Natural Compounds and Products from an Anti-Aging Perspective
Abstract
:1. Introduction
2. Vitamins
2.1. Vitamin C
2.2. Vitamin A
2.3. Vitamin E
3. Polyunsaturated Fatty Acids (PUFAs)
3.1. Omega-3 PUFA
3.2. Omega-6 PUFA
4. Trace Elements and Micronutrients
4.1. Zinc
4.2. Copper
4.3. Selenium
4.4. Role of Zn, Cu and Se in the Aging Prevention
5. Carnitine
6. Plant Metabolites
6.1. Polyphenols
6.1.1. Resveratrol
6.1.2. Curcumin
6.2. Terpenoids
6.3. Other Natural Compounds from Medicinal Plants
6.4. Alpha-Hydroxy Acids (AHAs)
6.5. Phytoestrogens
6.6. Cocoa Derivatives
6.7. Humic Substances
7. Marine-Derived Compounds
8. Honeybee Products
9. Mushrooms
10. Probiotics
11. Drinking Water
12. Natural Substances Dangerous to the Human Body
13. Concluding Remarks
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Conflicts of Interest
References
- Liu, J.K. Antiaging agents: Safe interventions to slow aging and healthy life span extension. Nat. Prod. Bioprospect. 2022, 12, 18. [Google Scholar] [CrossRef] [PubMed]
- Cao, X.; Li, W.; Wang, T.; Ran, D.; Davalos, V.; Planas-Serra, L.; Pujol, A.; Esteller, M.; Wang, X.; Yu, H. Accelerated biological aging in COVID-19 patients. Nat. Commun. 2022, 13, 2135. [Google Scholar] [CrossRef] [PubMed]
- Asgary, S.; Rastqar, A.; Keshvari, M. Functional food and cardiovascular disease prevention and treatment: A review. J. Am. Coll. Nutr. 2018, 37, 429–455. [Google Scholar] [CrossRef]
- Zia, A.; Farkhondeh, T.; Pourbagher-Shahri, A.M.; Samarghandian, S. The role of curcumin in aging and senescence: Molecular mechanisms. Biomed. Pharmacother. 2021, 134, 111119. [Google Scholar] [CrossRef] [PubMed]
- Rattan, S.I. Healthy ageing, but what is health? Biogerontology 2013, 14, 673–677. [Google Scholar] [CrossRef]
- Corrêa, R.C.; Peralta, R.M.; Haminiuk, C.W.; Maciel, G.M.; Bracht, A.; Ferreira, I.C. New phytochemicals as potential human anti-aging compounds: Reality, promise, and challenges. Crit. Rev. Food. Sci. Nutr. 2018, 58, 942–957. [Google Scholar] [CrossRef] [PubMed]
- Ding, A.-J.; Zheng, S.-Q.; Huang, X.-B.; Xing, T.-K.; Wu, G.-S.; Sun, H.-Y.; Qi, S.-H.; Luo, H.-R. Current perspective in the discovery of anti-aging agents from natural products. Nat. Prod. Bioprospect. 2017, 7, 335–404. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Tan, B.L.; Norhaizan, M.E. Carotenoids: How Effective Are They to Prevent Age-Related Diseases? Molecules 2019, 24, 1801. [Google Scholar] [CrossRef] [Green Version]
- Vranešić-Bender, D. The role of nutraceuticals in anti-aging medicine. Acta Clin. Croat. 2010, 49, 537–544. [Google Scholar]
- Wang, J.C.; Bennett, M. Aging and atherosclerosis: Mechanisms, functional consequences, and potential therapeutics for cellular senescence. Circ. Res. 2012, 111, 245–259. [Google Scholar] [CrossRef] [Green Version]
- Vaiserman, A.; Koliada, A.; Lushchak, O.; Castillo, M.J. Repurposing drugs to fight aging: The difficult path from bench to bedside. Med. Res. Rev. 2021, 41, 1676–1700. [Google Scholar] [CrossRef] [PubMed]
- Martin, R.E.; Postiglione, A.E.; Muday, G.K. Reactive oxygen species function as signaling molecules in controlling plant development and hormonal responses. Curr. Opin. Plant Biol. 2022, 69, 102293. [Google Scholar] [CrossRef] [PubMed]
- Shohag, S.; Akhter, S.; Islam, S.; Sarker, T.; Sifat, M.K.; Rahman, M.M.; Islam, M.R.; Sharma, R. Perspectives on the Molecular Mediators of Oxidative Stress and Antioxidant Strategies in the Context of Neuroprotection and Neurolongevity: An Extensive Review. Oxid. Med. Cell. Longev. 2022, 2022, 7743705. [Google Scholar] [CrossRef] [PubMed]
- Shayeghan, M.; Ansari, A.M.; Forouzesh, F.; Javidi, M.A. Reactive oxygen species, the trident of Neptune in the hands of hecate; role in different diseases, signaling pathways, and detection methods. Arch. Biochem. Biophys. 2022, 728, 109357. [Google Scholar] [CrossRef] [PubMed]
- Ho, Y.-S.; So, K.-F.; Chang, R.C.-C. Anti-aging herbal medicine—How and why can they be used in aging-associated neurodegenerative diseases? Ageing Res. Rev. 2010, 9, 354–362. [Google Scholar] [CrossRef]
- Prasad, S.; Gupta, S.C.; Tyagi, A.K. Reactive oxygen species (ROS) and cancer: Role of antioxidative nutraceuticals. Cancer Lett. 2017, 387, 95–105. [Google Scholar] [CrossRef]
- Bjørklund, G.; Dadar, M.; Martins, N.; Chirumbolo, S.; Goh, B.H.; Smetanina, K.; Lysiuk, R. Brief Challenges on Medicinal Plants: An Eye-Opening Look at Ageing-Related Disorders. Basic Clin. Pharmacol. Toxicol. 2018, 122, 539–558. [Google Scholar] [CrossRef] [Green Version]
- Bjørklund, G.; Dadar, M.; Chirumbolo, S.; Lysiuk, R. Flavonoids as detoxifying and pro-survival agents: What’s new? Food Chem. Toxicol. 2017, 110, 240–250. [Google Scholar] [CrossRef]
- Mehrandish, R.; Rahimian, A.; Shahriary, A. Heavy metals detoxification: A review of herbal compounds for chelation therapy in heavy metals toxicity. J. Herbmed. Pharmacol. 2019, 8, 69–77. [Google Scholar] [CrossRef] [Green Version]
- Ahmed, I.A.; Mikail, M.A.; Zamakshshari, N.; Abdullah, A.H. Natural anti-aging skincare: Role and potential. Biogerontology 2020, 21, 293–310. [Google Scholar] [CrossRef]
- Pullar, J.M.; Carr, A.C.; Vissers, M. The roles of vitamin C in skin health. Nutrients 2017, 9, 866. [Google Scholar] [CrossRef] [PubMed]
- Gašperlin, M.; Gosenca, M. Main approaches for delivering antioxidant vitamins through the skin to prevent skin ageing. Expert Opin. Drug Deliv. 2011, 8, 905–919. [Google Scholar] [CrossRef] [PubMed]
- Castiglione, D.; Platania, A.; Conti, A.; Falla, M.; D’Urso, M.; Marranzano, M. Dietary Micronutrient and Mineral Intake in the Mediterranean Healthy Eating, Ageing, and Lifestyle (MEAL) Study. Antioxidants 2018, 7, 79. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Lykkesfeldt, J. On the effect of vitamin C intake on human health: How to (mis)interprete the clinical evidence. Redox Biol. 2020, 34, 101532. [Google Scholar] [CrossRef]
- Brook, M.; Grimshaw, J. Vitamin C concentration of plasma and leukocytes as related to smoking habit, age, and sex of humans. Am. J. Clin. Nutr. 1968, 21, 1254–1258. [Google Scholar] [CrossRef] [PubMed]
- Delanghe, J.R.; Langlois, M.R.; De Buyzere, M.L.; Na, N.; Ouyang, J.; Speeckaert, M.M.; Torck, M.A. Vitamin C deficiency: More than just a nutritional disorder. Genes Nutr. 2011, 6, 341–346. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sharma, Y.; Miller, M.; Shahi, R.; Doyle, A.; Horwood, C.; Hakendorf, P.; Thompson, C. Vitamin C deficiency in Australian hospitalised patients: An observational study. Intern. Med. J. 2019, 49, 189–196. [Google Scholar] [CrossRef]
