- 1Department of Vascular Surgery, The Second Hospital of Jilin University, Changchun, China
- 2Department of Surgery, Changchun University of Chinese Medicine, Changchun, China
- 3Department of Biomedical Science and Engineering, Gwangju Institute of Science and Technology, Gwangju, Republic of Korea
- 4Korean Medicine Application Center, Korea Institute of Oriental Medicine, Daegu, Republic of Korea
- 5Department of Nephrology, Shandong Provincial Hospital Affiliated to Shandong First Medical University, Jinan, Shandong, China
- 6Department of Dermatology, Qingdao Women and Children’s Hospital, Qingdao University, Qingdao, Shandong, China
- 7Department of Korean Medical Science, School of Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, Republic of Korea
- 8Research Institute for Korean Medicine, Pusan National University, Yangsan, Gyeongsangnam-do, Republic of Korea
Developing treatments for age-related diseases requires cost-effective and efficient approaches. Nutrients and natural metabolites offer safer alternatives to synthetic drugs. Aging increases the need for solutions that protect health and repair cells. Recent studies show that nutrients and natural products reduce oxidative stress, regulate metabolism, and influence longevity-related genes. This review focuses on vitamins, minerals, antioxidants, and natural products that improve healthspan and combat aging. It also discusses challenges such as standardization, clinical validation, and regulatory approval. Finally, emerging trends, such as personalized nutrition and advanced delivery systems, highlight the potential of these metabolites for addressing aging.
1 Introduction
The increasing number of aged people in the world is causing more problems, especially diseases related to age (Prince et al., 2015; Jaul and Barron, 2017). This situation puts a lot of pressure on healthcare systems (Organization, 2001; Evans and Stoddart, 2017). Therefore, new treatment development is needed strongly that make life longer and improve the quality (Partridge et al., 2018). The increasing prevalence of age-related diseases, such as cardiovascular disorders and neurodegeneration, underscores the need for sustainable solutions (Collaborators G. A., 2022; Collaborators G. M. D., 2022). These solutions should focus on improving both lifespan and healthspan (López-Otín et al., 2013). To solve this issue, research on nutrients and natural products is becoming an important area (Cragg and Newman, 2013; Li et al., 2016c). Natural metabolites are good alternatives to synthetic drugs because synthetic drugs are effective but often cause side effects (Newman et al., 2003; Atanasov et al., 2015). Natural products from food and traditional medicine are safer to use because they do not cause side effects and can still improve health (Veeresham, 2012; Ekor, 2014).
To address these challenges, this review investigates the role of nutrients and natural products in aging. Researchers are studying natural metabolite functions that protect and repair aging cells at the molecular level (Choi and Friso, 2010; Yuan et al., 2016). These metabolites work to reduce oxidative stress, improve metabolism, and regulate aging-related genes (Harman, 1992; Chen et al., 2016). Recently aging studies aim to delay age-related conditions and reverse some cellular changes (López-Otín et al., 2013; Ristow and Schmeisser, 2014).
In this context, the paper focuses on how vitamins, minerals, antioxidants, and botanical drugs can prevent or reverse cellular damage caused by aging (Calder, 2017). These approaches represent a shift in healthcare, where the focus is moving from treating symptoms to preventing diseases (Kaeberlein et al., 2015). This review examines the increasing interest in using nutrients and natural products as potential drugs (Cragg and Newman, 2013; Atanasov et al., 2015). It also explains their important role in healthcare by analyzing recent research trends (Li et al., 2016c; Yuan et al., 2016).
This review focuses on vitamins and minerals with well-documented roles in aging-related pathways, particularly their effects on oxidative stress regulation, immune modulation, mitochondrial function, and neuroprotection. Metabolites were selected based on their mechanistic insights and clinical relevance, with priority given to randomized controlled trials (RCTs), preclinical studies, and systematic reviews/meta-analyses that provided mechanistic or clinical insights into aging interventions. Case reports, narrative reviews, and studies with insufficient methodological details were excluded.
Moreover, this review explores how natural products can be integrated into mainstream medical practices, while also addressing key challenges such as standardization, clinical validation, and regulatory approval. By adopting this approach, we aimed to provide a balanced evaluation of their therapeutic potential in aging-related pathways while discussing their translational challenges.
2 The role of nutrients in anti-aging drug development
Nutrients, the building blocks of life, play a crucial role in maintaining cellular health and preventing age-related decline (Kennedy et al., 2014; Ames, 2018). This section explores various vitamins, minerals, and antioxidants that have shown promise in slowing aging processes. Key nutrients like Vitamin C, Vitamin E, selenium, and omega-3 fatty acids are highlighted for their antioxidative and anti-inflammatory properties, which contribute to cellular longevity and resilience (Rayman, 2012; Li et al., 2016c; Calder, 2017). Recent studies illustrate how these nutrients not only prevent cellular damage but also enhance the efficacy of other therapeutic products, offering a dual advantage in anti-aging therapy (Traber and Atkinson, 2007; Ames, 2018). The discussion extends to the mechanisms through which these nutrients influence genetic pathways associated with aging, such as DNA repair and metabolic regulation (Harman, 2003; López-Otín et al., 2013). These nutrients and their effects and mechanisms are summarized in Table 1.
2.1 Omega-3 fatty acids
Omega-3 fatty acids are essential polyunsaturated fats primarily found in fish oil, flaxseeds, and walnuts (Calder, 2013). These fatty acids are known for their ability to reduce inflammation, support cardiovascular health, and enhance brain function, making them critical for healthy aging (Swanson et al., 2012; Calder, 2017). Their benefits arise mainly from their active forms, eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA) (Das, 2006). EPA serves as a precursor for anti-inflammatory eicosanoids, which inhibit pro-inflammatory mediators such as prostaglandins and leukotrienes (Serhan and Levy, 2018). DHA, as a structural metabolite of neuronal membranes, enhances membrane fluidity and modulates synaptic signaling, contributing to cognitive health and neuroprotection (Bazan, 2006). In the cardiovascular system, omega-3 fatty acids improve endothelial function by enhancing eNOS activity, which increases nitric oxide production and promotes vasodilation (Zgheel et al., 2014). They lower blood triglycerides, reduce low-density lipoprotein (LDL) oxidation, and suppress NF-κB signaling, contributing to cardiovascular health (Mozaffarian and Wu, 2011; Calder, 2017). In the brain, omega-3 fatty acids mitigate oxidative stress and neuroinflammation by reducing reactive oxygen species (ROS) and suppressing pro-inflammatory cytokines like IL-6 and TNF-α (Bazan, 2007). DHA activates PI3K/Akt signaling, supporting neuronal survival and reducing the risk of neurodegenerative conditions such as Alzheimer’s disease (Akbar et al., 2005; Bazan, 2006). Furthermore, omega-3 fatty acids influence brain-derived neurotrophic factor (BDNF) levels, promoting neurogenesis and cognitive (Cardoso et al., 2014; Dyall, 2015). These combined properties highlight their versatile role in promoting overall health and longevity (Simopoulos, 2008).
2.2 Vitamin A
Vitamin A (Retinoids), a fat-soluble vitamin found in foods like liver, carrots, and leafy green vegetables, plays a crucial role in vision, immune function, and cellular regulation (Blomhoff, 1994; Tanumihardjo, 2011). In vision, vitamin A is indispensable for forming rhodopsin, a light-sensitive pigment in the retina essential for low-light and color vision (Saari, 2012). It also maintains the health of the corneal epithelium and conjunctiva, reducing the risk of conditions like xerophthalmia and age-related macular degeneration (AMD) (Sommer, 2008). Retinoic acid, an active metabolite of vitamin A, regulates genes involved in photoreceptor differentiation and repair, preserving visual function with age (Saari, 2012). Vitamin A supports the immune system by strengthening epithelial barriers in the skin and mucous membranes, which act as the first line of defense against pathogens (Ross, 2012). It enhances the activity of T-cells and B-cells while promoting cytokine production essential for adaptive immunity (Stephensen, 2001; Ross, 2012). This is particularly vital in older populations, where weakened immunity heightens susceptibility to infections (Michaud et al., 2013). Additionally, vitamin A’s anti-inflammatory properties modulate NF-κB signaling, helping to reduce chronic inflammation associated with aging (Mora et al., 2008). At the cellular level, retinoic acid governs cell proliferation, differentiation, and apoptosis by activating nuclear receptors such as RARs (Retinoic Acid Receptors) and RXRs (Retinoid X Receptors) (Altucci and Gronemeyer, 2001). Retinoic acid also influences Wnt/β-catenin signaling, playing a key role in tissue regeneration and repair (Blum and Begemann, 2012).
2.3 Vitamin C
Vitamin C (Ascorbic acid), also known as ascorbic acid, is a water-soluble vitamin found in fruits and vegetables such as citrus fruits, strawberries, and bell peppers (Levine et al., 1996; Carr A. C. and Frei B., 1999). It plays a critical role in healthy aging through its antioxidant properties, immune support, and cardiovascular benefits (Carr and Maggini, 2017). As a potent antioxidant, vitamin C scavenges ROS and regenerates other antioxidants like vitamin E, preventing lipid peroxidation and DNA damage, which are key contributors to aging (Frei et al., 1989; Carr A. and Frei B., 1999). It also modulates inflammatory pathways by inhibiting NF-κB activation, reducing chronic inflammation in aging populations (Monacelli et al., 2017). In immune function, vitamin C enhances T-cell and macrophage activity, promoting pathogen clearance and adaptive immunity (Carr and Maggini, 2017). Additionally, it boosts interferon production, improving antiviral responses and reducing the severity of infections, particularly in older adults (Hemilä, 2017). Vitamin C supports cardiovascular health by activating eNOS to increase nitric oxide (NO) production, improving blood vessel dilation and reducing arterial stiffness (May and Harrison, 2013). Vitamin C prevents LDL cholesterol oxidation, reducing the risk of atherosclerosis (Frei, 1991). Additionally, clinical studies have shown its blood pressure-lowering effects, contributing to cardiovascular health (Juraschek et al., 2012).
2.4 Vitamin D
Vitamin D (Secosteroids), a fat-soluble vitamin, is synthesized in the skin through sunlight exposure and obtained from fatty fish, fortified foods, and supplements (Holick, 2007). It is crucial for bone health, immune regulation, and overall wellbeing, particularly in aging populations (Pludowski et al., 2013). In the immune system, vitamin D enhances innate immunity by stimulating antimicrobial peptides like cathelicidins to combat infections (Wang et al., 2004; Gombart et al., 2005). It modulates adaptive immunity by reducing excessive inflammation via NF-κB signaling, which is vital for preventing respiratory infections in older adults (Wang et al., 2004; Zhang et al., 2014). Vitamin D supports cardiovascular health by improving endothelial function and regulating the renin-angiotensin-aldosterone system (RAAS), lowering blood pressure and reducing arterial stiffness (Carrara et al., 2016; Al-Ishaq et al., 2021). Its anti-inflammatory effects further protect against vascular inflammation, a key factor in cardiovascular diseases (Norman and Powell, 2014; Yin and Agrawal, 2014). In the brain, vitamin D promotes neuronal survival and repair through neurotrophic factors (Khairy and Attia, 2021). Deficiency is linked to increased risks of neurodegenerative diseases and cognitive decline (Berridge, 2017; Feart et al., 2017). Supplementation may improve memory and in deficient individuals (Gowda et al., 2015; Pettersen, 2017).
2.5 Vitamin E
Vitamin E (Tocopherols) is a fat-soluble antioxidant primarily found in nuts, seeds, and vegetable oils (Bramley et al., 2000). It plays a crucial role in protecting cellular metabolites from oxidative damage and supports immune function and cardiovascular health, particularly during aging (Gombart et al., 2005; Traber and Stevens, 2011). Its primary function is to neutralize ROS and prevent lipid peroxidation, thereby safeguarding cell membranes from oxidative damage—a key contributor to aging and chronic diseases (Traber and Atkinson, 2007). Vitamin E also regenerates other antioxidants, such as vitamin C, to their active forms (Niki, 2014). In the immune system, Vitamin E enhances T-cell and natural killer (NK) cell function by stabilizing membranes and improving pathogen responsiveness. It reduces pro-inflammatory cytokines by inhibiting NF-κB signaling, mitigating chronic inflammation commonly seen in aging (Wu and Meydani, 2008). Vitamin E supports cardiovascular health by preventing LDL cholesterol oxidation, improving endothelial function, and reducing platelet aggregation (Freedman and Keaney, 2001; Meydani, 2001). These actions lower the risk of atherosclerosis, heart attacks, and strokes, particularly in individuals with low baseline levels of Vitamin E (Rimm et al., 1993). It also interacts with membrane lipids to optimize fluidity, promoting effective signal transduction for cellular communication (Traber and Packer, 1995; Zingg, 2019).
2.6 Vitamin K2
Vitamin K2 (Menaquinones) is a fat-soluble vitamin found in fermented foods, egg yolks, and dairy products (Khalil et al., 2021). It regulates calcium distribution to support bone and cardiovascular health, especially in aging populations (Hariri et al., 2021; Khalil et al., 2021). It activates osteocalcin, a protein responsible for binding calcium to the bone matrix, thereby improving bone mineral density and reducing the risk of fractures and osteoporosis (Na et al., 2022; Aaseth et al., 2024). Vitamin K2 also synergizes with vitamin D to support bone integrity and mitigate bone loss associated with aging (Kidd, 2010; Capozzi et al., 2020; Singh et al., 2022). In the cardiovascular system, vitamin K2 activates Matrix Gla Protein (MGP), which inhibits arterial calcium deposition (Shioi et al., 2020; Hariri et al., 2021). Vitamin K2 reduces vascular calcification and decreases the risk of atherosclerosis and cardiovascular diseases (Kurnatowska et al., 2015; Shioi et al., 2020; Hariri et al., 2021).
2.7 Coenzyme Q10 (CoQ10)
Coenzyme Q10 (CoQ10, Ubiquinone-10) is a lipid-soluble metabolite found in mitochondria, essential for cellular energy production and antioxidant defense (Turunen et al., 2004; Hidalgo-Gutiérrez et al., 2021). It is abundant in energy-demanding tissues like the heart, brain, and muscles but decreases with age, making it significant in aging research (López-Lluch, 2019; Gutierrez-Mariscal et al., 2021). CoQ10 supports ATP production by facilitating electron transfer in the mitochondrial electron transport chain (ETC) (Mantle and Dybring, 2020; Elgar, 2021). This ensures sufficient energy supply for tissues vulnerable to mitochondrial dysfunction during aging (Barcelos and Haas, 2019; Fišar and Hroudová, 2024). As a potent antioxidant, CoQ10 neutralizes ROS and protects mitochondrial membranes, proteins, and DNA from oxidative damage (Littarru and Tiano, 2007; Akbari et al., 2020). It also regenerates other antioxidants like vitamin E, enhancing overall antioxidant defense (Kagan et al., 2000). In the cardiovascular system, CoQ10 improves endothelial function by boosting nitric oxide (NO) production, promoting vasodilation, and reducing arterial stiffness (Rabanal-Ruiz et al., 2021). It inhibits lipid peroxidation, lowering the risk of atherosclerosis, and has been shown to improve heart function in heart failure patients (Rabanal-Ruiz et al., 2021). CoQ10 contributes to neuroprotection by reducing oxidative damage and improving mitochondrial efficiency in neurons (Young et al., 2007; Pradhan et al., 2021). These effects help maintain synaptic integrity and reduce the risk of neurodegenerative diseases such as Alzheimer’s and Parkinson’s (Kadian et al., 2022; Bagheri et al., 2023).
2.8 Selenium
Selenium is an essential trace element found in foods like Brazil nuts, seafood, and whole grains (Rayman, 2008). It acts as a cofactor for antioxidant enzymes, including glutathione peroxidase (GPx), and plays a crucial role in protecting cells from oxidative stress while supporting immune function (Beck et al., 2003; Santi et al., 2013). In the antioxidant system, selenium-dependent enzymes such as GPx reduce hydrogen peroxide and lipid hydroperoxides, preventing oxidative damage to lipids, proteins, and DNA (Ingold and Conrad, 2018; Weaver and Skouta, 2022). This action helps maintain cellular integrity and lowers the risk of age-related diseases, including cardiovascular and neurodegenerative disorders (Cai et al., 2019; Bjørklund et al., 2022a). Selenium also enhances the proliferation and function of T-cells and natural killer (NK) cells, modulating cytokine production to balance pro-inflammatory and anti-inflammatory responses (Hoffmann and Berry, 2008; Avery and Hoffmann, 2018). Emerging research suggests that selenium contributes to cognitive health by protecting neurons from oxidative damage and reducing neuroinflammation (Bai et al., 2024). Selenium-dependent enzymes like GPx and thioredoxin reductase help mitigate inflammation linked to neurodegenerative diseases such as Alzheimer’s and Parkinson’s (Bjørklund et al., 2022c; Oliveira et al., 2023). Beyond its antioxidant role, selenium influences mitochondrial dynamics by promoting mitochondrial fusion and preventing excessive fission, maintaining cellular energy balance (Kumari et al., 2012). Additionally, selenium-dependent enzymes help prevent protein misfolding, a key factor in neurodegenerative diseases (Cardoso et al., 2015; Zhang and Song, 2021).
2.9 Zinc
Zinc is an essential trace mineral found in foods such as meat, fish, and legumes (Fairweather-Tait, 1988). It is involved in various cellular processes, including enzymatic activity, DNA repair, and immune regulation, making it vital for healthy aging (Haase and Rink, 2009; Costa et al., 2023). In the immune system, zinc enhances the function of T-cells and natural killer (NK) cells, which are essential for adaptive and innate immunity (Shankar and Prasad, 1998). It also modulates cytokine production and prevents excessive inflammatory responses by inhibiting NF-κB signaling (Jarosz et al., 2017). This helps maintain immune balance and reduces chronic inflammation, a key contributor to aging-related diseases (Vasto et al., 2006; Wong et al., 2021). Zinc plays a crucial role in DNA repair and genomic stability by serving as a cofactor for enzymes such as DNA polymerase (Wu and Wu, 2023). By supporting DNA synthesis and repair, zinc helps prevent genomic instability, a key factor in cellular aging and age-related diseases (Dreosti, 2001; Costa et al., 2023). Zinc-finger transcription factors contribute to chromatin remodeling and gene regulation, further supporting cellular health during aging (Powell et al., 2019; Kamaliyan and Clarke, 2024). As an antioxidant, zinc stabilizes cell membranes and neutralizes ROS, preventing oxidative damage (Marreiro et al., 2017; Lee, 2018). It also supports the function of antioxidant enzymes like superoxide dismutase (SOD), which protects cells from oxidative stress (Mondola et al., 2016). This antioxidant role helps delay aging and lowers the risk of neurodegenerative and cardiovascular diseases (Prasad, 2014). Zinc regulates cellular senescence by modulating matrix metalloproteinases (MMPs), which remodel extracellular matrix metabolites and prevent tissue stiffness and fibrosis, common in aging tissues (Cabral-Pacheco et al., 2020).
