Synthetic and semi-synthetic antioxidants in medicine and food industry: a review
Status PubMed-not-MEDLINE Language English Country Switzerland Media electronic-ecollection
Document type Journal Article, Review
PubMed
40766767
PubMed Central
PMC12321848
DOI
10.3389/fphar.2025.1599816
PII: 1599816
Knihovny.cz E-resources
- Keywords
- chelation therapy, free radical scavenging, oxidative stress, semi-synthetic antioxidants, synthetic antioxidants,
- Publication type
- Journal Article MeSH
- Review MeSH
Oxidative stress is recognized as both a causative and contributing factor in many human diseases. As a result, significant research has been devoted to the development of synthetic and semi-synthetic antioxidants (ATs). This review summarizes the therapeutic potential of synthetic ATs, explores their possible clinical applications, and highlights novel structural modifications aimed at improving their pharmacological properties. Additionally, it presents ideas for refining current antioxidant testing methodologies. Despite the ongoing research, the therapeutic efficacy of synthetic ATs remains ambiguous for several reasons. These include the following: therapeutic benefits resulting from non-antioxidant mechanisms, insufficient dosage to elicit an antioxidant effect, poor oral bioavailability, a narrow therapeutic index, or toxicity that precludes clinical use. Nevertheless, some compounds, such as ebselen, edaravone, MitoQ10, and potentially N-acetylcysteine, have shown promising results. However, further studies are needed to confirm their efficacy and clarify whether their therapeutic effects are truly mediated through antioxidant mechanisms. Dietary antioxidants have achieved relatively higher clinical success, although their toxicity has also led to the withdrawal of some agents. One emerging therapeutic strategy involves inhibition of NADPH oxidase (NOX) enzymatic activity, with compounds such as ebselen, S17834, and GKT137831 showing potential across various disease models. Efforts to enhance antioxidant properties through molecular modifications, using advanced technologies such as prodrug strategies, nanotechnology, polymer complexation, targeted delivery systems, or conversion into inhalable formulations, have yielded variable success. Still, confirming the clinical relevance of newly developed antioxidants will require a paradigm shift in the testing approaches. Future studies must better define the molecular context of antioxidant action, including the following: which biomolecules are being protected, the specific radical species targeted, the tissue and subcellular distribution of the antioxidant, and how levels of endogenous antioxidants and reactive oxygen species (ROS) change post-administration (e.g., within the mitochondria). Despite extensive research, only a few synthetic antioxidants, such as edaravone, are currently used in clinical practice. Currently, no new antioxidant drugs are expected to receive regulatory approval in the near future.
Biotechnology Research and Application Center Cukurova University Adana Türkiye
Department of Food Science Cornell University Ithaca NY United States
Department of Seafood Processing Technology Faculty of Fisheries Çukurova University Adana Türkiye
See more in PubMed
Abe K., Morita S., Kikuchi T., Itoyama Y. (1997). Protective effect of a novel free radical scavenger, OPC-14117, on wobbler mouse motor neuron disease. J. Neurosci. Res. 48, 63–70. 10.1002/(sici)1097-4547(19970401)48:1<63::aid-jnr6>3.3.co;2-5 PubMed DOI
Adlam V. J., Harrison J. C., Porteous C. M., James A. M., Smith R. A. J., Murphy M. P., et al. (2005). Targeting an antioxidant to mitochondria decreases cardiac ischemia-reperfusion injury. FASEB J. 19, 1088–1095. 10.1096/fj.05-3718com PubMed DOI
Agus D. B., Gambhir S. S., Pardridge W. M., Spielholz C., Baselga J., Vera J. C., et al. (1997). Vitamin C crosses the blood-brain barrier in the oxidized form through the glucose transporters. J. Clin. Invest. 100, 2842–2848. 10.1172/JCI119832 PubMed DOI PMC
Ahola T., Lapatto R., Raivio K. O., Selander B., Stigson L., Jonsson B., et al. (2003). N-acetylcysteine does not prevent bronchopulmonary dysplasia in immature infants: a randomized controlled trial. J. Pediatr. 143, 713–719. 10.1067/S0022-3476(03)00419-0 PubMed DOI
Aliev G., Obrenovich M. E., Reddy V. P., Shenk J. C., Moreira P. I., Nunomura A., et al. (2008). Antioxidant therapy in Alzheimer’s disease: theory and practice. Mini Rev. Med. Chem. 8, 1395–1406. 10.2174/138955708786369582 PubMed DOI PMC
Altenhöfer S., Radermacher K. A., Kleikers P. W. M., Wingler K., Schmidt H. H. H. W. (2015). Evolution of NADPH oxidase inhibitors: selectivity and mechanisms for target engagement. Antioxid. Redox Signal. 23, 406–427. 10.1089/ars.2013.5814 PubMed DOI PMC
Amponsah P. S., Yahya G., Zimmermann J., Mai M., Mergel S., Mühlhaus T., et al. (2021). Peroxiredoxins couple metabolism and cell division in an ultradian cycle. Nat. Chem. Biol. 17, 477–484. 10.1038/s41589-020-00728-9 PubMed DOI
Andrus P. K., Fleck T. J., Oostveen J. A., Hall E. D. (1997). Neuroprotective effects of the novel brain-penetrating pyrrolopyrimidine antioxidants U-101033E and U-104067F against post-ischemic degeneration of nigrostriatal neurons. J. Neurosci. Res. 47, 650–654. 10.1002/(SICI)1097-4547(19970315)47:6<650::AID-JNR11>3.0.CO;2-Z PubMed DOI
Antonic A., Dottori M., Macleod M. R., Donnan G. A., Howells D. W. (2018). NXY-059, a failed stroke neuroprotectant, offers no protection to stem cell-derived human neurons. J. Stroke Cerebrovasc. Dis. 27, 2158–2165. 10.1016/j.jstrokecerebrovasdis.2018.03.015 PubMed DOI
Aquilina G., Bampidis V., Bastos M. L., Bories G., Chesson A., Cocconcelli P. S., et al. (2015). Safety and efficacy of ethoxyquin (6-ethoxy-1,2- dihydro-2,2,4-trimethylquinoline) for all animal species. EFSA J. 13, 4272. 10.2903/J.EFSA.2015.4272 PubMed DOI PMC
Arteaga S., Andrade-Cetto A., Cárdenas R. (2005). Larrea tridentata (creosote bush), an abundant plant of Mexican and US-American deserts and its metabolite nordihydroguaiaretic acid. J. Ethnopharmacol. 98, 231–239. 10.1016/j.jep.2005.02.002 PubMed DOI
Arts J., Bade S., Badrinas M., Ball N., Hindle S. (2018). Should DTPA, an aminocarboxylic acid (ethylenediamine-based) chelating agent, be considered a developmental toxicant? Regul. Toxicol. Pharmacol. 97, 197–208. 10.1016/j.yrtph.2018.06.019 PubMed DOI
Astete C. E., Dolliver D., Whaley M., Khachatryan L., Sabliov C. M. (2011). Antioxidant poly(lactic-co-glycolic) acid nanoparticles made with α-tocopherol-ascorbic acid surfactant. ACS Nano 5, 9313–9325. 10.1021/nn102845t PubMed DOI
Atamna H., Paler-Martínez A., Ames B. N. (2000). N-t-butyl hydroxylamine, a hydrolysis product of α-phenyl-N-t-butyl nitrone, is more potent in delaying senescence in human lung fibroblasts. J. Biol. Chem. 275, 6741–6748. 10.1074/jbc.275.10.6741 PubMed DOI
Augustin A. J., Boker T., Blumenroder S. H.-H., Lutz J., Spitznas M. (1994). Free radical scavenging and antioxidant activity of allopurinol and oxypurinol in experimental lens-induced uveitis. Invest. Ophthalmol. Vis. Sci. 35, 3897–3904. PubMed
Azzi A., Zingg J.-M. (2005). Vitamin E: textbooks require updating. Biochem. Mol. Biol. Educ. 33, 184–187. 10.1002/bmb.2005.494033032451 PubMed DOI
Bales B. C., Kodama T., Weledji Y. N., Pitié M., Meunier B., Greenberg M. M. (2005). Mechanistic studies on DNA damage by minor groove binding copper-phenanthroline conjugates. Nucleic Acids Res. 33, 5371–5379. 10.1093/nar/gki856 PubMed DOI PMC
Bartekova M., Barancik M., Ferenczyova K., Dhalla N. S. (2018). Beneficial effects of n-acetylcysteine and n-mercaptopropionylglycine on ischemia reperfusion injury in the heart. Curr. Med. Chem. 25, 355–366. 10.2174/0929867324666170608111917 PubMed DOI
Barzegar A., Pedersen J. Z., Incerpi S., Moosavi-Movahedi A. A., Saso L. (2011). The mechanism of antioxidant activity of IRFI005 as a synthetic hydrophilic analogue of vitamin e. Biochimie 93, 1880–1888. 10.1016/j.biochi.2011.07.019 PubMed DOI
Bayır H., Anthonymuthu T. S., Tyurina Y. Y., Patel S. J., Amoscato A. A., Lamade A. M., et al. (2020). Achieving life through death: redox biology of lipid peroxidation in ferroptosis. Cell Chem. Biol. 27, 387–408. 10.1016/j.chembiol.2020.03.014 PubMed DOI PMC
Beckman J. A., Goldfine A. B., Leopold J. A., Creager M. A. (2016). Ebselen does not improve oxidative stress and vascular function in patients with diabetes: a randomized, crossover trial. Am. J. Physiol. - Heart Circ. Physiol. 311, H1431–H1436. 10.1152/ajpheart.00504.2016 PubMed DOI PMC
Belayev L., Becker D. A., Alonso O. F., Liu Y., Busto R., Ley J. J., et al. (2002). Stilbazulenyl nitrone, a novel azulenyl nitrone antioxidant: improved neurological deficit and reduced contusion size after traumatic brain injury in rats. J. Neurosurg. 96, 1077–1083. 10.3171/jns.2002.96.6.1077 PubMed DOI
Bencini A., Lippolis V. (2010). 1,10-Phenanthroline: a versatile building block for the construction of ligands for various purposes. Coord. Chem. Rev. 254, 2096–2180. 10.1016/j.ccr.2010.04.008 DOI
Bergeron R. J., Wiegand J., Brittenham G. M. (1999). HBED: the continuing development of a potential alternative to deferoxamine for iron-chelating therapy. Blood 93, 370–375. 10.1182/blood.v93.1.370.401k29_370_375 PubMed DOI
Binbuga N., Chambers K., Henry W. P., Schultz T. P. (2005). Metal chelation studies relevant to wood preservation. 1. Complexation of propyl gallate with Fe2+. Holzforschung 59, 205–209. 10.1515/HF.2005.032 DOI
Bindoli A., Rigobello M., Favel A., Galzigna L. (1988). Antioxidant action and photosensitizing effects of three different chlorpromazines. J. Neurochem. 50, 138–141. 10.1111/j.1471-4159.1988.tb13240.x PubMed DOI
