Skip to main content
Log in

Recent Developments in the Probes and Assays for Measurement of the Activity of NADPH Oxidases

  • Original Paper
  • Published:
Cell Biochemistry and Biophysics Aims and scope Submit manuscript

Abstract

NADPH oxidases are a family of enzymes capable of transferring electrons from NADPH to molecular oxygen. A major function of NADPH oxidases is the activation of molecular oxygen into reactive oxygen species. Increased activity of NADPH oxidases has been implicated in various pathologies, including cardiovascular disease, neurological dysfunction, and cancer. Thus, NADPH oxidases have been identified as a viable target for the development of novel therapeutics exhibiting inhibitory effects on NADPH oxidases. Here, we describe the development of new assays for measuring the activity of NADPH oxidases enabling the high-throughput screening for NADPH oxidase inhibitors.

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
Fig. 2
Fig. 3
Fig. 4
Fig. 5
Fig. 6
Fig. 7
Fig. 8
Fig. 9
Fig. 10
Fig. 11
Fig. 12
Fig. 13
Fig. 14
Fig. 15
Fig. 16
Fig. 17

Similar content being viewed by others

Abbreviations

2-OH-E+ :

2-hydroxyethidium

2-OH-Pr2+ :

2-hydroxypropidium

BMPO:

5-tert-butoxycarbonyl-5-methyl-1-pyrroline-N-oxide

CAT:

catalase

CBA:

coumarin-7-boronic acod

CD:

cyclodextrin

COH:

7-hydroxycoumarin

cyt c 3+ :

ferricytochrome c

DEPMPO:

5-(diethoxyphosphoryl)-5-methyl-1-pyrroline-N-oxide

DIPPMPO:

5-(diisopropoxyphosphoryl)-5-methyl-1-pyrroline-N-oxide

DMPO:

5,5-dimethyl-1-pyrroline-N-oxide

DPI:

diphenyleneiodonium

E+ :

ethidium

E+-E+ :

diethidium

EPR:

electron paramagnetic resonance

GSH:

glutathione

HE:

hydroethidine (or dihydroethidium)

HE•+ :

HE radical cation

HPLC:

high-performance liquid chromatography

HPr+ :

hydropropidine

HRP:

horseradish peroxidase

HTS:

high-throughput screening

L-012:

8-amino-5-chloro-2,3-dihydro-7-phenyl-pyrido[3,4-d]pyridazine

MPO:

myeloperoxidase

NADPH:

nicotinamide adenine dinucleotide phosphate, reduced form

NBT:

nitroblue tetrazolium

Nox2:

NADPH oxidase-2

Nox4:

NADPH oxidase-4

Nox5:

NADPH oxidase-5

OCR:

oxygen consumption rate

PMA:

phorbol 12-myristate 13-acetate

SOD:

superoxide dismutase

VAS2870:

1,3-benzoxazol-2-yl-3-benzyl-3H-[1–3]triazolo[4,5-d]pyrimidin-7-yl sulfide

References

  1. Al Ghouleh, I., Khoo, N. K., Knaus, U. G., Griendling, K. K., Touyz, R. M., Thannickal, V. J., Barchowsky, A., Nauseef, W. M., Kelley, E. E., Bauer, P. M., Darley-Usmar, V., Shiva, S., Cifuentes-Pagano, E., Freeman, B. A., Gladwin, M. T., & Pagano, P. J. (2011). Oxidases and peroxidases in cardiovascular and lung disease: New concepts in reactive oxygen species signaling. Free Radical Biology & Medicine, 51, 1271–1288.

    Article  CAS  Google Scholar 

  2. Lambeth, J. D. (2004). NOX enzymes and the biology of reactive oxygen. Nature Reviews Immunology, 4, 181–189.

    Article  CAS  PubMed  Google Scholar 

  3. Leto, T. L., Morand, S., Hurt, D., & Ueyama, T. (2009). Targeting and regulation of reactive oxygen species generation by Nox family NADPH oxidases. Antioxidants & Redox Signaling, 11, 2607–2619.

    Article  CAS  Google Scholar 

  4. Bedard, K., & Krause, K. H. (2007). The NOX family of ROS-generating NADPH oxidases: Physiology and pathophysiology. Physiological Reviews, 87, 245–313.

    Article  CAS  PubMed  Google Scholar 

  5. Halliwell, B., & Gutteridge, J. M. (2015). Free radicals in biology and medicine. USA: Oxford University Press.

    Book  Google Scholar 

  6. Bonner, M. Y., & Arbiser, J. L. (2012). Targeting NADPH oxidases for the treatment of cancer and inflammation. Cellular and Molecular Life Sciences, 69, 2435–2442.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  7. Guzik, T. J., & Harrison, D. G. (2006). Vascular NADPH oxidases as drug targets for novel antioxidant strategies. Drug Discovery Today, 11, 524–533.