3 Natural products as innovators in aging therapeutics
Natural products, including botanical drugs and phytochemicals, have been used for centuries in traditional medicine, but now gaining prominence in modern pharmacology for their anti-aging properties (Bjørklund et al., 2022b). This paragraph examines key natural products such as resveratrol, curcumin, and ginkgo biloba, focusing on their roles in modulating age-related pathways (Pyo et al., 2020; Assi et al., 2023). Research findings are discussed that demonstrate how these products not only mitigate oxidative stress and inflammation but also activate longevity genes such as sirtuins and AMP-activated protein kinase (AMPK) (Shehzad et al., 2011; Chung et al., 2012; Li X. et al., 2020). This review also explores the potential of these natural products to reverse or delay aging symptoms. These natural products and their effects and mechanisms are summarized in Table 2.
3.1 Resveratrol
Resveratrol (3,5′,4′-trihydroxystilbene, [Polygonaceae]) is a polyphenolic metabolite found in red wine, grapes, and berries (Bonnefont-Rousselot, 2016). It is widely recognized for its antioxidant and anti-inflammatory properties and has been extensively studied for its role in promoting longevity and preventing age-related diseases (Bonnefont-Rousselot, 2016; Pyo et al., 2020). Resveratrol exerts its effects by scavenging ROS, thereby reducing oxidative stress and protecting lipids, proteins, and DNA from oxidative damage (Truong et al., 2018; Koushki et al., 2020). Since oxidative stress is a key contributor to aging and chronic diseases such as cardiovascular and neurodegenerative disorders, resveratrol’s antioxidant function plays a critical role in maintaining cellular health (Meng et al., 2020; Leyane et al., 2022). At the molecular level, resveratrol activates SIRT1 (Sirtuin 1), a key regulator of cellular metabolism, DNA repair, and cell survival (Borra et al., 2005; Jeong et al., 2007). By stimulating SIRT1, resveratrol mimics the effects of calorie restriction, enhancing mitochondrial function and promoting longevity (Rogina and Tissenbaum, 2024). Additionally, resveratrol increases mitochondrial biogenesis through the PGC-1α pathway, improving energy efficiency and reducing oxidative damage in aging cells (Higashida et al., 2013; Zhou et al., 2021). It also modulates the NAD+/NADH ratio, an essential factor in cellular metabolism, promoting adaptive stress responses and delaying cellular senescence (Jang et al., 2012; Desquiret-Dumas et al., 2013). Beyond its role in longevity, resveratrol has significant cardiovascular benefits. It improves endothelial function, enhances nitric oxide (NO) production, and increases blood flow, reducing the risk of heart disease (Xia et al., 2014). Additionally, it lowers LDL cholesterol oxidation, a critical factor in atherosclerosis, and improves blood vessel elasticity (Frémont et al., 1999; Cherniack and Troen, 2013; Breuss et al., 2019). Moreover, resveratrol exhibits anti-inflammatory properties by inhibiting NF-κB signaling, a central mediator of chronic inflammation (Ma et al., 2015; Meng et al., 2021).
3.2 Curcumin
Curcumin (Curcuma longa L., [Zingiberaceae]), the active metabolite in turmeric (Curcuma longa), is well known for its anti-inflammatory, antioxidant, and neuroprotective properties (Silvestro et al., 2021; Genchi et al., 2024). It has been extensively studied for its potential therapeutic effects in aging and age-related diseases (Sikora et al., 2010; Nunes et al., 2024). Curcumin exerts its anti-inflammatory effects primarily by inhibiting key inflammatory pathways such as NF-κB and COX-2, which regulate the production of pro-inflammatory cytokines and mediators involved in chronic inflammation (Yuan et al., 2018; Fu et al., 2022). By reducing inflammation, curcumin helps lower the risk of age-related conditions, including arthritis and cardiovascular diseases (He et al., 2015). As an antioxidant, curcumin neutralizes ROS and enhances cellular defense mechanisms (Sathyabhama et al., 2022). It stimulates antioxidant enzymes such as superoxide dismutase (SOD) and glutathione peroxidase, which protect cells from oxidative stress, a key driver of aging and neurodegenerative diseases (Khayatan et al., 2024). In the brain, curcumin’s antioxidant and anti-inflammatory properties contribute to its neuroprotective effects (Genchi et al., 2024). It mitigates oxidative damage and neuroinflammation, both implicated in Alzheimer’s disease, and promotes the clearance of amyloid plaques by activating autophagy and modulating microglial activity (Azzini et al., 2024). Curcumin also plays a crucial role in mitochondrial quality control by inducing mitophagy through the PINK1/Parkin pathway, thereby preventing age-related cellular decline (Cao et al., 2020; Jin et al., 2022). Additionally, it activates the Nrf2-Keap1 pathway, enhancing the production of endogenous antioxidant enzymes to counteract oxidative stress (Soetikno et al., 2013; Lin et al., 2019).
3.3 Querectin
Quercetin (3,3′,4′,5,7-Pentahydroxyflavone, [Fagaceae/Rutaceae]) is a flavonoid found in apples, onions, and green tea. It has strong antioxidant and anti-inflammatory properties, making it useful for aging and age-related diseases (Li et al., 2016b; Deepika and Maurya, 2022). Quercetin neutralizes ROS and reduces oxidative stress in cells, protecting them from damage (Costa et al., 2016). In the immune system, quercetin reduces inflammation by inhibiting the NF-κB pathway, which regulates pro-inflammatory cytokine production (Comalada et al., 2005). This makes quercetin useful in controlling chronic inflammation, especially in aging populations (David et al., 2016). Quercetin improves vascular health by enhancing blood vessel function and reducing LDL cholesterol oxidation, which helps prevent atherosclerosis (Jiang et al., 2020). It also improves circulation and lowers blood pressure (Serban et al., 2016). Studies show that quercetin may protect the brain from oxidative damage and neuroinflammation, both of which contribute to neurodegenerative diseases like Alzheimer’s (Khan et al., 2019). By crossing the blood-brain barrier, it helps sustain brain health throughout aging (Grewal et al., 2021). Quercetin also exerts its anti-aging effects by inhibiting the PI3K/AKT/mTOR pathway, which regulates cellular growth and aging (Granato et al., 2017; Li et al., 2018). By modulating this pathway, quercetin promotes autophagy, prevents cellular senescence, and strengthens intercellular junctions to enhance tissue integrity while reducing inflammation-induced damage (Rather and Bhagat, 2020).
3.4 Ginseng
Ginseng (panax ginseng, C.A.Mey., [Araliaceae]) is a well-known botanical drug used in traditional medicine for its adaptogenic properties, modulating immune functions and enhancing resilience to various stressors (Ratan et al., 2021). The active metabolites in ginseng, particularly ginsenosides, are responsible for its medicinal effects (Leung and Wong, 2010). Ginseng modulates the hypothalamic-pituitary-adrenal (HPA) axis, which regulates the body’s stress response (Lee and Rhee, 2017). By reducing the secretion of cortisol, a stress hormone, ginseng helps lower chronic inflammation and balance hormonal levels (Liao et al., 2018). In the immune system, ginseng enhances the activity of macrophages, T-cells, and natural killer (NK) cells, all of which are crucial for fighting infections and maintaining immune homeostasis (He et al., 2017). Ginsenosides activate pathways such as NF-κB and MAPK, which are involved in immune response and inflammation regulation, helping to strengthen the body’s defense mechanisms (Kim et al., 2017). Ginseng also supports cognitive function by protecting neurons from oxidative stress and improving synaptic plasticity (Shin et al., 2024). Ginseng’s active metabolites, ginsenosides, modulate AMPK signaling to enhance cellular energy sensing and lipid metabolism (Wang et al., 2022). This mechanism not only combats age-related metabolic disorders but also reduces chronic low-grade inflammation (Fan et al., 2020). Ginseng has also been shown to promote angiogenesis through VEGF signaling, aiding tissue repair and regeneration (Song et al., 2023).
3.5 Kaempferol
Kaempferol (3,4′,5,7-Tetrahydroxyflavone [Fabaceae/Brassicaceae]) is a flavonoid found in a variety of fruits, vegetables, and beverages, including broccoli, kale, beans, and tomato (Calderon-Montano et al., 2011). It has been widely studied for its antioxidant, anti-inflammatory, and anti-cancer properties, making it a promising metabolite for promoting health, particularly in aging (Gutiérrez-del-Río et al., 2016; Shahbaz et al., 2023). Kaempferol acts as a potent antioxidant by neutralizing ROS and preventing oxidative damage to cells (Wu et al., 2018). By scavenging free radicals, kaempferol reduces oxidative stress, which is a major contributor to aging and the development of chronic diseases, such as cardiovascular disease, cancer, and neurodegenerative disorders (Liao et al., 2016; Rahul and Siddique, 2021; Kamisah et al., 2023). In terms of inflammation, kaempferol modulates inflammatory pathways, particularly by inhibiting NF-κB and MAPK signaling, which are involved in the production of pro-inflammatory cytokines (Wang et al., 2020). In neuroprotection, kaempferol has been shown to protect neurons from oxidative stress and reduce neuroinflammation (Nezhad Salari et al., 2024). It may help in maintaining cognitive function by promoting the production of BDNF, which is involved in neuronal survival and synaptic plasticity (Silva dos Santos et al., 2021). Kaempferol inhibits the senescence-associated secretory phenotype (SASP) by reducing the production of inflammatory cytokines such as IL-6 and TNF-α (Lim et al., 2015; Hussain et al., 2024).
3.6 Apigenin
Apigenin (4′,5,7-Trihydroxyflavone) [Asteraceae] is a flavonoid found in a variety of plants, including parsley, celery, chamomile, and basil (Salehi et al., 2019b). Known for its antioxidant, anti-inflammatory, and anti-cancer properties, apigenin has been studied for its potential health benefits, particularly in aging (Madunić et al., 2018). Apigenin acts as a potent antioxidant by scavenging ROS and reducing oxidative stress in cells (Jung, 2014). In terms of inflammation, apigenin inhibits the NF-κB pathway, which is involved in the production of pro-inflammatory cytokines such as IL-1β, IL-6, and TNF-α (Patil et al., 2016). By modulating this pathway, apigenin helps reduce chronic inflammation, a common contributor to diseases like arthritis, heart disease, and neurodegenerative disorders in aging populations (Ali et al., 2017). In neuroprotection, apigenin has been shown to reduce oxidative stress and inflammation in the brain (Li R. et al., 2016). It increases the expression of BDNF, which is important for neuronal survival and synaptic plasticity (Sharma et al., 2020; Gao et al., 2023). These effects help maintain cognitive function and may reduce the risk of neurodegenerative diseases like Alzheimer’s (Zhao et al., 2013). Apigenin enhances mitochondrial function through increased expression of SOD2, reducing mitochondrial-derived oxidative stress (Jung, 2014; Pal et al., 2017). Apigenin’s ability to stabilize transcription factors like FOXO3 further contributes to its longevity-enhancing effects (Kawasaki et al., 2010; Lin et al., 2015).
3.7 Green tea extract
Green Tea Extract (Camellia sinensis (L.) Kuntze) [Theaceae] is derived from the leaves of Camellia sinensis and is rich in polyphenols, particularly epigallocatechin gallate (EGCG), which is the main natural products (Komes et al., 2010; Du et al., 2012). It is well-known for its antioxidant, anti-inflammatory, and metabolism-enhancing properties, making it a popular natural product for promoting healthy aging (Chen et al., 2009; Senanayake, 2013; Ohishi et al., 2016). Green tea extract exhibits strong antioxidant activity by neutralizing ROS and enhancing the activity of endogenous antioxidant enzymes (Elbling et al., 2005). The extract also exerts anti-inflammatory effects by inhibiting pathways like NF-κB and reducing levels of pro-inflammatory cytokines such as IL-6 and TNF-α (Ohishi et al., 2016). In addition, EGCG from green tea extract improves mitochondrial function and enhances energy metabolism by activating pathways like AMPK, which promotes cellular energy balance (Pournourmohammadi et al., 2017; Ha et al., 2018). Green tea extract also supports cognitive health by protecting neurons from oxidative stress and reducing neuroinflammation (Prasanth et al., 2019; Valverde-Salazar et al., 2023). Studies have shown its ability to enhance memory and learning in aging populations (Mancini et al., 2017). Green tea extract, rich in EGCG, enhances the DNA damage response (DDR) by upregulating ATM and ATR kinases, which repair double-strand breaks (Kuo et al., 2016; Majidinia et al., 2019). Additionally, EGCG modulates circadian clock genes, improving metabolic and cognitive functions in aging individuals (Liu et al., 2023).
3.8 Alpha-Lipoic Acid (ALA)
Alpha-Lipoic Acid (1,2-Dithiolane-3-pentanoic acid) (ALA) is a natural metabolite with strong antioxidant properties, crucial for energy metabolism and cellular protection (Tibullo et al., 2017; Superti and Russo, 2024). It is found in small amounts in foods like vegetables, futis and red meat (Salehi et al., 2019a). ALA’s water- and fat-solubility allows it to act in various cellular compartments, where it scavenges free radicals and regenerates other antioxidants, contributing to healthy aging (Shay et al., 2009; Nobakht-Haghighi et al., 2018). Additionally, ALA modulates NF-κB signaling, reducing inflammation and protecting tissues from chronic damage (Chang et al., 2017; Ishii et al., 2017). In metabolic health, ALA enhances insulin sensitivity by activating AMPK (AMP-activated protein kinase), which improves glucose uptake in muscle cells and lowers blood sugar levels (Lee et al., 2005; Targonsky et al., 2006; Shen et al., 2007).
3.9 Astaxanthin
Astaxanthin (3,3′-Dihydroxy-β,β-carotene-4,4′-dione) [Haematococcaceae] is a carotenoid pigment primarily found in microalgae, salmon, and krill (Johnson and An, 1991). It is known for its powerful antioxidant and anti-inflammatory properties, which contribute to its benefits for aging, skin health, and overall wellbeing (Pereira et al., 2021; Chae et al., 2022). As a powerful antioxidant, astaxanthin is significantly stronger than other carotenoids such as β-carotene and lutein (Naguib, 2000; Yuan et al., 2011; Ambati et al., 2014). It neutralizes ROS and protects cells from oxidative damage by scavenging free radicals (Mohammadi et al., 2021). Its unique structure enables it to safeguard both the lipid and aqueous phases of cells, effectively protecting various cellular metabolites, including cell membranes, proteins, and DNA (Ambati et al., 2014; Brotosudarmo et al., 2020). In addition to its antioxidant effects, astaxanthin exhibits anti-inflammatory properties by inhibiting key signaling pathways, including NF-κB and COX-2 (Choi et al., 2008; Farruggia et al., 2018). These pathways are involved in the production of pro-inflammatory cytokines and mediators, which contribute to chronic inflammation (Lee et al., 2003). By modulating these pathways, astaxanthin helps prevent the progression of inflammation-related diseases such as arthritis, cardiovascular disease, and neurodegeneration (Chang and Xiong, 2020). Astaxanthin also supports eye health by reducing oxidative stress and inflammation in the retina (Yeh et al., 2016; Giannaccare et al., 2020). It protects retinal cells from damage caused by light exposure and helps prevent AMD, a leading cause of vision loss in older adults (Otsuka et al., 2013; Giannaccare et al., 2020; Alugoju et al., 2023). Additionally, astaxanthin improves blood flow to the eyes, further enhancing visual function (Giannaccare et al., 2020).
4 Upcoming trends and challenges
As research continues to uncover the vast potential of nutrients and natural products in promoting health and longevity, several emerging trends and challenges are shaping the future of anti-aging therapies (Ros and Carrascosa, 2020; Bjørklund et al., 2022b). These developments highlight the importance of a holistic and evidence-based approach to leveraging natural metabolites for sustainable health benefits.
4.1 Personalized nutrition and precision medicine
One of the most promising trends is the move towards personalized nutrition and precision medicine. Advances in genomics and metabolomics are enabling a deeper understanding of individual differences in nutrient metabolism and response (Kaput and Rodriguez, 2006; Hadi, 2023). This personalized approach allows for tailored dietary and supplementation strategies that align with a person’s genetic profile, lifestyle, and health status, optimizing the efficacy of natural anti-aging interventions (Biesalski et al., 2009; Afshin et al., 2019). The integration of artificial intelligence and big data analytics further enhances the ability to develop customized health plans that promote longevity and mitigate age-related conditions (Topol, 2019).
Several clinical applications of precision medicine in nutrition are already in use (Wishart, 2016). Nutrigenomics-based dietary interventions enable personalized dietary recommendations based on genetic variants affecting nutrient metabolism, such as MTHFR polymorphisms influencing folate metabolism (Kiani et al., 2022; Andrade et al., 2025). Metabolomics-guided supplementation tailors nutrient intake by analyzing an individual’s metabolic profile to identify micronutrient deficiencies and optimize supplementation strategies (Rigamonti et al., 2023). Additionally, biomarker-driven antioxidant therapy assesses oxidative stress markers, such as the glutathione (GSH/GSSG) ratio, to guide personalized antioxidant interventions aimed at reducing cellular damage and improving metabolic health (Yagishita et al., 2020; Pan et al., 2024). These advancements illustrate how precision medicine strategies are being integrated into clinical practice to enhance the effectiveness of nutrient-based interventions (Bailey and Stover, 2023; Keijer et al., 2024).
4.2 Synergistic formulations
Another trend is the development of synergistic formulations that combine multiple nutrients and natural products to enhance their collective benefits (Ebrahimi et al., 2023). Research is increasingly focusing on the synergistic effects of metabolites such as vitamins, polyphenols, and omega-3 fatty acids (Liu, 2003; Li et al., 2016c; Calder, 2017; Ebrahimi et al., 2023). These combinations can potentially offer more significant health benefits than individual metabolites by targeting multiple pathways involved in aging (Liu, 2003). Formulating these combinations into easily accessible supplements and functional foods can provide practical solutions for aging populations seeking to maintain their health (Cencic and Chingwaru, 2010).
4.3 Advances in delivery systems
Many natural metabolites suffer from poor bioavailability due to low solubility, rapid metabolism, and degradation, limiting their therapeutic efficacy. Addressing these challenges, improving the bioavailability and efficacy of natural metabolites through advanced delivery systems is a crucial area of innovation (Obeid et al., 2017).
In vivo bioavailability is often restricted by multiple factors, including poor water solubility, instability in the gastrointestinal (GI) tract, extensive first-pass metabolism, and rapid systemic clearance. For instance, curcumin, a well-known polyphenol, has poor absorption due to its hydrophobic nature and rapid hepatic metabolism (Nelson et al., 2017). Coenzyme Q10 (CoQ10) also exhibits poor bioavailability due to its high molecular weight and low water solubility, which limit its intestinal absorption and systemic circulation (Zhang et al., 2022). Polyphenols such as quercetin and resveratrol also face challenges related to instability and rapid conjugation, leading to reduced systemic bioavailability (El Monfalouti and Kartah, 2024).