Bitto A., Minutoli L., Squadrito F., Polito F., Altavilla D. (2007). Raxofelast, (+/-)5-(acetyloxy)-2,3-dihydro-4,6,7-trimethyl-2-benzofuranacetic acid: a new antioxidant to modulate the inflammatory response during ischemia-reperfusion injury and impaired wound healing. Mini-Rev. Med. Chem. 7, 339–343. 10.2174/138955707780059835 PubMed DOI
Bjørklund G., Mutter J., Aaseth J. (2017). Metal chelators and neurotoxicity: lead, Mercury, and arsenic. Arch. Toxicol. 91, 3787–3797. 10.1007/s00204-017-2100-0 PubMed DOI
Błaszczyk A., Augustyniak A., Skolimowski J. (2013). Ethoxyquin: an antioxidant used in animal feed. Int. J. Food Sci. 2013, 585931. 10.1155/2013/585931 PubMed DOI PMC
Bloot A. P. M., Kalschne D. L., Amaral J. A. S., Baraldi I. J., Canan C. (2023). A review of phytic acid sources, obtention, and applications. Food Rev. Int. 39, 73–92. 10.1080/87559129.2021.1906697 DOI
Boshtam M., Kouhpayeh S., Amini F., Azizi Y., Najaflu M., Shariati L., et al. (2021). Anti-inflammatory effects of apocynin: a narrative review of the evidence. Life 14, 997–1010. 10.1080/26895293.2021.1990136 DOI
Bozic M., Goss C. H., Tirouvanziam R. M., Baines A., Kloster M., Antoine L., et al. (2020). Oral glutathione and growth in cystic fibrosis: a multicenter, randomized, placebo-controlled, double-blind trial. J. Pediatr. Gastroenterol. Nutr. 71, 771–777. 10.1097/MPG.0000000000002948 PubMed DOI PMC
Bracken M. B., Shepard M. J., Holford T. R., Leo-Summers L., Aldrich E. F., Fazl M., et al. (1998). Methylprednisolone or tirilazad mesylate administration after acute spinal cord injury: 1-Year follow up: results of the third national acute spinal cord injury randomized controlled trial. J. Neurosurg. 89, 699–706. 10.3171/jns.1998.89.5.0699 PubMed DOI
Braz M. G., Braz L. G., Freire C. M. M., Lucio L. M. C., Braz J. R. C., Tang G., et al. (2015). Isoflurane and propofol contribute to increasing the antioxidant status of patients during minor elective surgery a randomized clinical study. Med. U. S. 94, e1266. 10.1097/MD.0000000000001266 PubMed DOI PMC
Brittenham G. M., Nathan D. G., Olivieri N. F., Porter J. B., Pippard M., Vichinsky E. P., et al. (2003). Deferiprone and hepatic fibrosis. Blood 101, 5089–5091. 10.1182/blood-2002-10-3173 PubMed DOI
Brock N., Hilgard P., Pohl J., Ormstad K., Orrenius S. (1984). Pharmacokinetics and mechanism of action of detoxifying low-molecular-weight thiols. J. Cancer Res. Clin. Oncol. 108, 87–97. 10.1007/BF00390979 PubMed DOI PMC
Butterfield D. A., Halliwell B. (2019). Oxidative stress, dysfunctional glucose metabolism and alzheimer disease. Nat. Rev. Neurosci. 20, 148–160. 10.1038/s41583-019-0132-6 PubMed DOI PMC
Cacciatore I., Cornacchia C., Pinnen F., Mollica A., Di Stefano A. (2010). Prodrug approach for increasing cellular glutathione levels. Molecules 15, 1242–1264. 10.3390/molecules15031242 PubMed DOI PMC
Cahill L., Hall E. D. (2017). Is it time to resurrect “lazaroids”. J. Neurosci. Res. 95, 17–20. 10.1002/jnr.23842 PubMed DOI
Canistro D., Affatato A. A., Soleti A., Mollace V., Muscoli C., Sculco F., et al. (2010). The novel radical scavenger IAC is effective in preventing and protecting against post-ischemic brain damage in Mongolian gerbils. J. Neurol. Sci. 290, 90–95. 10.1016/j.jns.2009.10.023 PubMed DOI
Cappellini M. D. (2005). Iron-chelating therapy with the new oral agent ICL670 (exjade®). Best. Pract. Res. Clin. Haematol. 18, 289–298. 10.1016/j.beha.2004.09.002 PubMed DOI
Casas A. I., Nogales C., Mucke H. A. M., Petraina A., Cuadrado A., Rojo A. I., et al. (2020). On the clinical pharmacology of reactive oxygen species. Pharmacol. Rev. 72, 801–828. 10.1124/pr.120.019422 PubMed DOI
Cayatte A. J., Rupin A., Oliver-Krasinski J., Maitland K., Sansilvestri-Morel P., Boussard M.-F., et al. (2001). S17834, a new inhibitor of cell adhesion and atherosclerosis that targets NADPH oxidase. Arterioscler. Thromb. Vasc. Biol. 21, 1577–1584. 10.1161/hq1001.096723 PubMed DOI
Celebioglu A., Uyar T. (2017). Antioxidant vitamin E/cyclodextrin inclusion complex electrospun nanofibers: enhanced water solubility, prolonged shelf life, and photostability of vitamin E. J. Agric. Food Chem. 65, 5404–5412. 10.1021/acs.jafc.7b01562 PubMed DOI
Chabrier P. E., Auguet M. (2007). Pharmacological properties of BN82451: a novel multitargeting neuroprotective agent. CNS Drug Rev. 13, 317–332. 10.1111/j.1527-3458.2007.00018.x PubMed DOI PMC
Chan K. Y., Dusterhoft D. A., Chen T.-M. (1994). Determination of MDL 74,405, a synthetic analogue of α-tocopherol, in dog plasma and heart tissue by high-performance liquid chromatography with electrochemical detection. J. Chromatogr. B. Biomed. Sci. App. 656, 359–365. 10.1016/0378-4347(94)00120-0 PubMed DOI
Cheah I. K., Halliwell B. (2021). Ergothioneine, recent developments. Redox Biol. 42, 101868. 10.1016/j.redox.2021.101868 PubMed DOI PMC
Chen S.-H., Liang D.-C., Lin H.-C., Cheng S.-Y., Chen L.-J., Liu H.-C. (2005). Auditory and visual toxicity during deferoxamine therapy in transfusion-dependent patients. J. Pediatr. Hematol. Oncol. 27, 651–653. 10.1097/01.mph.0000194019.95096.b6 PubMed DOI
Chen Y., Shi J., Xia T. C., Xu R., He X., Xia Y. (2019). Preservation solutions for kidney transplantation: history, advances and mechanisms. Cell Transpl. 28, 1472–1489. 10.1177/0963689719872699 PubMed DOI PMC
Cho S., Kong B., Jung Y., Shin J., Park M., Chung W.-J., et al. (2023). Synthesis and physicochemical characterization of acyl myricetins as potential anti-neuroexocytotic agents. Sci. Rep. 13, 5136. 10.1038/s41598-023-32361-6 PubMed DOI PMC
Choi E.-Y., Kim E.-C., Oh H.-M., Kim S., Lee H.-J., Cho E.-Y., et al. (2004). Iron chelator triggers inflammatory signals in human intestinal epithelial cells: involvement of p38 and extracellular signal-regulated kinase signaling pathways. J. Immunol. 172, 7069–7077. 10.4049/jimmunol.172.11.7069 PubMed DOI
Chrószcz-Porębska M., Gadomska-Gajadhur A. (2024). Cysteine conjugation: an approach to obtain polymers with enhanced muco- and tissue adhesion. Int. J. Mol. Sci. 25, 12177. 10.3390/ijms252212177 PubMed DOI PMC
Cimmino G., De Nisco M., Alonso C., Gravina C., Piscopo V., Lemos R., et al. (2024). Novel synthesized seleno-glycoconjugates as cosmeceutical ingredients: antioxidant activity and DOI
Cipriano A., Viviano M., Feoli A., Milite C., Sarno G., Castellano S., et al. (2023). NADPH oxidases: from molecular mechanisms to current inhibitors. J. Med. Chem. 66, 11632–11655. 10.1021/acs.jmedchem.3c00770 PubMed DOI PMC
Ciriminna R., Pagliaro M. (2010). Industrial oxidations with organocatalyst TEMPO and its derivatives. Org. Process Res. Dev. 14, 245–251. 10.1021/op900059x DOI
Cohen G., Sinet P. M. (1982). The fenton reaction between ferrous-diethylenetriaminepentaacetic acid and hydrogen peroxide. FEBS Lett. 138, 258–260. 10.1016/0014-5793(82)80455-9 DOI
Collis C., Davies M., Rice-Evans C. (1993). The effects of n-methyl butyrohydroxamic acid and other monohydroxamates on reperfusion-induced damage to contractile function in the isolated rat-heart. Free Radic. Res. Commun. 18, 269–277. 10.3109/10715769309147494 PubMed DOI
Creighton-Gutteridge M., Tyrrell R. M. (2002). A novel iron chelator that does not induce HIF-1 activity. Free Radic. Biol. Med. 33, 356–363. 10.1016/S0891-5849(02)00884-5 PubMed DOI
Cui Z., Lockman P. R., Atwood C. S., Hsu C.-H., Gupte A., Allen D. D., et al. (2005). Novel D-penicillamine carrying nanoparticles for metal chelation therapy in Alzheimer’s and other CNS diseases. Eur. J. Pharm. Biopharm. 59, 263–272. 10.1016/j.ejpb.2004.07.009 PubMed DOI
Cuzzocrea S., Zingarelli B., Hake P., Salzman A. L., Szabo C. (1998). Antiinflammatory effects of mercaptoethylguanidine, a combined inhibitor of nitric oxide synthase and peroxynitrite scavenger, in carrageenan-induced models of inflammation. Free Radic. Biol. Med. 24, 450–459. 10.1016/S0891-5849(97)00280-3 PubMed DOI
Dandona P., Ghanim H., Brooks D. (2007). Antioxidant activity of carvedilol in cardiovascular disease. J. Hypertens. 25, 731–741. 10.1097/HJH.0b013e3280127948 PubMed DOI
Day B. J. (2004). Catalytic antioxidants: a radical approach to new therapeutics. Drug Discov. Today 9, 557–566. 10.1016/S1359-6446(04)03139-3 PubMed DOI
Day B. J. (2008). Antioxidants as potential therapeutics for lung fibrosis. Antioxid. Redox Signal. 10, 355–370. 10.1089/ars.2007.1916 PubMed DOI PMC
Deblonde G. J.-P., Kelley M. P., Su J., Batista E. R., Yang P., Booth C. H., et al. (2018). Spectroscopic and computational characterization of diethylenetriaminepentaacetic acid/transplutonium chelates: evidencing heterogeneity in the heavy Actinide(III) series. Angew. Chem. - Int. Ed. 57, 4521–4526. 10.1002/anie.201709183 PubMed DOI
De Bruin J. (1967). Ca-DTPA in the treatment of chronic lead poisoning. Ned. Tijdschr. Geneeskd. 111, 824–826. PubMed
Delanghe T., Huyghe J., Lee S., Priem D., Van Coillie S., Gilbert B., et al. (2021). Antioxidant and food additive BHA prevents TNF cytotoxicity by acting as a direct RIPK1 inhibitor. Cell Death Dis. 12, 699. 10.1038/s41419-021-03994-0 PubMed DOI PMC
de Matos D. G., Furnus C. C., Moses D. F., Baldassarre H. (1995). Effect of cysteamine on glutathione level and developmental capacity of bovine oocyte matured PubMed DOI
Demedts M., Behr J., Buhl R., Costabel U., Dekhuijzen R., Jansen H. M., et al. (2005). High-dose acetylcysteine in idiopathic pulmonary fibrosis. N. Engl. J. Med. 353, 2229–2242. 10.1056/NEJMoa042976 PubMed DOI
Deng S., Yan T., Nikolova T., Fuhrmann D., Nemecek A., Gödtel-Armbrust U., et al. (2015). The catalytic topoisomerase II inhibitor dexrazoxane induces DNA breaks, ATF3 and the DNA damage response in cancer cells. Br. J. Pharmacol. 172, 2246–2257. 10.1111/bph.13046 PubMed DOI PMC
Dewick P. M. (2009). Medicinal natural products: a biosynthetic approach. 3rd Edn. Chichester, UK: Wiley and Sons.
