The Potential Use of Vitamin D3 and Phytochemicals for Their Anti-Ageing Effects
Abstract
:1. Introduction
2. Methods
3. Vitamin D3
3.1. Vitamin D3 and Infectious Diseases
3.2. Vitamin D3 and Natural Immunity
3.3. Vitamin D3 and Chronic Inflammation
3.4. Vitamin D3, Gut Microbiota, and Gut Environment
4. Phytochemicals
4.1. Terpenoids
4.2. Carotenoids
4.3. Flavonoids
4.4. Attempts to Improve the Bioavailability and Activation of Phytochemicals in the Body
4.5. Consideration of the Affinity of Phytochemicals to the VDR
5. Synergy between Vitamin D3 and Phytochemicals
5.1. Vitamin D3 and Phytochemicals in Bone Metabolism
5.2. Vitamin D3 and Phytochemicals in the Prevention of Ageing
5.3. Vitamin D3 and Phytochemicals in the Suppression of Chronic Inflammation
5.4. Foods with Anti-Ageing Effects
6. Conclusions
Author Contributions
Funding
Acknowledgments
Conflicts of Interest
References
- Stevens, S.L. Fat-Soluble Vitamins. Nurs. Clin. N. Am. 2021, 56, 33–45. [Google Scholar] [CrossRef] [PubMed]
- Tardy, A.L.; Pouteau, E.; Marquez, D.; Yilmaz, C.; Scholey, A. Vitamins and Minerals for Energy, Fatigue and Cognition: A Narrative Review of the Biochemical and Clinical Evidence. Nutrients 2020, 12, 228. [Google Scholar] [CrossRef] [PubMed]
- Tripkovic, L.; Lambert, H.; Hart, K.; Smith, C.P.; Bucca, G.; Penson, S.; Chope, G.; Hyppönen, E.; Berry, J.; Vieth, R.; et al. Comparison of vitamin D2 and vitamin D3 supplementation in raising serum 25-hydroxyvitamin D status: A systematic review and meta-analysis. Am. J. Clin. Nutr. 2012, 95, 1357–1364. [Google Scholar] [CrossRef] [PubMed]
- Holick, M.F. The vitamin D deficiency pandemic: Approaches for diagnosis, treatment and prevention. Rev. Endocr. Metab. Disord. 2017, 18, 153–165. [Google Scholar] [CrossRef] [PubMed]
- Palacios, C.; Gonzalez, L. Is vitamin D deficiency a major global public health problem? J. Steroid Biochem. Mol. Biol. 2014, 144 Pt A, 138–145. [Google Scholar] [CrossRef]
- Weaver, C.M.; Alexander, D.D.; Boushey, C.J.; Dawson-Hughes, B.; Lappe, J.M.; LeBoff, M.S.; Liu, S.; Looker, A.C.; Wallace, T.C.; Wang, D.D. Calcium plus vitamin D supplementation and risk of fractures: An updated meta-analysis from the National Osteoporosis Foundation. Osteoporos. Int. 2016, 27, 367–376. [Google Scholar] [CrossRef] [PubMed]
- Płudowski, P.; Kos-Kudła, B.; Walczak, M.; Fal, A.; Zozulińska-Ziółkiewicz, D.; Sieroszewski, P.; Peregud-Pogorzelski, J.; Lauterbach, R.; Targowski, T.; Lewiński, A.; et al. Guidelines for Preventing and Treating Vitamin D Deficiency: A 2023 Update in Poland. Nutrients 2023, 15, 695. [Google Scholar] [CrossRef]
- Saponaro, F.; Saba, A.; Zucchi, R. An Update on Vitamin D Metabolism. Int. J. Mol. Sci. 2020, 21, 6573. [Google Scholar] [CrossRef]
- Hsu, S.; Zelnick, L.R.; Lin, Y.S.; Best, C.M.; Kestenbaum, B.R.; Thummel, K.E.; Hoofnagle, A.N.; de Boer, I.H. Validation of the 24,25-dihydroxyvitamin D3 to 25-hydroxyvitamin D3 ratio as a biomarker of 25-hydroxyvitamin D3 clearance. J. Steroid Biochem. Mol. Biol. 2022, 217, 106047. [Google Scholar] [CrossRef]
- Gorzynik-Debicka, M.; Przychodzen, P.; Cappello, F.; Kuban-Jankowska, A.; Marino Gammazza, A.; Knap, N.; Wozniak, M.; Gorska-Ponikowska, M. Potential Health Benefits of Olive Oil and Plant Polyphenols. Int. J. Mol Sci. 2018, 19, 686. [Google Scholar] [CrossRef]
- Scuto, M.; Trovato Salinaro, A.; Caligiuri, I.; Ontario, M.L.; Greco, V.; Sciuto, N.; Crea, R.; Calabrese, E.J.; Rizzolio, F.; Canzonieri, V.; et al. Redox modulation of vitagenes via plant polyphenols and vitamin D: Novel insights for chemoprevention and therapeutic interventions based on organoid technology. Mech. Ageing Dev. 2021, 199, 111551. [Google Scholar] [CrossRef] [PubMed]
- Prud’homme, G.J.; Kurt, M.; Wang, Q. Pathobiology of the Klotho Antiaging Protein and Therapeutic Considerations. Front. Aging 2022, 3, 931331. [Google Scholar] [CrossRef] [PubMed]
- Shen, J.; Shan, J.; Zhong, L.; Liang, B.; Zhang, D.; Li, M.; Tang, H. Dietary Phytochemicals that Can Extend Longevity by Regulation of Metabolism. Plant Foods Hum. Nutr. 2022, 77, 12–19. [Google Scholar] [CrossRef] [PubMed]
- Carlberg, C. Vitamin D and Its Target Genes. Nutrients 2022, 14, 1354. [Google Scholar] [CrossRef] [PubMed]
- Delrue, C.; Speeckaert, M.M. Vitamin D and Vitamin D-Binding Protein in Health and Disease. Int. J. Mol. Sci. 2023, 24, 4642. [Google Scholar] [CrossRef] [PubMed]
- Bikle, D.D. Vitamin D metabolism, mechanism of action, and clinical applications. Chem. Biol. 2014, 21, 319–329. [Google Scholar] [CrossRef] [PubMed]
- Li, J.J.; Kim, R.H.; Zhang, Q.; Ogata, Y.; Sodek, J. Characteristics of vitamin D3 receptor (VDR) binding to the vitamin D response element (VDRE) in rat bone sialoprotein gene promoter. Eur. J. Oral Sci. 1998, 106, 408–417. [Google Scholar] [CrossRef] [PubMed]