- Abdollahifar, M.-A.; Azad, N.; Sajadi, E.; Shams Mofarahe, Z.; Zare, F.; Moradi, A.; Rezaee, F.; Gholamin, M.; Abdi, S. Vitamin C restores ovarian follicular reservation in a mouse model of aging. Anat. Cell Biol. 2019, 52, 196–203. [Google Scholar] [CrossRef]
- Crisan, D.; Roman, I.; Crisan, M.; Scharffetter-Kochanek, K.; Badea, R. The role of vitamin C in pushing back the boundaries of skin aging: An ultrasonographic approach. Clin. Cosmet. Investig. Dermatol. 2015, 8, 463–470. [Google Scholar] [CrossRef] [Green Version]
- Alagl, A.S.; Bhat, S.G. Ascorbic acid: New role of an age-old micronutrient in the management of periodontal disease in older adults. Geriatr. Gerontol. Int. 2015, 15, 241–254. [Google Scholar] [CrossRef]
- Harrison, F.E. A critical review of vitamin C for the prevention of age-related cognitive decline and Alzheimer’s disease. J. Alzheimers Dis. 2012, 29, 711–726. [Google Scholar] [CrossRef] [PubMed]
- Shi, L.; Niedzwiecki, A.; Rath, M. Age and Dietary Vitamin C Intake Affect Brain Physiology in Genetically Modified Mice Expressing Human Lipoprotein(A) and Unable to Synthesize Vitamin C. Curr. Aging Sci. 2021, 14, 223–234. [Google Scholar] [CrossRef] [PubMed]
- Mumtaz, S.; Ali, S.; Tahir, H.M.; Kazmi, S.A.R.; Shakir, H.A.; Mughal, T.A.; Summer, M.; Farooq, M.A. Aging and its treatment with vitamin C: A comprehensive mechanistic review. Mol. Biol. Rep. 2021, 48, 8141–8153. [Google Scholar] [CrossRef] [PubMed]
- Kelly, M.E.; Ramkumar, S.; Sun, W.; Colon Ortiz, C.; Kiser, P.D.; Golczak, M.; von Lintig, J. The Biochemical Basis of Vitamin A Production from the Asymmetric Carotenoid beta-Cryptoxanthin. ACS Chem. Biol. 2018, 13, 2121–2129. [Google Scholar] [CrossRef]
- Mukherjee, S.; Date, A.; Patravale, V.; Korting, H.C.; Roeder, A.; Weindl, G. Retinoids in the treatment of skin aging: An overview of clinical efficacy and safety. Clin. Interv. Aging 2006, 1, 327–348. [Google Scholar] [CrossRef]
- Stratigos, A.J.; Katsambas, A.D. The role of topical retinoids in the treatment of photoaging. Drugs 2005, 65, 1061–1072. [Google Scholar] [CrossRef]
- Zasada, M.; Budzisz, E. Retinoids: Active molecules influencing skin structure formation in cosmetic and dermatological treatments. Postep. Dermatol. Alergol. 2019, 36, 392–397. [Google Scholar] [CrossRef] [Green Version]
- Kafi, R.; Kwak, H.S.; Schumacher, W.E.; Cho, S.; Hanft, V.N.; Hamilton, T.A.; King, A.L.; Neal, J.D.; Varani, J.; Fisher, G.J.; et al. Improvement of naturally aged skin with vitamin A (retinol). Arch. Dermatol. 2007, 143, 606–612. [Google Scholar] [CrossRef] [Green Version]
- Saari, J.C. Vitamin A and Vision. Subcell. Biochem. 2016, 81, 231–259. [Google Scholar] [CrossRef]
- Tucker-Samaras, S.; Zedayko, T.; Cole, C.; Miller, D.; Wallo, W.; Leyden, J.J. A stabilized 0.1% retinol facial moisturizer improves the appearance of photodamaged skin in an eight-week, double-blind, vehicle-controlled study. J. Drugs Dermatol. 2009, 8, 932–936. [Google Scholar]
- Margiana, R.; Pakpahan, C.; Pangestu, M. A systematic review of retinoic acid in the journey of spermatogonium to spermatozoa: From basic to clinical application. F1000Resarch 2022, 11, 552. [Google Scholar] [CrossRef] [PubMed]
- Michelazzo, F.B.; Oliveira, J.M.; Stefanello, J.; Luzia, L.A.; Rondo, P.H. The influence of vitamin A supplementation on iron status. Nutrients 2013, 5, 4399–4413. [Google Scholar] [CrossRef] [PubMed]
- Traber, M.G.; Atkinson, J. Vitamin E, antioxidant and nothing more. Free Radic. Biol. Med. 2007, 43, 4–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Brigelius-Flohe, R.; Davies, K.J. Is vitamin E an antioxidant, a regulator of signal transduction and gene expression, or a ‘junk’ food? Comments on the two accompanying papers: “Molecular mechanism of alpha-tocopherol action” by A. Azzi and “Vitamin E, antioxidant and nothing more” by M. Traber and J. Atkinson. Free Radic. Biol. Med. 2007, 43, 2–3. [Google Scholar] [CrossRef] [PubMed]
- Valentino, S.; Ghelfi, M.; Zunica, E.; Stamper, M.; Hickman, S.; Hwang, S.; Young, E.; Atkinson, J.; Manor, D. Antioxidant independent actions of vitamin E in modulating gene expression. Free Radic. Biol. Med. 2018, 128, S58–S59. [Google Scholar] [CrossRef]
- Keen, M.A.; Hassan, I. Vitamin E in dermatology. Indian Dermatol. Online J. 2016, 7, 311–315. [Google Scholar] [CrossRef]
- Reboul, E. Vitamin E Bioavailability: Mechanisms of Intestinal Absorption in the Spotlight. Antioxidants 2017, 6, 95. [Google Scholar] [CrossRef] [Green Version]
- Herrera, E.; Barbas, C. Vitamin E: Action, metabolism and perspectives. J. Physiol. Biochem. 2001, 57, 43–56. [Google Scholar] [CrossRef]
- Meydani, S.N.; Lewis, E.D.; Wu, D. Perspective: Should Vitamin E Recommendations for Older Adults Be Increased? Adv. Nutr. 2018, 9, 533–543. [Google Scholar] [CrossRef] [Green Version]
- La Fata, G.; Weber, P.; Mohajeri, M.H. Effects of vitamin E on cognitive performance during ageing and in Alzheimer’s disease. Nutrients 2014, 6, 5453–5472. [Google Scholar] [CrossRef] [Green Version]
- Kemnic, T.R.; Coleman, M. Vitamin E Deficiency; StatPearls: Treasure Island, FL, USA, 2022. [Google Scholar]
- Ubeda, N.; Achón, M.; Varela-Moreiras, G. Omega 3 fatty acids in the elderly. Br. J. Nutr. 2012, 107, S137–S151. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Molfino, A.; Gioia, G.; Fanelli, F.R.; Muscaritoli, M. The role for dietary omega-3 fatty acids supplementation in older adults. Nutrients 2014, 6, 4058–4072. [Google Scholar] [CrossRef] [PubMed]
- Huang, T.-H.; Wang, P.-W.; Yang, S.-C.; Chou, W.-L.; Fang, J.-Y. Cosmetic and therapeutic applications of fish oil’s fatty acids on the skin. Mar. Drugs 2018, 16, 256. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Whelan, J. (n-6) and (n-3) Polyunsaturated fatty acids and the aging brain: Food for thought. J. Nutr. 2008, 138, 2521–2522. [Google Scholar] [CrossRef] [Green Version]
- Abbatecola, A.M.; Cherubini, A.; Guralnik, J.M.; Andres Lacueva, C.; Ruggiero, C.; Maggio, M.; Bandinelli, S.; Paolisso, G.; Ferrucci, L. Plasma polyunsaturated fatty acids and age-related physical performance decline. Rejuvenation Res. 2009, 12, 25–32. [Google Scholar] [CrossRef] [Green Version]
- Chappus-McCendie, H.; Chevalier, L.; Roberge, C.; Plourde, M. Omega-3 PUFA metabolism and brain modifications during aging. Prog. Neuropsychopharmacol. Biol. Psychiatry 2019, 94, 109662. [Google Scholar] [CrossRef] [Green Version]
- Denis, I.; Potier, B.; Vancassel, S.; Heberden, C.; Lavialle, M. Omega-3 fatty acids and brain resistance to ageing and stress: Body of evidence and possible mechanisms. Ageing Res. Rev. 2013, 12, 579–594. [Google Scholar] [CrossRef]
- Xie, S.H.; Li, H.; Jiang, J.J.; Quan, Y.; Zhang, H.Y. Multi-Omics Interpretation of Anti-Aging Mechanisms for omega-3 Fatty Acids. Genes 2021, 12, 1691. [Google Scholar] [CrossRef]