Dickey A. S., La Spada A. R. (2018). Therapy development in huntington disease: from current strategies to emerging opportunities. Am. J. Med. Genet. 176, 842–861. 10.1002/ajmg.a.38494 PubMed DOI PMC
Di Costanzo A., Angelico R. (2019). Formulation strategies for enhancing the bioavailability of silymarin: the state of the art. Molecules 24, 2155. 10.3390/molecules24112155 PubMed DOI PMC
Diebold B. A., Smith S. M. E., Li Y., Lambeth J. D. (2015). NOX2 as a target for drug development: indications, possible complications, and progress. Antioxid. Redox Signal. 23, 375–405. 10.1089/ars.2014.5862 PubMed DOI PMC
Di Paola A., Tortora C., Argenziano M., Marrapodi M. M., Rossi F. (2022). Emerging roles of the iron chelators in inflammation. Int. J. Mol. Sci. 23, 7977. 10.3390/ijms23147977 PubMed DOI PMC
Doctrow S. R., Huffman K., Marcus C. B., Tocco G., Malfroy E., Adinolfi C. A., et al. (2002). Salen-manganese complexes as catalytic scavengers of hydrogen peroxide and cytoprotective agents: structure-Activity relationship studies. J. Med. Chem. 45, 4549–4558. 10.1021/jm020207y PubMed DOI
Doggrell S. A. (2002). Therapeutic potential of selective superoxide dismutase mimetics. Drugs Future 27, 385–390. 10.1358/dof.2002.027.04.664560 DOI
Duffy S. J., Biegelsen E. S., Holbrook M., Russell J. D., Gokce N., Keaney Jr., J. F., et al. (2001). Iron chelation improves endothelial function in patients with coronary artery disease. Circulation 103, 2799–2804. 10.1161/01.CIR.103.23.2799 PubMed DOI
Dupré-Crochet S., Erard M., Nüße O. (2013). ROS production in phagocytes: why, when, and where? J. Leukoc. Biol. 94, 657–670. 10.1189/jlb.1012544 PubMed DOI
Efrati S., Dishy V., Averbukh M., Blatt A., Krakover R., Weisgarten J., et al. (2003). The effect of N-acetylcysteine on renal function, nitric oxide, and oxidative stress after angiography. Kidney Int. 64, 2182–2187. 10.1046/j.1523-1755.2003.00322.x PubMed DOI
Egan T. J., Barthakur S. R., Aisen P. (1992). Catalysis of the haber-weiss reaction by iron-diethylenetriaminepentaacetate. J. Inorg. Biochem. 48, 241–249. 10.1016/0162-0134(92)84051-N PubMed DOI
El-Demerdash E., Ali A. A., Sayed-Ahmed M. M., Osman A.-M. M. (2003). New aspects in probucol cardioprotection against doxorubicin-induced cardiotoxicity. Cancer Chemother. Pharmacol. 52, 411–416. 10.1007/s00280-003-0676-y PubMed DOI
El -Far M., Essam A., El-Senduny F., El-Azim A., Yahia S., El-Sherbiny I. (2022). Potential use of nanoformulated ascorbyl palmitate as a promising anticancer agent: first comparative assessment between nano and free forms. J. Drug Deliv. Sci. Technol. 78, 103920. 10.1016/j.jddst.2022.103920 DOI
Eluashvili I. A., Prilipko L. L., Kagan V. E. (1978). Enzymic peroxidation of lipids and oxidative metabolism of chlorpromazine in brain microsomal fraction. Bull. Exp. Biol. Med. 86, 1321–1324. 10.1007/BF00800447 PubMed DOI
Eriksson O., Pollesello P., Saris N. E. L. (1992). Inhibition of lipid peroxidation in isolated rat liver mitochondria by the general anaesthetic propofol. Biochem. Pharmacol. 44, 391–393. 10.1016/0006-2952(92)90026-F PubMed DOI
Esazadeh K., Ezzati Nazhad Dolatabadi J., Andishmand H., Mohammadzadeh-Aghdash H., Mahmoudpour M., Naemi Kermanshahi M., et al. (2024). Cytotoxic and genotoxic effects of tert-butylhydroquinone, butylated hydroxyanisole and propyl gallate as synthetic food antioxidants. Food Sci. Nutr. 12, 7004–7016. 10.1002/fsn3.4373 PubMed DOI PMC
Fahlbusch S. A., Tsikas D., Mehls C., Gutzki F.-M., Böger R. H., Frölich J. C., et al. (2004). Effects of carvedilol on oxidative stress in human endothelial cells and healthy volunteers. Eur. J. Clin. Pharmacol. 60, 83–88. 10.1007/s00228-004-0729-0 PubMed DOI
Ferenci P. (2016). Silymarin in the treatment of liver diseases: what is the clinical evidence? Clin. Liver Dis. 7, 8–10. 10.1002/cld.522 PubMed DOI PMC
Firuzi O., Miri R., Tavakkoli M., Saso L. (2011). Antioxidant therapy: current status and future prospects. Curr. Med. Chem. 18, 3871–3888. 10.2174/092986711803414368 PubMed DOI
Flohé L., Toppo S., Orian L. (2022). The glutathione peroxidase family: discoveries and mechanism. Free Radic. Biol. Med. 187, 113–122. 10.1016/j.freeradbiomed.2022.05.003 PubMed DOI
Flora S. J. S., Pachauri V. (2010). Chelation in metal intoxication. Int. J. Environ. Res. Public. Health 7, 2745–2788. 10.3390/ijerph7072745 PubMed DOI PMC
Floyd R. A., Hensley K., Forster M. J., Kelleher-Andersson J. A., Wood P. L. (2002). Nitrones, their value as therapeutics and probes to understand aging. Mech. Ageing Dev. 123, 1021–1031. 10.1016/S0047-6374(01)00385-2 PubMed DOI
Forman H. J., Zhang H. (2021). Targeting oxidative stress in disease: promise and limitations of antioxidant therapy. Nat. Rev. Drug Discov. 20, 689–709. 10.1038/s41573-021-00233-1 PubMed DOI PMC
Frank M. (2022). Bucillamine in the treatment of patients with mild to moderate COVID-19: an interview with michael frank. Future Microbiol. 17, 157–159. 10.2217/fmb-2021-0277 PubMed DOI PMC
Fridavich I. (1995). Superoxide radical and superoxide dismutases. Annu. Rev. Biochem. 64, 97–112. 10.1146/annurev.bi.64.070195.000525 PubMed DOI
Fukuta T., Ikeda-Imafuku M., Iwao Y. (2023). Development of edaravone ionic liquids and their application for the treatment of cerebral ischemia/reperfusion injury. Mol. Pharm. 20, 3115–3126. 10.1021/acs.molpharmaceut.3c00103 PubMed DOI PMC
Gal S., Zheng H., Fridkin M., Youdim M. B. H. (2005). Novel multifunctional neuroprotective iron chelator-monoamine oxidase inhibitor drugs for neurodegenerative diseases. PubMed DOI
Galey J.-B., Destrée O., Dumats J., Génard S., Tachon P. (2000). Protection against oxidative damage by iron chelators: effect of lipophilic analogues and prodrugs of N,N’-bis(3,4,5- trimethoxybenzyl)ethylenediamine-N,N’-diacetic acid (OR10141). J. Med. Chem. 43, 1418–1421. 10.1021/jm9911635 PubMed DOI
Gambaro R. C., Berti I. R., Cacicedo M. L., Gehring S., Alvarez V. A., Castro G. R., et al. (2022). Colloidal delivery of vitamin E into solid lipid nanoparticles as a potential complement for the adverse effects of anemia treatment. Chem. Phys. Lipids 249, 105252. 10.1016/j.chemphyslip.2022.105252 PubMed DOI
Georgieva R., Momchilova A., Petkova D., Koumanov K., Staneva G. (2013). Effect of n-propyl gallate on lipid peroxidation in heterogenous model membranes. Biotechnol. Biotechnol. Equip. 27, 4145–4149. 10.5504/BBEQ.2013.0061 DOI
Gilbert N. C., Gerstmeier J., Schexnaydre E. E., Börner F., Garscha U., Neau D. B., et al. (2020). Structural and mechanistic insights into 5-lipoxygenase inhibition by natural products. Nat. Chem. Biol. 16, 783–790. 10.1038/s41589-020-0544-7 PubMed DOI PMC
Guillonneau C., Charton Y., Ginot Y.-M., Fouquier-D’Hérouël M.-V., Bertrand M., Lockhart B., et al. (2003). Synthesis and pharmacological evaluation of new 1,2-dithiolane based antioxidants. Eur. J. Med. Chem. 38, 1–11. 10.1016/s0223-5234(02)01424-1 PubMed DOI
Guo Y., Wang C., Cao J., Gao J., Zou X., Ren Y., et al. (2015). Antioxidant and lipid-regulating effects of probucol combined with atorvastatin in patients with acute coronary syndrome. J. Thorac. Dis. 7, 368–375. 10.3978/j.issn.2072-1439.2014.12.29 PubMed DOI PMC
Gutzmann H., Kühl K.-P., Hadler D., Rapp M. A. (2002). Safety and efficacy of idebenone PubMed DOI
Hadi H. A. R., Al Suwaidi J. A. (2007). Endothelial dysfunction in diabetes mellitus. Vasc. Health Risk Manag. 3, 853–876. PubMed PMC
Halliwell B. (1978). Superoxide-dependent formation of hydroxyl radicals in the presence of iron chelates. Is it a mechanism for hydroxyl radical production in biochemical systems? FEBS Lett. 92, 321–326. 10.1016/0014-5793(78)80779-0 PubMed DOI
Halliwell B. (2006). Oxidative stress and neurodegeneration: where are we now? J. Neurochem. 97, 1634–1658. 10.1111/j.1471-4159.2006.03907.x PubMed DOI
Halliwell B. (2009). The wanderings of a free radical. Free Radic. Biol. Med. 46, 531–542. 10.1016/j.freeradbiomed.2008.11.008 PubMed DOI
Halliwell B. (2012). Free radicals and antioxidants: updating a personal view. Nutr. Rev. 70, 257–265. 10.1111/j.1753-4887.2012.00476.x PubMed DOI
Halliwell B. (2022). Reactive oxygen species (ROS), oxygen radicals and antioxidants: where are we now, where is the field going and where should we go? Biochem. Biophys. Res. Commun. 633, 17–19. 10.1016/j.bbrc.2022.08.098 PubMed DOI
Halliwell B. (2024). Understanding mechanisms of antioxidant action in health and disease. Nat. Rev. Mol. Cell Biol. 25, 13–33. 10.1038/s41580-023-00645-4 PubMed DOI
Halliwell B., Cheah I. K., Tang R. M. Y. (2018). Ergothioneine – a diet-derived antioxidant with therapeutic potential. FEBS Lett. 592, 3357–3366. 10.1002/1873-3468.13123 PubMed DOI
Halliwell B., Gutteridge J. M. C. (2015). Free radicals in biology and medicine. 5th Edn. New York, USA: Oxford University Press.
