Potential Synergistic Supplementation of NAD+ Promoting Compounds as a Strategy for Increasing Healthspan
Abstract
:1. Introduction
2. NAD+ Homeostasis
2.1. NAD+, Sirtuins and Longevity-Promoting Pathway
2.2. NAD+ and Circadian Rhythm
2.3. CD38 and the Decline of NAD+
2.4. NMN as an NAD+ Boosting Therapeutic
2.5. A Combination Approach for Restoring Youthful NAD+ Levels during Aging
3. Potential Synergistic Nutraceutical Interventions
3.1. Stilbenes: Resveratrol and Pterostilbene
3.2. CoQ10
3.3. Betaine
3.4. Flavonoids: Quercetin, Fisetin, Luteolin/Luteolinidin, and Apigenin
3.5. Carotenoids: Astaxanthin and Lycopene
3.6. Curcumin
3.7. Alpha-Ketoglutarate
3.8. Epigallocatechin Gallate
4. Conclusive Remarks
Author Contributions
Funding
Institutional Review Board Statement
Informed Consent Statement
Data Availability Statement
Acknowledgments
Conflicts of Interest
References
- Niccoli, T.; Partridge, L. Ageing as a risk factor for disease. Curr. Biol. 2012, 22, R741–R752. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Jose, V.; Consuelo, B.; Miquel, J. Theories of ageing. IUBMB Life 2007, 59, 249–254. [Google Scholar] [CrossRef]
- Jin, K. Modern Biological Theories of Aging. Aging Dis. 2010, 1, 72–74. [Google Scholar]
- Partridge, L. Intervening in ageing to prevent the diseases of ageing. Trends Endocrinol. Metab. 2014, 25, 555–557. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Janson, M. Orthomolecular medicine: The therapeutic use of dietary supplements for anti-aging. Clin. Interv. Aging 2006, 1, 261–265. [Google Scholar] [CrossRef]
- Borumand, M.; Sibilla, S. Daily consumption of the collagen supplement Pure Gold Collagen® reduces visible signs of aging. Clin. Interv. Aging 2014, 9, 1747–1758. [Google Scholar] [CrossRef] [Green Version]
- Skovgaard, G.R.L.; Jensen, A.S.; Sigler, M.L. Effect of a novel dietary supplement on skin aging in post-menopausal women. Eur. J. Clin. Nutr. 2006, 60, 1201–1206. [Google Scholar] [CrossRef] [Green Version]
- Stephens, T.J.; Sigler, M.L.; Hino, P.D.; Moigne, A.L.; Dispensa, L. A Randomized, Double-blind, Placebo-controlled Clinical Trial Evaluating an Oral Anti-aging Skin Care Supplement for Treating Photodamaged Skin. J. Clin. Aesthet. Dermatol. 2016, 9, 25–32. [Google Scholar]
- Poddar, J.; Pradhan, M.; Ganguly, G.; Chakrabarti, S. Biochemical deficits and cognitive decline in brain aging: Intervention by dietary supplements. J. Chem. Neuroanat. 2018, 95, 70–80. [Google Scholar] [CrossRef]
- Lemon, J.A.; Boreham, D.R.; Rollo, C.D. A Complex Dietary Supplement Extends Longevity of Mice. J. Gerontol. Ser. A 2005, 60, 275–279. [Google Scholar] [CrossRef] [Green Version]
- Xie, X.; Gao, Y.; Zeng, M.; Wang, Y.; Wei, T.-F.; Lu, Y.-B.; Zhang, W.-P. Nicotinamide ribose ameliorates cognitive impairment of aged and Alzheimer’s disease model mice. Metab. Brain Dis. 2019, 34, 353–366. [Google Scholar] [CrossRef]
- Clement, J.; Wong, M.; Poljak, A.; Sachdev, P.; Braidy, N. The Plasma NAD+ Metabolome Is Dysregulated in “Normal” Aging. Rejuvenation Res. 2019, 22, 121–130. [Google Scholar] [CrossRef]
- Fang, E.F.; Lautrup, S.; Hou, Y.; Demarest, T.G.; Croteau, D.L.; Mattson, M.P.; Bohr, V.A. NAD+ in Aging: Molecular Mechanisms and Translational Implications. Trends Mol. Med. 2017, 23, 899–916. [Google Scholar] [CrossRef]
- Gomes, A.P.; Price, N.L.; Ling, A.J.Y.; Moslehi, J.J.; Montgomery, M.K.; Rajman, L.; White, J.P.; Teodoro, J.S.; Wrann, C.D.; Hubbard, B.P.; et al. Declining NAD+ induces a pseudohypoxic state disrupting nuclear-mitochondrial communication during aging. Cell 2013, 155, 1624–1638. [Google Scholar] [CrossRef] [Green Version]
- Radenkovic, D.; Reason; Verdin, E. Clinical Evidence for Targeting NAD Therapeutically. Pharmaceuticals 2020, 13, 247. [Google Scholar] [CrossRef]
- Liu, J.-K. Antiaging agents: Safe interventions to slow aging and healthy life span extension. Nat. Prod. Bioprospect. 2022, 12, 18. [Google Scholar] [CrossRef]
- Schultz, M.B.; Sinclair, D.A. Why NAD+ Declines during Aging: It’s Destroyed. Cell Metab. 2016, 23, 965–966. [Google Scholar] [CrossRef] [Green Version]
- Ray Chaudhuri, A.; Nussenzweig, A. The multifaceted roles of PARP1 in DNA repair and chromatin remodelling. Nat. Rev. Mol. Cell Biol. 2017, 18, 610–621. [Google Scholar] [CrossRef]
- Gerdts, J.; Brace, E.J.; Sasaki, Y.; DiAntonio, A.; Milbrandt, J. SARM1 activation triggers axon degeneration locally via NAD+ destruction. Science 2015, 348, 453. [Google Scholar] [CrossRef] [Green Version]
- Katsyuba, E.; Romani, M.; Hofer, D.; Auwerx, J. NAD+ homeostasis in health and disease. Nat. Metab. 2020, 2, 9–31. [Google Scholar] [CrossRef]
- Mateuszuk, Ł.; Campagna, R.; Kutryb-Zając, B.; Kuś, K.; Słominska, E.M.; Smolenski, R.T.; Chlopicki, S. Reversal of endothelial dysfunction by nicotinamide mononucleotide via extracellular conversion to nicotinamide riboside. Biochem. Pharmacol. 2020, 178, 114019. [Google Scholar] [CrossRef] [PubMed]
- Kannt, A.; Rajagopal, S.; Kadnur, S.V.; Suresh, J.; Bhamidipati, R.K.; Swaminathan, S.; Hallur, M.S.; Kristam, R.; Elvert, R.; Czech, J.; et al. A small molecule inhibitor of Nicotinamide N-methyltransferase for the treatment of metabolic disorders. Sci. Rep. 2018, 8, 3660. [Google Scholar] [CrossRef] [PubMed]
- Campagna, R.; Mateuszuk, Ł.; Wojnar-Lason, K.; Kaczara, P.; Tworzydło, A.; Kij, A.; Bujok, R.; Mlynarski, J.; Wang, Y.; Sartini, D.; et al. Nicotinamide N-methyltransferase in endothelium protects against oxidant stress-induced endothelial injury. Biochim. Biophys. Acta Mol. Cell Res. 2021, 1868, 119082. [Google Scholar] [CrossRef] [PubMed]
- Hong, S.; Moreno-Navarrete, J.M.; Wei, X.; Kikukawa, Y.; Tzameli, I.; Prasad, D.; Lee, Y.; Asara, J.M.; Fernandez-Real, J.M.; Maratos-Flier, E.; et al. Nicotinamide N-methyltransferase regulates hepatic nutrient metabolism through Sirt1 protein stabilization. Nat. Med. 2015, 21, 887–894. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Schmeisser, K.; Mansfeld, J.; Kuhlow, D.; Weimer, S.; Priebe, S.; Heiland, I.; Birringer, M.; Groth, M.; Segref, A.; Kanfi, Y.; et al. Role of sirtuins in lifespan regulation is linked to methylation of nicotinamide. Nat. Chem. Biol. 2013, 9, 693–700. [Google Scholar] [CrossRef] [Green Version]
- Van Haren, M.J.; Zhang, Y.; Thijssen, V.; Buijs, N.; Gao, Y.; Mateuszuk, L.; Fedak, F.A.; Kij, A.; Campagna, R.; Sartini, D.; et al. Macrocyclic peptides as allosteric inhibitors of nicotinamide N-methyltransferase (NNMT). RSC Chem. Biol. 2021, 2, 1546–1555. [Google Scholar] [CrossRef]
- Van Haren, M.J.; Gao, Y.; Buijs, N.; Campagna, R.; Sartini, D.; Emanuelli, M.; Mateuszuk, L.; Kij, A.; Chlopicki, S.; Escudé Martinez de Castilla, P.; et al. Esterase-Sensitive Prodrugs of a Potent Bisubstrate Inhibitor of Nicotinamide N-Methyltransferase (NNMT) Display Cellular Activity. Biomolecules 2021, 11, 1357. [Google Scholar] [CrossRef]