    Article  CAS  PubMed  Google Scholar 

  8. Hecker, L., Cheng, J., & Thannickal, V. J. (2012). Targeting NOX enzymes in pulmonary fibrosis. Cellular and Molecular Life Sciences, 69, 2365–2371.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  9. Streeter, J., Thiel, W., Brieger, K., & Miller, Jr., F. J. (2013). Opportunity Nox: The future of NADPH oxidases as therapeutic targets in cardiovascular disease. Cardiovascular Therapeutics, 31, 125–137.

    Article  CAS  PubMed  Google Scholar 

  10. Lambeth, J. D., Krause, K. H., & Clark, R. A. (2008). NOX enzymes as novel targets for drug development. Seminars in Immunopathology, 30, 339–363.

    Article  CAS  PubMed  Google Scholar 

  11. Krause, K. H., Lambeth, D., & Kronke, M. (2012). NOX enzymes as drug targets. Cellular and Molecular Life Sciences, 69, 2279–2282.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  12. Nayernia, Z., Jaquet, V., & Krause, K. H. (2014). New insights on NOX enzymes in the central nervous system. Antioxidants & Redox Signaling, 20, 2815–2837.

    Article  CAS  Google Scholar 

  13. Altenhofer, S., Radermacher, K. A., Kleikers, P. W., Wingler, K., & Schmidt, H. H. (2015). Evolution of NADPH oxidase inhibitors: Selectivity and mechanisms for target engagement. Antioxidants & Redox Signaling, 23, 406–427.

    Article  CAS  Google Scholar 

  14. Borbely, G., Szabadkai, I., Horvath, Z., Marko, P., Varga, Z., Breza, N., Baska, F., Vantus, T., Huszar, M., Geiszt, M., Hunyady, L., Buday, L., Orfi, L., & Keri, G. (2010). Small-molecule inhibitors of NADPH oxidase 4. Journal of Medicinal Chemistry, 53, 6758–6762.

    Article  CAS  PubMed  Google Scholar 

  15. Cifuentes-Pagano, E., Meijles, D. N., & Pagano, P. J. (2014). The quest for selective Nox inhibitors and therapeutics: Challenges, triumphs and pitfalls. Antioxidants & Redox Signaling, 20, 2741–2754.

    Article  CAS  Google Scholar 

  16. Drummond, G. R., Selemidis, S., Griendling, K. K., & Sobey, C. G. (2011). Combating oxidative stress in vascular disease: NADPH oxidases as therapeutic targets. Nature Reviews Drug Discovery, 10, 453–471.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  17. Jaquet, V., Scapozza, L., Clark, R. A., Krause, K. H., & Lambeth, J. D. (2009). Small-molecule NOX inhibitors: ROS-generating NADPH oxidases as therapeutic targets. Antioxidants & Redox Signaling, 11, 2535–2552.

    Article  CAS  Google Scholar 

  18. Wind, S., Beuerlein, K., Eucker, T., Muller, H., Scheurer, P., Armitage, M. E., Ho, H., Schmidt, H. H., & Wingler, K. (2010). Comparative pharmacology of chemically distinct NADPH oxidase inhibitors. British Journal of Pharmacology, 161, 885–898.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  19. Cifuentes-Pagano, E., Csanyi, G., & Pagano, P. J. (2012). NADPH oxidase inhibitors: A decade of discovery from Nox2ds to HTS. Cellular and Molecular Life Sciences, 69, 2315–2325.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  20. Laleu, B., Gaggini, F., Orchard, M., Fioraso-Cartier, L., Cagnon, L., Houngninou-Molango, S., Gradia, A., Duboux, G., Merlot, C., Heitz, F., Szyndralewiez, C., & Page, P. (2010). First in class, potent, and orally bioavailable NADPH oxidase isoform 4 (Nox4) inhibitors for the treatment of idiopathic pulmonary fibrosis. Journal of Medicinal Chemistry, 53, 7715–7730.

    Article  CAS  PubMed  Google Scholar 

  21. Cifuentes-Pagano, E., Saha, J., Csanyi, G., Ghouleh, I. A., Sahoo, S., Rodriguez, A., Wipf, P., Pagano, P. J., & Skoda, E. M. (2013). Bridged tetrahydroisoquinolines as selective NADPH oxidase 2 (Nox2) inhibitors. Medchemcomm., 4, 1085–1092.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  22. Hirano, K., Chen, W. S., Chueng, A. L., Dunne, A. A., Seredenina, T., Filippova, A., Ramachandran, S., Bridges, A., Chaudry, L., Pettman, G., Allan, C., Duncan, S., Lee, K. C., Lim, J., Ma, M. T., Ong, A. B., Ye, N. Y., Nasir, S., Mulyanidewi, S., Aw, C. C., Oon, P. P., Liao, S., Li, D., Johns, D. G., Miller, N. D., Davies, C. H., Browne, E. R., Matsuoka, Y., Chen, D. W., Jaquet, V., & Rutter, A. R. (2015). Discovery of GSK2795039, a novel small molecule NADPH oxidase 2 inhibitor. Antioxidants & Redox Signaling, 23, 358–374.