- Spanier, J.A.; Nashold, F.E.; Mayne, C.G.; Nelson, C.D.; Hayes, C.E. Vitamin D and estrogen synergy in Vdr-expressing CD4+ T cells is essential to induce Helios+FoxP3+ T cells and prevent autoimmune demyelinating disease. J. Neuroimmunol. 2015, 286, 48–58. [Google Scholar] [CrossRef]
- Cashman, K.D.; Dowling, K.G.; Škrabáková, Z.; Gonzalez-Gross, M.; Valtueña, J.; De Henauw, S.; Moreno, L.; Damsgaard, C.T.; Michaelsen, K.F.; Mølgaard, C.; et al. Vitamin D deficiency in Europe: Pandemic? Am. J. Clin. Nutr. 2016, 103, 1033–1044. [Google Scholar] [CrossRef]
- Teegarden, D.; Lyle, R.M.; Proulx, W.R.; Johnston, C.C.; Weaver, C.M. Previous milk consumption is associated with greater bone density in young women. Am. J. Clin. Nutr. 1999, 69, 1014–1017. [Google Scholar] [CrossRef]
- Lerch, C.; Meissner, T. Interventions for the prevention of nutritional rickets in term born children. Cochrane Database Syst. Rev. 2007, 2007, CD006164. [Google Scholar] [CrossRef] [PubMed]
- Dobson, R.; Cock, H.R.; Brex, P.; Giovannoni, G. Vitamin D supplementation. Pract. Neurol. 2018, 18, 35–42. [Google Scholar] [CrossRef] [PubMed]
- Pérez-López, F.R.; Brincat, M.; Erel, C.T.; Tremollieres, F.; Gambacciani, M.; Lambrinoudaki, I.; Moen, M.H.; Schenck-Gustafsson, K.; Vujovic, S.; Rozenberg, S.; et al. EMAS position statement: Vitamin D and postmenopausal health. Maturitas 2012, 71, 83–88. [Google Scholar] [CrossRef] [PubMed]
- Smith, T.J.; Tripkovic, L.; Lanham-New, S.A.; Hart, K.H. Vitamin D in adolescence: Evidence-based dietary requirements and implications for public health policy. Proc. Nutr. Soc. 2018, 77, 292–301. [Google Scholar] [CrossRef] [PubMed]
- Galmés, S.; Serra, F.; Palou, A. Current State of Evidence: Influence of Nutritional and Nutrigenetic Factors on Immunity in the COVID-19 Pandemic Framework. Nutrients 2020, 12, 2738. [Google Scholar] [CrossRef] [PubMed]
- Holick, M.F. Sunlight and vitamin D for bone health and prevention of autoimmune diseases, cancers, and cardiovascular disease. Am. J. Clin. Nutr. 2004, 80, 1678S–1688S. [Google Scholar] [CrossRef]
- Holick, M.F.; Binkley, N.C.; Bischoff-Ferrari, H.A.; Gordon, C.M.; Hanley, D.A.; Heaney, R.P.; Murad, M.H.; Weaver, C.M.; Endocrine Society. Evaluation, treatment, and prevention of vitamin D deficiency: An Endocrine Society clinical practice guideline. J. Clin. Endocrinol. Metab. 2011, 96, 1911–1930. [Google Scholar] [CrossRef]
- Holick, M.F. High prevalence of vitamin D inadequacy and implications for health. Mayo Clin. Proc. 2006, 81, 353–373. [Google Scholar] [CrossRef]
- Tong, X.; Zhang, Y.; Zhao, Y.; Li, Y.; Li, T.; Zou, H.; Yuan, Y.; Bian, J.; Liu, Z.; Gu, J. Vitamin D Alleviates Cadmium-Induced Inhibition of Chicken Bone Marrow Stromal Cells’ Osteogenic Differentiation In Vitro. Animals 2023, 13, 2544. [Google Scholar] [CrossRef] [PubMed]
- Vernia, F.; Valvano, M.; Longo, S.; Cesaro, N.; Viscido, A.; Latella, G. Vitamin D in Inflammatory Bowel Diseases. Mechanisms of Action and Therapeutic Implications. Nutrients 2022, 14, 269. [Google Scholar] [CrossRef]
- Rafique, A.; Rejnmark, L.; Heickendorff, L.; Møller, H.J. 25(OH)D3 and 1.25(OH)2D3 inhibits TNF-α expression in human monocyte derived macrophages. PLoS ONE 2019, 14, e0215383. [Google Scholar] [CrossRef] [PubMed]
- Ilie, P.C.; Stefanescu, S.; Smith, L. The role of vitamin D in the prevention of coronavirus disease 2019 infection and mortality. Aging Clin. Exp. Res. 2020, 32, 1195–1198. [Google Scholar] [CrossRef] [PubMed]
- Miyamoto, H.; Kawakami, D.; Hanafusa, N.; Nakanishi, T.; Miyasaka, M.; Furutani, Y.; Ikeda, Y.; Ito, K.; Kato, T.; Yokoyama, K.; et al. Determination of a Serum 25-Hydroxyvitamin D Reference Ranges in Japanese Adults Using Fully Automated Liquid Chromatography-Tandem Mass Spectrometry. J. Nutr. 2023, 153, 1253–1264. [Google Scholar] [CrossRef] [PubMed]
- Autier, P.; Mullie, P.; Macacu, A.; Dragomir, M.; Boniol, M.; Coppens, K.; Pizot, C.; Boniol, M. Effect of vitamin D supplementation on non-skeletal disorders: A systematic review of meta- analyses and randomised trials. Lancet Diabetes Endocrinol. 2017, 5, 986–1004. [Google Scholar] [CrossRef] [PubMed]
- Barbarawi, M.; Kheiri, B.; Zayed, Y.; Barbarawi, O.; Dhillon, H.; Swaid, B.; Yelangi, A.; Sundus, S.; Bachuwa, G.; Alkotob, M.L.; et al. Vitamin D Supplementation and Cardiovascular Disease Risks in More Than 83,000 Individuals in 21 Randomized Clinical Trials: A Meta- analysis. JAMA Cardiol. 2019, 4, 765–776. [Google Scholar] [CrossRef] [PubMed]
- Shenoy, S. Gut microbiome, Vitamin D, ACE2 interactions are critical factors in immune-senescence and inflammaging: Key for vaccine response and severity of COVID-19 infection. Inflamm. Res. 2022, 71, 13–26. [Google Scholar] [CrossRef] [PubMed]
