Antioxidant Metabolism Pathways in Vitamins, Polyphenols, and Selenium: Parallels and Divergences
Abstract
:1. Introduction
2. Relationship between RS, Aging, and Related Diseases
3. Molecular Antioxidant Capacity and Antioxidant Defenses
3.1. Definition and Features of Antioxidant Capacity
3.2. Antioxidant Defenses
3.2.1. Endogenous Defenses
3.2.2. Vitamins and Polyphenols as Exogenous Antioxidants
- (a)
- The 3-hydroxy-4-ketone groups on the C-ring (denoted “site 3-4”), resulting in a maltol-like coordination mode;
- (b)
- The 5-hydroxy group on the A-ring and the 4-carbonyl group on the C-ring (denoted “site 4-5”), resulting in a coordination mode similar to acetylacetone;
- (c)
- 3′,4′-dihydroxy groups located on the B-ring (denoted “3′-4′ site”), resulting in a coordination similar to the catechol mode;
- (d)
- The 6,7-dihydroxy groups on ring A (Figure 7).
3.2.3. Role of Selenium in Antioxidant Metabolism
4. Relationship of Antioxidant Metabolism Pathways, Sirtuins, and NRF2
4.1. Regulation of Sirtuins SIRT1 and SIRT3
4.2. Activation of the Transcription Factor NRF2 by Polyphenols, Vitamins, and Selenium
4.2.1. Transcription Factor NRF2 and Polyphenols
4.2.2. Transcription Factor NRF2 and Vitamins
4.2.3. Transcription Factor NRF2 and Selenium
5. Transcriptional Regulation of Polyphenols, Vitamins, and Selenium
5.1. Regulation of NF-κB
- -
- They can inhibit the activity of IKK, thereby preventing the phosphorylation and subsequent degradation of IκB proteins. This action blocks the translocation of NF-κB to the nucleus, preventing it from activating gene expression [195].
- -
- Indirectly inhibiting NF-κB activation due to its antioxidant properties [196].
- -
- Can influence the composition of NF-κB subunits, thereby altering the activity of the NF-κB complex. The p65 subunit, also known as RelA, is a key component of the NF-κB complex and plays a crucial role in the transcriptional activity of NF-κB. This inhibition can prevent the translocation of NF-κB into the nucleus and the transcription of pro-inflammatory cytokines [197].
- -
- Can disrupt upstream signaling pathways of NF-κB activation, as they can interfere with Toll-like receptors (TLRs) [198] or cytokine receptors [199], which are crucial for the initiation of NF-κB signaling cascades. By doing so, polyphenols can inhibit the activation of NF-κB, thereby potentially reducing the expression of NF-κB-dependent genes, many of which are involved in inflammatory responses.
5.2. Regulation of AP-1
5.3. Regulation of STAT3
5.4. Regulation of BACH1
6. Polyphenol-Mediated Enzyme Regulation
6.1. NADPH Oxidase
6.2. Cyclooxygenase 2
6.3. Lysyl Oxidase
6.4. Lipoxygenase
6.5. Xanthine Oxidase
6.6. α-Synuclein
6.7. Receptor Tyrosine Kinases
6.8. Histone Deacetylases
6.9. α-Amylase and α-Glucosidase
7. Is Bioavailability an Important Issue in the Functionality of Antioxidants?
8. Can Antioxidants Act as Pro-Oxidants?
- Increasing the formation of ROS: Certain substances can increase the production of ROS, which, in turn, can act as pro-oxidants themselves. This can lead to a cycle of oxidative damage and further increases in OS.
- Hindering the action of antioxidant enzymes and pathways: Pro-oxidants can interfere with the activity of antioxidant enzymes and pathways in the body, reducing their effectiveness in terms of neutralizing ROS and protecting against oxidative damage.
8.1. Pro-Oxidant Function of Vitamins
8.2. Pro-Oxidant Function of Polyphenols
8.3. Pro-Oxidant Function of Selenium
9. Conclusions
Author Contributions
Funding
Conflicts of Interest
Correction Statement
References
- Manisalidis, I.; Stavropoulou, E.; Stavropoulos, A.; Bezirtzoglou, E. Environmental and Health Impacts of Air Pollution: A Review. Front. Public Health 2020, 8, 14. [Google Scholar] [CrossRef] [PubMed]
- Ponnampalam, E.N.; Kiani, A.; Santhiravel, S.; Holman, B.W.B.; Lauridsen, C.; Dunshea, F.R. The Importance of Dietary Antioxidants on Oxidative Stress, Meat and Milk Production, and Their Preservative Aspects in Farm Animals: Antioxidant Action, Animal Health, and Product Quality—Invited Review. Animals 2022, 12, 3279. [Google Scholar] [CrossRef]
- Bjørklund, G.; Shanaida, M.; Lysiuk, R.; Butnariu, M.; Peana, M.; Sarac, I.; Strus, O.; Smetanina, K.; Chirumbolo, S. Natural Compounds and Products from an Anti-Aging Perspective. Molecules 2022, 27, 7084. [Google Scholar] [CrossRef]
- Murdaca, G.; Gangemi, S. Vitamin D in Health and Disease. Biomedicines 2023, 11, 10. [Google Scholar]
- Barbouti, A.; Goulas, V. Dietary Antioxidants in the Mediterranean Diet. Antioxidants 2021, 10, 1213. [Google Scholar] [CrossRef]
- Boeing, H.; Bechthold, A.; Bub, A.; Ellinger, S.; Haller, D.; Kroke, A.; Leschik-Bonnet, E.; Müller, M.J.; Oberritter, H.; Schulze, M.; et al. Critical review: Vegetables and fruit in the prevention of chronic diseases. Eur. J. Nutr. 2012, 51, 637–663. [Google Scholar] [CrossRef]
- Curieses Andrés, C.M.; Pérez de la Lastra, J.M.; Andrés Juan, C.; Plou, F.J.; Pérez-Lebeña, E. From reactive species to disease development: Effect of oxidants and antioxidants on the cellular biomarkers. J. Biochem. Mol. Toxicol. 2023, 37, e23455. [Google Scholar] [CrossRef]
- Liu, Z.; Zhou, T.; Ziegler, A.C.; Dimitrion, P.; Zuo, L. Oxidative Stress in Neurodegenerative Diseases: From Molecular Mechanisms to Clinical Applications. Oxidative Med. Cell. Longev. 2017, 2017, 2525967. [Google Scholar] [CrossRef]
- Bjelakovic, G.; Nikolova, D.; Gluud, L.L.; Simonetti, R.G.; Gluud, C. Antioxidant supplements for prevention of mortality in healthy participants and patients with various diseases. Cochrane Database Syst. Rev. 2012, 2012, CD007176. [Google Scholar] [CrossRef]
- Almeida, S.; Ozkan, S.; Gonçalves, D.; Paulo, I.; Queirós, C.S.G.P.; Ferreira, O.; Bordado, J.; Galhano dos Santos, R. A Brief Evaluation of Antioxidants, Antistatics, and Plasticizers Additives from Natural Sources for Polymers Formulation. Polymers 2023, 15, 6. [Google Scholar] [CrossRef]
- Juan, C.A.; Pérez de la Lastra, J.M.; Plou, F.J.; Pérez-Lebeña, E. The Chemistry of Reactive Oxygen Species (ROS) Revisited: Outlining Their Role in Biological Macromolecules (DNA, Lipids and Proteins) and Induced Pathologies. Int. J. Mol. Sci. 2021, 22, 4642. [Google Scholar] [CrossRef]
- Pérez de la Lastra, J.M.; Juan, C.A.; Plou, F.J.; Pérez-Lebeña, E. The Nitration of Proteins, Lipids and DNA by Peroxynitrite Derivatives-Chemistry Involved and Biological Relevance. Stresses 2022, 2, 53–64. [Google Scholar] [CrossRef]
- Childs, B.G.; Durik, M.; Baker, D.J.; van Deursen, J.M. Cellular senescence in aging and age-related disease: From mechanisms to therapy. Nat. Med. 2015, 21, 1424–1435. [Google Scholar] [CrossRef]
- Andrés, C.M.C.; Pérez de la Lastra, J.M.; Andrés Juan, C.; Plou, F.J.; Pérez-Lebeña, E. Superoxide Anion Chemistry—Its Role at the Core of the Innate Immunity. Int. J. Mol. Sci. 2023, 24, 1841. [Google Scholar] [CrossRef]
- Liguori, I.; Russo, G.; Curcio, F.; Bulli, G.; Aran, L.; Della-Morte, D.; Gargiulo, G.; Testa, G.; Cacciatore, F.; Bonaduce, D. Oxidative stress, aging, and diseases. Clin. Interv. Aging 2018, 13, 757–772. [Google Scholar] [CrossRef]
- Chandrasekaran, A.; Idelchik, M.d.P.S.; Melendez, J.A. Redox control of senescence and age-related disease. Redox Biol. 2017, 11, 91–102. [Google Scholar] [CrossRef]
- Ferrara, N.; Rinaldi, B.; Corbi, G.; Conti, V.; Stiuso, P.; Boccuti, S.; Rengo, G.; Rossi, F.; Filippelli, A. Exercise training promotes SIRT1 activity in aged rats. Rejuvenation Res. 2008, 11, 139–150. [Google Scholar] [CrossRef]
- Luo, J.; Mills, K.; le Cessie, S.; Noordam, R.; van Heemst, D. Ageing, age-related diseases and oxidative stress: What to do next? Ageing Res. Rev. 2020, 57, 100982. [Google Scholar] [CrossRef]
- Marquez-Exposito, L.; Tejedor-Santamaria, L.; Valentijn, F.A.; Tejera-Muñoz, A.; Rayego-Mateos, S.; Marchant, V.; Rodrigues-Diez, R.R.; Rubio-Soto, I.; Knoppert, S.N.; Ortiz, A.; et al. Oxidative Stress and Cellular Senescence Are Involved in the Aging Kidney. Antioxidants 2022, 11, 301. [Google Scholar] [CrossRef]
- Halliwell, B. How to Characterize a Biological Antioxidant. Free Radic. Res. Commun. 1990, 9, 1–32. [Google Scholar] [CrossRef]
- Lobo, V.; Patil, A.; Phatak, A.; Chandra, N. Free radicals, antioxidants and functional foods: Impact on human health. Pharmacogn. Rev. 2010, 4, 118–126. [Google Scholar] [CrossRef]
- Slemmer, J.E.; Shacka, J.J.; Sweeney, M.I.; Weber, J.T. Antioxidants and free radical scavengers for the treatment of stroke, traumatic brain injury and aging. Curr. Med. Chem. 2008, 15, 404–414. [Google Scholar] [CrossRef] [PubMed]
- Borba, C.M.; Tavares, M.N.; Macedo, L.P.; Araújo, G.S.; Furlong, E.B.; Dora, C.L.; Burkert, J.F.M. Physical and chemical stability of β-carotene nanoemulsions during storage and thermal process. Food Res. Int. 2019, 121, 229–237. [Google Scholar] [CrossRef] [PubMed]
- Munteanu, I.G.; Apetrei, C. Analytical Methods Used in Determining Antioxidant Activity: A Review. Int. J. Mol. Sci. 2021, 22, 3380. [Google Scholar] [CrossRef] [PubMed]
- Shahidi, F.; Zhong, Y. Measurement of antioxidant activity. J. Funct. Foods 2015, 18, 757–781. [Google Scholar] [CrossRef]
- Lü, J.-M.; Lin, P.H.; Yao, Q.; Chen, C. Chemical and molecular mechanisms of antioxidants: Experimental approaches and model systems. J. Cell. Mol. Med. 2010, 14, 840–860. [Google Scholar] [CrossRef] [PubMed]