To overcome these limitations, several advanced drug delivery technologies are being explored. Nanoencapsulation improves the solubility and stability of hydrophobic metabolites, facilitating better absorption (Livney, 2016). Liposomal delivery systems, composed of phospholipid bilayers, protect active metabolites from enzymatic degradation and enhance their permeability across biological membranes (Singh et al., 2019). Solid lipid nanoparticles and nanostructured lipid carriers further enhance drug stability and prolong systemic circulation, improving therapeutic efficacy (Safta et al., 2024).
Another promising approach is the use of cyclodextrin complexes, which form inclusion complexes with poorly soluble metabolites, increasing their water solubility and enhancing bioavailability (Loftsson and Brewster, 2012; Kurkov and Loftsson, 2013). Prodrug strategies, which involve modifying the chemical structure of a metabolite to improve its absorption and metabolic stability, are also gaining attention in the field of natural product-based therapeutics (Rautio et al., 2008).
These technologies can overcome the limitations of traditional supplementation, ensuring that active metabolites reach their target tissues at therapeutic levels. Further research is needed to refine these delivery strategies and optimize their application in clinical settings to maximize the therapeutic potential of natural metabolites.
4.4 Regulatory and quality assurance challenges
Despite the potential benefits, the widespread adoption of nutrient and natural product-based therapies faces significant regulatory and quality assurance challenges (Yadav et al., 2024). Ensuring the safety, efficacy, and consistency of these products requires stringent regulatory oversight and robust quality control measures (Bailey et al., 2013). The variability in the composition of natural products, influenced by factors such as sourcing, processing, and storage, necessitates standardized practices to guarantee their therapeutic potential (Kunle et al., 2012).
In addition, the interaction between natural metabolites and conventional medications remains a critical concern. Some nutrients and phytochemicals can influence drug metabolism by modulating cytochrome P450 enzymes, leading to altered drug efficacy or toxicity (Holst and Williamson, 2008; Johnson, 2008). For instance, St. John’s Wort induces CYP3A4, reducing the plasma concentration of certain drugs, whereas grapefruit juice inhibits the same enzyme, leading to potential drug accumulation and adverse effects (Dürr et al., 2000; Dahan and Altman, 2004; Rahimi and Abdollahi, 2012). Moreover, excessive intake of fat-soluble vitamins, iron, or polyphenols may lead to toxicity or interfere with drug absorption (Martin and Appel, 2009; Awuchi et al., 2020). Addressing these risks requires comprehensive safety assessments and clearer guidelines on the co-administration of natural products with pharmaceuticals (Colalto, 2010).
Furthermore, the lack of standardized labeling and dosage recommendations for natural supplements increases the risk of misuse (Avigan et al., 2016; Bailey, 2020). Unlike pharmaceuticals, where dosing is strictly regulated, natural metabolites often exhibit dose-dependent effects, making it challenging to establish universally safe and effective intake levels (Hasler, 2002; Wen et al., 2021). Certain natural products may also be contraindicated for individuals with specific health conditions. For example, high doses of omega-3 fatty acids may increase bleeding risk in individuals taking anticoagulants (Kapoor et al., 2021). Regulatory agencies must work towards harmonized guidelines that define safe consumption limits and potential contraindications, ensuring that natural product-based therapies are both effective and safe for consumers (Bast et al., 2002).
4.5 Ethical and accessibility considerations
As the popularity of anti-aging therapies grows, ethical and accessibility considerations must be addressed (Trothen, 2024). Ensuring that advancements in personalized nutrition and natural therapies are accessible to diverse populations, including underserved communities, is vital for promoting health equity (Petersen and Kwan, 2011). Additionally, ethical considerations regarding the use of genetic information and personalized health data must be carefully managed to protect individual privacy and autonomy (Vayena et al., 2018).
Furthermore, economic and environmental challenges must be considered when evaluating the scalability of natural metabolite-based interventions. Certain high-value natural metabolites, such as fish-derived omega-3 fatty acids and rare plant extracts, pose challenges in terms of cost, availability, and sustainability (Venegas-Calerón et al., 2010). The extraction and production of these metabolites often require significant resources, leading to high consumer prices and potential supply chain limitations (Palma et al., 2016). Additionally, overharvesting of rare botanical sources and intensive aquaculture practices for omega-3 production can contribute to ecological imbalances and biodiversity loss (Falkenberg et al., 2020; Zhang et al., 2024). To mitigate these challenges, research is increasingly focusing on alternative sources, such as microalgae-derived omega-3 and biotechnologically synthesized plant metabolites, which offer more sustainable and scalable solutions (Liu et al., 2022; Hassan Zadeh, 2024). Addressing these economic and environmental barriers is crucial for ensuring that natural therapies remain viable and accessible to a broader population.
4.6 Clinical trials investigating nutrients and natural products for age-related diseases
Several clinical trials have investigated the efficacy of nutrients and natural products in treating age-related diseases, including ocular, neurological, and muscular disorders (Dai et al., 2010; Suzuki et al., 2013; Dysken et al., 2014; Giannaccare et al., 2019; Voulgaropoulou et al., 2019). These studies provide critical insights into the therapeutic potential and limitations of natural metabolites in clinical settings. Table 3 summarizes key trials assessing the impact of these products on human health. While many studies demonstrate promising effects of natural products in in vitro models, these findings require further validation through well-designed in vivo and clinical studies to establish translational relevance (Denayer et al., 2014; Sorkin et al., 2020). While some studies demonstrate promising benefits, such as improved cognitive function and reduced oxidative stress, further large-scale, long-term trials are needed to confirm their effectiveness and establish optimal dosing strategies.
4.7 Essential amino acids in aging
Beyond vitamins, minerals, and natural products, essential amino acids also contribute to aging-related processes, particularly in muscle maintenance, metabolic regulation, and immune function (Roth, 2007). Branched-chain amino acids (BCAAs) such as leucine, isoleucine, and valine have been shown to support protein synthesis and mitochondrial function, while other amino acids like arginine and methionine play roles in oxidative stress modulation and immune response (Nie et al., 2018; Dai et al., 2020). While this review primarily focuses on non-protein nutrients, future research should explore the potential synergistic effects of essential amino acids with vitamins and natural metabolites in aging interventions.
4.8 Study limitations and research challenges
While numerous studies support the beneficial effects of nutrients and natural products in aging-related processes, many face limitations such as small sample sizes, lack of long-term follow-up, and variability in study designs, including differences in formulations, treatment duration, and administration routes (Ziegler et al., 2009; Puri et al., 2022; Li and Wang, 2024; Wimalawansa, 2025). Additionally, heterogeneity in study populations, inconsistent methodologies, and potential biases in self-reported dietary intake challenge the generalizability of findings (Behmer et al., 2002; Carocho and Ferreira, 2013; Mirmiran et al., 2021). Some clinical trials report positive outcomes, but confounding factors such as lifestyle, genetic variability, and concurrent interventions may influence results (Davey Smith and Ebrahim, 2003; Welch et al., 2011). Furthermore, preclinical studies, including In silico and In vitro models, provide valuable mechanistic insights but have inherent limitations in predicting clinical efficacy (Wu et al., 2020; Vashishat et al., 2024). Computational models rely on algorithm-based predictions that may not fully capture the complex biochemical interactions occurring in human physiology (Southern et al., 2008; Lee and Hu, 2019). Likewise, In vitro studies often utilize simplified cellular systems that lack systemic metabolic interactions, immune responses, and tissue-specific effects, making it difficult to extrapolate findings to whole-body physiology (Cook et al., 2012; McGonigle and Ruggeri, 2014).
A major controversy in the field is the lack of consensus on the effective doses of natural metabolites, which complicates their clinical translation (Singh et al., 2015). Unlike pharmaceuticals with well-defined dosing parameters, natural products often exhibit dose-dependent effects influenced by formulation, bioavailability, and individual metabolic differences (Boullata, 2005). For instance, curcumin requires high doses to achieve therapeutic effects due to its poor bioavailability, whereas lower doses may be sufficient for other polyphenols (Mahran et al., 2017; Abd El-Hack et al., 2021). Additionally, discrepancies in study methodologies, including variability in outcome measurements and data analysis approaches, contribute to inconsistent conclusions across studies (Kuhl, 2005; Argyropoulou et al., 2013; Sorkin et al., 2016). Many preclinical studies rely on in vitro models or animal experiments that do not fully replicate human physiological conditions, limiting their direct applicability (Henderson et al., 2013). While In vitro assays can provide initial screening for bioactivity, they lack the systemic complexity required to assess pharmacokinetics, organ-specific metabolism, and potential toxicity (Lipscomb and Poet, 2008; Astashkina et al., 2012). Therefore, findings from these models should be interpreted cautiously and validated through well-designed In vivo and clinical studies (Denayer et al., 2014).
In clinical trials, differences in intervention protocols, inconsistencies in sample selection, and inadequate control groups often result in conflicting findings (Jüni et al., 2001). Moreover, a lack of standardization in study endpoints and assessment methods further complicates comparative analysis, making it difficult to derive definitive conclusions about the efficacy of natural metabolites (Ziegelmann et al., 2020).
Another critical limitation is the insufficient understanding of the pharmacokinetics and bioavailability of many natural products (Hao et al., 2014; Pathak and Raghuvanshi, 2015). Variability in formulation types (e.g., whole extracts vs isolated metabolites), administration routes, and metabolic differences among individuals can significantly alter their biological activity (Possemiers et al., 2011; Sova and Saso, 2020). Without standardized dosing guidelines, it remains challenging to translate preclinical findings into effective therapeutic applications (Ioannidis et al., 2018).
Addressing these challenges requires standardized pharmacokinetic profiling and dose-response studies to establish optimal therapeutic windows for natural metabolites (Zeng et al., 2022). Future studies should also prioritize multicenter RCTs with larger and more diverse populations to improve the robustness of findings (Li W. et al., 2020). Furthermore, long-term clinical trials are necessary to determine the sustained effects and safety profiles of these metabolites over extended periods (Reginster et al., 2001; Zhou Y. et al., 2016). Given the variability in bioavailability and metabolic responses, future research should explore personalized approaches integrating metabolomics and nutrigenomics to tailor interventions to individual needs (Vyas et al., 2018; Lagoumintzis and Patrinos, 2023). Additionally, studies should focus on the synergistic effects of multiple natural metabolites, as combination therapies may enhance efficacy through complementary mechanisms (HemaIswarya and Doble, 2006; Zhou X. et al., 2016). Advanced drug delivery systems, such as nanoformulations and liposomal encapsulation, should also be investigated to improve bioavailability and therapeutic potential (Allen and Cullis, 2013; Khan et al., 2013). Additionally, In silico/In vitro methodologies should be supplemented with well-structured translational research strategies, ensuring that mechanistic findings can be effectively validated in physiologically relevant models (Bai et al., 2018; Rowland et al., 2018). Emerging technologies such as organ-on-a-chip systems and advanced 3D cell cultures may provide more predictive data, bridging the gap between early-stage research and clinical applications (Zhang and Radisic, 2017; Cavero et al., 2019). To establish more definitive conclusions, well-designed, large-scale, long-term clinical trials are needed (Piantadosi, 2024).
4.9 As the popularity future research directions
Ongoing research is critical to fully elucidate the mechanisms underlying the anti-aging effects of nutrients and natural products (Wu et al., 2024). Large-scale, long-term clinical trials are needed to confirm their efficacy and safety (Ahmed et al., 2023). Interdisciplinary collaborations among researchers, healthcare professionals, and industry stakeholders will drive innovation and translate scientific findings into practical applications (Bhavnani et al., 2017). Exploring new natural metabolites with anti-aging potential and understanding their interactions with existing therapies will continue to expand the repertoire of effective interventions (Wan et al., 2024).
5 Conclusion
The exploration of nutrients and natural products for sustainable drug development against offers significant health benefits. Unlike conventional pharmaceuticals, these natural metabolites often have fewer side effects while maintaining both efficacy and safety. By focusing on the therapeutic potential of metabolites derived from food and natural sources, researchers are uncovering new pathways to enhance health and longevity. Nutrients and natural products have demonstrated their capacity to modulate key biological pathways related to immune function, inflammation, and neuroprotection. Recent studies have highlighted the importance of targeting aging-related processes such as mitochondrial dysfunction, genomic instability, and cellular senescence.
In conclusion, leveraging the potential of nutrients and natural products provides a sustainable, effective approach to aging-related therapies. While this review primarily focuses on vitamins, minerals, and natural products, future research should also consider the role of essential amino acids in aging-related interventions. These metabolites address aging symptoms and target its underlying causes, offering the potential to significantly improve healthspan and quality of life. Anti-aging strategies could be transformed through continued exploration and advancements in delivery technologies and personalized medicine, benefiting individuals and healthcare systems worldwide.
Author contributions
FD: Conceptualization, Writing – original draft. YY: Data curation, Writing – original draft. YZ: Data curation, Writing – original draft. SW: Data curation, Writing – original draft. JH: Writing – original draft, Writing – review and editing. ZL: Conceptualization, Investigation, Writing – original draft. H-BJ: Investigation, Writing – original draft. DR: Writing – review and editing. WP: Funding acquisition, Writing – review and editing. K-TH: Funding acquisition, Writing – review and editing. LG: Conceptualization, Supervision, Writing – review and editing.
Funding
The author(s) declare that financial support was received for the research and/or publication of this article. This study was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF), funded by National Research Foundation of Korea (NRF) grants funded by the Korean government (MIST) (grant nos. 2022R1A2C2005130 and RS-2023-00237776).
Conflict of interest
The authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.
Generative AI statement
The author(s) declare that no Generative AI was used in the creation of this manuscript.
Publisher’s note
All claims expressed in this article are solely those of the authors and do not necessarily represent those of their affiliated organizations, or those of the publisher, the editors and the reviewers. Any product that may be evaluated in this article, or claim that may be made by its manufacturer, is not guaranteed or endorsed by the publisher.
References
Aaseth, J. O., Finnes, T. E., Askim, M., and Alexander, J. (2024). The importance of vitamin K and the combination of vitamins K and D for calcium metabolism and bone health: a review. Nutrients 16, 2420.
Abd El-Hack, M. E., El-Saadony, M. T., Swelum, A. A., Arif, M., Abo Ghanima, M. M., Shukry, M., et al. (2021). Curcumin, the active substance of turmeric: its effects on health and ways to improve its bioavailability. J. Sci. Food Agric. 101, 5747–5762. doi:10.1002/jsfa.11372
Afshin, A., Sur, P. J., Fay, K. A., Cornaby, L., Ferrara, G., Salama, J. S., et al. (2019). Health effects of dietary risks in 195 countries, 1990–2017: a systematic analysis for the Global Burden of Disease Study 2017. Lancet 393, 1958–1972. doi:10.1016/S0140-6736(19)30041-8
Ahmed, S. N., Ahmad, M., Zafar, M., Yaseen, G., Iqbal, N., Rashid, N., et al. (2023). “Herbal drugs: safety, cost-effectiveness, regulation, current trends, and future directions,” in Bioprospecting of tropical medicinal plants (Springer), 1479–1493.
Akbar, M., Calderon, F., Wen, Z., and Kim, H.-Y. (2005). Docosahexaenoic acid: a positive modulator of Akt signaling in neuronal survival. Proc. Natl. Acad. Sci. 102, 10858–10863. doi:10.1073/pnas.0502903102
Akbari, A., Mobini, G. R., Agah, S., Morvaridzadeh, M., Omidi, A., Potter, E., et al. (2020). Coenzyme Q10 supplementation and oxidative stress parameters: a systematic review and meta-analysis of clinical trials. Eur. J. Clin. Pharmacol. 76, 1483–1499. doi:10.1007/s00228-020-02919-8
Ali, F., Naz, F., Jyoti, S., and Siddique, Y. H. (2017). Health functionality of apigenin: a review. Int. J. Food Prop. 20, 1197–1238. doi:10.1080/10942912.2016.1207188
Al-Ishaq, R. K., Kubatka, P., Brozmanova, M., Gazdikova, K., Caprnda, M., and Büsselberg, D. (2021). Health implication of vitamin D on the cardiovascular and the renal system. Archives physiology Biochem. 127, 195–209. doi:10.1080/13813455.2019.1628064
Allen, T. M., and Cullis, P. R. (2013). Liposomal drug delivery systems: from concept to clinical applications. Adv. drug Deliv. Rev. 65, 36–48. doi:10.1016/j.addr.2012.09.037
Altucci, L., and Gronemeyer, H. (2001). The promise of retinoids to fight against cancer. Nat. Rev. Cancer 1, 181–193. doi:10.1038/35106036
Alugoju, P., Krishna Swamy, V., Anthikapalli, N. V. A., and Tencomnao, T. (2023). Health benefits of astaxanthin against age-related diseases of multiple organs: a comprehensive review. Crit. Rev. Food Sci. Nutr. 63, 10709–10774. doi:10.1080/10408398.2022.2084600
Ambati, R. R., Phang, S.-M., Ravi, S., and Aswathanarayana, R. G. (2014). Astaxanthin: sources, extraction, stability, biological activities and its commercial applications—a review. Mar. drugs 12, 128–152. doi:10.3390/md12010128
Ames, B. N. (2018). Prolonging healthy aging: longevity vitamins and proteins. Proc. Natl. Acad. Sci. 115, 10836–10844. doi:10.1073/pnas.1809045115
Andrade, P., Santamarina, A. B., De Freitas, J. A., Marum, A. B. R. F., and Pessoa, A. F. M. (2025). Personalized nutrition and precision medicine in perimenopausal women: a minireview of genetic polymorphisms COMT, FUT2, and MTHFR. Clinics 80, 100549. doi:10.1016/j.clinsp.2024.100549
Annweiler, C., and Beauchet, O. (2011). Vitamin D-mentia: randomized clinical trials should be the next step. Neuroepidemiology 37, 249–258. doi:10.1159/000334177
Annweiler, C., Rolland, Y., Schott, A. M., Blain, H., Vellas, B., Herrmann, F. R., et al. (2012). Higher vitamin D dietary intake is associated with lower risk of Alzheimer’s disease: a 7-year follow-up. Journals Gerontology Ser. A Biomed. Sci. Med. Sci. 67, 1205–1211. doi:10.1093/gerona/gls107
Argyropoulou, A., Aligiannis, N., Trougakos, I. P., and Skaltsounis, A.-L. (2013). Natural compounds with anti-ageing activity. Nat. Product. Rep. 30, 1412–1437. doi:10.1039/c3np70031c
Assi, A.-A., Farrag, M. M., Badary, D. M., Allam, E. A., and Nicola, M. A. (2023). Protective effects of curcumin and Ginkgo biloba extract combination on a new model of Alzheimer’s disease. Inflammopharmacology 31, 1449–1464. doi:10.1007/s10787-023-01164-6
Astashkina, A., Mann, B., and Grainger, D. W. (2012). A critical evaluation of in vitro cell culture models for high-throughput drug screening and toxicity. Pharmacol. and Ther. 134, 82–106. doi:10.1016/j.pharmthera.2012.01.001
Atanasov, A. G., Waltenberger, B., Pferschy-Wenzig, E.-M., Linder, T., Wawrosch, C., Uhrin, P., et al. (2015). Discovery and resupply of pharmacologically active plant-derived natural products: a review. Biotechnol. Adv. 33, 1582–1614. doi:10.1016/j.biotechadv.2015.08.001
Avery, J. C., and Hoffmann, P. R. (2018). Selenium, selenoproteins, and immunity. Nutrients 10, 1203. doi:10.3390/nu10091203
Avigan, M. I., Mozersky, R. P., and Seeff, L. B. (2016). Scientific and regulatory perspectives in herbal and dietary supplement associated hepatotoxicity in the United States. Int. J. Mol. Sci. 17, 331. doi:10.3390/ijms17030331
Awuchi, C. G., Igwe, V. S., and Amagwula, I. O. (2020). Nutritional diseases and nutrient toxicities: a systematic review of the diets and nutrition for prevention and treatment. Int. J. Adv. Acad. Res. 6, 1–46.