    Article  CAS  Google Scholar 

  23. Seredenina, T., Chiriano, G., Filippova, A., Nayernia, Z., Mahiout, Z., Fioraso-Cartier, L., Plastre, O., Scapozza, L., Krause, K. H., & Jaquet, V. (2015). A subset of N-substituted phenothiazines inhibits NADPH oxidases. Free Radical Biology & Medicine, 86, 239–249.

    Article  CAS  Google Scholar 

  24. Zielonka, J., Zielonka, M., VerPlank, L., Cheng, G., Hardy, M., Ouari, O., Ayhan, M. M., Podsiadly, R., Sikora, A., Lambeth, J. D., & Kalyanaraman, B. (2016). Mitigation of NADPH oxidase 2 activity as a strategy to inhibit peroxynitrite formation. The Journal of Biological Chemistry, 291, 7029–7044.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  25. Maghzal, G. J., Krause, K. H., Stocker, R., & Jaquet, V. (2012). Detection of reactive oxygen species derived from the family of NOX NADPH oxidases. Free Radical Biology & Medicine, 53, 1903–1918.

    Article  CAS  Google Scholar 

  26. Kalyanaraman, B., Hardy, M., Podsiadly, R., Cheng, G., & Zielonka, J. (2017). Recent developments in detection of superoxide radical anion and hydrogen peroxide: Opportunities, challenges, and implications in redox signaling. Archives of Biochemistry and Biophysics, 617, 38–47.

    Article  CAS  PubMed  Google Scholar 

  27. Debowska, K., Debski, D., Hardy, M., Jakubowska, M., Kalyanaraman, B., Marcinek, A., Michalski, R., Michalowski, B., Ouari, O., Sikora, A., Smulik, R., & Zielonka, J. (2015). Toward selective detection of reactive oxygen and nitrogen species with the use of fluorogenic probes--limitations, progress, and perspectives. Pharmacological Reports, 67, 756–764.

    Article  CAS  PubMed  Google Scholar 

  28. Kalyanaraman, B., Hardy, M., & Zielonka, J. (2016). A critical review of methodologies to detect reactive oxygen and nitrogen species stimulated by NADPH oxidase enzymes: Implications in pesticide toxicity. Current Pharmacology Reports, 2, 193–201.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  29. Wardman, P. (2007). Fluorescent and luminescent probes for measurement of oxidative and nitrosative species in cells and tissues: Progress, pitfalls, and prospects. Free Radical Biology & Medicine, 43, 995–1022.

    Article  CAS  Google Scholar 

  30. Koppenol, W. H., van Buuren, K. J., Butler, J., & Braams, R. (1976). The kinetics of the reduction of cytochrome c by the superoxide anion radical. Biochimica et Biophysica Acta, 449, 157–168.

    Article  CAS  PubMed  Google Scholar 

  31. Bielski, B. H. J., Shiue, G. G., & Bajuk, S. (1980). Reduction of nitro blue tetrazolium by CO2- and O2- radicals. The Journal of Physical Chemistry, 84, 830–833.

    Article  CAS  Google Scholar 

  32. Nauseef, W. M. (2014). Detection of superoxide anion and hydrogen peroxide production by cellular NADPH oxidases. Biochimica et Biophysica Acta, 1840, 757–767.

    Article  CAS  PubMed  Google Scholar 

  33. Butler, J., Koppenol, W. H., & Margoliash, E. (1982). Kinetics and mechanism of the reduction of ferricytochrome c by the superoxide anion. The Journal of Biological Chemistry, 257, 10747–10750.

    CAS  PubMed  Google Scholar 

  34. Nisimoto, Y., Otsuka-Murakami, H., & Iwata, S. (1994). NADPH-cytochrome c reductase from human neutrophil membranes: Purification, characterization and localization. The Biochemical Journal, 297(Pt 3), 585–593.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  35. Nisimoto, Y., Otsuka-Murakami, H., Iwata, S., Isogai, Y., & Iizuka, T. (1993). Characterization of superoxide dismutase-insensitive cytochrome c reductase activity in HL-60 cytosol as NADPH-cytochrome P450 reductase. Archives of Biochemistry and Biophysics, 302, 315–321.

