Hostname: page-component-f554764f5-44mx8 Total loading time: 0 Render date: 2025-04-21T20:13:36.750Z Has data issue: false hasContentIssue false

Anti-ageing natural supplements: the main players in promoting healthy lifespan

Published online by Cambridge University Press:  21 November 2024

Mehran Izadi
Affiliation:
Department of Infectious and Tropical Diseases, School of Medicine, Tehran University of Medical Sciences, Tehran, Iran Synapse Laboratory Diagnostic Technologies Accelerator, Tehran, Iran Department of Research & Technology, Zeenome Longevity Research Institute, Tehran, Iran
Nariman Sadri
Affiliation:
Synapse Laboratory Diagnostic Technologies Accelerator, Tehran, Iran Department of Research & Technology, Zeenome Longevity Research Institute, Tehran, Iran School of Medicine, Shahid beheshti University of Medical Sciences, Tehran, Iran
Amirhossein Abdi
Affiliation:
Synapse Laboratory Diagnostic Technologies Accelerator, Tehran, Iran Department of Research & Technology, Zeenome Longevity Research Institute, Tehran, Iran School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
Mohammad Mahdi Raeis Zadeh
Affiliation:
Synapse Laboratory Diagnostic Technologies Accelerator, Tehran, Iran Department of Research & Technology, Zeenome Longevity Research Institute, Tehran, Iran School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
Dorsa Jalaei
Affiliation:
Synapse Laboratory Diagnostic Technologies Accelerator, Tehran, Iran Department of Research & Technology, Zeenome Longevity Research Institute, Tehran, Iran School of Pharmacy, Zanjan University of Medical Sciences, Zanjan, Iran
Mohammad Mahdi Ghazimoradi
Affiliation:
Synapse Laboratory Diagnostic Technologies Accelerator, Tehran, Iran Department of Research & Technology, Zeenome Longevity Research Institute, Tehran, Iran School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
Sara Shouri
Affiliation:
Synapse Laboratory Diagnostic Technologies Accelerator, Tehran, Iran Department of Research & Technology, Zeenome Longevity Research Institute, Tehran, Iran School of Pharmacy, Tehran University of Medical Sciences, Tehran, Iran
Safa Tahmasebi*
Affiliation:
Synapse Laboratory Diagnostic Technologies Accelerator, Tehran, Iran Department of Research & Technology, Zeenome Longevity Research Institute, Tehran, Iran Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Iran
*
Corresponding author: Safa Tahmasebi; Email: Safa.tahmasebi@sbmu.ac.ir

Abstract

Ageing is an inevitable biological process accompanied by various physiological changes, and researchers have long sought interventions to promote healthy ageing. This article explores the effects of four natural compounds – omega-3 fatty acids, coenzyme Q10, gingerol and curcumin – on the ageing process. We delve into the scientific literature to examine the potential benefits and mechanisms behind these substances in mitigating age-related conditions. Omega-3’s anti-inflammatory properties, coenzyme Q10’s cellular energy support, gingerol’s antioxidant effects and curcumin’s anti-ageing properties are discussed. By shedding light on the impact of these compounds, this review aims to contribute to a better understanding of how natural substances may play a role in promoting longevity and enhancing the quality of life during the ageing journey.

Type
Review Article
Copyright
© The Author(s), 2024. Published by Cambridge University Press on behalf of The Nutrition Society

Access options

Get access to the full version of this content by using one of the access options below. (Log in options will check for institutional or personal access. Content may require purchase if you do not have access.)

Article purchase

Temporarily unavailable

References

Dziechciaż, M & Filip, R (2014) Biological psychological and social determinants of old age: bio-psycho-social aspects of human aging. Ann Agric Environ Med: AAEM 21, 835838.Google Scholar
Farr, JN & Almeida, M (2018) The spectrum of fundamental basic science discoveries contributing to organismal aging. J Bone Miner Res 33, 15681584.Google Scholar
Sourada, L & Kuglík, P (2020) Genetic mechanisms of aging. Cas Lek Cesk 159, 8187.Google Scholar
Khrapko, K & Turnbull, D (2014) Mitochondrial DNA mutations in aging. Prog Mol Biol Transl Sci 127, 2962.Google Scholar
Fathi, E, Charoudeh, HN, Sanaat, Z & Farahzadi, R (2019) Telomere shortening as a hallmark of stem cell senescence. Stem Cell Investig 6, 7.Google Scholar
Turner, KJ, Vasu, V & Griffin, DK (2019) Telomere biology and human phenotype. Cells 8, 73. doi: 10.3390/cells8010073.Google Scholar
Jiang, F, Xu, XR, Li, WM, Xia, K, Wang, LF & Yang, XC (2020) Monotropein alleviates H2O2-induced inflammation, oxidative stress and apoptosis via NF-κB/AP-1 signaling. Mol Med Rep 22, 48284836.Google Scholar
Coluzzi, E, Leone, S & Sgura, A (2019) Oxidative stress induces telomere dysfunction and senescence by replication fork arrest. Cells 8, 19.Google Scholar
Lundgren, CA, Sjöstrand, D, Biner, O, Bennett, M, Rudling, A, Johansson, A-L, et al. (2018) Scavenging of superoxide by a membrane-bound superoxide oxidase. Nat Chem Biol 14, 788793.Google Scholar
Labunskyy, VM & Gladyshev, VN (2013) Role of reactive oxygen species-mediated signaling in aging. Antioxid Redox Signal 19, 13621372.Google Scholar
Giorgi, C, Marchi, S, Simoes, IC, Ren, Z, Morciano, G, Perrone, M, et al. (2018) Mitochondria and reactive oxygen species in aging and age-related diseases. Int Rev Cell Mol Biol 340, 209344.Google Scholar
Franceschi, C & Campisi, J (2014) Chronic inflammation (inflammaging) and its potential contribution to age-associated diseases. J Gerontol Ser A: Biomed Sci Med Sci 69, Suppl_1, S4S9.Google Scholar
Akazawa, H (2017) Aging and homeostasis. Chronic inflammation and aging. Clin Calcium 27, 963968.Google Scholar
Harris, SE, Riggio, V, Evenden, L, Gilchrist, T, McCafferty, S, Murphy, L, et al. (2017) Age-related gene expression changes, and transcriptome wide association study of physical and cognitive aging traits, in the Lothian Birth Cohort 1936. Aging 9, 2489.Google Scholar
Cuanalo-Contreras, K, Mukherjee, A & Soto, C (2013) Role of protein misfolding and proteostasis deficiency in protein misfolding diseases and aging. Int J Cell Biol 2013, 638083. doi: 10.1155/2013/638083.Google Scholar
Forloni, G, Terreni, L, Bertani, I, Fogliarino, S, Invernizzi, R, Assini, A, et al. (2002) Protein misfolding in Alzheimer’s and Parkinson’s disease: genetics and molecular mechanisms. Neurobiol Aging 23, 957976.Google Scholar
Copinschi, G & Caufriez, A (2013) Sleep and hormonal changes in aging. Endocrinol Metab Clin 42, 371389.Google Scholar
Sánchez Macarro, M, Martínez Rodríguez, JP, Bernal Morell, E, Pérez-Piñero, S, Victoria-Montesinos, D, García-Muñoz, AM, et al. (2020) Effect of a combination of citrus flavones and flavanones and olive polyphenols for the reduction of cardiovascular disease risk: an exploratory randomized, double-blind, placebo-controlled study in healthy subjects. Nutrients 12, 1475. doi: 10.3390/nu12051475.Google Scholar
Tajaldini, M, Samadi, F, Khosravi, A, Ghasemnejad, A & Asadi, J (2020) Protective and anticancer effects of orange peel extract and naringin in doxorubicin treated esophageal cancer stem cell xenograft tumor mouse model. Biomed Pharmacother 121, 109594.Google Scholar
Asgharian, P, Tazekand, AP, Hosseini, K, Forouhandeh, H, Ghasemnejad, T, Ranjbar, M, et al. (2022) Potential mechanisms of quercetin in cancer prevention: focus on cellular and molecular targets. Cancer Cell Int 22, 257.Google Scholar