- de Magalhães, J.P.; Müller, M.; Rainger, G.E.; Steegenga, W. Fish oil supplements, longevity and aging. Aging 2016, 8, 1578. [Google Scholar] [CrossRef] [Green Version]
- Pedersen, A.M. Calanus® Oil. Utilization, Composition and Digestion. Ph.D. Thesis, UiT The Arctic University of Norway, Tromsø, Norway, 2016. [Google Scholar]
- Park, K. Role of micronutrients in skin health and function. Biomol. Ther. 2015, 23, 207. [Google Scholar] [CrossRef] [Green Version]
- Denis, I.; Potier, B.; Heberden, C.; Vancassel, S. Omega-3 polyunsaturated fatty acids and brain aging. Curr. Opin. Clin. Nutr. Metab. Care 2015, 18, 139–146. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Cutuli, D.; Pagani, M.; Caporali, P.; Galbusera, A.; Laricchiuta, D.; Foti, F.; Neri, C.; Spalletta, G.; Caltagirone, C.; Petrosini, L. Effects of omega-3 fatty acid supplementation on cognitive functions and neural substrates: A voxel-based morphometry study in aged mice. Front. Aging Neurosci. 2016, 8, 38. [Google Scholar] [CrossRef] [PubMed]
- Gellert, S.; Schuchardt, J.P.; Hahn, A. Low long chain omega-3 fatty acid status in middle-aged women. Prostaglandins Leukot. Essent. Fatty Acids 2017, 117, 54–59. [Google Scholar] [CrossRef]
- Cutuli, D. Functional and structural benefits induced by omega-3 polyunsaturated fatty acids during aging. Curr. Neuropharmacol. 2017, 15, 534–542. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Maltais, M.; de Souto Barreto, P.; Bowman, G.L.; Smith, A.D.; Cantet, C.; Andrieu, S.; Rolland, Y. Omega-3 Supplementation for the Prevention of Cognitive Decline in Older Adults: Does It Depend on Homocysteine Levels? J. Nutr. Health Aging 2022, 26, 615–620. [Google Scholar] [CrossRef]
- Andrieu, S.; Guyonnet, S.; Coley, N.; Cantet, C.; Bonnefoy, M.; Bordes, S.; Bories, L.; Cufi, M.N.; Dantoine, T.; Dartigues, J.F.; et al. Effect of long-term omega 3 polyunsaturated fatty acid supplementation with or without multidomain intervention on cognitive function in elderly adults with memory complaints (MAPT): A randomised, placebo-controlled trial. Lancet Neurol. 2017, 16, 377–389. [Google Scholar] [CrossRef]
- Hooper, C.; De Souto Barreto, P.; Coley, N.; Cantet, C.; Cesari, M.; Andrieu, S.; Vellas, B. Cognitive Changes with Omega-3 Polyunsaturated Fatty Acids in Non-Demented Older Adults with Low Omega-3 Index. Nutr. Health Aging 2017, 21, 988–993. [Google Scholar] [CrossRef]
- O’Rourke, E.J.; Kuballa, P.; Xavier, R.; Ruvkun, G. omega-6 Polyunsaturated fatty acids extend life span through the activation of autophagy. Genes Dev. 2013, 27, 429–440. [Google Scholar] [CrossRef] [Green Version]
- Lapierre, L.R.; Melendez, A.; Hansen, M. Autophagy links lipid metabolism to longevity in C. elegans. Autophagy 2012, 8, 144–146. [Google Scholar] [CrossRef] [Green Version]
- Wang, K.; Zhong, Y.; Yang, F.; Hu, C.; Liu, X.; Zhu, Y.; Yao, K. Causal Effects of N-6 Polyunsaturated Fatty Acids on Age-related Macular Degeneration: A Mendelian Randomization Study. J. Clin. Endocrinol. Metab. 2021, 106, e3565–e3572. [Google Scholar] [CrossRef]
- Patterson, E.; Wall, R.; Fitzgerald, G.F.; Ross, R.P.; Stanton, C. Health implications of high dietary omega-6 polyunsaturated Fatty acids. J. Nutr. Metab. 2012, 2012, 539426. [Google Scholar] [CrossRef] [PubMed]
- Boyd, J.T.; LoCoco, P.M.; Furr, A.R.; Bendele, M.R.; Tram, M.; Li, Q.; Chang, F.M.; Colley, M.E.; Samenuk, G.M.; Arris, D.A.; et al. Elevated dietary omega-6 polyunsaturated fatty acids induce reversible peripheral nerve dysfunction that exacerbates comorbid pain conditions. Nat. Metab. 2021, 3, 762–773. [Google Scholar] [CrossRef] [PubMed]
- Zoroddu, M.A.; Aaseth, J.; Crisponi, G.; Medici, S.; Peana, M.; Nurchi, V.M. The essential metals for humans: A brief overview. J. Inorg. Biochem. 2019, 195, 120–129. [Google Scholar] [CrossRef] [PubMed]
- Chasapis, C.T.; Ntoupa, P.A.; Spiliopoulou, C.A.; Stefanidou, M.E. Recent aspects of the effects of zinc on human health. Arch. Toxicol. 2020, 94, 1443–1460. [Google Scholar] [CrossRef]
- Choi, S.; Liu, X.; Pan, Z. Zinc deficiency and cellular oxidative stress: Prognostic implications in cardiovascular diseases. Acta Pharmacol. Sin. 2018, 39, 1120–1132. [Google Scholar] [CrossRef] [Green Version]
- Cabrera, A.J. Zinc, aging, and immunosenescence: An overview. Pathobiol. Aging Age Relat. Dis. 2015, 5, 25592. [Google Scholar] [CrossRef]
- Jarosz, M.; Olbert, M.; Wyszogrodzka, G.; Mlyniec, K.; Librowski, T. Antioxidant and anti-inflammatory effects of zinc. Zinc-dependent NF-kappaB signaling. Inflammopharmacology 2017, 25, 11–24. [Google Scholar] [CrossRef] [Green Version]
- Institute of Medicine (US) Panel on Micronutrients. 12, Zinc. In Dietary Reference Intakes for Vitamin A, Vitamin K, Arsenic, Boron, Chromium, Copper, Iodine, Iron, Manganese, Molybdenum, Nickel, Silicon, Vanadium, and Zinc; National Academies Press (US): Washington, DC, USA, 2001. [Google Scholar] [CrossRef] [Green Version]
- Pickart, L.; Margolina, A. Skin regenerative and anti-cancer actions of copper peptides. Cosmetics 2018, 5, 29. [Google Scholar] [CrossRef] [Green Version]
- Bjørklund, G.; Shanaida, M.; Lysiuk, R.; Antonyak, H.; Klishch, I.; Shanaida, V.; Peana, M. Selenium: An Antioxidant with a Critical Role in Anti-Aging. Molecules 2022, 27, 6613. [Google Scholar] [CrossRef]
- Solovyev, N.; Drobyshev, E.; Bjørklund, G.; Dubrovskii, Y.; Lysiuk, R.; Rayman, M.P. Selenium, selenoprotein P, and Alzheimer’s disease: Is there a link? Free Radic. Biol. Med. 2018, 127, 124–133. [Google Scholar] [CrossRef]
- Bjørklund, G.; Zou, L.; Wang, J.; Chasapis, C.T.; Peana, M. Thioredoxin reductase as a pharmacological target. Pharmacol. Res. 2021, 174, 105854. [Google Scholar] [CrossRef] [PubMed]
- Kohrle, J. Selenium and the control of thyroid hormone metabolism. Thyroid 2005, 15, 841–853. [Google Scholar] [CrossRef] [PubMed]
- Kobayashi, R.; Hasegawa, M.; Kawaguchi, C.; Ishikawa, N.; Tomiwa, K.; Shima, M.; Nogami, K. Thyroid function in patients with selenium deficiency exhibits high free T4 to T3 ratio. Clin. Pediatr. Endocrinol. 2021, 30, 19–26. [Google Scholar] [CrossRef] [PubMed]
- Olivieri, O.; Girelli, D.; Azzini, M.; Stanzial, A.M.; Russo, C.; Ferroni, M.; Corrocher, R. Low selenium status in the elderly influences thyroid hormones. Clin. Sci. 1995, 89, 637–642. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fulop, T.; Larbi, A.; Dupuis, G.; Le Page, A.; Frost, E.H.; Cohen, A.A.; Witkowski, J.M.; Franceschi, C. Immunosenescence and Inflamm-Aging As Two Sides of the Same Coin: Friends or Foes? Front. Immunol. 2017, 8, 1960. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kazemi, T.; Moodi, M.; Rajabi, S.; Sharifi, F.; Samarghandian, S.; Khorashadizadeh, M.; Farkhondeh, T. Trace element concentration and cognitive dysfunction in elderly residents in Birjand. Curr. Alzheimer Res. 2022. [Google Scholar] [CrossRef]