- Neelakantan, H.; Vance, V.; Wetzel, M.D.; Wang, H.-Y.L.; McHardy, S.F.; Finnerty, C.C.; Hommel, J.D.; Watowich, S.J. Selective and membrane-permeable small molecule inhibitors of nicotinamide N-methyltransferase reverse high fat diet-induced obesity in mice. Biochem. Pharmacol. 2018, 147, 141–152. [Google Scholar] [CrossRef]
- Gao, Y.; van Haren, M.J.; Buijs, N.; Innocenti, P.; Zhang, Y.; Sartini, D.; Campagna, R.; Emanuelli, M.; Parsons, R.B.; Jespers, W.; et al. Potent Inhibition of Nicotinamide N-Methyltransferase by Alkene-Linked Bisubstrate Mimics Bearing Electron Deficient Aromatics. J. Med. Chem. 2021, 64, 12938–12963. [Google Scholar] [CrossRef]
- Mendelsohn, A.R.; Larrick, J.W. The NAD+/PARP1/SIRT1 Axis in Aging. Rejuvenation Res. 2017, 20, 244–247. [Google Scholar] [CrossRef]
- Yaku, K.; Okabe, K.; Nakagawa, T. NAD metabolism: Implications in aging and longevity. Ageing Res. Rev. 2018, 47, 1–17. [Google Scholar] [CrossRef]
- Chini, C.C.S.; Tarragó, M.G.; Chini, E.N. NAD and the aging process: Role in life, death and everything in between. Mol. Cell. Endocrinol. 2017, 455, 62. [Google Scholar] [CrossRef]
- Aguiar, S.S.; Rosa, T.S.; Neves, R.V.P.; Leite, P.L.A.; Maciel, L.A.; Gutierrez, S.D.; Rosa, E.C.; Andrade, R.V.; Degens, H.; Korhonen, M.T.; et al. Telomere Length, SIRT1, and Insulin in Male Master Athletes: The Path to Healthy Longevity? Int. J. Sport. Med. 2022, 43, 29–33. [Google Scholar] [CrossRef]
- Tang, B.L. Sirt1 and the Mitochondria. Mol. Cells 2016, 39, 87–95. [Google Scholar] [CrossRef] [Green Version]
- Brunet, A.; Sweeney, L.B.; Sturgill, J.F.; Chua, K.F.; Greer, P.L.; Lin, Y.; Tran, H.; Ross, S.E.; Mostoslavsky, R.; Cohen, H.Y.; et al. Stress-dependent regulation of FOXO transcription factors by the SIRT1 deacetylase. Science 2004, 303, 2011–2015. [Google Scholar] [CrossRef] [Green Version]
- Greer, E.L.; Brunet, A. FOXO transcription factors at the interface between longevity and tumor suppression. Oncogene 2005, 24, 7410–7425. [Google Scholar] [CrossRef] [Green Version]
- Zhao, Y.; Liu, Y.-S. Longevity Factor FOXO3: A Key Regulator in Aging-Related Vascular Diseases. Front. Cardiovasc. Med. 2021, 8, 778674. [Google Scholar] [CrossRef]
- Kaletsky, R.; Lakhina, V.; Arey, R.; Williams, A.; Landis, J.; Ashraf, J.; Murphy, C.T. The C. elegans adult neuronal IIS/FOXO transcriptome reveals adult phenotype regulators. Nature 2016, 529, 92–96. [Google Scholar] [CrossRef] [Green Version]
- Slack, C.; Giannakou, M.E.; Foley, A.; Goss, M.; Partridge, L. dFOXO-independent effects of reduced insulin-like signaling in Drosophila. Aging Cell 2011, 10, 735–748. [Google Scholar] [CrossRef] [Green Version]
- Brenmoehl, J.; Hoeflich, A. Dual control of mitochondrial biogenesis by sirtuin 1 and sirtuin 3. Mitochondrion 2013, 13, 755–761. [Google Scholar] [CrossRef]
- Chan, M.C.; Arany, Z. The Many roles of PGC-1α in Muscle—Recent Developments. Metabolism 2014, 63, 441–451. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Summermatter, S.; Shui, G.; Maag, D.; Santos, G.; Wenk, M.R.; Handschin, C. PGC-1α improves glucose homeostasis in skeletal muscle in an activity-dependent manner. Diabetes 2013, 62, 85–95. [Google Scholar] [CrossRef] [PubMed]
- Cantó, C.; Gerhart-Hines, Z.; Feige, J.N.; Lagouge, M.; Noriega, L.; Milne, J.C.; Elliott, P.J.; Puigserver, P.; Auwerx, J. AMPK regulates energy expenditure by modulating NAD+ metabolism and SIRT1 activity. Nature 2009, 458, 1056–1060. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kauppinen, A.; Suuronen, T.; Ojala, J.; Kaarniranta, K.; Salminen, A. Antagonistic crosstalk between NF-κB and SIRT1 in the regulation of inflammation and metabolic disorders. Cell. Signal. 2013, 25, 1939–1948. [Google Scholar] [CrossRef] [PubMed]
- Mao, K.; Zhang, G. The role of PARP1 in neurodegenerative diseases and aging. FEBS J. 2022, 289, 2013–2024. [Google Scholar] [CrossRef]
- Amjad, S.; Nisar, S.; Bhat, A.A.; Shah, A.R.; Frenneaux, M.P.; Fakhro, K.; Haris, M.; Reddy, R.; Patay, Z.; Baur, J.; et al. Role of NAD+ in regulating cellular and metabolic signaling pathways. Mol. Metab. 2021, 49, 101195. [Google Scholar] [CrossRef]
- Sedlackova, L.; Korolchuk, V.I. The crosstalk of NAD, ROS and autophagy in cellular health and ageing. Biogerontology 2020, 21, 381–397. [Google Scholar] [CrossRef] [Green Version]
- Levine, D.C.; Hong, H.; Weidemann, B.J.; Ramsey, K.M.; Affinati, A.H.; Schmidt, M.S.; Cedernaes, J.; Omura, C.; Braun, R.; Lee, C.; et al. NAD+ Controls Circadian Reprogramming through PER2 Nuclear Translocation to Counter Aging. Mol. Cell 2020, 78, 835–849.e7. [Google Scholar] [CrossRef]
- Peek, C.B.; Affinati, A.H.; Ramsey, K.M.; Kuo, H.-Y.; Yu, W.; Sena, L.A.; Ilkayeva, O.; Marcheva, B.; Kobayashi, Y.; Omura, C.; et al. Circadian Clock NAD+ Cycle Drives Mitochondrial Oxidative Metabolism in Mice. Science 2013, 342, 1243417. [Google Scholar] [CrossRef] [Green Version]
- Rehan, L.; Laszki-Szcząchor, K.; Sobieszczańska, M.; Polak-Jonkisz, D. SIRT1 and NAD as regulators of ageing. Life Sci. 2014, 105, 1–6. [Google Scholar] [CrossRef]
- Chu, X.; Raju, R.P. Regulation of NAD+ metabolism in aging and disease. Metabolism 2022, 126, 154923. [Google Scholar] [CrossRef] [PubMed]
- Gilmour, B.C.; Gudmundsrud, R.; Frank, J.; Hov, A.; Lautrup, S.; Aman, Y.; Røsjø, H.; Brenner, C.; Ziegler, M.; Tysnes, O.-B.; et al. Targeting NAD+ in translational research to relieve diseases and conditions of metabolic stress and ageing. Mech. Ageing Dev. 2020, 186, 111208. [Google Scholar] [CrossRef] [PubMed]
- Zhang, M.; Ying, W. NAD+ Deficiency Is a Common Central Pathological Factor of a Number of Diseases and Aging: Mechanisms and Therapeutic Implications. Antioxid. Redox Signal. 2019, 30, 890–905. [Google Scholar] [CrossRef]
- Zhou, F.-Q. NAD+, Senolytics, or Pyruvate for Healthy Aging? Nutr. Metab. Insights 2021, 14, 11786388211053408. [Google Scholar] [CrossRef] [PubMed]
- Li, W.; Sauve, A.A. NAD+ content and its role in mitochondria. Methods Mol. Biol. 2015, 1241, 39–48. [Google Scholar] [CrossRef]
- Cerutti, R.; Pirinen, E.; Lamperti, C.; Marchet, S.; Sauve, A.A.; Li, W.; Leoni, V.; Schon, E.A.; Dantzer, F.; Auwerx, J.; et al. NAD(+)-dependent activation of Sirt1 corrects the phenotype in a mouse model of mitochondrial disease. Cell Metab. 2014, 19, 1042–1049. [Google Scholar] [CrossRef] [Green Version]
- Li, W.; Li, Y.; Jin, X.; Liao, Q.; Chen, Z.; Peng, H.; Zhou, Y. CD38: A Significant Regulator of Macrophage Function. Front. Oncol. 2022, 12, 775649. [Google Scholar] [CrossRef]
- Aksoy, P.; Escande, C.; White, T.A.; Thompson, M.; Soares, S.; Benech, J.C.; Chini, E.N. Regulation of SIRT 1 mediated NAD dependent deacetylation: A novel role for the multifunctional enzyme CD38. Biochem. Biophys. Res. Commun. 2006, 349, 353–359. [Google Scholar] [CrossRef]
- Bu, X.; Kato, J.; Hong, J.A.; Merino, M.J.; Schrump, D.S.; Lund, F.E.; Moss, J. CD38 knockout suppresses tumorigenesis in mice and clonogenic growth of human lung cancer cells. Carcinogenesis 2018, 39, 242–251. [Google Scholar] [CrossRef] [Green Version]