    Article  CAS  PubMed  Google Scholar 

  36. Ginsburgh, C. L., & Everse, J. (1978). Studies on the reduction of cytochrome c by thiols. Bioorganic Chemistry, 7, 481–492.

    Article  CAS  Google Scholar 

  37. Hu, T. M., & Ho, S. C. (2011). Kinetics of redox interaction between cytochrome c and thiols. Journal of Medical Sciences, 31, 109–115.

    Google Scholar 

  38. Choi, H. S., Kim, J. W., Cha, Y. N., & Kim, C. (2006). A quantitative nitroblue tetrazolium assay for determining intracellular superoxide anion production in phagocytic cells. Journal of Immunoassay & Immunochemistry, 27, 31–44.

    Article  CAS  Google Scholar 

  39. Sun, Y., Oberley, L. W., & Li, Y. (1988). A simple method for clinical assay of superoxide dismutase. Clinical Chemistry, 34, 497–500.

    CAS  PubMed  Google Scholar 

  40. Keshari, R. S., Verma, A., Barthwal, M. K., & Dikshit, M. (2013). Reactive oxygen species-induced activation of ERK and p38 MAPK mediates PMA-induced NETs release from human neutrophils. Journal of Cellular Biochemistry, 114, 532–540.

    Article  CAS  PubMed  Google Scholar 

  41. Thayer, W. S. (1990). Superoxide-dependent and superoxide-independent pathways for reduction of nitroblue tetrazolium in isolated rat cardiac myocytes. Archives of Biochemistry and Biophysics, 276, 139–145.

    Article  CAS  PubMed  Google Scholar 

  42. Schor, N. A., Stedman, R. B., Epstein, N., & Schally, G. (1982). Rat splenic D-T diaphorase and NAD(P)H-nitroblue tetrazolium reductase. Their use to assess the action of polycyclic hydrocarbons in the lymphatic system. Virchows Archiv. B, Cell Pathology Including Molecular Pathology, 41, 83–93.

    Article  CAS  PubMed  Google Scholar 

  43. Auclair, C., Torres, M., & Hakim, J. (1978). Superoxide anion involvement in nbt reduction catalyzed by NADPH-cytochrome P-450 reductase: A pitfall. FEBS Letters, 89, 26–28.

    Article  CAS  PubMed  Google Scholar 

  44. Picker, S. D., & Fridovich, I. (1984). On the mechanism of production of superoxide radical by reaction mixtures containing NADH, phenazine methosulfate, and nitroblue tetrazolium. Archives of Biochemistry and Biophysics, 228, 155–158.

    Article  CAS  PubMed  Google Scholar 

  45. Faulkner, K., & Fridovich, I. (1993). Luminol and lucigenin as detectors for O2. Free Radical Biology & Medicine, 15, 447–451.

    Article  CAS  Google Scholar 

  46. Liochev, S. I., & Fridovich, I. (1997). Lucigenin (bis-n-methylacridinium) as a mediator of superoxide anion production. Archives of Biochemistry and Biophysics, 337, 115–120.

    Article  CAS  PubMed  Google Scholar 

  47. Vasquez-Vivar, J., Hogg, N., Pritchard, Jr., K. A., Martasek, P., & Kalyanaraman, B. (1997). Superoxide anion formation from lucigenin: An electron spin resonance spin-trapping study. FEBS Letters, 403, 127–130.

    Article  CAS  PubMed  Google Scholar 

  48. Vasquez-Vivar, J., Martasek, P., Hogg, N., Karoui, H., Masters, B. S., Pritchard, Jr., K. A., & Kalyanaraman, B. (1999). Electron spin resonance spin-trapping detection of superoxide generated by neuronal nitric oxide synthase. Methods in Enzymology, 301, 169–177.

    Article  CAS  PubMed  Google Scholar 

  49. Wardman, P., Burkitt, M. J., Patel, K. B., Lawrence, A., Jones, C. M., Everett, S. A., & Vojnovic, B. (2002). Pitfalls in the use of common luminescent probes for oxidative and nitrosative stress. Journal of Fluorescence, 12, 65–68.

    Article  Google Scholar 

  50. Rezende, F., Prior, K. K., Lowe, O., Wittig, I., Strecker, V., Moll, F., Helfinger, V., Schnutgen, F., Kurrle, N., Wempe, F., Walter, M., Zukunft, S., Luck, B., Fleming, I., Weissmann, N., Brandes, R. P., & Schroder, K. (2017). Cytochrome P450 enzymes but not NADPH oxidases are the source of the NADPH-dependent lucigenin chemiluminescence in membrane assays. Free Radical Biology & Medicine, 102, 57–66.