- Calder, P.C.; Ortega, E.F.; Meydani, S.N.; Adkins, Y.; Stephensen, C.B.; Thompson, B.; Zwickey, H. Nutrition, Immunosenescence, and Infectious Disease: An Overview of the Scientific Evidence on Micronutrients and on Modulation of the Gut Microbiota. Adv. Nutr. 2022, 13, S1–S26. [Google Scholar] [CrossRef]
- Cai, Z.; Zhang, J.; Li, H. Selenium, aging and aging-related diseases. Aging Clin. Exp. Res. 2019, 31, 1035–1047. [Google Scholar] [CrossRef]
- Wong, C.P.; Magnusson, K.R.; Sharpton, T.J.; Ho, E. Effects of zinc status on age-related T cell dysfunction and chronic inflammation. Biometals 2021, 34, 291–301. [Google Scholar] [CrossRef]
- Haase, H.; Rink, L. The immune system and the impact of zinc during aging. Immun. Ageing 2009, 6, 9. [Google Scholar] [CrossRef] [Green Version]
- Baarz, B.R.; Laurentius, T.; Wolf, J.; Wessels, I.; Bollheimer, L.C.; Rink, L. Short-term zinc supplementation of zinc-deficient seniors counteracts CREMalpha—Mediated IL-2 suppression. Immun. Ageing 2022, 19, 40. [Google Scholar] [CrossRef] [PubMed]
- Borkow, G. Using Copper to Improve the Well-Being of the Skin. Curr. Chem. Biol. 2014, 8, 89–102. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Baek, J.H.; Yoo, M.A.; Koh, J.S.; Borkow, G. Reduction of facial wrinkles depth by sleeping on copper oxide-containing pillowcases: A double blind, placebo controlled, parallel, randomized clinical study. J. Cosmet. Dermatol. 2012, 11, 193–200. [Google Scholar] [CrossRef] [PubMed]
- Canfield, C.-A.; Bradshaw, P.C. Amino acids in the regulation of aging and aging-related diseases. Transl. Med. Aging 2019, 3, 70–89. [Google Scholar] [CrossRef]
- Kageyama, H.; Waditee-Sirisattha, R. Antioxidative, anti-inflammatory, and anti-aging properties of mycosporine-like amino acids: Molecular and Cellular Mechanisms in the Protection of Skin-aging. Mar. Drugs 2019, 17, 222. [Google Scholar] [CrossRef] [Green Version]
- Crozier, A.; Clifford, M.N.; Ashihara, H. Plant Secondary Metabolites: Occurrence, Structure and Role in the Human Diet; John Wiley & Sons: Hoboken, NJ, USA, 2008. [Google Scholar]
- Tundis, R.; Loizzo, M.; Bonesi, M.; Menichini, F. Potential role of natural compounds against skin aging. Curr. Med. Chem. 2015, 22, 1515–1538. [Google Scholar] [CrossRef]
- Warsito, M.F.; Kusumawati, I. The Impact of Herbal Products in the Prevention, Regeneration and Delay of Skin Aging. In Reviews on Biomarker Studies in Aging and Anti-Aging Research; Springer: Berlin/Heidelberg, Germany, 2019; pp. 155–174. [Google Scholar]
- Queen, B.L.; Tollefsbol, T.O. Polyphenols and aging. Curr. Aging Sci. 2010, 3, 34–42. [Google Scholar] [CrossRef]
- Nichols, J.A.; Katiyar, S.K. Skin photoprotection by natural polyphenols: Anti-inflammatory, antioxidant and DNA repair mechanisms. Arch. Dermatol. Res. 2010, 302, 71–83. [Google Scholar] [CrossRef] [Green Version]
- Petruk, G.; Del Giudice, R.; Rigano, M.M.; Monti, D.M. Antioxidants from plants protect against skin photoaging. Oxid. Med. Cell Longev. 2018, 2018, 1454936. [Google Scholar] [CrossRef] [Green Version]
- Yücel, Ç.; Şeker Karatoprak, G.; Değim, İ.T. Anti-aging formulation of rosmarinic acid-loaded ethosomes and liposomes. J. Microencapsul. 2019, 36, 180–191. [Google Scholar] [CrossRef]
- Liu, Y.; Song, X.; Zhang, D.; Zhou, F.; Wang, D.; Wei, Y.; Gao, F.; Xie, L.; Jia, G.; Wu, W. Blueberry anthocyanins: Protection against ageing and light-induced damage in retinal pigment epithelial cells. Br. J. Nutr. 2012, 108, 16–27. [Google Scholar] [CrossRef] [PubMed]
- Li, H.; Chen, F.J.; Yang, W.L.; Qiao, H.Z.; Zhang, S.J. Quercetin improves cognitive disorder in aging mice by inhibiting NLRP3 inflammasome activation. Food Funct. 2021, 12, 717–725. [Google Scholar] [CrossRef] [PubMed]
- Katiyar, S.K. Green tea prevents non-melanoma skin cancer by enhancing DNA repair. Arch. Biochem. Biophys. 2011, 508, 152–158. [Google Scholar] [CrossRef] [Green Version]
- Peluso, I.; Serafini, M. Antioxidants from black and green tea: From dietary modulation of oxidative stress to pharmacological mechanisms. Br. J. Pharmacol. 2017, 174, 1195–1208. [Google Scholar] [CrossRef] [Green Version]
- Shanaida, M.; Golembiovska, O.; Hudz, N.; Wieczorek, P.P. Phenolic compounds of herbal infusions obtained from some species of the family. Curr. Issues Pharm. Med. Sci. 2018, 31, 194–199. [Google Scholar] [CrossRef] [Green Version]
- Ayaz, M.; Sadiq, A.; Junaid, M.; Ullah, F.; Ovais, M.; Ullah, I.; Ahmed, J.; Shahid, M. Flavonoids as prospective neuroprotectants and their therapeutic propensity in aging associated neurological disorders. Front. Aging Neurosci. 2019, 11, 155. [Google Scholar] [CrossRef] [Green Version]
- Rawal, G.; Yadav, S.; Nagayach, M.S. Phytosterols and the health. Med. Res. Chron. 2015, 2, 441–444. [Google Scholar]
- Luo, J.; Si, H.; Jia, Z.; Liu, D. Dietary Anti-Aging Polyphenols and Potential Mechanisms. Antioxidants 2021, 10, 283. [Google Scholar] [CrossRef]
- Fan, X.; Fan, Z.; Yang, Z.; Huang, T.; Tong, Y.; Yang, D.; Mao, X.; Yang, M. Flavonoids-Natural Gifts to Promote Health and Longevity. Int. J. Mol. Sci. 2022, 23, 2176. [Google Scholar] [CrossRef]
- Tsao, R. Chemistry and biochemistry of dietary polyphenols. Nutrients 2010, 2, 1231–1246. [Google Scholar] [CrossRef] [Green Version]
- Gasmi, A.; Mujawdiya, P.K.; Lysiuk, R.; Shanaida, M.; Peana, M.; Gasmi Benahmed, A.; Beley, N.; Kovalska, N.; Bjørklund, G. Quercetin in the Prevention and Treatment of Coronavirus Infections: A Focus on SARS-CoV-2. Pharmaceuticals 2022, 15, 1049. [Google Scholar] [CrossRef] [PubMed]
- Gasmi, A.; Mujawdiya, P.K.; Noor, S.; Lysiuk, R.; Darmohray, R.; Piscopo, S.; Lenchyk, L.; Antonyak, H.; Dehtiarova, K.; Shanaida, M.; et al. Polyphenols in Metabolic Diseases. Molecules 2022, 27, 6280. [Google Scholar] [CrossRef] [PubMed]
- Justice, J.N.; Nambiar, A.M.; Tchkonia, T.; LeBrasseur, N.K.; Pascual, R.; Hashmi, S.K.; Prata, L.; Masternak, M.M.; Kritchevsky, S.B.; Musi, N.; et al. Senolytics in idiopathic pulmonary fibrosis: Results from a first-in-human, open-label, pilot study. EBioMedicine 2019, 40, 554–563. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Fan, T.; Du, Y.; Zhang, M.; Zhu, A.R.; Zhang, J. Senolytics Cocktail Dasatinib and Quercetin Alleviate Human Umbilical Vein Endothelial Cell Senescence via the TRAF6-MAPK-NF-kappaB Axis in a YTHDF2-Dependent Manner. Gerontology 2022, 68, 920–934. [Google Scholar] [CrossRef]