Abdolmohammadi, K, Mahmoudi, T, Alimohammadi, M, Tahmasebi, S, Zavvar, M & Hashemi, SM (2023) Mesenchymal stem cell-based therapy as a new therapeutic approach for acute inflammation. Life Sci 312, 121206.Google Scholar
Jafari, D, Mousavi, MJ, Keshavarz Shahbaz, S, Jafarzadeh, L, Tahmasebi, S, Spoor, J, et al. (2021) E3 ubiquitin ligase Casitas B lineage lymphoma-b and its potential therapeutic implications for immunotherapy. Clin Exp Immunol 204, 1431.Google Scholar
Marofi, F, Azizi, R, Motavalli, R, Vahedi, G, Nasimi, M, Yousefi, M, et al. (2021) COVID-19: our current knowledge of epidemiology, pathology, therapeutic approaches, and diagnostic methods. Anti-Cancer Agents Med Chem 21, 21422162.Google Scholar
Zare Rafie, M, Esmaeilzadeh, A, Ghoreishi, A, Tahmasebi, S, Faghihzadeh, E, Elahi, R (2021) IL-38 as an early predictor of the ischemic stroke prognosis. Cytokine 146, 155626.Google Scholar
Liu, RH (2003) Health benefits of fruit and vegetables are from additive and synergistic combinations of phytochemicals. Am J Clin Nutr 78, 3 Suppl, 517S520S.Google Scholar
Tressera-Rimbau, A, Arranz, S, Eder, M & Vallverdú-Queralt, A (2017) Dietary polyphenols in the prevention of stroke. Oxid Med Cell Longy 2017, 7467962.Google Scholar
Calder, PC & Grimble, RF (2002) Polyunsaturated fatty acids, inflammation and immunity. Eur J Clin Nutr 56, Suppl 3, S14S19.Google Scholar
Ander, BP, Dupasquier, CM, Prociuk, MA & Pierce, GN (2003) Polyunsaturated fatty acids and their effects on cardiovascular disease. Exp Clin Cardiol 8, 164172.Google Scholar
Endo, J & Arita, M (2016) Cardioprotective mechanism of omega-3 polyunsaturated fatty acids. J Cardiol 67, 2227.Google Scholar
Paixão, EMdS, Oliveira, ACdM, Pizato, N, Muniz-Junqueira, MI, Magalhães, KG, Nakano, EY, et al. (2017) The effects of EPA and DHA enriched fish oil on nutritional and immunological markers of treatment naïve breast cancer patients: a randomized double-blind controlled trial. Nutr J 16, 111.Google Scholar
Chagas, TR, Borges, D, de Oliveira, PF, Mocellin, MC, Barbosa, AM, Camargo, C, et al. (2017) Oral fish oil positively influences nutritional-inflammatory risk in patients with haematological malignancies during chemotherapy with an impact on long-term survival: a randomised clinical trial. J Human Nutr Diet 30, 681692.Google Scholar
Simopoulos, AP (2002) The importance of the ratio of omega-6/omega-3 essential fatty acids. Biomed Pharmacother 56, 365379.Google Scholar
Su, KP, Tseng, PT, Lin, PY, Okubo, R, Chen, TY, Chen, YW, et al. (2018) Association of use of omega-3 polyunsaturated fatty acids with changes in severity of anxiety symptoms: a systematic review and meta-analysis. JAMA Netw Open 1, e182327.Google Scholar
Brigham, EP, Woo, H, McCormack, M, Rice, J, Koehler, K, Vulcain, T, et al. (2019) Omega-3 and omega-6 intake modifies asthma severity and response to indoor air pollution in children. Am J Respir Crit Care Med 199, 14781486.Google Scholar
Dighriri, IM, Alsubaie, AM, Hakami, FM, Hamithi, DM, Alshekh, MM, Khobrani, FA, et al. (2022) Effects of omega-3 polyunsaturated fatty acids on brain functions: a systematic review. Cureus 14, e30091.Google Scholar
Welty, FK (2023) Omega-3 fatty acids and cognitive function. Curr Opin Lipidol 34, 1221.Google Scholar
Wei, BZ, Li, L, Dong, CW, Tan, CC & Xu, W (2023) The relationship of omega-3 fatty acids with dementia and cognitive decline: evidence from prospective cohort studies of supplementation, dietary intake, and blood markers. Am J Clin Nutr 117, 10961109.Google Scholar
Krupa, K, Fritz, K & Parmar, M (2023) Omega-3 fatty acids. StatPearls. Treasure Island, FL: StatPearls Publishing. Copyright ©, StatPearls Publishing LLC.Google Scholar
Jacobsen, C (2010) Enrichment of foods with omega-3 fatty acids: a multidisciplinary challenge. Ann N Y Acad Sci 1190, 141150. doi: 10.1111/j.1749-6632.2009.05263.x.Google Scholar
Shahidi, F & Ambigaipalan, P (2018) Omega-3 polyunsaturated fatty acids and their health benefits. Annu Rev Food Sci Technol 9, 345381.Google Scholar
Ito, MK (2015) A comparative overview of prescription omega-3 fatty acid products. P T 40, 826857.Google Scholar
Schuchardt, JP & Hahn, A (2013) Bioavailability of long-chain omega-3 fatty acids. Prostaglandins Leukot Essent Fatty Acids 89, 18.Google Scholar
Innis, SM (2014) Omega-3 fatty acid biochemistry: perspectives from human nutrition. Mil Med 179, 11 Suppl, 8287.Google Scholar
Yang, X, Yi, X, Zhang, F, Li, F, Lang, L, Ling, M, et al. (2022) Cytochrome P450 epoxygenase-derived EPA and DHA oxylipins 17,18-epoxyeicosatetraenoic acid and 19,20-epoxydocosapentaenoic acid promote BAT thermogenesis and WAT browning through the GPR120-AMPKα signaling pathway. Food Funct 13, 12321245.Google Scholar
Cholewski, M, Tomczykowa, M & Tomczyk, M (2018) A comprehensive review of chemistry, sources and bioavailability of Omega-3 fatty acids. Nutrients 10, 1662. doi: 10.3390/nu10111662.Google Scholar
Maki, KC & Dicklin, MR (2019) Strategies to improve bioavailability of omega-3 fatty acids from ethyl ester concentrates. Curr Opin Clin Nutr Metab Care 22, 116123.Google Scholar
Vannice, G & Rasmussen, H (2014) Position of the academy of nutrition and dietetics: dietary fatty acids for healthy adults. J Acad Nutr Diet 114, 136153.Google Scholar
EFSA (2012) Panel on Dietetic Products N, Allergies. Scientific Opinion on the Tolerable Upper Intake Level of eicosapentaenoic acid (EPA), docosahexaenoic acid (DHA) and docosapentaenoic acid (DPA). EFSA J 10, 2815.Google Scholar
Medicine Io (2005) Dietary reference intakes for energy, carbohydrate, fiber, fat, fatty acids, cholesterol, protein, and amino acids. Washington, DC: The National Academies Press, 1358.Google Scholar
McDonnell, SL, French, CB, Baggerly, CA & Harris, WS (2019) Cross-sectional study of the combined associations of dietary and supplemental eicosapentaenoic acid + docosahexaenoic acid on Omega-3 Index. Nutr Res 71, 4355.Google Scholar
Zhang, Z, Fulgoni, VL, Kris-Etherton, PM & Mitmesser, SH (2018) Dietary intakes of EPA and DHA omega-3 fatty acids among US childbearing-age and pregnant women: an analysis of NHANES 2001–2014. Nutrients 10, 416. doi: 10.3390/nu10040416.Google Scholar
Mazereeuw, G, Lanctôt, KL, Chau, SA, Swardfager, W & Herrmann, N (2012) Effects of ω-3 fatty acids on cognitive performance: a meta-analysis. Neurobiol Aging 33, 1482.e171482.e29.Google Scholar
Lev-Tzion, R, Griffiths, AM, Leder, O & Turner, D (2014) Omega 3 fatty acids (fish oil) for maintenance of remission in Crohn’s disease. Cochrane Database Syst Rev 2014, Cd006320.Google Scholar
Bjerve, KS, Fischer, S, Wammer, F & Egeland, T (1989) alpha-Linolenic acid and long-chain omega-3 fatty acid supplementation in three patients with omega-3 fatty acid deficiency: effect on lymphocyte function, plasma and red cell lipids, and prostanoid formation. Am J Clin Nutr 49, 290300.Google Scholar
Sakamoto, A, Saotome, M, Iguchi, K & Maekawa, Y (2019) Marine-derived omega-3 polyunsaturated fatty acids and heart failure: current understanding for basic to clinical relevance. Int J Mol Sci 20, 4025. doi: 10.3390/ijms20164025.Google Scholar
Calder, PC (2017) Omega-3 fatty acids and inflammatory processes: from molecules to man. Biochem Soc Trans 45, 11051115.Google Scholar
Calder, PC (2013) Omega-3 polyunsaturated fatty acids and inflammatory processes: nutrition or pharmacology? Br J Clin Pharmacol 75, 645662.Google Scholar
Krupa, K, Fritz, K & Parmar, M (2023) Omega-3 fatty acids. [Updated 2023 Jan 17]. StatPearls [Internet]. Treasure Island, FL: StatPearls Publishing; 2023 Jan-: NCBI.Google Scholar