- Camacho-Pereira, J.; Tarragó, M.G.; Chini, C.C.S.; Nin, V.; Escande, C.; Warner, G.M.; Puranik, A.S.; Schoon, R.A.; Reid, J.M.; Galina, A.; et al. CD38 Dictates Age-Related NAD Decline and Mitochondrial Dysfunction through an SIRT3-Dependent Mechanism. Cell Metab. 2016, 23, 1127–1139. [Google Scholar] [CrossRef] [Green Version]
- Domínguez-Pantoja, M.; López-Herrera, G.; Romero-Ramírez, H.; Santos-Argumedo, L.; Chávez-Rueda, A.K.; Hernández-Cueto, Á.; Flores-Muñoz, M.; Rodríguez-Alba, J.C. CD38 protein deficiency induces autoimmune characteristics and its activation enhances IL-10 production by regulatory B cells. Scand. J. Immunol. 2018, 87, e12664. [Google Scholar] [CrossRef] [Green Version]
- Chini, C.C.S.; Peclat, T.R.; Warner, G.M.; Kashyap, S.; Espindola-Netto, J.M.; de Oliveira, G.C.; Gomez, L.S.; Hogan, K.A.; Tarragó, M.G.; Puranik, A.S.; et al. CD38 ecto-enzyme in immune cells is induced during aging and regulates NAD+ and NMN levels. Nat. Metab. 2020, 2, 1284–1304. [Google Scholar] [CrossRef]
- Chini, C.; Hogan, K.A.; Warner, G.M.; Tarragó, M.G.; Peclat, T.R.; Tchkonia, T.; Kirkland, J.L.; Chini, E. The NADase CD38 is induced by factors secreted from senescent cells providing a potential link between senescence and age-related cellular NAD+ decline. Biochem. Biophys. Res. Commun. 2019, 513, 486–493. [Google Scholar] [CrossRef]
- Covarrubias, A.J.; Kale, A.; Perrone, R.; Lopez-Dominguez, J.A.; Pisco, A.O.; Kasler, H.G.; Schmidt, M.S.; Heckenbach, I.; Kwok, R.; Wiley, C.D.; et al. Senescent cells promote tissue NAD+ decline during ageing via the activation of CD38+ macrophages. Nat. Metab. 2020, 2, 1265–1283. [Google Scholar] [CrossRef]
- Covarrubias, A.J.; Kale, A.; Perrone, R.; Lopez-Dominguez, J.A.; Pisco, A.O.; Kasler, H.G.; Schmidt, M.S.; Wiley, C.D.; Iyer, S.S.; Basisty, N.; et al. Aging-related inflammation driven by cellular senescence enhances NAD consumption via activation of CD38+ pro-inflammatory macrophages. biorXiv 2019, 609438. [Google Scholar] [CrossRef] [Green Version]
- Yoshino, J.; Baur, J.A.; Imai, S.-I. NAD+ Intermediates: The Biology and Therapeutic Potential of NMN and NR. Cell Metab. 2018, 27, 513–528. [Google Scholar] [CrossRef] [Green Version]
- Rajman, L.; Chwalek, K.; Sinclair, D.A. Therapeutic potential of NAD-boosting molecules: The in vivo evidence. Cell Metab. 2018, 27, 529–547. [Google Scholar] [CrossRef] [Green Version]
- Roberts, M.D.; Osburn, S.C.; Godwin, J.S.; Ruple, B.A.; La Monica, M.B.; Raub, B.; Sandrock, J.E.; Ziegenfuss, T.N.; Lopez, H.L. Enhance Trial: Effects of NAD3® on Hallmarks of Aging and Clinical Endpoints of Health in Middle Aged Adults: A Subset Analysis Focused on Blood Cell NAD+ Concentrations and Lipid Metabolism. Physiologia 2022, 2, 20–31. [Google Scholar] [CrossRef]
- Zhong, O.; Wang, J.; Tan, Y.; Lei, X.; Tang, Z. Effects of NAD+ precursor supplementation on glucose and lipid metabolism in humans: A meta-analysis. Nutr. Metab. 2022, 19, 20. [Google Scholar] [CrossRef]
- Nadeeshani, H.; Li, J.; Ying, T.; Zhang, B.; Lu, J. Nicotinamide mononucleotide (NMN) as an anti-aging health product—Promises and safety concerns. J. Adv. Res. 2022, 37, 267–278. [Google Scholar] [CrossRef]
- Shade, C. The Science Behind NMN—A Stable, Reliable NAD+Activator and Anti-Aging Molecule. Integr. Med. 2020, 19, 12–14. [Google Scholar]
- Aman, Y.; Qiu, Y.; Tao, J.; Fang, E.F. Therapeutic potential of boosting NAD+ in aging and age-related diseases. Transl. Med. Aging 2018, 2, 30–37. [Google Scholar] [CrossRef]
- Huang, H. A Multicentre, Randomised, Double Blind, Parallel Design, Placebo Controlled Study to Evaluate the Efficacy and Safety of Uthever (NMN Supplement), an Orally Administered Supplementation in Middle Aged and Older Adults. Front. Aging 2022, 3, 851698. [Google Scholar] [CrossRef] [PubMed]
- Niu, K.-M.; Bao, T.; Gao, L.; Ru, M.; Li, Y.; Jiang, L.; Ye, C.; Wang, S.; Wu, X. The Impacts of Short-Term NMN Supplementation on Serum Metabolism, Fecal Microbiota, and Telomere Length in Pre-Aging Phase. Front. Nutr. 2021, 8, 756243. [Google Scholar] [CrossRef] [PubMed]
- Huang, P.; Jiang, A.; Wang, X.; Zhou, Y.; Tang, W.; Ren, C.; Qian, X.; Zhou, Z.; Gong, A. NMN Maintains Intestinal Homeostasis by Regulating the Gut Microbiota. Front. Nutr. 2021, 8, 714604. [Google Scholar] [CrossRef] [PubMed]
- Kiss, T.; Giles, C.B.; Tarantini, S.; Yabluchanskiy, A.; Balasubramanian, P.; Gautam, T.; Csipo, T.; Nyúl-Tóth, Á.; Lipecz, A.; Szabo, C.; et al. Nicotinamide mononucleotide (NMN) supplementation promotes anti-aging miRNA expression profile in the aorta of aged mice, predicting epigenetic rejuvenation and anti-atherogenic effects. Geroscience 2019, 41, 419–439. [Google Scholar] [CrossRef] [PubMed]
- Liao, B.; Zhao, Y.; Wang, D.; Zhang, X.; Hao, X.; Hu, M. Nicotinamide mononucleotide supplementation enhances aerobic capacity in amateur runners: A randomized, double-blind study. J. Int. Soc. Sport. Nutr. 2021, 18, 54. [Google Scholar] [CrossRef]
- Igarashi, M.; Nakagawa-Nagahama, Y.; Miura, M.; Kashiwabara, K.; Yaku, K.; Sawada, M.; Sekine, R.; Fukamizu, Y.; Sato, T.; Sakurai, T.; et al. Chronic nicotinamide mononucleotide supplementation elevates blood nicotinamide adenine dinucleotide levels and alters muscle function in healthy older men. npj Aging 2022, 8, 5. [Google Scholar] [CrossRef]
- Kim, M.; Seol, J.; Sato, T.; Fukamizu, Y.; Sakurai, T.; Okura, T. Effect of 12-Week Intake of Nicotinamide Mononucleotide on Sleep Quality, Fatigue, and Physical Performance in Older Japanese Adults: A Randomized, Double-Blind Placebo-Controlled Study. Nutrients 2022, 14, 755. [Google Scholar] [CrossRef]
- Maric, T.; Bazhin, A.; Khodakivskyi, P.; Mikhaylov, G.; Solodnikova, E.; Yevtodiyenko, A.; Giordano Attianese, G.M.P.; Coukos, G.; Irving, M.; Joffraud, M.; et al. A bioluminescent-based probe for in vivo non-invasive monitoring of nicotinamide riboside uptake reveals a link between metastasis and NAD+ metabolism. Biosens. Bioelectron. 2023, 220, 114826. [Google Scholar] [CrossRef]
- Cros, C.; Cannelle, H.; Laganier, L.; Grozio, A.; Canault, M. Safety evaluation after acute and sub-chronic oral administration of high purity nicotinamide mononucleotide (NMN-C®) in Sprague-Dawley rats. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2021, 150, 112060. [Google Scholar] [CrossRef]
- Yoshino, M.; Yoshino, J.; Kayser, B.D.; Patti, G.J.; Franczyk, M.P.; Mills, K.F.; Sindelar, M.; Pietka, T.; Patterson, B.W.; Imai, S.-I.; et al. Nicotinamide mononucleotide increases muscle insulin sensitivity in prediabetic women. Science 2021, 372, 1224–1229. [Google Scholar] [CrossRef]