- Sierra-Ramirez, A.; Lopez-Aceituno, J.L.; Costa-Machado, L.F.; Plaza, A.; Barradas, M.; Fernandez-Marcos, P.J. Transient metabolic improvement in obese mice treated with navitoclax or dasatinib/quercetin. Aging 2020, 12, 11337–11348. [Google Scholar] [CrossRef]
- Hickson, L.J.; Langhi Prata, L.G.P.; Bobart, S.A.; Evans, T.K.; Giorgadze, N.; Hashmi, S.K.; Herrmann, S.M.; Jensen, M.D.; Jia, Q.; Jordan, K.L.; et al. Senolytics decrease senescent cells in humans: Preliminary report from a clinical trial of Dasatinib plus Quercetin in individuals with diabetic kidney disease. EBioMedicine 2019, 47, 446–456. [Google Scholar] [CrossRef] [Green Version]
- Jing, W.; Xiaolan, C.; Yu, C.; Feng, Q.; Haifeng, Y. Pharmacological effects and mechanisms of tannic acid. Biomed. Pharmacother. 2022, 154, 113561. [Google Scholar] [CrossRef]
- Navarro-Cruz, A.; Ramírez y Ayala, R.; Ochoa-Velasco, C.; Brambila, E.; Avila-Sosa, R.; Pérez-Fernández, S.; Morales-Medina, J.; Aguilar-Alonso, P. Effect of chronic administration of resveratrol on cognitive performance during aging process in rats. Oxid. Med. Cell Longev. 2017, 2017, 8510761. [Google Scholar] [CrossRef] [Green Version]
- Pallauf, K.; Rimbach, G.; Rupp, P.M.; Chin, D.; MA Wolf, I. Resveratrol and lifespan in model organisms. Curr. Med. Chem. 2016, 23, 4639–4680. [Google Scholar] [CrossRef]
- Reinisalo, M.; Kårlund, A.; Koskela, A.; Kaarniranta, K.; Karjalainen, R.O. Polyphenol stilbenes: Molecular mechanisms of defence against oxidative stress and aging-related diseases. Oxid. Med. Cell Longev. 2015, 2015, 340520. [Google Scholar] [CrossRef] [Green Version]
- McCubrey, J.A.; Lertpiriyapong, K.; Steelman, L.S.; Abrams, S.L.; Yang, L.V.; Murata, R.M.; Rosalen, P.L.; Scalisi, A.; Neri, L.M.; Cocco, L.; et al. Effects of resveratrol, curcumin, berberine and other nutraceuticals on aging, cancer development, cancer stem cells and microRNAs. Aging 2017, 9, 1477–1536. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chedea, V.S.; Tomoiaga, L.L.; Macovei, S.O.; Magureanu, D.C.; Iliescu, M.L.; Bocsan, I.C.; Buzoianu, A.D.; Vosloban, C.M.; Pop, R.M. Antioxidant/Pro-Oxidant Actions of Polyphenols From Grapevine and Wine By-Products-Base for Complementary Therapy in Ischemic Heart Diseases. Front. Cardiovasc. Med. 2021, 8, 750508. [Google Scholar] [CrossRef] [PubMed]
- Ma, S.; Feng, J.; Zhang, R.; Chen, J.; Han, D.; Li, X.; Yang, B.; Fan, M.; Li, C.; Tian, Z.; et al. SIRT1 Activation by Resveratrol Alleviates Cardiac Dysfunction via Mitochondrial Regulation in Diabetic Cardiomyopathy Mice. Oxid. Med. Cell Longev. 2017, 2017, 4602715. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bhullar, K.S.; Hubbard, B.P. Lifespan and healthspan extension by resveratrol. Biochim. Biophys. Acta 2015, 1852, 1209–1218. [Google Scholar] [CrossRef] [Green Version]
- Shailaja, M.; Gowda, K.D.; Vishakh, K.; Kumari, N.S. Anti-aging role of curcumin by modulating the inflammatory markers in albino wistar rats. J. Natl. Med. Assoc. 2017, 109, 9–13. [Google Scholar] [CrossRef]
- Shen, L.R.; Parnell, L.D.; Ordovas, J.M.; Lai, C.Q. Curcumin and aging. Biofactors 2013, 39, 133–140. [Google Scholar] [CrossRef]
- Fleenor, B.S.; Sindler, A.L.; Marvi, N.K.; Howell, K.L.; Zigler, M.L.; Yoshizawa, M.; Seals, D.R. Curcumin ameliorates arterial dysfunction and oxidative stress with aging. Exp. Gerontol. 2013, 48, 269–276. [Google Scholar] [CrossRef] [Green Version]
- Agatonovic-Kustrin, S.; Kustrin, E.; Morton, D.W. Essential oils and functional herbs for healthy aging. Neural Regen. Res. 2019, 14, 441. [Google Scholar] [CrossRef]
- Loizzo, M.R.; Jemia, M.B.; Senatore, F.; Bruno, M.; Menichini, F.; Tundis, R. Chemistry and functional properties in prevention of neurodegenerative disorders of five Cistus species essential oils. Food Chem. Toxicol. 2013, 59, 586–594. [Google Scholar] [CrossRef]
- Shanaida, M. Antioxidant activity of essential oils obtained from aerial part of some Lamiaceae species. Int. J. Green Pharm. 2018, 12, 200–204. [Google Scholar] [CrossRef]
- Hancianu, M.; Cioanca, O.; Mihasan, M.; Hritcu, L. Neuroprotective effects of inhaled lavender oil on scopolamine-induced dementia via anti-oxidative activities in rats. Phytomedicine 2013, 20, 446–452. [Google Scholar] [CrossRef] [PubMed]
- Gharibi, S.; Bakhtiari, N.; Elham Moslemee, J.; Bakhtiari, F. Ursolic Acid Mediates Hepatic Protection through Enhancing of anti-aging Biomarkers. Curr. Aging Sci. 2018, 11, 16–23. [Google Scholar] [CrossRef] [PubMed]
- Maccioni, R.B.; Calfio, C.; Gonzalez, A.; Luttges, V. Novel Nutraceutical Compounds in Alzheimer Prevention. Biomolecules 2022, 12, 249. [Google Scholar] [CrossRef] [PubMed]
- Yasin, Z.A.; Ibrahim, F.; Rashid, N.N.; Razif, M.F.; Yusof, R. The importance of some plant extracts as skin anti-aging resources: A review. Curr. Pharm. Biotechnol. 2017, 18, 864–876. [Google Scholar] [CrossRef]
- Zeb, I.; Ahmadi, N.; Nasir, K.; Kadakia, J.; Larijani, V.N.; Flores, F.; Li, D.; Budoff, M.J. Aged garlic extract and coenzyme Q10 have favorable effect on inflammatory markers and coronary atherosclerosis progression: A randomized clinical trial. J. Cardiovasc. Dis. Res. 2012, 3, 185–190. [Google Scholar] [CrossRef] [Green Version]
- Chen, P.H.; Chang, C.H.; Lin, W.S.; Nagabhushanam, K.; Ho, C.T.; Pan, M.H. S-Allylcysteine Ameliorates Aging Features via Regulating Mitochondrial Dynamics in Naturally Aged C57BL/6J Mice. Mol. Nutr. Food. Res. 2022, 66, e2101077. [Google Scholar] [CrossRef]
- Bjørklund, G.; Rahaman, M.S.; Shanaida, M.; Lysiuk, R.; Oliynyk, P.; Lenchyk, L.; Chirumbolo, S.; Chasapis, C.T.; Peana, M. Natural Dietary Compounds in the Treatment of Arsenic Toxicity. Molecules 2022, 27, 4871. [Google Scholar] [CrossRef]
- Bjørklund, G.; Oliinyk, P.; Lysiuk, R.; Rahaman, M.S.; Antonyak, H.; Lozynska, I.; Lenchyk, L.; Peana, M. Arsenic intoxication: General aspects and chelating agents. Arch. Toxicol. 2020, 94, 1879–1897. [Google Scholar] [CrossRef]
- Zhu, S.Y.; Dong, Y.; Tu, J.; Zhou, Y.; Zhou, X.H.; Xu, B. Silybum marianum oil attenuates oxidative stress and ameliorates mitochondrial dysfunction in mice treated with D-galactose. Pharmacogn. Mag. 2014, 10, S92. [Google Scholar] [CrossRef] [Green Version]
- Zuo, W.; Yan, F.; Zhang, B.; Li, J.; Mei, D. Advances in the studies of Ginkgo biloba leaves extract on aging-related diseases. Aging Dis. 2017, 8, 812. [Google Scholar] [CrossRef] [Green Version]
- Budoff, M.J.; Ahmadi, N.; Gul, K.M.; Liu, S.T.; Flores, F.R.; Tiano, J.; Takasu, J.; Miller, E.; Tsimikas, S. Aged garlic extract supplemented with B vitamins, folic acid and L-arginine retards the progression of subclinical atherosclerosis: A randomized clinical trial. Prev. Med. 2009, 49, 101–107. [Google Scholar] [CrossRef] [PubMed]
- Moghimipour, E. Hydroxy acids, the most widely used anti-aging agents. Jundishapur J. Nat. Pharm. Prod. 2012, 7, 9–10. [Google Scholar] [CrossRef] [PubMed]