Calder, PC (2015) Marine omega-3 fatty acids and inflammatory processes: effects, mechanisms and clinical relevance. Biochim Biophys Acta (BBA) – Mol Cell Biol Lipids 1851, 469484.Google Scholar
Sethi, S, Ziouzenkova, O, Ni, H, Wagner, DD, Plutzky, J & Mayadas, TN (2002) Oxidized omega-3 fatty acids in fish oil inhibit leukocyte-endothelial interactions through activation of PPAR alpha. Blood 100, 13401346.Google Scholar
Calder, PC (2020) n-3 PUFA and inflammation: from membrane to nucleus and from bench to bedside. Proc Nutr Soc, 113. doi: 10.1017/S0029665120007077.Google Scholar
Nielsen, AA, Jørgensen, LG, Nielsen, JN, Eivindson, M, Grønbaek, H, Vind, I, et al. (2005) Omega-3 fatty acids inhibit an increase of proinflammatory cytokines in patients with active Crohn’s disease compared with omega-6 fatty acids. Aliment Pharmacol Ther 22, 11211128.Google Scholar
Serhan, CN & Levy, BD (2018) Resolvins in inflammation: emergence of the pro-resolving superfamily of mediators. J Clin Invest 128, 26572669.Google Scholar
Wellhauser, L & Belsham, DD (2014) Activation of the omega-3 fatty acid receptor GPR120 mediates anti-inflammatory actions in immortalized hypothalamic neurons. J Neuroinflamm 11, 60.Google Scholar
Hashimoto, M & Hossain, S (2018) Fatty acids: from membrane ingredients to signaling molecules. In: Viduranga, W (ed.), Biochemistry and health benefits of fatty acids. Rijeka: IntechOpen, pp. 120.Google Scholar
Calder, PC (2016) Docosahexaenoic acid. Ann Nutr Metab 69, Suppl. 1, 821.Google Scholar
de Carvalho, C & Caramujo, MJ (2018) The various roles of fatty acids. Molecules 23, 2583. doi: 10.3390/molecules23102583.Google Scholar
Wood, AHR, Chappell, HF & Zulyniak, MA (2022) Dietary and supplemental long-chain omega-3 fatty acids as moderators of cognitive impairment and Alzheimer’s disease. Eur J Nutr 61, 589604.Google Scholar
Sala-Vila, A, Satizabal, CL, Tintle, N, Melo van Lent, D, Vasan, RS, Beiser, AS, et al. (2022) Red blood cell DHA is inversely associated with risk of incident Alzheimer’s disease and all-cause dementia: Framingham offspring study. Nutrients 14, 2408. doi: 10.3390/nu14122408.Google Scholar
Tully, AM, Roche, HM, Doyle, R, Fallon, C, Bruce, I, Lawlor, B, et al. (2003) Low serum cholesteryl ester-docosahexaenoic acid levels in Alzheimer’s disease: a case-control study. Br J Nutr 89, 483489.Google Scholar
Whalley, LJ, Deary, IJ, Starr, JM, Wahle, KW, Rance, KA, Bourne, VJ, et al. (2008) n-3 Fatty acid erythrocyte membrane content, APOE varepsilon4, and cognitive variation: an observational follow-up study in late adulthood. Am J Clin Nutr 87, 449454.Google Scholar
Phillips, MA, Childs, CE, Calder, PC, Rogers, PJ (2015) No effect of omega-3 fatty acid supplementation on cognition and mood in individuals with cognitive impairment and probable Alzheimer’s disease: a randomised controlled trial. Int J Mol Sci 16, 2460024613.Google Scholar
van de Rest, O, Wang, Y, Barnes, LL, Tangney, C, Bennett, DA & Morris, MC (2016) APOE ε4 and the associations of seafood and long-chain omega-3 fatty acids with cognitive decline. Neurology 86, 20632070.Google Scholar
Li, L, Xu, W, Tan, CC, Cao, XP, Wei, BZ, Dong, CW, et al. (2022) A gene-environment interplay between omega-3 supplementation and APOE ε4 provides insights for Alzheimer’s disease precise prevention amongst high-genetic-risk population. Eur J Neurol 29, 422431.Google Scholar
Jeong, W, Lee, H, Cho, S & Seo, J (2019) ApoE4-induced cholesterol dysregulation and its brain cell type-specific implications in the pathogenesis of Alzheimer’s disease. Mol Cells 42, 739746.Google Scholar
Beydoun, MA, Kaufman, JS, Satia, JA, Rosamond, W & Folsom, AR (2007) Plasma n-3 fatty acids and the risk of cognitive decline in older adults: the Atherosclerosis Risk in Communities Study. Am J Clin Nutr 85, 11031111.Google Scholar
Barberger-Gateau, P, Samieri, C, Féart, C & Plourde, M (2011) Dietary omega 3 polyunsaturated fatty acids and Alzheimer’s disease: interaction with apolipoprotein E genotype. Curr Alzheimer Res 8, 479491.Google Scholar
Qi, W, Gutierrez, GE, Gao, X, Dixon, H, McDonough, JA, Marini, AM, et al. (2017) The ω-3 fatty acid α-linolenic acid extends Caenorhabditis elegans lifespan via NHR-49/PPARα and oxidation to oxylipins. Aging Cell 16, 11251135.Google Scholar
Mora, I, Pérez-Santamaria, A, Tortajada-Pérez, J, Vázquez-Manrique, RP, Arola, L & Puiggròs, F (2023) Structured docosahexaenoic acid (DHA) enhances motility and promotes the antioxidant capacity of aged C. elegans . Cells 12, 1932. doi: 10.3390/cells12151932.Google Scholar
Champigny, CM, Cormier, RPJ, Simard, CJ, St-Coeur, PD, Fortin, S & Pichaud, N (2018) Omega-3 monoacylglyceride effects on longevity, mitochondrial metabolism and oxidative stress: insights from Drosophila melanogaster . Mar Drugs 16, 453. doi: 10.3390/md16110453.Google Scholar
Saini, A, Sharples, AP, Al-Shanti, N & Stewart, CE (2017) Omega-3 fatty acid EPA improves regenerative capacity of mouse skeletal muscle cells exposed to saturated fat and inflammation. Biogerontology 18, 109129.Google Scholar
Su, HM (2010) Mechanisms of n-3 fatty acid-mediated development and maintenance of learning memory performance. J Nutr Biochem 21, 364373.Google Scholar
Jolly, CA, Muthukumar, A, Avula, CP, Troyer, D & Fernandes, G (2001) Life span is prolonged in food-restricted autoimmune-prone (NZB x NZW)F(1) mice fed a diet enriched with (n-3) fatty acids. J Nutr 131, 27532760.Google Scholar
Fernandes, G (2008) Progress in nutritional immunology. Immunol Res 40, 244261.Google Scholar
Halade, GV, Williams, PJ, Lindsey, ML & Fernandes, G (2011) Fish oil decreases inflammation and reduces cardiac remodeling in rosiglitazone treated aging mice. Pharmacol Res 63, 300307.Google Scholar
Dyall, SC, Michael, GJ, Michael-Titus, AT (2010) Omega-3 fatty acids reverse age-related decreases in nuclear receptors and increase neurogenesis in old rats. J Neurosci Res 88, 20912102.Google Scholar
Qureshi, AW, Altamimy, R, El Habhab, A, El Itawi, H, Farooq, MA, Zobairi, F, et al. (2020) Ageing enhances the shedding of splenocyte microvesicles with endothelial pro-senescent effect that is prevented by a short-term intake of omega-3 PUFA EPA:DHA 6:1. Biochem Pharmacol 173, 113734.Google Scholar
Dyall, SC, Michael, GJ, Whelpton, R, Scott, AG, Michael-Titus, AT (2007) Dietary enrichment with omega-3 polyunsaturated fatty acids reverses age-related decreases in the GluR2 and NR2B glutamate receptor subunits in rat forebrain. Neurobiol Aging 28, 424439.Google Scholar
Razgonova, MP, Zakharenko, AM, Golokhvast, KS, Thanasoula, M, Sarandi, E, Nikolouzakis, K, et al. (2020) Telomerase and telomeres in aging theory and chronographic aging theory (Review). Mol Med Rep 22, 16791694.Google Scholar
Epel, ES, Blackburn, EH, Lin, J, Dhabhar, FS, Adler, NE, Morrow, JD, et al. (2004) Accelerated telomere shortening in response to life stress. Proc Natl Acad Sci USA 101, 1731217315.Google Scholar
Chen, J, Wei, Y, Chen, X, Jiao, J & Zhang, Y (2017) Polyunsaturated fatty acids ameliorate aging via redox-telomere-antioncogene axis. Oncotarget 8, 7301.Google Scholar
Ogłuszka, M, Te Pas, MFW, Poławska, E, Nawrocka, A, Stepanow, K & Pierzchała, M (2020) Omega-3 Alpha-Linolenic fatty acid affects the level of telomere binding protein TRF1 in porcine skeletal muscle. Animals (Basel) 10, 1090. doi: 10.3390/ani10061090.Google Scholar
Varela-Lopez, A, Pérez-López, MP, Ramirez-Tortosa, CL, Battino, M, Granados-Principal, S, del Carmen Ramirez-Tortosa, M, et al. (2018) Gene pathways associated with mitochondrial function, oxidative stress and telomere length are differentially expressed in the liver of rats fed lifelong on virgin olive, sunflower or fish oils. J Nutr Biochem 52, 3644.Google Scholar