Azzini, E., Peña-Corona, S. I., Hernández-Parra, H., Chandran, D., Saleena, L. a.K., Sawikr, Y., et al. (2024). Neuroprotective and anti-inflammatory effects of curcumin in Alzheimer's disease: targeting neuroinflammation strategies. Phytotherapy Res. 38, 3169–3189. doi:10.1002/ptr.8200
Bagheri, S., Haddadi, R., Saki, S., Kourosh-Arami, M., Rashno, M., Mojaver, A., et al. (2023). Neuroprotective effects of coenzyme Q10 on neurological diseases: a review article. Front. Neurosci. 17, 1188839. doi:10.3389/fnins.2023.1188839
Bai, Q., Li, L., Liu, S., Xiao, S., and Guo, Y. (2018). Drug design progress of in silico, in vitro and in vivo researches. In-vitro In-vivo In-silico J. 1, 16–37.
Bai, Y.-Z., Zhang, Y., and Zhang, S.-Q. (2024). New horizons for the role of selenium on cognitive function: advances and challenges. Metab. Brain Dis. 39, 1255–1268. doi:10.1007/s11011-024-01375-y
Bailey, R. L. (2020). Current regulatory guidelines and resources to support research of dietary supplements in the United States. Crit. Rev. food Sci. Nutr. 60, 298–309. doi:10.1080/10408398.2018.1524364
Bailey, R. L., Gahche, J. J., Miller, P. E., Thomas, P. R., and Dwyer, J. T. (2013). Why US adults use dietary supplements. JAMA Intern. Med. 173, 355–361. doi:10.1001/jamainternmed.2013.2299
Bailey, R. L., and Stover, P. J. (2023). Precision nutrition: the hype is exceeding the science and evidentiary standards needed to inform public health recommendations for prevention of chronic disease. Annu. Rev. Nutr. 43, 385–407. doi:10.1146/annurev-nutr-061021-025153
Barcelos, I. P. D., and Haas, R. H. (2019). CoQ10 and aging. Biology 8, 28. doi:10.3390/biology8020028
Bast, A., Chandler, R. F., Choy, P. C., Delmulle, L. M., Gruenwald, J., Halkes, S. B. A., et al. (2002). Botanical health products, positioning and requirements for effective and safe use. Environ. Toxicol. Pharmacol. 12, 195–211. doi:10.1016/s1382-6689(02)00035-2
Bazan, N. G. (2006). Cell survival matters: docosahexaenoic acid signaling, neuroprotection and photoreceptors. Trends Neurosci. 29, 263–271. doi:10.1016/j.tins.2006.03.005
Bazan, N. G. (2007). Omega-3 fatty acids, pro-inflammatory signaling and neuroprotection. Curr. Opin. Clin. Nutr. and Metabolic Care 10, 136–141. doi:10.1097/MCO.0b013e32802b7030
Beck, M. A., Levander, O. A., and Handy, J. (2003). Selenium deficiency and viral infection. J. Nutr. 133, 1463S–1467S. doi:10.1093/jn/133.5.1463S
Behmer, S. T., Simpson, S. J., and Raubenheimer, D. (2002). Herbivore foraging in chemically heterogeneous environments: nutrients and secondary metabolites. Ecology 83, 2489–2501. doi:10.2307/3071809
Berridge, M. J. (2017). Vitamin D deficiency accelerates ageing and age-related diseases: a novel hypothesis. J. physiology 595, 6825–6836. doi:10.1113/JP274887
Bhargava, R., Chandra, M., Bansal, U., Singh, D., Ranjan, S., and Sharma, S. (2016a). A randomized controlled trial of omega 3 fatty acids in rosacea patients with dry eye symptoms. Curr. eye Res. 41, 1274–1280. doi:10.3109/02713683.2015.1122810
Bhargava, R., and Kumar, P. (2015). Oral omega-3 fatty acid treatment for dry eye in contact lens wearers. Cornea 34, 413–420. doi:10.1097/ICO.0000000000000386
Bhargava, R., Kumar, P., and Arora, Y. (2016b). Short-term omega 3 fatty acids treatment for dry eye in young and middle-aged visual display terminal users. Eye and contact lens 42, 231–236. doi:10.1097/ICL.0000000000000179
Bhargava, R., Kumar, P., Kumar, M., Mehra, N., and Mishra, A. (2013). A randomized controlled trial of omega-3 fatty acids in dry eye syndrome. Int. J. Ophthalmol. 6, 811–816. doi:10.3980/j.issn.2222-3959.2013.06.13
Bhavnani, S. P., Parakh, K., Atreja, A., Druz, R., Graham, G. N., Hayek, S. S., et al. (2017). 2017 Roadmap for innovation—ACC health policy statement on healthcare transformation in the era of digital health, big data, and precision health: a report of the American College of Cardiology Task Force on Health Policy Statements and Systems of Care. J. Am. Coll. Cardiol. 70, 2696–2718. doi:10.1016/j.jacc.2017.10.018
Biesalski, H.-K., Dragsted, L. O., Elmadfa, I., Grossklaus, R., Müller, M., Schrenk, D., et al. (2009). Bioactive compounds: safety and efficacy. Nutrition 25, 1206–1211. doi:10.1016/j.nut.2009.06.014
Bjørklund, G., Shanaida, M., Lysiuk, R., Antonyak, H., Klishch, I., Shanaida, V., et al. (2022a). Selenium: an antioxidant with a critical role in anti-aging. Molecules 27, 6613. doi:10.3390/molecules27196613
Bjørklund, G., Shanaida, M., Lysiuk, R., Butnariu, M., Peana, M., Sarac, I., et al. (2022b). Natural compounds and products from an anti-aging perspective. Molecules 27, 7084. doi:10.3390/molecules27207084
Bjørklund, G., Zou, L., Peana, M., Chasapis, C. T., Hangan, T., Lu, J., et al. (2022c). The role of the thioredoxin system in brain diseases. Antioxidants 11, 2161. doi:10.3390/antiox11112161
Blum, N., and Begemann, G. (2012). Retinoic acid signaling controls the formation, proliferation and survival of the blastema during adult zebrafish fin regeneration. Development 139, 107–116. doi:10.1242/dev.065391
Bonnefont-Rousselot, D. (2016). Resveratrol and cardiovascular diseases. Nutrients 8, 250. doi:10.3390/nu8050250
Borra, M. T., Smith, B. C., and Denu, J. M. (2005). Mechanism of human SIRT1 activation by resveratrol. J. Biol. Chem. 280, 17187–17195. doi:10.1074/jbc.M501250200
Boullata, J. (2005). Natural health product interactions with medication. Nutr. Clin. Pract. 20, 33–51. doi:10.1177/011542650502000133
Bramley, P. M., Elmadfa, I., Kafatos, A., Kelly, F. J., Manios, Y., Roxborough, H. E., et al. (2000). Vitamin E. J. Sci. Food Agric. 80, 913–938. doi:10.1002/(sici)1097-0010(20000515)80:7<913::aid-jsfa600>3.3.co;2-v
Breuss, J. M., Atanasov, A. G., and Uhrin, P. (2019). Resveratrol and its effects on the vascular system. Int. J. Mol. Sci. 20, 1523. doi:10.3390/ijms20071523
Brignole-Baudouin, F., Baudouin, C., Aragona, P., Rolando, M., Labetoulle, M., Pisella, P. J., et al. (2011). A multicentre, double-masked, randomized, controlled trial assessing the effect of oral supplementation of omega-3 and omega-6 fatty acids on a conjunctival inflammatory marker in dry eye patients. Acta Ophthalmol. 89, e591–e597. doi:10.1111/j.1755-3768.2011.02196.x
Brotosudarmo, T. H. P., Limantara, L., Setiyono, E., and Heriyanto, (2020). Structures of astaxanthin and their consequences for therapeutic application. Int. J. Food Sci. 2020, 2156582. doi:10.1155/2020/2156582
Cabral-Pacheco, G. A., Garza-Veloz, I., Castruita-De La Rosa, C., Ramirez-Acuña, J. M., Perez-Romero, B. A., Guerrero-Rodriguez, J. F., et al. (2020). The roles of matrix metalloproteinases and their inhibitors in human diseases. Int. J. Mol. Sci. 21, 9739. doi:10.3390/ijms21249739
Cai, Z., Zhang, J., and Li, H. (2019). Selenium, aging and aging-related diseases. Aging Clin. Exp. Res. 31, 1035–1047. doi:10.1007/s40520-018-1086-7
Calder, P. C. (2013). Omega-3 polyunsaturated fatty acids and inflammatory processes: nutrition or pharmacology? Br. J. Clin. Pharmacol. 75, 645–662. doi:10.1111/j.1365-2125.2012.04374.x
Calder, P. C. (2017). Omega-3 fatty acids and inflammatory processes: from molecules to man. Biochem. Soc. Trans. 45, 1105–1115. doi:10.1042/BST20160474
Calderon-Montano, J., Burgos-Morón, E., Pérez-Guerrero, C., and López-Lázaro, M. (2011). A review on the dietary flavonoid kaempferol. Mini Rev. Med. Chem. 11, 298–344. doi:10.2174/138955711795305335
Cao, S., Wang, C., Yan, J., Li, X., Wen, J., and Hu, C. (2020). Curcumin ameliorates oxidative stress-induced intestinal barrier injury and mitochondrial damage by promoting Parkin dependent mitophagy through AMPK-TFEB signal pathway. Free Radic. Biol. Med. 147, 8–22. doi:10.1016/j.freeradbiomed.2019.12.004
Capozzi, A., Scambia, G., and Lello, S. (2020). Calcium, vitamin D, vitamin K2, and magnesium supplementation and skeletal health. Maturitas 140, 55–63. doi:10.1016/j.maturitas.2020.05.020
Cardoso, B. R., Roberts, B. R., Bush, A. I., and Hare, D. J. (2015). Selenium, selenoproteins and neurodegenerative diseases. Metallomics 7, 1213–1228. doi:10.1039/c5mt00075k
Cardoso, H. D., Dos Santos Junior, E. F., De Santana, D. F., Gonçalves-Pimentel, C., Angelim, M. K., Isaac, A. R., et al. (2014). Omega-3 deficiency and neurodegeneration in the substantia nigra: involvement of increased nitric oxide production and reduced BDNF expression. Biochimica Biophysica Acta (BBA)-General Subj. 1840, 1902–1912. doi:10.1016/j.bbagen.2013.12.023
Carocho, M., and Ferreira, I. C. (2013). A review on antioxidants, prooxidants and related controversy: natural and synthetic compounds, screening and analysis methodologies and future perspectives. Food Chem. Toxicol. 51, 15–25. doi:10.1016/j.fct.2012.09.021
Carr, A., and Frei, B. (1999a). Does vitamin C act as a pro-oxidant under physiological conditions? FASEB J. 13, 1007–1024. doi:10.1096/fasebj.13.9.1007
Carr, A. C., and Frei, B. (1999b). Toward a new recommended dietary allowance for vitamin C based on antioxidant and health effects in humans. Am. J. Clin. Nutr. 69, 1086–1107. doi:10.1093/ajcn/69.6.1086
Carr, A. C., and Maggini, S. (2017). Vitamin C and immune function. Nutrients 9, 1211. doi:10.3390/nu9111211
Carrara, D., Bruno, R. M., Bacca, A., Taddei, S., Duranti, E., Ghiadoni, L., et al. (2016). Cholecalciferol treatment downregulates renin–angiotensin system and improves endothelial function in essential hypertensive patients with hypovitaminosid D. J. Hypertens. 34, 2199–2205. doi:10.1097/HJH.0000000000001072
Cavero, I., Guillon, J.-M., and Holzgrefe, H. H. (2019). Human organotypic bioconstructs from organ-on-chip devices for human-predictive biological insights on drug candidates. Expert Opin. Drug Saf. 18, 651–677. doi:10.1080/14740338.2019.1634689
Cencic, A., and Chingwaru, W. (2010). The role of functional foods, nutraceuticals, and food supplements in intestinal health. Nutrients 2, 611–625. doi:10.3390/nu2060611
Chae, S. Y., Park, R., and Hong, S. W. (2022). Surface-mediated high antioxidant and anti-inflammatory effects of astaxanthin-loaded ultrathin graphene oxide film that inhibits the overproduction of intracellular reactive oxygen species. Biomaterials Res. 26, 30. doi:10.1186/s40824-022-00276-4
Chang, M. X., and Xiong, F. (2020). Astaxanthin and its effects in inflammatory responses and inflammation-associated diseases: recent advances and future directions. Molecules 25, 5342. doi:10.3390/molecules25225342
Chang, P., Liu, J., Yu, Y., Cui, S.-Y., Guo, Z.-H., Chen, G.-M., et al. (2017). Alpha-lipoic acid suppresses extracellular histone-induced release of the infammatory mediator tumor necrosis factor-α by macrophages. Cell. Physiology Biochem. 42, 2559–2568. doi:10.1159/000480217
Chen, N., Bezzina, R., Hinch, E., Lewandowski, P. A., Cameron-Smith, D., Mathai, M. L., et al. (2009). Green tea, black tea, and epigallocatechin modify body composition, improve glucose tolerance, and differentially alter metabolic gene expression in rats fed a high-fat diet. Nutr. Res. 29, 784–793. doi:10.1016/j.nutres.2009.10.003
Chen, W., Jia, Z., Pan, M.-H., and Babu, P. V. A. (2016). Natural products for the prevention of oxidative stress-related diseases: mechanisms and strategies. Oxidative Med. Cell. Longev. 2016, 4628502. doi:10.1155/2016/4628502
Cherniack, E. P., and Troen, B. R. (2013). Resveratrol: effects on lipids and cardiovascular risk. Curr. Cardiovasc. risk Rep. 7, 9–16. doi:10.1007/s12170-012-0289-2
Choi, S.-K., Park, Y.-S., Choi, D.-K., and Chang, H.-I. (2008). Effects of astaxanthin on the production of NO and the expression of COX-2 and iNOS in LPS-stimulated BV2 microglial cells. J. Microbiol. Biotechnol. 18, 1990–1996.
Choi, S.-W., and Friso, S. (2010). Epigenetics: a new bridge between nutrition and health. Adv. Nutr. 1, 8–16. doi:10.3945/an.110.1004
Chung, J. H., Manganiello, V., and Dyck, J. R. (2012). Resveratrol as a calorie restriction mimetic: therapeutic implications. Trends Cell Biol. 22, 546–554. doi:10.1016/j.tcb.2012.07.004
Colalto, C. (2010). Herbal interactions on absorption of drugs: mechanisms of action and clinical risk assessment. Pharmacol. Res. 62, 207–227. doi:10.1016/j.phrs.2010.04.001
Collaborators, G. A. (2022a). Global, regional, and national burden of diseases and injuries for adults 70 years and older: systematic analysis for the Global Burden of Disease 2019 Study. bmj 376, e068208. doi:10.1136/bmj-2021-068208
Collaborators, G. M. D. (2022b). Global, regional, and national burden of 12 mental disorders in 204 countries and territories, 1990–2019: a systematic analysis for the Global Burden of Disease Study 2019. Lancet Psychiatry 9, 137–150. doi:10.1016/S2215-0366(21)00395-3
Comalada, M., Camuesco, D., Sierra, S., Ballester, I., Xaus, J., Gálvez, J., et al. (2005). In vivo quercitrin anti-inflammatory effect involves release of quercetin, which inhibits inflammation through down-regulation of the NF-kappaB pathway. Eur. J. Immunol. 35, 584–592. doi:10.1002/eji.200425778
Cook, N., Jodrell, D. I., and Tuveson, D. A. (2012). Predictive in vivo animal models and translation to clinical trials. Drug Discov. today 17, 253–260. doi:10.1016/j.drudis.2012.02.003
Costa, L. G., Garrick, J. M., Roquè, P. J., and Pellacani, C. (2016). Mechanisms of neuroprotection by quercetin: counteracting oxidative stress and more. Oxidative Med. Cell. Longev. 2016, 2986796. doi:10.1155/2016/2986796
Costa, M. I., Sarmento-Ribeiro, A. B., and Gonçalves, A. C. (2023). Zinc: from biological functions to therapeutic potential. Int. J. Mol. Sci. 24, 4822. doi:10.3390/ijms24054822
Cox, K. H., Pipingas, A., and Scholey, A. B. (2015). Investigation of the effects of solid lipid curcumin on cognition and mood in a healthy older population. J. Psychopharmacol. 29, 642–651. doi:10.1177/0269881114552744
Cragg, G. M., and Newman, D. J. (2013). Natural products: a continuing source of novel drug leads. Biochimica Biophysica Acta (BBA)-General Subj. 1830, 3670–3695. doi:10.1016/j.bbagen.2013.02.008
Dahan, A., and Altman, H. (2004). Food–drug interaction: grapefruit juice augments drug bioavailability—mechanism, extent and relevance. Eur. J. Clin. Nutr. 58, 1–9. doi:10.1038/sj.ejcn.1601736
Dai, H., Coleman, D., Hu, L., Martinez-Cortés, I., Wang, M., Parys, C., et al. (2020). Methionine and arginine supplementation alter inflammatory and oxidative stress responses during lipopolysaccharide challenge in bovine mammary epithelial cells in vitro. J. Dairy Sci. 103, 676–689. doi:10.3168/jds.2019-16631
Dai, Y.-L., Luk, T.-H., Siu, C.-W., Yiu, K.-H., Chan, H.-T., Lee, S. W., et al. (2010). Mitochondrial dysfunction induced by statin contributes to endothelial dysfunction in patients with coronary artery disease. Cardiovasc. Toxicol. 10, 130–138. doi:10.1007/s12012-010-9071-1
Das, U. N. (2006). Essential fatty acids: biochemistry, physiology and pathology. Biotechnol. J. Healthc. Nutr. Technol. 1, 420–439. doi:10.1002/biot.200600012
Davey Smith, G., and Ebrahim, S. (2003). Mendelian randomization’: can genetic epidemiology contribute to understanding environmental determinants of disease? Int. J. Epidemiol. 32, 1–22. doi:10.1093/ije/dyg070
David, A. V. A., Arulmoli, R., and Parasuraman, S. (2016). Overviews of biological importance of quercetin: a bioactive flavonoid. Pharmacogn. Rev. 10, 84–89. doi:10.4103/0973-7847.194044
Deepika, P., and Maurya, P. K. (2022). Health benefits of quercetin in age-related diseases. Molecules 27, 2498. doi:10.3390/molecules27082498
Denayer, T., Stöhr, T., and Van Roy, M. (2014). Animal models in translational medicine: validation and prediction. New Horizons Transl. Med. 2, 5–11.
