrogen peroxide production in FRTL-5 thyroid cells is mediated not by adenosine 3′, 5′-monophosphate, but by Ca2+ signaling followed by phospholipase-A2 activation and potentiated by an adenosine derivative. Endocrinology 136, 116–123 (1995).

    Article  CAS  PubMed  Google Scholar 

  27. Bae, Y. S. et al. Epidermal growth factor (EGF)-induced generation of hydrogen peroxide. Role in EGF receptor-mediated tyrosine phosphorylation. J. Biol. Chem. 272, 217–221 (1997).

    Article  CAS  PubMed  Google Scholar 

  28. Mukhin, Y. V. et al. 5-Hydroxytryptamine1A receptor/Giβγ stimulates mitogen-activated protein kinase via NAD(P)H oxidase and reactive oxygen species upstream of src in chinese hamster ovary fibroblasts. Biochem. J. 347, 61–67 (2000).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Dickinson, B. C., Peltier, J., Stone, D., Schaffer, D. V. & Chang, C. J. Nox2 redox signaling maintains essential cell populations in the brain. Nat. Chem. Biol. 7, 106–112 (2011).

    Article  CAS  PubMed  Google Scholar 

  30. Kamsler, A. & Segal, M. Hydrogen peroxide modulation of synaptic plasticity. J. Neurosci. 23, 269–276 (2003).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  31. Tejada-Simon, M. V. et al. Synaptic localization of a functional NADPH oxidase in the mouse hippocampus. Mol. Cell. Neurosci. 29, 97–106 (2005).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  32. Brennan, A. M. et al. NADPH oxidase is the primary source of superoxide induced by NMDA receptor activation. Nat. Neurosci. 12, 857–863 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  33. De Pasquale, R., Beckhauser, T. F., Hernandes, M. S. & Giorgetti Britto, L. R. LTP and LTD in the visual cortex require the activation of NOX2. J. Neurosci. 34, 12778–12787 (2014).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  34. Le Belle, J. E. et al. Proliferative neural stem cells have high endogenous ROS levels that regulate self-renewal and neurogenesis in a PI3K/Akt-dependant manner. Cell Stem Cell 8, 59–71 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  35. Xu, C., Luo, J., He, L., Montell, C. & Perrimon, N. Oxidative stress induces stem cell proliferation via TRPA1/RyR-mediated Ca2+ signaling in the Drosophila midgut. eLife 6, e22441 (2017).

    Article  PubMed  PubMed Central  Google Scholar 

  36. O’Neill, J. S. & Reddy, A. B. Circadian clocks in human red blood cells. Nature 469, 498–503 (2011).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  37. Wible, R. S. et al. NRF2 regulates core and stabilizing circadian clock loops, coupling redox and timekeeping in Mus musculus. eLife 7, e31656 (2018).

    Article  PubMed  PubMed Central  Google Scholar 

  38. Pei, J.-F. et al. Diurnal oscillations of endogenous H2O2 sustained by p66Shc regulate circadian clocks. Nat. Cell Biol. 21, 1553–1564 (2019).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  39. Niethammer, P., Grabher, C., Look, A. T. & Mitchison, T. J. A tissue-scale gradient of hydrogen peroxide mediates rapid wound detection in zebrafish. Nature 459, 996–999 (2009).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  40. Hervera, A. et al. Reactive oxygen species regulate axonal regeneration through the release of exosomal NADPH oxidase 2 complexes into injured axons. Nat. Cell Biol. 20, 307–319 (2018).

    Article  CAS  PubMed  Google Scholar 

  41. Lippert, A. R., Van de Bittner, G. C. & Chang, C. J. Boronate oxidation as a bioorthogonal reaction approach for studying the chemistry of hydrogen peroxide in living systems. Acc. Chem. Res. 44, 793–804 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  42. Chan, J., Dodani, S. C. & Chang, C. J. Reaction-based small-molecule fluorescent probes for chemoselective bioimaging. Nat. Chem. 4, 973–984 (2012).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  43. Brewer, T. F., Garcia, F. J., Onak, C. S., Carroll, K. S. & Chang, C. J. Chemical approaches to discovery and study of sources and targets of hydrogen peroxide redox signaling through NADPH oxidase proteins. Annu. Rev. Biochem. 84, 765–790 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  44. Bruemmer, K. J., Crossley, S. W. M. & Chang, C. J. Activity-based sensing: a synthetic methods approach for selective molecular imaging and beyond. Angew. Chem. Int. Ed. 59, 13734–13762 (2019).

