Elevated Cellular Oxidative Stress in Circulating Immune Cells in Otherwise Healthy Young People Who Use Electronic Cigarettes in a Cross-Sectional Single-Center Study: Implications for Future Cardiovascular Risk

Theodoros Kelesidis, Elizabeth Tran, Sara Arastoo, Karishma Lakhani, Rachel Heymans, Jeffrey Gornbein, Holly R Middlekauff, Theodoros Kelesidis, Elizabeth Tran, Sara Arastoo, Karishma Lakhani, Rachel Heymans, Jeffrey Gornbein, Holly R Middlekauff

Abstract

Background Tobacco cigarettes (TCs) increase oxidative stress and inflammation, both instigators of atherosclerotic cardiac disease. It is unknown if electronic cigarettes (ECs) also increase immune cell oxidative stress. We hypothesized an ordered, "dose-response" relationship, with tobacco-product type as "dose" (lowest in nonsmokers, intermediate in EC vapers, and highest in TC smokers), and the "response" being cellular oxidative stress (COS) in immune cell subtypes, in otherwise, healthy young people. Methods and Results Using flow cytometry and fluorescent probes, COS was determined in immune cell subtypes in 33 otherwise healthy young people: nonsmokers (n=12), EC vapers (n=12), and TC smokers (n=9). Study groups had similar baseline characteristics, including age, sex, race, and education level. A dose-response increase in proinflammatory monocytes and lymphocytes, and their COS content among the 3 study groups was found: lowest in nonsmokers, intermediate in EC vapers, and highest in TC smokers. These findings were most striking in CD14dimCD16+ and CD14++CD16+ proinflammatory monocytes and were reproduced with 2 independent fluorescent probes of COS. Conclusions These findings portend the development of premature cardiovascular disease in otherwise healthy young people who chronically vape ECs. On the other hand, that the COS is lower in EC vapers compared with TC smokers warrants additional investigation to determine if switching to ECs may form part of a harm-reduction strategy. Registration URL: https://www.clinicaltrials.gov; Unique identifier: NCT03823885.

Keywords: electronic cigarettes; monocytes; nicotine; reactive oxidative species; tobacco cigarettes.

Conflict of interest statement

None.

Figures

Figure 1. Frequency of immune cell types…
Figure 1. Frequency of immune cell types among smoker groups.
Flow cytometry was used to determine the percentage of different immune cell types in CD45+ immune cells (A through J). The compared groups were nonsmokers (NSs; white), electronic cigarette vapers (EC vapers; light gray), and tobacco cigarette smokers (TC smokers; dark gray). Summary of data (% cellular marker+ of parent population) is shown for CD45+CD15+CD16+CD14−hi‐SSC neutrophils (A), CD45+CD14++CD16− classic monocytes (B), CD45+CD14++CD16+ intermediate monocytes (C), CD45+CD14dimCD16+ nonclassic (patrolling or CD14+CD16++) monocytes (D), CD45+CD14+CD16+ total proinflammatory monocytes (intermediate and nonclassic) (E), CD45+CD3+ T cells (F), CD45+CD3+CD4+ T cells (G), CD45+CD3+CD8+ T cells (H), CD45+CD3−CD56+CD16+ natural killer (NK) cells (I), and CD45+CD19+ B cells (J). Data represent box‐and‐whisker boxes that display the minimum, mean, and maximum (n=9–12 participants per group). The ANOVA statistical test was used to compare 3 groups, and the t test was used to compare 2 groups. The trend P analysis tested the continuum of the difference in measures among groups in an ordered direction (NSs→EC vapers→TC smokers). *P<0.05, **P<0.01, ***P<0.001.
Figure 2. Cellular oxidative stress in CD45…
Figure 2. Cellular oxidative stress in CD45+ immune cells among smoker groups.