- Dimitrov, V.; White, J.H. Vitamin D signaling in intestinal innate immunity and homeostasis. Mol. Cell. Endocrinol. 2017, 453, 68–78. [Google Scholar] [CrossRef] [PubMed]
- Kurosu, H.; Yamamoto, M.; Clark, J.D.; Pastor, J.V.; Nandi, A.; Gurnani, P.; McGuinness, O.P.; Chikuda, H.; Yamaguchi, M.; Kawaguchi, H.; et al. Suppression of aging in mice by the hormone Klotho. Science 2005, 309, 1829–1833. [Google Scholar] [CrossRef]
- Battistini, C.; Ballan, R.; Herkenhoff, M.E.; Saad, S.M.I.; Sun, J. Vitamin D Modulates Intestinal Microbiota in Inflammatory Bowel Diseases. Int. J. Mol. Sci. 2020, 22, 362. [Google Scholar] [CrossRef]
- Krutzik, S.R.; Hewison, M.; Liu, P.T.; Robles, J.A.; Stenger, S.; Adams, J.S.; Modlin, R.L. IL-15 links TLR2/1-induced macrophage differentiation to the vitamin D-dependent antimicrobial pathway. J. Immunol. 2008, 181, 7115–7120. [Google Scholar] [CrossRef]
- Takahashi, K.; Nakayama, Y.; Horiuchi, H.; Ohta, T.; Komoriya, K.; Ohmori, H.; Kamimura, T. Human neutrophils express messenger RNA of vitamin D receptor and respond to 1α,25-dihydroxyvitamin D3. Immunopharmacol. Immunotoxicol. 2002, 24, 335–347. [Google Scholar] [CrossRef]
- Li, W.; Che, X.; Chen, X.; Zhou, M.; Luo, X.; Liu, T. Study of calcitriol anti-aging effects on human natural killer cells in vitro. Bioengineered 2021, 12, 6844–6854. [Google Scholar] [CrossRef]
- Bhatt, T.; Dam, B.; Khedkar, S.U.; Lall, S.; Pandey, S.; Kataria, S.; Ajnabi, J.; Gulzar, S.E.; Dias, P.M.; Waskar, M.; et al. Niacinamide enhances cathelicidin mediated SARS-CoV-2 membrane disruption. Front. Immunol. 2023, 14, 1255478. [Google Scholar] [CrossRef]
- Wimalawansa, S.J. Infections and Autoimmunity-The Immune System and Vitamin D: A Systematic Review. Nutrients 2023, 15, 3842. [Google Scholar] [CrossRef]
- Carlberg, C.; Muñoz, A. An update on vitamin D signaling and cancer. Semin. Cancer Biol. 2022, 79, 217–230. [Google Scholar] [CrossRef]
- Pfotenhauer, K.M.; Shubrook, J.H. Vitamin D Deficiency, Its Role in Health and Disease, and Current Supplementation Recommendations. J. Am. Osteopath Assoc. 2017, 117, 301–305. [Google Scholar] [CrossRef] [PubMed]
- Kennel, K.A.; Drake, M.T.; Hurley, D.L. Vitamin D deficiency in adults: When to test and how to treat. Mayo Clin. Proc. 2010, 85, 752–758. [Google Scholar] [CrossRef] [PubMed]
- Amrein, K.; Scherkl, M.; Hoffmann, M.; Neuwersch-Sommeregger, S.; Köstenberger, M.; Tmava Berisha, A.; Martucci, G.; Pilz, S.; Malle, O. Vitamin D deficiency 2.0: An update on the current status worldwide. Eur. J. Clin. Nutr. 2020, 74, 1498–1513. [Google Scholar] [CrossRef] [PubMed]
- Vranić, L.; Mikolašević, I.; Milić, S. Vitamin D Deficiency: Consequence or Cause of Obesity? Medicina 2019, 55, 541. [Google Scholar] [CrossRef] [PubMed]
- Chen, Z.F.; Zhang, H.; Wang, H.; Matsumura, K.; Wong, Y.H.; Ravasi, T.; Qian, P.Y. Quantitative proteomics study of larval settlement in the Barnacle Balanus amphitrite. PLoS ONE 2014, 9, e88744. [Google Scholar] [CrossRef] [PubMed]
- Zhang, R.; Wang, M.; Wang, M.; Zhang, L.; Ding, Y.; Tang, Z.; Fu, Z.; Fan, H.; Zhang, W.; Wang, J. Vitamin D Level and Vitamin D Receptor Genetic Variation Were Involved in the Risk of Non-Alcoholic Fatty Liver Disease: A Case-Control Study. Front. Endocrinol. 2021, 12, 648844. [Google Scholar] [CrossRef]
- Chen, J.; Zhang, J.; Li, J.; Qin, R.; Lu, N.; Goltzman, D.; Miao, D.; Yang, R. 1,25-Dihydroxyvitamin D Deficiency Accelerates Aging-related Osteoarthritis via Downregulation of Sirt1 in Mice. Int. J. Biol. Sci. 2023, 19, 610–624. [Google Scholar] [CrossRef] [PubMed]
- Capozzi, A.; Scambia, G.; Lello, S. Calcium, vitamin D, vitamin K2, and magnesium supplementation and skeletal health. Maturitas 2020, 140, 55–63. [Google Scholar] [CrossRef] [PubMed]
- Sekikawa, A.; Wharton, W.; Butts, B.; Veliky, C.V.; Garfein, J.; Li, J.; Goon, S.; Fort, A.; Li, M.; Hughes, T.M. Potential Protective Mechanisms of S-equol, a Metabolite of Soy Isoflavone by the Gut Microbiome, on Cognitive Decline and Dementia. Int. J. Mol. Sci. 2022, 23, 11921. [Google Scholar] [CrossRef]
- Zhang, D.; Xu, Y.; Chen, H.; Wang, D.; Geng, Z.; Chen, Y.; Chen, Y.; Xiong, D.; Yang, R.; Liu, X.; et al. Fagopyrum dibotrys extract alleviates hepatic steatosis and insulin resistance, and alters autophagy and gut microbiota diversity in mouse models of high-fat diet-induced non-alcoholic fatty liver disease. Front. Nutr. 2022, 9, 993501. [Google Scholar] [CrossRef] [PubMed]
- Qin, J.; Li, Y.; Cai, Z.; Li, S.; Zhu, J.; Zhang, F.; Liang, S.; Zhang, W.; Guan, Y.; Shen, D.; et al. A metagenome-wide association study of gut microbiota in type 2 diabetes. Nature 2012, 490, 55–60. [Google Scholar] [CrossRef] [PubMed]