- Kurutas, E.B. The importance of antioxidants which play the role in cellular response against oxidative/nitrosative stress: Current state. Nutr. J. 2016, 15, 71. [Google Scholar] [CrossRef] [PubMed]
- Birben, E.; Sahiner, U.M.; Sackesen, C.; Erzurum, S.; Kalayci, O. Oxidative stress and antioxidant defense. World Allergy Organ. J. 2012, 5, 9–19. [Google Scholar] [CrossRef] [PubMed]
- Andrés, C.M.C.; Pérez de la Lastra, J.M.; Juan, C.A.; Plou, F.J.; Pérez-Lebeña, E. Chemistry of Hydrogen Peroxide Formation and Elimination in Mammalian Cells, and Its Role in Various Pathologies. Stresses 2022, 2, 256–274. [Google Scholar] [CrossRef]
- Ren, X.; Zou, L.; Zhang, X.; Branco, V.; Wang, J.; Carvalho, C.; Holmgren, A.; Lu, J. Redox Signaling Mediated by Thioredoxin and Glutathione Systems in the Central Nervous System. Antioxid. Redox Signal. 2017, 27, 989–1010. [Google Scholar] [CrossRef]
- Aranda-Rivera, A.K.; Cruz-Gregorio, A.; Arancibia-Hernández, Y.L.; Hernández-Cruz, E.Y.; Pedraza-Chaverri, J. RONS and Oxidative Stress: An Overview of Basic Concepts. Oxygen 2022, 2, 437–478. [Google Scholar] [CrossRef]
- Flohé, L.; Toppo, S.; Orian, L. The glutathione peroxidase family: Discoveries and mechanism. Free Radic. Biol. Med. 2022, 187, 113–122. [Google Scholar] [CrossRef]
- Masuda, R.; Kimura, R.; Karasaki, T.; Sase, S.; Goto, K. Modeling the Catalytic Cycle of Glutathione Peroxidase by Nuclear Magnetic Resonance Spectroscopic Analysis of Selenocysteine Selenenic Acids. J. Am. Chem. Soc. 2021, 143, 6345–6350. [Google Scholar] [CrossRef]
- Orian, L.; Mauri, P.; Roveri, A.; Toppo, S.; Benazzi, L.; Bosello-Travain, V.; De Palma, A.; Maiorino, M.; Miotto, G.; Zaccarin, M.; et al. Selenocysteine oxidation in glutathione peroxidase catalysis: An MS-supported quantum mechanics study. Free Radic. Biol. Med. 2015, 87, 1–14. [Google Scholar] [CrossRef]
- Winterbourn, C.C. Toxicity of iron and hydrogen peroxide: The Fenton reaction. Toxicol. Lett. 1995, 82–83, 969–974. [Google Scholar] [CrossRef]
- Karimi, Z.; Bahadoran, Z.; Abedini, S.; Rad, A.H.; Rashidkhani, B. Dietary total antioxidant capacity and the risk of breast cancer: A case-control study. EMHJ-East. Mediterr. Health J. 2015, 21, 564–571. [Google Scholar] [CrossRef]
- Halvorsen, B.L.; Carlsen, M.H.; Phillips, K.M.; Bøhn, S.K.; Holte, K.; Jacobs, D.R., Jr.; Blomhoff, R. Content of redox-active compounds (ie, antioxidants) in foods consumed in the United States. Am. J. Clin. Nutr. 2006, 84, 95–135. [Google Scholar] [CrossRef]
- Shahidi, F. Antioxidants in food and food antioxidants. Nahrung 2000, 44, 158–163. [Google Scholar] [CrossRef]
- Cooke, M.S.; Evans, M.D.; Mistry, N.; Lunec, J. Role of dietary antioxidants in the prevention of in vivo oxidative DNA damage. Nutr. Res. Rev. 2002, 15, 19–42. [Google Scholar] [CrossRef]
- Wongama Given, P.; Dirk Jacobus, B.; Adriaan Johannes, E.; Guillaume, A. Dietary Antioxidant Properties of Vegetable Oils and Nuts—The Race Against Cardiovascular Disease Progression. In Antioxidant-Antidiabetic Agents and Human Health; Oluwafemi, O., Ed.; IntechOpen: Rijeka, Croatia, 2014. [Google Scholar]
- Mendonça, J.d.S.; Guimarães, R.d.C.A.; Zorgetto-Pinheiro, V.A.; Fernandes, C.D.P.; Marcelino, G.; Bogo, D.; Freitas, K.d.C.; Hiane, P.A.; de Pádua Melo, E.S.; Vilela, M.L.B.; et al. Natural Antioxidant Evaluation: A Review of Detection Methods. Molecules 2022, 27, 3563. [Google Scholar] [CrossRef]
- Zehiroglu, C.; Ozturk Sarikaya, S.B. The importance of antioxidants and place in today’s scientific and technological studies. J. Food Sci. Technol. 2019, 56, 4757–4774. [Google Scholar] [CrossRef]
- Huang, Z.; Liu, Y.; Qi, G.; Brand, D.; Zheng, S.G. Role of Vitamin A in the Immune System. J. Clin. Med. 2018, 7, 258. [Google Scholar] [CrossRef]
- Black, R.E.; Allen, L.H.; Bhutta, Z.A.; Caulfield, L.E.; De Onis, M.; Ezzati, M.; Mathers, C.; Rivera, J. Maternal and child undernutrition: Global and regional exposures and health consequences. Lancet 2008, 371, 243–260. [Google Scholar] [CrossRef]
- Sommer, A.; Vyas, K.S. A global clinical view on vitamin A and carotenoids. Am. J. Clin. Nutr. 2012, 96, 1204S–1206S. [Google Scholar] [CrossRef]
- Palace, V.P.; Khaper, N.; Qin, Q.; Singal, P.K. Antioxidant potentials of vitamin A and carotenoids and their relevance to heart disease. Free Radic. Biol. Med. 1999, 26, 746–761. [Google Scholar] [CrossRef]
- Lewis, E.D.; Meydani, S.N.; Wu, D. Regulatory role of vitamin E in the immune system and inflammation. IUBMB Life 2019, 71, 487–494. [Google Scholar] [CrossRef]
- Mahir, M.A.; Mohd Fauzi, M.S.H.; Mohamed Rehan, A.; Mohammed, E. Production of natural food-derived vitamin c from orange juice. In Chemical Process and Sustainability in Medical Biotechnology; UTHM: Parit Raja, Malaysia, 2022. [Google Scholar]
- Neves, J.R.; Grether-Beck, S.; Krutmann, J.; Correia, P.; Gonçalves, J.E., Jr.; Sant’Anna, B.; Kerob, D. Efficacy of a topical serum containing L-ascorbic acid, neohesperidin, pycnogenol, tocopherol, and hyaluronic acid in relation to skin aging signs. J. Cosmet. Dermatol. 2022, 21, 4462–4469. [Google Scholar] [CrossRef]
- Chambial, S.; Dwivedi, S.; Shukla, K.K.; John, P.J.; Sharma, P. Vitamin C in disease prevention and cure: An overview. Indian. J. Clin. Biochem. 2013, 28, 314–328. [Google Scholar] [CrossRef]
- Padayatty, S.J.; Katz, A.; Wang, Y.; Eck, P.; Kwon, O.; Lee, J.-H.; Chen, S.; Corpe, C.; Dutta, A.; Dutta, S.K.; et al. Vitamin C as an Antioxidant: Evaluation of Its Role in Disease Prevention. J. Am. Coll. Nutr. 2003, 22, 18–35. [Google Scholar] [CrossRef]
- Aune, D. Plant Foods, Antioxidant Biomarkers, and the Risk of Cardiovascular Disease, Cancer, and Mortality: A Review of the Evidence. Adv. Nutr. 2019, 10, S404–S421. [Google Scholar] [CrossRef]
- de Lourdes Samaniego-Vaesken, M.; Alonso-Aperte, E.; Varela-Moreiras, G. Vitamin food fortification today. Food Nutr. Res. 2012, 56. [Google Scholar] [CrossRef]
- Hever, J.; Cronise, R.J. Plant-based nutrition for healthcare professionals: Implementing diet as a primary modality in the prevention and treatment of chronic disease. J. Geriatr. Cardiol. 2017, 14, 355–368. [Google Scholar] [CrossRef]
- Rizvi, S.; Raza, S.T.; Ahmed, F.; Ahmad, A.; Abbas, S.; Mahdi, F. The role of vitamin E in human health and some diseases. Sultan Qaboos Univ. Med. J. 2014, 14, e157. [Google Scholar]
- Poudel, P.; Petropoulos, S.A.; Di Gioia, F. Plant Tocopherols and Phytosterols and Their Bioactive Properties. In Natural Secondary Metabolites: From Nature, Through Science, to Industry; Carocho, M., Heleno, S.A., Barros, L., Eds.; Springer International Publishing: Cham, Switzerland, 2023; pp. 285–319. [Google Scholar]
- Szewczyk, K.; Chojnacka, A.; Górnicka, M. Tocopherols and Tocotrienols—Bioactive Dietary Compounds; What Is Certain, What Is Doubt? Int. J. Mol. Sci. 2021, 22, 6222. [Google Scholar] [CrossRef]
- Brglez Mojzer, E.; Knez Hrnčič, M.; Škerget, M.; Knez, Ž.; Bren, U. Polyphenols: Extraction Methods, Antioxidative Action, Bioavailability and Anticarcinogenic Effects. Molecules 2016, 21, 901. [Google Scholar] [CrossRef]
- Pandey, K.B.; Rizvi, S.I. Plant Polyphenols as Dietary Antioxidants in Human Health and Disease. Oxidative Med. Cell. Longev. 2009, 2, 270–278. [Google Scholar] [CrossRef] [PubMed]
- Marín, L.; Miguélez, E.M.; Villar, C.J.; Lombó, F. Bioavailability of dietary polyphenols and gut microbiota metabolism: Antimicrobial properties. Biomed. Res. Int. 2015, 2015, 905215. [Google Scholar] [CrossRef] [PubMed]
- Smeriglio, A.; Barreca, D.; Bellocco, E.; Trombetta, D. Proanthocyanidins and hydrolysable tannins: Occurrence, dietary intake and pharmacological effects. Br. J. Pharmacol. 2017, 174, 1244–1262. [Google Scholar] [CrossRef]
- Rudrapal, M.; Khairnar, S.J.; Khan, J.; Dukhyil, A.B.; Ansari, M.A.; Alomary, M.N.; Alshabrmi, F.M.; Palai, S.; Deb, P.K.; Devi, R. Dietary Polyphenols and Their Role in Oxidative Stress-Induced Human Diseases: Insights Into Protective Effects, Antioxidant Potentials and Mechanism(s) of Action. Front. Pharmacol. 2022, 13, 806470. [Google Scholar] [CrossRef]
- Bistrian, B.R. Dietary Modulation of Cell Signaling Pathways. Gastroenterology 2009, 137, 737. [Google Scholar] [CrossRef]
- Leri, M.; Scuto, M.; Ontario, M.L.; Calabrese, V.; Calabrese, E.J.; Bucciantini, M.; Stefani, M. Healthy Effects of Plant Polyphenols: Molecular Mechanisms. Int. J. Mol. Sci. 2020, 21, 1250. [Google Scholar] [CrossRef] [PubMed]
- Costa, M.; Sezgin-Bayindir, Z.; Losada-Barreiro, S.; Paiva-Martins, F.; Saso, L.; Bravo-Díaz, C. Polyphenols as Antioxidants for Extending Food Shelf-Life and in the Prevention of Health Diseases: Encapsulation and Interfacial Phenomena. Biomedicines 2021, 9, 1909. [Google Scholar] [CrossRef] [PubMed]
- Pérez de la Lastra, J.M.; Juan, C.A.; Plou, F.J.; Pérez-Lebeña, E. Nitration of Flavonoids and Tocopherols as Potential Modulators of Nitrosative Stress—A Study Based on Their Conformational Structures and Energy Content. Stresses 2022, 2, 213–230. [Google Scholar] [CrossRef]
- Lakey-Beitia, J.; Burillo, A.M.; La Penna, G.; Hegde, M.L.; Rao, K.S. Polyphenols as Potential Metal Chelation Compounds Against Alzheimer’s Disease. J. Alzheimers Dis. 2021, 82, S335–S357. [Google Scholar] [CrossRef] [PubMed]
- Pietta, P.G. Flavonoids as antioxidants. J. Nat. Prod. 2000, 63, 1035–1042. [Google Scholar] [CrossRef] [PubMed]
