Summary

Alternative Methods for the Detection of Superoxide Anion Generation in Platelets

Published: March 29, 2024
doi:

Summary

The generation of superoxide anion is essential for the stimulation of platelets and, if dysregulated, critical for thrombotic diseases. Here, we present three protocols for the selective detection of superoxide anions and the study of redox-dependent platelet regulation.

Abstract

Reactive oxygen species (ROS) are highly unstable oxygen-containing molecules. Their chemical instability makes them extremely reactive and gives them the ability to react with important biological molecules such as proteins, nucleic acids, and lipids. Superoxide anions are important ROS generated by the reduction of molecular oxygen reduction (i.e., acquisition of one electron). Despite their initial implication exclusively in aging, degenerative, and pathogenic processes, their participation in important physiological responses has recently become apparent. In the vascular system, superoxide anions have been shown to modulate the differentiation and function of vascular smooth muscle cells, the proliferation and migration of vascular endothelial cells in angiogenesis, the immune response, and the activation of platelets in hemostasis. The role of superoxide anions is particularly important in the dysregulation of platelets and the cardiovascular complications associated with a plethora of conditions, including cancer, infection, inflammation, diabetes, and obesity. It has, therefore, become extremely relevant in cardiovascular research to be able to effectively measure the generation of superoxide anions by human platelets, understand the redox-dependent mechanisms regulating the balance between hemostasis and thrombosis and, eventually, identify novel pharmacological tools for the modulation of platelet responses leading to thrombosis and cardiovascular complications. This study presents three experimental protocols successfully adopted for the detection of superoxide anions in platelets and the study of the redox-dependent mechanisms regulating hemostasis and thrombosis: 1) dihydroethidium (DHE)-based superoxide anion detection by flow cytometry; 2) DHE-based superoxide anion visualization and analysis by single platelet imaging; and 3) spin probe-based quantification of superoxide anion output in platelets by electron paramagnetic resonance (EPR).

Introduction

The superoxide anion (O2•-) is the most functionally relevant ROS generated in platelets1. O2•- is the product of the reduction of molecular oxygen and the precursor of many different ROS 2. The dismutation of O2•- leads to the generation of hydrogen peroxide (H2O2) via spontaneous reactions in aqueous solution or reactions catalyzed by superoxide dismutases (SODs3). Although different enzymatic sources have been suggested (e.g., xanthine oxidase4, lipoxygenase5, cyclooxygenase6, and nitric oxide synthase7), mitochondrial respiration8,9 and nicotinamide adenine dinucleotide phosphate-oxidases (NOXs)10 are the most prominent sources of superoxide anion in eukaryotic cells. This also seems to be the case in platelets, where electron leakage from mitochondrial respiration11,12 and the enzymatic activity of NOXs13,14 have been described as the main contributors to the superoxide anion output.

Although several studies have focused on the regulation of platelets by O2•-, there is no consensus regarding the molecular mechanisms responsible. The modulation of surface receptor activity via direct oxidation and disulfide bond formation has been proposed for different platelet receptors. The positive regulation of integrin αIIbβ3 by ROS via direct oxidation of cysteine residues has been suggested15,16,17. Similarly, since platelet responses to collagen depend on disulfide-dependent dimerization and consequent dimerization of the glycoprotein VI (GPVI)18, receptor activity potentiation by ROS-dependent oxidation has been proposed19, although not fully proven experimentally. Finally, ROS-induced oxidation of the sulfhydryl groups of glycoprotein Ib (GPIb) was shown to promote platelet adhesion and platelet-leukocyte interaction during inflammation20. Conversely, as a possible consequence of decreased sulfhydryl group oxidation and receptor activation, the shedding of the ectodomain of both GPVI and GPIb is diminished by reducing conditions21.