- Karlsson, F.H.; Tremaroli, V.; Nookaew, I.; Bergström, G.; Behre, C.J.; Fagerberg, B.; Nielsen, J.; Bäckhed, F. Gut metagenome in European women with normal, impaired and diabetic glucose control. Nature 2013, 498, 99–103. [Google Scholar] [CrossRef] [PubMed]
- Hashimoto, Y.; Hamaguchi, M.; Kaji, A.; Sakai, R.; Osaka, T.; Inoue, R.; Kashiwagi, S.; Mizushima, K.; Uchiyama, K.; Takagi, T.; et al. Intake of sucrose affects gut dysbiosis in patients with type 2 diabetes. J. Diabetes Investig. 2020, 11, 1623–1634. [Google Scholar] [CrossRef]
- Tangestani, H.; Boroujeni, H.K.; Djafarian, K.; Emamat, H.; Shab-Bidar, S. Vitamin D and The Gut Microbiota: A Narrative Literature Review. Clin. Nutr. Res. 2021, 10, 181–191. [Google Scholar] [CrossRef]
- Bellerba, F.; Muzio, V.; Gnagnarella, P.; Facciotti, F.; Chiocca, S.; Bossi, P.; Cortinovis, D.; Chiaradonna, F.; Serrano, D.; Raimondi, S.; et al. The Association between Vitamin D and Gut Microbiota: A Systematic Review of Human Studies. Nutrients 2021, 13, 3378. [Google Scholar] [CrossRef]
- Li, Q.; Chan, H.; Liu, W.X.; Liu, C.A.; Zhou, Y.; Huang, D.; Wang, X.; Li, X.; Xie, C.; Liu, W.Y.; et al. Carnobacterium maltaromaticum boosts intestinal vitamin D production to suppress colorectal cancer in female mice. Cancer Cell 2023, 41, 1450–1465.e8. [Google Scholar] [CrossRef]
- Odamaki, T.; Kato, K.; Sugahara, H.; Hashikura, N.; Takahashi, S.; Xiao, J.Z.; Abe, F.; Osawa, R. Age-related changes in gut microbiota composition from newborn to centenarian: A cross-sectional study. BMC Microbiol. 2016, 16, 90. [Google Scholar] [CrossRef]
- Johansen, J.; Atarashi, K.; Arai, Y.; Hirose, N.; Sørensen, S.J.; Vatanen, T.; Knip, M.; Honda, K.; Xavier, R.J.; Rasmussen, S.; et al. Centenarians have a diverse gut virome with the potential to modulate metabolism and promote healthy lifespan. Nat. Microbiol. 2023, 8, 1064–1078. [Google Scholar] [CrossRef]
- Yoshimoto, S.; Mitsuyama, E.; Yoshida, K.; Odamaki, T.; Xiao, J.Z. Enriched metabolites that potentially promote age-associated diseases in subjects with an elderly-type gut microbiota. Gut Microbes 2021, 13, 1865705. [Google Scholar] [CrossRef]
- Sato, Y.; Atarashi, K.; Plichta, D.R.; Arai, Y.; Sasajima, S.; Kearney, S.M.; Suda, W.; Takeshita, K.; Sasaki, T.; Okamoto, S.; et al. Novel bile acid biosynthetic pathways are enriched in the microbiome of centenarians. Nature 2021, 599, 458–464. [Google Scholar] [CrossRef]
- Santa, K.; Kumazawa, Y.; Nagaoka, I. Prevention of Metabolic Syndrome by Phytochemicals and Vitamin D. Int. J. Mol. Sci. 2023, 24, 2627. [Google Scholar] [CrossRef]
- Santa, K.; Kumazawa, Y.; Nagaoka, I. The Potential Use of Grape Phytochemicals for Preventing the Development of Intestine-Related and Subsequent Inflammatory Diseases. Endocr. Metab. Immune Disord. Drug Targets 2019, 19, 794–802. [Google Scholar] [CrossRef] [PubMed]
- Santa, K.; Watanabe, K.; Kumazawa, Y.; Nagaoka, I. Phytochemicals and Vitamin D for a Healthy Life and Prevention of Diseases. Int. J. Mol. Sci. 2023, 24, 12167. [Google Scholar] [CrossRef] [PubMed]
- Santa, K. Grape Phytochemicals and Vitamin D in the Alleviation of Lung Disorders. Endocr. Metab. Immune Disord. Drug Targets 2022, 22, 1276–1292. [Google Scholar] [CrossRef] [PubMed]
- Santa, K. Healthy Diet, Grape Phytochemicals, and Vitamin D: Preventing Chronic Inflammation and Keeping Good Microbiota. Endocr. Metab. Immune Disord. Drug Targets 2023, 23, 777–800. [Google Scholar] [CrossRef] [PubMed]
- Trullemans, L.; Koelewijn, S.F.; Boonen, I.; Cooreman, E.; Hendrickx, T.; Preegel, G.; Van Aelst, J.; Witters, H.; Elskens, M.; Van Puyvelde, P.; et al. Renewable and safer bisphenol A substitutes enabled by selective zeolite alkylation. Nat. Sustain. 2023, 6, 1693–1704. [Google Scholar] [CrossRef]
- Marko, M.; Pawliczak, R. Resveratrol and Its Derivatives in Inflammatory Skin Disorders-Atopic Dermatitis and Psoriasis: A Review. Antioxidants 2023, 12, 1954. [Google Scholar] [CrossRef] [PubMed]
- Bernatoniene, J.; Kopustinskiene, D.M. The Role of Catechins in Cellular Responses to Oxidative Stress. Molecules 2018, 23, 965. [Google Scholar] [CrossRef] [PubMed]
- Shan, Z.; Nisar, M.F.; Li, M.; Zhang, C.; Wan, C.C. Theaflavin Chemistry and Its Health Benefits. Oxidative Med. Cell. Longev. 2021, 2021, 6256618. [Google Scholar] [CrossRef]
- Alappat, B.; Alappat, J. Anthocyanin Pigments: Beyond Aesthetics. Molecules 2020, 25, 5500. [Google Scholar] [CrossRef]
- Xiong, H.; Wang, J.; Ran, Q.; Lou, G.; Peng, C.; Gan, Q.; Hu, J.; Sun, J.; Yao, R.; Huang, Q. Hesperidin: A Therapeutic Agent For Obesity. Drug Des. Dev. Ther. 2019, 13, 3855–3866. [Google Scholar] [CrossRef] [PubMed]
- Křížová, L.; Dadáková, K.; Kašparovská, J.; Kašparovský, T. Isoflavones. Molecules 2019, 24, 1076. [Google Scholar] [CrossRef] [PubMed]