Flow cytometry was used to determine total (nuclear and cytoplasmic) and cytoplasmic reactive oxygen species. The compared groups were nonsmokers (NSs; white), electronic cigarette vapers (EC vapers; light gray), and tobacco cigarette smokers (TC smokers; dark gray). Representative data of percentage of immune (CD45+) cells that had positive staining for CELLROX Green among compared groups are shown (A). Summary of data for (A) is shown (B). Representative data of percentage of CD45+ cells that had positive staining for CELLROX Deep Red among compared groups are shown (C). Summary of data for C is shown (D). Representative data of CellROX Green change in mean fluorescence intensity (∆MFI) in CD45+ cells are shown (E). Fluorescence intensity of a positive cell population was compared with a negative cell population (fluorescence minus one negative control for staining) (∆MFI). Summary of data for E is shown (F). Representative data of CellROX Deep Red ∆MFI in CD45+ cells is shown (G). Summary of data for G is shown (H). Data represent box‐and‐whisker boxes that display the minimum, mean, and maximum (n=9–12 participants per group). The ANOVA statistical test was used to compare 3 groups, and the t test was used to compare 2 groups. The trend P analysis tested the continuum of the difference in measures among groups in an ordered direction (NSs→EC vapers→TC smokers). *P<0.05, **P<0.01. SSC indicates side scatter.
Figure 3. Cellular oxidative stress in neutrophils,…
Figure 3. Cellular oxidative stress in neutrophils, natural killer (NK) cells, and B cells among smoker groups.
Flow cytometry was used to determine total (nuclear and cytoplasmic) and cytoplasmic reactive oxygen species. The compared groups were nonsmokers (NSs; white), electronic cigarette vapers (EC vapers; light gray), and tobacco cigarette smokers (TC smokers; dark gray). Summary data of percentage of immune cells that had positive staining for CELLROX Green (A, E, I) and CELLROX Deep Red (C, G, K) and for change in mean fluorescence intensity (∆MFI) CellROX Green (B, F, J) and ∆MFI CellROX Deep Red in cells (D, H, L) among compared groups are shown for CD45+CD15+CD16+CD14−hi‐SSC neutrophils (A through D), CD45+CD3−CD56+CD16+ NK cells (E through H), CD45+CD19+ B cells (I through L). Data represent box‐and‐whisker boxes that display the minimum, mean, and maximum (n=9–12 participants per group). The ANOVA statistical test was used to compare 3 groups, and the t test was used to compare 2 groups. The trend P analysis tested the continuum of the difference in measures among groups in an ordered direction (NSs→EC vapers→TC smokers). *P<0.05, **P<0.01, ***P<0.001.
Figure 4. Cellular oxidative stress in T…
Figure 4. Cellular oxidative stress in T cell subsets among smoker groups.
Flow cytometry was used to determine total (nuclear and cytoplasmic) and cytoplasmic reactive oxygen species. The compared groups were nonsmokers (NSs; white), electronic cigarette vapers (EC vapers; light gray), and tobacco cigarette smokers (TC smokers; dark gray). Summary data of percentage of immune cells that had positive staining for CELLROX Green (A, E, and I) and CELLROX Deep Red (C, G, and K) and for change in mean fluorescence intensity (∆MFI) CellROX Green (B, F, and J) and ∆MFI CellROX Deep Red in cells (D, H, and L) among compared groups are shown for CD45+CD3+ T cells (A through D), CD45+CD3+CD4+ T cells (E through H), and CD45+CD3+CD8+ T cells (I through L). Data represent box‐and‐whisker boxes that display the minimum, mean, and maximum (n=9–12 participants per group). The ANOVA statistical test was used to compare 3 groups, and the t test was used to compare 2 groups. The trend P analysis tested the continuum of the difference in measures among groups in an ordered direction (NSs→EC vapers→TC smokers). *P<0.05, ***P<0.001.
Figure 5. Cellular oxidative stress in monocyte…
Figure 5. Cellular oxidative stress in monocyte subsets among smoker groups.