    Article  CAS  Google Scholar 

  45. Chang, M. C. Y., Pralle, A., Isacoff, E. Y. & Chang, C. J. A selective, cell-permeable optical probe for hydrogen peroxide in living cells. J. Am. Chem. Soc. 126, 15392–15393 (2004).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  46. Miller, E. W., Tulyathan, O., Isacoff, E. Y. & Chang, C. J. Molecular imaging of hydrogen peroxide produced for cell signaling. Nat. Chem. Biol. 3, 263–267 (2007).

    Article  CAS  PubMed  Google Scholar 

  47. Dickinson, B. C., Huynh, C. & Chang, C. J. A palette of fluorescent probes with varying emission colors for imaging hydrogen peroxide signaling in living cells. J. Am. Chem. Soc. 132, 5906–5915 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  48. Srikun, D., Miller, E. W., Domaille, D. W. & Chang, C. J. An ICT-based approach to ratiometric fluorescence imaging of hydrogen peroxide produced in living cells. J. Am. Chem. Soc. 130, 4596–4597 (2008).

    Article  CAS  PubMed  Google Scholar 

  49. Albers, A. E., Okreglak, V. S. & Chang, C. J. A FRET-based approach to ratiometric fluorescence detection of hydrogen peroxide. J. Am. Chem. Soc. 128, 9640–9641 (2006).

    Article  CAS  PubMed  Google Scholar 

  50. Chung, C., Srikun, D., Lim, C. S., Chang, C. J. & Cho, B. R. A two-photon fluorescent probe for ratiometric imaging of hydrogen peroxide in live tissue. Chem. Commun. 47, 9618–9620 (2011).

    Article  CAS  Google Scholar 

  51. Dickinson, B. C. & Chang, C. J. A targetable fluorescent probe for imaging hydrogen peroxide in the mitochondria of living cells. J. Am. Chem. Soc. 130, 9638–9639 (2008).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  52. Dickinson, B. C., Tang, Y., Chang, Z. & Chang, C. J. A nuclear-localized fluorescent hydrogen peroxide probe for monitoring sirtuin-mediated oxidative stress responses in vivo. Chem. Biol. 18, 943–948 (2011).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  53. Miller, E. W., Dickinson, B. C. & Chang, C. J. Aquaporin-3 mediates hydrogen peroxide uptake to regulate downstream intracellular signaling. Proc. Natl Acad. Sci. USA. 107, 15681–15686 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  54. Iwashita, H., Castillo, E., Messina, M. S., Swanson, R. A. & Chang, C. J. A tandem activity-based sensing and labeling strategy enables imaging of transcellular hydrogen peroxide signaling. Proc. Natl Acad. Sci. USA. 118, e2018513118 (2021).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  55. Van de Bittner, G. C., Dubikovskaya, E. A., Bertozzi, C. R. & Chang, C. J. In vivo imaging of hydrogen peroxide production in a murine tumor model with a chemoselective bioluminescent reporter. Proc. Natl Acad. Sci. USA. 107, 21316–21321 (2010).

    Article  PubMed  PubMed Central  Google Scholar 

  56. Van de Bittner, G. C., Bertozzi, C. R. & Chang, C. J. Strategy for dual-analyte luciferin imaging: in vivo bioluminescence detection of hydrogen peroxide and caspase activity in a murine model of acute Inflammation. J. Am. Chem. Soc. 135, 1783–1795 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  57. Jin, L. et al. Glutamate dehydrogenase 1 signals through antioxidant glutathione peroxidase 1 to regulate redox homeostasis and tumor growth. Cancer Cell 27, 257–270 (2015).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  58. Schoenfeld, J. D. et al. O2 and H2O2–mediated disruption of Fe metabolism causes the differential susceptibility of NSCLC and GBM cancer cells to pharmacological ascorbate. Cancer Cell 31, 487–500 (2017).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Chung, C. Y.-S., Timblin, G. A., Saijo, K. & Chang, C. J. Versatile histochemical approach to detection of hydrogen peroxide in cells and tissues based on puromycin staining. J. Am. Chem. Soc. 140, 6109–6121 (2018).

    Article  PubMed Central  CAS  Google Scholar 

  60. Dhibi, M. et al. The intake of high fat diet with different trans fatty acid levels differentially induces oxidative stress and non alcoholic fatty liver disease (NAFLD) in rats. Nutr. Metab. 8, 65–77 (2011).

    Article  CAS  Google Scholar 

  61. Bilan Dmitry, S. & Belousov Vsevolod, V. In vivo imaging of hydrogen peroxide with HyPer probes. Antioxid. Redox Signal. 29, 569–584 (2018).