- Irie, J.; Inagaki, E.; Fujita, M.; Nakaya, H.; Mitsuishi, M.; Yamaguchi, S.; Yamashita, K.; Shigaki, S.; Ono, T.; Yukioka, H.; et al. Effect of oral administration of nicotinamide mononucleotide on clinical parameters and nicotinamide metabolite levels in healthy Japanese men. Endocr. J. 2020, 67, 153–160. [Google Scholar] [CrossRef]
- Fukamizu, Y.; Uchida, Y.; Shigekawa, A.; Sato, T.; Kosaka, H.; Sakurai, T. Safety evaluation of β-nicotinamide mononucleotide oral administration in healthy adult men and women. Sci. Rep. 2022, 12, 14442. [Google Scholar] [CrossRef]
- Yi, L.; Maier, A.B.; Tao, R.; Lin, Z.; Vaidya, A.; Pendse, S.; Thasma, S.; Andhalkar, N.; Avhad, G.; Kumbhar, V. The efficacy and safety of β-nicotinamide mononucleotide (NMN) supplementation in healthy middle-aged adults: A randomized, multicenter, double-blind, placebo-controlled, parallel-group, dose-dependent clinical trial. GeroScience 2022, 1–15. [Google Scholar] [CrossRef]
- Pencina, K.; Lavu, S.; Dos Santos, M.; Beleva, Y.M.; Cheng, M.; Livingston, D.; Bhasin, S. MIB-626, an Oral Formulation of a Microcrystalline Unique Polymorph of β-Nicotinamide Mononucleotide, Increases Circulating Nicotinamide Adenine Dinucleotide and its Metabolome in Middle-aged and Older Adults. J. Gerontol. A Biol. Sci. Med. Sci. 2022, glac049. [Google Scholar] [CrossRef]
- Tomé-Carneiro, J.; Larrosa, M.; González-Sarrías, A.; Tomás-Barberán, F.A.; García-Conesa, M.T.; Espín, J.C. Resveratrol and Clinical Trials: The Crossroad from In Vitro Studies to Human Evidence. Curr. Pharm. Des. 2013, 19, 6064–6093. [Google Scholar] [CrossRef] [Green Version]
- Resveratrol and pterostilbene: A comparative overview of their chemistry, biosynthesis, plant sources and pharmacological properties. J. App. Pharm. Sci. 2019, 9, 124–129. [CrossRef] [Green Version]
- Riche, D.M.; McEwen, C.L.; Riche, K.D.; Sherman, J.J.; Wofford, M.R.; Deschamp, D.; Griswold, M. Analysis of Safety from a Human Clinical Trial with Pterostilbene. J. Toxicol. 2013, 2013, e463595. [Google Scholar] [CrossRef] [Green Version]
- Ramírez-Garza, S.L.; Laveriano-Santos, E.P.; Marhuenda-Muñoz, M.; Storniolo, C.E.; Tresserra-Rimbau, A.; Vallverdú-Queralt, A.; Lamuela-Raventós, R.M. Health Effects of Resveratrol: Results from Human Intervention Trials. Nutrients 2018, 10, 1892. [Google Scholar] [CrossRef] [Green Version]
- Li, Y.-R.; Li, S.; Lin, C.-C. Effect of resveratrol and pterostilbene on aging and longevity. Biofactors 2018, 44, 69–82. [Google Scholar] [CrossRef] [PubMed]
- Beghelli, D.; Zallocco, L.; Barbalace, M.C.; Paglia, S.; Strocchi, S.; Cirilli, I.; Marzano, V.; Putignani, L.; Lupidi, G.; Hrelia, S.; et al. Pterostilbene Promotes Mean Lifespan in Both Male and Female Drosophila Melanogaster Modulating Different Proteins in the Two Sexes. Oxid. Med. Cell. Longev. 2022, 2022, 1744408. [Google Scholar] [CrossRef] [PubMed]
- McCormack, D.; McFadden, D. A Review of Pterostilbene Antioxidant Activity and Disease Modification. Oxid. Med. Cell. Longev. 2013, 2013, 575482. [Google Scholar] [CrossRef] [PubMed]
- Knutson, M.D.; Leeuwenburgh, C. Resveratrol and novel potent activators of SIRT1: Effects on aging and age-related diseases. Nutr. Rev. 2008, 66, 591–596. [Google Scholar] [CrossRef] [PubMed]
- Yabluchanskiy, A.; Ma, Y.; Iyer, R.P.; Hall, M.E.; Lindsey, M.L. Matrix Metalloproteinase-9: Many Shades of Function in Cardiovascular Disease. Physiology 2013, 28, 391–403. [Google Scholar] [CrossRef] [Green Version]
- Moens, U.; Kostenko, S.; Sveinbjørnsson, B. The Role of Mitogen-Activated Protein Kinase-Activated Protein Kinases (MAPKAPKs) in Inflammation. Genes 2013, 4, 101–133. [Google Scholar] [CrossRef]
- De la Lastra, C.A.; Villegas, I. Resveratrol as an anti-inflammatory and anti-aging agent: Mechanisms and clinical implications. Mol. Nutr. Food Res. 2005, 49, 405–430. [Google Scholar] [CrossRef]
- Teng, W.-L.; Huang, P.-H.; Wang, H.-C.; Tseng, C.-H.; Yen, F.-L. Pterostilbene Attenuates Particulate Matter-Induced Oxidative Stress, Inflammation and Aging in Keratinocytes. Antioxidants 2021, 10, 1552. [Google Scholar] [CrossRef]
- Kasiotis, K.M.; Pratsinis, H.; Kletsas, D.; Haroutounian, S.A. Resveratrol and related stilbenes: Their anti-aging and anti-angiogenic properties. Food Chem. Toxicol. Int. J. Publ. Br. Ind. Biol. Res. Assoc. 2013, 61, 112–120. [Google Scholar] [CrossRef]
- Hecker, A.; Schellnegger, M.; Hofmann, E.; Luze, H.; Nischwitz, S.P.; Kamolz, L.; Kotzbeck, P. The impact of resveratrol on skin wound healing, scarring, and aging. Int. Wound J. 2021, 19, 9–28. [Google Scholar] [CrossRef]
- Escolme, S.M.; Wakeling, L.A.; Alatawi, F.; Valentine, R.; Ford, D. Does resveratrol act independently of SIRT1 to affect genes relevant to ageing? Proc. Nutr. Soc. 2013, 72, E191. [Google Scholar] [CrossRef] [Green Version]
- Fernández, A.F.; Fraga, M.F. The effects of the dietary polyphenol resveratrol on human healthy aging and lifespan. Epigenetics 2011, 6, 870–874. [Google Scholar] [CrossRef]
- Hu, Y.; Liu, J.; Wang, J.; Liu, Q. The controversial links among calorie restriction, SIRT1, and resveratrol. Free Radic. Biol. Med. 2011, 51, 250–256. [Google Scholar] [CrossRef]
- Bonkowski, M.S.; Sinclair, D.A. Slowing ageing by design: The rise of NAD+ and sirtuin-activating compounds. Nat. Rev. Mol. Cell Biol. 2016, 17, 679–690. [Google Scholar] [CrossRef]
- Zhou, D.-D.; Luo, M.; Huang, S.-Y.; Saimaiti, A.; Shang, A.; Gan, R.-Y.; Li, H.-B. Effects and Mechanisms of Resveratrol on Aging and Age-Related Diseases. Oxid. Med. Cell. Longev. 2021, 2021, 9932218. [Google Scholar] [CrossRef]
- Lachiondo-Ortega, S.; Delgado, T.C.; Baños-Jaime, B.; Velázquez-Cruz, A.; Díaz-Moreno, I.; Martínez-Chantar, M.L. Hu Antigen R (HuR) Protein Structure, Function and Regulation in Hepatobiliary Tumors. Cancers 2022, 14, 2666. [Google Scholar] [CrossRef]
- Barger, J.L.; Kayo, T.; Vann, J.M.; Arias, E.B.; Wang, J.; Hacker, T.A.; Wang, Y.; Raederstorff, D.; Morrow, J.D.; Leeuwenburgh, C.; et al. A low dose of dietary resveratrol partially mimics caloric restriction and retards aging parameters in mice. PLoS ONE 2008, 3, e2264. [Google Scholar] [CrossRef]
- Li, J.; Zhang, C.-X.; Liu, Y.-M.; Chen, K.-L.; Chen, G. A comparative study of anti-aging properties and mechanism: Resveratrol and caloric restriction. Oncotarget 2017, 8, 65717–65729. [Google Scholar] [CrossRef] [Green Version]
- Uddin, M.J.; Farjana, M.; Moni, A.; Hossain, K.S.; Hannan, M.A.; Ha, H. Prospective Pharmacological Potential of Resveratrol in Delaying Kidney Aging. Int. J. Mol. Sci. 2021, 22, 8258. [Google Scholar] [CrossRef]
- Kim, E.N.; Kim, M.Y.; Lim, J.H.; Kim, Y.; Shin, S.J.; Park, C.W.; Kim, Y.-S.; Chang, Y.S.; Yoon, H.E.; Choi, B.S. The protective effect of resveratrol on vascular aging by modulation of the renin-angiotensin system. Atherosclerosis 2018, 270, 123–131. [Google Scholar] [CrossRef]
- Caldeira, C.A.; Santos, M.A.; Araújo, G.R.; Lara, R.C.; Franco, F.N.; Chaves, M.M. Resveratrol: Change of SIRT 1 and AMPK signaling pattern during the aging process. Exp. Gerontol. 2021, 146, 111226. [Google Scholar] [CrossRef] [PubMed]