Flow cytometry was used to determine total (nuclear and cytoplasmic) and cytoplasmic reactive oxygen species. The compared groups were nonsmokers (NSs; white), electronic cigarette vapers (EC vapers; light gray), and tobacco cigarette smokers (TC smokers; dark gray). Summary data of percentage of immune cells that had positive staining for CELLROX Green (A, E, I, and M) and CELLROX Deep Red (C, G, K, and O) and for change in mean fluorescence intensity (∆MFI) CellROX Green (B, F, J, and N) and ∆MFI CellROX Deep Red in cells (D, H, L, and P) among compared groups are shown for CD45+CD15+CD16+CD14−hi‐SSC neutrophils (A through D), CD45+CD3−CD56+CD16+ natural killer cells (E through H), CD45+CD19+ B cells (I through L), and CD45+CD3+ T cells (M through P). Data represent box‐and‐whisker boxes that display the minimum, mean, and maximum (n=9–12 participants per group). The ANOVA statistical test was used to compare 3 groups, and the t test was used to compare 2 groups. The trend P analysis tested the continuum of the difference in measures among groups in an ordered direction (NSs→EC vapers→TC smokers). *P<0.05, **P<0.01, ***P<0.001. MNC indicates monocytes.
Figure 6. Ordered, “dose‐response” relationship in cellular…
Figure 6. Ordered, “dose‐response” relationship in cellular oxidative stress among immune cell types and smoker groups, with tobacco‐product type as “dose.”
EC indicates electronic cigarette; NK, natural killer; ROS, reactive oxygen species; and TC, tobacco cigarette.

References

    1. Sack MN, Fyhrquist FY, Saijonmaa OJ, Fuster V, Kovacic JC. Basic biology of oxidative stress and the cardiovascular system: part 1 of a 3-part series. J Am Coll Cardiol. 2017;70:196–211.
    1. Csiszar A, Podlutsky A, Wolin MS, Losonczy G, Pacher P, Ungvari Z. Oxidative stress and accelerated vascular aging: implications for cigarette smoking. Front Biosci (Landmark Ed). 2009;14:3128–3144.
    1. Ambrose JA, Barua RS. The pathophysiology of cigarette smoking and cardiovascular disease: an update. J Am Coll Cardiol. 2004;43:1731–1737.
    1. National Center for Chronic Disease Prevention and Health Promotion (US) Office on Smoking and Health . The Health Consequences of Smoking—50 Years of Progress: A Report of the Surgeon General. Atlanta, GA: Centers for Disease Control and Prevention (US); 2014.
    1. Pryor WA, Stone K. Oxidants in cigarette smoke: radicals, hydrogen peroxide, peroxynitrate, and peroxynitrite. Ann N Y Acad Sci. 1993;686:12–27; discussion 27-28.
    1. US Food & Drug Administration . Chemicals in Cigarettes: From Plant to Product to Puff. June 3, 2020. Available at: . Accessed June 23, 2020.
    1. Qiu F, Liang CL, Liu H, Zeng YQ, Hou S, Huang S, Lai X, Dai Z. Impacts of cigarette smoking on immune responsiveness: up and down or upside down? Oncotarget. 2017;8:268–284.
    1. Cathcart MK, Morel DW, Chisolm GM III. Monocytes and neutrophils oxidize low density lipoprotein making it cytotoxic. J Leukoc Biol. 1985;38:341–350.
    1. Shahab L, Goniewicz ML, Blount BC, Brown J, McNeill A, Alwis KU, Feng J, Wang L, West R. Nicotine, carcinogen, and toxin exposure in long‐term e‐cigarette and nicotine replacement therapy users: a cross‐sectional study. Ann Intern Med. 2017;166:390–400.
    1. Miech R, Johnston L, O'Malley PM, Bachman JG, Patrick ME. Trends in adolescent vaping, 2017–2019. N Engl J Med. 2019;381:1490–1491.
    1. Yang Z, Choi H. Single‐cell, time‐lapse reactive oxygen species detection in E. Coli . Curr Protoc Cell Biol. 2018;80:e60.
    1. Kalyanaraman B, Darley‐Usmar V, Davies KJ, Dennery PA, Forman HJ, Grisham MB, Mann GE, Moore K, Roberts LJ II, Ischiropoulos H. Measuring reactive oxygen and nitrogen species with fluorescent probes: challenges and limitations. Free Radic Biol Med. 2012;52:1–6.
    1. McBee ME, Chionh YH, Sharaf ML, Ho P, Cai MW, Dedon PC. Production of superoxide in bacteria is stress- and cell state‐dependent: a gating‐optimized flow cytometry method that minimizes ROS measurement artifacts with fluorescent dyes. Front Microbiol. 2017;8:459.