    Article  CAS  Google Scholar 

  51. Rezende, F., Lowe, O., Helfinger, V., Prior, K. K., Walter, M., Zukunft, S., Fleming, I., Weissmann, N., Brandes, R. P., & Schroder, K. (2016). Unchanged NADPH oxidase activity in Nox1-Nox2-Nox4 triple knockout mice: What do NADPH-stimulated chemiluminescence assays really detect? Antioxidants & Redox Signaling, 24, 392–399.

    Article  CAS  Google Scholar 

  52. Nishinaka, Y., Aramaki, Y., Yoshida, H., Masuya, H., Sugawara, T., & Ichimori, Y. (1993). A new sensitive chemiluminescence probe, L-012, for measuring the production of superoxide anion by cells. Biochemical and Biophysical Research Communications, 193, 554–559.

    Article  CAS  PubMed  Google Scholar 

  53. Imada, I., Sato, E. F., Miyamoto, M., Ichimori, Y., Minamiyama, Y., Konaka, R., & Inoue, M. (1999). Analysis of reactive oxygen species generated by neutrophils using a chemiluminescence probe L-012. Analytical Biochemistry, 271, 53–58.

    Article  CAS  PubMed  Google Scholar 

  54. Sohn, H. Y., Gloe, T., Keller, M., Schoenafinger, K., & Pohl, U. (1999). Sensitive superoxide detection in vascular cells by the new chemiluminescence dye L-012. Journal of Vascular Research, 36, 456–464.

    Article  CAS  PubMed  Google Scholar 

  55. Daiber, A., August, M., Baldus, S., Wendt, M., Oelze, M., Sydow, K., Kleschyov, A. L., & Munzel, T. (2004). Measurement of NAD(P)H oxidase-derived superoxide with the luminol analogue L-012. Free Radical Biology & Medicine, 36, 101–111.

    Article  CAS  Google Scholar 

  56. Merenyi, G., Lind, J., & Eriksen, T. E. (1990). Luminol chemiluminescence: Chemistry, excitation, emitter. Journal of Bioluminescence and Chemiluminescence, 5, 53–56.

    Article  CAS  PubMed  Google Scholar 

  57. Zielonka, J., Lambeth, J. D., & Kalyanaraman, B. (2013). On the use of L-012, a luminol-based chemiluminescent probe, for detecting superoxide and identifying inhibitors of NADPH oxidase: A reevaluation. Free Radical Biology & Medicine, 65, 1310–1314.

    Article  CAS  Google Scholar 

  58. Li, Y., Ganesh, T., Diebold, B. A., Zhu, Y., McCoy, J. W., Smith, S. M., Sun, A., & Lambeth, J. D. (2015). Thioxo-dihydroquinazolin-one compounds as novel inhibitors of myeloperoxidase. ACS Medicinal Chemistry Letters, 6, 1047–1052.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  59. Rothe, G., & Valet, G. (1990). Flow cytometric analysis of respiratory burst activity in phagocytes with hydroethidine and 2’,7’-dichlorofluorescin. Journal of Leukocyte Biology, 47, 440–448.

    CAS  PubMed  Google Scholar 

  60. Zhao, H., Joseph, J., Fales, H. M., Sokoloski, E. A., Levine, R. L., Vasquez-Vivar, J., & Kalyanaraman, B. (2005). Detection and characterization of the product of hydroethidine and intracellular superoxide by HPLC and limitations of fluorescence. Proceedings of the National Academy of Sciences of the United States of America, 102, 5727–5732.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  61. Michalski, R., Michalowski, B., Sikora, A., Zielonka, J., & Kalyanaraman, B. (2014). On the use of fluorescence lifetime imaging and dihydroethidium to detect superoxide in intact animals and ex vivo tissues: A reassessment. Free Radical Biology & Medicine, 67, 278–284.

    Article  CAS  Google Scholar 

  62. Zielonka, J., & Kalyanaraman, B. (2010). Hydroethidine- and MitoSOX-derived red fluorescence is not a reliable indicator of intracellular superoxide formation: Another inconvenient truth. Free Radical Biology & Medicine, 48, 983–1001.

    Article  CAS  Google Scholar 

  63. Zielonka, J., Zhao, H., Xu, Y., & Kalyanaraman, B. (2005). Mechanistic similarities between oxidation of hydroethidine by fremy’s salt and superoxide: Stopped-flow optical and EPR studies. Free Radical Biology & Medicine, 39, 853–863.

    Article  CAS  Google Scholar 

  64. Zielonka, J., Sarna, T., Roberts, J. E., Wishart, J. F., & Kalyanaraman, B. (2006). Pulse radiolysis and steady-state analyses of the reaction between hydroethidine and superoxide and other oxidants. Archives of Biochemistry and Biophysics, 456, 39–47.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  65. Zielonka, J., Srinivasan, S., Hardy, M., Ouari, O., Lopez, M., Vasquez-Vivar, J., Avadhani, N. G., & Kalyanaraman, B. (2008). Cytochrome c-mediated oxidation of hydroethidine and mito-hydroethidine in mitochondria: Identification of homo- and heterodimers. Free Radical Biology & Medicine, 44, 835–846.