- Chung, B.H.; Lee, J.J.; Kim, J.D.; Jeoung, D.; Lee, H.; Choe, J.; Ha, K.S.; Kwon, Y.G.; Kim, Y.M. Angiogenic activity of sesamin through the activation of multiple signal pathways. Biochem. Biophys. Res. Commun. 2010, 391, 254–260. [Google Scholar] [CrossRef]
- Li, S.; Yin, S.; Ding, H.; Shao, Y.; Zhou, S.; Pu, W.; Han, L.; Wang, T.; Yu, H. Polyphenols as potential metabolism mechanisms regulators in liver protection and liver cancer prevention. Cell Prolif. 2023, 56, e13346. [Google Scholar] [CrossRef]
- Lawson, L.D.; Hunsaker, S.M. Allicin Bioavailability and Bioequivalence from Garlic Supplements and Garlic Foods. Nutrients 2018, 10, 812. [Google Scholar] [CrossRef]
- Milani, A.; Basirnejad, M.; Shahbazi, S.; Bolhassani, A. Carotenoids: Biochemistry, pharmacology and treatment. Br. J. Pharmacol. 2017, 174, 1290–1324. [Google Scholar] [CrossRef]
- Castellano, J.M.; Ramos-Romero, S.; Perona, J.S. Oleanolic Acid: Extraction, Characterization and Biological Activity. Nutrients 2022, 14, 623. [Google Scholar] [CrossRef]
- Alemi, F.; Poole, D.P.; Chiu, J.; Schoonjans, K.; Cattaruzza, F.; Grider, J.R.; Bunnett, N.W.; Corvera, C.U. The receptor TGR5 mediates the prokinetic actions of intestinal bile acids and is required for normal defecation in mice. Gastroenterology 2013, 144, 145–154. [Google Scholar] [CrossRef]
- Chávez-Talavera, O.; Tailleux, A.; Lefebvre, P.; Staels, B. Bile Acid Control of Metabolism and Inflammation in Obesity, Type 2 Diabetes, Dyslipidemia, and Nonalcoholic Fatty Liver Disease. Gastroenterology 2017, 152, 1679–1694.e3. [Google Scholar] [CrossRef]
- Sakuma, K.; Hamada, K.; Yamaguchi, A.; Aoi, W. Current Nutritional and Pharmacological Approaches for Attenuating Sarcopenia. Cells 2023, 12, 2422. [Google Scholar] [CrossRef] [PubMed]
- Langi, P.; Kiokias, S.; Varzakas, T.; Proestos, C. Carotenoids: From Plants to Food and Feed Industries. Methods Mol. Biol. 2018, 1852, 57–71. [Google Scholar] [PubMed]
- Dewett, D.; Lam-Kamath, K.; Poupault, C.; Khurana, H.; Rister, J. Mechanisms of vitamin A metabolism and deficiency in the mammalian and fly visual system. Dev. Biol. 2021, 476, 68–78. [Google Scholar] [CrossRef] [PubMed]
- Serafini, M.; Peluso, I.; Raguzzini, A. Flavonoids as anti-inflammatory agents. Proc. Nutr. Soc. 2010, 69, 273–278. [Google Scholar] [CrossRef] [PubMed]
- Deepika Maurya, P.K. Health Benefits of Quercetin in Age-Related Diseases. Molecules 2022, 27, 2498. [Google Scholar] [CrossRef] [PubMed]
- 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] [PubMed]
- Sannappa Gowda, N.G.; Shiragannavar, V.D.; Puttahanumantharayappa, L.D.; Shivakumar, A.T.; Dallavalasa, S.; Basavaraju, C.G.; Bhat, S.S.; Prasad, S.K.; Vamadevaiah, R.M.; Madhunapantula, S.V.; et al. Quercetin activates vitamin D receptor and ameliorates breast cancer induced hepatic inflammation and fibrosis. Front. Nutr. 2023, 10, 1158633. [Google Scholar] [CrossRef]
- Kawaguchi, K.; Kikuchi, S.; Hasunuma, R.; Maruyama, H.; Yoshikawa, T.; Kumazawa, Y. A citrus flavonoid hesperidin suppresses infection-induced endotoxin shock in mice. Biol. Pharm. Bull. 2004, 27, 679–683. [Google Scholar] [CrossRef]
- Xie, J.; Yang, Z.; Zhou, C.; Zhu, J.; Lee, R.J.; Teng, L. Nanotechnology for the delivery of phytochemicals in cancer therapy. Biotechnol. Adv. 2016, 34, 343–353. [Google Scholar] [CrossRef]
- Kim, B.; Park, J.E.; Im, E.; Cho, Y.; Lee, J.; Lee, H.J.; Sim, D.Y.; Park, W.Y.; Shim, B.S.; Kim, S.H. Recent Advances in Nanotechnology with Nano-Phytochemicals: Molecular Mechanisms and Clinical Implications in Cancer Progression. Int. J. Mol. Sci. 2021, 22, 3571. [Google Scholar] [CrossRef]
- Pyrzynska, K. Hesperidin: A Review on Extraction Methods, Stability and Biological Activities. Nutrients 2022, 14, 2387. [Google Scholar] [CrossRef]
- Amaretti, A.; Raimondi, S.; Leonardi, A.; Quartieri, A.; Rossi, M. Hydrolysis of the rutinose-conjugates flavonoids rutin and hesperidin by the gut microbiota and bifidobacteria. Nutrients 2015, 7, 2788–2800. [Google Scholar] [CrossRef]
- Kawai, Y.; Nishikawa, T.; Shiba, Y.; Saito, S.; Murota, K.; Shibata, N.; Kobayashi, M.; Kanayama, M.; Uchida, K.; Terao, J. Macrophage as a target of quercetin glucuronides in human atherosclerotic arteries: Implication in the anti-atherosclerotic mechanism of dietary flavonoids. J. Biol. Chem. 2008, 283, 9424–9434. [Google Scholar] [CrossRef] [PubMed]
- Gagliardi, S.; Franco, V.; Sorrentino, S.; Zucca, S.; Pandini, C.; Rota, P.; Bernuzzi, S.; Costa, A.; Sinforiani, E.; Pansarasa, O.; et al. Curcumin and Novel Synthetic Analogs in Cell-Based Studies of Alzheimer’s Disease. Front. Pharmacol. 2018, 9, 1404. [Google Scholar] [CrossRef] [PubMed]