- Perron, N.R.; Brumaghim, J.L. A review of the antioxidant mechanisms of polyphenol compounds related to iron binding. Cell Biochem. Biophys. 2009, 53, 75–100. [Google Scholar] [CrossRef]
- Zhou, B.; Wu, L.M.; Yang, L.; Liu, Z.L. Evidence for alpha-tocopherol regeneration reaction of green tea polyphenols in SDS micelles. Free Radic. Biol. Med. 2005, 38, 78–84. [Google Scholar] [CrossRef]
- Saini, R.K.; Prasad, P.; Lokesh, V.; Shang, X.; Shin, J.; Keum, Y.-S.; Lee, J.-H. Carotenoids: Dietary Sources, Extraction, Encapsulation, Bioavailability, and Health Benefits—A Review of Recent Advancements. Antioxidants 2022, 11, 795. [Google Scholar] [CrossRef]
- Ratnasari, N.; Walters, M.; Tsopmo, A. Antioxidant and lipoxygenase activities of polyphenol extracts from oat brans treated with polysaccharide degrading enzymes. Heliyon 2017, 3, e00351. [Google Scholar] [CrossRef]
- Owczarek, K.; Lewandowska, U. The Impact of Dietary Polyphenols on COX-2 Expression in Colorectal Cancer. Nutr. Cancer 2017, 69, 1105–1118. [Google Scholar] [CrossRef]
- Nastasijević, B.; Lazarević-Pašti, T.; Dimitrijević-Branković, S.; Pašti, I.; Vujačić, A.; Joksić, G.; Vasić, V. Inhibition of myeloperoxidase and antioxidative activity of Gentiana lutea extracts. J. Pharm. Biomed. Anal. 2012, 66, 191–196. [Google Scholar] [CrossRef]
- Maraldi, T. Natural compounds as modulators of NADPH oxidases. Oxidative Med. Cell. Longev. 2013, 2013, 271602. [Google Scholar] [CrossRef]
- Borges, F.; Fernandes, E.; Roleira, F. Progress towards the discovery of xanthine oxidase inhibitors. Curr. Med. Chem. 2002, 9, 195–217. [Google Scholar] [CrossRef] [PubMed]
- Mileo, A.M.; Miccadei, S. Polyphenols as Modulator of Oxidative Stress in Cancer Disease: New Therapeutic Strategies. Oxidative Med. Cell Longev. 2016, 2016, 6475624. [Google Scholar] [CrossRef] [PubMed]
- Ingold, K.U. Inhibition of the Autoxidation of Organic Substances in the Liquid Phase. Chem. Rev. 1961, 61, 563–589. [Google Scholar] [CrossRef]
- Kammoun, M.; Miladi, S.; Ali, Y.B.; Damak, M.; Gargouri, Y.; Bezzine, S. In vitro study of the PLA2 inhibition and antioxidant activities of Aloe vera leaf skin extracts. Lipids Health Dis. 2011, 10, 30. [Google Scholar] [CrossRef]
- Kanner, J. Polyphenols by Generating H2O2, Affect Cell Redox Signaling, Inhibit PTPs and Activate Nrf2 Axis for Adaptation and Cell Surviving: In Vitro, In Vivo and Human Health. Antioxidants 2020, 9, 797. [Google Scholar] [CrossRef]
- Rodríguez-Sureda, V.; Vilches, Á.; Sánchez, O.; Audí, L.; Domínguez, C. Intracellular oxidant activity, antioxidant enzyme defense system, and cell senescence in fibroblasts with trisomy 21. Oxidative Med. Cell Longev. 2015, 2015, 509241. [Google Scholar] [CrossRef]
- Kurek-Górecka, A.; Rzepecka-Stojko, A.; Górecki, M.; Stojko, J.; Sosada, M.; Świerczek-Zięba, G. Structure and Antioxidant Activity of Polyphenols Derived from Propolis. Molecules 2014, 19, 78–101. [Google Scholar] [CrossRef]
- Ramirez-Tortosa, C.; Andersen, Ø.M.; Gardner, P.T.; Morrice, P.C.; Wood, S.G.; Duthie, S.J.; Collins, A.R.; Duthie, G.G. Anthocyanin-rich extract decreases indices of lipid peroxidation and DNA damage in vitamin E-depleted rats. Free Radic. Biol. Med. 2001, 31, 1033–1037. [Google Scholar] [CrossRef]
- Manal Azat, A.; Abdulkareem Shehab, D.; Abeer Abdulrazak, M. Antioxidant Categories and Mode of Action. In Antioxidants; Emad, S., Ed.; IntechOpen: Rijeka, Croatia, 2019. [Google Scholar]
- Juana, M.M.-R.; Pilar, H.-S. Oxidative Stress and Antioxidant Defenses Induced by Physical Exercise. In Basic Principles and Clinical Significance of Oxidative Stress; Sivakumar Joghi Thatha, G., Ed.; IntechOpen: Rijeka, Croatia, 2015. [Google Scholar]
- Cherrak, S.A.; Mokhtari-Soulimane, N.; Berroukeche, F.; Bensenane, B.; Cherbonnel, A.; Merzouk, H.; Elhabiri, M. In Vitro Antioxidant versus Metal Ion Chelating Properties of Flavonoids: A Structure-Activity Investigation. PLoS ONE 2016, 11, e0165575. [Google Scholar] [CrossRef] [PubMed]
- Han, X.; Shen, T.; Lou, H. Dietary polyphenols and their biological significance. Int. J. Mol. Sci. 2007, 8, 950–988. [Google Scholar] [CrossRef]
- Tan, Y.; Kim, J.; Cheng, J.; Ong, M.; Lao, W.-G.; Jin, X.-L.; Lin, Y.-G.; Xiao, L.; Zhu, X.-Q.; Qu, X.-Q. Green tea polyphenols ameliorate non-alcoholic fatty liver disease through upregulating AMPK activation in high fat fed Zucker fatty rats. World J. Gastroenterol. 2017, 23, 3805. [Google Scholar] [CrossRef]
- He, L.; He, T.; Farrar, S.; Ji, L.; Liu, T.; Ma, X. Antioxidants Maintain Cellular Redox Homeostasis by Elimination of Reactive Oxygen Species. Cell Physiol. Biochem. 2017, 44, 532–553. [Google Scholar] [CrossRef]
- Yousefian, M.; Shakour, N.; Hosseinzadeh, H.; Hayes, A.W.; Hadizadeh, F.; Karimi, G. The natural phenolic compounds as modulators of NADPH oxidases in hypertension. Phytomedicine 2019, 55, 200–213. [Google Scholar] [CrossRef]
- Remigante, A.; Spinelli, S.; Straface, E.; Gambardella, L.; Caruso, D.; Falliti, G.; Dossena, S.; Marino, A.; Morabito, R. Antioxidant activity of quercetin in a H2O2-induced oxidative stress model in red blood cells: Functional role of band 3 protein. Int. J. Mol. Sci. 2022, 23, 10991. [Google Scholar] [CrossRef]
- Bocsan, I.C.; Măgureanu, D.C.; Pop, R.M.; Levai, A.M.; Macovei, Ș.O.; Pătrașca, I.M.; Chedea, V.S.; Buzoianu, A.D. Antioxidant and Anti-Inflammatory Actions of Polyphenols from Red and White Grape Pomace in Ischemic Heart Diseases. Biomedicines 2022, 10, 2337. [Google Scholar] [CrossRef] [PubMed]
- Miličević, A.; Raos, N. Modelling of protective mechanism of iron (II)-polyphenol binding with OH-related molecular descriptors. Croat. Chem. Acta 2016, 89, 511–515. [Google Scholar] [CrossRef]
- Cheng, I.F.; Breen, K. On the ability of four flavonoids, baicilein, luteolin, naringenin, and quercetin, to suppress the Fenton reaction of the iron-ATP complex. Biometals 2000, 13, 77–83. [Google Scholar] [CrossRef]
- Selvaraj, S.; Krishnaswamy, S.; Devashya, V.; Sethuraman, S.; Krishnan, U.M. Flavonoid-metal ion complexes: A novel class of therapeutic agents. Med. Res. Rev. 2014, 34, 677–702. [Google Scholar] [CrossRef]
- Yang, J.G.; Yu, H.N.; Sun, S.L.; Zhang, L.C.; He, G.Q.; Das, U.N.; Ruan, H.; Shen, S.R. Epigallocatechin-3-gallate affects the growth of LNCaP cells via membrane fluidity and distribution of cellular zinc. J. Zhejiang Univ. Sci. B 2009, 10, 411–421. [Google Scholar] [CrossRef]
- Kejík, Z.; Kaplánek, R.; Masařík, M.; Babula, P.; Matkowski, A.; Filipenský, P.; Veselá, K.; Gburek, J.; Sýkora, D.; Martásek, P.; et al. Iron Complexes of Flavonoids-Antioxidant Capacity and Beyond. Int. J. Mol. Sci. 2021, 22, 646. [Google Scholar] [CrossRef]
- Kostyuk, V.A.; Potapovich, A.I.; Strigunova, E.N.; Kostyuk, T.V.; Afanas’ev, I.B. Experimental evidence that flavonoid metal complexes may act as mimics of superoxide dismutase. Arch. Biochem. Biophys. 2004, 428, 204–208. [Google Scholar] [CrossRef] [PubMed]
- Mira, L.; Fernandez, M.T.; Santos, M.; Rocha, R.; Florêncio, M.H.; Jennings, K.R. Interactions of flavonoids with iron and copper ions: A mechanism for their antioxidant activity. Free Radic. Res. 2002, 36, 1199–1208. [Google Scholar] [CrossRef] [PubMed]
- Leopoldini, M.; Russo, N.; Chiodo, S.; Toscano, M. Iron chelation by the powerful antioxidant flavonoid quercetin. J. Agric. Food Chem. 2006, 54, 6343–6351. [Google Scholar] [CrossRef]
- Fernandez, M.T.; Mira, M.L.; Florêncio, M.H.; Jennings, K.R. Iron and copper chelation by flavonoids: An electrospray mass spectrometry study. J. Inorg. Biochem. 2002, 92, 105–111. [Google Scholar] [CrossRef] [PubMed]
- Satterfield, M.; Brodbelt, J.S. Enhanced detection of flavonoids by metal complexation and electrospray ionization mass spectrometry. Anal. Chem. 2000, 72, 5898–5906. [Google Scholar] [CrossRef]
- Kim, Y.A.; Tarahovsky, Y.S.; Yagolnik, E.A.; Kuznetsova, S.M.; Muzafarov, E.N. Lipophilicity of flavonoid complexes with iron(II) and their interaction with liposomes. Biochem. Biophys. Res. Commun. 2013, 431, 680–685. [Google Scholar] [CrossRef]
- Baccan, M.M.; Chiarelli-Neto, O.; Pereira, R.M.; Espósito, B.P. Quercetin as a shuttle for labile iron. J. Inorg. Biochem. 2012, 107, 34–39. [Google Scholar] [CrossRef]
- Martins, I.L.; Charneira, C.; Gandin, V.; Ferreira da Silva, J.L.; Justino, G.C.; Telo, J.P.; Vieira, A.J.; Marzano, C.; Antunes, A.M. Selenium-containing chrysin and quercetin derivatives: Attractive scaffolds for cancer therapy. J. Med. Chem. 2015, 58, 4250–4265. [Google Scholar] [CrossRef]
- Khater, M.; Ravishankar, D.; Greco, F.; Osborn, H.M. Metal complexes of flavonoids: Their synthesis, characterization and enhanced antioxidant and anticancer activities. Future Med. Chem. 2019, 11, 2845–2867. [Google Scholar] [CrossRef]
- Minich, W.B. Selenium Metabolism and Biosynthesis of Selenoproteins in the Human Body. Biochemistry 2022, 87, S168–S177. [Google Scholar] [CrossRef]
- Genchi, G.; Lauria, G.; Catalano, A.; Sinicropi, M.S.; Carocci, A. Biological Activity of Selenium and Its Impact on Human Health. Int. J. Mol. Sci. 2023, 24, 2633. [Google Scholar] [CrossRef] [PubMed]
- Perrone, D.; Monteiro, M.; Nunes, J.C. The Chemistry of Selenium. In Selenium: Chemistry, Analysis, Function and Effects; Preedy, V.R., Ed.; The Royal Society of Chemistry: London, UK, 2015. [Google Scholar]