Gao, J, Xiao, H, Li, J, Guo, X, Cai, W & Li, D (2019) N-3 polyunsaturated fatty acids decrease long-term diabetic risk of offspring of gestational diabetes rats by postponing shortening of hepatic telomeres and modulating liver metabolism. Nutrients 11, 1699.Google Scholar
Chen, J, Wei, Y, Chen, X, Jiao, J & Zhang, Y (2016) Polyunsaturated fatty acids ameliorate aging via redox-telomere-antioncogene axis. Oncotarget 8, 73017314. doi: 10.18632/oncotarget.14236.Google Scholar
Ogłuszka, M, Lipiński, P & Starzyński, RR (2022) Effect of Omega-3 fatty acids on telomeres-are they the elixir of youth? Nutrients 14, 3723. doi: 10.3390/nu14183723.Google Scholar
Farzaneh-Far, R, Lin, J, Epel, ES, Harris, WS, Blackburn, EH & Whooley, MA (2010) Association of marine omega-3 fatty acid levels with telomeric aging in patients with coronary heart disease. JAMA 303, 250257.Google Scholar
Liu, X, Shi, Q, Fan, X, Chen, H, Chen, N, Zhao, Y, et al. (2022) Associations of maternal polyunsaturated fatty acids with telomere length in the cord blood and placenta in Chinese population. Front Nutr 8, 779306.Google Scholar
Kiecolt-Glaser, JK, Epel, ES, Belury, MA, Andridge, R, Lin, J, Glaser, R, et al. (2013) Omega-3 fatty acids, oxidative stress, and leukocyte telomere length: a randomized controlled trial. Brain, Behav Immun 28, 1624.Google Scholar
Barden, A, O’Callaghan, N, Burke, V, Mas, E, Beilin, LJ, Fenech, M, et al. (2016) n-3 fatty acid supplementation and leukocyte telomere length in patients with chronic kidney disease. Nutrients 8, 175.Google Scholar
O’Callaghan, N, Parletta, N, Milte, CM, Benassi-Evans, B, Fenech, M, Howe, PR (2014) Telomere shortening in elderly individuals with mild cognitive impairment may be attenuated with ω-3 fatty acid supplementation: a randomized controlled pilot study. Nutrition 30, 489491.Google Scholar
Madison, AA, Belury, MA, Andridge, R, Renna, ME, Rosie Shrout, M, Malarkey, WB, et al. (2021) Omega-3 supplementation and stress reactivity of cellular aging biomarkers: an ancillary substudy of a randomized, controlled trial in midlife adults. Mol Psychiatry 26, 30343042.Google Scholar
Lai, HT, de Oliveira Otto, MC, Lemaitre, RN, McKnight, B, Song, X, King, IB, et al. (2018) Serial circulating omega 3 polyunsaturated fatty acids and healthy ageing among older adults in the Cardiovascular Health Study: prospective cohort study. BMJ 363, k4067.Google Scholar
Harris, WS, Tintle, NL, Imamura, F, Qian, F, Korat, AVA, Marklund, M, et al. (2021) Blood n-3 fatty acid levels and total and cause-specific mortality from 17 prospective studies. Nat Commun 12, 2329.Google Scholar
Mozaffarian, D, Lemaitre, RN, King, IB, Song, X, Huang, H, Sacks, FM, et al. (2013) Plasma phospholipid long-chain ω-3 fatty acids and total and cause-specific mortality in older adults: a cohort study. Ann Intern Med 158, 515525.Google Scholar
Abbatecola, AM, Cherubini, A, Guralnik, JM, Andres Lacueva, C, Ruggiero, C, Maggio, M, et al. (2009) Plasma polyunsaturated fatty acids and age-related physical performance decline. Rejuvenation Res 12, 2532.Google Scholar
Kuszewski, JC, Howe, PR & Wong, RH (2020) Evaluation of cognitive performance following fish-oil and curcumin supplementation in middle-aged and older adults with overweight or obesity. J Nutr 150, 31903199.Google Scholar
Malik, A, Ramadan, A, Vemuri, B, Siddiq, W, Amangurbanova, M, Ali, A, et al. (2021) ω-3 Ethyl ester results in better cognitive function at 12 and 30 months than control in cognitively healthy subjects with coronary artery disease: a secondary analysis of a randomized clinical trial. Am J Clin Nutr 113, 11681176.Google Scholar
Gillies, D, Leach, MJ & Algorta, GP (2023) Polyunsaturated fatty acids (PUFA) for attention deficit hyperactivity disorder (ADHD) in children and adolescents. Cochrane Database Syst Rev 4, CD007986. doi: 10.1002/14651858.Google Scholar
Yurko-Mauro, K, McCarthy, D, Rom, D, Nelson, EB, Ryan, AS, Blackwell, A, et al. (2010) Beneficial effects of docosahexaenoic acid on cognition in age-related cognitive decline. Alzheimers Dement 6, 456464.Google Scholar
Iroku-Malize, T & Kirsch, S (2016) Eye conditions in older adults: age-related macular degeneration. FP Essent 445, 2428.Google Scholar
Christen, WG, Schaumberg, DA, Glynn, RJ & Buring, JE (2011) Dietary ω-3 fatty acid and fish intake and incident age-related macular degeneration in women. Arch Ophthalmol 129, 921929.Google Scholar
Augood, C, Chakravarthy, U, Young, I, Vioque, J, de Jong, PT, Bentham, G, et al. (2008) Oily fish consumption, dietary docosahexaenoic acid and eicosapentaenoic acid intakes, and associations with neovascular age-related macular degeneration. Am J Clin Nutr 88, 398406.Google Scholar
Seddon, JM, George, S, Rosner, B (2006) Cigarette smoking, fish consumption, omega-3 fatty acid intake, and associations with age-related macular degeneration: the US Twin Study of age-related macular degeneration. Arch Ophthalmol 124, 9951001.Google Scholar
Skulas-Ray, AC, Wilson, PWF, Harris, WS, Brinton, EA, Kris-Etherton, PM, Richter, CK, et al. (2019) Omega-3 fatty acids for the management of hypertriglyceridemia: a science advisory from the American Heart Association. Circulation 140, e673e691.Google Scholar
Laredj, LN, Licitra, F & Puccio, HM (2014) The molecular genetics of coenzyme Q biosynthesis in health and disease. Biochimie 100, 7887.Google Scholar
Gutierrez-Mariscal, FM, Yubero-Serrano, EM, Villalba, JM, Lopez-Miranda, J (2019) Coenzyme Q(10): From bench to clinic in aging diseases, a translational review. Crit Rev Food Sci Nutr 59, 22402257.Google Scholar
Maciejewska-Stupska, K, Czarnecka, K & Szymański, P (2024) Bioavailability enhancement of coenzyme Q(10): An update of novel approaches. Arch Pharmazie 357, e2300676.Google Scholar
Garrido-Maraver, J, Cordero, MD, Oropesa-Avila, M, Vega, AF, de la Mata, M, Pavon, AD, et al. (2014) Clinical applications of coenzyme Q10. Front Biosci 19, 619633.Google Scholar
Zozina, VI, Covantev, S, Goroshko, OA, Krasnykh, LM & Kukes, VG (2018) Coenzyme Q10 in cardiovascular and metabolic diseases: current state of the problem. Curr Cardiol Rev 14, 164174.Google Scholar
Lenaz, G, Fato, R, Di Bernardo, S, Jarreta, D, Costa, A, Genova, ML, et al. (1999) Localization and mobility of coenzyme Q in lipid bilayers and membranes. BioFactors 9, 8793.Google Scholar
Olson, RE & Rudney, H (1983) Biosynthesis of ubiquinone. Vitam Hormones 40, 143.Google Scholar
Alcázar-Fabra, M, Navas, P & Brea-Calvo, G (2016) Coenzyme Q biosynthesis and its role in the respiratory chain structure. Biochim Biophys Acta 1857, 10731078.Google Scholar
Lenaz, G, Fato, R, Castelluccio, C, Genova, ML, Bovina, C, Estornell, E, et al. (1993) The function of coenzyme Q in mitochondria. Clin Invest 71, 8 Suppl, S66S70.Google Scholar
Harman, D (1972) The biologic clock: the mitochondria? J Am Geriatr Soc 20, 145147.Google Scholar
Eirin, A, Lerman, A & Lerman, LO (2017) The emerging role of mitochondrial targeting in kidney disease. Handb Exp Pharmacol 240, 229250. doi: 10.1007/164_2016_6.Google Scholar
Poulose, N & Raju, R (2014) Aging and injury: alterations in cellular energetics and organ function. Aging Dis 5, 101.Google Scholar
Schniertshauer, D, Gebhard, D, van Beek, H, Nöth, V, Schon, J & Bergemann, J (2020) The activity of the DNA repair enzyme hOGG1 can be directly modulated by ubiquinol. DNA Repair 87, 102784.Google Scholar