    1. Ray PD, Huang BW, Tsuji Y. Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling. Cell Signal. 2012;24:981–990.
    1. Pinke KH, Lima HG, Cunha FQ, Lara VS. Mast cells phagocyte Candida albicans and produce nitric oxide by mechanisms involving TLR2 and Dectin-1. Immunobiology. 2016;221:220–227.
    1. Esakky P, Hansen DA, Drury AM, Cusumano A, Moley KH. Cigarette smoke‐induced cell death of a spermatocyte cell line can be prevented by inactivating the aryl hydrocarbon receptor. Cell Death Discov. 2015;1:15050.
    1. Wang H, Yang Y, Chen H, Dan J, Cheng J, Guo S, Sun X, Wang W, Ai Y, Li S, et al. The predominant pathway of apoptosis in THP-1 macrophage‐derived foam cells induced by 5-aminolevulinic acid‐mediated sonodynamic therapy is the mitochondria‐caspase pathway despite the participation of endoplasmic reticulum stress. Cell Physiol Biochem. 2014;33:1789–1801.
    1. Bartholomew DJ. A test of homogeneity for ordered alternatives. Biometrika. 1959;46:36–48.
    1. Sager HB, Kessler T, Schunkert H. Monocytes and macrophages in cardiac injury and repair. J Thorac Dis. 2017;9:S30–S35.
    1. Poitou C, Dalmas E, Renovato M, Benhamo V, Hajduch F, Abdennour M, Kahn JF, Veyrie N, Rizkalla S, Fridman WH, et al. CD14dimCD16+ and CD14+CD16+ monocytes in obesity and during weight loss: relationships with fat mass and subclinical atherosclerosis. Arterioscler Thromb Vasc Biol. 2011;31:2322–2330.
    1. Feise RJ. Do multiple outcome measures require p‐value adjustment? BMC Med Res Methodol. 2002;2:8.
    1. Althouse AD. Adjust for multiple comparisons? It's not that simple. Ann Thorac Surg. 2016;101:1644–1645.
    1. Chen Y, Zhou Z, Min W. Mitochondria, oxidative stress and innate immunity. Front Physiol. 2018;9:1487.
    1. Chen J, Bullen C, Dirks K. A comparative health risk assessment of electronic cigarettes and conventional cigarettes. Int J Environ Res Public Health. 2017;14:382.
    1. Carnevale R, Sciarretta S, Violi F, Nocella C, Loffredo L, Perri L, Peruzzi M, Marullo AG, De Falco E, Chimenti I, et al. Acute impact of tobacco vs electronic cigarette smoking on oxidative stress and vascular function. Chest. 2016;150:606–612.
    1. Singh KP, Lawyer G, Muthumalage T, Maremanda KP, Khan NA, McDonough SR, Ye D, McIntosh S, Rahman I. Systemic biomarkers in electronic cigarette users: implications for noninvasive assessment of vaping‐associated pulmonary injuries. ERJ Open Res. 2019;5:00182 -2019.
    1. Reidel B, Radicioni G, Clapp PW, Ford AA, Abdelwahab S, Rebuli ME, Haridass P, Alexis NE, Jaspers I, Kesimer M. E‐cigarette use causes a unique innate immune response in the lung, involving increased neutrophilic activation and altered mucin secretion. Am J Respir Crit Care Med. 2018;197:492–501.
    1. Scott A, Lugg ST, Aldridge K, Lewis KE, Bowden A, Mahida RY, Grudzinska FS, Dosanjh D, Parekh D, Foronjy R, et al. Pro‐inflammatory effects of e‐cigarette vapour condensate on human alveolar macrophages. Thorax. 2018;73:1161–1169.
    1. Hiemstra PS, Bals R. Basic science of electronic cigarettes: assessment in cell culture and in vivo models. Respir Res. 2016;17:127.
    1. Moheimani RS, Bhetraratana M, Yin F, Peters KM, Gornbein J, Araujo JA, Middlekauff HR. Increased cardiac sympathetic activity and oxidative stress in habitual electronic cigarette users: implications for cardiovascular risk. JAMA Cardiol. 2017;2:278–284.