Desquiret-Dumas, V., Gueguen, N., Leman, G., Baron, S., Nivet-Antoine, V., Chupin, S., et al. (2013). Resveratrol induces a mitochondrial complex I-dependent increase in NADH oxidation responsible for sirtuin activation in liver cells. J. Biol. Chem. 288, 36662–36675. doi:10.1074/jbc.M113.466490
Dreosti, I. E. (2001). Zinc and the gene. Mutat. Research/Fundamental Mol. Mech. Mutagen. 475, 161–167. doi:10.1016/s0027-5107(01)00067-7
Du, G.-J., Zhang, Z., Wen, X.-D., Yu, C., Calway, T., Yuan, C.-S., et al. (2012). Epigallocatechin Gallate (EGCG) is the most effective cancer chemopreventive polyphenol in green tea. Nutrients 4, 1679–1691. doi:10.3390/nu4111679
Dürr, D., Stieger, B., Kullak-Ublick, G. A., Rentsch, K. M., Steinert, H. C., Meier, P. J., et al. (2000). St John's Wort induces intestinal P-glycoprotein/MDR1 and intestinal and hepatic CYP3A4. Clin. Pharmacol. and Ther. 68, 598–604. doi:10.1067/mcp.2000.112240
Dyall, S. C. (2015). Long-chain omega-3 fatty acids and the brain: a review of the independent and shared effects of EPA, DPA and DHA. Front. aging Neurosci. 7, 52. doi:10.3389/fnagi.2015.00052
Dysken, M. W., Sano, M., Asthana, S., Vertrees, J. E., Pallaki, M., Llorente, M., et al. (2014). Effect of vitamin E and memantine on functional decline in Alzheimer disease: the TEAM-AD VA cooperative randomized trial. Jama 311, 33–44. doi:10.1001/jama.2013.282834
Ebrahimi, B., Baroutian, S., Li, J., Zhang, B., Ying, T., and Lu, J. (2023). Combination of marine bioactive compounds and extracts for the prevention and treatment of chronic diseases. Front. Nutr. 9, 1047026. doi:10.3389/fnut.2022.1047026
Ekor, M. (2014). The growing use of herbal medicines: issues relating to adverse reactions and challenges in monitoring safety. Front. Pharmacol. 4, 177. doi:10.3389/fphar.2013.00177
Elbling, L., Weiss, R.-M., Teufelhofer, O., Uhl, M., Knasmueller, S., Schulte-Hermann, R., et al. (2005). Green tea extract and (−)-epigallocatechin-3-gallate, the major tea catechin, exert oxidant but lack antioxidant activities. FASEB J. 19, 807–809. doi:10.1096/fj.04-2915fje
El Monfalouti, H., and Kartah, B. E. (2024). Enhancing polyphenol bioavailability through nanotechnology: current trends and challenges. IntechOpen: London.
Evans, R. G., and Stoddart, G. L. (2017). “Producing health, consuming health care,” in Why are some people healthy and others not? (Routledge), 27–64.
Fairweather-Tait, S. J. (1988). Zinc in human nutrition. Nutr. Res. Rev. 1, 23–37. doi:10.1079/NRR19880005
Falkenberg, L. J., Bellerby, R. G., Connell, S. D., Fleming, L. E., Maycock, B., Russell, B. D., et al. (2020). Ocean acidification and human health. Int. J. Environ. Res. Public Health 17, 4563. doi:10.3390/ijerph17124563
Fan, W., Huang, Y., Zheng, H., Li, S., Li, Z., Yuan, L., et al. (2020). Ginsenosides for the treatment of metabolic syndrome and cardiovascular diseases: pharmacology and mechanisms. Biomed. and Pharmacother. 132, 110915. doi:10.1016/j.biopha.2020.110915
Farruggia, C., Kim, M.-B., Bae, M., Lee, Y., Pham, T. X., Yang, Y., et al. (2018). Astaxanthin exerts anti-inflammatory and antioxidant effects in macrophages in NRF2-dependent and independent manners. J. Nutr. Biochem. 62, 202–209. doi:10.1016/j.jnutbio.2018.09.005
Feart, C., Helmer, C., Merle, B., Herrmann, F. R., Annweiler, C., Dartigues, J.-F., et al. (2017). Associations of lower vitamin D concentrations with cognitive decline and long-term risk of dementia and Alzheimer's disease in older adults. Alzheimer's and Dementia 13, 1207–1216. doi:10.1016/j.jalz.2017.03.003
Fišar, Z., and Hroudová, J. (2024). CoQ10 and mitochondrial dysfunction in Alzheimer’s disease. Antioxidants 13, 191. doi:10.3390/antiox13020191
Freedman, J. E., and Keaney, J. F. (2001). Vitamin E inhibition of platelet aggregation is independent of antioxidant activity. J. Nutr. 131, 374S–377S. doi:10.1093/jn/131.2.374S
Frei, B. (1991). Ascorbic acid protects lipids in human plasma and low-density lipoprotein against oxidative damage. Am. J. Clin. Nutr. 54, 1113S–1118S. doi:10.1093/ajcn/54.6.1113s
Frei, B., England, L., and Ames, B. N. (1989). Ascorbate is an outstanding antioxidant in human blood plasma. Proc. Natl. Acad. Sci. 86, 6377–6381. doi:10.1073/pnas.86.16.6377
Frémont, L., Belguendouz, L., and Delpal, S. (1999). Antioxidant activity of resveratrol and alcohol-free wine polyphenols related to LDL oxidation and polyunsaturated fatty acids. Life Sci. 64, 2511–2521. doi:10.1016/s0024-3205(99)00209-x
Fu, Y.-S., Kang, N., Yu, Y., Mi, Y., Guo, J., Wu, J., et al. (2022). Polyphenols, flavonoids and inflammasomes: the role of cigarette smoke in COPD. Eur. Respir. Rev. 31, 220028. doi:10.1183/16000617.0028-2022
Gao, A. X., Xia, T. C. X., Lin, L. S. Y., Dong, T. T. X., and Tsim, K. W. K. (2023). The neurotrophic activities of brain-derived neurotrophic factor are potentiated by binding with apigenin, a common flavone in vegetables, in stimulating the receptor signaling. CNS Neurosci. and Ther. 29, 2787–2799. doi:10.1111/cns.14230
Genchi, G., Lauria, G., Catalano, A., Carocci, A., and Sinicropi, M. S. (2024). Neuroprotective effects of curcumin in neurodegenerative diseases. Foods 13, 1774. doi:10.3390/foods13111774
Giannaccare, G., Pellegrini, M., Sebastiani, S., Bernabei, F., Roda, M., Taroni, L., et al. (2019). Efficacy of omega-3 fatty acid supplementation for treatment of dry eye disease: a meta-analysis of randomized clinical trials. Cornea 38, 565–573. doi:10.1097/ICO.0000000000001884
Giannaccare, G., Pellegrini, M., Senni, C., Bernabei, F., Scorcia, V., and Cicero, A. F. G. (2020). Clinical applications of astaxanthin in the treatment of ocular diseases: emerging insights. Mar. drugs 18, 239. doi:10.3390/md18050239
Gombart, A. F., Borregaard, N., and Koeffler, H. P. (2005). Human cathelicidin antimicrobial peptide (CAMP) gene is a direct target of the vitamin D receptor and is strongly up-regulated in myeloid cells by 1, 25-dihydroxyvitamin D3. FASEB J. 19, 1067–1077. doi:10.1096/fj.04-3284com
Gowda, U., Mutowo, M. P., Smith, B. J., Wluka, A. E., and Renzaho, A. M. (2015). Vitamin D supplementation to reduce depression in adults: meta-analysis of randomized controlled trials. Nutrition 31, 421–429. doi:10.1016/j.nut.2014.06.017
Granato, M., Rizzello, C., Montani, M. S. G., Cuomo, L., Vitillo, M., Santarelli, R., et al. (2017). Quercetin induces apoptosis and autophagy in primary effusion lymphoma cells by inhibiting PI3K/AKT/mTOR and STAT3 signaling pathways. J. Nutr. Biochem. 41, 124–136. doi:10.1016/j.jnutbio.2016.12.011
Grewal, A. K., Singh, T. G., Sharma, D., Sharma, V., Singh, M., Rahman, M. H., et al. (2021). Mechanistic insights and perspectives involved in neuroprotective action of quercetin. Biomed. and Pharmacother. 140, 111729. doi:10.1016/j.biopha.2021.111729
Grundman, M. (2000). Vitamin E and Alzheimer disease: the basis for additional clinical trials. Am. J. Clin. Nutr. 71, 630S–636S. doi:10.1093/ajcn/71.2.630s
Gutiérrez-Del-Río, I., Villar, C. J., and Lombó, F. (2016). Therapeutic uses of kaempferol: anticancer and antiinflammatory activity. Biosynth. food sources Ther. uses 15, 71.
Gutierrez-Mariscal, F. M., De La Cruz-Ares, S., Torres-Peña, J. D., Alcalá-Diaz, J. F., Yubero-Serrano, E. M., and López-Miranda, J. (2021). Coenzyme Q10 and cardiovascular diseases. Antioxidants 10, 906. doi:10.3390/antiox10060906
Ha, T., Kim, M. K., Park, K.-S., Jung, W., Choo, H., and Chong, Y. (2018). Structural modification of (−)-epigallocatechin gallate (EGCG) shows significant enhancement in mitochondrial biogenesis. J. Agric. food Chem. 66, 3850–3859. doi:10.1021/acs.jafc.8b00364
Haase, H., and Rink, L. (2009). The immune system and the impact of zinc during aging. Immun. and Ageing 6, 9–17. doi:10.1186/1742-4933-6-9
Hadi, P. (2023). A review of the methods and approaches for integrating genetics and metabolomics into personalized nutrition. J. Pharm. Negat. Results, 3823–3830. doi:10.47750/pnr.2023.14.02.447
Hao, H., Zheng, X., and Wang, G. (2014). Insights into drug discovery from natural medicines using reverse pharmacokinetics. Trends Pharmacol. Sci. 35, 168–177. doi:10.1016/j.tips.2014.02.001
Hariri, E., Kassis, N., Iskandar, J.-P., Schurgers, L. J., Saad, A., Abdelfattah, O., et al. (2021). Vitamin K2—a neglected player in cardiovascular health: a narrative review. Open Heart 8, e001715. doi:10.1136/openhrt-2021-001715
Harman, D. (1992). Free radical theory of aging. Mutat. Research/DNAging 275, 257–266. doi:10.1016/0921-8734(92)90030-s
Harman, D. (2003). The free radical theory of aging. Antioxidants Redox Signal. 5, 557–561. doi:10.1089/152308603770310202
Hasler, C. M. (2002). Functional foods: benefits, concerns and challenges—a position paper from the American Council on Science and Health. J. Nutr. 132, 3772–3781. doi:10.1093/jn/132.12.3772
He, L.-X., Ren, J.-W., Liu, R., Chen, Q.-H., Zhao, J., Wu, X., et al. (2017). Ginseng (Panax ginseng Meyer) oligopeptides regulate innate and adaptive immune responses in mice via increased macrophage phagocytosis capacity, NK cell activity and Th cells secretion. Food and Funct. 8, 3523–3532. doi:10.1039/c7fo00957g
He, Y., Yue, Y., Zheng, X., Zhang, K., Chen, S., and Du, Z. (2015). Curcumin, inflammation, and chronic diseases: how are they linked? Molecules 20, 9183–9213. doi:10.3390/molecules20059183
Hemaiswarya, S., and Doble, M. (2006). Potential synergism of natural products in the treatment of cancer. Phytotherapy Res. Int. J. Devoted Pharmacol. Toxicol. Eval. Nat. Prod. Deriv. 20, 239–249. doi:10.1002/ptr.1841
Henderson, V. C., Kimmelman, J., Fergusson, D., Grimshaw, J. M., and Hackam, D. G. (2013). Threats to validity in the design and conduct of preclinical efficacy studies: a systematic review of guidelines for in vivo animal experiments. PLoS Med. 10, e1001489. doi:10.1371/journal.pmed.1001489
Hidalgo-Gutiérrez, A., González-García, P., Díaz-Casado, M. E., Barriocanal-Casado, E., López-Herrador, S., Quinzii, C. M., et al. (2021). Metabolic targets of coenzyme Q10 in mitochondria. Antioxidants 10, 520. doi:10.3390/antiox10040520
Higashida, K., Kim, S. H., Jung, S. R., Asaka, M., Holloszy, J. O., and Han, D.-H. (2013). Effects of resveratrol and SIRT1 on PGC-1α activity and mitochondrial biogenesis: a reevaluation. PLoS Biol. 11, e1001603. doi:10.1371/journal.pbio.1001603
Hishikawa, N., Takahashi, Y., Amakusa, Y., Tanno, Y., Tuji, Y., Niwa, H., et al. (2012). Effects of turmeric on Alzheimer's disease with behavioral and psychological symptoms of dementia. AYU (An international quarterly journal of research in Ayurveda) 33, 499–504. doi:10.4103/0974-8520.110524
Hoffmann, P. R., and Berry, M. J. (2008). The influence of selenium on immune responses. Molecular nutrition and food research 52, 1273–1280. doi:10.1002/mnfr.200700330
Holick, M. F. (2007). Vitamin D deficiency. N. Engl. journal of medicine 357, 266–281. doi:10.1056/NEJMra070553
Holst, B., and Williamson, G. (2008). Nutrients and phytochemicals: from bioavailability to bioefficacy beyond antioxidants. Current opinion in biotechnology 19, 73–82. doi:10.1016/j.copbio.2008.03.003
Hussain, M. S., Altamimi, A. S. A., Afzal, M., Almalki, W. H., Kazmi, I., Alzarea, S. I., et al. (2024). Kaempferol: paving the path for advanced treatments in aging-related diseases. Experimental gerontology 188, 112389. doi:10.1016/j.exger.2024.112389
Ingold, I., and Conrad, M. (2018). Oxidative stress, selenium redox systems including GPX/TXNRD families. Selenium, 111–135. doi:10.1007/978-3-319-95390-8_6
Ioannidis, J. P., Kim, B. Y., and Trounson, A. (2018). How to design preclinical studies in nanomedicine and cell therapy to maximize the prospects of clinical translation. Nature biomedical engineering 2, 797–809. doi:10.1038/s41551-018-0314-y
Ishii, H. M., Murakashi, E., Igarashi-Takeuchi, H., Shoji, H., and Numabe, Y. (2017). Alpha-lipoic acid inhibits NF-κB signal transduced inflammatory cytokines secretion in LPS-induced human gingival fibroblasts. Nihon Shishubyo Gakkai Kaishi (Journal of the Japanese Society of Periodontology) 59, 28–38. doi:10.2329/perio.59.28
Jang, S.-Y., Kang, H. T., and Hwang, E. S. (2012). Nicotinamide-induced mitophagy: event mediated by high NAD+/NADH ratio and SIRT1 protein activation. Journal of Biological Chemistry 287, 19304–19314. doi:10.1074/jbc.M112.363747
Jarosz, M., Olbert, M., Wyszogrodzka, G., Młyniec, K., and Librowski, T. (2017). Antioxidant and anti-inflammatory effects of zinc. Zinc-dependent NF-κB signaling. Inflammopharmacology 25, 11–24. doi:10.1007/s10787-017-0309-4
Jaul, E., and Barron, J. (2017). Age-related diseases and clinical and public health implications for the 85 years old and over population. Frontiers in public health 5, 335. doi:10.3389/fpubh.2017.00335
Jeong, J., Juhn, K., Lee, H., Kim, S.-H., Min, B.-H., Lee, K.-M., et al. (2007). SIRT1 promotes DNA repair activity and deacetylation of Ku70. Experimental and molecular medicine 39, 8–13. doi:10.1038/emm.2007.2
Jiang, Y.-H., Jiang, L.-Y., Wang, Y.-C., Ma, D.-F., and Li, X. (2020). Quercetin attenuates atherosclerosis via modulating oxidized LDL-induced endothelial cellular senescence. Frontiers in pharmacology 11, 512. doi:10.3389/fphar.2020.00512
Jin, Z., Chang, B., Wei, Y., Yang, Y., Zhang, H., Liu, J., et al. (2022). Curcumin exerts chondroprotective effects against osteoarthritis by promoting AMPK/PINK1/Parkin-mediated mitophagy. Biomedicine and Pharmacotherapy 151, 113092. doi:10.1016/j.biopha.2022.113092
Johnson, E. A., and An, G.-H. (1991). Astaxanthin from microbial sources. Critical reviews in Biotechnology 11, 297–326. doi:10.3109/07388559109040622
Johnson, W. W. (2008). Cytochrome P450 inactivation by pharmaceuticals and phytochemicals: therapeutic relevance. Drug metabolism reviews 40, 101–147. doi:10.1080/03602530701836704
Jung, W.-W. (2014). Protective effect of apigenin against oxidative stress-induced damage in osteoblastic cells. International journal of molecular medicine 33, 1327–1334. doi:10.3892/ijmm.2014.1666
Jüni, P., Altman, D. G., and Egger, M. (2001). Systematic reviews in health care: assessing the quality of controlled clinical trials. Bmj 323, 42–46. doi:10.1136/bmj.323.7303.42
Juraschek, S. P., Guallar, E., Appel, L. J., and Miller Iii, E. R. (2012). Effects of vitamin C supplementation on blood pressure: a meta-analysis of randomized controlled trials. The American journal of clinical nutrition 95, 1079–1088. doi:10.3945/ajcn.111.027995
Kadian, M., Sharma, G., Pandita, S., Sharma, K., Shrivasatava, K., Saini, N., et al. (2022). The impact of coenzyme Q10 on neurodegeneration: a comprehensive review. Current Pharmacology Reports 8, 1–19. doi:10.1007/s40495-021-00273-6
Kaeberlein, M., Rabinovitch, P. S., and Martin, G. M. (2015). Healthy aging: the ultimate preventative medicine. Science 350, 1191–1193. doi:10.1126/science.aad3267
Kagan, V., Fabisiak, J., and Quinn, P. (2000). Coenzyme Q and vitamin E need each other as antioxidants. Protoplasma 214, 11–18. doi:10.1007/bf02524257
Kamaliyan, Z., and Clarke, T. L. (2024). Zinc finger proteins: guardians of genome stability. Frontiers in Cell and Developmental Biology 12, 1448789. doi:10.3389/fcell.2024.1448789
Kamisah, Y., Jalil, J., Yunos, N. M., and Zainalabidin, S. (2023). Cardioprotective properties of kaempferol: a review. Plants 12, 2096. doi:10.3390/plants12112096
Kapoor, K., Alfaddagh, A., Al Rifai, M., Bhatt, D. L., Budoff, M. J., Nasir, K., et al. (2021). Association between omega-3 fatty acid levels and risk for incident major bleeding events and atrial fibrillation: MESA. Journal of the American Heart Association 10, e021431. doi:10.1161/JAHA.121.021431
Kaput, J., and Rodriguez, R. L. (2006). Nutritional genomics: discovering the path to personalized nutrition. John Wiley and Sons.