- Barchielli, G.; Capperucci, A.; Tanini, D. The Role of Selenium in Pathologies: An Updated Review. Antioxidants 2022, 11, 251. [Google Scholar] [CrossRef] [PubMed]
- Newton, T.D.; Bolton, S.G.; Garcia, A.C.; Chouinard, J.E.; Golledge, S.L.; Zakharov, L.N.; Pluth, M.D. Hydrolysis-Based Small-Molecule Hydrogen Selenide (H2Se) Donors for Intracellular H2Se Delivery. J. Am. Chem. Soc. 2021, 143, 19542–19550. [Google Scholar] [CrossRef] [PubMed]
- Tangjaidee, P.; Swedlund, P.; Xiang, J.; Yin, H.; Quek, S.Y. Selenium-enriched plant foods: Selenium accumulation, speciation, and health functionality. Front. Nutr. 2022, 9, 962312. [Google Scholar] [CrossRef] [PubMed]
- Rayman, M.P. The importance of selenium to human health. Lancet 2000, 356, 233–241. [Google Scholar] [CrossRef]
- Dodig, S.; Cepelak, I. The facts and controversies about selenium. Acta Pharm. 2004, 54, 261–276. [Google Scholar] [PubMed]
- Hu, Y.; Chai, X.; Men, J.; Rao, S.; Cong, X.; Cheng, S.; Qiao, Z. Does Methionine Status Influence the Outcome of Selenomethinione Supplementation? A Comparative Study of Metabolic and Selenium Levels in HepG2 Cells. Nutrients 2022, 14, 3705. [Google Scholar] [CrossRef]
- Hariharan, S.; Dharmaraj, S. Selenium and selenoproteins: It’s role in regulation of inflammation. Inflammopharmacology 2020, 28, 667–695. [Google Scholar] [CrossRef]
- Zhang, Y.; Roh, Y.J.; Han, S.-J.; Park, I.; Lee, H.M.; Ok, Y.S.; Lee, B.C.; Lee, S.-R. Role of Selenoproteins in Redox Regulation of Signaling and the Antioxidant System: A Review. Antioxidants 2020, 9, 383. [Google Scholar] [CrossRef] [PubMed]
- Zhang, F.; Li, X.; Wei, Y. Selenium and Selenoproteins in Health. Biomolecules 2023, 13, 799. [Google Scholar] [CrossRef] [PubMed]
- Radomska, D.; Czarnomysy, R.; Radomski, D.; Bielawska, A.; Bielawski, K. Selenium as a Bioactive Micronutrient in the Human Diet and Its Cancer Chemopreventive Activity. Nutrients 2021, 13, 1649. [Google Scholar] [CrossRef] [PubMed]
- Pei, J.; Pan, X.; Wei, G.; Hua, Y. Research progress of glutathione peroxidase family (GPX) in redoxidation. Front. Pharmacol. 2023, 14, 1147414. [Google Scholar] [CrossRef]
- Vašková, J.; Kočan, L.; Vaško, L.; Perjési, P. Glutathione-Related Enzymes and Proteins: A Review. Molecules 2023, 28, 1447. [Google Scholar] [CrossRef] [PubMed]
- Alkadi, H. A Review on Free Radicals and Antioxidants. Infect. Disord. Drug Targets 2020, 20, 16–26. [Google Scholar] [CrossRef] [PubMed]
- Cardoso, B.R.; Cominetti, C.; Seale, L.A. Editorial: Selenium, Human Health and Chronic Disease. Front. Nutr. 2021, 8, 827759. [Google Scholar] [CrossRef]
- Bononi, G.; Flori, L.; Citi, V.; Acciai, C.; Nocilla, V.; Martelli, A.; Poli, G.; Tuccinardi, T.; Granchi, C.; Testai, L.; et al. New Synthetic Analogues of Natural Polyphenols as Sirtuin 1-Activating Compounds. Pharmaceuticals 2022, 15, 339. [Google Scholar] [CrossRef]
- Lu, C.; Zhao, H.; Liu, Y.; Yang, Z.; Yao, H.; Liu, T.; Gou, T.; Wang, L.; Zhang, J.; Tian, Y.; et al. Novel Role of the SIRT1 in Endocrine and Metabolic Diseases. Int. J. Biol. Sci. 2023, 19, 484–501. [Google Scholar] [CrossRef]
- Wong, A.; Woodcock, E.A. FoxO proteins and cardiac pathology. Adv. Exp. Med. Biol. 2010, 665, 78–89. [Google Scholar]
- Gu, X.; Han, D.; Chen, W.; Zhang, L.; Lin, Q.; Gao, J.; Fanning, S.; Han, B. SIRT1-mediated FoxOs pathways protect against apoptosis by promoting autophagy in osteoblast-like MC3T3-E1 cells exposed to sodium fluoride. Oncotarget 2016, 7, 65218. [Google Scholar] [CrossRef]
- Jiao, F.; Gong, Z. The Beneficial Roles of SIRT1 in Neuroinflammation-Related Diseases. Oxidative Med. Cell. Longev. 2020, 2020, 6782872. [Google Scholar] [CrossRef] [PubMed]
- Zhang, J.; Tao, J.; Ling, Y.; Li, F.; Zhu, X.; Xu, L.; Wang, M.; Zhang, S.; McCall, C.E.; Liu, T.F. Switch of NAD Salvage to de novo Biosynthesis Sustains SIRT1-RelB-Dependent Inflammatory Tolerance. Front. Immunol. 2019, 10, 2358. [Google Scholar] [CrossRef] [PubMed]
- Anand, S.; Bharadvaja, N. Potential Benefits of Nutraceuticals for Oxidative Stress Management. Rev. Bras. Farm. 2022, 32, 211–220. [Google Scholar] [CrossRef] [PubMed]
- Iside, C.; Scafuro, M.; Nebbioso, A.; Altucci, L. SIRT1 Activation by Natural Phytochemicals: An Overview. Front. Pharmacol. 2020, 11, 1225. [Google Scholar] [CrossRef] [PubMed]
- Yang, Y.; Paik, J.H.; Cho, D.; Cho, J.A.; Kim, C.W. Resveratrol induces the suppression of tumor-derived CD4+CD25+ regulatory T cells. Int. Immunopharmacol. 2008, 8, 542–547. [Google Scholar] [CrossRef] [PubMed]
- Yeung, F.; Hoberg, J.E.; Ramsey, C.S.; Keller, M.D.; Jones, D.R.; Frye, R.A.; Mayo, M.W. Modulation of NF-kappaB-dependent transcription and cell survival by the SIRT1 deacetylase. EMBO J. 2004, 23, 2369–2380. [Google Scholar] [CrossRef] [PubMed]
- Longo, V.D.; Kennedy, B.K. Sirtuins in aging and age-related disease. Cell 2006, 126, 257–268. [Google Scholar] [CrossRef] [PubMed]
- David, A.V.A.; Arulmoli, R.; Parasuraman, S. Overviews of biological importance of quercetin: A bioactive flavonoid. Pharmacogn. Rev. 2016, 10, 84. [Google Scholar]
- Ciumărnean, L.; Milaciu, M.V.; Runcan, O.; Vesa, Ș.C.; Răchișan, A.L.; Negrean, V.; Perné, M.-G.; Donca, V.I.; Alexescu, T.-G.; Para, I.; et al. The Effects of Flavonoids in Cardiovascular Diseases. Molecules 2020, 25, 4320. [Google Scholar] [CrossRef]
- Zhang, F.; Feng, J.; Zhang, J.; Kang, X.; Qian, D. Quercetin modulates AMPK/SIRT1/NF-κB signaling to inhibit inflammatory/oxidative stress responses in diabetic high fat diet-induced atherosclerosis in the rat carotid artery. Exp. Ther. Med. 2020, 20, 280. [Google Scholar] [CrossRef] [PubMed]
- Bazzucchi, I.; Patrizio, F.; Ceci, R.; Duranti, G.; Sabatini, S.; Sgrò, P.; Di Luigi, L.; Sacchetti, M. Quercetin Supplementation Improves Neuromuscular Function Recovery from Muscle Damage. Nutrients 2020, 12, 2850. [Google Scholar] [CrossRef] [PubMed]
- Sgrò, P.; Ceci, R.; Lista, M.; Patrizio, F.; Sabatini, S.; Felici, F.; Sacchetti, M.; Bazzucchi, I.; Duranti, G.; Di Luigi, L. Quercetin Modulates IGF-I and IGF-II Levels After Eccentric Exercise-Induced Muscle-Damage: A Placebo-Controlled Study. Front. Endocrinol. 2021, 12, 745959. [Google Scholar] [CrossRef] [PubMed]
- Yu, Y.; Sun, J.; Wang, R.; Liu, J.; Wang, P.; Wang, C. Curcumin Management of Myocardial Fibrosis and its Mechanisms of Action: A Review. Am. J. Chin. Med. 2019, 47, 1675–1710. [Google Scholar] [CrossRef] [PubMed]
- Gupta, S.C.; Patchva, S.; Koh, W.; Aggarwal, B.B. Discovery of curcumin, a component of golden spice, and its miraculous biological activities. Clin. Exp. Pharmacol. Physiol. 2012, 39, 283–299. [Google Scholar] [CrossRef] [PubMed]
- Zendedel, E.; Butler, A.E.; Atkin, S.L.; Sahebkar, A. Impact of curcumin on sirtuins: A review. J. Cell. Biochem. 2018, 119, 10291–10300. [Google Scholar] [CrossRef]
- Chen, Y.; Fu, L.L.; Wen, X.; Wang, X.Y.; Liu, J.; Cheng, Y.; Huang, J. Sirtuin-3 (SIRT3), a therapeutic target with oncogenic and tumor-suppressive function in cancer. Cell Death Dis. 2014, 5, e1047. [Google Scholar] [CrossRef]
- Zhang, J.; Xiang, H.; Liu, J.; Chen, Y.; He, R.R.; Liu, B. Mitochondrial Sirtuin 3: New emerging biological function and therapeutic target. Theranostics 2020, 10, 8315–8342. [Google Scholar] [CrossRef]
- Rivas-Chacón, L.d.M.; Yanes-Díaz, J.; de Lucas, B.; Riestra-Ayora, J.I.; Madrid-García, R.; Sanz-Fernández, R.; Sánchez-Rodríguez, C. Cocoa Polyphenol Extract Inhibits Cellular Senescence via Modulation of SIRT1 and SIRT3 in Auditory Cells. Nutrients 2023, 15, 544. [Google Scholar] [CrossRef]
- Wei, W.; Li, L.; Zhang, Y.; Geriletu; Yang, J.; Zhang, Y.; Xing, Y. Vitamin C protected human retinal pigmented epithelium from oxidant injury depending on regulating SIRT1. Sci. World J. 2014, 2014, 750634. [Google Scholar] [CrossRef]
- Clifford, T.; Acton, J.P.; Cocksedge, S.P.; Davies, K.A.B.; Bailey, S.J. The effect of dietary phytochemicals on nuclear factor erythroid 2-related factor 2 (Nrf2) activation: A systematic review of human intervention trials. Mol. Biol. Rep. 2021, 48, 1745–1761. [Google Scholar] [CrossRef] [PubMed]
- Gugliandolo, A.; Bramanti, P.; Mazzon, E. Activation of Nrf2 by Natural Bioactive Compounds: A Promising Approach for Stroke? Int. J. Mol. Sci. 2020, 21, 4875. [Google Scholar] [CrossRef] [PubMed]
- Vomhof-DeKrey, E.E.; Picklo, M.J. The Nrf2-antioxidant response element pathway: A target for regulating energy metabolism. J. Nutr. Biochem. 2012, 23, 1201–1206. [Google Scholar] [CrossRef] [PubMed]
- Musial, C.; Kuban-Jankowska, A.; Gorska-Ponikowska, M. Beneficial Properties of Green Tea Catechins. Int. J. Mol. Sci. 2020, 21, 1744. [Google Scholar] [CrossRef] [PubMed]
- Hayes, J.D.; Dinkova-Kostova, A.T. The Nrf2 regulatory network provides an interface between redox and intermediary metabolism. Trends Biochem. Sci. 2014, 39, 199–218. [Google Scholar] [CrossRef] [PubMed]
- Jenkins, T.; Gouge, J. Nrf2 in Cancer, Detoxifying Enzymes and Cell Death Programs. Antioxidants 2021, 10, 1030. [Google Scholar] [CrossRef]
- Brigelius-Flohé, R.; Flohé, L. Basic principles and emerging concepts in the redox control of transcription factors. Antioxid. Redox Signal 2011, 15, 2335–2381. [Google Scholar] [CrossRef]
- Covas, G.; Marinho, H.S.; Cyrne, L.; Antunes, F. Chapter Nine—Activation of Nrf2 by H2O2: De Novo Synthesis Versus Nuclear Translocation. In Methods in Enzymology; Cadenas, E., Packer, L., Eds.; Academic Press: Cambridge, MA, USA, 2013; Volume 528, pp. 157–171. [Google Scholar]