    1. Yoshida N, Yamamoto H, Shinke T, Otake H, Kuroda M, Terashita D, Takahashi H, Sakaguchi K, Hirota Y, Emoto T, et al. Impact of CD14(++)CD16(+) monocytes on plaque vulnerability in diabetic and non‐diabetic patients with asymptomatic coronary artery disease: a cross‐sectional study. Cardiovasc Diabetol. 2017;16:96.
    1. Kapellos TS, Bonaguro L, Gemund I, Reusch N, Saglam A, Hinkley ER, Schultze JL. Human monocyte subsets and phenotypes in major chronic inflammatory diseases. Front Immunol. 2019;10:2035.
    1. Rogacev KS, Cremers B, Zawada AM, Seiler S, Binder N, Ege P, Grosse‐Dunker G, Heisel I, Hornof F, Jeken J, et al. CD14++CD16+ monocytes independently predict cardiovascular events: a cohort study of 951 patients referred for elective coronary angiography. J Am Coll Cardiol. 2012;60:1512–1520.
    1. Cros J, Cagnard N, Woollard K, Patey N, Zhang SY, Senechal B, Puel A, Biswas SK, Moshous D, Picard C, et al. Human CD14dim monocytes patrol and sense nucleic acids and viruses via TLR7 and TLR8 receptors. Immunity. 2010;33:375–386.
    1. Urbanski K, Ludew D, Filip G, Filip M, Sagan A, Szczepaniak P, Grudzien G, Sadowski J, Jasiewicz‐Honkisz B, Sliwa T, et al. CD14(+)CD16(++) "nonclassical" monocytes are associated with endothelial dysfunction in patients with coronary artery disease. Thromb Haemost. 2017;117:971–980.
    1. Zhao C, Tan YC, Wong WC, Sem X, Zhang H, Han H, Ong SM, Wong KL, Yeap WH, Sze SK, et al. The CD14(+/low)CD16(+) monocyte subset is more susceptible to spontaneous and oxidant‐induced apoptosis than the CD14(+)CD16(-) subset. Cell Death Dis. 2010;1:e95.
    1. Brandes RP, Rezende F, Schroder K. Redox regulation beyond ROS: why ROS should not be measured as often. Circ Res. 2018;123:326–328.
    1. Dikalov SI, Harrison DG. Methods for detection of mitochondrial and cellular reactive oxygen species. Antioxid Redox Signal. 2014;20:372–382.
    1. Tanase M, Urbanska AM, Zolla V, Clement CC, Huang L, Morozova K, Follo C, Goldberg M, Roda B, Reschiglian P, et al. Role of carbonyl modifications on aging‐associated protein aggregation. Sci Rep. 2016;6:19311.
    1. Bone B, Seredick B, Olszowy M. Reactive oxygen probes—a broad range of colors with easier labeling and compatibility with fixation: novel CellROX® reagents from Molecular Probes® (P3295). J Immunol. 2013;190:211.215.
    1. Genov M, Kreiseder B, Nagl M, Drucker E, Wiederstein M, Muellauer B, Krebs J, Grohmann T, Pretsch D, Baumann K, et al. Tetrahydroanthraquinone derivative (+/-)-4-deoxyaustrocortilutein induces cell cycle arrest and apoptosis in melanoma cells via upregulation of p21 and p53 and downregulation of NF‐kappaB. J Cancer. 2016;7:555–568.
    1. Choi H, Yang Z, Weisshaar JC. Oxidative stress induced in E. coli by the human antimicrobial peptide ll-37. PLoS Pathog. 2017;13:e1006481.
    1. de Castro LS, de Assis PM, Siqueira AF, Hamilton TR, Mendes CM, Losano JD, Nichi M, Visintin JA, Assumpcao ME. Sperm oxidative stress is detrimental to embryo development: a dose‐dependent study model and a new and more sensitive oxidative status evaluation. Oxid Med Cell Longev. 2016;2016:8213071.