- Bartik, L.; Whitfield, G.K.; Kaczmarska, M.; Lowmiller, C.L.; Moffet, E.W.; Furmick, J.K.; Hernandez, Z.; Haussler, C.A.; Haussler, M.R.; Jurutka, P.W. Curcumin: A novel nutritionally derived ligand of the vitamin D receptor with implications for colon cancer chemoprevention. J. Nutr. Biochem. 2010, 21, 1153–1161. [Google Scholar] [CrossRef] [PubMed]
- Haussler, M.R.; Whitfield, G.K.; Kaneko, I.; Haussler, C.A.; Hsieh, D.; Hsieh, J.C.; Jurutka, P.W. Molecular mechanisms of vitamin D action. Calcif. Tissue Int. 2013, 92, 77–98. [Google Scholar] [CrossRef] [PubMed]
- Farghali, M.; Ruga, S.; Morsanuto, V.; Uberti, F. Can Brain Health Be Supported by Vitamin D-Based Supplements? A Critical Review. Brain Sci. 2020, 10, 660. [Google Scholar] [CrossRef]
- Kaneko, M.; Takimoto, H.; Sugiyama, T.; Seki, Y.; Kawaguchi, K.; Kumazawa, Y. Suppressive effects of the flavonoids quercetin and luteolin on the accumulation of lipid rafts after signal transduction via receptors. Immunopharmacol. Immunotoxicol. 2008, 30, 867–882. [Google Scholar] [CrossRef] [PubMed]
- Kondo, Y.; Ikeda, K.; Tokuda, N.; Nishitani, C.; Ohto, U.; Akashi-Takamura, S.; Ito, Y.; Uchikawa, M.; Kuroki, Y.; Taguchi, R.; et al. TLR4-MD-2 complex is negatively regulated by an endogenous ligand, globotetraosylceramide. Proc. Natl. Acad. Sci. USA 2013, 110, 4714–4719. [Google Scholar] [CrossRef]
- Wang, Q.; Lin, H.; Shen, C.; Zhang, M.; Wang, X.; Yuan, M.; Yuan, M.; Jia, S.; Cao, Z.; Wu, C.; et al. Gut microbiota regulates postprandial GLP-1 response via ileal bile acid-TGR5 signaling. Gut Microbes 2023, 15, 2274124. [Google Scholar] [CrossRef]
- Wei, H.J.; Pareek, T.K.; Liu, Q.; Letterio, J.J. A unique tolerizing dendritic cell phenotype induced by the synthetic triterpenoid CDDO-DFPA (RTA-408) is protective against EAE. Sci. Rep. 2017, 7, 9886. [Google Scholar] [CrossRef]
- Li, W.; Zeng, H.; Xu, M.; Huang, C.; Tao, L.; Li, J.; Zhang, T.; Chen, H.; Xia, J.; Li, C.; et al. Oleanolic Acid Improves Obesity-Related Inflammation and Insulin Resistance by Regulating Macrophages Activation. Front. Pharmacol. 2021, 12, 697483. [Google Scholar] [CrossRef]
- Welch, A.A.; Hardcastle, A.C. The effects of flavonoids on bone. Curr. Osteoporos. Rep. 2014, 12, 205–210. [Google Scholar] [CrossRef]
- Maeda-Yamamoto, M.; Ohtani, T. Development of functional agricultural products utilizing the new health claim labeling system in Japan. Biosci. Biotechnol. Biochem. 2018, 82, 554–563. [Google Scholar] [CrossRef]
- Campisi, J.; Kapahi, P.; Lithgow, G.J.; Melov, S.; Newman, J.C.; Verdin, E. From discoveries in ageing research to therapeutics for healthy ageing. Nature 2019, 571, 183–192. [Google Scholar] [CrossRef]
- Bhatti, J.S.; Bhatti, G.K.; Reddy, P.H. Mitochondrial dysfunction and oxidative stress in metabolic disorders—A step towards mitochondria based therapeutic strategies. Biochim. Biophys. Acta Mol. Basis Dis. 2017, 1863, 1066–1077. [Google Scholar] [CrossRef]
- Rando, T.A.; Chang, H.Y. Aging, rejuvenation, and epigenetic reprogramming: Resetting the aging clock. Cell 2012, 148, 46–57. [Google Scholar] [CrossRef]
- Kondratova, A.A.; Kondratov, R.V. The circadian clock and pathology of the ageing brain. Nat. Rev. Neurosci. 2012, 13, 325–335. [Google Scholar] [CrossRef] [PubMed]
- Napoleão, A.; Fernandes, L.; Miranda, C.; Marum, A.P. Effects of Calorie Restriction on Health Span and Insulin Resistance: Classic Calorie Restriction Diet vs. Ketosis-Inducing Diet. Nutrients 2021, 13, 1302. [Google Scholar] [CrossRef]
- Guarente, L.; Picard, F. Calorie restriction--the SIR2 connection. Cell 2005, 120, 473–482. [Google Scholar] [CrossRef]
- Tao, Z.; Jin, Z.; Wu, J.; Cai, G.; Yu, X. Sirtuin family in autoimmune diseases. Front. Immunol. 2023, 14, 1186231. [Google Scholar] [CrossRef] [PubMed]
- Ma, L.; Wang, R.; Wang, H.; Zhang, Y.; Zhao, Z. Long-term caloric restriction activates the myocardial SIRT1/AMPK/PGC-1α pathway in C57BL/6J male mice. Food Nutr. Res. 2020, 64, 3668. [Google Scholar] [CrossRef]
- Chen, M.; Huang, N.; Liu, J.; Huang, J.; Shi, J.; Jin, F. AMPK: A bridge between diabetes mellitus and Alzheimer’s disease. Behav. Brain Res. 2021, 400, 113043. [Google Scholar] [CrossRef]
- Wu, Q.J.; Zhang, T.N.; Chen, H.H.; Yu, X.F.; Lv, J.L.; Liu, Y.Y.; Liu, Y.S.; Zheng, G.; Zhao, J.Q.; Wei, Y.F.; et al. The sirtuin family in health and disease. Signal Transduct. Target. Ther. 2022, 7, 402. [Google Scholar] [CrossRef]
- Lee, S.H.; Lee, J.H.; Lee, H.Y.; Min, K.J. Sirtuin signaling in cellular senescence and aging. BMB Rep. 2019, 52, 24–34. [Google Scholar] [CrossRef] [PubMed]
- Bielach-Bazyluk, A.; Zbroch, E.; Mysliwiec, H.; Rydzewska-Rosolowska, A.; Kakareko, K.; Flisiak, I.; Hryszko, T. Sirtuin 1 and Skin: Implications in Intrinsic and Extrinsic Aging-A Systematic Review. Cells 2021, 10, 813. [Google Scholar] [CrossRef] [PubMed]
- Ghamar Talepoor, A.; Doroudchi, M. Immunosenescence in atherosclerosis: A role for chronic viral infections. Front. Immunol. 2022, 13, 945016. [Google Scholar] [CrossRef] [PubMed]