    Article  CAS  Google Scholar 

  66. Maghzal, G. J., Cergol, K. M., Shengule, S. R., Suarna, C., Newington, D., Kettle, A. J., Payne, R. J., & Stocker, R. (2014). Assessment of myeloperoxidase activity by the conversion of hydroethidine to 2-chloroethidium. The Journal of Biological Chemistry, 289, 5580–5595.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  67. Talib, J., Maghzal, G. J., Cheng, D., & Stocker, R. (2016). Detailed protocol to assess in vivo and ex vivo myeloperoxidase activity in mouse models of vascular inflammation and disease using hydroethidine. Free Radical Biology & Medicine, 97, 124–135.

    Article  CAS  Google Scholar 

  68. Kalyanaraman, B., Dranka, B. P., Hardy, M., Michalski, R., & Zielonka, J. (2014). HPLC-based monitoring of products formed from hydroethidine-based fluorogenic probes--the ultimate approach for intra- and extracellular superoxide detection. Biochimica et Biophysica Acta, 1840, 739–744.

    Article  CAS  PubMed  Google Scholar 

  69. Zielonka, J., Hardy, M., & Kalyanaraman, B. (2009). HPLC study of oxidation products of hydroethidine in chemical and biological systems: Ramifications in superoxide measurements. Free Radical Biology & Medicine, 46, 329–338.

    Article  CAS  Google Scholar 

  70. Zielonka, J., Vasquez-Vivar, J., & Kalyanaraman, B. (2008). Detection of 2-hydroxyethidium in cellular systems: A unique marker product of superoxide and hydroethidine. Nature Protocols, 3, 8–21.

    Article  CAS  PubMed  Google Scholar 

  71. Fernandes, D. C., Gonçalves, R. C., & Laurindo, F. R. M. (2017). Measurement of superoxide production and NADPH oxidase activity by HPLC analysis of dihydroethidium oxidation. In R. M. Touyz & E. L. Schiffrin (Ed.), Hypertension: Methods and protocols (pp. 233–249). New York, NY: Springer New York.

    Chapter  Google Scholar 

  72. Michalski, R., Zielonka, J., Hardy, M., Joseph, J., & Kalyanaraman, B. (2013). Hydropropidine: A novel, cell-impermeant fluorogenic probe for detecting extracellular superoxide. Free Radical Biology & Medicine, 54, 135–147.

    Article  CAS  Google Scholar 

  73. Ouari, O., Hardy, M., Karoui, H., & Tordo, P. (2011). Recent developments and applications of the coupled EPR/spin trapping technique (EPR/st). Electron Paramagnetic Resonance, 22, 1–40.

    CAS  Google Scholar 

  74. Abbas, K., Babic, N., & Peyrot, F. (2016). Use of spin traps to detect superoxide production in living cells by electron paramagnetic resonance (EPR) spectroscopy. Methods, 109, 31–43.

    Article  CAS  PubMed  Google Scholar 

  75. Davies, M. J. (2016). Detection and characterisation of radicals using electron paramagnetic resonance (EPR) spin trapping and related methods. Methods, 109, 21–30.

    Article  CAS  PubMed  Google Scholar 

  76. Britigan, B. E., Rosen, G. M., Chai, Y., & Cohen, M. S. (1986). Do human neutrophils make hydroxyl radical? Determination of free radicals generated by human neutrophils activated with a soluble or particulate stimulus using electron paramagnetic resonance spectrometry. The Journal of Biological Chemistry, 261, 4426–4431.

    CAS  PubMed  Google Scholar 

  77. Britigan, B. E., Rosen, G. M., Thompson, B. Y., Chai, Y., & Cohen, M. S. (1986). Stimulated human neutrophils limit iron-catalyzed hydroxyl radical formation as detected by spin-trapping techniques. The Journal of Biological Chemistry, 261, 17026–17032.

    CAS  PubMed  Google Scholar 

  78. Pou, S., Cohen, M. S., Britigan, B. E., & Rosen, G. M. (1989). Spin-trapping and human neutrophils. Limits of detection of hydroxyl radical. The Journal of Biological Chemistry, 264, 12299–12302.