- Zhou, Y.; Jiang, Z.; Lu, H.; Xu, Z.; Tong, R.; Shi, J.; Jia, G. Recent Advances of Natural Polyphenols Activators for Keap1-Nrf2 Signaling Pathway. Chem. Biodivers. 2019, 16, e1900400. [Google Scholar] [CrossRef]
- Scapagnini, G.; Sonya, V.; Nader, A.G.; Calogero, C.; Zella, D.; Fabio, G. Modulation of Nrf2/ARE Pathway by Food Polyphenols: A Nutritional Neuroprotective Strategy for Cognitive and Neurodegenerative Disorders. Mol. Neurobiol. 2011, 44, 192–201. [Google Scholar] [CrossRef]
- Campbell, N.K.; Fitzgerald, H.K.; Dunne, A. Regulation of inflammation by the antioxidant haem oxygenase 1. Nat. Rev. Immunol. 2021, 21, 411–425. [Google Scholar] [CrossRef]
- Chen, K.; Gunter, K.; Maines, M.D. Neurons Overexpressing Heme Oxygenase-1 Resist Oxidative Stress-Mediated Cell Death. J. Neurochem. 2000, 75, 304–313. [Google Scholar] [CrossRef]
- Erlank, H.; Elmann, A.; Kohen, R.; Kanner, J. Polyphenols activate Nrf2 in astrocytes via H2O2, semiquinones, and quinones. Free Radic. Biol. Med. 2011, 51, 2319–2327. [Google Scholar] [CrossRef]
- Wragg, D.; Leoni, S.; Casini, A. Aquaporin-driven hydrogen peroxide transport: A case of molecular mimicry? RSC Chem. Biol. 2020, 1, 390–394. [Google Scholar] [CrossRef]
- Dunlap, T.; Piyankarage, S.C.; Wijewickrama, G.T.; Abdul-Hay, S.; Vanni, M.; Litosh, V.; Luo, J.; Thatcher, G.R.J. Quinone-Induced Activation of Keap1/Nrf2 Signaling by Aspirin Prodrugs Masquerading as Nitric Oxide. Chem. Res. Toxicol. 2012, 25, 2725–2736. [Google Scholar] [CrossRef] [PubMed]
- Klopčič, I.; Dolenc, M.S. Chemicals and Drugs Forming Reactive Quinone and Quinone Imine Metabolites. Chem. Res. Toxicol. 2019, 32, 1–34. [Google Scholar] [CrossRef]
- Ito, S.; Sugumaran, M.; Wakamatsu, K. Chemical Reactivities of ortho-Quinones Produced in Living Organisms: Fate of Quinonoid Products Formed by Tyrosinase and Phenoloxidase Action on Phenols and Catechols. Int. J. Mol. Sci. 2020, 21, 6080. [Google Scholar] [CrossRef] [PubMed]
- Zhou, X.; Iqbal, A.; Li, J.; Liu, C.; Murtaza, A.; Xu, X.; Pan, S.; Hu, W. Changes in Browning Degree and Reducibility of Polyphenols during Autoxidation and Enzymatic Oxidation. Antioxidants 2021, 10, 1809. [Google Scholar] [CrossRef]
- Unoki, T.; Akiyama, M.; Kumagai, Y. Nrf2 Activation and Its Coordination with the Protective Defense Systems in Response to Electrophilic Stress. Int. J. Mol. Sci. 2020, 21, 545. [Google Scholar] [CrossRef]
- Kobayashi, M.; Li, L.; Iwamoto, N.; Nakajima-Takagi, Y.; Kaneko, H.; Nakayama, Y.; Eguchi, M.; Wada, Y.; Kumagai, Y.; Yamamoto, M. The antioxidant defense system Keap1-Nrf2 comprises a multiple sensing mechanism for responding to a wide range of chemical compounds. Mol. Cell Biol. 2009, 29, 493–502. [Google Scholar] [CrossRef] [PubMed]
- Andrés, C.M.C.; Pérez de la Lastra, J.M.; Juan, C.A.; Plou, F.J.; Pérez-Lebeña, E. Polyphenols as Antioxidant/Pro-Oxidant Compounds and Donors of Reducing Species: Relationship with Human Antioxidant Metabolism. Processes 2023, 11, 2771. [Google Scholar] [CrossRef]
- Huang, Y.; Li, W.; Su, Z.Y.; Kong, A.N. The complexity of the Nrf2 pathway: Beyond the antioxidant response. J. Nutr. Biochem. 2015, 26, 1401–1413. [Google Scholar] [CrossRef]
- Chen, S.H.; Li, C.W. Detection and Characterization of Catechol Quinone-Derived Protein Adducts Using Biomolecular Mass Spectrometry. Front. Chem. 2019, 7, 571. [Google Scholar] [CrossRef]
- Bolton, J.L.; Dunlap, T. Formation and Biological Targets of Quinones: Cytotoxic versus Cytoprotective Effects. Chem. Res. Toxicol. 2017, 30, 13–37. [Google Scholar] [CrossRef]
- Sirakawin, C.; Lin, D.; Zhou, Z.; Wang, X.; Kelleher, R.; Huang, S.; Long, W.; Pires-daSilva, A.; Liu, Y.; Wang, J.; et al. SKN-1/NRF2 upregulation by vitamin A is conserved from nematodes to mammals and is critical for lifespan extension in Caenorhabditis elegans. Aging Cell 2023, e14064. [Google Scholar] [CrossRef]
- Xu, L.L.; Zhao, B.; Sun, S.L.; Yu, S.F.; Wang, Y.M.; Ji, R.; Yang, Z.T.; Ma, L.; Yao, Y.; Chen, Y.; et al. High-dose vitamin C alleviates pancreatic injury via the NRF2/NQO1/HO-1 pathway in a rat model of severe acute pancreatitis. Ann. Transl. Med. 2020, 8, 852. [Google Scholar] [CrossRef]
- Mishra, P.; Paital, B.; Jena, S.; Swain, S.S.; Kumar, S.; Yadav, M.K.; Chainy, G.B.N.; Samanta, L. Possible activation of NRF2 by Vitamin E/Curcumin against altered thyroid hormone induced oxidative stress via NFĸB/AKT/mTOR/KEAP1 signalling in rat heart. Sci. Rep. 2019, 9, 7408. [Google Scholar] [CrossRef]
- Nakai, K.; Fujii, H.; Kono, K.; Goto, S.; Kitazawa, R.; Kitazawa, S.; Hirata, M.; Shinohara, M.; Fukagawa, M.; Nishi, S. Vitamin D Activates the Nrf2-Keap1 Antioxidant Pathway and Ameliorates Nephropathy in Diabetic Rats. Am. J. Hypertens. 2013, 27, 586–595. [Google Scholar] [CrossRef]
- Liou, G.Y.; Storz, P. Reactive oxygen species in cancer. Free Radic. Res. 2010, 44, 479–496. [Google Scholar] [CrossRef]
- Moon, E.J.; Giaccia, A. Dual roles of NRF2 in tumor prevention and progression: Possible implications in cancer treatment. Free Radic. Biol. Med. 2015, 79, 292–299. [Google Scholar] [CrossRef] [PubMed]
- Mostafavi-Pour, Z.; Ramezani, F.; Keshavarzi, F.; Samadi, N. The role of quercetin and vitamin C in Nrf2-dependent oxidative stress production in breast cancer cells. Oncol. Lett. 2017, 13, 1965–1973. [Google Scholar] [CrossRef]
- Yim, S.H.; Clish, C.B.; Gladyshev, V.N. Selenium Deficiency Is Associated with Pro-longevity Mechanisms. Cell Rep. 2019, 27, 2785–2797. [Google Scholar] [CrossRef]
- Müller, M.; Banning, A.; Brigelius-Flohé, R.; Kipp, A. Nrf2 target genes are induced under marginal selenium-deficiency. Genes Nutr. 2010, 5, 297–307. [Google Scholar] [CrossRef]
- Lin, T.; Tao, J.; Chen, Y.; Zhang, Y.; Li, F.; Zhang, Y.; Han, X.; Zhao, Z.; Liu, G.; Li, H. Selenium Deficiency Leads to Changes in Renal Fibrosis Marker Proteins and Wnt/β-Catenin Signaling Pathway Components. Biol. Trace Elem. Res. 2022, 200, 1127–1139. [Google Scholar] [CrossRef]
- Ng, L.F.; Kaur, P.; Bunnag, N.; Suresh, J.; Sung, I.C.H.; Tan, Q.H.; Gruber, J.; Tolwinski, N.S. WNT Signaling in Disease. Cells 2019, 8, 826. [Google Scholar] [CrossRef]
- Kang, G.G.; Francis, N.; Hill, R.; Waters, D.; Blanchard, C.; Santhakumar, A.B. Dietary Polyphenols and Gene Expression in Molecular Pathways Associated with Type 2 Diabetes Mellitus: A Review. Int. J. Mol. Sci. 2019, 21, 140. [Google Scholar] [CrossRef] [PubMed]
- Ding, S.; Jiang, H.; Fang, J. Regulation of Immune Function by Polyphenols. J. Immunol. Res. 2018, 2018, 1264074. [Google Scholar] [CrossRef] [PubMed]
- Chen, Z.; Hao, W.; Gao, C.; Zhou, Y.; Zhang, C.; Zhang, J.; Wang, R.; Wang, Y.; Wang, S. A polyphenol-assisted IL-10 mRNA delivery system for ulcerative colitis. Acta Pharm. Sin. B 2022, 12, 3367–3382. [Google Scholar] [CrossRef] [PubMed]
- Borsoi, F.T.; Neri-Numa, I.A.; de Oliveira, W.Q.; de Araújo, F.F.; Pastore, G.M. Dietary polyphenols and their relationship to the modulation of non-communicable chronic diseases and epigenetic mechanisms: A mini-review. Food Chem. 2023, 6, 100155. [Google Scholar] [CrossRef] [PubMed]
- Montero Vega, M.T.; de Andrés Martín, A. Toll-like receptors: A family of innate sensors of danger that alert and drive immunity. Allergol. Immunopathol. 2008, 36, 347–357. [Google Scholar] [CrossRef] [PubMed]
- Jimi, E.; Huang, F.; Nakatomi, C. NF-κB Signaling Regulates Physiological and Pathological Chondrogenesis. Int. J. Mol. Sci. 2019, 20, 6275. [Google Scholar] [CrossRef]
- Pramanik, K.C.; Makena, M.R.; Bhowmick, K.; Pandey, M.K. Advancement of NF-κB Signaling Pathway: A Novel Target in Pancreatic Cancer. Int. J. Mol. Sci. 2018, 19, 3890. [Google Scholar] [CrossRef]
- Hayden, M.S.; Ghosh, S. NF-κB, the first quarter-century: Remarkable progress and outstanding questions. Genes Dev. 2012, 26, 203–234. [Google Scholar] [CrossRef]
- Bhatt, D.; Ghosh, S. Regulation of the NF-κB-Mediated Transcription of Inflammatory Genes. Front. Immunol. 2014, 5, 71. [Google Scholar] [CrossRef]
- Liu, T.; Zhang, L.; Joo, D.; Sun, S.C. NF-κB signaling in inflammation. Signal Transduct. Target. Ther. 2017, 2, 17023. [Google Scholar] [CrossRef]
- Verzella, D.; Pescatore, A.; Capece, D.; Vecchiotti, D.; Ursini, M.V.; Franzoso, G.; Alesse, E.; Zazzeroni, F. Life, death, and autophagy in cancer: NF-κB turns up everywhere. Cell Death Dis. 2020, 11, 210. [Google Scholar] [CrossRef] [PubMed]
- Notarbartolo, M.; Cervello, M.; Dusonchet, L.; Cusimano, A.; D’Alessandro, N. Resistance to diverse apoptotic triggers in multidrug resistant HL60 cells and its possible relationship to the expression of P-glycoprotein, Fas and of the novel anti-apoptosis factors IAP (inhibitory of apoptosis proteins). Cancer Lett. 2002, 180, 91–101. [Google Scholar] [CrossRef] [PubMed]
- Yahfoufi, N.; Alsadi, N.; Jambi, M.; Matar, C. The Immunomodulatory and Anti-Inflammatory Role of Polyphenols. Nutrients 2018, 10, 1618. [Google Scholar] [CrossRef] [PubMed]
- Endale, M.; Park, S.C.; Kim, S.; Kim, S.H.; Yang, Y.; Cho, J.Y.; Rhee, M.H. Quercetin disrupts tyrosine-phosphorylated phosphatidylinositol 3-kinase and myeloid differentiation factor-88 association, and inhibits MAPK/AP-1 and IKK/NF-κB-induced inflammatory mediators production in RAW 264.7 cells. Immunobiology 2013, 218, 1452–1467. [Google Scholar] [CrossRef]