Mantle, D & Dybring, A (2020) Bioavailability of coenzyme Q10: an overview of the absorption process and subsequent metabolism. Antioxidants 9, 386.Google Scholar
Olivieri, F, Lazzarini, R, Babini, L, Prattichizzo, F, Rippo, MR, Tiano, L, et al. (2013) Anti-inflammatory effect of ubiquinol-10 on young and senescent endothelial cells via miR-146a modulation. Free Radical Biol Med 63, 410420.Google Scholar
Fan, L, Feng, Y, Chen, GC, Qin, LQ, Fu, CL & Chen, LH (2017) Effects of coenzyme Q10 supplementation on inflammatory markers: a systematic review and meta-analysis of randomized controlled trials. Pharmacol Res 119, 128136.Google Scholar
Schmelzer, C, Lindner, I, Rimbach, G, Niklowitz, P, Menke, T & Döring, F (2008) Functions of coenzyme Q10 in inflammation and gene expression. BioFactors 32, 179183.Google Scholar
Sharma, A, Fonarow, GC, Butler, J, Ezekowitz, JA & Felker, GM (2016) Coenzyme Q10 and heart failure: a state-of-the-art review. Circ Heart Failure 9, e002639.Google Scholar
Jorat, MV, Tabrizi, R, Mirhosseini, N, Lankarani, KB, Akbari, M, Heydari, ST, et al. (2018) The effects of coenzyme Q10 supplementation on lipid profiles among patients with coronary artery disease: a systematic review and meta-analysis of randomized controlled trials. Lipids Health Dis 17, 230.Google Scholar
Pérez-Sánchez, C, Aguirre, M, Ruiz-Limón, P, Ábalos-Aguilera, MC, Jiménez-Gómez, Y, Arias-de la Rosa, I, et al. (2017) Ubiquinol effects on antiphospholipid syndrome prothrombotic profile: a randomized, placebo-controlled trial. Arterioscler Thromb Vasc Biol 37, 19231932.Google Scholar
Wilcock, DM & Griffin, WS (2013) Down’s syndrome, neuroinflammation, and Alzheimer neuropathogenesis. J Neuroinflamm 10, 84.Google Scholar
Zaki, ME, El-Bassyouni, HT, Tosson, A, Youness, E & Hussein, J (2017) Coenzyme Q10 and pro-inflammatory markers in children with Down syndrome: clinical and biochemical aspects☆. J Pediatr 93, 100104.Google Scholar
Wolters, M & Hahn, A (2003) Plasma ubiquinone status and response to six-month supplementation combined with multivitamins in healthy elderly women – results of a randomized, double-blind, placebo-controlled study. Int J Vitamin Nutr Res 73, 207214.Google Scholar
Kalén, A, Appelkvist, EL & Dallner, G (1989) Age-related changes in the lipid compositions of rat and human tissues. Lipids 24, 579584.Google Scholar
Söderberg, M, Edlund, C, Kristensson, K & Dallner, G (1990) Lipid compositions of different regions of the human brain during aging. J Neurochem 54, 415423.Google Scholar
Edlund, C, Söderberg, M, Kristensson, K, Dallner, G (1992) Ubiquinone, dolichol, and cholesterol metabolism in aging and Alzheimer’s disease. Biochem Cell Biol 70, 422428.Google Scholar
Nagase, M, Yamamoto, Y, Matsumoto, N, Arai, Y & Hirose, N (2018) Increased oxidative stress and coenzyme Q10 deficiency in centenarians. J Clin Biochem Nutr 63, 129136.Google Scholar
Hathcock, JN & Shao, A (2006) Risk assessment for coenzyme Q10 (ubiquinone). Regul Toxicol Pharmacol 45, 282288.Google Scholar
Singletary, K (2010) Ginger: an overview of health benefits. Nutr Today 45, 171183.Google Scholar
Abolaji, AO, Ojo, M, Afolabi, TT, Arowoogun, MD, Nwawolor, D & Farombi, EO (2017) Protective properties of 6-gingerol-rich fraction from Zingiber officinale (ginger) on chlorpyrifos-induced oxidative damage and inflammation in the brain, ovary and uterus of rats. Chem-Biol Interact 270, 1523.Google Scholar
Mohd Sahardi, NFN & Makpol, S (2019) Ginger (Zingiber officinale Roscoe) in the prevention of ageing and degenerative diseases: review of current evidence. Evid-Based Complement Altern Med: eCAM 2019, 5054395.Google Scholar
Ballester, P, Cerdá, B, Arcusa, R, Marhuenda, J, Yamedjeu, K & Zafrilla, P (2022) Effect of ginger on inflammatory diseases. Molecules 27, 7223. doi: 10.3390/molecules27217223.Google Scholar
Kim, SO, Chun, KS, Kundu, JK & Surh, YJ (2004) Inhibitory effects of [6]-gingerol on PMA-induced COX-2 expression and activation of NF-kappaB and p38 MAPK in mouse skin. BioFactors 21, 2731.Google Scholar
Miyoshi, N, Nakamura, Y, Ueda, Y, Abe, M, Ozawa, Y, Uchida, K, et al. (2003) Dietary ginger constituents, galanals A and B, are potent apoptosis inducers in human T lymphoma Jurkat cells. Cancer Lett 199, 113119.Google Scholar
Hu, W, Yu, A, Wang, S, Bai, Q, Tang, H, Yang, B, et al. (2023) Extraction, purification, structural characteristics, biological activities, and applications of the polysaccharides from Zingiber officinale Roscoe. (Ginger): a review. Molecules 28, 3855. doi: 10.3390/molecules28093855.Google Scholar
Butt, MS & Sultan, MT (2011) Ginger and its health claims: molecular aspects. Crit Rev Food Sci Nutr 51, 383393.Google Scholar
Izadi, M, Sadri, N, Abdi, A, Serajian, S, Jalalei, D & Tahmasebi, S (2024) Epigenetic biomarkers in aging and longevity: current and future application. Life Sci 351, 122842.Google Scholar
Ozkur, M, Benlier, N, Takan, I, Vasileiou, C, Georgakilas, AG, Pavlopoulou, A, et al. (2022) Ginger for healthy ageing: a systematic review on current evidence of its antioxidant, anti-inflammatory, and anticancer properties. Oxid Med Cell Longevity 2022, 4748447.Google Scholar
Arulkumar, R, Bang, E, Noh, S-G, Yokozawa, T & Chung, HY (2019) Role of garlic and ginger in anti-oxidative and anti-inflammatory effects in aging. SDRP J Food Sci Technol 4, 788795.Google Scholar
Ziada, AS, Smith, MR & Côté, HCF (2020) Updating the free radical theory of aging. Front Cell Develop Biol 8, 575645.Google Scholar
Checa, J & Aran, JM (2020) Reactive oxygen species: drivers of physiological and pathological processes. J Inflamm Res 13, 10571073.Google Scholar
Ilkhanizadeh, B, Shirpoor, A, Khadem Ansari, MH, Nemati, S & Rasmi, Y (2016) Protective effects of ginger (Zingiber officinale) extract against diabetes-induced heart abnormality in rats. Diabetes Metab J 40, 4653.Google Scholar
Shaukat, MN, Nazir, A & Fallico, B (2023) Ginger bioactives: a comprehensive review of health benefits and potential food applications. Antioxidants (Basel) 12, 2015. doi: 10.3390/antiox12112015.Google Scholar
Romero, A, Forero, M, Sequeda-Castañeda, LG, Grismaldo, A, Iglesias, J, Celis-Zambrano, CA, et al. (2018) Effect of ginger extract on membrane potential changes and AKT activation on a peroxide-induced oxidative stress cell model. J King Saud Univ – Sci 30, 263269.Google Scholar
Shanmugam, KR, Mallikarjuna, K, Nishanth, K, Kuo, CH, Reddy, KS (2011) Protective effect of dietary ginger on antioxidant enzymes and oxidative damage in experimental diabetic rat tissues. Food Chem 124, 14361442.Google Scholar
Thomas, CM, Fuller, CJ, Whittles, CE & Sharif, M (2007) Chondrocyte death by apoptosis is associated with cartilage matrix degradation. Osteoarthritis Cartilage 15, 2734.Google Scholar
Asl, SS, Pourheydar, B, Dabaghian, F, Nezhadi, A, Roointan, A & Mehdizadeh, M (2013) Ecstasy-induced caspase expression alters following ginger treatment. Basic Clin Neurosci 4, 329333.Google Scholar
Kim, CY, Seo, Y, Lee, C, Park, GH & Jang, JH (2018) Neuroprotective effect and molecular mechanism of [6]-gingerol against scopolamine-induced amnesia in C57BL/6 mice. Evid-Based Complement Altern Med: eCAM 2018, 8941564.Google Scholar
Al Syaad, KM, Elsaid, FG, Abdraboh, ME & Al-Doaiss, AA (2019) Effect of graviola (Annona muricata l.) and ginger (Zingiber officinale Roscoe) on diabetes mellitus induced in male Wistar albino rats. Folia Biol 65, 275284.Google Scholar
Baechle, JJ, Chen, N, Makhijani, P, Winer, S, Furman, D & Winer, DA (2023) Chronic inflammation and the hallmarks of aging. Mol Metab 74, 101755.Google Scholar