Modes of action independently of a direct oxidation of platelet surface receptors have also been proposed. ROS, including O2•-, have been shown to positively modulate the collagen receptor GPVI by attenuating the activity of the Src homology region 2-containing protein tyrosine phosphatase 2 (SHP-2), which negatively regulates the signaling cascade of this receptor22. Moreover, O2•- can generate ONOO(peroxynitrite) by rapid reaction with nitric oxide (NO), which normally inhibits platelets through the NO-sensitive guanylyl cyclase (NO-GC) and the generation of the negative platelet regulator cyclic GMP (cGMP)23,24. The resulting decrease in NO levels can lead to platelet potentiation. Alternatively, the generation of O2•- by NOX2 has been suggested to contribute to lipid peroxidation and isoprostane formation, which is essential for platelet activation and adhesion25. Finally, mitogen-activated protein kinase (MAPK) extracellular signal-regulated kinase 5 (ERK5), a protein kinase proposed as a redox stress sensor in platelets26, is activated by O2•- and induces a procoagulant phenotype in platelets (as estimated by flow cytometry-based measurement of phosphatidylserine externalization)27.

The dysregulation of O2•- and other ROS generation in platelets has been associated with the exaggerated hemostatic response leading to thrombotic cardiovascular complications associated with atherosclerosis, diabetes mellitus, hypertension, obesity, and cancer28,29. In these pathological settings, the ROS output by platelets is increased, which leads to a potentiation of their adhesive and aggregatory responses. In addition to the effect on platelet responses, the free radical output of platelets may have consequences on other blood cells and vascular structures, which is a poorly understood and underinvestigated area of cardiovascular health30. Despite our limited understanding of the molecular mechanisms linking oxidative stress to thrombotic conditions, the clinical relevance of antioxidants for the protection against cardiovascular disease has received considerable attention. Plasma antioxidant levels have been shown to inversely correlate with the risk of developing cardiovascular conditions, and dietary antioxidant consumption has been shown to protect against coronary artery disease31,32. Consequently, the use of dietary antioxidants has been advocated as a promising approach for cardiovascular disease prevention33,34,35. Amongst the effects of ROS generation in platelets, the increase in apoptosis can have important pathophysiological effects36,37. Overall, reliable protocols to detect and quantify the O2•- output by platelets are increasingly relevant in cardiovascular research.

Currently, available techniques for the detection of ROS have important limitations of specificity (i.e., the chemical nature of the oxidant molecules detected is unknown) and reliability (i.e., the unwanted interaction with biological molecules and experimental reagents leads to biased non-physiological results)38,39. The most commonly used approach for the detection of ROS in platelets is based on the use of dichlorodihydrofluorescein diacetate (DCFDA), which is converted to dichlorodihydrofluorescein (DCFH) by intracellular esterases and consequently to the highly fluorescent dichlorofluorescein (DCF) by cellular oxidants, including hydroxyl radicals and peroxidase-H2O2 intermediates40,41. Despite its wide use, serious questions have been raised regarding the reliability of this approach for the measurement of intracellular ROS38. The oxidation of DCFH to DCF can be, in fact, induced by transition metal ions (e.g., Fe2+) or heme-containing enzymes (e.g., cytochromes) instead of ROS42. Moreover, DCFDA is converted by cell peroxidases to its semiquinone free radical form (DCF•-), which is in turn oxidized to DCF by reaction with molecular oxygen (O2) with the release of O2•-, which leads to the artificial amplification of oxidative responses41,43,44. Therefore, the detection of intracellular ROS by DCFDA is useful for obtaining initial insights but requires cautious consideration and extensive experimental controls38,39.

This study presents three alternative techniques for the detection and measurement of the key regulator of platelet function O2•-1. The first technique is the detection using DHE and flow cytometry, which offers advantages of reliability and specificity over DCFDA. The second technique proposed here also utilizes DHE, but the detection method is live-platelet fluorescence imaging, which allows the study of the generation of O2•- upon platelet signaling with fast kinetics and single-cell resolution. Finally, a protocol based on the use of the hydroxylamine spin probe 1-hydroxy-3-methoxycarbonyl-2,2,5,5-tetramethylpyrrolidine (CMH) in EPR resonance experiments offers the possibility of quantifying the rate of O2•- generation by platelets and comparing it in different conditions.