- Shanaida, M.; Lysiuk, R.; Mykhalkiv, M.; Shanaida, V. Chromatographic profiles of carboxylic acids in the raw materials of some mentheae dumort. Species. PharmacologyOnLine 2021, 3, 30–37. [Google Scholar]
- Green, B.A.; Yu, R.J.; Van Scott, E.J. Clinical and cosmeceutical uses of hydroxyacids. Clin. Dermatol. 2009, 27, 495–501. [Google Scholar] [CrossRef]
- Brooks, J.D.; Ward, W.E.; Lewis, J.E.; Hilditch, J.; Nickell, L.; Wong, E.; Thompson, L.U. Supplementation with flaxseed alters estrogen metabolism in postmenopausal women to a greater extent than does supplementation with an equal amount of soy. Am. J. Clin. Nutr. 2004, 79, 318–325. [Google Scholar] [CrossRef] [Green Version]
- Jefremov, V.; Zilmer, M.; Zilmer, K.; Bogdanovic, N.; Karelson, E. Antioxidative effects of plant polyphenols: From protection of G protein signaling to prevention of age-related pathologies. Ann. N. Y. Acad. Sci. 2007, 1095, 449–457. [Google Scholar] [CrossRef]
- Si, H.; Lai, C.Q.; Liu, D. Dietary Epicatechin, A Novel Anti-aging Bioactive Small Molecule. Curr. Med. Chem. 2021, 28, 3–18. [Google Scholar] [CrossRef]
- Latif, R. Chocolate/cocoa and human health: A review. Neth. J. Med. 2013, 71, 63–68. [Google Scholar]
- Montagna, M.T.; Diella, G.; Triggiano, F.; Caponio, G.R.; De Giglio, O.; Caggiano, G.; Di Ciaula, A.; Portincasa, P. Chocolate, “Food of the Gods”: History, Science, and Human Health. Int. J. Environ. Res. Public Health 2019, 16, 4960. [Google Scholar] [CrossRef] [Green Version]
- Sorrenti, V.; Ali, S.; Mancin, L.; Davinelli, S.; Paoli, A.; Scapagnini, G. Cocoa Polyphenols and Gut Microbiota Interplay: Bioavailability, Prebiotic Effect, and Impact on Human Health. Nutrients 2020, 12, 1908. [Google Scholar] [CrossRef]
- Esser, D.; Mars, M.; Oosterink, E.; Stalmach, A.; Muller, M.; Afman, L.A. Dark chocolate consumption improves leukocyte adhesion factors and vascular function in overweight men. Faseb J. 2014, 28, 1464–1473. [Google Scholar] [CrossRef] [PubMed]
- Lopez-Otin, C.; Galluzzi, L.; Freije, J.M.P.; Madeo, F.; Kroemer, G. Metabolic Control of Longevity. Cell 2016, 166, 802–821. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mirza, M.A. Future of Humic substances as Pharmaceutical Excipient. Pharm. Sci. Anal. Res. J. 2018, 1, 180004. [Google Scholar]
- Chauke, T.L. Evaluating the Efficacy, Safety and Possible Mechanism of Action of Potassium Humate with Selenium. Master’s Thesis, University of Pretoria, Pretoria, South Africa, 2013. [Google Scholar]
- Aeschbacher, M.; Graf, C.; Schwarzenbach, R.P.; Sander, M. Antioxidant properties of humic substances. Environ. Sci. Technol. 2012, 46, 4916–4925. [Google Scholar] [CrossRef] [PubMed]
- de Melo, B.A.; Motta, F.L.; Santana, M.H. Humic acids: Structural properties and multiple functionalities for novel technological developments. Mater. Sci. Eng. C Mater. Biol. Appl. 2016, 62, 967–974. [Google Scholar] [CrossRef] [PubMed]
- Jacob, K.K.; Prashob, P.K.J.; Chandramohanakumar, N. Humic substances as a potent biomaterials for therapeutic and drug delivery system—A review. Int. J. Appl. Pharm. 2019, 11, 1–4. [Google Scholar] [CrossRef] [Green Version]
- Kučerík, J.; Bakajová, B.; Pekař, M. Antioxidant effect of lignite humic acids and its salts on the thermo-oxidative stability/degradation of polyvinyl alcohol blends. Environ. Chem. Lett. 2008, 6, 241–245. [Google Scholar] [CrossRef]
- Vysokogorskii, V.E.; Nozdrunova, A.A.; Plaksin, G.V.; Krivonos, O.I.; Mkrtchan, O.Z.; Petrosyan, L.Y. Antioxidant activity of liquid products of heat-treated sapropels. Pharm. Chem. J. 2009, 43, 191–194. [Google Scholar] [CrossRef]
- Avvakumova, N.P.; Gerchikov, A.Y.; Khairullina, V.R.; Zhdanova, A.V. Antioxidant properties of humic substances isolated from peloids. Pharm. Chem. J. 2011, 45, 192. [Google Scholar] [CrossRef]
- Vaskova, J.; Velika, B.; Pilatova, M.; Kron, I.; Vasko, L. Effects of humic acids in vitro. In Vitro Cell Dev. Biol. Anim. 2011, 47, 376–382. [Google Scholar] [CrossRef]
- Tarasova, A.S.; Stom, D.I.; Kudryasheva, N.S. Antioxidant activity of humic substances via bioluminescent monitoring in vitro. Environ. Monit. Assess. 2015, 187, 89. [Google Scholar] [CrossRef] [PubMed]
- Khil’ko, S.L.; Efimova, I.V.; Smirnova, O.V. Antioxidant properties of humic acids from brown coal. Solid Fuel Chem. 2011, 45, 367–371. [Google Scholar] [CrossRef]
- Jackson, W.R. Humic, Fulvic and Microbial Balance: Organic Soil Conditioning; Jackson Research Center: Evergreen, CO, USA, 1993. [Google Scholar]
- Shenyuan, Y. Application of Fulvic Acid and Its Derivatives in the Fields of Agriculture and Medicine, 1st ed.; IHSS: Sevilla, Spain, 1993. [Google Scholar]
- Kinoshita, H.; Kinoshita, M.; Takahashi, A.; Yuasa, S.; Fukuda, K. Effect of fulvic acid on ultraviolet induced skin aging: The effect of fulvic acid on fibroblasts and matrix metalloproteinase. Nishinihon J. Dermatol. 2012, 74, 427–431. [Google Scholar] [CrossRef]
- Pant, K.; Gupta, A.; Gupta, P.; Ashraf, A.; Yadav, A.; Venugopal, S. Anti-proliferative and anticancer properties of fulvic acid on hepatic cancer cells. J. Clin. Exp. Hepatol. 2015, 5, S2. [Google Scholar] [CrossRef]
- Aykac, A.; Becer, E.; Okcanoğlu, T.B.; Güvenir, M.; Süer, K.; Vatansever, S. The Cytotoxic Effects of Humic Acid on Human Breast Cancer Cells. Proceedings 2018, 2, 1565. [Google Scholar]
- Martini, S.; D’Addario, C.; Bonechi, C.; Leone, G.; Tognazzi, A.; Consumi, M.; Magnani, A.; Rossi, C. Increasing photostability and water-solubility of carotenoids: Synthesis and characterization of beta-carotene-humic acid complexes. J. Photochem. Photobiol. B 2010, 101, 355–361. [Google Scholar] [CrossRef]
- Ghosal, S. Delivery System for Pharmaceutical, Nutritional and Cosmetic Ingredients. U.S. Patent US6558712B1, 6 May 2003. [Google Scholar]
- Khanna, R.; Agarwal, S.P.; Khar, R.K. Fulvic Acids and Humic Acids as Novel Complexing Agents and a Process. Indian Patent 249172, 14 October 2011. [Google Scholar]
- Alves, A.; Sousa, E.; Kijjoa, A.; Pinto, M. Marine-Derived Compounds with Potential Use as Cosmeceuticals and Nutricosmetics. Molecules 2020, 25, 2536. [Google Scholar] [CrossRef]
- Gasmi, A.; Mujawdiya, P.K.; Shanaida, M.; Ongenae, A.; Lysiuk, R.; Dosa, M.D.; Tsal, O.; Piscopo, S.; Chirumbolo, S.; Bjorklund, G. Calanus oil in the treatment of obesity-related low-grade inflammation, insulin resistance, and atherosclerosis. Appl. Microbiol. Biotechnol. 2020, 104, 967–979. [Google Scholar] [CrossRef]
- Wang, X.; Zhang, Z.; Zhang, S.; Yang, F.; Yang, M.; Zhou, J.; Hu, Z.; Xu, X.; Mao, G.; Chen, G.; et al. Antiaging compounds from marine organisms. Food. Res. Int. 2021, 143, 110313. [Google Scholar] [CrossRef] [PubMed]