Ham J., Lim W., Park S., Bae H., You S., Song G. (2019). Synthetic phenolic antioxidant propyl gallate induces Male infertility through disruption of calcium homeostasis and mitochondrial function. Environ. Pollut. 248, 845–856. 10.1016/j.envpol.2019.02.087 PubMed DOI
Han J., Tao W., Cui W., Chen J. (2022). Propofol PubMed DOI PMC
Han Y., Liu Y., Rockenbauer A., Zweier J. L., Durand G., Villamena F. A. (2009). Lipophilic β-cyclodextrin cyclic-nitrone conjugate: synthesis and spin trapping studies. J. Org. Chem. 74, 5369–5380. 10.1021/jo900856x PubMed DOI PMC
Hanachi N., Charef N., Baghiani A., Khennouf S., Derradji Y., Boumerfeg S., et al. (2009). Comparison of xanthine oxidase levels in synovial fluid from patients with rheumatoid arthritis and other joint inflammations. Saudi Med. J. 30, 1422–1425. PubMed
Hansberg W. (2022). Monofunctional heme-catalases. Antioxidants 11, 2173. 10.3390/antiox11112173 PubMed DOI PMC
Harcárová P., Lomozová Z., Kallivretaki M., Karlícková J., Kucera R., Mladenka P. (2025). Different behavior of food-related benzoic acids toward iron and copper. Food Chem. 462, 141014. 10.1016/j.foodchem.2024.141014 PubMed DOI
Hasaniya N. W., Premaratne S., Zhang W. W., Razzuk A., Abdul-Ghani A. A., Dashwood R. H., et al. (2011). Amelioration of ischemia-reperfusion injury in an isolated rabbit lung model using OXANOH. Vasc. Endovasc. Surg. 45, 581–591. 10.1177/1538574410390715 PubMed DOI
Hatcher H. C., Singh R. N., Torti F. M., Torti S. V. (2009). Synthetic and natural iron chelators: therapeutic potential and clinical use. Future Med. Chem. 1, 1643–1670. 10.4155/fmc.09.121 PubMed DOI PMC
Horwitz L. D. (2003). Bucillamine: a potent thiol donor with multiple clinical applications. Cardiovasc. Drug Rev. 21, 77–90. 10.1111/j.1527-3466.2003.tb00107.x PubMed DOI
Hsu C., Mahdi H., Pourahmadi M., Ahmadi S. (2013). Cysteamine cream as a new skin depigmenting product. J. Am. Acad. Dermatol. 68, AB189. 10.1016/j.jaad.2012.12.781 DOI
Inoue Y., Isobe M., Hayashi H. (2003). The combined effect of topical CX-659S, a novel diaminouracil derivative, with topical corticosteroid on the three types of allergic responses in mice or Guinea pigs. J. Pharmacol. Sci. 91, 71–78. 10.1254/jphs.91.71 PubMed DOI
Islam M. R., Jony M. H., Thufa G. K., Akash S., Dhar P. S., Rahman M. M., et al. (2024). A clinical study and future prospects for bioactive compounds and semi-synthetic molecules in the therapies for Huntington’s disease. Mol. Neurobiol. 61, 1237–1270. 10.1007/s12035-023-03604-4 PubMed DOI
Ismail H., Shakkour Z., Tabet M., Abdelhady S., Kobaisi A., Abedi R., et al. (2020). Traumatic brain injury: oxidative stress and novel anti-oxidants such as mitoquinone and edaravone. Antioxidants 9, 943–18. 10.3390/antiox9100943 PubMed DOI PMC
Jacobs H., Peters R., Den Hartog G. J. M., Van Der Vijgh W. J. F., Bast A., Haenen G. R. M. M. (2011). Identification of the metabolites of the antioxidant flavonoid 7-mono-O-(β-hydroxyethyl)-rutoside in mice. Drug Metab. Dispos. 39, 750–756. 10.1124/dmd.110.036525 PubMed DOI
Javed S., Petropoulos I. N., Alam U., Malik R. A. (2015). Treatment of painful diabetic neuropathy. Ther. Adv. Chronic Dis. 6, 15–28. 10.1177/2040622314552071 PubMed DOI PMC
Jung J.-I., Park K.-Y., Lee Y., Park M., Kim J. (2018). Vitamin C-linker–conjugated tripeptide AHK stimulates BMP-2-induced osteogenic differentiation of mouse myoblast C2C12 cells. Differentiation 101, 1–7. 10.1016/j.diff.2018.03.001 PubMed DOI
Kalamkar S., Acharya J., Madathil A., Gajjar V., Divate U., Karandikar-Iyer S., et al. (2022). Randomized clinical trial of how long-term glutathione supplementation offers protection from oxidative damage and improves hba1c in elderly type 2 diabetic patients. Antioxidants 11, 1026. 10.3390/antiox11051026 PubMed DOI PMC
Kalinowski D. S., Richardson D. R. (2005). The evolution of iron chelators for the treatment of iron overload disease and cancer. Pharmacol. Rev. 57, 547–583. 10.1124/pr.57.4.2 PubMed DOI
Kassahun K., Pearson P. G., Tang W., McIntosh I., Leung K., Elmore C., et al. (2001). Studies on the metabolism of troglitazone to reactive intermediates PubMed DOI
Kato N., Yanaka K., Nagase S., Hirayama A., Nose T., Mathiesen T. (2003). The antioxidant EPC-K1 ameliorates brain injury by inhibiting lipid peroxidation in a rat model of transient focal cerebral ischaemia. Acta Neurochir. (Wien) 145, 489–493. 10.1007/s00701-003-0036-z PubMed DOI
Kavanagh R. J., Kam P. C. A. (2001). Lazaroids: efficacy and mechanism of action of the 21-aminosteroids in neuroprotection. Br. J. Anaesth. 86, 110–119. 10.1093/bja/86.1.110 PubMed DOI
Kezic A., Spasojevic I., Lezaic V., Bajcetic M. (2016). Mitochondria-targeted antioxidants: future perspectives in kidney ischemia reperfusion injury. Oxid. Med. Cell. Longev. 2016, 2950503. 10.1155/2016/2950503 PubMed DOI PMC
Khan N., Singh A. K., Saneja A. (2023). Preparation, characterization, and antioxidant activity of L-Ascorbic Acid/HP-β-Cyclodextrin inclusion complex-incorporated electrospun nanofibers. Foods 12, 1363. 10.3390/foods12071363 PubMed DOI PMC
Khatib S. Y., Farah H., El-Migdadi F. (2001). Allopurinol enhances adenine nucleotide levels and improves myocardial function in isolated hypoxic rat heart. Biochem. Mosc. 66, 328–333. 10.1023/A:1010264216357 PubMed DOI
Khayata N., Abdelwahed W., Chehna M. F., Charcosset C., Fessi H. (2012). Preparation of vitamin e loaded nanocapsules by the nanoprecipitation method: from laboratory scale to large scale using a membrane contactor. Int. J. Pharm. 423, 419–427. 10.1016/j.ijpharm.2011.12.016 PubMed DOI
Kitazawa M., Ishitsuka Y., Kobayashi M., Nakano T., Iwasaki K., Sakamoto K., et al. (2005). Protective effects of an antioxidant derived from serine and vitamin B 6 on skin photoaging in hairless mice. Photochem. Photobiol. 81, 970–974. 10.1562/2004-09-18-RA-320 PubMed DOI
Klein E. A., Thompson Jr., I. M., Tangen C. M., Crowley J. J., Lucia S., Goodman P. J., et al. (2011). Vitamin E and the risk of prostate cancer: the selenium and vitamin E cancer prevention trial (SELECT). JAMA - J. Am. Med. Assoc. 306, 1549–1556. 10.1001/jama.2011.1437 PubMed DOI PMC
Kleschyov A. L., Sen’ V., Golubev V., Münnemann K., Hinderberger D., Lackner K. J., et al. (2012). Heparin-polynitroxides: synthesis and preliminary evaluation as cardiovascular EPR/MR imaging probes and extracellular space-targeted antioxidants. Eur. J. Med. Chem. 58, 265–271. 10.1016/j.ejmech.2012.09.028 PubMed DOI
Klivenyi P., Ferrante R. J., Gardian G., Browne S., Chabrier P.-E., Beal M. F. (2003). Increased survival and neuroprotective effects of BN82451 in a transgenic mouse model of Huntington’s disease. J. Neurochem. 86, 267–272. 10.1046/j.1471-4159.2003.t01-1-01868.x PubMed DOI
Kobayashi S., Fukuma S., Ikenoue T., Fukuhara S., Kobayashi S. (2019). Effect of edaravone on neurological symptoms in real-world patients with acute ischemic stroke: japan stroke data bank. Stroke 50, 1805–1811. 10.1161/STROKEAHA.118.024351 PubMed DOI
Konorev E. A., Kotamraju S., Zhao H., Kalivendi S., Joseph J., Kalyanaraman B. (2002). Paradoxical effects of metalloporphyrins on doxorubicin-induced apoptosis: scavenging of reactive oxygen species PubMed DOI
Kontoghiorghes G. J. (2013). Turning a blind eye to deferasirox’s toxicity? Lancet 381, 1183–1184. 10.1016/S0140-6736(13)60799-0 PubMed DOI
Koufaki M., Kiziridi C., Nikoloudaki F., Alexis M. N. (2007). Design and synthesis of 1,2-dithiolane derivatives and evaluation of their neuroprotective activity. Bioorg. Med. Chem. Lett. 17, 4223–4227. 10.1016/j.bmcl.2007.05.036 PubMed DOI
Kumar S., Balkrishna S. J., Kumar S., Azad G. K., Bhakuni B. S., Panini P., et al. (2014). An ebselen like catalyst with enhanced GPx activity PubMed DOI
Kuo B.-S., Mathews B. S., Stuhler J. D., Carrel B. K., Ho J., Rose J. Q. (1995). Disposition and cardioselectivity of MDL 74,405, a vitamin E-like free radical scavenger, in rats and dogs after intravenous infusion. Drug Metab. Dispos. 23, 757–764. 10.1016/s0090-9556(25)06654-1 PubMed DOI
Kurajoh M., Fukumoto S., Yoshida S., Akari S., Murase T., Nakamura T., et al. (2021). Uric acid shown to contribute to increased oxidative stress level independent of xanthine oxidoreductase activity in MedCity21 health examination registry. Sci. Rep. 11, 7378. 10.1038/s41598-021-86962-0 PubMed DOI PMC
Kwon T.-H., Chao D. L., Malloy K., Sun D., Alessandri B., Bullock M. R. (2003). Tempol, a novel stable nitroxide, reduces brain damage and free radical production, after acute subdural hematoma in the rat. J. Neurotrauma 20, 337–345. 10.1089/089771503765172291 PubMed DOI
Lamas G. A., Goertz C., Boineau R., Mark D. B., Rozema T., Nahin R. L., et al. (2013). Effect of disodium EDTA chelation regimen on cardiovascular events in patients with previous myocardial infarction: the TACT randomized trial. JAMA 309, 1241–1250. 10.1001/jama.2013.2107 PubMed DOI PMC
Lee J.-S., Song I.-H., Shinde P. B., Nimse S. B. (2020). Macrocycles and supramolecules as antioxidants: excellent scaffolds for development of potential therapeutic agents. Antioxidants 9, 859. 10.3390/antiox9090859 PubMed DOI PMC
Leggio L., Addolorato G., Abenavoli L., Gasbarrini G. (2005). Wilson’s disease: clinical, genetic and pharmacological findings. Int. J. Immunopathol. Pharmacol. 18, 7–14. 10.1177/039463200501800102 PubMed DOI
Levtchenko E. N., de Graaf-Hess A., Wilmer M., van den Heuvel L., Monnens L., Blom H. (2005). Altered status of glutathione and its metabolites in cystinotic cells. Nephrol. Dial. Transpl. 20, 1828–1832. 10.1093/ndt/gfh932 PubMed DOI
Ley J. J., Belayev L., Saul I., Becker D. A., Ginsberg M. D. (2007). Neuroprotective effect of STAZN, a novel azulenyl nitrone antioxidant, in focal cerebral ischemia in rats: dose-Response and therapeutic window. Brain Res. 1180, 101–110. 10.1016/j.brainres.2007.05.028 PubMed DOI PMC
Ley J. J., Prado R., Wei J. Q., Bishopric N. H., Becker D. A., Ginsberg M. D. (2008). Neuroprotective antioxidant STAZN protects against myocardial ischemia/reperfusion injury. Biochem. Pharmacol. 75, 448–456. 10.1016/j.bcp.2007.09.001 PubMed DOI PMC
Ley J. J., Vigdorchik A., Belayev L., Zhao W., Busto R., Khoutorova L., et al. (2005). Stilbazulenyl nitrone, a second-generation azulenyl nitrone antioxidant, confers enduring neuroprotection in experimental focal cerebral ischemia in the rat: Neurobehavior, histopathology, and pharmacokinetics. J. Pharmacol. Exp. Ther. 313, 1090–1100. 10.1124/jpet.105.083386 PubMed DOI
Li X., Hua S., Zhong D., Zhou M., Ding Z. (2025). Metal-organic framework-edaravone nanoparticles for radiotherapy-induced brain injury treatment. Biomaterials 314, 122868. 10.1016/j.biomaterials.2024.122868 PubMed DOI
Li Y., Davis J. M. (2003). Delivering antioxidants by zip code. Am. J. Physiol. - Lung Cell. Mol. Physiol. 285, L281–L282. 10.1152/ajplung.00092.2003 PubMed DOI