    1. He Y, Li W, Zheng Z, Zhao L, Li W, Wang Y, Li H. Inhibition of protein arginine methyltransferase 6 reduces reactive oxygen species production and attenuates aminoglycoside- and cisplatin‐induced hair cell death. Theranostics. 2020;10:133–150.
    1. Schattauer SS, Bedini A, Summers F, Reilly‐Treat A, Andrews MM, Land BB, Chavkin C. Reactive oxygen species (ROS) generation is stimulated by kappa opioid receptor activation through phosphorylated c‐Jun N‐terminal kinase and inhibited by p38 mitogen‐activated protein kinase (MAPK) activation. J Biol Chem. 2019;294:16884–16896.
    1. Rao VR, Lautz JD, Kaja S, Foecking EM, Lukacs E, Stubbs EB Jr. Mitochondrial‐targeted antioxidants attenuate TGF‐beta2 signaling in human trabecular meshwork cells. Invest Ophthalmol Vis Sci. 2019;60:3613–3624.
    1. Skonieczna M, Hudy D, Poterala‐Hejmo A, Hejmo T, Buldak RJ, Dziedzic A. Effects of resveratrol, berberine and their combinations on reactive oxygen species, survival and apoptosis in human squamous carcinoma (SCC-25) cells. Anticancer Agents Med Chem. 2019;19:1161–1171.
    1. Chovancova B, Hudecova S, Lencesova L, Babula P, Rezuchova I, Penesova A, Grman M, Moravcik R, Zeman M, Krizanova O. Melatonin‐induced changes in cytosolic calcium might be responsible for apoptosis induction in tumour cells. Cell Physiol Biochem. 2017;44:763–777.
    1. Lilli NL, Revy D, Robelet S, Lejeune B. Effect of the micro‐immunotherapy medicine 2LPARK((r)) on rat primary dopaminergic neurons after 6-OHDA injury: oxidative stress and survival evaluation in an in vitro model of Parkinson's disease. Degener Neurol Neuromuscul Dis. 2019;9:79–88.
    1. Stelmashook EV, Genrikhs EE, Kapkaeva MR, Zelenova EA, Isaev NK. N‐acetyl‐L‐cysteine in the presence of Cu(2+) induces oxidative stress and death of granule neurons in dissociated cultures of rat cerebellum. Biochemistry (Mosc). 2017;82:1176–1182.
    1. Isaev NK, Genrikhs EE, Aleksandrova OP, Zelenova EA, Stelmashook EV. Glucose deprivation stimulates Cu(2+) toxicity in cultured cerebellar granule neurons and Cu(2+)-dependent zinc release. Toxicol Lett. 2016;250-251:29–34.
    1. Ahn HY, Fairfull‐Smith KE, Morrow BJ, Lussini V, Kim B, Bondar MV, Bottle SE, Belfield KD. Two‐photon fluorescence microscopy imaging of cellular oxidative stress using profluorescent nitroxides. J Am Chem Soc. 2012;134:4721–4730.
    1. Rodrigues MB. An efficient technique to detect sperm reactive oxygen species: the CellROX Deep Red® fluorescent probe. Biochem Physiol. 2015;4:1–5.
    1. Plaza Davila M, Martin Munoz P, Tapia JA, Ortega Ferrusola C, Balao da Silva CC, Pena FJ. Inhibition of mitochondrial complex I leads to decreased motility and membrane integrity related to increased hydrogen peroxide and reduced ATP production, while the inhibition of glycolysis has less impact on sperm motility. PLoS One. 2015;10:e0138777.
    1. Korkmaz F, Malama E, Siuda M, Leiding C, Bollwein H. Effects of sodium pyruvate on viability, synthesis of reactive oxygen species, lipid peroxidation and DNA integrity of cryopreserved bovine sperm. Anim Reprod Sci. 2017;185:18–27.
    1. Varela E, Rey J, Plaza E, Munoz de Propios P, Ortiz‐Rodriguez JM, Alvarez M, Anel‐Lopez L, Anel L, De Paz P, Gil MC, et al. How does the microbial load affect the quality of equine cool‐stored semen? Theriogenology. 2018;114:212–220.