Rea, IM, Gibson, DS, McGilligan, V, McNerlan, SE, Alexander, HD & Ross, OA (2018) Age and age-related diseases: role of inflammation triggers and cytokines. Front Immunol 9, 586.Google Scholar
Ezzat, SM, Ezzat, MI, Okba, MM, Menze, ET & Abdel-Naim, AB (2018) The hidden mechanism beyond ginger (Zingiber officinale Rosc.) potent in vivo and in vitro anti-inflammatory activity. J Ethnopharmacol 214, 113123.Google Scholar
Mao, QQ, Xu, XY, Cao, SY, Gan, RY, Corke, H, Beta, T, et al. (2019) Bioactive compounds and bioactivities of ginger (Zingiber officinale Roscoe). Foods 8, 185. doi: 10.3390/foods8060185.Google Scholar
Rastogi, N, Gara, RK, Trivedi, R, Singh, A, Dixit, P, Maurya, R, et al. (2014) (6)-Gingerolinduced myeloid leukemia cell death is initiated by reactive oxygen species and activation of miR-27b expression. Free Radical Biol Med 68, 288301.Google Scholar
Deorukhkar, A, Ahuja, N, Mercado, AL, Diagaradjane, P, Raju, U, Patel, N, et al. (2015) Zerumbone increases oxidative stress in a thiol-dependent ROS-independent manner to increase DNA damage and sensitize colorectal cancer cells to radiation. Cancer Med 4, 278292.Google Scholar
Bawadood, AS, Al-Abbasi, FA, Anwar, F, El-Halawany, AM & Al-Abd, AM (2020) 6-Shogaol suppresses the growth of breast cancer cells by inducing apoptosis and suppressing autophagy via targeting notch signaling pathway. Biomed Pharmacother 128, 110302.Google Scholar
Akimoto, M, Iizuka, M, Kanematsu, R, Yoshida, M & Takenaga, K (2015) Anticancer effect of ginger extract against pancreatic cancer cells mainly through reactive oxygen species-mediated autotic cell death. PLoS One 10, e0126605.Google Scholar
Chakraborty, D, Bishayee, K, Ghosh, S, Biswas, R, Mandal, SK, Khuda-Bukhsh, AR (2012) [6]-Gingerol induces caspase 3 dependent apoptosis and autophagy in cancer cells: drug-DNA interaction and expression of certain signal genes in HeLa cells. Eur J Pharmacol 694, 2029.Google Scholar
Farombi, EO, Ajayi, BO & Adedara, IA (2020) 6-Gingerol delays tumorigenesis in benzo[a]pyrene and dextran sulphate sodium-induced colorectal cancer in mice. Food Chem Toxicol 142, 111483.Google Scholar
Saenghong, N, Wattanathorn, J, Muchimapura, S, Tongun, T, Piyavhatkul, N, Banchonglikitkul, C, et al. (2012) Zingiber officinale improves cognitive function of the middle-aged healthy women. Evid-Based Complement Altern Med: eCAM 2012, 383062.Google Scholar
Ghayur, MN, Gilani, AH, Ahmed, T, Khalid, A, Nawaz, SA, Agbedahunsi, JM, et al. (2008) Muscarinic, Ca(++) antagonist and specific butyrylcholinesterase inhibitory activity of dried ginger extract might explain its use in dementia. J Phar Pharmacol 60, 13751383.Google Scholar
Zeng, GF, Zhang, ZY, Lu, L, Xiao, DQ, Zong, SH & He, JM (2013) Protective effects of ginger root extract on Alzheimer disease-induced behavioral dysfunction in rats. Rejuvenat Res 16, 124133.Google Scholar
Zhao, Y & Chen, ZY (2018) Roles of spicy foods and their bioactive compounds in management of hypercholesterolemia. J Agric Food Chem 66, 86628671.Google Scholar
Li, J, Wang, S, Yao, L, Ma, P, Chen, Z, Han, TL, et al. (2019) 6-gingerol ameliorates age-related hepatic steatosis: association with regulating lipogenesis, fatty acid oxidation, oxidative stress and mitochondrial dysfunction. Toxicol Appl Pharmacol 362, 125135.Google Scholar
Khezri, K, Maleki Dizaj, S, Rahbar Saadat, Y, Sharifi, S, Shahi, S, Ahmadian, E, et al. (2021) Osteogenic differentiation of mesenchymal stem cells via curcumin-containing nanoscaffolds. Stem Cells Int 2021, 1520052.Google Scholar
Mashhadi, NS, Ghiasvand, R, Askari, G, Hariri, M, Darvishi, L & Mofid, MR (2013) Anti-oxidative and anti-inflammatory effects of ginger in health and physical activity: review of current evidence. Int J Prevent Med 4, Suppl 1, S36S42.Google Scholar
Leelapornpisid, P, Wickett, RR, Chansakaow, S & Wongwattananukul, N (2015) Potential of native Thai aromatic plant extracts in antiwrinkle body creams. J Cosmet Sci 66, 219231.Google Scholar
Ryan, JL & Morrow, GR (2010) Ginger. Oncol Nurse Ed 24, 4649.Google Scholar
Tyagi, AK, Prasad, S, Yuan, W, Li, S & Aggarwal, BB (2015) Identification of a novel compound (β-sesquiphellandrene) from turmeric (Curcuma longa) with anticancer potential: comparison with curcumin. Invest New Drugs 33, 11751186.Google Scholar
Liu, W, Zhai, Y, Heng, X, Che, FY, Chen, W, Sun, D, et al. (2016) Oral bioavailability of curcumin: problems and advancements. J Drug Target 24, 694702.Google Scholar
Shoba, G, Joy, D, Joseph, T, Majeed, M, Rajendran, R & Srinivas, PS (1998) Influence of piperine on the pharmacokinetics of curcumin in animals and human volunteers. Plant Med 64, 353356.Google Scholar
Dey, S & Sreenivasan, K (2014) Conjugation of curcumin onto alginate enhances aqueous solubility and stability of curcumin. Carbohydr Polym 99, 499507.Google Scholar
Abbaspour-Aghdam, S, Hazrati, A, Abdolmohammadi-Vahid, S, Tahmasebi, S, Mohseni, J, Valizadeh, H, et al. (2022) Immunomodulatory role of nanocurcumin in COVID-19 patients with dropped natural killer cells frequency and function. Eur J Pharmacol 933, 175267.Google Scholar
Esmaeilzadeh, A, Jafari, D, Tahmasebi, S, Elahi, R & Khosh, E (2021) Immune-based therapy for COVID-19. Adv Exp Med Biol 1318, 449468.Google Scholar
Tahmasebi, S, Saeed, BQ, Temirgalieva, E, Yumashev, AV, El-Esawi, MA, Navashenaq, JG, et al. (2021) Nanocurcumin improves Treg cell responses in patients with mild and severe SARS-CoV2. Life Sci 276, 119437.Google Scholar
Cheng, AL, Hsu, CH, Lin, JK, Hsu, MM, Ho, YF, Shen, TS, et al. (2001) Phase I clinical trial of curcumin, a chemopreventive agent, in patients with high-risk or pre-malignant lesions. Anticancer Res 21, 28952900.Google Scholar
Gupta, SC, Kismali, G & Aggarwal, BB (2013) Curcumin, a component of turmeric: from farm to pharmacy. BioFactors 39, 213.Google Scholar
Calder, PC, Bosco, N, Bourdet-Sicard, R, Capuron, L, Delzenne, N, Doré, J, et al. (2017) Health relevance of the modification of low grade inflammation in ageing (inflammageing) and the role of nutrition. Ageing Res Rev 40, 95119.Google Scholar
Sikora, E, Scapagnini, G & Barbagallo, M (2010) Curcumin, inflammation, ageing and age-related diseases. Immun Ageing: I & A 7, 1.Google Scholar
Sandur, SK, Ichikawa, H, Pandey, MK, Kunnumakkara, AB, Sung, B, Sethi, G, et al. (2007) Role of pro-oxidants and antioxidants in the anti-inflammatory and apoptotic effects of curcumin (diferuloylmethane). Free Radical Biol Med 43, 568580.Google Scholar
Olszanecki, R, Jawień, J, Gajda, M, Mateuszuk, L, Gebska, A, Korabiowska, M, et al. (2005) Effect of curcumin on atherosclerosis in apoE/LDLR-double knockout mice. J Physiol Pharmacol: Off J Polish Physiol Soc 56, 627635.Google Scholar
He, Y, Yue, Y, Zheng, X, Zhang, K, Chen, S & Du, Z (2015) Curcumin, inflammation, and chronic diseases: how are they linked? Molecules 20, 91839213.Google Scholar
Sun, Y, Hu, X, Hu, G, Xu, C & Jiang, H (2015) Curcumin attenuates hydrogen peroxide-induced premature senescence via the activation of SIRT1 in human umbilical vein endothelial cells. Biol Pharm Bull 38, 11341141.Google Scholar
Kitani, K, Osawa, T & Yokozawa, T (2007) The effects of tetrahydrocurcumin and green tea polyphenol on the survival of male C57BL/6 mice. Biogerontology 8, 567573.Google Scholar
Bielak-Zmijewska, A, Sikora-Polaczek, M, Nieznanski, K, Mosieniak, G, Kolano, A, Maleszewski, M, et al. (2010) Curcumin disrupts meiotic and mitotic divisions via spindle impairment and inhibition of CDK1 activity. Cell Proliferation 43, 354364.Google Scholar
Hansen, J (1998) Common cancers in the elderly. Drugs Aging 13, 467478.Google Scholar