- Ogrodnik, M.; Evans, S.A.; Fielder, E.; Victorelli, S.; Kruger, P.; Salmonowicz, H.; Weigand, B.M.; Patel, A.D.; Pirtskhalava, T.; Inman, C.L.; et al. Whole-body senescent cell clearance alleviates age-related brain inflammation and cognitive impairment in mice. Aging Cell 2021, 20, e13296. [Google Scholar] [CrossRef]
- 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]
- Ma, X.R.; Zhu, X.; Xiao, Y.; Gu, H.M.; Zheng, S.S.; Li, L.; Wang, F.; Dong, Z.J.; Wang, D.X.; Wu, Y.; et al. Restoring nuclear entry of Sirtuin 2 in oligodendrocyte progenitor cells promotes remyelination during ageing. Nat. Commun. 2022, 13, 1225. [Google Scholar] [CrossRef]
- Absalon, C.; Fabre, S.; Tarascou, I.; Fouquet, E.; Pianet, I. New strategies to study the chemical nature of wine oligomeric procyanidins. Anal. Bioanal. Chem. 2011, 401, 1485–1495. [Google Scholar] [CrossRef]
- Gill, V.; Kumar, V.; Singh, K.; Kumar, A.; Kim, J.J. Advanced Glycation End Products (AGEs) May Be a Striking Link Between Modern Diet and Health. Biomolecules 2019, 9, 888. [Google Scholar] [CrossRef] [PubMed]
- Filošević Vujnović, A.; Jović, K.; Pištan, E.; Andretić Waldowski, R. Influence of Dopamine on Fluorescent Advanced Glycation End Products Formation Using Drosophila melanogaster. Biomolecules 2021, 11, 453. [Google Scholar] [CrossRef] [PubMed]
- Münch, G.; Kuhla, B.; Lüth, H.J.; Arendt, T.; Robinson, S.R. Anti-AGEing defences against Alzheimer’s disease. Biochem. Soc. Trans. 2003, 31, 1397–1399. [Google Scholar] [CrossRef]
- Szczechowiak, K.; Diniz, B.S.; Leszek, J. Diet and Alzheimer’s dementia—Nutritional approach to modulate inflammation. Pharmacol. Biochem. Behav. 2019, 184, 172743. [Google Scholar] [CrossRef]
- Zhou, M.; Graves, D.T. Impact of the host response and osteoblast lineage cells on periodontal disease. Front. Immunol. 2022, 13, 998244. [Google Scholar] [CrossRef]
- Borsa, L.; Dubois, M.; Sacco, G.; Lupi, L. Analysis the Link between Periodontal Diseases and Alzheimer’s Disease: A Systematic Review. Int. J. Environ. Res. Public Health 2021, 18, 9312. [Google Scholar] [CrossRef]
- Mahomoodally, M.F.; Aumeeruddy, M.Z.; Legoabe, L.J.; Dall’Acqua, S.; Zengin, G. Plants’ bioactive secondary metabolites in the management of sepsis: Recent findings on their mechanism of action. Front. Pharmacol. 2022, 13, 1046523. [Google Scholar] [CrossRef] [PubMed]
- Bostanci, N.; Bao, K.; Greenwood, D.; Silbereisen, A.; Belibasakis, G.N. Periodontal disease: From the lenses of light microscopy to the specs of proteomics and next-generation sequencing. Adv. Clin. Chem. 2019, 93, 263–290. [Google Scholar] [PubMed]
- Maria, S.; Swanson, M.H.; Enderby, L.T.; D’Amico, F.; Enderby, B.; Samsonraj, R.M.; Dudakovic, A.; van Wijnen, A.J.; Witt-Enderby, P.A. Melatonin-micronutrients Osteopenia Treatment Study (MOTS): A translational study assessing melatonin, strontium (citrate), vitamin D3 and vitamin K2 (MK7) on bone density, bone marker turnover and health related quality of life in postmenopausal osteopenic women following a one-year double-blind RCT and on osteoblast-osteoclast co-cultures. Aging 2017, 9, 256–285. [Google Scholar] [PubMed]
- Nakamura, Y.; Suzuki, T.; Kamimura, M.; Ikegami, S.; Uchiyama, S.; Kato, H. Alfacalcidol Increases the Therapeutic Efficacy of Ibandronate on Bone Mineral Density in Japanese Women with Primary Osteoporosis. Tohoku J. Exp. Med. 2017, 241, 319–326. [Google Scholar] [CrossRef] [PubMed]
- Harriden, B.; D’Cunha, N.M.; Kellett, J.; Isbel, S.; Panagiotakos, D.B.; Naumovski, N. Are dietary patterns becoming more processed? The effects of different dietary patterns on cognition: A review. Nutr. Health 2022, 28, 341–356. [Google Scholar] [CrossRef] [PubMed]
- D’Alessandro, A.; De Pergola, G. Mediterranean diet pyramid: A proposal for Italian people. Nutrients 2014, 6, 4302–4316. [Google Scholar] [CrossRef] [PubMed]
- Davis, C.; Bryan, J.; Hodgson, J.; Murphy, K. Definition of the Mediterranean Diet; a Literature Review. Nutrients 2015, 7, 9139–9153. [Google Scholar] [CrossRef] [PubMed]
- Mazza, E.; Ferro, Y.; Pujia, R.; Mare, R.; Maurotti, S.; Montalcini, T.; Pujia, A. Mediterranean Diet in Healthy Aging. J. Nutr. Health Aging 2021, 25, 1076–1083. [Google Scholar] [CrossRef]
- Lucerón-Lucas-Torres, M.; Saz-Lara, A.; Díez-Fernández, A.; Martínez-García, I.; Martínez-Vizcaíno, V.; Cavero-Redondo, I.; Álvarez-Bueno, C. Association between Wine Consumption with Cardiovascular Disease and Cardiovascular Mortality: A Systematic Review and Meta-Analysis. Nutrients 2023, 15, 2785. [Google Scholar] [CrossRef]
- Detopoulou, P.; Demopoulos, C.A.; Antonopoulou, S. Micronutrients, Phytochemicals and Mediterranean Diet: A Potential Protective Role against COVID-19 through Modulation of PAF Actions and Metabolism. Nutrients 2021, 13, 462. [Google Scholar] [CrossRef] [PubMed]