    CAS  PubMed  Google Scholar 

  79. Zielonka, J., Cheng, G., Zielonka, M., Ganesh, T., Sun, A., Joseph, J., Michalski, R., O’Brien, W. J., Lambeth, J. D., & Kalyanaraman, B. (2014). High-throughput assays for superoxide and hydrogen peroxide: Design of a screening workflow to identify inhibitors of NADPH oxidases. The Journal of Biological Chemistry, 289, 16176–16189.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  80. Abbas, K., Hardy, M., Poulhes, F., Karoui, H., Tordo, P., Ouari, O., & Peyrot, F. (2015). Medium-throughput ESR detection of superoxide production in undetached adherent cells using cyclic nitrone spin traps. Free Radical Research, 49, 1122–1128.

    Article  CAS  PubMed  Google Scholar 

  81. Hardy, M., Bardelang, D., Karoui, H., Rockenbauer, A., Finet, J. P., Jicsinszky, L., Rosas, R., Ouari, O., & Tordo, P. (2009). Improving the trapping of superoxide radical with a beta-cyclodextrin- 5-diethoxyphosphoryl-5-methyl-1-pyrroline-n-oxide (DEPMPO) conjugate. Chemistry, 15, 11114–11118.

    Article  CAS  PubMed  Google Scholar 

  82. Zhou, M., Diwu, Z., Panchuk-Voloshina, N., & Haugland, R. P. (1997). A stable nonfluorescent derivative of resorufin for the fluorometric determination of trace hydrogen peroxide: Applications in detecting the activity of phagocyte NADPH oxidase and other oxidases. Analytical Biochemistry, 253, 162–168.

    Article  CAS  PubMed  Google Scholar 

  83. Votyakova, T. V., & Reynolds, I. J. (2004). Detection of hydrogen peroxide with Amplex Red: Interference by NADH and reduced glutathione auto-oxidation. Archives of Biochemistry and Biophysics, 431, 138–144.

    Article  CAS  PubMed  Google Scholar 

  84. Kashem, M. A., & Dunford, H. B. (1986). Kinetics of the oxidation of reduced nicotinamide adenine dinucleotide by horseradish peroxidase compounds I and II. Biochemistry and Cell Biology, 64, 323–327.

    Article  CAS  PubMed  Google Scholar 

  85. Harman, L. S., Carver, D. K., Schreiber, J., & Mason, R. P. (1986). One- and two-electron oxidation of reduced glutathione by peroxidases. The Journal of Biological Chemistry, 261, 1642–1648.

    CAS  PubMed  Google Scholar 

  86. Zhao, B., Summers, F. A., & Mason, R. P. (2012). Photooxidation of Amplex Red to resorufin: Implications of exposing the Amplex Red assay to light. Free Radical Biology & Medicine, 53, 1080–1087.

    Article  CAS  Google Scholar 

  87. Debski, D., Smulik, R., Zielonka, J., Michalowski, B., Jakubowska, M., Debowska, K., Adamus, J., Marcinek, A., Kalyanaraman, B., & Sikora, A. (2016). Mechanism of oxidative conversion of Amplex(R) Red to resorufin: Pulse radiolysis and enzymatic studies. Free Radical Biology & Medicine, 95, 323–332.

    Article  CAS  Google Scholar 

  88. Summers, F. A., Zhao, B., Ganini, D., & Mason, R. P. (2013). Photooxidation of Amplex Red to resorufin: Implications of exposing the Amplex Red assay to light. Methods in Enzymology, 526, 1–17.

    Article  CAS  PubMed  Google Scholar 

  89. Zhao, B., Ranguelova, K., Jiang, J., & Mason, R. P. (2011). Studies on the photosensitized reduction of resorufin and implications for the detection of oxidative stress with Amplex Red. Free Radical Biology & Medicine, 51, 153–159.

    Article  CAS  Google Scholar 

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

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  91. Zielonka, J., Sikora, A., Hardy, M., Joseph, J., Dranka, B. P., & Kalyanaraman, B. (2012). Boronate probes as diagnostic tools for real time monitoring of peroxynitrite and hydroperoxides. Chemical Research in Toxicology, 25, 1793–1799.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  92. Sikora, A., Zielonka, J., Lopez, M., Joseph, J., & Kalyanaraman, B. (2009). Direct oxidation of boronates by peroxynitrite: Mechanism and implications in fluorescence imaging of peroxynitrite. Free Radical Biology & Medicine, 47, 1401–1407.