Protocol

The collection of peripheral blood from consenting volunteers is approved by the local Ethics Committee and the National Health Service Health Research Authority (REC reference: 21/SC/0215; IRAS ID: 283854). 1. Method 1: Superoxide anion detection using DHE by flow cytometry Pre-warm (37 °C) sodium citrate solution (4% w/v) and use it as an anti-coagulant by adding it directly to the blood at the time of venepuncture at a ratio of 1:7 (0.5% w/v final). <…

Representative Results

For flow cytometry detection of DHE fluorescence, we show representative results for platelets either resting (Figure 3A) or stimulated with 0.1 unit/mL thrombin (Figure 3B). The O2•- output was quantified as platelet mean fluorescence intensity (MFI), as shown for stimulation with 0.1 unit/mL thrombin (Figure 3C) or 3 µg/mL collagen-related peptide (CRP) (Figure 3D</s…

Discussion

In this manuscript, we present three different techniques with the potential to advance the capability to investigate the redox-dependent regulation of platelet function via the selective detection of O2. The first two methods are an improvement on existing techniques because of the redox probe utilized (DHE instead of the more common but less reliable DCFDA). These techniques are, therefore, easily accessible, and most laboratories can adopt them effectively without particular eq…

Disclosures

The authors have nothing to disclose.

Acknowledgements

This work was funded by the British Heart Foundation (PG/15/40/31522), Alzheimer Research UK (ARUK-PG2017A-3), and European Research Council (#10102507) grants to G. Pula.

Materials

1-hydroxy-3-methoxycarbonyl-2,2,5,5-tetramethylpyrrolidine (CMH) Noxygen Science trasfer and Diagnostics GmbH NOX-02.1-50mg Reagent for EPR (spin probe)
BD FACSAria III BD Biosciences  NA Flow cytometer
Bovine Serum Albumin Merck/Sigma A7030 For μ-slide coating
Bruker E-scan M (Noxyscan) Noxygen Science trasfer and Diagnostics GmbH  NOX-E.11-BES  EPR spectrometer
Catalase–polyethylene glycol (PEG-Cat.) Merck/Sigma C4963 Hydrogen peroxide scavenger (specificity control)
ChronoLog Model 490+4 Labmedics/Chronolog NA Aggregometer
CM radical Noxygen Science trasfer and Diagnostics GmbH NOX-20.1-100mg  Reagent for EPR (calibration control)
deferoxamine  Noxygen Science trasfer and Diagnostics GmbH NOX-09.1-100mg  Reagent for EPR
diethyldithiocarbamate (DETC)  Noxygen Science trasfer and Diagnostics GmbH NOX-10.1-1g  Reagent for EPR
Dihydroethidium Thermo Fisher Scientifics D11347 Superoxide anion probe
Dimethyl sulfoxide Merck/Sigma 34869 For stock solution preparation 
EPR sealing wax plates Noxygen Science trasfer and Diagnostics GmbH NOX-A.3-VPM Consumable for EPR
EPR-grade water Noxygen Science trasfer and Diagnostics GmbH NOX-07.7.1-0.5L  Reagent for EPR
Fibrinogen from human plasma Merck/Sigma F4883 For μ-slide coating
FITC anti-human CD41 Antibody BioLegend 303704 Platelet-specific staining for flow cytometry
Glass cuvettes  Labmedics/Chronolog P/N 312 Consumable for incubation in aggregometer
Horm Collagen Labmedics/Chronolog P/N 385 For platelet stimulation
ImageJ  National Institutes of Health (NIH) NA ImageJ 1.53t (Wayne Rasband)
Indomethacin Merck/Sigma I7378 For platelet isolation
Micropipettes DURAN 50µl Noxygen Science trasfer and Diagnostics GmbH NOX-G.6.1-50µL Consumable for EPR
Poly-L-lysine hydrochloride Merck/Sigma P2658 For μ-slide coating
Prostaglandin E1 (PGE1) Merck/Sigma P5515 For platelet isolation
Sodium citrate (4% w/v solution) Merck/Sigma S5770 For platelet isolation
Stirring bars (Teflon-coated) Labmedics/Chronolog P/N 313 Consumable for incubation in aggregometer
Superoxide dismutase–polyethylene glycol (PEG-SOD) Merck/Sigma S9549 Superoxide anion scavenger (specificity control)
Thrombin from human plasma Merck/Sigma T6884 For platelet stimulation and μ-slide coating
VAS2870 Enzo Life Science BML-EI395 NOX inhibitor
Zeiss 510 LSM confocal microscope Zeiss NA Confocal microscope