- Vauzour, D. Dietary polyphenols as modulators of brain functions: Biological actions and molecular mechanisms underpinning their beneficial effects. Oxidative Med. Cell Longev. 2012, 2012, 914273. [Google Scholar] [CrossRef] [PubMed]
- Da Cunha, L.R.; Muniz-Junqueira, M.I.; Dos Santos Borges, T.K. Impact of polyphenols in phagocyte functions. J. Inflamm. Res. 2019, 12, 205–217. [Google Scholar] [CrossRef] [PubMed]
- Azam, S.; Jakaria, M.; Kim, I.S.; Kim, J.; Haque, M.E.; Choi, D.K. Regulation of Toll-Like Receptor (TLR) Signaling Pathway by Polyphenols in the Treatment of Age-Linked Neurodegenerative Diseases: Focus on TLR4 Signaling. Front. Immunol. 2019, 10, 1000. [Google Scholar] [CrossRef]
- Karunaweera, N.; Raju, R.; Gyengesi, E.; Münch, G. Plant polyphenols as inhibitors of NF-κB induced cytokine production-a potential anti-inflammatory treatment for Alzheimer’s disease? Front. Mol. Neurosci. 2015, 8, 24. [Google Scholar] [CrossRef] [PubMed]
- Cárcamo, J.M.; Pedraza, A.; Bórquez-Ojeda, O.; Golde, D.W. Vitamin C suppresses TNF alpha-induced NF kappa B activation by inhibiting I kappa B alpha phosphorylation. Biochemistry 2002, 41, 12995–13002. [Google Scholar] [CrossRef] [PubMed]
- Austenaa, L.M.; Carlsen, H.; Ertesvag, A.; Alexander, G.; Blomhoff, H.K.; Blomhoff, R. Vitamin A status significantly alters nuclear factor-kappaB activity assessed by in vivo imaging. FASEB J. 2004, 18, 1255–1257. [Google Scholar] [CrossRef] [PubMed]
- Glauert, H.P. Vitamin E and NF-kappaB activation: A review. Vitam. Horm. 2007, 76, 135–153. [Google Scholar] [CrossRef] [PubMed]
- Kretz-Remy, C.; Arrigo, A.P. Selenium: A key element that controls NF-kappa B activation and I kappa B alpha half life. Biofactors 2001, 14, 117–125. [Google Scholar] [CrossRef]
- Karin, M.; Liu, Z.; Zandi, E. AP-1 function and regulation. Curr. Opin. Cell Biol. 1997, 9, 240–246. [Google Scholar] [CrossRef] [PubMed]
- Kyriakis, J.M. Activation of the AP-1 transcription factor by inflammatory cytokines of the TNF family. Gene Expr. 1999, 7, 217–231. [Google Scholar]
- Atsaves, V.; Leventaki, V.; Rassidakis, G.Z.; Claret, F.X. AP-1 Transcription Factors as Regulators of Immune Responses in Cancer. Cancers 2019, 11, 1037. [Google Scholar] [CrossRef]
- Spencer, J.P. The interactions of flavonoids within neuronal signalling pathways. Genes. Nutr. 2007, 2, 257–273. [Google Scholar] [CrossRef]
- Meng, T.; Xiao, D.; Muhammed, A.; Deng, J.; Chen, L.; He, J. Anti-Inflammatory Action and Mechanisms of Resveratrol. Molecules 2021, 26, 229. [Google Scholar] [CrossRef] [PubMed]
- Mattmiller, S.A.; Carlson, B.A.; Sordillo, L.M. Regulation of inflammation by selenium and selenoproteins: Impact on eicosanoid biosynthesis. J. Nutr. Sci. 2013, 2, e28. [Google Scholar] [CrossRef]
- Mitchell, T.J.; John, S. Signal transducer and activator of transcription (STAT) signalling and T-cell lymphomas. Immunology 2005, 114, 301–312. [Google Scholar] [CrossRef] [PubMed]
- Mengie Ayele, T.; Tilahun Muche, Z.; Behaile Teklemariam, A.; Bogale Kassie, A.; Chekol Abebe, E. Role of JAK2/STAT3 Signaling Pathway in the Tumorigenesis, Chemotherapy Resistance, and Treatment of Solid Tumors: A Systemic Review. J. Inflamm. Res. 2022, 15, 1349–1364. [Google Scholar] [CrossRef] [PubMed]
- Yang, J.; Wang, L.; Guan, X.; Qin, J.J. Inhibiting STAT3 signaling pathway by natural products for cancer prevention and therapy: In vitro and in vivo activity and mechanisms of action. Pharmacol. Res. 2022, 182, 106357. [Google Scholar] [CrossRef]
- Silveira, A.C.; Dias, J.P.; Santos, V.M.; Oliveira, P.F.; Alves, M.G.; Rato, L.; Silva, B.M. The Action of Polyphenols in Diabetes Mellitus and Alzheimer’s Disease: A Common Agent for Overlapping Pathologies. Curr. Neuropharmacol. 2019, 17, 590–613. [Google Scholar] [CrossRef] [PubMed]
- Zhang, M.; Wei, J.; Pan, X.; Shan, H.; Yan, R.; Xue, J.; Zhu, Y.; Lin, L. Change of cardiac mitochondrial STAT3 activity in rats with selenium deficiency and its relation with myocardial injury. Nan Fang Yi Ke Da Xue Xue Bao 2013, 33, 967–971. [Google Scholar]
- Mafra, D.; Alvarenga, L.; Cardozo, L.; Stockler-Pinto, M.B.; Nakao, L.S.; Stenvinkel, P.; Shiels, P.G. Inhibiting BTB domain and CNC homolog 1 (Bach1) as an alternative to increase Nrf2 activation in chronic diseases. Biochim. Biophys. Acta Gen. Subj. 2022, 1866, 130129. [Google Scholar] [CrossRef]
- Ozono, R. New biotechnological methods to reduce oxidative stress in the cardiovascular system: Focusing on the Bach1/heme oxygenase-1 pathway. Curr. Pharm. Biotechnol. 2006, 7, 87–93. [Google Scholar] [CrossRef]
- Jiang, P.; Li, F.; Liu, Z.; Hao, S.; Gao, J.; Li, S. BTB and CNC homology 1 (Bach1) induces lung cancer stem cell phenotypes by stimulating CD44 expression. Respir. Res. 2021, 22, 320. [Google Scholar] [CrossRef]
- Arunachalam, A.; Lakshmanan, D.K.; Ravichandran, G.; Paul, S.; Manickam, S.; Kumar, P.V.; Thilagar, S. Regulatory mechanisms of heme regulatory protein BACH1: A potential therapeutic target for cancer. Med. Oncol. 2021, 38, 122. [Google Scholar] [CrossRef]
- Song, Q.; Mao, X.; Jing, M.; Fu, Y.; Yan, W. Pathophysiological role of BACH transcription factors in digestive system diseases. Front. Physiol. 2023, 14, 1121353. [Google Scholar] [CrossRef]
- Dhakshinamoorthy, S.; Jain, A.K.; Bloom, D.A.; Jaiswal, A.K. Bach1 competes with Nrf2 leading to negative regulation of the antioxidant response element (ARE)-mediated NAD(P)H:quinone oxidoreductase 1 gene expression and induction in response to antioxidants. J. Biol. Chem. 2005, 280, 16891–16900. [Google Scholar] [CrossRef]
- Ahuja, M.; Kaidery, N.A.; Dutta, D.; Attucks, O.C.; Kazakov, E.H.; Gazaryan, I.; Matsumoto, M.; Igarashi, K.; Sharma, S.M.; Thomas, B. Harnessing the Therapeutic Potential of the Nrf2/Bach1 Signaling Pathway in Parkinson’s Disease. Antioxidants 2022, 11, 1780. [Google Scholar] [CrossRef]
- Kasai, S.; Mimura, J.; Ozaki, T.; Itoh, K. Emerging Regulatory Role of Nrf2 in Iron, Heme, and Hemoglobin Metabolism in Physiology and Disease. Front. Vet. Sci. 2018, 5, 242. [Google Scholar] [CrossRef]
- Waxman, E.A. Bach2 is a potent repressor of Nrf2-mediated antioxidant enzyme expression in dopaminergic neurons. bioRxiv 2019. [Google Scholar] [CrossRef]
- Reichard, J.F.; Sartor, M.A.; Puga, A. BACH1 is a specific repressor of HMOX1 that is inactivated by arsenite. J. Biol. Chem. 2008, 283, 22363–22370. [Google Scholar] [CrossRef]
- Su, C.; Liu, Z.; Wang, Y.; Wang, Y.; Song, E.; Song, Y. The electrophilic character of quinones is essential for the suppression of Bach1. Toxicology 2017, 387, 17–26. [Google Scholar] [CrossRef]
- Wang, T.; Dong, Y.; Huang, Z.; Zhang, G.; Zhao, Y.; Yao, H.; Hu, J.; Tüksammel, E.; Cai, H.; Liang, N.; et al. Antioxidants stimulate BACH1-dependent tumor angiogenesis. J. Clin. Investig. 2023, 133, e169671. [Google Scholar] [CrossRef]
- Feng, Y.; Jin, C.; Lv, S.; Zhang, H.; Ren, F.; Wang, J. Molecular Mechanisms and Applications of Polyphenol-Protein Complexes with Antioxidant Properties: A Review. Antioxidants 2023, 12, 1577. [Google Scholar] [CrossRef]
- Chio, I.I.C.; Tuveson, D.A. ROS in Cancer: The Burning Question. Trends Mol. Med. 2017, 23, 411–429. [Google Scholar] [CrossRef]
- Konaté, M.M.; Antony, S.; Doroshow, J.H. Inhibiting the Activity of NADPH Oxidase in Cancer. Antioxid. Redox Signal. 2020, 33, 435–454. [Google Scholar] [CrossRef]
- Gong, S.; Wang, S.; Shao, M. NADPH Oxidase 4: A Potential Therapeutic Target of Malignancy. Front. Cell Dev. Biol. 2022, 10, 884412. [Google Scholar] [CrossRef]
- Jang, J.Y.; Min, J.H.; Wang, S.B.; Chae, Y.H.; Baek, J.Y.; Kim, M.; Ryu, J.S.; Chang, T.S. Resveratrol inhibits collagen-induced platelet stimulation through suppressing NADPH oxidase and oxidative inactivation of SH2 domain-containing protein tyrosine phosphatase-2. Free Radic. Biol. Med. 2015, 89, 842–851. [Google Scholar] [CrossRef]
- Sul, O.-J.; Ra, S.W. Quercetin Prevents LPS-Induced Oxidative Stress and Inflammation by Modulating NOX2/ROS/NF-kB in Lung Epithelial Cells. Molecules 2021, 26, 6949. [Google Scholar] [CrossRef]
- Zhu, W.; Oteiza, P.I. NADPH oxidase 1: A target in the capacity of dimeric ECG and EGCG procyanidins to inhibit colorectal cancer cell invasion. Redox Biol. 2023, 65, 102827. [Google Scholar] [CrossRef]
- Fan, Z.; Duan, X.; Cai, H.; Wang, L.; Li, M.; Qu, J.; Li, W.; Wang, Y.; Wang, J. Curcumin inhibits the invasion of lung cancer cells by modulating the PKCα/Nox-2/ROS/ATF-2/MMP-9 signaling pathway. Oncol. Rep. 2015, 34, 691–698. [Google Scholar] [CrossRef]
- Jaquet, V.; Marcoux, J.; Forest, E.; Leidal, K.G.; McCormick, S.; Westermaier, Y.; Perozzo, R.; Plastre, O.; Fioraso-Cartier, L.; Diebold, B.; et al. NADPH oxidase (NOX) isoforms are inhibited by celastrol with a dual mode of action. Br. J. Pharmacol. 2011, 164, 507–520. [Google Scholar] [CrossRef]
- Serino, A.; Salazar, G. Protective Role of Polyphenols against Vascular Inflammation, Aging and Cardiovascular Disease. Nutrients 2018, 11, 53. [Google Scholar] [CrossRef]
- Jin, K.; Qian, C.; Lin, J.; Liu, B. Cyclooxygenase-2-Prostaglandin E2 pathway: A key player in tumor-associated immune cells. Front. Oncol. 2023, 13, 1099811. [Google Scholar] [CrossRef]
- Tyagi, A.; Kamal, M.A.; Poddar, N.K. Integrated Pathways of COX-2 and mTOR: Roles in Cell Sensing and Alzheimer’s Disease. Front. Neurosci. 2020, 14, 693. [Google Scholar] [CrossRef] [PubMed]