- Ghosh, S.; Sarkar, T.; Pati, S.; Kari, Z.A.; Edinur, H.A.; Chakraborty, R. Novel bioactive compounds from marine sources as a tool for functional food development. Front. Mar. Sci. 2022, 9, 10-3389. [Google Scholar] [CrossRef]
- Zhou, X.; Cao, Q.; Orfila, C.; Zhao, J.; Zhang, L. Systematic Review and Meta-Analysis on the Effects of Astaxanthin on Human Skin Ageing. Nutrients 2021, 13, 2917. [Google Scholar] [CrossRef] [PubMed]
- Singh, K.N.; Patil, S.; Barkate, H. Protective effects of astaxanthin on skin: Recent scientific evidence, possible mechanisms, and potential indications. J. Cosmet. Dermatol. 2020, 19, 22–27. [Google Scholar] [CrossRef]
- Chan, K.C.; Mong, M.C.; Yin, M.C. Antioxidative and anti-inflammatory neuroprotective effects of astaxanthin and canthaxanthin in nerve growth factor differentiated PC12 cells. J. Food Sci. 2009, 74, H225–H231. [Google Scholar] [CrossRef]
- Bjørklund, G.; Gasmi, A.; Lenchyk, L.; Shanaida, M.; Zafar, S.; Mujawdiya, P.; Lysiukh, R.; Antonyak, H.; Noor, S.; Akram, M.; et al. The Role of Astaxanthin as a Neutraceutical in Health and Aging. Molecules 2022, in press. [Google Scholar]
- Ahn, J.H.; Kim, D.W.; Park, C.W.; Kim, B.; Sim, H.; Kim, H.S.; Lee, T.K.; Lee, J.C.; Yang, G.E.; Her, Y.; et al. Laminarin Attenuates Ultraviolet-Induced Skin Damage by Reducing Superoxide Anion Levels and Increasing Endogenous Antioxidants in the Dorsal Skin of Mice. Mar. Drugs 2020, 18, 345. [Google Scholar] [CrossRef]
- Cao, L.; Lee, S.G.; Lim, K.T.; Kim, H.R. Potential Anti-Aging Substances Derived from Seaweeds. Mar. Drugs 2020, 18, 564. [Google Scholar] [CrossRef]
- Havas, F.; Krispin, S.; Cohen, M.; Loing, E.; Farge, M.; Suere, T.; Attia-Vigneau, J. A Dunaliella salina Extract Counteracts Skin Aging under Intense Solar Irradiation Thanks to Its Antiglycation and Anti-Inflammatory Properties. Mar. Drugs 2022, 20, 104. [Google Scholar] [CrossRef]
- Kim, J.H.; Lee, J.E.; Kim, K.H.; Kang, N.J. Beneficial Effects of Marine Algae-Derived Carbohydrates for Skin Health. Mar. Drugs. 2018, 16, 459. [Google Scholar] [CrossRef] [Green Version]
- Cornara, L.; Biagi, M.; Xiao, J.; Burlando, B. Therapeutic Properties of Bioactive Compounds from Different Honeybee Products. Front. Pharmacol. 2017, 8, 412. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Giampieri, F.; Quiles, J.L.; Cianciosi, D.; Forbes-Hernandez, T.Y.; Orantes-Bermejo, F.J.; Alvarez-Suarez, J.M.; Battino, M. Bee Products: An Emblematic Example of Underutilized Sources of Bioactive Compounds. J. Agric. Food Chem. 2022, 70, 6833–6848. [Google Scholar] [CrossRef] [PubMed]
- Kurek-Gorecka, A.; Gorecki, M.; Rzepecka-Stojko, A.; Balwierz, R.; Stojko, J. Bee Products in Dermatology and Skin Care. Molecules 2020, 25, 556. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Collazo, N.; Carpena, M.; Nunez-Estevez, B.; Otero, P.; Simal-Gandara, J.; Prieto, M.A. Health Promoting Properties of Bee Royal Jelly: Food of the Queens. Nutrients 2021, 13, 543. [Google Scholar] [CrossRef] [PubMed]
- Kunugi, H.; Mohammed Ali, A. Royal Jelly and Its Components Promote Healthy Aging and Longevity: From Animal Models to Humans. Int. J. Mol. Sci. 2019, 20, 4662. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhang, C.; Gao, Z.; Hu, C.; Zhang, J.; Sun, X.; Rong, C.; Jia, L. Antioxidant, antibacterial and anti-aging activities of intracellular zinc polysaccharides from Grifola frondosa SH-05. Int. J. Biol. Macromol. 2017, 95, 778–787. [Google Scholar] [CrossRef]
- Wu, J.Y.; Siu, K.C.; Geng, P. Bioactive Ingredients and Medicinal Values of Grifola frondosa (Maitake). Foods 2021, 10, 95. [Google Scholar] [CrossRef]
- Wang, J.; Cao, B.; Zhao, H.; Feng, J. Emerging Roles of Ganoderma Lucidum in Anti-Aging. Aging Dis. 2017, 8, 691–707. [Google Scholar] [CrossRef] [Green Version]
- Lolou, V.; Panayiotidis, M.I. Functional Role of Probiotics and Prebiotics on Skin Health and Disease. Fermentation 2019, 5, 41. [Google Scholar] [CrossRef]
- Sivamaruthi, B.S.; Kesika, P.; Chaiyasut, C. A review on anti-aging properties of probiotics. Int. J. Appl. Pharm. 2018, 10, 23–27. [Google Scholar] [CrossRef] [Green Version]
- Roudsari, M.R.; Karimi, R.; Sohrabvandi, S.; Mortazavian, A. Health effects of probiotics on the skin. Crit. Rev. Food Sci. Nutr. 2015, 55, 1219–1240. [Google Scholar] [CrossRef] [PubMed]
- Gasmi, A.; Tippairote, T.; Mujawdiya, P.K.; Peana, M.; Menzel, A.; Dadar, M.; Benahmed, A.G.; Bjørklund, G. The microbiota-mediated dietary and nutritional interventions for COVID-19. Clin. Immunol. 2021, 226, 108725. [Google Scholar] [CrossRef] [PubMed]
- Christensen, K.V.; Morch, M.G.; Morthorst, T.H.; Lykkemark, S.; Olsen, A. Microbiota, probiotic bacteria and ageing. In Ageing: Lessons from C. elegans; Springer: Berlin/Heidelberg, Germany, 2017; pp. 411–429. [Google Scholar]
- Inglis, J.E.; Ilich, J.Z. The microbiome and osteosarcopenic obesity in older individuals in long-term care facilities. Curr. Osteoporos. Rep. 2015, 13, 358–362. [Google Scholar] [CrossRef] [PubMed]
- Tremaroli, V.; Bäckhed, F. Functional interactions between the gut microbiota and host metabolism. Nature 2012, 489, 242. [Google Scholar] [CrossRef]
- El-Abbadi, N.H.; Dao, M.C.; Meydani, S.N. Yogurt: Role in healthy and active aging. Am. J. Clin. Nutr. 2014, 99, 1263S–1270S. [Google Scholar] [CrossRef] [Green Version]
- Hooper, L.; Bunn, D.; Jimoh, F.O.; Fairweather-Tait, S.J. Water-loss dehydration and aging. Mech. Ageing Dev. 2014, 136, 50–58. [Google Scholar] [CrossRef] [Green Version]
- Palma, L.; Marques, L.T.; Bujan, J.; Rodrigues, L.M. Dietary water affects human skin hydration and biomechanics. Clin. Cosmet. Investig. Dermatol. 2015, 8, 413. [Google Scholar] [CrossRef] [Green Version]
- Martino, D. The Effects of Chlorinated Drinking Water on the Assembly of the Intestinal Microbiome. Challenges 2019, 10, 10. [Google Scholar] [CrossRef] [Green Version]
- Forbes, J.D.; Van Domselaar, G.; Sargent, M.; Green, C.; Springthorpe, S.; Krause, D.O.; Bernstein, C.N. Microbiome profiling of drinking water in relation to incidence of inflammatory bowel disease. Can. J. Microbiol. 2016, 62, 781–793. [Google Scholar] [CrossRef]
- Perrin, Y.; Bouchon, D.; Delafont, V.; Moulin, L.; Héchard, Y. Microbiome of drinking water: A full-scale spatio-temporal study to monitor water quality in the Paris distribution system. Water Res. 2019, 149, 375–385. [Google Scholar] [CrossRef]
- Jafri, A.B. Aging and toxins. Clin. Geriatr. Med. 2011, 27, 609–628. [Google Scholar] [CrossRef] [PubMed]
- World Health Organization. Natural Toxins in Food. Available online: https://www.who.int/news-room/fact-sheets/detail/natural-toxins-in-food (accessed on 23 August 2022).