- Demidenko, Z.N.; Blagosklonny, M.V. At concentrations that inhibit mTOR, resveratrol suppresses cellular senescence. Cell Cycle 2009, 8, 1901–1904. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zhou, J.; Xue, Z.; He, H.-N.; Liu, X.; Yin, S.-Y.; Wu, D.-Y.; Zhang, X.; Schatten, H.; Miao, Y.-L. Resveratrol delays postovulatory aging of mouse oocytes through activating mitophagy. Aging 2019, 11, 11504–11519. [Google Scholar] [CrossRef]
- Sun, Y.-L.; Tang, S.-B.; Shen, W.; Yin, S.; Sun, Q.-Y. Roles of Resveratrol in Improving the Quality of Postovulatory Aging Oocytes In Vitro. Cells 2019, 8, 1132. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Chandrasekaran, K.; Salimian, M.; Konduru, S.R.; Choi, J.; Kumar, P.; Long, A.; Klimova, N.; Ho, C.-Y.; Kristian, T.; Russell, J.W. Overexpression of Sirtuin 1 protein in neurons prevents and reverses experimental diabetic neuropathy. Brain 2019, 142, 3737–3752. [Google Scholar] [CrossRef]
- Moraes, D.S.; Moreira, D.C.; Andrade, J.M.O.; Santos, S.H.S. Sirtuins, brain and cognition: A review of resveratrol effects. IBRO Rep. 2020, 9, 46–51. [Google Scholar] [CrossRef]
- Gocmez, S.S.; Gacar, N.; Utkan, T.; Gacar, G.; Scarpace, P.J.; Tumer, N. Protective effects of resveratrol on aging-induced cognitive impairment in rats. Neurobiol. Learn Mem. 2016, 131, 131–136. [Google Scholar] [CrossRef]
- Oomen, C.A.; Farkas, E.; Roman, V.; Van Der Beek, E.M.; Luiten, P.G.; Meerlo, P. Resveratrol preserves cerebrovascular density and cognitive function in aging mice. Front. Aging Neurosci. 2009, 1, 4. [Google Scholar] [CrossRef] [Green Version]
- Szczurek-Janicka, P.; Ropka-Molik, K.; Oczkowicz, M.; Orczewska-Dudek, S.; Pietras, M.; Pieszka, M. Expression Profile of Brain Aging and Metabolic Function are Altered by Resveratrol or α-Ketoglutarate Supplementation in Rats Fed a High-Fat Diet. Pol. J. Food Nutr. Sci. 2021, 71, 255–268. [Google Scholar] [CrossRef]
- Dubowitz, N.; Xue, W.; Long, Q.; Ownby, J.G.; Olson, D.E.; Barb, D.; Rhee, M.K.; Mohan, A.V.; Watson-Williams, P.I.; Jackson, S.L.; et al. Aging is associated with increased HbA1c levels, independently of glucose levels and insulin resistance, and also with decreased HbA1c diagnostic specificity. Diabet. Med. J. Br. Diabet. Assoc. 2014, 31, 927–935. [Google Scholar] [CrossRef]
- Witte, A.V.; Kerti, L.; Margulies, D.S.; Flöel, A. Effects of Resveratrol on Memory Performance, Hippocampal Functional Connectivity, and Glucose Metabolism in Healthy Older Adults. J. Neurosci. 2014, 34, 7862–7870. [Google Scholar] [CrossRef] [Green Version]
- Moussa, C.; Hebron, M.; Huang, X.; Ahn, J.; Rissman, R.A.; Aisen, P.S.; Turner, R.S. Resveratrol regulates neuro-inflammation and induces adaptive immunity in Alzheimer’s disease. J. Neuroinflam. 2017, 14, 1. [Google Scholar] [CrossRef] [Green Version]
- Mahjabeen, W.; Khan, D.A.; Mirza, S.A. Role of resveratrol supplementation in regulation of glucose hemostasis, inflammation and oxidative stress in patients with diabetes mellitus type 2: A randomized, placebo-controlled trial. Complement. Ther. Med. 2022, 66, 102819. [Google Scholar] [CrossRef]
- Wong, R.H.; Thaung Zaw, J.J.; Xian, C.J.; Howe, P.R. Regular Supplementation with Resveratrol Improves Bone Mineral Density in Postmenopausal Women: A Randomized, Placebo-Controlled Trial. J. Bone Miner. Res. Off. J. Am. Soc. Bone Miner. Res. 2020, 35, 2121–2131. [Google Scholar] [CrossRef]
- Thaung Zaw, J.J.; Howe, P.R.C.; Wong, R.H.X. Long-term effects of resveratrol on cognition, cerebrovascular function and cardio-metabolic markers in postmenopausal women: A 24-month randomised, double-blind, placebo-controlled, crossover study. Clin. Nutr. 2021, 40, 820–829. [Google Scholar] [CrossRef]
- Zaw, J.J.T.; Howe, P.R.C.; Wong, R.H.X. Sustained Cerebrovascular and Cognitive Benefits of Resveratrol in Postmenopausal Women. Nutrients 2020, 12, 828. [Google Scholar] [CrossRef] [Green Version]
- Mo, Z.; Zheng, S.; Lv, Z.; Zhuang, Y.; Lan, X.; Wang, F.; Lu, X.; Zhao, Y.; Zhou, S. Senescence marker protein 30 (SMP30) serves as a potential prognostic indicator in hepatocellular carcinoma. Sci. Rep. 2016, 6, 39376. [Google Scholar] [CrossRef] [Green Version]
- Torregrosa-Muñumer, R.; Vara, E.; Fernández-Tresguerres, J.Á.; Gredilla, R. Resveratrol supplementation at old age reverts changes associated with aging in inflammatory, oxidative and apoptotic markers in rat heart. Eur. J. Nutr. 2021, 60, 2683–2693. [Google Scholar] [CrossRef]
- Smoliga, J.M.; Baur, J.; Hausenblas, H.A. Resveratrol and health—A comprehensive review of human clinical trials. Mol. Nutr. Food Res. 2011, 55, 1129–1141. [Google Scholar] [CrossRef]
- Mehta, S.; Young, C.C.; Warren, M.R.; Akhtar, S.; Shefelbine, S.J.; Crane, J.D.; Bajpayee, A.G. Resveratrol and Curcumin Attenuate Ex Vivo Sugar-Induced Cartilage Glycation, Stiffening, Senescence, and Degeneration. Cartilage 2021, 13, 1214S–1228S. [Google Scholar] [CrossRef]
- Furuyama, T.; Kitayama, K.; Yamashita, H.; Mori, N. Forkhead transcription factor FOXO1 (FKHR)-dependent induction of PDK4 gene expression in skeletal muscle during energy deprivation. Biochem. J. 2003, 375, 365–371. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sin, T.K.; Yung, B.Y.; Siu, P.M. Modulation of SIRT1-Foxo1 signaling axis by resveratrol: Implications in skeletal muscle aging and insulin resistance. Cell Physiol. Biochem. Int. J. Exp. Cell Physiol. Biochem. Pharmacol. 2015, 35, 541–552. [Google Scholar] [CrossRef] [PubMed]
- Ramis, M.R.; Esteban, S.; Miralles, A.; Tan, D.-X.; Reiter, R.J. Caloric restriction, resveratrol and melatonin: Role of SIRT1 and implications for aging and related-diseases. Mech. Ageing Dev. 2015, 146–148, 28–41. [Google Scholar] [CrossRef] [PubMed]
- Corrêa, R.C.G.; Peralta, R.M.; Haminiuk, C.W.I.; Maciel, G.M.; Bracht, A.; Ferreira, I.C.F.R. 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]
- Smoliga, J.M.; Colombo, E.S.; Campen, M.J. A healthier approach to clinical trials evaluating resveratrol for primary prevention of age-related diseases in healthy populations. Aging 2013, 5, 495–506. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Bai, L.; Yau, L.; Tong, T.; Chan, W.; Zhang, W.; Jiang, Z. Improvement of tissue-specific distribution and biotransformation potential of nicotinamide mononucleotide in combination with ginsenosides or resveratrol. Pharmacol. Res. Perspect. 2022, 10, e00986. [Google Scholar] [CrossRef] [PubMed]
- Grant, R. Resveratrol Increases Intracellular NAD+ Levels Through Up regulation of the NAD+ Synthetic Enzyme Nicotinamide Mononucleotide Adenylyltransferase. Nat. Preced. 2010, 4421, 1–8. [Google Scholar] [CrossRef]
- Simic, P.; Vela Parada, X.F.; Parikh, S.M.; Dellinger, R.; Guarente, L.P.; Rhee, E.P. Nicotinamide riboside with pterostilbene (NRPT) increases NAD+ in patients with acute kidney injury (AKI): A randomized, double-blind, placebo-controlled, stepwise safety study of escalating doses of NRPT in patients with AKI. BMC Nephrol. 2020, 21, 342. [Google Scholar] [CrossRef]