Kawasaki, I., Jeong, M.-H., Oh, B.-K., and Shim, Y.-H. (2010). Apigenin inhibits larval growth of Caenorhabditis elegans through DAF-16 activation. FEBS letters 584, 3587–3591. doi:10.1016/j.febslet.2010.07.026
Keijer, J., Escoté, X., Galmés, S., Palou-March, A., Serra, F., Aldubayan, M. A., et al. (2024). Omics biomarkers and an approach for their practical implementation to delineate health status for personalized nutrition strategies. Critical reviews in food science and nutrition 64, 8279–8307. doi:10.1080/10408398.2023.2198605
Kennedy, B. K., Berger, S. L., Brunet, A., Campisi, J., Cuervo, A. M., Epel, E. S., et al. (2014). Geroscience: linking aging to chronic disease. Cell 159, 709–713. doi:10.1016/j.cell.2014.10.039
Khairy, E. Y., and Attia, M. M. (2021). Protective effects of vitamin D on neurophysiologic alterations in brain aging: role of brain-derived neurotrophic factor (BDNF). Nutritional neuroscience 24, 650–659. doi:10.1080/1028415X.2019.1665854
Khalil, Z., Alam, B., Akbari, A. R., and Sharma, H. (2021). The medical benefits of vitamin K2 on calcium-related disorders. Nutrients 13, 691. doi:10.3390/nu13020691
Khan, A. A., Mudassir, J., Mohtar, N., and Darwis, Y. (2013). Advanced drug delivery to the lymphatic system: lipid-based nanoformulations. International journal of nanomedicine 8, 2733–2744. doi:10.2147/IJN.S41521
Khan, H., Ullah, H., Aschner, M., Cheang, W. S., and Akkol, E. K. (2019). Neuroprotective effects of quercetin in Alzheimer’s disease. Biomolecules 10, 59. doi:10.3390/biom10010059
Khayatan, D., Razavi, S. M., Arab, Z. N., Hosseini, Y., Niknejad, A., Momtaz, S., et al. (2024). Superoxide dismutase: a key target for the neuroprotective effects of curcumin. Molecular and Cellular Biochemistry 479, 693–705. doi:10.1007/s11010-023-04757-5
Kiani, A. K., Bonetti, G., Donato, K., Kaftalli, J., Herbst, K. L., Stuppia, L., et al. (2022). Polymorphisms, diet and nutrigenomics. Journal of preventive medicine and hygiene 63, E125–E141. doi:10.15167/2421-4248/jpmh2022.63.2S3.2754
Kidd, P. M. (2010). Vitamins D and K as pleiotropic nutrients: clinical importance to the skeletal and cardiovascular systems and preliminary evidence for synergy. Altern Med Rev 15, 199–222.
Kim, J. H., Yi, Y.-S., Kim, M.-Y., and Cho, J. Y. (2017). Role of ginsenosides, the main active components of Panax ginseng, in inflammatory responses and diseases. Journal of ginseng research 41, 435–443. doi:10.1016/j.jgr.2016.08.004
Komes, D., Horžić, D., Belščak, A., Ganić, K. K., and Vulić, I. (2010). Green tea preparation and its influence on the content of bioactive compounds. Food research international 43, 167–176. doi:10.1016/j.foodres.2009.09.022
Koushki, M., Lakzaei, M., Khodabandehloo, H., Hosseini, H., Meshkani, R., and Panahi, G. (2020). Therapeutic effect of resveratrol supplementation on oxidative stress: a systematic review and meta-analysis of randomised controlled trials. Postgraduate Medical Journal 96, 197–205. doi:10.1136/postgradmedj-2019-136415
Kuhl, H. (2005). Pharmacology of estrogens and progestogens: influence of different routes of administration. Climacteric 8, 3–63. doi:10.1080/13697130500148875
Kumari, S., Mehta, S. L., and Li, P. A. (2012). Glutamate induces mitochondrial dynamic imbalance and autophagy activation: preventive effects of selenium. PloS one 7, e39382. doi:10.1371/journal.pone.0039382
Kunle, O. F., Egharevba, H. O., and Ahmadu, P. O. (2012). Standardization of herbal medicines-A review. International journal of biodiversity and conservation 4, 101–112. doi:10.5897/ijbc11.163
Kuo, C.-Y., Zupkó, I., Chang, F.-R., Hunyadi, A., Wu, C.-C., Weng, T.-S., et al. (2016). Dietary flavonoid derivatives enhance chemotherapeutic effect by inhibiting the DNA damage response pathway. Toxicology and Applied Pharmacology 311, 99–105. doi:10.1016/j.taap.2016.09.019
Kurkov, S. V., and Loftsson, T. (2013). Cyclodextrins. International journal of pharmaceutics 453, 167–180. doi:10.1016/j.ijpharm.2012.06.055
Kurnatowska, I., Grzelak, P., Masajtis-Zagajewska, A., Kaczmarska, M., Stefańczyk, L., Vermeer, C., et al. (2015). Effect of vitamin K2 on progression of atherosclerosis and vascular calcification in nondialyzed patients with chronic kidney disease stages 3-5. Polskie Archiwum Medycyny Wewnętrznej 125, 631–640. doi:10.20452/pamw.3041
Lagoumintzis, G., and Patrinos, G. P. (2023). Triangulating nutrigenomics, metabolomics and microbiomics toward personalized nutrition and healthy living. Human Genomics 17, 109. doi:10.1186/s40246-023-00561-w
Lee, M. Y., and Hu, T. (2019). Computational methods for the discovery of metabolic markers of complex traits. Metabolites 9, 66. doi:10.3390/metabo9040066
Lee, S., and Rhee, D.-K. (2017). Effects of ginseng on stress-related depression, anxiety, and the hypothalamic–pituitary–adrenal axis. Journal of ginseng research 41, 589–594. doi:10.1016/j.jgr.2017.01.010
Lee, S.-J., Bai, S.-K., Lee, K.-S., Namkoong, S., Na, H.-J., Ha, K.-S., et al. (2003). Astaxanthin inhibits nitric oxide production and inflammatory gene expression by suppressing IκB kinase-dependent NF-κB activation. Molecules and cells 16, 97–105. doi:10.1016/s1016-8478(23)13772-1
Lee, S. R. (2018). Critical role of zinc as either an antioxidant or a prooxidant in cellular systems. Oxidative medicine and cellular longevity 2018, 9156285. doi:10.1155/2018/9156285
Lee, W. J., Song, K.-H., Koh, E. H., Won, J. C., Kim, H. S., Park, H.-S., et al. (2005). Alpha-lipoic acid increases insulin sensitivity by activating AMPK in skeletal muscle. Biochemical and biophysical research communications 332, 885–891. doi:10.1016/j.bbrc.2005.05.035
Leung, K. W., and Wong, A.S.-T. (2010). Pharmacology of ginsenosides: a literature review. Chinese medicine 5, 20–27. doi:10.1186/1749-8546-5-20
Levine, M., Conry-Cantilena, C., Wang, Y., Welch, R. W., Washko, P. W., Dhariwal, K. R., et al. (1996). Vitamin C pharmacokinetics in healthy volunteers: evidence for a recommended dietary allowance. Proceedings of the National Academy of Sciences 93, 3704–3709. doi:10.1073/pnas.93.8.3704
Leyane, T. S., Jere, S. W., and Houreld, N. N. (2022). Oxidative stress in ageing and chronic degenerative pathologies: molecular mechanisms involved in counteracting oxidative stress and chronic inflammation. International journal of molecular sciences 23, 7273. doi:10.3390/ijms23137273
Li, C., and Wang, J. (2024). Clinical applications and safety of aromatic medicinal plants: efficacy evaluation and standard formulation. Medicinal Plant Research 14. doi:10.5376/mpr.2024.14.0007
Li, R., Wang, X., Qin, T., Qu, R., and Ma, S. (2016a). Apigenin ameliorates chronic mild stress-induced depressive behavior by inhibiting interleukin-1β production and NLRP3 inflammasome activation in the rat brain. Behavioural Brain research 296, 318–325. doi:10.1016/j.bbr.2015.09.031
Li, W., Van Wely, M., Gurrin, L., and Mol, B. W. (2020a). Integrity of randomized controlled trials: challenges and solutions. Fertility and Sterility 113, 1113–1119. doi:10.1016/j.fertnstert.2020.04.018
Li, X., Lu, L., Chen, J., Zhang, C., Chen, H., and Huang, H. (2020b). New insight into the mechanisms of ginkgo biloba extract in vascular aging prevention. Current Vascular Pharmacology 18, 334–345. doi:10.2174/1570161117666190621150725
Li, X., Zhou, N., Wang, J., Liu, Z., Wang, X., Zhang, Q., et al. (2018). Quercetin suppresses breast cancer stem cells (CD44+/CD24−) by inhibiting the PI3K/Akt/mTOR-signaling pathway. Life sciences 196, 56–62. doi:10.1016/j.lfs.2018.01.014
Li, Y., Yao, J., Han, C., Yang, J., Chaudhry, M. T., Wang, S., et al. (2016b). Quercetin, inflammation and immunity. Nutrients 8, 167. doi:10.3390/nu8030167
Li, Y., Zhang, J.-J., Xu, D.-P., Zhou, T., Zhou, Y., Li, S., et al. (2016c). Bioactivities and health benefits of wild fruits. International journal of molecular sciences 17, 1258. doi:10.3390/ijms17081258
Liao, L.-Y., He, Y.-F., Li, L., Meng, H., Dong, Y.-M., Yi, F., et al. (2018). A preliminary review of studies on adaptogens: comparison of their bioactivity in TCM with that of ginseng-like herbs used worldwide. Chinese medicine 13, 57–12. doi:10.1186/s13020-018-0214-9
Liao, W., Chen, L., Ma, X., Jiao, R., Li, X., and Wang, Y. (2016). Protective effects of kaempferol against reactive oxygen species-induced hemolysis and its antiproliferative activity on human cancer cells. European Journal of Medicinal Chemistry 114, 24–32. doi:10.1016/j.ejmech.2016.02.045
Lim, H., Park, H., and Kim, H. P. (2015). Effects of flavonoids on senescence-associated secretory phenotype formation from bleomycin-induced senescence in BJ fibroblasts. Biochemical Pharmacology 96, 337–348. doi:10.1016/j.bcp.2015.06.013
Lin, C.-H., Chang, C.-Y., Lee, K.-R., Lin, H.-J., Chen, T.-H., and Wan, L. (2015). Flavones inhibit breast cancer proliferation through the Akt/FOXO3a signaling pathway. BMC cancer 15, 958–1012. doi:10.1186/s12885-015-1965-7
Lin, X., Bai, D., Wei, Z., Zhang, Y., Huang, Y., Deng, H., et al. (2019). Curcumin attenuates oxidative stress in RAW264. 7 cells by increasing the activity of antioxidant enzymes and activating the Nrf2-Keap1 pathway. PloS one 14, e0216711. doi:10.1371/journal.pone.0216711
Lipscomb, J. C., and Poet, T. S. (2008). In vitro measurements of metabolism for application in pharmacokinetic modeling. Pharmacology and therapeutics 118, 82–103. doi:10.1016/j.pharmthera.2008.01.006
Littarru, G. P., and Tiano, L. (2007). Bioenergetic and antioxidant properties of coenzyme Q 10: recent developments. Molecular biotechnology 37, 31–37. doi:10.1007/s12033-007-0052-y
Liu, R. H. (2003). Health benefits of fruit and vegetables are from additive and synergistic combinations of phytochemicals. The American journal of clinical nutrition 78, 517S–520S. doi:10.1093/ajcn/78.3.517S
Liu, S., Cheng, L., Liu, Y., Zhan, S., Wu, Z., and Zhang, X. (2023). Relationship between dietary polyphenols and gut microbiota: new clues to improve cognitive disorders, mood disorders and circadian rhythms. Foods 12, 1309. doi:10.3390/foods12061309
Liu, Y., Ren, X., Fan, C., Wu, W., Zhang, W., and Wang, Y. (2022). Health benefits, food applications, and sustainability of microalgae-derived N-3 PUFA. Foods 11, 1883. doi:10.3390/foods11131883
Livney, Y. D. (2016). “Nanoencapsulation technologies,” in Engineering foods for bioactives stability and delivery (Springer), 143–169.
Loftsson, T., and Brewster, M. E. (2012). Cyclodextrins as functional excipients: methods to enhance complexation efficiency. Journal of pharmaceutical sciences 101, 3019–3032. doi:10.1002/jps.23077
López-Otín, C., Blasco, M. A., Partridge, L., Serrano, M., and Kroemer, G. (2013). The hallmarks of aging. Cell 153, 1194–1217. doi:10.1016/j.cell.2013.05.039
Ma, C., Wang, Y., Dong, L., Li, M., and Cai, W. (2015). Anti-inflammatory effect of resveratrol through the suppression of NF-κB and JAK/STAT signaling pathways. Acta biochimica et biophysica Sinica 47, 207–213. doi:10.1093/abbs/gmu135
Madunić, J., Madunić, I. V., Gajski, G., Popić, J., and Garaj-Vrhovac, V. (2018). Apigenin: a dietary flavonoid with diverse anticancer properties. Cancer letters 413, 11–22. doi:10.1016/j.canlet.2017.10.041
Mahran, R. I., Hagras, M. M., Sun, D., and Brenner, D. E. (2017). Bringing curcumin to the clinic in cancer prevention: a review of strategies to enhance bioavailability and efficacy. The AAPS journal 19, 54–81. doi:10.1208/s12248-016-0003-2
Majidinia, M., Bishayee, A., and Yousefi, B. (2019). Polyphenols: major regulators of key components of DNA damage response in cancer. DNA repair 82, 102679. doi:10.1016/j.dnarep.2019.102679
Mancini, E., Beglinger, C., Drewe, J., Zanchi, D., Lang, U. E., and Borgwardt, S. (2017). Green tea effects on cognition, mood and human brain function: a systematic review. Phytomedicine 34, 26–37. doi:10.1016/j.phymed.2017.07.008
Mantle, D., and Dybring, A. (2020). Bioavailability of coenzyme Q10: an overview of the absorption process and subsequent metabolism. Antioxidants 9, 386. doi:10.3390/antiox9050386
Marreiro, D. D. N., Cruz, K. J. C., Morais, J. B. S., Beserra, J. B., Severo, J. S., and De Oliveira, A. R. S. (2017). Zinc and oxidative stress: current mechanisms. Antioxidants 6, 24. doi:10.3390/antiox6020024
Martin, K. R., and Appel, C. L. (2009). Polyphenols as dietary supplements: a double-edged sword. Nutrition and Dietary Supplements, 1–12. doi:10.2147/nds.s6422
May, J. M., and Harrison, F. E. (2013). Role of vitamin C in the function of the vascular endothelium. Antioxidants and redox signaling 19, 2068–2083. doi:10.1089/ars.2013.5205
Mcgonigle, P., and Ruggeri, B. (2014). Animal models of human disease: challenges in enabling translation. Biochemical pharmacology 87, 162–171. doi:10.1016/j.bcp.2013.08.006
Meng, T., Xiao, D., Muhammed, A., Deng, J., Chen, L., and He, J. (2021). Anti-inflammatory action and mechanisms of resveratrol. Molecules 26, 229. doi:10.3390/molecules26010229
Meng, X., Zhou, J., Zhao, C.-N., Gan, R.-Y., and Li, H.-B. (2020). Health benefits and molecular mechanisms of resveratrol: a narrative review. Foods 9, 340. doi:10.3390/foods9030340
Meydani, M. (2001). Vitamin E and atherosclerosis: beyond prevention of LDL oxidation. The Journal of nutrition 131, 366S–368S. doi:10.1093/jn/131.2.366S
Michaud, M., Balardy, L., Moulis, G., Gaudin, C., Peyrot, C., Vellas, B., et al. (2013). Proinflammatory cytokines, aging, and age-related diseases. Journal of the American Medical Directors Association 14, 877–882. doi:10.1016/j.jamda.2013.05.009
Mirmiran, P., Bahadoran, Z., and Gaeini, Z. (2021). Common limitations and challenges of dietary clinical trials for translation into clinical practices. International journal of endocrinology and metabolism 19, e108170. doi:10.5812/ijem.108170
Mohammadi, S., Barzegari, A., Dehnad, A., Barar, J., and Omidi, Y. (2021). Astaxanthin protects mesenchymal stem cells from oxidative stress by direct scavenging of free radicals and modulation of cell signaling. Chemico-biological interactions 333, 109324. doi:10.1016/j.cbi.2020.109324
Monacelli, F., Acquarone, E., Giannotti, C., Borghi, R., and Nencioni, A. (2017). Vitamin C, aging and Alzheimer’s disease. Nutrients 9, 670. doi:10.3390/nu9070670
Mondola, P., Damiano, S., Sasso, A., and Santillo, M. (2016). The Cu, Zn superoxide dismutase: not only a dismutase enzyme. Frontiers in physiology 7, 594. doi:10.3389/fphys.2016.00594
Mora, J. R., Iwata, M., and Von Andrian, U. H. (2008). Vitamin effects on the immune system: vitamins A and D take centre stage. Nature reviews immunology 8, 685–698. doi:10.1038/nri2378
Mortensen, S. A., Rosenfeldt, F., Kumar, A., Dolliner, P., Filipiak, K. J., Pella, D., et al. (2014). The effect of coenzyme Q10 on morbidity and mortality in chronic heart failure: results from Q-SYMBIO: a randomized double-blind trial. JACC Heart Failure 2, 641–649. doi:10.1016/j.jchf.2014.06.008
Mozaffarian, D., and Wu, J. H. (2011). Omega-3 fatty acids and cardiovascular disease: effects on risk factors, molecular pathways, and clinical events. Journal of the American College of Cardiology 58, 2047–2067. doi:10.1016/j.jacc.2011.06.063
Na, S., Ahmad, S., Karim, S., Ibrahim, I., Alkreathy, H., Alsieni, M., and Khan, M. (2022). Effect of vitamin K on bone mineral density and fracture risk in adults: systematic review and meta-analysis. Biomedicines 10, 1048. doi:10.3390/biomedicines10051048
Naguib, Y. M. (2000). Antioxidant activities of astaxanthin and related carotenoids. Journal of agricultural and food chemistry 48, 1150–1154. doi:10.1021/jf991106k
Nelson, K. M., Dahlin, J. L., Bisson, J., Graham, J., Pauli, G. F., and Walters, M. A. (2017). The essential medicinal chemistry of curcumin: miniperspective. Journal of medicinal chemistry 60, 1620–1637. doi:10.1021/acs.jmedchem.6b00975
Newman, D. J., Cragg, G. M., and Snader, K. M. (2003). Natural products as sources of new drugs over the period 1981− 2002. Journal of natural products 66, 1022–1037. doi:10.1021/np030096l
Nezhad Salari, A. M., Rasoulizadeh, Z., Shabgah, A. G., Vakili-Ghartavol, R., Sargazi, G., and Gholizadeh Navashenaq, J. (2024). Exploring the mechanisms of kaempferol in neuroprotection: implications for neurological disorders. Cell Biochemistry and Function 42, e3964. doi:10.1002/cbf.3964
Nie, C., He, T., Zhang, W., Zhang, G., and Ma, X. (2018). Branched chain amino acids: beyond nutrition metabolism. International journal of molecular sciences 19, 954. doi:10.3390/ijms19040954
Niki, E. (2014). Role of vitamin E as a lipid-soluble peroxyl radical scavenger: in vitro and in vivo evidence. Free Radical Biology and Medicine 66, 3–12. doi:10.1016/j.freeradbiomed.2013.03.022
Nobakht-Haghighi, N., Rahimifard, M., Baeeri, M., Rezvanfar, M. A., Moini Nodeh, S., Haghi-Aminjan, H., et al. (2018). Regulation of aging and oxidative stress pathways in aged pancreatic islets using alpha-lipoic acid. Molecular and Cellular Biochemistry 449, 267–276. doi:10.1007/s11010-018-3363-3
Norman, P., and Powell, J. (2014). Vitamin D and cardiovascular disease. Circulation research 114, 379–393. doi:10.1161/CIRCRESAHA.113.301241
Nunes, Y. C., Mendes, N. M., Pereira De Lima, E., Chehadi, A. C., Lamas, C. B., Haber, J. F., et al. (2024). Curcumin: a golden approach to healthy aging: a systematic review of the evidence. Nutrients 16, 2721. doi:10.3390/nu16162721
Obeid, M. A., Al Qaraghuli, M. M., Alsaadi, M., Alzahrani, A. R., Niwasabutra, K., and Ferro, V. A. (2017). Delivering natural products and biotherapeutics to improve drug efficacy. Therapeutic delivery 8, 947–956. doi:10.4155/tde-2017-0060
Ohishi, T., Goto, S., Monira, P., Isemura, M., and Nakamura, Y. (2016). Anti-inflammatory action of green tea. Anti-Inflammatory and Anti-Allergy Agents in Medicinal Chemistry (Formerly Current Medicinal Chemistry-Anti-Inflammatory and Anti-Allergy Agents) 15, 74–90. doi:10.2174/1871523015666160915154443
Oliveira, C. S., Piccoli, B. C., Nogara, P. A., Pereira, M. E., De Carvalho, K. a.T., Skalny, A. V., et al. (2023). “Selenium neuroprotection in neurodegenerative disorders,” in Handbook of neurotoxicity (Springer), 2489–2523.