Izadi, M, Sadri, N, Abdi, A, Zadeh, MMR, Jalaei, D, Ghazimoradi, MM, et al. (2024) Longevity and anti-aging effects of curcumin supplementation. GeroScience 46, 29332950.Google Scholar
Luo, J, Mills, K, le Cessie, S, Noordam, R & van Heemst, D (2020) Ageing, age-related diseases and oxidative stress: what to do next? Ageing Res Rev 57, 100982.Google Scholar
Pizzino, G, Irrera, N, Cucinotta, M, Pallio, G, Mannino, F, Arcoraci, V, et al. (2017) Oxidative stress: harms and benefits for human health. Oxid Med Cell Longev 2017, 8416763. doi: 10.1155/2017/8416763.Google Scholar
Huang, MT (1997). Antioxidant and antitumorigenic properties of curcumin. In Ohigashi, H, Osawa, T, Terao, J, Watanabe, S, Yoshikawa, T (eds.), Food Factors for Cancer Prevention. Tokyo: Springer. doi: 10.1007/978-4-431-67017-9_50.Google Scholar
Abo-Salem, O, Harisa, G, Ali, T, El-Sayed, E & Abou-Elnour, F (2014) Curcumin ameliorates streptozotocin-induced heart injury in rats: Curcumin attenuates diabetic heart injury. J Biochem Mol Toxicol 28, 263270.Google Scholar
Oyetayo, BO, Abolaji, AO, Fasae, KD & Aderibigbe, A (2020) Ameliorative role of diets fortified with curcumin in a Drosophila melanogaster model of aluminum chloride-induced neurotoxicity. J Funct Foods 71, 104035.Google Scholar
Zhang, J, Zheng, Y, Luo, Y, Du, Y, Zhang, X & Fu, J (2019) Curcumin inhibits LPS-induced neuroinflammation by promoting microglial M2 polarization via TREM2/ TLR4/ NF-κB pathways in BV2 cells. Mol Immunol 116, 2937.Google Scholar
Tahmasebi, S, Alimohammadi, M, Khorasani, S & Rezaei, N (2022) Pro-tumorigenic and anti-tumorigenic roles of pro-inflammatory cytokines in cancer. In Rezaei, N (ed.), Handbook of cancer and immunology. Cham: Springer International Publishing, pp. 125.Google Scholar
Salminen, A, Huuskonen, J, Ojala, J, Kauppinen, A, Kaarniranta, K & Suuronen, T (2008) Activation of innate immunity system during aging: NF-kB signaling is the molecular culprit of inflamm-aging. Ageing Rese Rev 7, 83105.Google Scholar
Yousefzadeh, MJ, Schafer, MJ, Noren Hooten, N, Atkinson, EJ, Evans, MK, Baker, DJ, et al. (2018) Circulating levels of monocyte chemoattractant protein-1 as a potential measure of biological age in mice and frailty in humans. Aging Cell 17, e12706.Google Scholar
Kunnumakkara, AB, Bordoloi, D, Padmavathi, G, Monisha, J, Roy, NK, Prasad, S, et al. (2017) Curcumin, the golden nutraceutical: multitargeting for multiple chronic diseases. Br J Pharmacol 174, 13251348.Google Scholar
Hansen, M, Rubinsztein, DC & Walker, DW (2018) Autophagy as a promoter of longevity: insights from model organisms. Nat Rev Mol Cell Biol 19, 579593.Google Scholar
Brown-Borg, HM & Bartke, A (2012) GH and IGF1: roles in energy metabolism of long-living GH mutant mice. J Gerontol Ser A, Biol Sci Med Sci 67, 652660.Google Scholar
Sikora, E, Bielak-Zmijewska, A, Mosieniak, G & Piwocka, K (2010) The promise of slow down ageing may come from curcumin. Curr Pharmaceut Design 16, 884892.Google Scholar
Jiao, D, Wang, J, Lu, W, Tang, X, Chen, J, Mou, H, et al. (2016) Curcumin inhibited HGF-induced EMT and angiogenesis through regulating c-Met dependent PI3K/Akt/mTOR signaling pathways in lung cancer. Mol Ther Oncolyt 3, 16018.Google Scholar
Zhang, J, Wang, J, Xu, J, Lu, Y, Jiang, J, Wang, L, et al. (2016) Curcumin targets the TFEB-lysosome pathway for induction of autophagy. Oncotarget 7, 7565975671.Google Scholar
Maiti, P, Rossignol, J & Dunbar, GL (2017) Curcumin modulates molecular chaperones and autophagy-lysosomal pathways in vitro after exposure to Aβ42. J Alzheimers Dis Parkinsonism 7, 299. doi: 10.4172/2161-0460.1000299.Google Scholar
de Oliveira, MR, Jardim, FR, Setzer, WN, Nabavi, SM & Nabavi, SF (2016) Curcumin, mitochondrial biogenesis, and mitophagy: Exploring recent data and indicating future needs. Biotechnol Adv 34, 813826.Google Scholar
Giordano, S, Darley-Usmar, V & Zhang, J (2014) Autophagy as an essential cellular antioxidant pathway in neurodegenerative disease. Redox Biol 2, 8290.Google Scholar
Hunter, DJ, Schofield, D & Callander, E (2014) The individual and socioeconomic impact of osteoarthritis. Nat Rev Rheumatol 10, 437441.Google Scholar
Henrotin, Y, Priem, F & Mobasheri, A (2013) Curcumin: a new paradigm and therapeutic opportunity for the treatment of osteoarthritis: curcumin for osteoarthritis management. SpringerPlus 2, 56.Google Scholar
Belcaro, G, Hosoi, M, Pellegrini, L, Appendino, G, Ippolito, E, Ricci, A, et al. (2014) A controlled study of a lecithinized delivery system of curcumin (Meriva®) to alleviate the adverse effects of cancer treatment. Phytother Res: PTR 28, 444450.Google Scholar
Panahi, Y, Rahimnia, AR, Sharafi, M, Alishiri, G, Saburi, A & Sahebkar, A (2014) Curcuminoid treatment for knee osteoarthritis: a randomized double-blind placebo-controlled trial. Phytother Res: PTR 28, 16251631.Google Scholar
Qin, S, Huang, L, Gong, J, Shen, S, Huang, J, Tang, Y, et al. (2018) Meta-analysis of randomized controlled trials of 4 weeks or longer suggest that curcumin may afford some protection against oxidative stress. Nutr Res 60, 112.Google Scholar
Na, LX, Li, Y, Pan, HZ, Zhou, XL, Sun, DJ, Meng, M, et al. (2013) Curcuminoids exert glucose-lowering effect in type 2 diabetes by decreasing serum free fatty acids: a double-blind, placebo-controlled trial. Mol Nutr Food Res 57, 15691577.Google Scholar
Bradford, PG (2013) Curcumin and obesity. BioFactors 39, 7887.Google Scholar
Hlavačková, L, Janegová, A, Uličná, O, Janega, P, Cerná, A & Babál, P (2011) Spice up the hypertension diet – curcumin and piperine prevent remodeling of aorta in experimental L-NAME induced hypertension. Nutr Metab 8, 72.Google Scholar
Sahebkar, A (2014) Are curcuminoids effective C-reactive protein-lowering agents in clinical practice? Evidence from a meta-analysis. Phytother Res: PTR 28, 633642.Google Scholar
Ak, T & Gülçin, I (2008) Antioxidant and radical scavenging properties of curcumin. Chem-Biol Interact 174, 2737.Google Scholar
Baum, L, Lam, CW, Cheung, SK, Kwok, T, Lui, V, Tsoh, J, et al. (2008) Six-month randomized, placebo-controlled, double-blind, pilot clinical trial of curcumin in patients with Alzheimer disease. J Clin Psychopharmacol 28, 110113.Google Scholar
Chen, M, Du, ZY, Zheng, X, Li, DL, Zhou, RP & Zhang, K (2018) Use of curcumin in diagnosis, prevention, and treatment of Alzheimer’s disease. Neural Regenerat Res 13, 742752.Google Scholar
Hewlings, SJ & Kalman, DS (2017) Curcumin: a review of its effects on human health. Foods 6, 92. doi: 10.3390/foods6100092.Google Scholar
Ibrahim Fouad, G (2020) Synergistic anti-atherosclerotic role of combined treatment of omega-3 and co-enzyme Q10 in hypercholesterolemia-induced obese rats. Heliyon 6, e03659.Google Scholar
Xiao, B, Zhang, Z, Viennois, E, Kang, Y, Zhang, M, Han, MK, et al. (2016) Combination therapy for ulcerative colitis: orally targeted nanoparticles prevent mucosal damage and relieve inflammation. Theranostics 6, 22502266.Google Scholar
Muhammad, I, Wang, X, Li, S, Li, R & Zhang, X (2018) Curcumin confers hepatoprotection against AFB(1)-induced toxicity via activating autophagy and ameliorating inflammation involving Nrf2/HO-1 signaling pathway. Mol Biol Rep 45, 17751785.Google Scholar
Chen, F, Tang, Y, Sun, Y, Veeraraghavan, VP, Mohan, SK & Cui, C (2019) 6-shogaol, a active constiuents of ginger prevents UVB radiation mediated inflammation and oxidative stress through modulating NrF2 signaling in human epidermal keratinocytes (HaCaT cells). J Photochem Photobiol B, Biol 197, 111518.Google Scholar