Liang L.-P., Fulton R., Bradshaw-Pierce E. L., Pearson-Smith J., Day B. J., Patel M. (2021). Optimization of lipophilic metalloporphyrins modifies disease outcomes in a rat model of parkinsonism. J. Pharmacol. Exp. Ther. 377, 1–10. 10.1124/jpet.120.000229 PubMed DOI
Liang L.-P., Huang J., Fulton R., Day B. J., Patel M. (2007). An orally active catalytic metalloporphyrin protects against 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine neurotoxicity PubMed DOI PMC
Link G., Ponka P., Konijn A., Breuer W., Cabantchik Z., Hershko C. (2003). Effects of combined chelation treatment with pyridoxal isonicotinoyl hydrazone analogs and deferoxamine in hypertransfused rats and in iron-loaded rat heart cells. Blood 101, 4172–4179. 10.1182/blood-2002-08-2382 PubMed DOI
Liu J., Wang L.-N. (2014). Mitochondrial enhancement for neurodegenerative movement disorders: a systematic review of trials involving creatine, coenzyme Q10, idebenone and mitoquinone. CNS Drugs 28, 63–68. 10.1007/s40263-013-0124-4 PubMed DOI
Liu X., Yang L., Zhang G., Ling J. (2023). Neuroprotective effects of phenolic antioxidant tert-butylhydroquinone (tBHQ) in brain diseases. Mol. Neurobiol. 60, 4909–4923. 10.1007/s12035-023-03370-3 PubMed DOI
Lobo V., Patil A., Phatak A., Chandra N. (2010). Free radicals, antioxidants and functional foods: impact on human health. Pharmacogn. Rev. 4, 118–126. 10.4103/0973-7847.70902 PubMed DOI PMC
Lopez S. G., Luderer U. (2004). Effects of cyclophosphamide and buthionine sulfoximine on ovarian glutathione and apoptosis. Free Radic. Biol. Med. 36, 1366–1377. 10.1016/j.freeradbiomed.2004.02.067 PubMed DOI
Lu W. J., Ferlito V., Xu C., Flockhart D. A., Caccamese S. (2011). Enantiomers of naringenin as pleiotropic, stereoselective inhibitors of cytochrome P450 isoforms. Chirality 23, 891–896. 10.1002/chir.21005 PubMed DOI PMC
Lu X., Mestres G., Singh V. P., Effati P., Poon J.-F., Engman L., et al. (2017). Selenium- and tellurium-based antioxidants for modulating inflammation and effects on osteoblastic activity. Antioxidants 6, 13. 10.3390/antiox6010013 PubMed DOI PMC
Lu Y., Wang J. T.-W., Li N., Zhu X., Li Y., Bansal S., et al. (2023). Intranasal administration of edaravone nanoparticles improves its stability and brain bioavailability. J. Control. Release 359, 257–267. 10.1016/j.jconrel.2023.06.001 PubMed DOI
Lucente-Schultz R. M., Moore C. M., Leonard A. D., Price B. K., Kosynkin D. V., Lu M., et al. (2009). Antioxidant single-walled carbon nanotubes. J. Am. Chem. Soc. 131, 3934–3941. 10.1021/ja805721p PubMed DOI
Lynch E. D., Gu R., Pierce C., Kil J. (2005). Combined oral delivery of ebselen and allopurinol reduces multiple cisplatin toxicities in rat breast and ovarian cancer models while enhancing anti-tumor activity. Anticancer. Drugs 16, 569–579. 10.1097/00001813-200506000-00013 PubMed DOI
Lyseng-Williamson K. A., Perry C. M. (2003). Micronised purified flavonoid fraction: a review of its use in chronic venous insufficiency, venous ulcers and haemorrhoids. Drugs 63, 71–100. 10.2165/00003495-200363010-00005 PubMed DOI
MacDonald-Wicks L. K., Wood L. G., Garg M. L. (2006). Methodology for the determination of biological antioxidant capacity DOI
Maini R. (2004). Infliximab treatment of rheumatoid arthritis. Rheum. Dis. Clin. N. Am. 30, 329–347. 10.1016/j.rdc.2004.01.009 PubMed DOI
Maldonado-Sanabria L. A., Rodriguez-Saavedra I. N., Reyes-Peña I. V., Castillo-Aguirre A., Maldonado M., Crespo A., et al. (2024). Comparative study of the antioxidant activity of the conformers of c-tetra(4-methoxyphenyl)calix[4]resorcinarene. Int. J. Mol. Sci. 25, 10010. 10.3390/ijms251810010 PubMed DOI PMC
Malig T., Xiao Z., Chen S. R. W., Back T. G. (2016). Suppression of Store overload-induced calcium release by hydroxylated metabolites of carvedilol. Bioorg. Med. Chem. Lett. 26, 149–153. 10.1016/j.bmcl.2015.11.008 PubMed DOI
Manach C., Donovan J. L. (2004). Pharmacokinetics and metabolism of dietary flavonoids in humans. Free Radic. Res. 38, 771–785. 10.1080/10715760410001727858 PubMed DOI
Manda G., Rojo A. I., Martínez-Klimova E., Pedraza-Chaverri J., Cuadrado A. (2020). Nordihydroguaiaretic acid: from herbal medicine to clinical development for cancer and chronic diseases. Front. Pharmacol. 11, 151. 10.3389/fphar.2020.00151 PubMed DOI PMC
Maples K. R., Green A. R., Floyd R. A. (2004). Nitrone-related therapeutics: potential of NXY-059 for the treatment of acute ischaemic stroke. CNS Drugs 18, 1071–1084. 10.2165/00023210-200418150-00003 PubMed DOI
Maples K. R., Ma F., Zhang Y.-K. (2001). Comparison of the radical trapping ability of PBN, S-PPBN and NXY-059. Free Radic. Res. 34, 417–426. 10.1080/10715760100300351 PubMed DOI
Marco-Contelles J. (2024). α-Phenyl-N-tert-Butylnitrone and analogous α-Aryl-N-alkylnitrones as neuroprotective antioxidant agents for stroke. Antioxidants 13, 440. 10.3390/antiox13040440 PubMed DOI PMC
Marsanasco M., Márquez A. L., Wagner J. R., del V., Alonso S., Chiaramoni N. S. (2011). Liposomes as vehicles for vitamins E and C: an alternative to fortify Orange juice and offer vitamin C protection after heat treatment. Food Res. Int. 44, 3039–3046. 10.1016/j.foodres.2011.07.025 DOI
Marshall L. F., Marshall S. B. (1995). Pitfalls and advances from the international tirilazad trial in moderate and severe head injury. J. Neurotrauma 12, 929–932. 10.1089/neu.1995.12.929 PubMed DOI
Matteucci M., Habibe M., Robson K., Baldassano A., Riffenburgh R., Tanen D. (2006). Effect of oral calcium disodium EDTA on iron absorption in a human model of iron overdose. Clin. Toxicol. 44, 39–43. 10.1080/15563650500394761 PubMed DOI
May S. W. (2016). Selenium-based drug design. Top. Med. Chem. 17, 87–118. 10.1007/7355_2015_86 DOI
Mazzanti C. M., Spanevello R., Ahmed M., Pereira L. B., Gonçalves J. F., Corrêa M., et al. (2009). Pre-treatment with ebselen and vitamin E modulate acetylcholinesterase activity: interaction with demyelinating agents. Int. J. Dev. Neurosci. 27, 73–80. 10.1016/j.ijdevneu.2008.09.005 PubMed DOI
McDonald R. (1966). Deferoxamine and diethylenetriaminepentaacetic acid (DTPA) in thalassemia. J. Pediatr. 69, 563–571. 10.1016/S0022-3476(66)80041-0 PubMed DOI
Mello-Filho A. C., Meneghini R. (1991). Iron is the intracellular metal involved in the production of DNA damage by oxygen radicals. Mutat. Res. - Fundam. Mol. Mech. Mutagen. 251, 109–113. 10.1016/0027-5107(91)90220-I PubMed DOI
Menon S., Vartak R., Patel K., Billack B. (2021). Evaluation of the antifungal activity of an ebselen-loaded nanoemulsion in a mouse model of vulvovaginal candidiasis. Nanomedicine Nanotechnol. Biol. Med. 37, 102428. 10.1016/j.nano.2021.102428 PubMed DOI
Merlot A. M., Kalinowski D. S., Richardson D. R. (2013). Novel chelators for cancer treatment: where are we now? Antioxid. Redox Signal. 18, 973–1006. 10.1089/ars.2012.4540 PubMed DOI
Mittal M., Siddiqui M. R., Tran K., Reddy S. P., Malik A. B. (2014). Reactive oxygen species in inflammation and tissue injury. Antioxid. Redox Signal. 20, 1126–1167. 10.1089/ars.2012.5149 PubMed DOI PMC
Miyaji Y., Yoshimura S., Sakai N., Yamagami H., Egashira Y., Shirakawa M., et al. (2015). Effect of edaravone on favorable outcome in patients with acute cerebral large vessel occlusion: subanalysis of RESCUE-japan registry. Neurol. Med. Chir. (Tokyo) 55, 241–247. 10.2176/nmc.ra.2014-0219 PubMed DOI PMC
Moad G., Rizzardo E., Thang S. H. (2008). Toward living radical polymerization. Acc. Chem. Res. 41, 1133–1142. 10.1021/ar800075n PubMed DOI
Monnier L., Colette C., Michel F., Cristol J.-P., Owens D. R. (2011). Insulin therapy has a complex relationship with measure of oxidative stress in type 2 diabetes: a case for further study. Diabetes Metab. Res. Rev. 27, 348–353. 10.1002/dmrr.1174 PubMed DOI
Monteiro K. K. A. C., Shiroma M. E., Damous L. L., Simões M. D. J., Simões R. D. S., Cipolla-Neto J., et al. (2024). Antioxidant actions of melatonin: a systematic review of animal studies. Antioxidants 13, 439. 10.3390/antiox13040439 PubMed DOI PMC
Moorhouse P. C., Grootveld M., Halliwell B., Quinlan J. G., Gutteridge J. M. C. (1987). Allopurinol and oxypurinol are hydroxyl radical scavengers. FEBS Lett. 213, 23–28. 10.1016/0014-5793(87)81458-8 PubMed DOI
Moosmann B., Behl C. (2002). Antioxidants as treatment for neurodegenerative disorders. Expert Opin. Investig. Drugs 11, 1407–1435. 10.1517/13543784.11.10.1407 PubMed DOI
Mozafari F., Rashidzadeh H., Bijani S., Zare-Molaei F., Islambulchilar Z., Danafar H., et al. (2023). Enhancing the neuroprotection potential of edaravone in transient global ischemia treatment with glutathione- (GSH-) conjugated poly(methacrylic acid) nanogel as a promising carrier for targeted brain drug delivery. Oxid. Med. Cell. Longev. 2023, 7643280. 10.1155/2023/7643280 PubMed DOI PMC
Mückter H., Liebl B., Reichl F.-X., Hunder G., Walther U., Fichtl B. (1997). Are we ready to replace dimercaprol (BAL) as an arsenic antidote? Hum. Exp. Toxicol. 16, 460–465. 10.1177/096032719701600807 PubMed DOI
Muldrew K. M., Franks A. M. (2009). Succinobucol: review of the metabolic, antiplatelet and cardiovascular effects. Expert Opin. Investig. Drugs 18, 531–539. 10.1517/13543780902849244 PubMed DOI
Murakami K., Yoshino M. (2020). Generation of reactive oxygen species by hydroxypyridone compound/iron complexes. Redox Rep. 25, 59–63. 10.1080/13510002.2020.1787662 PubMed DOI PMC
Murphy M. P., Holmgren A., Larsson N.-G., Halliwell B., Chang C. J., Kalyanaraman B., et al. (2011). Unraveling the biological roles of reactive oxygen species. Cell Metab. 13, 361–366. 10.1016/j.cmet.2011.03.010 PubMed DOI PMC
Murphy P. G., Myers D. S., Davies M. J., Webster N. R., Jones J. G. (1992). The antioxidant potential of propofol (2,6-diisopropylphenol). Br. J. Anaesth. 68, 613–618. 10.1093/bja/68.6.613 PubMed DOI
Murrell G. A. C., Hooper G. (1992). An insight into Dupuytren’s contracture. Ann. R. Coll. Surg. Engl. 74, 156–161. PubMed PMC
Nair C. K. K., Parida D. K., Nomura T. (2001). Radioprotectors in radiotherapy. J. Radiat. Res. (Tokyo) 42, 21–37. 10.1269/jrr.42.21 PubMed DOI
Nasi S., Castelblanco M., Chobaz V., Ehirchiou D., So A., Bernabei I., et al. (2021). Xanthine oxidoreductase is involved in chondrocyte mineralization and expressed in osteoarthritic damaged cartilage. Front. Cell Dev. Biol. 9, 612440. 10.3389/fcell.2021.612440 PubMed DOI PMC
Neupane G. P., Kim D.-M. (2009). Comparison of the effects of deferasirox, deferiprone, and deferoxamine on the growth and virulence of Vibrio vulnificus. Transfus. Paris. 49, 1762–1769. 10.1111/j.1537-2995.2009.02186.x PubMed DOI
Nick H., Acklin P., Lattmann R., Buehlmayer P., Hauffe S., Schupp J., et al. (2003). Development of tridentate iron chelators: from desferrithiocin to ICL670. Curr. Med. Chem. 10, 1065–1076. 10.2174/0929867033457610 PubMed DOI
Noda Y., Tanaka H., Ito Y., Morimoto N., Tsuruma K., Shimazawa M., et al. (2013). Neuroprotective effects of SUN N8075 on neuronal cell death in the Huntington’s disease mice. J. Pharmacol. Sci. 121, 142P.