    Article  CAS  Google Scholar 

  93. Zielonka, J., Sikora, A., Joseph, J., & Kalyanaraman, B. (2010). Peroxynitrite is the major species formed from different flux ratios of co-generated nitric oxide and superoxide: Direct reaction with boronate-based fluorescent probe. The Journal of Biological Chemistry, 285, 14210–14216.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  94. Sikora, A., Zielonka, J., Lopez, M., Dybala-Defratyka, A., Joseph, J., Marcinek, A., & Kalyanaraman, B. (2011). Reaction between peroxynitrite and boronates: EPR spin-trapping, HPLC analyses, and quantum mechanical study of the free radical pathway. Chemical Research in Toxicology, 24, 687–697.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  95. Zielonka, J., Zielonka, M., Sikora, A., Adamus, J., Joseph, J., Hardy, M., Ouari, O., Dranka, B. P., & Kalyanaraman, B. (2012). Global profiling of reactive oxygen and nitrogen species in biological systems: High-throughput real-time analyses. The Journal of Biological Chemistry, 287, 2984–2995.

    Article  CAS  PubMed  Google Scholar 

  96. Michalski, R., Zielonka, J., Gapys, E., Marcinek, A., Joseph, J., & Kalyanaraman, B. (2014). Real-time measurements of amino acid and protein hydroperoxides using coumarin boronic acid. The Journal of Biological Chemistry, 289, 22536–22553.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  97. Zielonka, J., Sikora, A., Adamus, J., & Kalyanaraman, B. (2015). Detection and differentiation between peroxynitrite and hydroperoxides using mitochondria-targeted arylboronic acid. Methods in Molecular Biology, 1264, 171–181.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  98. Zielonka, J., Podsiadly, R., Zielonka, M., Hardy, M., & Kalyanaraman, B. (2016). On the use of peroxy-caged luciferin (PCL-1) probe for bioluminescent detection of inflammatory oxidants in vitro and in vivo—identification of reaction intermediates and oxidant-specific minor products. Free Radical Biology & Medicine, 99, 32–42.

    Article  CAS  Google Scholar 

  99. Smulik, R., Debski, D., Zielonka, J., Michalowski, B., Adamus, J., Marcinek, A., Kalyanaraman, B., & Sikora, A. (2014). Nitroxyl (HNO) reacts with molecular oxygen and forms peroxynitrite at physiological pH. Biological implications. The Journal of Biological Chemistry, 289, 35570–35581.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  100. Woolley, J. F., Naughton, R., Stanicka, J., Gough, D. R., Bhatt, L., Dickinson, B. C., Chang, C. J., & Cotter, T. G. (2012). H2O2 production downstream of FLT3 is mediated by p22phox in the endoplasmic reticulum and is required for STAT5 signalling. PLoS ONE, 7, e34050.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  101. Dickinson, B. C., Peltier, J., Stone, D., Schaffer, D. V., & Chang, C. J. (2011). Nox2 redox signaling maintains essential cell populations in the brain. Nature Chemical Biology, 7, 106–112.

    Article  CAS  PubMed  Google Scholar 

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

    Article  CAS  PubMed  Google Scholar 

  103. Nisimoto, Y., Diebold, B. A., Cosentino-Gomes, D., & Lambeth, J. D. (2014). Nox4: A hydrogen peroxide-generating oxygen sensor. Biochemistry, 53, 5111–5120.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  104. Serrander, L., Cartier, L., Bedard, K., Banfi, B., Lardy, B., Plastre, O., Sienkiewicz, A., Forro, L., Schlegel, W., & Krause, K. H. (2007). Nox4 activity is determined by mRNA levels and reveals a unique pattern of ROS generation. The Biochemical Journal, 406, 105–114.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  105. Takac, I., Schroder, K., Zhang, L., Lardy, B., Anilkumar, N., Lambeth, J. D., Shah, A. M., Morel, F., & Brandes, R. P. (2011). The E-loop is involved in hydrogen peroxide formation by the NADPH oxidase Nox4. The Journal of Biological Chemistry, 286, 13304–13313.

    Article  CAS  PubMed  PubMed Central  Google Scholar 

  106. Heumuller, S., Wind, S., Barbosa-Sicard, E., Schmidt, H. H., Busse, R., Schroder, K., & Brandes, R. P. (2008). Apocynin is not an inhibitor of vascular NADPH oxidases but an antioxidant. Hypertension, 51, 211–217.

    Article  PubMed  CAS  Google Scholar 

Download references

Funding

This work was supported by a grant from NIH (R01 AA022986) to B.K. and from the French National Research Agency (ANR-16-CE07-0023-01) to O.O. and M.H. A.S. and R.M. were supported by a grant from Polish National Science Centre, No. 2015/18/E/ST4/00235.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Jacek Zielonka.

Ethics declarations

Conflict of Interests

The authors declare that they have no competing interests.

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Zielonka, J., Hardy, M., Michalski, R. et al. Recent Developments in the Probes and Assays for Measurement of the Activity of NADPH Oxidases. Cell Biochem Biophys 75, 335–349 (2017). https://doi.org/10.1007/s12013-017-0813-6

Download citation

  • Received:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s12013-017-0813-6

Keywords

Navigation