Zhou, X, Afzal, S, Wohlmuth, H, Münch, G, Leach, D, Low, M, et al. (2022) Synergistic anti-inflammatory activity of ginger and turmeric extracts in inhibiting lipopolysaccharide and interferon-γ-induced proinflammatory mediators. Molecules 27, 3877. doi: 10.3390/molecules27123877.Google Scholar
Zhou, X, Münch, G, Wohlmuth, H, Afzal, S, Kao, MT, Al-Khazaleh, A, et al. (2022) Synergistic Inhibition of pro-inflammatory pathways by ginger and turmeric extracts in RAW 264.7 cells. Front Pharmacol 13, 818166.Google Scholar
Bakhtiari Aqmasjed, S, Sajjadi, MM, Falahatkar, B & Safari, R (2023) Effects of dietary ginger (Zingiber officinale) extract and curcumin on growth, hematology, immunity, and antioxidant status in rainbow trout (Oncorhynchus mykiss). Aquacult Rep 32, 101714.Google Scholar
(2024) Synergistic effect of Zingiber officinale (ginger) and Curcuma longa L. (curcumin analogs) for anti-inflammatory, anti-nociceptive activity and analgesic potentials. J Popul Ther Clin Pharmacol 31, 13631369. https://doi.org/10.53555/jptcp.v31i3.5132 Google Scholar
Thota, RN, Acharya, SH & Garg, ML (2019) Curcumin and/or omega-3 polyunsaturated fatty acids supplementation reduces insulin resistance and blood lipids in individuals with high risk of type 2 diabetes: a randomised controlled trial. Lipids Health Dis 18, 31.Google Scholar
Hoeijmakers, JH (2009) DNA damage, aging, and cancer. N Engl J Med 361, 14751485.Google Scholar
Burtner, CR & Kennedy, BK (2010) Progeria syndromes and ageing: what is the connection? Nat Rev Mol Cell Biol 11, 567578.Google Scholar
Moskalev, AA, Shaposhnikov, MV, Plyusnina, EN, Zhavoronkov, A, Budovsky, A, Yanai, H, et al. (2013) The role of DNA damage and repair in aging through the prism of Koch-like criteria. Ageing Res Rev 12, 661684.Google Scholar
Wu, H & Roks, AJ (2014) Genomic instability and vascular aging: a focus on nucleotide excision repair. Trends Cardiovasc Med 24, 6168.Google Scholar
Fumagalli, M, Rossiello, F, Clerici, M, Barozzi, S, Cittaro, D, Kaplunov, JM, et al. (2012) Telomeric DNA damage is irreparable and causes persistent DNA-damage-response activation. Nat Cell Biol 14, 355365.Google Scholar
Palm, W & de Lange, T (2008) How shelterin protects mammalian telomeres. Annu Rev Genet 42, 301334.Google Scholar
Bodnar, AG, Ouellette, M, Frolkis, M, Holt, SE, Chiu, C-P, Morin, GB, et al. (1998) Extension of life-span by introduction of telomerase into normal human cells. Science 279, 349352.Google Scholar
Blackburn, EH, Epel, ES & Lin, J (2015) Human telomere biology: A contributory and interactive factor in aging, disease risks, and protection. Science 350, 11931198.Google Scholar
Fraga, MF & Esteller, M (2007) Epigenetics and aging: the targets and the marks. Trends Genet 23, 413418.Google Scholar
Talens, RP, Christensen, K, Putter, H, Willemsen, G, Christiansen, L, Kremer, D, et al. (2012) Epigenetic variation during the adult lifespan: cross-sectional and longitudinal data on monozygotic twin pairs. Aging Cell 11, 694703.Google Scholar
van Ham, TJ, Holmberg, MA, van der Goot, AT, Teuling, E, Garcia-Arencibia, M, Kim, H-E, et al. (2010) Identification of MOAG-4/SERF as a regulator of age-related proteotoxicity. Cell 142, 601612.Google Scholar
Taylor, RC & Dillin, A (2011) Aging as an event of proteostasis collapse. Cold Spring Harb Perspect Biol 3, a004440. doi: 10.1101/cshperspect.a004440.Google Scholar
Yu, G & Hyun, S (2021) Proteostasis-associated aging: lessons from a Drosophila model. Genes Genomics 43, 19.Google Scholar
Barzilai, N, Huffman, DM, Muzumdar, RH & Bartke, A (2012) The critical role of metabolic pathways in aging. Diabetes 61, 13151322.Google Scholar
Fontana, L, Partridge, L & Longo, VD (2010) Extending healthy life span—from yeast to humans. Science 328, 321326.Google Scholar
Kujoth, GC, Hiona, A, Pugh, TD, Someya, S, Panzer, K, Wohlgemuth, SE, et al. (2005) Mitochondrial DNA mutations, oxidative stress, and apoptosis in mammalian aging. Science 309, 481484.Google Scholar
Trifunovic, A, Wredenberg, A, Falkenberg, M, Spelbrink, JN, Rovio, AT, Bruder, CE, et al. (2004) Premature ageing in mice expressing defective mitochondrial DNA polymerase. Nature 429, 417423.Google Scholar
Vermulst, M, Wanagat, J, Kujoth, GC, Bielas, JH, Rabinovitch, PS, Prolla, TA, et al. (2008) DNA deletions and clonal mutations drive premature aging in mitochondrial mutator mice. Nature Genet 40, 392394.Google Scholar
Matheu, A, Maraver, A, Collado, M, Garcia-Cao, I, Cañamero, M, Borras, C, et al. (2009) Anti-aging activity of the Ink4/Arf locus. Aging Cell 8, 152161.Google Scholar
Baker, DJ, Wijshake, T, Tchkonia, T, LeBrasseur, NK, Childs, BG, Van De Sluis, B, et al. (2011) Clearance of p16Ink4a-positive senescent cells delays ageing-associated disorders. Nature 479, 232236.Google Scholar
Rando, TA & Chang, HY (2012) Aging, rejuvenation, and epigenetic reprogramming: resetting the aging clock. Cell 148, 4657.Google Scholar
Conboy, IM & Rando, TA (2012) Heterochronic parabiosis for the study of the effects of aging on stem cells and their niches. Cell Cycle 11, 22602267.Google Scholar
da Silva, PFL & Schumacher, B (2021) Principles of the molecular and cellular mechanisms of aging. J Invest Dermatol 141, 951960.Google Scholar
Chen, S, Gan, D, Lin, S, Zhong, Y, Chen, M, Zou, X, et al. (2022) Metformin in aging and aging-related diseases: clinical applications and relevant mechanisms. Theranostics 12, 27222740.Google Scholar
Trumbo, P, Schlicker, S, Yates, AA & Poos, M (2002) Dietary reference intakes for energy, carbohydrate, fiber, fat, fatty acids, cholesterol, protein and amino acids. J Am Diet Assoc 102, 16211630.Google Scholar
Andrew, M, Michael, M, Karl, K, Elisabeth, AdB, Flint, B, Karen, M, et al. (2017) A randomized, double-blind, placebo-controlled trial of coenzyme Q10 in Huntington disease. Neurology 88, 152.Google Scholar
Fotino, AD, Thompson-Paul, AM & Bazzano, LA (2013) Effect of coenzyme Q10 supplementation on heart failure: a meta-analysis. Am J Clin Nutr 97, 268275.Google Scholar
Alehagen, U, Aaseth, J & Johansson, P (2015) Reduced cardiovascular mortality 10 years after supplementation with selenium and coenzyme Q10 for four years: follow-up results of a prospective randomized double-blind placebo-controlled trial in elderly citizens. PLoS One 10, e0141641.Google Scholar
Alehagen, U, Alexander, J & Aaseth, J (2016) Supplementation with selenium and coenzyme Q10 reduces cardiovascular mortality in elderly with low selenium status. A secondary analysis of a randomised clinical trial. PLoS One 11, e0157541.Google Scholar
Moradi, M, Haghighatdoost, F, Feizi, A, Larijani, B & Azadbakht, L (2016) Effect of coenzyme Q10 supplementation on diabetes biomarkers: a systematic review and meta-analysis of randomized controlled clinical trials. Arch Iran Med 19.Google Scholar
Abdollahzad, H, Aghdashi, MA, Jafarabadi, MA & Alipour, B (2015) Effects of coenzyme Q10 supplementation on inflammatory cytokines (TNF-α, IL-6) and oxidative stress in rheumatoid arthritis patients: a randomized controlled trial. Arch Med Res 46, 527533.Google Scholar
Rivara, MB, Yeung, CK, Robinson-Cohen, C, Phillips, BR, Ruzinski, J, Rock, D, et al. (2017) Effect of coenzyme Q10 on biomarkers of oxidative stress and cardiac function in hemodialysis patients: the CoQ10 biomarker trial. Am J Kidney Dis 69, 389399.Google Scholar
Heidari, A, Hamidi, G, Soleimani, A, Aghadavod, E & Asemi, Z (2018) Effects of Coenzyme Q10 supplementation on gene expressions related to insulin, lipid, and inflammation pathways in patients with diabetic nephropathy. Iran J Kidney Dis 12, 1421.Google Scholar