Organization, W. H. (2001). The world health report 2001: mental health: new understanding. new hope.
Otsuka, T., Shimazawa, M., Nakanishi, T., Ohno, Y., Inoue, Y., Tsuruma, K., et al. (2013). Protective effects of a dietary carotenoid, astaxanthin, against light-induced retinal damage. Journal of Pharmacological Sciences 123, 209–218. doi:10.1254/jphs.13066fp
Pal, M. K., Jaiswar, S. P., Dwivedi, A., Goyal, S., Dwivedi, V. N., Pathak, A. K., et al. (2017). Synergistic effect of graphene oxide coated nanotised apigenin with paclitaxel (GO-NA/PTX): a ROS dependent mitochondrial mediated apoptosis in ovarian cancer. Anti-Cancer Agents in Medicinal Chemistry (Formerly Current Medicinal Chemistry-Anti-Cancer Agents) 17, 1721–1732. doi:10.2174/1871520617666170425094549
Palma, M. A., Ribera, L. A., and Knutson, R. D. (2016). The era of the functional consumer. Journal of food products marketing 22, 555–570. doi:10.1080/10454446.2015.1121425
Pan, Q., Xie, L., Zhu, H., Zong, Z., Wu, D., Liu, R., et al. (2024). Curcumin-incorporated EGCG-based nano-antioxidants alleviate colon and kidney inflammation via antioxidant and anti-inflammatory therapy. Regenerative Biomaterials 11, rbae122. doi:10.1093/rb/rbae122
Partridge, L., Deelen, J., and Slagboom, P. E. (2018). Facing up to the global challenges of ageing. Nature 561, 45–56. doi:10.1038/s41586-018-0457-8
Pathak, K., and Raghuvanshi, S. (2015). Oral bioavailability: issues and solutions via nanoformulations. Clinical pharmacokinetics 54, 325–357. doi:10.1007/s40262-015-0242-x
Patil, R. H., Babu, R., Naveen Kumar, M., Kiran Kumar, K., Hegde, S. M., Nagesh, R., et al. (2016). Anti-inflammatory effect of apigenin on LPS-induced pro-inflammatory mediators and AP-1 factors in human lung epithelial cells. Inflammation 39, 138–147. doi:10.1007/s10753-015-0232-z
Pereira, C. P. M., Souza, A. C. R., Vasconcelos, A. R., Prado, P. S., and Name, J. J. (2021). Antioxidant and anti‑inflammatory mechanisms of action of astaxanthin in cardiovascular diseases (Review). International journal of molecular medicine 47, 37–48. doi:10.3892/ijmm.2020.4783
Petersen, P. E., and Kwan, S. (2011). Equity, social determinants and public health programmes–the case of oral health. Community dentistry and oral epidemiology 39, 481–487. doi:10.1111/j.1600-0528.2011.00623.x
Pettersen, J. A. (2017). Does high dose vitamin D supplementation enhance cognition? a randomized trial in healthy adults. Experimental gerontology 90, 90–97. doi:10.1016/j.exger.2017.01.019
Pludowski, P., Holick, M. F., Pilz, S., Wagner, C. L., Hollis, B. W., Grant, W. B., et al. (2013). Vitamin D effects on musculoskeletal health, immunity, autoimmunity, cardiovascular disease, cancer, fertility, pregnancy, dementia and mortality—a review of recent evidence. Autoimmunity reviews 12, 976–989. doi:10.1016/j.autrev.2013.02.004
Possemiers, S., Bolca, S., Verstraete, W., and Heyerick, A. (2011). The intestinal microbiome: a separate organ inside the body with the metabolic potential to influence the bioactivity of botanicals. Fitoterapia 82, 53–66. doi:10.1016/j.fitote.2010.07.012
Pournourmohammadi, S., Grimaldi, M., Stridh, M. H., Lavallard, V., Waagepetersen, H. S., Wollheim, C. B., et al. (2017). Epigallocatechin-3-gallate (EGCG) activates AMPK through the inhibition of glutamate dehydrogenase in muscle and pancreatic ß-cells: a potential beneficial effect in the pre-diabetic state? The international journal of biochemistry and Cell biology 88, 220–225. doi:10.1016/j.biocel.2017.01.012
Powell, M. D., Read, K. A., Sreekumar, B. K., and Oestreich, K. J. (2019). Ikaros zinc finger transcription factors: regulators of cytokine signaling pathways and CD4+ T helper cell differentiation. Frontiers in immunology 10, 1299. doi:10.3389/fimmu.2019.01299
Pradhan, N., Singh, C., and Singh, A. (2021). Coenzyme Q10 a mitochondrial restorer for various brain disorders. Naunyn-Schmiedeberg's Archives of Pharmacology 394, 2197–2222. doi:10.1007/s00210-021-02161-8
Prasad, A. S. (2014). Zinc is an antioxidant and anti-inflammatory agent: its role in human health. Frontiers in nutrition 1, 100515. doi:10.3389/fnut.2014.00014
Prasanth, M. I., Sivamaruthi, B. S., Chaiyasut, C., and Tencomnao, T. (2019). A review of the role of green tea (Camellia sinensis) in antiphotoaging, stress resistance, neuroprotection, and autophagy. Nutrients 11, 474. doi:10.3390/nu11020474
Prince, M. J., Wu, F., Guo, Y., Robledo, L. M. G., O'donnell, M., Sullivan, R., et al. (2015). The burden of disease in older people and implications for health policy and practice. The lancet 385, 549–562. doi:10.1016/S0140-6736(14)61347-7
Puri, V., Nagpal, M., Singh, I., Singh, M., Dhingra, G. A., Huanbutta, K., et al. (2022). A comprehensive review on nutraceuticals: therapy support and formulation challenges. Nutrients 14, 4637. doi:10.3390/nu14214637
Pyo, I. S., Yun, S., Yoon, Y. E., Choi, J.-W., and Lee, S.-J. (2020). Mechanisms of aging and the preventive effects of resveratrol on age-related diseases. Molecules 25, 4649. doi:10.3390/molecules25204649
Rabanal-Ruiz, Y., Llanos-González, E., and Alcain, F. J. (2021). The use of coenzyme Q10 in cardiovascular diseases. Antioxidants 10, 755. doi:10.3390/antiox10050755
Rahimi, R., and Abdollahi, M. (2012). An update on the ability of St. John's wort to affect the metabolism of other drugs. Expert opinion on drug metabolism and toxicology 8, 691–708. doi:10.1517/17425255.2012.680886
Rahul, Y., and Siddique, Y. H. (2021). Neurodegenerative diseases and flavonoids: special reference to kaempferol. CNS and Neurological Disorders-Drug Targets-CNS and Neurological Disorders 20, 327–342. doi:10.2174/1871527320666210129122033
Rainey-Smith, S. R., Brown, B. M., Sohrabi, H. R., Shah, T., Goozee, K. G., Gupta, V. B., et al. (2016). Curcumin and cognition: a randomised, placebo-controlled, double-blind study of community-dwelling older adults. British Journal of Nutrition 115, 2106–2113. doi:10.1017/S0007114516001203
Ratan, Z. A., Youn, S. H., Kwak, Y.-S., Han, C.-K., Haidere, M. F., Kim, J. K., et al. (2021). Adaptogenic effects of Panax ginseng on modulation of immune functions. Journal of ginseng research 45, 32–40. doi:10.1016/j.jgr.2020.09.004
Rather, R. A., and Bhagat, M. (2020). Quercetin as an innovative therapeutic tool for cancer chemoprevention: molecular mechanisms and implications in human health. Cancer medicine 9, 9181–9192. doi:10.1002/cam4.1411
Rautio, J., Kumpulainen, H., Heimbach, T., Oliyai, R., Oh, D., Järvinen, T., et al. (2008). Prodrugs: design and clinical applications. Nature reviews Drug discovery 7, 255–270. doi:10.1038/nrd2468
Rayman, M. P. (2008). Food-chain selenium and human health: emphasis on intake. British journal of nutrition 100, 254–268. doi:10.1017/S0007114508939830
Rayman, M. P. (2012). Selenium and human health. The Lancet 379, 1256–1268. doi:10.1016/S0140-6736(11)61452-9
Reginster, J. Y., Deroisy, R., Rovati, L. C., Lee, R. L., Lejeune, E., Bruyere, O., et al. (2001). Long-term effects of glucosamine sulphate on osteoarthritis progression: a randomised, placebo-controlled clinical trial. The Lancet 357, 251–256. doi:10.1016/S0140-6736(00)03610-2
Rigamonti, A. E., Frigerio, G., Caroli, D., De Col, A., Cella, S. G., Sartorio, A., et al. (2023). A metabolomics-based investigation of the effects of a short-term body weight reduction program in a cohort of adolescents with obesity: a prospective interventional clinical study. Nutrients 15, 529. doi:10.3390/nu15030529
Rimm, E. B., Stampfer, M. J., Ascherio, A., Giovannucci, E., Colditz, G. A., and Willett, W. C. (1993). Vitamin E consumption and the risk of coronary heart disease in men. N. Engl. Journal of Medicine 328, 1450–1456. doi:10.1056/NEJM199305203282004
Ristow, M., and Schmeisser, K. (2014). Mitohormesis: promoting health and lifespan by increased levels of reactive oxygen species (ROS). Dose-response 12, 288–341. doi:10.2203/dose-response.13-035.Ristow
Rogina, B., and Tissenbaum, H. A. (2024). SIRT1, resveratrol and aging. Frontiers in Genetics 15, 1393181. doi:10.3389/fgene.2024.1393181
Ros, M., and Carrascosa, J. M. (2020). Current nutritional and pharmacological anti-aging interventions. Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease 1866, 165612. doi:10.1016/j.bbadis.2019.165612
Ross, A. C. (2012). Vitamin A and retinoic acid in T cell–related immunity. The American journal of clinical nutrition 96, 1166S–1172S. doi:10.3945/ajcn.112.034637
Roth, E. (2007). Immune and cell modulation by amino acids. Clinical nutrition 26, 535–544. doi:10.1016/j.clnu.2007.05.007
Rowland, I., Gibson, G., Heinken, A., Scott, K., Swann, J., Thiele, I., et al. (2018). Gut microbiota functions: metabolism of nutrients and other food components. European journal of nutrition 57, 1–24. doi:10.1007/s00394-017-1445-8
Saari, J. C. (2012). Vitamin A metabolism in rod and cone visual cycles. Annual review of nutrition 32, 125–145. doi:10.1146/annurev-nutr-071811-150748
Safta, D. A., Bogdan, C., and Moldovan, M.-L. (2024). SLNs and NLCs for skin applications: enhancing the bioavailability of natural bioactives. Pharmaceutics 16, 1270. doi:10.3390/pharmaceutics16101270
Salehi, B., Berkay Yılmaz, Y., Antika, G., Boyunegmez Tumer, T., Fawzi Mahomoodally, M., Lobine, D., et al. (2019a). Insights on the use of α-lipoic acid for therapeutic purposes. Biomolecules 9, 356. doi:10.3390/biom9080356
Salehi, B., Venditti, A., Sharifi-Rad, M., Kręgiel, D., Sharifi-Rad, J., Durazzo, A., et al. (2019b). The therapeutic potential of apigenin. International Journal of Molecular Sciences 20, 1305. doi:10.3390/ijms20061305
Sano, M., Ernesto, C., Thomas, R. G., Klauber, M. R., Schafer, K., Grundman, M., et al. (1997). A controlled trial of selegiline, alpha-tocopherol, or both as treatment for Alzheimer's disease. The Alzheimer's Disease Cooperative Study. N. Engl. Journal of Medicine 336, 1216–1222. doi:10.1056/NEJM199704243361704
Santi, C., Tidei, C., Scalera, C., Piroddi, M., and Galli, F. (2013). Selenium containing compounds from poison to drug candidates: a review on the GPx-like activity. Current Chemical Biology 7, 25–36. doi:10.2174/2212796811307010003
Sathyabhama, M., Priya Dharshini, L. C., Karthikeyan, A., Kalaiselvi, S., and Min, T. (2022). The credible role of curcumin in oxidative stress-mediated mitochondrial dysfunction in mammals. Biomolecules 12, 1405. doi:10.3390/biom12101405
Senanayake, S. N. (2013). Green tea extract: chemistry, antioxidant properties and food applications–A review. Journal of functional foods 5, 1529–1541. doi:10.1016/j.jff.2013.08.011
Serban, M. C., Sahebkar, A., Zanchetti, A., Mikhailidis, D. P., Howard, G., Antal, D., et al. (2016). Effects of quercetin on blood pressure: a systematic review and meta-analysis of randomized controlled trials. Journal of the American Heart Association 5, e002713. doi:10.1161/JAHA.115.002713
Serhan, C. N., and Levy, B. D. (2018). Resolvins in inflammation: emergence of the pro-resolving superfamily of mediators. The Journal of clinical investigation 128, 2657–2669. doi:10.1172/JCI97943
Shahbaz, M., Imran, M., Alsagaby, S. A., Naeem, H., Al Abdulmonem, W., Hussain, M., et al. (2023). Anticancer, antioxidant, ameliorative and therapeutic properties of kaempferol. International Journal of Food Properties 26, 1140–1166. doi:10.1080/10942912.2023.2205040
Shankar, A. H., and Prasad, A. S. (1998). Zinc and immune function: the biological basis of altered resistance to infection. The American journal of clinical nutrition 68, 447S–463S. doi:10.1093/ajcn/68.2.447S
Sharma, P., Sharma, S., and Singh, D. (2020). Apigenin reverses behavioural impairments and cognitive decline in kindled mice via CREB-BDNF upregulation in the hippocampus. Nutritional Neuroscience 23, 118–127. doi:10.1080/1028415X.2018.1478653
Shay, K. P., Moreau, R. F., Smith, E. J., Smith, A. R., and Hagen, T. M. (2009). Alpha-lipoic acid as a dietary supplement: molecular mechanisms and therapeutic potential. Biochimica et Biophysica Acta (BBA)-General Subjects 1790, 1149–1160. doi:10.1016/j.bbagen.2009.07.026
Shehzad, A., Ha, T., Subhan, F., and Lee, Y. S. (2011). New mechanisms and the anti-inflammatory role of curcumin in obesity and obesity-related metabolic diseases. European journal of nutrition 50, 151–161. doi:10.1007/s00394-011-0188-1
Shen, Q. W., Zhu, M. J., Tong, J., Ren, J., and Du, M. (2007). Ca2+/calmodulin-dependent protein kinase kinase is involved in AMP-activated protein kinase activation by α-lipoic acid in C2C12 myotubes. American Journal of Physiology-Cell Physiology 293, C1395–C1403. doi:10.1152/ajpcell.00115.2007
Shin, M.-S., Lee, Y. J., Cho, I.-H., and Yang, H.-J. (2024). Brain plasticity and ginseng. Journal of Ginseng Research 48, 286–297. doi:10.1016/j.jgr.2024.03.007
Shioi, A., Morioka, T., Shoji, T., and Emoto, M. (2020). The inhibitory roles of vitamin K in progression of vascular calcification. Nutrients 12, 583. doi:10.3390/nu12020583
Shults, C. W., Oakes, D., Kieburtz, K., Beal, M. F., Haas, R., Plumb, S., et al. (2002). Effects of coenzyme Q10 in early Parkinson disease: evidence of slowing of the functional decline. Archives of neurology 59, 1541–1550. doi:10.1001/archneur.59.10.1541
Sikora, E., Scapagnini, G., and Barbagallo, M. (2010). Curcumin, inflammation, ageing and age-related diseases. Immunity and Ageing 7, 1–4. doi:10.1186/1742-4933-7-1
Silva Dos Santos, J., Goncalves Cirino, J. P., De Oliveira Carvalho, P., and Ortega, M. M. (2021). The pharmacological action of kaempferol in central nervous system diseases: a review. Frontiers in Pharmacology 11, 565700. doi:10.3389/fphar.2020.565700
Silvestro, S., Sindona, C., Bramanti, P., and Mazzon, E. (2021). A state of the art of antioxidant properties of curcuminoids in neurodegenerative diseases. International Journal of Molecular Sciences 22, 3168. doi:10.3390/ijms22063168
Simopoulos, A. P. (2008). The importance of the omega-6/omega-3 fatty acid ratio in cardiovascular disease and other chronic diseases. Experimental biology and medicine 233, 674–688. doi:10.3181/0711-MR-311
Singh, C. K., Ndiaye, M. A., and Ahmad, N. (2015). Resveratrol and cancer: challenges for clinical translation. Biochimica et Biophysica Acta (BBA)-Molecular Basis of Disease 1852, 1178–1185. doi:10.1016/j.bbadis.2014.11.004
Singh, M., Devi, S., Rana, V. S., Mishra, B. B., Kumar, J., and Ahluwalia, V. (2019). Delivery of phytochemicals by liposome cargos: recent progress, challenges and opportunities. Journal of microencapsulation 36, 215–235. doi:10.1080/02652048.2019.1617361
Singh, V., Jain, S., Prakash, S., and Thakur, M. (2022). Studies on the synergistic interplay of vitamin D and K for improving bone and cardiovascular health. Current Research in Nutrition and Food Science Journal 10, 840–857. doi:10.12944/crnfsj.10.3.3
Soetikno, V., Sari, F. R., Lakshmanan, A. P., Arumugam, S., Harima, M., Suzuki, K., et al. (2013). Curcumin alleviates oxidative stress, inflammation, and renal fibrosis in remnant kidney through the N rf2–keap1 pathway. Molecular nutrition and food research 57, 1649–1659. doi:10.1002/mnfr.201200540
Sommer, A. (2008). Vitamin A deficiency and clinical disease: an historical overview. The Journal of nutrition 138, 1835–1839. doi:10.1093/jn/138.10.1835
Song, X., Gao, W., Shi, Y., Li, J., and Zheng, Z. (2023). Panax ginseng and its derivatives: promoting angiogenesis in ischemic diseases–a mechanistic overview. Journal of Functional Foods 109, 105762. doi:10.1016/j.jff.2023.105762
Sorkin, B. C., Kuszak, A. J., Bloss, G., Fukagawa, N. K., Hoffman, F. A., Jafari, M., et al. (2020). Improving natural product research translation: from source to clinical trial. The FASEB Journal 34, 41–65. doi:10.1096/fj.201902143R
Sorkin, B. C., Kuszak, A. J., Williamson, J. S., Hopp, D. C., and Betz, J. M. (2016). The challenge of reproducibility and accuracy in nutrition research: resources and pitfalls. Advances in Nutrition 7, 383–389. doi:10.3945/an.115.010595
Southern, J., Pitt-Francis, J., Whiteley, J., Stokeley, D., Kobashi, H., Nobes, R., et al. (2008). Multi-scale computational modelling in biology and physiology. Progress in biophysics and molecular biology 96, 60–89. doi:10.1016/j.pbiomolbio.2007.07.019
Sova, M., and Saso, L. (2020). Natural sources, pharmacokinetics, biological activities and health benefits of hydroxycinnamic acids and their metabolites. Nutrients 12, 2190. doi:10.3390/nu12082190
Stephensen, C. B. (2001). Vitamin A, infection, and immune function. Annual review of nutrition 21, 167–192. doi:10.1146/annurev.nutr.21.1.167
Superti, F., and Russo, R. (2024). Alpha-Lipoic acid: biological mechanisms and health benefits. Antioxidants 13, 1228. doi:10.3390/antiox13101228
Suzuki, M., Yoshioka, M., Hashimoto, M., Murakami, M., Noya, M., Takahashi, D., et al. (2013). Randomized, double-blind, placebo-controlled trial of vitamin D supplementation in Parkinson disease. The American journal of clinical nutrition 97, 1004–1013. doi:10.3945/ajcn.112.051664
Swanson, D., Block, R., and Mousa, S. A. (2012). Omega-3 fatty acids EPA and DHA: health benefits throughout life. Advances in nutrition 3, 1–7. doi:10.3945/an.111.000893
Tanumihardjo, S. A. (2011). Vitamin A: biomarkers of nutrition for development. The American journal of clinical nutrition 94, 658S–665S. doi:10.3945/ajcn.110.005777
Targonsky, E., Dai, F., Koshkin, V., Karaman, G., Gyulkhandanyan, A., Zhang, Y., et al. (2006). alpha-lipoic acid regulates AMP-activated protein kinase and inhibits insulin secretion from beta cells. Diabetologia 49, 1587–1598. doi:10.1007/s00125-006-0265-9
Tibullo, D., Li Volti, G., Giallongo, C., Grasso, S., Tomassoni, D., Anfuso, C. D., et al. (2017). Biochemical and clinical relevance of alpha lipoic acid: antioxidant and anti-inflammatory activity, molecular pathways and therapeutic potential. Inflammation Research 66, 947–959. doi:10.1007/s00011-017-1079-6
Topol, E. J. (2019). High-performance medicine: the convergence of human and artificial intelligence. Nature medicine 25, 44–56. doi:10.1038/s41591-018-0300-7
Traber, M. G., and Atkinson, J. (2007). Vitamin E, antioxidant and nothing more. Free radical biology and medicine 43, 4–15. doi:10.1016/j.freeradbiomed.2007.03.024
Traber, M. G., and Packer, L. (1995). Vitamin E: beyond antioxidant function. The American journal of clinical nutrition 62, 1501S–1509S. doi:10.1093/ajcn/62.6.1501S
Traber, M. G., and Stevens, J. F. (2011). Vitamins C and E: beneficial effects from a mechanistic perspective. Free radical biology and medicine 51, 1000–1013. doi:10.1016/j.freeradbiomed.2011.05.017
Trothen, T. J. (2024). “Anti-aging or enhancing-aging technologies? social and religious implications of radical life extension,” in An interdisciplinary approach to aging, biohacking and technology (Routledge), 44–62.