- Chao, C.L.; Weng, C.S.; Chang, N.C.; Lin, J.S.; Kao, S.T.; Ho, F.M. Naringenin more effectively inhibits inducible nitric oxide synthase and cyclooxygenase-2 expression in macrophages than in microglia. Nutr. Res. 2010, 30, 858–864. [Google Scholar] [CrossRef]
- Ferruelo, A.; de Las Heras, M.M.; Redondo, C.; Ramón de Fata, F.; Romero, I.; Angulo, J.C. Wine polyphenols exert antineoplasic effect on androgen resistant PC-3 cell line through the inhibition of the transcriptional activity of COX-2 promoter mediated by NF-kβ. Actas Urol. Esp. 2014, 38, 429–437. [Google Scholar] [CrossRef] [PubMed]
- Rodríguez, C.; Martínez-González, J.; Raposo, B.; Alcudia, J.F.; Guadall, A.; Badimon, L. Regulation of lysyl oxidase in vascular cells: Lysyl oxidase as a new player in cardiovascular diseases. Cardiovasc. Res. 2008, 79, 7–13. [Google Scholar] [CrossRef] [PubMed]
- Raposo, B.; Rodríguez, C.; Martínez-González, J.; Badimon, L. High levels of homocysteine inhibit lysyl oxidase (LOX) and downregulate LOX expression in vascular endothelial cells. Atherosclerosis 2004, 177, 1–8. [Google Scholar] [CrossRef] [PubMed]
- Wang, H.; Poe, A.; Pak, L.; Nandakumar, K.; Jandu, S.; Steppan, J.; Löser, R.; Santhanam, L. An in situ activity assay for lysyl oxidases. Commun. Biol. 2021, 4, 840. [Google Scholar] [CrossRef] [PubMed]
- Añazco, C.; Riedelsberger, J.; Vega-Montoto, L.; Rojas, A. Exploring the Interplay between Polyphenols and Lysyl Oxidase Enzymes for Maintaining Extracellular Matrix Homeostasis. Int. J. Mol. Sci. 2023, 24, 10985. [Google Scholar] [CrossRef]
- Singh, P.; Arif, Y.; Miszczuk, E.; Bajguz, A.; Hayat, S. Specific Roles of Lipoxygenases in Development and Responses to Stress in Plants. Plants 2022, 11, 979. [Google Scholar] [CrossRef]
- Chrisnasari, R.; Hennebelle, M.; Vincken, J.P.; van Berkel, W.J.H.; Ewing, T.A. Bacterial lipoxygenases: Biochemical characteristics, molecular structure and potential applications. Biotechnol. Adv. 2022, 61, 108046. [Google Scholar] [CrossRef]
- Kuhn, H.; Banthiya, S.; van Leyen, K. Mammalian lipoxygenases and their biological relevance. Biochim. Biophys. Acta 2015, 1851, 308–330. [Google Scholar] [CrossRef]
- Hussain, T.; Tan, B.; Yin, Y.; Blachier, F.; Tossou, M.C.; Rahu, N. Oxidative Stress and Inflammation: What Polyphenols Can Do for Us? Oxidative Med. Cell Longev. 2016, 2016, 7432797. [Google Scholar] [CrossRef]
- Battelli, M.G.; Polito, L.; Bortolotti, M.; Bolognesi, A. Xanthine Oxidoreductase in Drug Metabolism: Beyond a Role as a Detoxifying Enzyme. Curr. Med. Chem. 2016, 23, 4027–4036. [Google Scholar] [CrossRef] [PubMed]
- Spanou, C.; Veskoukis, A.S.; Kerasioti, T.; Kontou, M.; Angelis, A.; Aligiannis, N.; Skaltsounis, A.L.; Kouretas, D. Flavonoid glycosides isolated from unique legume plant extracts as novel inhibitors of xanthine oxidase. PLoS ONE 2012, 7, e32214. [Google Scholar] [CrossRef]
- Bhattacharyya, A.; Chattopadhyay, R.; Mitra, S.; Crowe, S.E. Oxidative stress: An essential factor in the pathogenesis of gastrointestinal mucosal diseases. Physiol. Rev. 2014, 94, 329–354. [Google Scholar] [CrossRef] [PubMed]
- Dwibedi, V.; Jain, S.; Singhal, D.; Mittal, A.; Rath, S.K.; Saxena, S. Inhibitory activities of grape bioactive compounds against enzymes linked with human diseases. Appl. Microbiol. Biotechnol. 2022, 106, 1399–1417. [Google Scholar] [CrossRef] [PubMed]
- Gómez-Benito, M.; Granado, N.; García-Sanz, P.; Michel, A.; Dumoulin, M.; Moratalla, R. Modeling Parkinson’s Disease with the Alpha-Synuclein Protein. Front. Pharmacol. 2020, 11, 356. [Google Scholar] [CrossRef] [PubMed]
- Meade, R.M.; Fairlie, D.P.; Mason, J.M. Alpha-synuclein structure and Parkinson’s disease—Lessons and emerging principles. Mol. Neurodegener. 2019, 14, 29. [Google Scholar] [CrossRef] [PubMed]
- Ono, K.; Tsuji, M.; Yamasaki, T.R.; Pasinetti, G.M. Anti-aggregation Effects of Phenolic Compounds on α-synuclein. Molecules 2020, 25, 2444. [Google Scholar] [CrossRef]
- Sudhesh Dev, S.; Zainal Abidin, S.A.; Farghadani, R.; Othman, I.; Naidu, R. Receptor Tyrosine Kinases and Their Signaling Pathways as Therapeutic Targets of Curcumin in Cancer. Front. Pharmacol. 2021, 12, 772510. [Google Scholar] [CrossRef]
- Maiuolo, J.; Gliozzi, M.; Carresi, C.; Musolino, V.; Oppedisano, F.; Scarano, F.; Nucera, S.; Scicchitano, M.; Bosco, F.; Macri, R.; et al. Nutraceuticals and Cancer: Potential for Natural Polyphenols. Nutrients 2021, 13, 3834. [Google Scholar] [CrossRef]
- Paul, D.; Mahanta, S.; Tag, H.; Das, S.K.; Das Gupta, D.; Tanti, B.; Ananthan, R.; Das, R.; Jambhulkar, S.; Hui, P.K. Identification of tyrosine kinase inhibitors from Panax bipinnatifidus and Panax pseudoginseng for RTK-HER2 and VEGFR2 receptors, by in silico approach. Mol. Divers. 2022, 26, 1933–1955. [Google Scholar] [CrossRef] [PubMed]
- Seto, E.; Yoshida, M. Erasers of histone acetylation: The histone deacetylase enzymes. Cold Spring Harb. Perspect. Biol. 2014, 6, a018713. [Google Scholar] [CrossRef] [PubMed]
- Bannister, A.J.; Kouzarides, T. Regulation of chromatin by histone modifications. Cell Res. 2011, 21, 381–395. [Google Scholar] [CrossRef]
- Li, Y.; Seto, E. HDACs and HDAC Inhibitors in Cancer Development and Therapy. Cold Spring Harb. Perspect. Med. 2016, 6, a026831. [Google Scholar] [CrossRef] [PubMed]
- Eckschlager, T.; Plch, J.; Stiborova, M.; Hrabeta, J. Histone Deacetylase Inhibitors as Anticancer Drugs. Int. J. Mol. Sci. 2017, 18, 1414. [Google Scholar] [CrossRef]
- Rizwan, H.; Pal, S.; Sabnam, S.; Pal, A. High glucose augments ROS generation regulates mitochondrial dysfunction and apoptosis via stress signalling cascades in keratinocytes. Life Sci. 2020, 241, 117148. [Google Scholar] [CrossRef]
- Choi, S.Y.; Piao, Z.H.; Jin, L.; Kim, J.H.; Kim, G.R.; Ryu, Y.; Lin, M.Q.; Kim, H.S.; Kee, H.J.; Jeong, M.H. Piceatannol Attenuates Renal Fibrosis Induced by Unilateral Ureteral Obstruction via Downregulation of Histone Deacetylase 4/5 or p38-MAPK Signaling. PLoS ONE 2016, 11, e0167340. [Google Scholar] [CrossRef]
- McCrory, M.A.; Hamaker, B.R.; Lovejoy, J.C.; Eichelsdoerfer, P.E. Pulse consumption, satiety, and weight management. Adv. Nutr. 2010, 1, 17–30. [Google Scholar] [CrossRef]
- Gong, L.; Feng, D.; Wang, T.; Ren, Y.; Liu, Y.; Wang, J. Inhibitors of α-amylase and α-glucosidase: Potential linkage for whole cereal foods on prevention of hyperglycemia. Food Sci. Nutr. 2020, 8, 6320–6337. [Google Scholar] [CrossRef]
- Proença, C.; Ribeiro, D.; Freitas, M.; Fernandes, E. Flavonoids as potential agents in the management of type 2 diabetes through the modulation of α-amylase and α-glucosidase activity: A review. Crit. Rev. Food Sci. Nutr. 2022, 62, 3137–3207. [Google Scholar] [CrossRef]
- Xiao, J.B.; Högger, P. Dietary polyphenols and type 2 diabetes: Current insights and future perspectives. Curr. Med. Chem. 2015, 22, 23–38. [Google Scholar] [CrossRef]
- Lo Piparo, E.; Scheib, H.; Frei, N.; Williamson, G.; Grigorov, M.; Chou, C.J. Flavonoids for controlling starch digestion: Structural requirements for inhibiting human alpha-amylase. J. Med. Chem. 2008, 51, 3555–3561. [Google Scholar] [CrossRef]
- Żyżelewicz, D.; Oracz, J. Bioavailability and Bioactivity of Plant Antioxidants. Antioxidants. 2022, 11, 2336. [Google Scholar] [CrossRef]
- Rahman, M.; Akter, R.; Bhattacharya, T.; Abdel-Daim, M.M.; Alkahtani, S.; Arafah, M.W.; Al-Johani, N.S.; Alhoshani, N.M.; Alkeraishan, N.; Alhenaky, A. Resveratrol and neuroprotection: Impact and its therapeutic potential in Alzheimer’s disease. Front. Pharmacol. 2020, 11, 619024. [Google Scholar] [CrossRef] [PubMed]
- Lykkesfeldt, J.; Tveden-Nyborg, P. The Pharmacokinetics of Vitamin C. Nutrients 2019, 11, 2412. [Google Scholar] [CrossRef] [PubMed]
- Institute of Medicine (US) Panel on Dietary Antioxidants and Related Compounds. Dietary Reference Intakes for Vitamin C, Vitamin E, Selenium, and Carotenoids; National Academies Press (US): Washington, DC, USA, 2000. [Google Scholar]
- Feitosa, B.F.; de Alcântara, C.M.; de Lima, A.B.S.; Silva, A.S.; Araújo, A.D.S.; Cavalcanti, M.T.; Mori, E.; Araújo, I.M.; de Farias, P.A.M.; Wilairatana, P.; et al. Bioactive Natural Products for Chemical Control of Microorganisms: Scientific Prospecting (2001–2021) and Systematic Review. Molecules 2022, 27, 5917. [Google Scholar] [CrossRef] [PubMed]
- Yap, S.P.; Yuen, K.H. Influence of lipolysis and droplet size on tocotrienol absorption from self-emulsifying formulations. Int. J. Pharm. 2004, 281, 67–78. [Google Scholar] [CrossRef]
- Tso, P.; Fujimoto, K. The absorption and transport of lipids by the small intestine. Brain Res. Bull. 1991, 27, 477–482. [Google Scholar] [CrossRef] [PubMed]
- Borel, P.; Pasquier, B.; Armand, M.; Tyssandier, V.; Grolier, P.; Alexandre-Gouabau, M.-C.; Andre, M.; Senft, M.; Peyrot, J.; Jaussan, V. Processing of vitamin A and E in the human gastrointestinal tract. Am. J. Physiol.-Gastrointest. Liver Physiol. 2001, 280, G95–G103. [Google Scholar] [CrossRef]
- Borel, P.; Preveraud, D.; Desmarchelier, C. Bioavailability of vitamin E in humans: An update. Nutr. Rev. 2013, 71, 319–331. [Google Scholar] [CrossRef] [PubMed]
- Habauzit, V.; Morand, C. Evidence for a protective effect of polyphenols-containing foods on cardiovascular health: An update for clinicians. Ther. Adv. Chronic Dis. 2012, 3, 87–106. [Google Scholar] [CrossRef]
- Hu, M. Commentary: Bioavailability of flavonoids and polyphenols: Call to arms. Mol. Pharm. 2007, 4, 803–806. [Google Scholar] [CrossRef]