- Dellinger, R.W.; Santos, S.R.; Morris, M.; Evans, M.; Alminana, D.; Guarente, L.; Marcotulli, E. Repeat dose NRPT (nicotinamide riboside and pterostilbene) increases NAD+ levels in humans safely and sustainably: A randomized, double-blind, placebo-controlled study. npj Aging Mech. Dis. 2017, 3, 17. [Google Scholar] [CrossRef] [Green Version]
- Yang, Y.; Liu, Z.; Zhao, B.; Joseph, S.; Ma, Y.; Xu, H.; Cheng, H. Resveratrol and Quercetin Potentiate the Cell Protection and Rescue Effects of NAD+ Precursors in HEK293 Cells Challenged by DNA Damaging Agent, N-Methyl-N′-nitro-N-nitrosoguanidine. Nat. Prod. Commun. 2021, 16, 1934578X211045465. [Google Scholar] [CrossRef]
- Garrido-Maraver, J.; Cordero, M.D.; Oropesa-Ávila, M.; Fernández Vega, A.; de la Mata, M.; Delgado Pavón, A.; de Miguel, M.; Pérez Calero, C.; Villanueva Paz, M.; Cotán, D.; et al. Coenzyme q10 therapy. Mol. Syndromol. 2014, 5, 187–197. [Google Scholar] [CrossRef] [Green Version]
- Garrido-Maraver, J.; Cordero, M.D.; Oropesa-Avila, M.; Vega, A.F.; de la Mata, M.; Pavon, A.D.; Alcocer-Gomez, E.; Calero, C.P.; Paz, M.V.; Alanis, M.; et al. Clinical applications of coenzyme Q10. Front. Biosci. 2014, 19, 619–633. [Google Scholar] [CrossRef] [Green Version]
- Martelli, A.; Testai, L.; Colletti, A.; Cicero, A.F.G. Coenzyme Q10: Clinical Applications in Cardiovascular Diseases. Antioxidants 2020, 9, 341. [Google Scholar] [CrossRef]
- Hernández-Camacho, J.D.; Bernier, M.; López-Lluch, G.; Navas, P. Coenzyme Q10 Supplementation in Aging and Disease. Front. Physiol. 2018, 9, 44. [Google Scholar] [CrossRef] [Green Version]
- Gutierrez-Mariscal, F.M.; Yubero-Serrano, E.M.; Villalba, J.M.; Lopez-Miranda, J. Coenzyme Q10: From bench to clinic in aging diseases, a translational review. Crit. Rev. Food Sci. Nutr. 2019, 59, 2240–2257. [Google Scholar] [CrossRef] [PubMed]
- Díaz-Casado, M.E.; Quiles, J.L.; Barriocanal-Casado, E.; González-García, P.; Battino, M.; López, L.C.; Varela-López, A. The Paradox of Coenzyme Q10 in Aging. Nutrients 2019, 11, 2221. [Google Scholar] [CrossRef] [Green Version]
- González-Guardia, L.; Yubero-Serrano, E.M.; Delgado-Lista, J.; Perez-Martinez, P.; Garcia-Rios, A.; Marin, C.; Camargo, A.; Delgado-Casado, N.; Roche, H.M.; Perez-Jimenez, F.; et al. Effects of the Mediterranean diet supplemented with coenzyme q10 on metabolomic profiles in elderly men and women. J. Gerontol. A Biol. Sci. Med. Sci. 2015, 70, 78–84. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Castro-Marrero, J.; Sáez-Francàs, N.; Segundo, M.J.; Calvo, N.; Faro, M.; Aliste, L.; Fernández de Sevilla, T.; Alegre, J. Effect of coenzyme Q10 plus nicotinamide adenine dinucleotide supplementation on maximum heart rate after exercise testing in chronic fatigue syndrome—A randomized, controlled, double-blind trial. Clin. Nutr. 2016, 35, 826–834. [Google Scholar] [CrossRef] [Green Version]
- Castro-Marrero, J.; Cordero, M.D.; Segundo, M.J.; Sáez-Francàs, N.; Calvo, N.; Román-Malo, L.; Aliste, L.; Fernández de Sevilla, T.; Alegre, J. Does oral coenzyme Q10 plus NADH supplementation improve fatigue and biochemical parameters in chronic fatigue syndrome? Antioxid. Redox Signal. 2015, 22, 679–685. [Google Scholar] [CrossRef] [Green Version]
- Zeisel, S. Choline, Other Methyl-Donors and Epigenetics. Nutrients 2017, 9, 445. [Google Scholar] [CrossRef] [Green Version]
- Go, E.K.; Jung, K.J.; Kim, J.Y.; Yu, B.P.; Chung, H.Y. Betaine suppresses proinflammatory signaling during aging: The involvement of nuclear factor-kappaB via nuclear factor-inducing kinase/IkappaB kinase and mitogen-activated protein kinases. J. Gerontol. A Biol. Sci. Med. Sci. 2005, 60, 1252–1264. [Google Scholar] [CrossRef] [Green Version]
- Zhao, G.; He, F.; Wu, C.; Li, P.; Li, N.; Deng, J.; Zhu, G.; Ren, W.; Peng, Y. Betaine in Inflammation: Mechanistic Aspects and Applications. Front. Immunol. 2018, 9, 1070. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Sun, W.-P.; Zhai, M.-Z.; Li, D.; Zhou, Y.; Chen, N.-N.; Guo, M.; Zhou, S.-S. Comparison of the effects of nicotinic acid and nicotinamide degradation on plasma betaine and choline levels. Clin. Nutr. 2017, 36, 1136–1142. [Google Scholar] [CrossRef] [PubMed]
- Li, D.; Tian, Y.-J.; Guo, J.; Sun, W.-P.; Lun, Y.-Z.; Guo, M.; Luo, N.; Cao, Y.; Cao, J.-M.; Gong, X.-J.; et al. Nicotinamide supplementation induces detrimental metabolic and epigenetic changes in developing rats. Br. J. Nutr. 2013, 110, 2156–2164. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Kashyap, D.; Garg, V.K.; Tuli, H.S.; Yerer, M.B.; Sak, K.; Sharma, A.K.; Kumar, M.; Aggarwal, V.; Sandhu, S.S. Fisetin and Quercetin: Promising Flavonoids with Chemopreventive Potential. Biomolecules 2019, 9, 174. [Google Scholar] [CrossRef] [Green Version]
- Yousefzadeh, M.J.; Zhu, Y.; McGowan, S.J.; Angelini, L.; Fuhrmann-Stroissnigg, H.; Xu, M.; Ling, Y.Y.; Melos, K.I.; Pirtskhalava, T.; Inman, C.L.; et al. Fisetin is a senotherapeutic that extends health and lifespan. eBioMedicine 2018, 36, 18–28. [Google Scholar] [CrossRef] [Green Version]
- Jang, S.-Y.; Kang, H.T.; Hwang, E.S. Nicotinamide-induced Mitophagy. J. Biol. Chem. 2012, 287, 19304–19314. [Google Scholar] [CrossRef] [Green Version]
- Deepika; Maurya, P.K. Health Benefits of Quercetin in Age-Related Diseases. Molecules 2022, 27, 2498. [Google Scholar] [CrossRef]
- Geng, L.; Liu, Z.; Zhang, W.; Li, W.; Wu, Z.; Wang, W.; Ren, R.; Su, Y.; Wang, P.; Sun, L.; et al. Chemical screen identifies a geroprotective role of quercetin in premature aging. Protein Cell 2018, 10, 417–435. [Google Scholar] [CrossRef] [Green Version]
- Escande, C.; Nin, V.; Price, N.L.; Capellini, V.; Gomes, A.P.; Barbosa, M.T.; O’Neil, L.; White, T.A.; Sinclair, D.A.; Chini, E.N. Flavonoid Apigenin Is an Inhibitor of the NAD+ase CD38. Diabetes 2013, 62, 1084–1093. [Google Scholar] [CrossRef] [Green Version]
- Zhang, F.; Feng, J.; Zhang, J.; Kang, X.; Qian, D. Quercetin modulates AMPK/SIRT1/NF-κB signaling to inhibit inflammatory/oxidative stress responses in diabetic high fat diet-induced atherosclerosis in the rat carotid artery. Exp. Ther. Med. 2020, 20, 280. [Google Scholar] [CrossRef]
- Buss, G.D.; Constantin, J.; de Lima, L.C.N.; Teodoro, G.R.; Comar, J.F.; Ishii-Iwamoto, E.L.; Bracht, A. The Action of Quercetin on the Mitochondrial NADH to NAD+ Ratio in the Isolated Perfused Rat Liver. Planta Med. 2005, 71, 1118–1122. [Google Scholar] [CrossRef]
- Gendrisch, F.; Esser, P.R.; Schempp, C.M.; Wölfle, U. Luteolin as a modulator of skin aging and inflammation. BioFactors 2021, 47, 170–180. [Google Scholar] [CrossRef]
- Compound in Celery, Peppers Reduces Age-Related Memory Deficits. Available online: https://www.sciencedaily.com/releases/2010/10/101013122601.htm (accessed on 11 November 2022).