Novelli M., D’aleo V., Lupi R., Paolini M., Soleti A., Marchetti P., et al. (2007). Reduction of oxidative stress by a new low-molecular-weight antioxidant improves metabolic alterations in a Nonobese mouse diabetes model. Pancreas 35, e10–e17. 10.1097/mpa.0b013e318150e4f2 PubMed DOI
Ojino K., Shimazawa M., Ohno Y., Otsuka T., Tsuruma K., Hara H. (2014). Protective effect of SUN N8075, a free radical scavenger, against excessive light-induced retinal damage in mice. Biol. Pharm. Bull. 37, 424–430. 10.1248/bpb.b13-00778 PubMed DOI
Oliver D. M. A., Reddy P. H. (2019). Small molecules as therapeutic drugs for Alzheimer’s disease. Mol. Cell. Neurosci. 96, 47–62. 10.1016/j.mcn.2019.03.001 PubMed DOI PMC
Orian L., Mauri P., Roveri A., Toppo S., Benazzi L., Bosello-Travain V., et al. (2015). Selenocysteine oxidation in glutathione peroxidase catalysis: an MS-supported quantum mechanics study. Free Radic. Biol. Med. 87, 1–14. 10.1016/j.freeradbiomed.2015.06.011 PubMed DOI
Pacher P., Nivorozhkin A., Szabó C. (2006). Therapeutic effects of xanthine oxidase inhibitors: Renaissance half a century after the discovery of allopurinol. Pharmacol. Rev. 58, 87–114. 10.1124/pr.58.1.6 PubMed DOI PMC
Padayatty S. J., Levine M. (2016). Vitamin C: the known and the unknown and goldilocks. Oral Dis. 22, 463–493. 10.1111/odi.12446 PubMed DOI PMC
Panés J., Perry M., Granger D. N. (1999). Leukocyte-endothelial cell adhesion: avenues for therapeutic intervention. Br. J. Pharmacol. 126, 537–550. 10.1038/sj.bjp.0702328 PubMed DOI PMC
Pariagh S., Tasker K. M., Fry F. H., Holme A. L., Collins C. A., Okarter N., et al. (2005). Asymmetric organotellurides as potent antioxidants and building blocks of protein conjugates. Org. Biomol. Chem. 3, 975–980. 10.1039/b500409h PubMed DOI
Parnham M. J., Sies H. (2013). The early research and development of ebselen. Biochem. Pharmacol. 86, 1248–1253. 10.1016/j.bcp.2013.08.028 PubMed DOI
Pearson A. G., Pullar J. M., Cook J., Spencer E. S., Vissers M. C., Carr A. C., et al. (2021). Peroxiredoxin 2 oxidation reveals hydrogen peroxide generation within erythrocytes during high-dose vitamin C administration. Redox Biol. 43, 101980. 10.1016/j.redox.2021.101980 PubMed DOI PMC
Pendyala L., Creaven P. J., Schwartz G., Meropol N. J., Bolanowska-Higdon W., Zdanowicz J., et al. (2000). Intravenous ifosfamide/mesna is associated with depletion of plasma thiols without depletion of leukocyte glutathione. Clin. Cancer Res. 6, 1314–1321. PubMed
Persson H. L., Richardson D. R. (2005). Iron-binding drugs targeted to lysosomes: a potential strategy to treat inflammatory lung disorders. Expert Opin. Investig. Drugs 14, 997–1008. 10.1517/13543784.14.8.997 PubMed DOI
Petrov D., Mansfield C., Moussy A., Hermine O. (2017). ALS clinical trials review: 20 years of failure. Are we any closer to registering a new treatment? Front. Aging Neurosci. 9, 68. 10.3389/fnagi.2017.00068 PubMed DOI PMC
Pimentel L. S., Allard S., Do Carmo S., Weinreb O., Danik M., Hanzel C. E., et al. (2015). The multi-target drug M30 shows pro-cognitive and anti-inflammatory effects in a rat model of Alzheimer’s disease. J. Alzheimers Dis. 47, 373–383. 10.3233/JAD-143126 PubMed DOI
Poeggeler B., Durand G., Polidori A., Pappolla M. A., Vega-Naredo I., Coto-Montes A., et al. (2005). Mitochondrial medicine: neuroprotection and life extension by the new amphiphilic nitrone LPBNAH acting as a highly potent antioxidant agent. J. Neurochem. 95, 962–973. 10.1111/j.1471-4159.2005.03425.x PubMed DOI
Poli G., Cheeseman K. H., Biasi F., Chiarpotto E., Dianzani M. U., Esterbauer H., et al. (1989). Promethazine inhibits the formation of aldehydic products of lipid peroxidation but not covalent binding resulting from the exposure of rat liver fractions to CCl4. Biochem. J. 264, 527–532. 10.1042/bj2640527 PubMed DOI PMC
Potashkin J. A., Blume S. R., Runkle N. K. (2011). Limitations of animal models of Parkinson’s disease. Park. Dis. 2011, 658083. 10.4061/2011/658083 PubMed DOI PMC
Praticò D. (2008). Evidence of oxidative stress in Alzheimer’s disease brain and antioxidant therapy: lights and shadows. Ann. N. Y. Acad. Sci. 1147, 70–78. 10.1196/annals.1427.010 PubMed DOI
Primas N., Lano G., Brun D., Curti C., Sallée M., Sampol-Manos E., et al. (2023). Stability study of parenteral N-acetylcysteine, and chemical inhibition of its dimerization. Pharmaceuticals 16, 72. 10.3390/ph16010072 PubMed DOI PMC
Puoliväli J., Nurmi A., Miettinen T.-K., Soleti A., Riccardino F., Kalesnykas G., et al. (2011). The radical scavenger IAC (bis(1-hydroxy-2,2,6,6-tetramethyl-4-piperidinyl) decantionate) decreases mortality, enhances cognitive functions in water maze and reduces amyloid plaque burden in hAβPP transgenic mice. J. Alzheimers Dis. 27, 499–510. 10.3233/JAD-2011-110881 PubMed DOI
Puri V., Chaudhary K. R., Singh A., Singh C. (2022). Inhalation potential of N-Acetylcysteine loaded PLGA nanoparticles for the management of tuberculosis: PubMed DOI PMC
Putman S., Anderson F., Morrison M. R., Turtle P., Passini B. T., Wilner A. N., et al. (1994). Safety study of tirilazad mesylate in patients with acute ischemic stroke (stipas). Stroke 25, 418–423. 10.1161/01.STR.25.2.418 PubMed DOI
Qin F., Siwik D. A., Luptak I., Hou X., Wang L., Higuchi A., et al. (2012). The polyphenols resveratrol and S17834 prevent the structural and functional sequelae of diet-induced metabolic heart disease in mice. Circulation 125, 1757–S6. 10.1161/CIRCULATIONAHA.111.067801 PubMed DOI PMC
Rahhal S., Richter H. W. (1988). Reduction of hydrogen peroxide by the ferrous iron chelate of diethylenetriamine-N, N, N’, N’’, N’’-pentaacetate. J. Am. Chem. Soc. 110, 3126–3133. 10.1021/ja00218a022 DOI
Ramachandran A., Jaeschke H. (2021). Oxidant stress and acetaminophen hepatotoxicity: mechanism-Based drug development. Antioxid. Redox Signal. 35, 718–733. 10.1089/ars.2021.0102 PubMed DOI PMC
Ramli F. F., Cowen P. J., Godlewska B. R. (2022). The potential use of ebselen in treatment-resistant depression. Pharmaceuticals 15, 485. 10.3390/ph15040485 PubMed DOI PMC
Ravi P., Isaq M., Ramachandra Y. L., Somu P., Rai P. S., Poojari C. C., et al. (2023). “Therapeutic effectiveness of phytochemicals targeting specific cancer cells: a review of the evidence,” in Recent frontiers of phytochemicals: applications in food, pharmacy, cosmetics, and biotechnology (Amsterdam, Netherlands: Elsevier; ), 247–259. 10.1016/B978-0-443-19143-5.00039-6 DOI
Ren H., Han R., Chen X., Liu X., Wan J., Wang L., et al. (2020). Potential therapeutic targets for intracerebral hemorrhage-associated inflammation: an update. J. Cereb. Blood Flow. Metab. 40, 1752–1768. 10.1177/0271678X20923551 PubMed DOI PMC
Riganti C., Costamagna C., Bosia A., Ghigo D. (2006). The NADPH oxidase inhibitor apocynin (acetovanillone) induces oxidative stress. Toxicol. Appl. Pharmacol. 212, 179–187. 10.1016/j.taap.2005.07.011 PubMed DOI
Riganti C., Gazzano E., Polimeni M., Costamagna C., Bosia A., Ghigo D. (2004). Diphenyleneiodonium inhibits the cell redox metabolism and induces oxidative stress. J. Biol. Chem. 279, 47726–47731. 10.1074/jbc.M406314200 PubMed DOI
Robinson I., De Serna D. G., Gutierrez A., Schade D. S. (2006). Vitamin E in humans: an explanation of clinical trial failure. Endocr. Pract. 12, 576–582. 10.4158/EP.12.5.576 PubMed DOI
Sadowska-Bartosz I., Bartosz G. (2024). The cellular and organismal effects of nitroxides and nitroxide-containing nanoparticles. Int. J. Mol. Sci. 25, 1446. 10.3390/ijms25031446 PubMed DOI PMC