Truong, V. L., Jun, M., and Jeong, W. S. (2018). Role of resveratrol in regulation of cellular defense systems against oxidative stress. Biofactors 44, 36–49. doi:10.1002/biof.1399
Turunen, M., Olsson, J., and Dallner, G. (2004). Metabolism and function of coenzyme Q. Biochimica et Biophysica Acta (BBA)-Biomembranes 1660, 171–199. doi:10.1016/j.bbamem.2003.11.012
Valverde-Salazar, V., Ruiz-Gabarre, D., and García-Escudero, V. (2023). Alzheimer’s disease and green tea: epigallocatechin-3-gallate as a modulator of inflammation and oxidative stress. Antioxidants 12, 1460. doi:10.3390/antiox12071460
Vashishat, A., Patel, P., Das Gupta, G., and Das Kurmi, B. (2024). Alternatives of animal models for biomedical research: a comprehensive review of modern approaches. Stem Cell Reviews and Reports 20, 881–899. doi:10.1007/s12015-024-10701-x
Vasto, S., Mocchegiani, E., Candore, G., Listì, F., Colonna-Romano, G., Lio, D., et al. (2006). Inflammation, genes and zinc in ageing and age-related diseases. Biogerontology 7, 315–327. doi:10.1007/s10522-006-9046-6
Vayena, E., Blasimme, A., and Cohen, I. G. (2018). Machine learning in medicine: addressing ethical challenges. PLoS medicine 15, e1002689. doi:10.1371/journal.pmed.1002689
Veeresham, C. (2012). Natural products derived from plants as a source of drugs. Journal of advanced pharmaceutical technology and research 3, 200–201. doi:10.4103/2231-4040.104709
Venegas-Calerón, M., Sayanova, O., and Napier, J. A. (2010). An alternative to fish oils: metabolic engineering of oil-seed crops to produce omega-3 long chain polyunsaturated fatty acids. Progress in lipid research 49, 108–119. doi:10.1016/j.plipres.2009.10.001
Voulgaropoulou, S. D., Van Amelsvoort, T., Prickaerts, J., and Vingerhoets, C. (2019). The effect of curcumin on cognition in Alzheimer’s disease and healthy aging: a systematic review of pre-clinical and clinical studies. Brain research 1725, 146476. doi:10.1016/j.brainres.2019.146476
Vyas, P., Singh, D., Singh, N., Kumar, V., and Dhaliwal, H. S. (2018). Nutrigenomics: advances, opportunities and challenges in understanding the nutrient-gene interactions. Current Nutrition and Food Science 14, 104–115. doi:10.2174/1573401313666170614094410
Wan, R., Cai, R., Hu, H., Wu, J., Jiang, X., Li, L., et al. (2024). Identification of active anti-aging ingredients from Asparagus cochinchinensis merill based on the spectrum-effect relationship. Preprint. SSRN
Wang, D.-S., Wang, J.-M., Zhang, F.-R., Lei, F.-J., Wen, X., Song, J., et al. (2022). Ameliorative effects of malonyl ginsenoside from Panax ginseng on glucose-lipid metabolism and insulin resistance via IRS1/PI3K/Akt and AMPK signaling pathways in type 2 diabetic mice. The American Journal of Chinese Medicine 50, 863–882. doi:10.1142/S0192415X22500367
Wang, T.-T., Nestel, F. P., Bourdeau, V., Nagai, Y., Wang, Q., Liao, J., et al. (2004). Cutting edge: 1, 25-dihydroxyvitamin D3 is a direct inducer of antimicrobial peptide gene expression. The Journal of Immunology 173, 2909–2912. doi:10.4049/jimmunol.173.5.2909
Wang, Z., Sun, W., Sun, X., Wang, Y., and Zhou, M. (2020). Kaempferol ameliorates Cisplatin induced nephrotoxicity by modulating oxidative stress, inflammation and apoptosis via ERK and NF-κB pathways. Amb Express 10, 58–11. doi:10.1186/s13568-020-00993-w
Weaver, K., and Skouta, R. (2022). The selenoprotein glutathione peroxidase 4: from molecular mechanisms to novel therapeutic opportunities. Biomedicines 10, 891. doi:10.3390/biomedicines10040891
Welch, R. W., Antoine, J.-M., Berta, J.-L., Bub, A., De Vries, J., Guarner, F., et al. (2011). Guidelines for the design, conduct and reporting of human intervention studies to evaluate the health benefits of foods. British Journal of Nutrition 106, S3–S15. doi:10.1017/S0007114511003606
Wen, T., Song, L., and Hua, S. (2021). Perspectives and controversies regarding the use of natural products for the treatment of lung cancer. Cancer Medicine 10, 2396–2422. doi:10.1002/cam4.3660
Wimalawansa, S. J. (2025). Enhancing the design of nutrient clinical trials for disease prevention—a focus on vitamin D: a systematic review. Nutrition Reviews, nuae164. doi:10.1093/nutrit/nuae164
Wishart, D. S. (2016). Emerging applications of metabolomics in drug discovery and precision medicine. Nature reviews Drug discovery 15, 473–484. doi:10.1038/nrd.2016.32
Wong, C. P., Magnusson, K. R., Sharpton, T. J., and Ho, E. (2021). Effects of zinc status on age-related T cell dysfunction and chronic inflammation. Biometals 34, 291–301. doi:10.1007/s10534-020-00279-5
Wu, D., and Meydani, S. N. (2008). Age-associated changes in immune and inflammatory responses: impact of vitamin E intervention. Journal of Leucocyte Biology 84, 900–914. doi:10.1189/jlb.0108023
Wu, F., Zhou, Y., Li, L., Shen, X., Chen, G., Wang, X., et al. (2020). Computational approaches in preclinical studies on drug discovery and development. Frontiers in chemistry 8, 726. doi:10.3389/fchem.2020.00726
Wu, F.Y.-H., and Wu, C.-W. (2023). The role of zinc in DNA and RNA polymerases. Metal ions in biological systems, 157–192. doi:10.1201/9781003418092-4
Wu, P., Meng, X., Zheng, H., Zeng, Q., Chen, T., Wang, W., et al. (2018). Kaempferol attenuates ROS-induced hemolysis and the molecular mechanism of its induction of apoptosis on bladder cancer. Molecules 23, 2592. doi:10.3390/molecules23102592
Wu, W., Mi, Y., Meng, Q., Li, N., Li, W., Wang, P., et al. (2024). Natural polyphenols as novel interventions for aging and age-related diseases: exploring efficacy, mechanisms of action and implications for future research. Chinese Herbal Medicines 17, 279–291. doi:10.1016/j.chmed.2024.09.001
Xia, N., Förstermann, U., and Li, H. (2014). Resveratrol and endothelial nitric oxide. Molecules 19, 16102–16121. doi:10.3390/molecules191016102
Yadav, N., Singh Chandel, S., Venkatachalam, T., and Fathima, S. N. (2024). “Herbal Medicine Formulation, standardization, and Commercialization challenges and sustainable strategies for improvement,” in Herbal medicine phytochemistry: applications and trends (Springer), 1769–1795.
Yagishita, Y., Gatbonton-Schwager, T. N., Mccallum, M. L., and Kensler, T. W. (2020). Current landscape of NRF2 biomarkers in clinical trials. Antioxidants 9, 716. doi:10.3390/antiox9080716
Yeh, P.-T., Huang, H.-W., Yang, C.-M., Yang, W.-S., and Yang, C.-H. (2016). Astaxanthin inhibits expression of retinal oxidative stress and inflammatory mediators in streptozotocin-induced diabetic rats. PLoS One 11, e0146438. doi:10.1371/journal.pone.0146438
Yin, K., and Agrawal, D. K. (2014). Vitamin D and inflammatory diseases. Journal of inflammation research 7, 69–87. doi:10.2147/JIR.S63898
Young, A. J., Johnson, S., Steffens, D. C., and Doraiswamy, P. M. (2007). Coenzyme Q10: a review of its promise as a neuroprotectant. CNS spectrums 12, 62–68. doi:10.1017/s1092852900020538
Yuan, H., Ma, Q., Ye, L., and Piao, G. (2016). The traditional medicine and modern medicine from natural products. Molecules 21, 559. doi:10.3390/molecules21050559
Yuan, J., Liu, R., Ma, Y., Zhang, Z., and Xie, Z. (2018). Curcumin attenuates airway inflammation and airway remolding by inhibiting NF-κB signaling and COX-2 in cigarette smoke-induced COPD mice. Inflammation 41, 1804–1814. doi:10.1007/s10753-018-0823-6
Yuan, J. P., Peng, J., Yin, K., and Wang, J. H. (2011). Potential health-promoting effects of astaxanthin: a high-value carotenoid mostly from microalgae. Molecular nutrition and food research 55, 150–165. doi:10.1002/mnfr.201000414
Zeng, M., Guo, D., Fernández-Varo, G., Zhang, X., Fu, S., Ju, S., et al. (2022). The integration of nanomedicine with traditional Chinese medicine: drug delivery of natural products and other opportunities. Molecular pharmaceutics 20, 886–904. doi:10.1021/acs.molpharmaceut.2c00882
Zgheel, F., Alhosin, M., Rashid, S., Burban, M., Auger, C., and Schini-Kerth, V. B. (2014). Redox-sensitive induction of Src/PI3-kinase/Akt and MAPKs pathways activate eNOS in response to EPA: DHA 6: 1. PloS one 9, e105102. doi:10.1371/journal.pone.0105102
Zhang, B., and Radisic, M. (2017). Organ-on-a-chip devices advance to market. Lab on a Chip 17, 2395–2420. doi:10.1039/c6lc01554a
Zhang, Y., Du, Z., Zhou, Q., Wang, Y., and Li, J. (2014). Remifentanil attenuates lipopolysaccharide-induced acute lung injury by downregulating the NF-κB signaling pathway. Inflammation 37, 1654–1660. doi:10.1007/s10753-014-9893-2
Zhang, Y., Huang, X., Liu, N., Liu, M., Sun, C., Qi, B., et al. (2022). Discovering the potential value of coenzyme Q10 in oxidative stress: enlightenment from a synthesis of clinical evidence based on various population. Frontiers in Pharmacology 13, 936233. doi:10.3389/fphar.2022.936233
Zhang, Z., Liu, H., Jin, J., Zhu, X., Han, D., and Xie, S. (2024). Towards a low-carbon footprint: current status and prospects for aquaculture. Water Biology and Security 3, 100290. doi:10.1016/j.watbs.2024.100290
Zhang, Z.-H., and Song, G.-L. (2021). Roles of selenoproteins in brain function and the potential mechanism of selenium in Alzheimer’s disease. Frontiers in Neuroscience 15, 646518. doi:10.3389/fnins.2021.646518
Zhao, L., Wang, J.-L., Liu, R., Li, X.-X., Li, J.-F., and Zhang, L. (2013). Neuroprotective, anti-amyloidogenic and neurotrophic effects of apigenin in an Alzheimer’s disease mouse model. Molecules 18, 9949–9965. doi:10.3390/molecules18089949
Zhou, J., Yang, Z., Shen, R., Zhong, W., Zheng, H., Chen, Z., et al. (2021). Resveratrol improves mitochondrial biogenesis function and activates PGC-1α pathway in a preclinical model of early brain injury following subarachnoid hemorrhage. Frontiers in molecular biosciences 8, 620683. doi:10.3389/fmolb.2021.620683
Zhou, X., Seto, S. W., Chang, D., Kiat, H., Razmovski-Naumovski, V., Chan, K., et al. (2016a). Synergistic effects of Chinese herbal medicine: a comprehensive review of methodology and current research. Frontiers in pharmacology 7, 201. doi:10.3389/fphar.2016.00201
Zhou, Y., Wang, J., Gu, Z., Wang, S., Zhu, W., AceñA, J. L., et al. (2016b). Next generation of fluorine-containing pharmaceuticals, compounds currently in phase II–III clinical trials of major pharmaceutical companies: new structural trends and therapeutic areas. Chemical reviews 116, 422–518. doi:10.1021/acs.chemrev.5b00392
Ziegelmann, M. J., Trost, L. W., Russo, G. I., and Levine, L. A. (2020). Peyronie's disease intervention studies: an exploration of modern-era challenges in study design and evaluating treatment outcomes. The Journal of Sexual Medicine 17, 364–377. doi:10.1016/j.jsxm.2019.11.271
Ziegler, O., Sirveaux, M.-A., Brunaud, L., Reibel, N., and Quilliot, D. (2009). Medical follow up after bariatric surgery: nutritional and drug issues General recommendations for the prevention and treatment of nutritional deficiencies. Diabetes and metabolism 35, 544–557. doi:10.1016/S1262-3636(09)73464-0
Keywords: aging, nutrients, natural products, pharmaceuticals, healthspan
Citation: Ding F, Yu Y, Zhang Y, Wei S, Han JH, Li Z, Jiang H-B, Ryu D, Park W, Ha K-T and Geng L (2025) Harnessing nutrients and natural products for sustainable drug development against aging. Front. Pharmacol. 16:1579266. doi: 10.3389/fphar.2025.1579266
Received: 19 February 2025; Accepted: 04 April 2025;
Published: 28 April 2025.
Edited by:
Maurizio Ragni, University of Milan, ItalyReviewed by:
Loredana Bergandi, University of Turin, ItalyElisabetta Caiazzo, University of Naples Federico II, Italy
Copyright © 2025 Ding, Yu, Zhang, Wei, Han, Li, Jiang, Ryu, Park, Ha and Geng. This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
*Correspondence: Wonyoung Park, amlubGluZzAxMjJAcHVzYW4uYWMua3I=; Ki-Tae Ha, aGFnaXNAcHVzYW4uYWMua3I=; Li Geng, Z2VuZ2xpQGpsdS5lZHUuY24=