- Kellenberger, E.; Kuhn, I.; Schuber, F.; Muller-Steffner, H. Flavonoids as inhibitors of human CD38. Bioorg. Med. Chem. Lett. 2011, 21, 3939–3942. [Google Scholar] [CrossRef]
- Boslett, J.; Hemann, C.; Zhao, Y.J.; Lee, H.-C.; Zweier, J.L. Luteolinidin Protects the Postischemic Heart through CD38 Inhibition with Preservation of NAD(P)(H). J. Pharmacol. Exp. Ther. 2017, 361, 99–108. [Google Scholar] [CrossRef] [Green Version]
- Wissler Gerdes, E.O.; Misra, A.; Netto, J.M.E.; Tchkonia, T.; Kirkland, J.L. Strategies for late phase preclinical and early clinical trials of senolytics. Mech. Ageing Dev. 2021, 200, 111591. [Google Scholar] [CrossRef]
- Farkas, O.; Palócz, O.; Pászti-Gere, E.; Gálfi, P. Polymethoxyflavone Apigenin-Trimethylether Suppresses LPS-Induced Inflammatory Response in Nontransformed Porcine Intestinal Cell Line IPEC-J2. Oxidative Med. Cell. Longev. 2015, 2015, e673847. [Google Scholar] [CrossRef] [Green Version]
- Ali, F.; Rahul; Naz, F.; Jyoti, S.; Siddique, Y.H. Health functionality of apigenin: A review. Int. J. Food Prop. 2017, 20, 1197–1238. [Google Scholar] [CrossRef]
- Ahmed, S.A.; Parama, D.; Daimari, E.; Girisa, S.; Banik, K.; Harsha, C.; Dutta, U.; Kunnumakkara, A.B. Rationalizing the therapeutic potential of apigenin against cancer. Life Sci. 2021, 267, 118814. [Google Scholar] [CrossRef]
- Qiu, J.-G.; Wang, L.; Liu, W.-J.; Wang, J.-F.; Zhao, E.-J.; Zhou, F.-M.; Ji, X.-B.; Wang, L.-H.; Xia, Z.-K.; Wang, W.; et al. Apigenin Inhibits IL-6 Transcription and Suppresses Esophageal Carcinogenesis. Front. Pharmacol. 2019, 10, 1002. [Google Scholar] [CrossRef] [Green Version]
- Budhraja, A.; Gao, N.; Zhang, Z.; Son, Y.-O.; Cheng, S.; Wang, X.; Ding, S.; Hitron, A.; Chen, G.; Luo, J.; et al. Apigenin induces apoptosis in human leukemia cells and exhibits anti-leukemic activity in vivo. Mol. Cancer Ther. 2012, 11, 132–142. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Alam, W.; Rocca, C.; Khan, H.; Hussain, Y.; Aschner, M.; De Bartolo, A.; Amodio, N.; Angelone, T.; Cheang, W.S. Current Status and Future Perspectives on Therapeutic Potential of Apigenin: Focus on Metabolic-Syndrome-Dependent Organ Dysfunction. Antioxidants 2021, 10, 1643. [Google Scholar] [CrossRef] [PubMed]
- Ginwala, R.; Bhavsar, R.; Chigbu, D.G.I.; Jain, P.; Khan, Z.K. Potential Role of Flavonoids in Treating Chronic Inflammatory Diseases with a Special Focus on the Anti-Inflammatory Activity of Apigenin. Antioxidants 2019, 8, 35. [Google Scholar] [CrossRef] [PubMed]
- Choi, W.H.; Son, H.J.; Jang, Y.J.; Ahn, J.; Jung, C.H.; Ha, T.Y. Apigenin Ameliorates the Obesity-Induced Skeletal Muscle Atrophy by Attenuating Mitochondrial Dysfunction in the Muscle of Obese Mice. Mol. Nutr. Food Res. 2017, 61, 1700218. [Google Scholar] [CrossRef]
- Su, T.; Chunhua, H.; Yang, C.; Jiang, T.; Su, J.; Chen, M.; Fatima, S.; Gong, R.-H.; Hu, X.; Bian, Z.-X.; et al. Apigenin inhibits STAT3/CD36 signaling axis and reduces visceral obesity. Pharmacol. Res. 2019, 152, 104586. [Google Scholar] [CrossRef]
- Clayton, Z.S.; Hutton, D.A.; Brunt, V.E.; VanDongen, N.S.; Ziemba, B.P.; Casso, A.G.; Greenberg, N.T.; Mercer, A.N.; Rossman, M.J.; Campisi, J.; et al. Apigenin restores endothelial function by ameliorating oxidative stress, reverses aortic stiffening, and mitigates vascular inflammation with aging. Am. J. Physiol. Heart Circ. Physiol. 2021, 321, H185–H196. [Google Scholar] [CrossRef]
- Sharma, A.; Ghani, A.; Sak, K.; Tuli, H.S.; Sharma, A.K.; Setzer, W.N.; Sharma, S.; Das, A.K. Probing into Therapeutic Anti-cancer Potential of Apigenin: Recent Trends and Future Directions. Recent. Pat. Inflamm. Allergy Drug Discov. 2019, 13, 124–133. [Google Scholar] [CrossRef]
- Li, B.S.; Zhu, R.Z.; Lim, S.-H.; Seo, J.H.; Choi, B.-M. Apigenin Alleviates Oxidative Stress-Induced Cellular Senescence via Modulation of the SIRT1-NAD+-CD38 Axis. Am. J. Chin. Med. 2021, 49, 1235–1250. [Google Scholar] [CrossRef]
- Ogura, Y.; Kitada, M.; Xu, J.; Monno, I.; Koya, D. CD38 inhibition by apigenin ameliorates mitochondrial oxidative stress through restoration of the intracellular NAD+/NADH ratio and Sirt3 activity in renal tubular cells in diabetic rats. Aging 2020, 12, 11325–11336. [Google Scholar] [CrossRef]
- Sztretye, M.; Dienes, B.; Gönczi, M.; Czirják, T.; Csernoch, L.; Dux, L.; Szentesi, P.; Keller-Pintér, A. Astaxanthin: A Potential Mitochondrial-Targeted Antioxidant Treatment in Diseases and with Aging. Oxid. Med. Cell. Longev. 2019, 2019, 3849692. [Google Scholar] [CrossRef] [Green Version]
- Zhang, X.-S.; Lu, Y.; Li, W.; Tao, T.; Peng, L.; Wang, W.-H.; Gao, S.; Liu, C.; Zhuang, Z.; Xia, D.-Y.; et al. Astaxanthin ameliorates oxidative stress and neuronal apoptosis via SIRT1/NRF2/Prx2/ASK1/p38 after traumatic brain injury in mice. Br. J. Pharmacol. 2021, 178, 1114–1132. [Google Scholar] [CrossRef]
- Zhang, J.; Wang, Q.; Zhao, S.; Ji, X.; Qiu, J.; Wang, J.; Zhou, Y.; Cai, Q.; Zhang, J.; Gao, H. Astaxanthin attenuated pressure overload-induced cardiac dysfunction and myocardial fibrosis: Partially by activating SIRT1. Biochim. Biophys. Acta BBA Gen. Subj. 2017, 1861, 1715–1728. [Google Scholar] [CrossRef]
- Shatoor, A.S.; Al Humayed, S. Astaxanthin Ameliorates high-fat diet-induced cardiac damage and fibrosis by upregulating and activating SIRT1. Saudi J. Biol. Sci. 2021, 28, 7012–7021. [Google Scholar] [CrossRef]
- Gao, D.; Wang, H.; Xu, Y.; Zheng, D.; Zhang, Q.; Li, W. Protective effect of astaxanthin against contrast-induced acute kidney injury via SIRT1-p53 pathway in rats. Int. Urol. Nephrol. 2019, 51, 351–358. [Google Scholar] [CrossRef]
- Zhang, D.; Zhao, L.; Wong, L. β-Nicotinamide Monucleotide Supplement with Astaxanthin and Blood Orange Enhanced NAD+ Bioavailability and Mitigated Age-Associated Physiological Decline in Zebrafish. Curr. Dev. Nutr. 2022, 6, 54. [Google Scholar] [CrossRef]
- Petyaev, I.M. Lycopene Deficiency in Ageing and Cardiovascular Disease. Oxid. Med. Cell. Longev. 2016, 2016, 3218605. [Google Scholar] [CrossRef] [Green Version]
- Li, J.; Zhang, Y.; Zeng, X.; Cheng, Y.; Tang, L.; Hong, D.; Yang, X. Lycopene ameliorates insulin resistance and increases muscle capillary density in aging via activation of SIRT1. J. Nutr. Biochem. 2022, 99, 108862. [Google Scholar] [CrossRef]
- Liu, X.; Dilxat, T.; Shi, Q.; Qiu, T.; Lin, J. The combination of nicotinamide mononucleotide and lycopene prevents cognitive impairment and attenuates oxidative damage in D-galactose induced aging models via Keap1-Nrf2 signaling. Gene 2022, 822, 146348. [Google Scholar] [CrossRef] [PubMed]
- Bielak-Zmijewska, A.; Grabowska, W.; Ciolko, A.; Bojko, A.; Mosieniak, G.; Bijoch, Ł.; Sikora, E. The Role of Curcumin in the Modulation of Ageing. Int. J. Mol. Sci. 2019, 20, 1239. [Google Scholar] [CrossRef] [PubMed] [Green Version]
- Zendedel, E.; E Butler, A.; Atkin, S.L.; Sahebkar, A. Impact of curcumin on sirtuins: A review. J. Cell. Biochem. 2018, 119, 10291–10300. [Google Scholar] [CrossRef] [PubMed]
- Hu, A.; Huang, J.-J.; Li, R.-L.; Lu, Z.-Y.; Duan, J.-L.; Xu, W.-H.; Chen, X.-P.; Fan, J.-P. Curcumin as therapeutics for the treatment of head and neck squamous cell carcinoma by activating SIRT1. Sci. Rep. 2015, 5, 13429. [Google Scholar] [CrossRef] [Green Version]