- Cicerale, S.; Lucas, L.; Keast, R. Biological activities of phenolic compounds present in virgin olive oil. Int. J. Mol. Sci. 2010, 11, 458–479. [Google Scholar] [CrossRef]
- Manach, C.; Williamson, G.; Morand, C.; Scalbert, A.; Rémésy, C. Bioavailability and bioefficacy of polyphenols in humans. I. Review of 97 bioavailability studies. Am. J. Clin. Nutr. 2005, 81, 230S–242S. [Google Scholar] [CrossRef]
- Xu, Y.; Le Sayec, M.; Roberts, C.; Hein, S.; Rodriguez-Mateos, A.; Gibson, R. Dietary Assessment Methods to Estimate (Poly)phenol Intake in Epidemiological Studies: A Systematic Review. Adv. Nutr. 2021, 12, 1781–1801. [Google Scholar] [CrossRef]
- Ros, E.; Martínez-González, M.A.; Estruch, R.; Salas-Salvadó, J.; Fitó, M.; Martínez, J.A.; Corella, D. Mediterranean diet and cardiovascular health: Teachings of the PREDIMED study. Adv. Nutr. 2014, 5, 330S–336S. [Google Scholar] [CrossRef]
- Di Lorenzo, C.; Colombo, F.; Biella, S.; Stockley, C.; Restani, P. Polyphenols and human health: The role of bioavailability. Nutrients 2021, 13, 273. [Google Scholar] [CrossRef]
- Del Bo’, C.; Bernardi, S.; Marino, M.; Porrini, M.; Tucci, M.; Guglielmetti, S.; Cherubini, A.; Carrieri, B.; Kirkup, B.; Kroon, P. Systematic review on polyphenol intake and health outcomes: Is there sufficient evidence to define a health-promoting polyphenol-rich dietary pattern? Nutrients 2019, 11, 1355. [Google Scholar] [PubMed]
- Flieger, J.; Flieger, W.; Baj, J.; Maciejewski, R. Antioxidants: Classification, natural sources, activity/capacity measurements, and usefulness for the synthesis of nanoparticles. Materials 2021, 14, 4135. [Google Scholar] [CrossRef] [PubMed]
- Schürks, M.; Glynn, R.J.; Rist, P.M.; Tzourio, C.; Kurth, T. Effects of vitamin E on stroke subtypes: Meta-analysis of randomised controlled trials. BMJ 2010, 341, c5702. [Google Scholar] [CrossRef] [PubMed]
- Klein, E.A.; Thompson, I.M.; Tangen, C.M.; Crowley, J.J.; Lucia, M.S.; Goodman, P.J.; Minasian, L.M.; Ford, L.G.; Parnes, H.L.; Gaziano, J.M.; et al. Vitamin E and the Risk of Prostate Cancer: The Selenium and Vitamin E Cancer Prevention Trial (SELECT). JAMA 2011, 306, 1549–1556. [Google Scholar] [CrossRef] [PubMed]
- Ye, G.; Metreveli, N.S.; Donthi, R.V.; Xia, S.; Xu, M.; Carlson, E.C.; Epstein, P.N. Catalase protects cardiomyocyte function in models of type 1 and type 2 diabetes. Diabetes 2004, 53, 1336–1343. [Google Scholar] [CrossRef] [PubMed]
- Ray, P.D.; Huang, B.-W.; Tsuji, Y. Reactive oxygen species (ROS) homeostasis and redox regulation in cellular signaling. Cell Signal. 2012, 24, 981–990. [Google Scholar] [CrossRef]
- Amorim, J.A.; Coppotelli, G.; Rolo, A.P.; Palmeira, C.M.; Ross, J.M.; Sinclair, D.A. Mitochondrial and metabolic dysfunction in ageing and age-related diseases. Nat. Rev. Endocrinol. 2022, 18, 243–258. [Google Scholar] [CrossRef]
- Kaźmierczak-Barańska, J.; Boguszewska, K.; Adamus-Grabicka, A.; Karwowski, B.T. Two Faces of Vitamin C-Antioxidative and Pro-Oxidative Agent. Nutrients 2020, 12, 1501. [Google Scholar] [CrossRef] [PubMed]
- Sotler, R.; Poljšak, B.; Dahmane, R.; Jukić, T.; Pavan Jukić, D.; Rotim, C.; Trebše, P.; Starc, A. Prooxidant Activities of Antioxidants and Their Impact on Health. Acta Clin. Croat. 2019, 58, 726–736. [Google Scholar] [CrossRef]
- Pauling, L. Vitamin C and common cold. JAMA 1971, 216, 332. [Google Scholar] [CrossRef]
- Bast, A.; Haenen, G.R.; Doelman, C.J. Oxidants and antioxidants: State of the art. Am. J. Med. 1991, 91, S2–S13. [Google Scholar] [CrossRef]
- Mishra, S.; Stierman, B.; Gahche, J.J.; Potischman, N. Dietary Supplement Use among Adults: United States, 2017–2018; NCHS Data Brief; NCHS: Hyattsville, MD, USA, 2021; pp. 1–8. [Google Scholar]
- GeríŸ, J.; Köpcke, W. The questionable association of vitamin E supplementation and mortality-inconsistent results of different meta-analytic approaches. Cell. Mol. Biol. 2009, 55, 1111–1120. [Google Scholar]
- Pérez Trueba, G. Los flavonoides: Antioxidantes o prooxidantes. Rev. Cuba. Investig. Bioméd. 2003, 22, 48–57. [Google Scholar]
- Speisky, H.; Shahidi, F.; Costa de Camargo, A.; Fuentes, J. Revisiting the Oxidation of Flavonoids: Loss, Conservation or Enhancement of Their Antioxidant Properties. Antioxidants 2022, 11, 133. [Google Scholar] [CrossRef]
- Skibola, C.F.; Smith, M.T. Potential health impacts of excessive flavonoid intake. Free Radic. Biol. Med. 2000, 29, 375–383. [Google Scholar] [CrossRef] [PubMed]
- Rahal, A.; Kumar, A.; Singh, V.; Yadav, B.; Tiwari, R.; Chakraborty, S.; Dhama, K. Oxidative Stress, Prooxidants, and Antioxidants: The Interplay. BioMed Res. Int. 2014, 2014, 761264. [Google Scholar] [CrossRef]
- Da Silva, J.; Herrmann, S.; Heuser, V.; Peres, W.; Marroni, N.P.; González-Gallego, J.; Erdtmann, B. Evaluation of the genotoxic effect of rutin and quercetin by comet assay and micronucleus test. Food Chem. Toxicol. 2002, 40, 941–947. [Google Scholar] [CrossRef]
- Zeraik, M.L.; Petrônio, M.S.; Coelho, D.; Regasini, L.O.; Silva, D.H.; da Fonseca, L.M.; Machado, S.A.; Bolzani, V.S.; Ximenes, V.F. Improvement of pro-oxidant capacity of protocatechuic acid by esterification. PLoS ONE 2014, 9, e110277. [Google Scholar] [CrossRef] [PubMed]
- Fordyce, F. Selenium geochemistry and health. Ambio 2007, 36, 94–97. [Google Scholar] [CrossRef]
- Petrović, M. Selenium: Widespread yet scarce, essential yet toxic. ChemTexts 2021, 7, 11. [Google Scholar] [CrossRef]
- Hartikainen, H.; Xue, T.; Piironen, V. Selenium as an anti-oxidant and pro-oxidant in ryegrass. Plant Soil 2000, 225, 193–200. [Google Scholar] [CrossRef]
- Hernández-Díaz, J.A.; Garza-García, J.J.; León-Morales, J.M.; Zamudio-Ojeda, A.; Arratia-Quijada, J.; Velázquez-Juárez, G.; López-Velázquez, J.C.; García-Morales, S. Antibacterial Activity of Biosynthesized Selenium Nanoparticles Using Extracts of Calendula officinalis against Potentially Clinical Bacterial Strains. Molecules 2021, 26, 5929. [Google Scholar] [CrossRef]
- Lee, K.H.; Jeong, D. Bimodal actions of selenium essential for antioxidant and toxic pro-oxidant activities: The selenium paradox (Review). Mol. Med. Rep. 2012, 5, 299–304. [Google Scholar] [CrossRef]
- Garbo, S.; Di Giacomo, S.; Łażewska, D.; Honkisz-Orzechowska, E.; Di Sotto, A.; Fioravanti, R.; Zwergel, C.; Battistelli, C. Selenium-Containing Agents Acting on Cancer-A New Hope? Pharmaceutics 2022, 15, 104. [Google Scholar] [CrossRef]
- Ehudin, M.A.; Golla, U.; Trivedi, D.; Potlakayala, S.D.; Rudrabhatla, S.V.; Desai, D.; Dovat, S.; Claxton, D.; Sharma, A. Therapeutic Benefits of Selenium in Hematological Malignancies. Int. J. Mol. Sci. 2022, 23, 7972. [Google Scholar] [CrossRef]
- Sanmartín, C.; Plano, D.; Sharma, A.K.; Palop, J.A. Selenium compounds, apoptosis and other types of cell death: An overview for cancer therapy. Int. J. Mol. Sci. 2012, 13, 9649–9672. [Google Scholar] [CrossRef]
- Debnath, S.; Agarwal, A.; Kumar, N.R.; Bedi, A. Selenium-Based Drug Development for Antioxidant and Anticancer Activity. Future Pharmacol. 2022, 2, 595–607. [Google Scholar] [CrossRef]
- Brozmanová, J.; Mániková, D.; Vlčková, V.; Chovanec, M. Selenium: A double-edged sword for defense and offence in cancer. Arch. Toxicol. 2010, 84, 919–938. [Google Scholar] [CrossRef] [PubMed]
- Radomska, D.; Czarnomysy, R.; Radomski, D.; Bielawski, K. Selenium Compounds as Novel Potential Anticancer Agents. Int. J. Mol. Sci. 2021, 22, 1009. [Google Scholar] [CrossRef] [PubMed]
- Ali, W.; Chen, Y.; Gandahi, J.A.; Qazi, I.H.; Sun, J.; Wang, T.; Liu, Z.; Zou, H. Cross-Talk Between Selenium Nanoparticles and Cancer Treatment Through Autophagy. Biol. Trace Elem. Res. 2023. [Google Scholar] [CrossRef] [PubMed]
- Zambonino, M.C.; Quizhpe, E.M.; Mouheb, L.; Rahman, A.; Agathos, S.N.; Dahoumane, S.A. Biogenic Selenium Nanoparticles in Biomedical Sciences: Properties, Current Trends, Novel Opportunities and Emerging Challenges in Theranostic Nanomedicine. Nanomaterials 2023, 13, 424. [Google Scholar] [CrossRef]
Structure | R1 | R2 | R3 | Name |
---|---|---|---|---|
CH3 | CH3 | CH3 | α- | |
CH3 | H | CH3 | β- | |
H | CH3 | CH3 | γ- | |
H | H | CH3 | δ- |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Andrés, C.M.C.; Pérez de la Lastra, J.M.; Juan, C.A.; Plou, F.J.; Pérez-Lebeña, E. Antioxidant Metabolism Pathways in Vitamins, Polyphenols, and Selenium: Parallels and Divergences. Int. J. Mol. Sci. 2024, 25, 2600. https://doi.org/10.3390/ijms25052600
Andrés CMC, Pérez de la Lastra JM, Juan CA, Plou FJ, Pérez-Lebeña E. Antioxidant Metabolism Pathways in Vitamins, Polyphenols, and Selenium: Parallels and Divergences. International Journal of Molecular Sciences. 2024; 25(5):2600. https://doi.org/10.3390/ijms25052600
Chicago/Turabian StyleAndrés, Celia María Curieses, José Manuel Pérez de la Lastra, Celia Andrés Juan, Francisco J. Plou, and Eduardo Pérez-Lebeña. 2024. "Antioxidant Metabolism Pathways in Vitamins, Polyphenols, and Selenium: Parallels and Divergences" International Journal of Molecular Sciences 25, no. 5: 2600. https://doi.org/10.3390/ijms25052600
APA StyleAndrés, C. M. C., Pérez de la Lastra, J. M., Juan, C. A., Plou, F. J., & Pérez-Lebeña, E. (2024). Antioxidant Metabolism Pathways in Vitamins, Polyphenols, and Selenium: Parallels and Divergences. International Journal of Molecular Sciences, 25(5), 2600. https://doi.org/10.3390/ijms25052600