References

  1. Vara, D., Cifuentes-Pagano, E., Pagano, P. J., Pula, G. A novel combinatorial technique for simultaneous quantification of oxygen radicals and aggregation reveals unexpected redox patterns in the activation of platelets by different physiopathological stimuli. Haematologica. 104 (9), 1879-1891 (2019).
  2. Fridovich, I. Superoxide radical: An endogenous toxicant. Annu Rev Pharmacol Toxicol. 23, 239-257 (1983).
  3. Keele, B. B., Mccord, J. M., Fridovich, I. Superoxide dismutase from Escherichia coli B. A new manganese-containing enzyme. J Biol Chem. 245 (22), 6176-6181 (1970).
  4. Berry, C. E., Hare, J. M. Xanthine oxidoreductase and cardiovascular disease: Molecular mechanisms and pathophysiological implications. J Physiol. 555, 589-606 (2004).
  5. Kim, C., Kim, J. Y., Kim, J. H. Cytosolic phospholipase a(2), lipoxygenase metabolites, and reactive oxygen species. BMB Rep. 41 (2), 555-559 (2008).
  6. Armstead, W. M., Mirro, R., Busija, D. W., Leffler, C. W. Postischemic generation of superoxide anion by newborn pig brain. Am J Physiol. 255 (2), H401-H403 (1988).
  7. Mayer, B., et al. Nitric oxide synthase-catalyzed activation of oxygen and reduction of cytochromes: Reaction mechanisms and possible physiological implications. J Cardiovasc Pharmacol. 20, S54-S56 (1992).
  8. Du, G., Mouithys-Mickalad, A., Sluse, F. E. Generation of superoxide anion by mitochondria and impairment of their functions during anoxia and reoxygenation in vitro. Free Radic Biol Med. 25 (9), 1066-1074 (1998).
  9. Turrens, J. F. Mitochondrial formation of reactive oxygen species. J Physiol. 552, 335-344 (2003).
  10. Jiang, F., Zhang, Y., Dusting, G. J. Nadph oxidase-mediated redox signaling: Roles in cellular stress response, stress tolerance, and tissue repair. Pharmacol Rev. 63 (1), 218-242 (2011).
  11. Wang, Z., et al. The role of mitochondria-derived reactive oxygen species in hyperthermia-induced platelet apoptosis. PLoS One. 8 (9), e75044 (2013).
  12. Wachowicz, B., Olas, B., Zbikowska, H. M., Buczynski, A. Generation of reactive oxygen species in blood platelets. Platelets. 13 (3), 175-182 (2002).
  13. Vara, D., et al. Nadph oxidases are required for full platelet activation in vitro and thrombosis in vivo but dispensable for plasma coagulation and hemostasis. Arterioscler Thromb Vasc Biol. 41 (2), 683-697 (2021).
  14. Violi, F., Pignatelli, P. Platelet nox, a novel target for anti-thrombotic treatment. Thromb Haemost. 111 (5), 817-823 (2014).
  15. Begonja, A. J., et al. Platelet NAD(P)H-oxidase-generated ros production regulates alphaiibbeta3-integrin activation independent of the NO/CGMP pathway. Blood. 106 (8), 2757-2760 (2005).
  16. Vara, D. S., et al. Autocrine amplification of integrin αIIbβ3 activation and platelet adhesive responses by deoxyribose-1-phosphate. Thromb Haemost. 109 (6), 1108-1119 (2013).
  17. Essex, D. W. The role of thiols and disulfides in platelet function. Antioxid Redox Signal. 6 (4), 736-746 (2004).
  18. Arthur, J. F., et al. Ligand binding rapidly induces disulfide-dependent dimerization of glycoprotein vi on the platelet plasma membrane. J Biol Chem. 282 (42), 30434-30441 (2007).
  19. Arthur, J. F., Gardiner, E. E., Kenny, D., Andrews, R. K., Berndt, M. C. Platelet receptor redox regulation. Platelets. 19 (1), 1-8 (2008).
  20. Kim, K., Li, J., Tseng, A., Andrews, R. K., Cho, J. Nox2 is critical for heterotypic neutrophil-platelet interactions during vascular inflammation. Blood. 126 (16), 1952-1964 (2015).