- Bańkowski, S.; Petr, M.; Rozpara, M.; Sadowska-Krępa, E. Effect of 6-week curcumin supplementation on aerobic capacity, antioxidant status and sirtuin 3 level in middle-aged amateur long-distance runners. Redox Rep. 2022, 27, 186–192. [Google Scholar] [CrossRef]
- Rhoads, T.W.; Anderson, R.M. Alpha-Ketoglutarate, the Metabolite that Regulates Aging in Mice. Cell Metab. 2020, 32, 323–325. [Google Scholar] [CrossRef]
- Chin, R.M.; Fu, X.; Pai, M.Y.; Vergnes, L.; Hwang, H.; Deng, G.; Diep, S.; Lomenick, B.; Meli, V.S.; Monsalve, G.C.; et al. The metabolite α-ketoglutarate extends lifespan by inhibiting ATP synthase and TOR. Nature 2014, 510, 397–401. [Google Scholar] [CrossRef] [Green Version]
- Bayliak, M.M.; Lushchak, V.I. Pleiotropic effects of alpha-ketoglutarate as a potential anti-ageing agent. Ageing Res. Rev. 2021, 66, 101237. [Google Scholar] [CrossRef]
- Demidenko, O.; Barardo, D.; Budovskii, V.; Finnemore, R.; Palmer, F.R.; Kennedy, B.K.; Budovskaya, Y.V. Rejuvant®, a potential life-extending compound formulation with alpha-ketoglutarate and vitamins, conferred an average 8 year reduction in biological aging, after an average of 7 months of use, in the TruAge DNA methylation test. Aging 2021, 13, 24485–24499. [Google Scholar] [CrossRef]
- Asadi Shahmirzadi, A.; Edgar, D.; Liao, C.-Y.; Hsu, Y.-M.; Lucanic, M.; Asadi Shahmirzadi, A.; Wiley, C.D.; Gan, G.; Kim, D.E.; Kasler, H.G.; et al. Alpha-Ketoglutarate, an Endogenous Metabolite, Extends Lifespan and Compresses Morbidity in Aging Mice. Cell Metab. 2020, 32, 447–456.e6. [Google Scholar] [CrossRef]
- Payne, A.; Nahashon, S.; Taka, E.; Adinew, G.M.; Soliman, K.F.A. Epigallocatechin-3-Gallate (EGCG): New Therapeutic Perspectives for Neuroprotection, Aging, and Neuroinflammation for the Modern Age. Biomolecules 2022, 12, 371. [Google Scholar] [CrossRef]
- Niu, Y.; Na, L.; Feng, R.; Gong, L.; Zhao, Y.; Li, Q.; Li, Y.; Sun, C. The phytochemical, EGCG, extends lifespan by reducing liver and kidney function damage and improving age-associated inflammation and oxidative stress in healthy rats. Aging Cell 2013, 12, 1041–1049. [Google Scholar] [CrossRef]
- Pai, P.-Y.; Chou, W.-C.; Chan, S.-H.; Wu, S.-Y.; Chen, H.-I.; Li, C.-W.; Hsieh, P.-L.; Chu, P.-M.; Chen, Y.-A.; Ou, H.-C.; et al. Epigallocatechin Gallate Reduces Homocysteine-Caused Oxidative Damages through Modulation SIRT1/AMPK Pathway in Endothelial Cells. Am. J. Chin. Med. 2020, 49, 113–129. [Google Scholar] [CrossRef]
- Ayissi, V.B.O.; Ebrahimi, A.; Schluesenner, H. Epigenetic effects of natural polyphenols: A focus on SIRT1-mediated mechanisms. Mol. Nutr. Food Res. 2013, 58, 22–32. [Google Scholar] [CrossRef] [PubMed]
- Jiang, S.; Huang, C.; Zheng, G.; Yi, W.; Wu, B.; Tang, J.; Liu, X.; Huang, B.; Wu, D.; Yan, T.; et al. EGCG Inhibits Proliferation and Induces Apoptosis Through Downregulation of SIRT1 in Nasopharyngeal Carcinoma Cells. Front. Nutr. 2022, 9, 851972. [Google Scholar] [CrossRef] [PubMed]
- Ageing and Health. Available online: https://www.who.int/news-room/fact-sheets/detail/ageing-and-health (accessed on 11 November 2022).
- Peclat, T.R.; Thompson, K.L.; Warner, G.M.; Chini, C.C.S.; Tarragó, M.G.; Mazdeh, D.Z.; Zhang, C.; Zavala-Solorio, J.; Kolumam, G.; Liang Wong, Y.; et al. CD38 inhibitor 78c increases mice lifespan and healthspan in a model of chronological aging. Aging Cell 2022, 21, e13589. [Google Scholar] [CrossRef] [PubMed]
Clinical Trials | Compound of Interest |
---|---|
Nicotinamide Mononucleotide Increases Muscle Insulin Sensitivity in Prediabetic Women [82] | NMN |
Effect of Oral Administration of Nicotinamide Mononucleotide on Clinical Parameters and Nicotinamide Metabolite Levels in Healthy Japanese Men [83] | NMN |
Nicotinamide Mononucleotide Supplementation Enhances Aerobic Capacity in Amateur Runners: a randomized, double-blind study [77] | NMN |
Effect of 12-Week Intake of Nicotinamide Mononucleotide on Sleep Quality, Fatigue, and Physical Performance in Older Japanese Adults: a randomized, double-blind placebo-controlled study [79] | NMN |
Safety Evaluation of Beta-nicotinamide Mononucleotide Oral Administration in Healthy Adult Men and Women [84] | NMN |
The Efficacy and Safety of Beta-nicotinamide Mononucleotide (NMN) Supplementation in Healthy Middle-aged Adults: a randomized, multicenter, double-blind, placebo-controlled, parallel-group dose-dependent clinical trial [85] | NMN |
A Multicenter, Randomized, Double-Blind, Parallel Design, Placebo-Controlled Study to Evaluate the Efficacy and Safety of Uthever (NMN Supplement), an Orally Administered Supplementation, in Middle-Aged and Older Adults [73] | NMN |
MIB-626, an Oral Formulation of a Microcrystalline Unique Polymorph of Beta-Nicotinamide Mononucleotide, Increases Circulating Nicotinamide Adenine Dinucleotide and its Metabolome in Middle-Aged and Older Adults [86] | MIB-626 |
Phase 2a MIB-626 vs. Placebo COVID-19 (NCT05038488) | MIB-626 |
Effect of Oral NAD+ Precursors Administration on Blood NAD+ Concentration in Healthy Adults (NICO) (NCT05517122) | NAM, NR, and NMN |
Effect of NMN Supplementation on Organ System Biology (VAN) (NCT04571008) | NMN |
Pharmacodynamics and Tolerance of Nicotinamide Mononucleotide (NMN, 400mg/Day) in Healthy Adults (NCT04862338) | NMN |
Study to Evaluate the Effect of Nicotinamide Mononucleotide (NMN) As an Adjuvant to Standard of Care (SOC) On Fatigue Associated with Covid-19 Infection (NCT05175768) | NMN |
Nicotinamide Mononucleotide in Hypertensive Patients (NCT04903210) | NMN |
Safety and Pharmacokinetics of Nicotinamide Mononucleotide (NMN) in Healthy Adults (NCT04910061) | NMN |
Effect of NMN (Nicotinamide Mononucleotide) on Polycystic Ovary Syndrome (NMN) (NCT05305677) | NMN |
Effect of NMN (Nicotinamide Mononucleotide) on Diminished Ovarian Reserve (Including Premature Ovarian Insufficiency) (NCT05485610) | NMN |
Effect of NMN on Muscle Recovery and Physical Capacity in Healthy Volunteers with Moderate Physical Activity (NCT04664361) | NMN |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2023 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
Sharma, A.; Chabloz, S.; Lapides, R.A.; Roider, E.; Ewald, C.Y. Potential Synergistic Supplementation of NAD+ Promoting Compounds as a Strategy for Increasing Healthspan. Nutrients 2023, 15, 445. https://doi.org/10.3390/nu15020445
Sharma A, Chabloz S, Lapides RA, Roider E, Ewald CY. Potential Synergistic Supplementation of NAD+ Promoting Compounds as a Strategy for Increasing Healthspan. Nutrients. 2023; 15(2):445. https://doi.org/10.3390/nu15020445
Chicago/Turabian StyleSharma, Arastu, Sophie Chabloz, Rebecca A. Lapides, Elisabeth Roider, and Collin Y. Ewald. 2023. "Potential Synergistic Supplementation of NAD+ Promoting Compounds as a Strategy for Increasing Healthspan" Nutrients 15, no. 2: 445. https://doi.org/10.3390/nu15020445
APA StyleSharma, A., Chabloz, S., Lapides, R. A., Roider, E., & Ewald, C. Y. (2023). Potential Synergistic Supplementation of NAD+ Promoting Compounds as a Strategy for Increasing Healthspan. Nutrients, 15(2), 445. https://doi.org/10.3390/nu15020445