Saez J. C., Ward P. H., Gunther B., Vivaldi E. (1984). Superoxide radical involvement in the pathogenesis of burn shock. Circ. Shock 12, 229–239. PubMed
Saha T., Sinha S., Harfoot R., Quiñones-Mateu M. E., Das S. C. (2023). Spray-dried inhalable microparticles combining remdesivir and ebselen against SARS-CoV-2 infection. Pharmaceutics 15, 2229. 10.3390/pharmaceutics15092229 PubMed DOI PMC
Sahoo P., Lenka D. R., Batabyal M., Pain P. K., Kumar S., Manna D., et al. (2023). Detailed insights into the inhibitory mechanism of new ebselen derivatives against main protease (mpro) of severe acute respiratory syndrome coronavirus-2 (SARS-CoV-2). ACS Pharmacol. Transl. Sci. 6, 171–180. 10.1021/acsptsci.2c00203 PubMed DOI PMC
Sandhir R., Mahajan N., Mehrotra A., Aggarwal A., Sunkaria A. (2015). 4-Hydroxy tempo improves mitochondrial and neurobehavioral deficits in experimental model of Huntington’s disease. Synapse 69, 128–138. 10.1002/syn.21793 PubMed DOI
Sarkar C., Chandra G., Peng S., Zhang Z., Liu A., Mukherjee A. B. (2013). Neuroprotection and lifespan extension in Ppt1-/- mice by NtBuHA: therapeutic implications for INCL. Nat. Neurosci. 16, 1608–1617. 10.1038/nn.3526 PubMed DOI PMC
Sarma B. K., Mugesh G. (2005). Glutathione peroxidase (GPx)-like antioxidant activity of the organoselenium drug ebselen: unexpected complications with thiol exchange reactions. J. Am. Chem. Soc. 127, 11477–11485. 10.1021/ja052794t PubMed DOI
Sasaninia K., Kelley M., Abnousian A., Badaoui A., Alexander L., Sheren N., et al. (2023). Topical absorption of glutathione–cyclodextrin nanoparticle complex in healthy human subjects improves immune response against PubMed DOI PMC
Schmitt B., Vicenzi M., Garrel C., Denis F. M. (2015). Effects of N-acetylcysteine, oral glutathione (GSH) and a novel sublingual form of GSH on oxidative stress markers: a comparative crossover study. Redox Biol. 6, 198–205. 10.1016/j.redox.2015.07.012 PubMed DOI PMC
Seet R. C.-S., Lee C.-Y. J., Lim E. C. H., Quek A. M. L., Huang S.-H., Khoo C.-M., et al. (2010). Markers of oxidative damage are not elevated in otherwise healthy individuals with the metabolic syndrome. Diabetes Care 33, 1140–1142. 10.2337/dc09-2124 PubMed DOI PMC
Seixas J. D., Chaves-Ferreira M., Montes-Grajales D., Gonçalves A. M., Marques A. R., Saraiva L. M., et al. (2015). An N-acetyl cysteine ruthenium tricarbonyl conjugate enables simultaneous release of CO and ablation of reactive oxygen species. Chem. - Eur. J. 21, 14708–14712. 10.1002/chem.201502474 PubMed DOI PMC
Sesso H. D., Buring J. E., Christen W. G., Kurth T., Belanger C., MacFadyen J., et al. (2008). Vitamins E and C in the prevention of cardiovascular disease in men: the physicians’ health study II randomized controlled trial. JAMA - J. Am. Med. Assoc. 300, 2123–2133. 10.1001/jama.2008.600 PubMed DOI PMC
Shams F., Livingstone I., Oladiwura D., Ramaesh K. (2014). Treatment of corneal cystine crystal accumulation in patients with cystinosis. Clin. Ophthalmol. 8, 2077–2084. 10.2147/OPTH.S36626 PubMed DOI PMC
Shaw I. C. (2018). Food safety: the science of keeping food safe. 2nd Edn. Hoboken, USA: Wiley and Sons.
Sherif A. H., Khalil R. H., Talaat T. S., Baromh M. Z., Elnagar M. A. (2024). Dietary nanocomposite of vitamin C and vitamin E enhanced the performance of nile tilapia. Sci. Rep. 14, 15648. 10.1038/s41598-024-65507-1 PubMed DOI PMC
Shih M. F., Cherng J. Y. (2014). Reduction of adhesion molecule production and alteration of eNOS and endothelin-1 mRNA expression in endothelium by Euphorbia hirta L. through its beneficial β- amyrin molecule. Molecules 19, 10534–10545. 10.3390/molecules190710534 PubMed DOI PMC
Shimizu K., Rajapakse N., Horiguchi T., Payne R. M., Busija D. W. (2003). Neuroprotection against hypoxia-ischemia in neonatal rat brain by novel superoxide dismutase mimetics. Neurosci. Lett. 346, 41–44. 10.1016/S0304-3940(03)00558-5 PubMed DOI
Siles A. M., Martínez-Hernández E., Araque J., Diaz-Manera J., Rojas-Garcia R., Gallardo E., et al. (2018). Antibodies against cell adhesion molecules and neural structures in paraneoplastic neuropathies. Ann. Clin. Transl. Neurol. 5, 559–569. 10.1002/acn3.554 PubMed DOI PMC
Silva F. S. G., Simões R. F., Couto R., Oliveira P. J. (2016). Targeting mitochondria in cardiovascular diseases. Curr. Pharm. Des. 22, 5698–5717. 10.2174/1381612822666160822150243 PubMed DOI
Simons J. M., ’t Hart B. A., Ip V. C., Theo R. A. M., Van Dijk H., Labadie R. P. (1990). Metabolic activation of natural phenols into selective oxidative burst agonists by activated human neutrophils. Free Radic. Biol. Med. 8, 251–258. 10.1016/0891-5849(90)90070-Y PubMed DOI
Simunek T., Boer C., Bouwman R., Vlasblom R., Versteilen A., Sterba M., et al. (2005). SIH -: a novel lipophilic iron chelator -: protects H9c2 cardiomyoblasts from oxidative stress-induced mitochondrial injury and cell death. J. Mol. Cell. Cardiol. 39, 345–354. 10.1016/j.yjmcc.2005.05.008 PubMed DOI
Smith R. A. J., Hartley R. C., Murphy M. P. (2012). “Mitochondria-targeted antioxidants,” in Mitochondrial signaling in health and disease (Boca Raton, USA: CRC Press; ), 111–127. Available online at: https://www.scopus.com/inward/record.uri?eid=2-s2.0-85028106843&partnerID=40&md5=8eb1b2275a047321ee94daff69c8051d.
Soares P., Silva C., Chavarria D., Silva F. S. G., Oliveira P. J., Borges F. (2023). Drug discovery and amyotrophic lateral sclerosis: emerging challenges and therapeutic opportunities. Ageing Res. Rev. 83, 101790. 10.1016/j.arr.2022.101790 PubMed DOI
Spinnewyn B., Mautino G., Marin J.-G., Rocher M. N., Grandoulier A. S., Ferrandis E., et al. (2011). BN82451 attenuates l-dopa-induced dyskinesia in 6-OHDA-lesioned rat model of Parkinson’s disease. Neuropharmacology 60, 692–700. 10.1016/j.neuropharm.2010.11.019 PubMed DOI
Stef G., Csiszar A., Xiangmin Z., Ferdinandy P., Ungvari Z., Veress G. (2007). Inhibition of NAD(P)H oxidase attenuates aggregation of platelets from high-risk cardiac patients with aspirin resistance. Pharmacol. Rep. 59, 428–436. PubMed
Steinhubl S. R. (2008). Why have antioxidants failed in clinical trials? Am. J. Cardiol. 101, S14–S19. 10.1016/j.amjcard.2008.02.003 PubMed DOI
Sugita S., Inomata M., Kono Y., Shiroshita H., Etoh T., Shiraishi N., et al. (2013). Effect of the new synthetic vitamin E derivative ETS-GS on radiation enterocolitis symptoms in a rat model. Oncol. Lett. 6, 1229–1233. 10.3892/ol.2013.1581 PubMed DOI PMC
Sunitha K., Hemshekhar M., Thushara R. M., Santhosh M. S., Yariswamy M., Kemparaju K., et al. (2013). N-Acetylcysteine amide: a derivative to fulfill the promises of N-Acetylcysteine. Free Radic. Res. 47, 357–367. 10.3109/10715762.2013.781595 PubMed DOI
Sutton H. C., Winterbourn C. C. (1984). Chelated iron-catalyzed OH. Formation from paraquat radicals and H2O2: mechanism of formate oxidation. Arch. Biochem. Biophys. 235, 106–115. 10.1016/0003-9861(84)90259-5 PubMed DOI
Tambyraja A. L., Mitchell R., Driscoll P. J., Deans C., Parks R. W., Rahman I., et al. (2007). Glutathione supplementation to university of Wisconsin solution causes endothelial dysfunction. Transpl. Immunol. 18, 146–150. 10.1016/j.trim.2007.06.002 PubMed DOI
Tanaka H., Ito Y., Shimazaki H., Tsuruma K., Shimazawa M., Hara H. (2010). SUN N8075 delays disease onset and progression in mice model of amyotrophic lateral sclerosis. J. Pharmacol. Sci. 112, 68P.