- Naureen, Z.; Dhuli, K.; Donato, K.; Aquilanti, B.; Velluti, V.; Matera, G.; Iaconelli, A.; Bertelli, M. Foods of the Mediterranean diet: Citrus, cucumber and grape. J. Prev. Med. Hyg. 2022, 63, E21–E27. [Google Scholar] [PubMed]
- Kushida, M.; Sugawara, S.; Asano, M.; Yamamoto, K.; Fukuda, S.; Tsuduki, T. Effects of the 1975 Japanese diet on the gut microbiota in younger adults. J. Nutr. Biochem. 2019, 64, 121–127. [Google Scholar] [CrossRef] [PubMed]
- Johmura, Y.; Yamanaka, T.; Omori, S.; Wang, T.W.; Sugiura, Y.; Matsumoto, M.; Suzuki, N.; Kumamoto, S.; Yamaguchi, K.; Hatakeyama, S.; et al. Senolysis by glutaminolysis inhibition ameliorates various age-associated disorders. Science 2021, 371, 265–270. [Google Scholar] [CrossRef] [PubMed]
- Hiraoka, M.; Kagawa, Y. Genetic polymorphisms and folate status. Congenit. Anom. 2017, 57, 142–149. [Google Scholar] [CrossRef] [PubMed]
- Kagawa, Y.; Hiraoka, M.; Kageyama, M.; Kontai, Y.; Yurimoto, M.; Nishijima, C.; Sakamoto, K. Medical cost savings in Sakado City and worldwide achieved by preventing disease by folic acid fortification. Congenit. Anom. 2017, 57, 157–165. [Google Scholar] [CrossRef] [PubMed]
- Parletta, N.; Zarnowiecki, D.; Cho, J.; Wilson, A.; Bogomolova, S.; Villani, A.; Itsiopoulos, C.; Niyonsenga, T.; Blunden, S.; Meyer, B.; et al. A Mediterranean-style dietary intervention supplemented with fish oil improves diet quality and mental health in people with depression: A randomized controlled trial (HELFIMED). Nutr. Neurosci. 2019, 22, 474–487. [Google Scholar] [CrossRef]
- Von Schacky, C. Importance of EPA and DHA Blood Levels in Brain Structure and Function. Nutrients 2021, 13, 1074. [Google Scholar] [CrossRef]
- Wu, X.; Le, T.K.; Maeda-Minami, A.; Yoshino, T.; Horiba, Y.; Mimura, M.; Watanabe, K. Relationship Between Conventional Medicine Chapters in ICD-10 and Kampo Pattern Diagnosis: A Cross-Sectional Study. Front. Pharmacol. 2021, 12, 751403. [Google Scholar] [CrossRef]
- Maeda-Minami, A.; Ihara, K.; Yoshino, T.; Horiba, Y.; Mimura, M.; Watanabe, K. A prediction model of qi stagnation: A prospective observational study referring to two existing models. Comput. Biol. Med. 2022, 146, 105619. [Google Scholar] [CrossRef]
- Maeda-Minami, A.; Yoshino, T.; Horiba, Y.; Nakamura, T.; Watanabe, K. Inter-Rater Reliability of Kampo Diagnosis for Chronic Diseases. J. Altern. Complement. Med. 2021, 27, 613–616. [Google Scholar] [CrossRef] [PubMed]
Flavonoids | Effects | Ref. |
---|---|---|
Quercetin, naringin, nobiletin | Anti-inflammatory effects in metabolic syndrome prevention | [66] |
Flavan-3-ols, flavon-3-ols, anthocyanidins | Anti-inflammatory effects in intestinal disorders | [67] |
Quercetin, hesperidin, nobiletin, rutin | Anti-inflammatory effects for healthy longevity and prevention of “ME-BYO” | [68] |
Catechins | Cellular antioxidant properties | [73] |
Theaflavin | Health-related effects and chemistry | [74] |
Anthocyanin | Pharmaceutical and nutraceutical effects other than use as colour pigments | [75] |
Hesperidin | Hesperidin in obesity treatment | [76] |
Isoflavone | Therapeutic effects from oestrogen activity | [77] |
Sesamin | Effects of sesamin in angiogenic processes | [78] |
Flavonoids | Anti-inflammation effects of flavonoids | [88] |
Quercetin | Anti-ageing effects | [89] |
Quercetin, fisetin | Effects of flavonoids in cancer chemotherapy | [90] |
Quercetin, luteolin | Suppression of TNF-α production of flavonoids in cellular process | [91] |
Hesperidin | Suppression of endotoxic shock | [92] |
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content. |
© 2024 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (https://creativecommons.org/licenses/by/4.0/).
Share and Cite
Santa, K.; Kumazawa, Y.; Watanabe, K.; Nagaoka, I. The Potential Use of Vitamin D3 and Phytochemicals for Their Anti-Ageing Effects. Int. J. Mol. Sci. 2024, 25, 2125. https://doi.org/10.3390/ijms25042125
Santa K, Kumazawa Y, Watanabe K, Nagaoka I. The Potential Use of Vitamin D3 and Phytochemicals for Their Anti-Ageing Effects. International Journal of Molecular Sciences. 2024; 25(4):2125. https://doi.org/10.3390/ijms25042125
Chicago/Turabian StyleSanta, Kazuki, Yoshio Kumazawa, Kenji Watanabe, and Isao Nagaoka. 2024. "The Potential Use of Vitamin D3 and Phytochemicals for Their Anti-Ageing Effects" International Journal of Molecular Sciences 25, no. 4: 2125. https://doi.org/10.3390/ijms25042125
APA StyleSanta, K., Kumazawa, Y., Watanabe, K., & Nagaoka, I. (2024). The Potential Use of Vitamin D3 and Phytochemicals for Their Anti-Ageing Effects. International Journal of Molecular Sciences, 25(4), 2125. https://doi.org/10.3390/ijms25042125