    1. Saidani C, Hammoudi‐Triki D, Laraba‐Djebari F, Taub M. In vitro studies with renal proximal tubule cells show direct cytotoxicity of androctonus australis hector scorpion venom triggered by oxidative stress, caspase activation and apoptosis. Toxicon. 2016;120:29–37.
    1. Celeghini ECC, Alves MBR, de Arruda RP, de Rezende GM, Florez‐Rodriguez SA, de Sa Filho MF. Efficiency of CellROX deep red((R)) and CellROX orange((R)) fluorescent probes in identifying reactive oxygen species in sperm samples from high and low fertility bulls. Anim Biotechnol. 2019:1–7. 10.1080/10495398.2019.1654485.
    1. Gallardo Bolanos JM, Balao da Silva CM, Martin Munoz P, Morillo Rodriguez A, Plaza Davila M, Rodriguez‐Martinez H, Aparicio IM, Tapia JA, Ortega Ferrusola C, Pena FJ. Phosphorylated AKT preserves stallion sperm viability and motility by inhibiting caspases 3 and 7. Reproduction. 2014;148:221–235.
    1. Wood JW, Bas E, Gupta C, Selman Y, Eshraghi A, Telischi FF, Van De Water TR. Otoprotective properties of mannitol against gentamicin induced hair cell loss. Otol Neurotol. 2014;35:e187–e194.
    1. Annu AS, Kaur G, Sharma P, Singh S, Ikram S. Evaluation of the antioxidant, antibacterial and anticancer (lung cancer cell line A549) activity of Punica granatum mediated silver nanoparticles. Toxicol Res (Camb). 2018;7:923–930.
    1. Chong CM, Zheng W. Artemisinin protects human retinal pigment epithelial cells from hydrogen peroxide‐induced oxidative damage through activation of ERK/CREB signaling. Redox Biol. 2016;9:50–56.
    1. DeLoughery Z, Luczak MW, Zhitkovich A. Monitoring cr intermediates and reactive oxygen species with fluorescent probes during chromate reduction. Chem Res Toxicol. 2014;27:843–851.
    1. Tormos AM, Talens‐Visconti R, Bonora‐Centelles A, Perez S, Sastre J. Oxidative stress triggers cytokinesis failure in hepatocytes upon isolation. Free Radic Res. 2015;49:927–934.
    1. Murota Y, Tabu K, Taga T. Requirement of ABC transporter inhibition and Hoechst 33342 dye deprivation for the assessment of side population‐defined C6 glioma stem cell metabolism using fluorescent probes. BMC Cancer. 2016;16:847.
    1. Sung HK, Song E, Jahng JWS, Pantopoulos K, Sweeney G. Iron induces insulin resistance in cardiomyocytes via regulation of oxidative stress. Sci Rep. 2019;9:4668.
    1. Leurgans TM, Bloksgaard M, Brewer JR, Bagatolli LA, Fredgart MH, Rosenstand K, Hansen ML, Rasmussen LM, Irmukhamedov A, De Mey JG. Endothelin-1 shifts the mediator of bradykinin‐induced relaxation from NO to H2 O2 in resistance arteries from patients with cardiovascular disease. Br J Pharmacol. 2016;173:1653–1664.
    1. Fang J, Zhao X, Li S, Xing X, Wang H, Lazarovici P, Zheng W. Protective mechanism of artemisinin on rat bone marrow‐derived mesenchymal stem cells against apoptosis induced by hydrogen peroxide via activation of c‐Raf‐Erk1/2-p90(rsk)-CREB pathway. Stem Cell Res Ther. 2019;10:312.
    1. Marrocco I, Altieri F, Peluso I. Measurement and clinical significance of biomarkers of oxidative stress in humans. Oxid Med Cell Longev. 2017;2017:6501046.
    1. Rouaud F, Romero‐Perez M, Wang H, Lobysheva I, Ramassamy B, Henry E, Tauc P, Giacchero D, Boucher JL, Deprez E, et al. Regulation of NADPH‐dependent nitric oxide and reactive oxygen species signalling in endothelial and melanoma cells by a photoactive NADPH analogue. Oncotarget. 2014;5:10650–10664.

Source: PubMed

3
Subscribe