Tang X.-L., Kaur H., Sun J.-Z., Qiu Y., Park S.-W., Schleman M., et al. (1995a). Effect of the hydrophilic α-tocopherol analog MDL 74,405 on detection of hydroxyl radicals in stunned myocardium in dogs. Am. Heart J. 130, 940–948. 10.1016/0002-8703(95)90192-2 PubMed DOI
Tang X.-L., Mccay P. B., Sun J.-Z., Hartley C. J., Schleman M., Bolli R. (1995b). Inhibitory effect of a hydrophilic αtocopherol analogue, MDL 74, 405, on generation of free radicals in stunned myocardium in dogs. Free Radic. Res. 22, 293–302. 10.3109/10715769509145641 PubMed DOI
Tangirala R. K., Casanada F., Miller E., Witztum J. L., Steinberg D., Palinski W. (1995). Effect of the antioxidant N,N’-Diphenyl 1,4-phenylenediamine (DPPD) on atherosclerosis in ApoE-deficient mice. Arterioscler. Thromb. Vasc. Biol. 15, 1625–1630. 10.1161/01.ATV.15.10.1625 PubMed DOI
Tanguy S., Durand G., Reboul C., Polidori A., Pucci B., Dauzat M., et al. (2006). Protection against reactive oxygen species injuries in rat isolated perfused hearts: effect of LPBNAH, a new amphiphilic spin-trap derived from PBN. Cardiovasc. Drugs Ther. 20, 147–149. 10.1007/s10557-006-6754-8 PubMed DOI
Tauchen J., Huml L., Rimpelova S., Jurášek M. (2020). Flavonoids and related members of the aromatic polyketide group in human health and disease: do they really work? Molecules 25, 25173846. 10.3390/molecules25173846 PubMed DOI PMC
Tayade K., Yeom G., Sahoo S., Puschmann H., Nimse S., Kuwar A. (2022). Exploration of molecular structure, dft calculations, and antioxidant activity of a hydrazone derivative. Antioxidants 11, 2138. 10.3390/antiox11112138 PubMed DOI PMC
Ten Freyhaus H., Huntgeburth M., Wingler K., Schnitker J., Bäumer A. T., Vantler M., et al. (2006). Novel nox inhibitor VAS2870 attenuates PDGF-Dependent smooth muscle cell chemotaxis, but not proliferation. Cardiovasc. Res. 71, 331–341. 10.1016/j.cardiores.2006.01.022 PubMed DOI
Terpstra M., Torkelson C., Emir U., Hodges J. S., Raatz S. (2011). Noninvasive quantification of human brain antioxidant concentrations after an intravenous bolus of vitamin C. NMR Biomed. 24, 521–528. 10.1002/nbm.1619 PubMed DOI PMC
Thakare R., Kaul G., Shukla M., Kesharwani P., Srinivas N., Dasgupta A., et al. (2020). “Chapter 5 - repurposing nonantibiotic drugs as antibacterials,” in Drug discovery targeting drug-resistant bacteria. Editors Kesharwani P., Chopra S., Dasgupta A. (Academic Press; ), 105–138. 10.1016/B978-0-12-818480-6.00005-9 DOI
Traber M. G., Head B. (2021). Vitamin E: how much is enough, too much and why. Free Radic. Biol. Med. 177, 212–225. 10.1016/j.freeradbiomed.2021.10.028 PubMed DOI
Tsai M.-S., Liou G.-G., Liao J.-W., Lai P.-Y., Yang D.-J., Wu S.-H., et al. (2024). N-acetyl cysteine overdose induced acute toxicity and hepatic microvesicular steatosis by disrupting GSH and interfering lipid metabolisms in normal mice. Antioxidants 13, 832. 10.3390/antiox13070832 PubMed DOI PMC
Ursini F., Maiorino M., Forman H. J. (2016). Redox homeostasis: the golden mean of healthy living. Redox Biol. 8, 205–215. 10.1016/j.redox.2016.01.010 PubMed DOI PMC
Valdivia A., Pérez-Álvarez S., Aroca-Aguilar J. D., Ikuta I., Jordán J. (2009). Superoxide dismutases: a physiopharmacological update. J. Physiol. Biochem. 65, 195–208. 10.1007/BF03179070 PubMed DOI
Valgimigli L., Amorati R. (2019). “CHAPTER 11: Vitamin E. Inspired synthetic antioxidants,”. Vitamin E: chemistry and nutritional benefits (food chemistry, function and analysis. Editor Niki E. (London, UK: The Royal Society of Chemistry; ), 11, 151–164. 10.1039/9781788016216-00151 DOI
Valko M., Leibfritz D., Moncol J., Cronin M. T. D., Mazur M., Telser J. (2007). Free radicals and antioxidants in normal physiological functions and human disease. Int. J. Biochem. Cell Biol. 39, 44–84. 10.1016/j.biocel.2006.07.001 PubMed DOI
VanOrden H. E., Hagemann T. M. (2006). Deferasirox - an oral agent for chronic iron overload. Ann. Pharmacother. 40, 1110–1117. 10.1345/aph.1G566 PubMed DOI
Vasina V., Broccoli M., Ursino M. G., Bellot S. F., Soleti A., Paolini M., et al. (2009). Effects of the non-peptidyl low molecular weight radical scavenger IAC in DNBS-Induced colitis in rats. Eur. J. Pharmacol. 614, 137–145. 10.1016/j.ejphar.2009.04.021 PubMed DOI
Wang X., Guo S., Li Z., Luo Q., Dai Y., Zhang H., et al. (2021). Amphiphilic branched polymer-nitroxides conjugate as a nanoscale agent for potential magnetic resonance imaging of multiple objects PubMed DOI PMC
Wang Z., Li W., Wang Y., Li X., Huang L., Li X. (2016). Design, synthesis and evaluation of clioquinol-ebselen hybrids as multi-target-directed ligands against Alzheimer’s disease. RSC Adv. 6, 7139–7158. 10.1039/c5ra26797h DOI
Winterbourn C. C., Metodiewa D. (1999). Reactivity of biologically important thiol compounds with superoxide and hydrogen peroxide. Free Radic. Biol. Med. 27, 322–328. 10.1016/S0891-5849(99)00051-9 PubMed DOI
Wiśniewski M., Bieniek A., Roszek K., Czarnecka J., Bolibok P., Ferrer P., et al. (2018). Cystine-based MBioF for maintaining the antioxidant-oxidant balance in airway diseases. ACS Med. Chem. Lett. 9, 1280–1284. 10.1021/acsmedchemlett.8b00468 PubMed DOI PMC
Witting P. K., Mohr D., Stocker R. (1999). Assessment of prooxidant activity of vitamin E in human low-density lipoprotein and plasma. Methods Enzymol. 299, 362–375. 10.1016/S0076-6879(99)99036-5 PubMed DOI
Woo K. J., Lee T.-J., Park J.-W., Kwon T. K. (2006). Desferrioxamine, an iron chelator, enhances HIF-1alpha accumulation PubMed DOI
Xiao J., Lv Y., Lin B., Tipoe G. L., Youdim M. B. H., Xing F., et al. (2015). A novel antioxidant multitarget iron chelator M30 protects hepatocytes against ethanol-induced injury. Oxid. Med. Cell. Longev. 2015, 607271. 10.1155/2015/607271 PubMed DOI PMC
Xu J., Wang A., Meng X., Yalkun G., Xu A., Gao Z., et al. (2021). Edaravone dexborneol PubMed DOI
Xu S., Jiang B., Hou X., Shi C., Bachschmid M. M., Zang M., et al. (2011). High-fat diet increases and the polyphenol, s17834, decreases acetylation of the sirtuin-1-dependent lysine-382 on p53 and apoptotic signaling in atherosclerotic lesion-prone aortic endothelium of normal mice. J. Cardiovasc. Pharmacol. 58, 263–271. 10.1097/FJC.0b013e3182239eb7 PubMed DOI PMC
Yamaguchi T., Sano K., Takakura K., Saito I., Shinohara Y., Asano T., et al. (1998). Ebselen in acute ischemic stroke: a placebo-controlled, double-blind clinical trial. Ebselen study group. Stroke 29, 12–17. 10.1161/01.STR.29.1.12 PubMed DOI
Yamamoto S., Hagiwara S., Hidaka S., Shingu C., Goto K., Kashima K., et al. (2011). The antioxidant EPC-K1 attenuates renal ischemia-reperfusion injury in a rat model. Am. J. Nephrol. 33, 485–490. 10.1159/000327820 PubMed DOI
Yang L., Calingasan N. Y., Chen J., Ley J. J., Becker D. A., Beal M. F. (2005). A novel azulenyl nitrone antioxidant protects against MPTP and 3-nitropropionic acid neurotoxicities. Exp. Neurol. 191, 86–93. 10.1016/j.expneurol.2004.07.012 PubMed DOI
Yang X., Song W., Liu N., Sun Z., Liu R., Liu Q. S., et al. (2018). Synthetic phenolic antioxidants cause perturbation in steroidogenesis PubMed DOI
Yao X., Zhang Y., Hao J., Duan H.-Q., Zhao C.-X., Sun C., et al. (2019). Deferoxamine promotes recovery of traumatic spinal cord injury by inhibiting ferroptosis. Neural Regen. Res. 14, 532–541. 10.4103/1673-5374.245480 PubMed DOI PMC
Yasui F., Sudoh M., Arai M., Kohara M. (2013). Synthetic lipophilic antioxidant BO-653 suppresses HCV replication. J. Med. Virol. 85, 241–249. 10.1002/jmv.23466 PubMed DOI
Ye M., Lin W., Zheng J., Lin S. (2021). N-acetylcysteine for chronic kidney disease: a systematic review and meta-analysis. Am. J. Transl. Res. 13, 2472–2485. PubMed PMC
Yonekuta Y., Oyaizu K., Nishide H. (2007). Structural implication of oxoammonium cations for reversible organic one-electron redox reaction to nitroxide radicals. Chem. Lett. 36, 866–867. 10.1246/cl.2007.866 DOI
Younus H. (2018). Therapeutic potentials of superoxide dismutase. Int. J. Health Sci. Qassim 12, 88–93. PubMed PMC
Yuan Y., Zha H., Rangarajan P., Ling E.-A., Wu C. (2014). Anti-inflammatory effects of edaravone and scutellarin in activated microglia in experimentally induced ischemia injury in rats and in BV-2 microglia. BMC Neurosci. 15, 125. 10.1186/s12868-014-0125-3 PubMed DOI PMC
Yue T.-L., Gu J.-L., Lysko P. G., Cheng H.-Y., Barone F. C., Feuerstein G. (1992). Neuroprotective effects of phenyl-t-butyl-nitrone in gerbil global brain ischemia and in cultured rat cerebellar neurons. Brain Res. 574, 193–197. 10.1016/0006-8993(92)90816-R PubMed DOI
Zang M., Xu S., Maitland-Toolan K. A., Zuccollo A., Hou X., Jiang B., et al. (2006). Polyphenols stimulate AMP-Activated protein kinase, lower lipids, and inhibit accelerated atherosclerosis in diabetic LDL receptor-deficient mice. Diabetes 55, 2180–2191. 10.2337/db05-1188 PubMed DOI
Zeng L., Tan L., Li H., Zhang Q., Li Y., Guo J. (2018). Deferoxamine therapy for intracerebral hemorrhage: a systematic review. PLoS ONE 13, e0193615. 10.1371/journal.pone.0193615 PubMed DOI PMC
Zesiewicz T., Salemi J. L., Perlman S., Sullivan K. L., Shaw J. D., Huang Y., et al. (2018). Double-blind, randomized and controlled trial of EPI-743 in Friedreich’s ataxia. Neurodegener. Dis. Manag. 8, 233–242. 10.2217/nmt-2018-0013 PubMed DOI
Zhang J., Zhou X., Wu W., Wang J., Xie H., Wu Z. (2017). Regeneration of glutathione by α-lipoic acid PubMed DOI
Zhang R., Pinson A., Samuni A. (1998). Both hydroxylamine and nitroxide protect cardiomyocytes from oxidative stress. Free Radic. Biol. Med. 24, 66–75. 10.1016/S0891-5849(97)00165-2 PubMed DOI
Zhang W. R., Hayashi T., Sasaki C., Sato K., Nagano I., Manabe Y., et al. (2001). Attenuation of oxidative DNA damage with a novel antioxidant EPC-K1 in rat brain neuronal cells after transient middle cerebral artery occlusion. Neurol. Res. 23, 676–680. 10.1179/016164101101199027 PubMed DOI
Zhang X.-R., Zhou W.-X., Zhang Y.-X. (2018). Improvements in SOD mimic AEOL-10150, a potent broad-spectrum antioxidant. Mil. Med. Res. 5, 30. 10.1186/s40779-018-0176-3 PubMed DOI PMC
Zondagh L. S., Malan S. F., Joubert J. (2020). Design, synthesis and biological evaluation of edaravone derivatives bearing the N-benzyl pyridinium moiety as multifunctional anti-alzheimer’s agents. J. Enzyme Inhib. Med. Chem. 35, 1596–1605. 10.1080/14756366.2020.1801673 PubMed DOI PMC
Zou L., Lu J., Wang J., Ren X., Zhang L., Gao Y., et al. (2017). Synergistic antibacterial effect of silver and ebselen against multidrug-resistant Gram-negative bacterial infections. EMBO Mol. Med. 9, 1165–1178. 10.15252/emmm.201707661 PubMed DOI PMC