  21. Hosseini, E., Solouki, A., Roudsari, Z. O., Kargar, F., Ghasemzadeh, M. Reducing state attenuates ectodomain shedding of GPVI while restoring adhesion capacities of stored platelets: Evidence addressing the controversy around the effects of redox condition on thrombosis. J Thromb Thrombolysis. 50 (1), 123-134 (2020).
  22. Jang, J. Y., et al. Reactive oxygen species play a critical role in collagen-induced platelet activation via SHP-2 oxidation. Antioxid Redox Signal. 20 (16), 2528-2540 (2014).
  23. Beckman, J. S., Koppenol, W. H. Nitric oxide, superoxide, and peroxynitrite: The good, the bad, and ugly. Am J Physiol. 271 (5), C1424-C1437 (1996).
  24. Koppenol, W. H., Kissner, R., Beckman, J. S. Syntheses of peroxynitrite: To go with the flow or on solid grounds. Methods Enzymol. 269, 296-302 (1996).
  25. Cammisotto, V., et al. Nox2-mediated platelet activation by glycoprotein (GP) VI: Effect of rivaroxaban alone and in combination with aspirin. Biochem Pharmacol. 163, 111-118 (2019).
  26. Yang, M., et al. Platelet CD36 promotes thrombosis by activating redox sensor ERK5 in hyperlipidemic conditions. Blood. 129 (21), 2917-2927 (2017).
  27. Yang, M., et al. Platelet CD36 signaling through ERK5 promotes caspase-dependent procoagulant activity and fibrin deposition in vivo. Blood Adv. 2 (21), 2848-2861 (2018).
  28. Freedman, J. E. Oxidative stress and platelets. Arterioscler Thromb Vasc Biol. 28 (3), s11-s16 (2008).
  29. Masselli, E., et al. ROS in platelet biology: Functional aspects and methodological insights. Int J Mol Sci. 21 (14), 4866 (2020).
  30. Madamanchi, N. R., Vendrov, A., Runge, M. S. Oxidative stress and vascular disease. Arterioscler Thromb Vasc Biol. 25 (1), 29-38 (2005).
  31. Riemersma, R. A., et al. Risk of angina pectoris and plasma concentrations of vitamins a, c, and e and carotene. Lancet. 337 (8732), 1-5 (1991).
  32. Rimm, E. B., et al. Vitamin E consumption and the risk of coronary heart disease in men. N Engl J Med. 328 (20), 1450-1456 (1993).
  33. Zhang, W., Zheng, Y., Yan, F., Dong, M., Ren, Y. Research progress of quercetin in cardiovascular disease. Front Cardiovasc Med. 10, 1203713 (2023).
  34. Russell, C., Keshavamurthy, S., Saha, S. Nutraceuticals in the management of cardiovascular risk factors: Where is the evidence. Cardiovasc Hematol Disord Drug Targets. 21 (3), 150-161 (2021).
  35. Rotariu, D., et al. Oxidative stress – complex pathological issues concerning the hallmark of cardiovascular and metabolic disorders. Biomed Pharmacother. 152, 113238 (2022).
  36. Hosseini, E., Ghasemzadeh, M., Atashibarg, M., Haghshenas, M. R. O. S. scavenger, n-acetyl-l-cysteine and NOX specific inhibitor, VAS2870 reduce platelets apoptosis while enhancing their viability during storage. Transfusion. 59 (4), 1333-1343 (2019).
  37. Hosseini, E., Nodeh, F. K., Ghasemzadeh, M. Gamma irradiation induces a pro-apoptotic state in longer stored platelets, without progressing to an overt apoptosis by day 7 of storage. Apoptosis. 28 (7-8), 1141-1153 (2023).
  38. Griendling, K. K., et al. Measurement of reactive oxygen species, reactive nitrogen species, and redox-dependent signaling in the cardiovascular system: A scientific statement from the American heart association. Circ Res. 119 (5), e39-e75 (2016).
  39. Murphy, M. P., et al. Guidelines for measuring reactive oxygen species and oxidative damage in cells and in vivo. Nat Metab. 4 (6), 651-662 (2022).
  40. Hempel, S. L., Buettner, G. R., O’malley, Y. Q., Wessels, D. A., Flaherty, D. M. Dihydrofluorescein diacetate is superior for detecting intracellular oxidants: Comparison with 2′,7′-dichlorodihydrofluorescein diacetate, 5(and 6)-carboxy-2′,7′-dichlorodihydrofluorescein diacetate, and dihydrorhodamine 123. Free Radic Biol Med. 27 (1-2), 146-159 (1999).