- Melnikova, D.I.; Khotimchenko, Y.S.; Magarlamov, T.Y. Addressing the Issue of Tetrodotoxin Targeting. Mar. Drugs 2018, 16, 352. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kohane, D.S.; Yieh, J.; Lu, N.T.; Langer, R.; Strichartz, G.R.; Berde, C.B. A re-examination of tetrodotoxin for prolonged duration local anesthesia. Anesthesiology 1998, 89, 119–131. [Google Scholar] [CrossRef] [PubMed]
- Falconer, I.R.; Humpage, A.R. Health risk assessment of cyanobacterial (blue-green algal) toxins in drinking water. Int. J. Environ. Res Public. Health 2005, 2, 43–50. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Mahmood, N.A.; Carmichael, W.W. Paralytic shellfish poisons produced by the freshwater cyanobacterium Aphanizomenon flos-aquae NH-5. Toxicon 1986, 24, 175–186. [Google Scholar] [CrossRef]
- Cusick, K.D.; Sayler, G.S. An overview on the marine neurotoxin, saxitoxin: Genetics, molecular targets, methods of detection and ecological functions. Mar. Drugs 2013, 11, 991–1018. [Google Scholar] [CrossRef] [Green Version]
- Agnihotri, V.K. Anabaena flos-aquae. Crit. Rev. Environ. Sci. Technol. 2014, 44, 1995–2037. [Google Scholar] [CrossRef]
- Elleman, T.C.; Falconer, I.R.; Jackson, A.R.; Runnegar, M.T. Isolation, characterization and pathology of the toxin from a Microcystis aeruginosa (= Anacystis cyanea) bloom. Aust. J. Biol. Sci. 1978, 31, 209–218. [Google Scholar] [CrossRef]
- Sivonen, K.; Carmichael, W.W.; Namikoshi, M.; Rinehart, K.L.; Dahlem, A.M.; Niemela, S.I. Isolation and characterization of hepatotoxic microcystin homologs from the filamentous freshwater cyanobacterium Nostoc sp. strain 152. Appl. Environ. Microbiol. 1990, 56, 2650–2657. [Google Scholar] [CrossRef]
- Bennett, J.W.; Klich, M. Mycotoxins. Clin. Microbiol. Rev. 2003, 16, 497–516. [Google Scholar] [CrossRef] [Green Version]
- Yazar, S.; Omurtag, G.Z. Fumonisins, trichothecenes and zearalenone in cereals. Int. J. Mol. Sci. 2008, 9, 2062–2090. [Google Scholar] [CrossRef] [PubMed]
- Rahmani, A.; Jinap, S.; Soleimany, F. Qualitative and Quantitative Analysis of Mycotoxins. Compr. Rev. Food Sci. Food Saf. 2009, 8, 202–251. [Google Scholar] [CrossRef] [PubMed]
- Bertero, A.; Moretti, A.; Spicer, L.J.; Caloni, F. Fusarium Molds and Mycotoxins: Potential Species-Specific Effects. Toxins 2018, 10, 244. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Marijani, E.; Kigadye, E.; Okoth, S. Occurrence of Fungi and Mycotoxins in Fish Feeds and Their Impact on Fish Health. Int. J. Microbiol. 2019, 2019, 6743065. [Google Scholar] [CrossRef] [Green Version]
- Ferrigo, D.; Raiola, A.; Causin, R. Fusarium Toxins in Cereals: Occurrence, Legislation, Factors Promoting the Appearance and Their Management. Molecules 2016, 21, 627. [Google Scholar] [CrossRef] [Green Version]
- Diaz, J.H. Poisoning by Herbs and Plants: Rapid Toxidromic Classification and Diagnosis. Wilderness Environ. Med. 2016, 27, 136–152. [Google Scholar] [CrossRef] [Green Version]
- Ma, L.; Gu, R.; Tang, L.; Chen, Z.E.; Di, R.; Long, C. Important poisonous plants in tibetan ethnomedicine. Toxins 2015, 7, 138–155. [Google Scholar] [CrossRef] [Green Version]
- Birnbaum, L.S. The effect of environmental chemicals on human health. Fertil. Steril. 2008, 89, e31. [Google Scholar] [CrossRef]
- Otter, J.; D’Orazio, J.L. Strychnine Toxicity; StatPearls Publishing: Treasure Island, FL, USA, 2021. [Google Scholar]
- Moreira, R.; Pereira, D.M.; Valentao, P.; Andrade, P.B. Pyrrolizidine Alkaloids: Chemistry, Pharmacology, Toxicology and Food Safety. Int. J. Mol. Sci. 2018, 19, 1668. [Google Scholar] [CrossRef]
- Schramm, S.; Kohler, N.; Rozhon, W. Pyrrolizidine Alkaloids: Biosynthesis, Biological Activities and Occurrence in Crop Plants. Molecules 2019, 24, 498. [Google Scholar] [CrossRef] [Green Version]
- Bradberry, S.M.; Dickers, K.J.; Rice, P.; Griffiths, G.D.; Vale, J.A. Ricin poisoning. Toxicol. Rev. 2003, 22, 65–70. [Google Scholar] [CrossRef] [PubMed]
- Moshiri, M.; Hamid, F.; Etemad, L. Ricin Toxicity: Clinical and Molecular Aspects. Rep. Biochem. Mol. Biol. 2016, 4, 60–65. [Google Scholar] [PubMed]
- Onojah, P.; Odin, E. Cyanogenic glycoside in food plants. Int. J. Innov. Sci. Math. 2015, 3, 2347–9051. [Google Scholar]
- Senica, M.; Stampar, F.; Veberic, R.; Mikulic-Petkovsek, M. Fruit Seeds of the Rosaceae Family: A Waste, New Life, or a Danger to Human Health? J. Agric. Food Chem. 2017, 65, 10621–10629. [Google Scholar] [CrossRef]
- Bruni, R.; Barreca, D.; Protti, M.; Brighenti, V.; Righetti, L.; Anceschi, L.; Mercolini, L.; Benvenuti, S.; Gattuso, G.; Pellati, F. Botanical Sources, Chemistry, Analysis, and Biological Activity of Furanocoumarins of Pharmaceutical Interest. Molecules 2019, 24, 2163. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Modi, G.M.; Doherty, C.B.; Katta, R.; Orengo, I.F. Irritant contact dermatitis from plants. Dermatitis 2009, 20, 63–78. [Google Scholar] [CrossRef]
- Otang, W.M.; Grierson, D.S.; Afolayan, A.J. A survey of plants responsible for causing irritant contact dermatitis in the Amathole district, Eastern Cape, South Africa. J. Ethnopharmacol. 2014, 157, 274–284. [Google Scholar] [CrossRef]
- Rozas-Munoz, E.; Lepoittevin, J.P.; Pujol, R.M.; Gimenez-Arnau, A. Allergic contact dermatitis to plants: Understanding the chemistry will help our diagnostic approach. Actas Dermosifiliogr. 2012, 103, 456–477. [Google Scholar] [CrossRef]
- Jack, A.R.; Norris, P.L.; Storrs, F.J. Allergic contact dermatitis to plant extracts in cosmetics. Semin. Cutan. Med. Surg. 2013, 32, 140–146. [Google Scholar] [CrossRef]
- Quinn, J.C.; Kessell, A.; Weston, L.A. Secondary plant products causing photosensitization in grazing herbivores: Their structure, activity and regulation. Int. J. Mol. Sci. 2014, 15, 1441–1465. [Google Scholar] [CrossRef]
- Vinceti, M.; Wei, E.T.; Malagoli, C.; Bergomi, M.; Vivoli, G. Adverse health effects of selenium in humans. Rev. Environ. Health 2001, 16, 233–251. [Google Scholar] [CrossRef] [PubMed]
- Koller, L.D.; Exon, J.H.; Talcott, P.A.; Osborne, C.A.; Henningsen, G.M. Immune responses in rats supplemented with selenium. Clin. Exp. Immunol. 1986, 63, 570–576. [Google Scholar] [PubMed]
- Gau, R.J.; Yang, H.L.; Chow, S.N.; Suen, J.L.; Lu, F.J. Humic acid suppresses the LPS-induced expression of cell-surface adhesion proteins through the inhibition of NF-kappaB activation. Toxicol. Appl. Pharmacol. 2000, 166, 59–67. [Google Scholar] [CrossRef] [PubMed]
- Hseu, Y.C.; Lu, F.J.; Engelking, L.R.; Chen, C.L.; Chen, Y.H.; Yang, H.L. Humic acid-induced echinocyte transformation in human erythrocytes: Characterization of morphological changes and determination of the mechanism underlying damage. J. Toxicol. Environ. Health A 2000, 60, 215–230. [Google Scholar] [CrossRef]
- Alija, A.J.; Bresgen, N.; Sommerburg, O.; Langhans, C.D.; Siems, W.; Eckl, P.M. Cyto- and genotoxic potential of beta-carotene and cleavage products under oxidative stress. Biofactors 2005, 24, 159–163. [Google Scholar] [CrossRef] [PubMed]
- Virtamo, J.; Taylor, P.R.; Kontto, J.; Männistö, S.; Utriainen, M.; Weinstein, S.J.; Huttunen, J.; Albanes, D. Effects of α-tocopherol and β-carotene supplementation on cancer incidence and mortality: 18-year postintervention follow-up of the Alpha-tocopherol, Beta-carotene Cancer Prevention Study. Int. J. Cancer 2014, 135, 178–185. [Google Scholar] [CrossRef] [Green Version]
- Alija, A.; Bresgen, N.; Langhans, C.D.; Siems, W.; Sommerburg, O.; Eckl, P. β-carotene under oxidative stress induces genotoxicity. Research2019 2020, 24, 107–122. [Google Scholar]
- Soto-Blanco, B. Chapter 12—Herbal glycosides in healthcare. In Herbal Biomolecules in Healthcare Applications; Mandal, S.C., Nayak, A.K., Dhara, A.K., Eds.; Academic Press: Cambridge, MA, USA, 2022; pp. 239–282. [Google Scholar] [CrossRef]
- Peixoto, H.; Roxo, M.; Röhrig, T.; Richling, E.; Wang, X.; Wink, M. Anti-Aging and Antioxidant Potential of Paullinia cupana var. sorbilis: Findings in Caenorhabditis elegans Indicate a New Utilization for Roasted Seeds of Guarana. Medicines 2017, 4, 61. [Google Scholar] [CrossRef]
- Triana-Martínez, F.; Picallos-Rabina, P.; Da Silva-Álvarez, S.; Pietrocola, F.; Llanos, S.; Rodilla, V.; Soprano, E.; Pedrosa, P.; Ferreirós, A.; Barradas, M.; et al. Identification and characterization of Cardiac Glycosides as senolytic compounds. Nat. Commun. 2019, 10, 4731. [Google Scholar] [CrossRef]
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations. |
© 2022 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
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. https://doi.org/10.3390/molecules27207084
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(20):7084. https://doi.org/10.3390/molecules27207084
Chicago/Turabian StyleBjørklund, Geir, Mariia Shanaida, Roman Lysiuk, Monica Butnariu, Massimiliano Peana, Ioan Sarac, Oksana Strus, Kateryna Smetanina, and Salvatore Chirumbolo. 2022. "Natural Compounds and Products from an Anti-Aging Perspective" Molecules 27, no. 20: 7084. https://doi.org/10.3390/molecules27207084