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  63. Srikun, D., Albers, A. E., Nam, C. I., Iavarone, A. T. & Chang, C. J. Organelle-targetable fluorescent probes for imaging hydrogen peroxide in living cells via SNAP-Tag protein labeling. J. Am. Chem. Soc. 132, 4455–4465 (2010).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  64. Dickinson, B. C., Lin, V. S. & Chang, C. J. Preparation and use of MitoPY1 for imaging hydrogen peroxide in mitochondria of live cells. Nat. Protoc. 8, 1249–1259 (2013).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

  65. Szweda, P. A., Tsai, L. & Szweda, L. I. Immunochemical detection of a fluorophore derived from the lipid peroxidation product 4-hydroxy-2-nonenal and lysine. In Oxidants and Antioxidants: Ultrastructure and Molecular Biology Protocols (ed. Armstrong, D.) Vol. 196 277–290 (Humana Press, 2002).

  66. Spangler, B. et al. A reactivity-based probe of the intracellular labile ferrous iron pool. Nat. Chem. Biol. 12, 680–685 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Schmidt, E. K., Clavarino, G., Ceppi, M. & Pierre, P. SUnSET, a nonradioactive method to monitor protein synthesis. Nat. Methods 6, 275–277 (2009).

    Article  CAS  PubMed  Google Scholar 

  68. Su, K.-H. et al. HSF1 critically attunes proteotoxic stress sensing by mTORC1 to combat stress and promote growth. Nat. Cell Biol. 18, 527–539 (2016).

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  69. tom Dieck, S. et al. Direct visualization of newly synthesized target proteins in situ. Nat. Methods 12, 411–414 (2015).

    Article  CAS  PubMed  Google Scholar 

  70. Deliu, L. P., Ghosh, A. & Grewal, S. S. Investigation of protein synthesis in Drosophila larvae using puromycin labelling. Biol. Open 6, 1229–1234 (2017).

    CAS  PubMed  PubMed Central  Google Scholar 

  71. Bielczyk-Maczyńska, E. et al. The ribosome biogenesis protein Nol9 is essential for definitive hematopoiesis and pancreas morphogenesis in zebrafish. PLoS Genet. 11, e1005677 (2015).

    Article  PubMed  PubMed Central  CAS  Google Scholar 

Download references

Acknowledgements

We thank the NIH (R01 GM 79465, R01 GM 139245 and R01 ES 28096 to C.J.C.) for research support. K.H. thanks the College of Chemistry for a summer undergraduate research fellowship. J.O. thanks the Japan Society for the Promotion of Science for a postdoctoral fellowship. C.Y.-S.C. thanks the Croucher Foundation for a postdoctoral fellowship. M.S.M. thanks the UC President’s Postdoctoral Fellowship Program, Chinook-Berkeley Postdoctoral Fellowship Program and an NIH MOSAIC K99/R00 (1K99GM143573-01) award for funding. C.J.C. is a CIFAR Fellow. We thank Alison Killilea and Carissa Tasto (UC Berkeley Tissue Culture Facility) for expert technical assistance.

Author information

Authors and Affiliations

Authors

Contributions

K.H., J.O., C.Y.-S.C., M.S.M. and C.J.C. wrote the manuscript. K.H. and J.O. developed the new synthesis. C.Y.-S.C. performed the imaging experiments. M.S.M. reproduced the synthesis and provided full characterization of the compounds. M.S.M. performed imaging experiments required for revisions.

Corresponding author

Correspondence to Christopher J. Chang.

Ethics declarations

Competing interests

A patent application has been filed for the Peroxymycin-1 probe. The patent application number is PCT/US2019/023242.

Peer review

Peer review information

Nature Protocols thanks Deju Ye and the other, anonymous, reviewer(s) for their contribution to the peer review of this work.

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

Chung, C. et al. J. Am. Chem. Soc. 140, 6109–6121 (2018): https://doi.org/10.1021/jacs.8b02279

Spangler, B. et al. Nat. Chem. Biol. 12, 680–685 (2016): https://doi.org/10.1038/nchembio.2116

Supplementary information

Supplementary Information

Supplementary Text and Supplementary Figs. 1–8.

Source data

Source Data Fig. 3

Unprocessed HPLC data

Source Data Fig. 4

Fluorescence intensity data and statistical analysis

Source Data Fig. 5

Fluorescence intensity data and statistical analysis

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Hoshi, K., Messina, M.S., Ohata, J. et al. A puromycin-dependent activity-based sensing probe for histochemical staining of hydrogen peroxide in cells and animal tissues. Nat Protoc 17, 1691–1710 (2022). https://doi.org/10.1038/s41596-022-00694-7

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1038/s41596-022-00694-7

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