  41. Kalyanaraman, B., et al. Measuring reactive oxygen and nitrogen species with fluorescent probes: Challenges and limitations. Free Radic Biol Med. 52 (1), 1-6 (2012).
  42. Burkitt, M. J., Wardman, P. Cytochrome c is a potent catalyst of dichlorofluorescin oxidation: Implications for the role of reactive oxygen species in apoptosis. Biochem Biophys Res Commun. 282 (1), 329-333 (2001).
  43. Rota, C., Fann, Y. C., Mason, R. P. Phenoxyl free radical formation during the oxidation of the fluorescent dye 2′,7′-dichlorofluorescein by horseradish peroxidase. Possible consequences for oxidative stress measurements. J Biol Chem. 274 (40), 28161-28168 (1999).
  44. Rota, C., Chignell, C. F., Mason, R. P. Evidence for free radical formation during the oxidation of 2′-7′-dichlorofluorescin to the fluorescent dye 2′-7′-dichlorofluorescein by horseradish peroxidase: Possible implications for oxidative stress measurements. Free Radic Biol Med. 27 (7-8), 873-881 (1999).
  45. Rodig, S. J. Attaching suspension cells to slides for staining. Cold Spring Harb Protoc. 2020 (12), (2020).
  46. Abubaker, A. A., Vara, D., Eggleston, I., Canobbio, I., Pula, G. A novel flow cytometry assay using dihydroethidium as redox-sensitive probe reveals NADPH oxidase-dependent generation of superoxide anion in human platelets exposed to amyloid peptide beta. Platelets. 30 (2), 181-189 (2019).
  47. Gaspar, R. S., et al. Protein disulphide isomerase and NADPH oxidase 1 cooperate to control platelet function and are associated with cardiometabolic disease risk factors. Antioxidants (Basel). 10 (3), 497 (2021).
  48. Gaspar, R. S., Ferreira, P. M., Mitchell, J. L., Pula, G., Gibbins, J. M. Platelet-derived extracellular vesicles express NADPH oxidase-1 (NOX-1), generate superoxide and modulate platelet function. Free Radic Biol Med. 165, 395-400 (2021).
  49. Vara, D., et al. NADPH oxidase 1 is a novel pharmacological target for the development of an antiplatelet drug without bleeding side effects. FASEB J. 34 (10), 13959-13977 (2020).
  50. Lopes-Pires, M. E., et al. Zinc regulates reactive oxygen species generation in platelets. Platelets. 32 (3), 368-377 (2021).
  51. Zielonka, J., Kalyanaraman, B. Hydroethidine- and mitoSOX-derived red fluorescence is not a reliable indicator of intracellular superoxide formation: Another inconvenient truth. Free Radic Biol Med. 48 (8), 983-1001 (2010).
  52. Linden, M. D. Platelet flow cytometry. Methods Mol Biol. 992, 241-262 (2013).
  53. Kauffman, M. E., et al. MitoSOX-based flow cytometry for detecting mitochondrial ROS. React Oxyg Species (Apex). 2 (5), 361-370 (2016).
  54. Sonkar, V. K., et al. NOX2 NADPH oxidase is dispensable for platelet activation or arterial thrombosis in mice. Blood Adv. 3 (8), 1272-1284 (2019).
  55. Dikalov, S. I., Kirilyuk, I. A., Voinov, M., Grigor’ev, I. A. EPR detection of cellular and mitochondrial superoxide using cyclic hydroxylamines. Free Radic Res. 45 (4), 417-430 (2011).
  56. Dikalov, S. I., Polienko, Y. F., Kirilyuk, I. Electron paramagnetic resonance measurements of reactive oxygen species by cyclic hydroxylamine spin probes. Antioxid Redox Signal. 28 (15), 1433-1443 (2018).
This article has been published
Video Coming Soon
Keep me updated:

.

Cite This Article
Vara, D., Leonard, S., Calaminus, S., Pula, G. Alternative Methods for the Detection of Superoxide Anion Generation in Platelets. J. Vis. Exp. (205), e66647, doi:10.3791/66647 (2024).

View Video