Next Article in Journal
How Does Our Brain Process Sugars and Non-Nutritive Sweeteners Differently: A Systematic Review on Functional Magnetic Resonance Imaging Studies
Next Article in Special Issue
Dietary Protein Intake Patterns and Inadequate Protein Intake in Older Adults from Four Countries
Previous Article in Journal
Effects of the Human Gut Microbiota on Cognitive Performance, Brain Structure and Function: A Narrative Review
Previous Article in Special Issue
Metabolic Defects Caused by High-Fat Diet Modify Disease Risk through Inflammatory and Amyloidogenic Pathways in a Mouse Model of Alzheimer’s Disease
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Plant Fortification of the Diet for Anti-Ageing Effects: A Review

1
School of Bioengineering and Biosciences, Lovely Professional University, Phagwara 144411, Punjab, India
2
School of Bioengineering and Food Technology, Shoolini University of Biotechnology and Management Sciences, Solan 173229, Himachal Pradesh, India
3
School of Biological and Environmental Sciences, Shoolini University of Biotechnology and Management Sciences, Solan 173229, Himachal Pradesh, India
4
Department of Chemistry, Faculty of Science, University of Hradec Kralove, 50003 Hradec Kralove, Czech Republic
*
Authors to whom correspondence should be addressed.
These authors contributed equally to this work.
Submission received: 31 August 2020 / Revised: 27 September 2020 / Accepted: 29 September 2020 / Published: 30 September 2020
(This article belongs to the Special Issue Nutrition, Diet Quality, Aging and Frailty)

Abstract

:
Ageing is an enigmatic and progressive biological process which undermines the normal functions of living organisms with time. Ageing has been conspicuously linked to dietary habits, whereby dietary restrictions and antioxidants play a substantial role in slowing the ageing process. Oxygen is an essential molecule that sustains human life on earth and is involved in the synthesis of reactive oxygen species (ROS) that pose certain health complications. The ROS are believed to be a significant factor in the progression of ageing. A robust lifestyle and healthy food, containing dietary antioxidants, are essential for improving the overall livelihood and decelerating the ageing process. Dietary antioxidants such as adaptogens, anthocyanins, vitamins A/D/C/E and isoflavones slow the ageing phenomena by reducing ROS production in the cells, thereby improving the life span of living organisms. This review highlights the manifestations of ageing, theories associated with ageing and the importance of diet management in ageing. It also discusses the available functional foods as well as nutraceuticals with anti-ageing potential.

1. Introduction

Ageing is a progressive biological process which affects the normal functions of cells and tissue, thereby imperiling the person towards diseases and mortality [1]. For a layman, it is the process of maturing and growing old. Both internal and external factors play an integral role in ageing [2]. Internal factors comprise the usual biological processes of the cell, whereas the external factors involve chronic sun-exposure, hormonal imbalance, nutritional deficiencies, ultraviolet (UV) irradiation and other factors such as pollution and smoking [3]. The hallmarks associated with ageing have been illustrated in Figure 1. Skin ageing, characterized by wrinkling, can be reduced via suitable preventive measures involving the consumption of antioxidant-rich supplements, a balanced diet and undertaking skincare [4]. By opting for these measures, the harmful effects induced by free radicals can be restrained [5].
Over the past few decades, the relationship between nutrition and ageing has been extensively studied in both animals and humans [6]. Nutraceuticals are nutritional elements with medicinal characteristics; hence the name, where “Nutra” stands for food and “ceutical” means therapeutic properties [7]. As per the definition of Foundation for Innovation in Medicine (FIM), nutraceuticals are the “food and food products” that have medicinal value and provide health benefits, especially in preventing and treating age-related diseases [8]. These products include functional foods, dietary supplements and herbal extracts, which provide health benefits in the long-run when consumed as supplements in the diet [9]. Even researchers have suggested that antioxidants have propitious effects on both chronic as well as age-related diseases, especially neurodegenerative diseases and cancer [10]. Various food supplements that exhibit an antioxidant potential, such as carotenoids, flavonoids and vitamins, prevent and treat ROS-associated chronic conditions, which results in healthier and longer lifespans [10]. Food supplements produce antagonistic effects against the degenerative and inflammatory processes in the body, and have beneficial effects on the immune and digestive system, hence improving the quality of life [11].
The current review focusses on highlighting the manifestations of ageing and theories associated with ageing. Additionally, it also discusses the importance of diet management in ageing and functional food, as well as nutraceuticals with anti-ageing potential.

2. Manifestation of Ageing

Clinical manifestations of intrinsic ageing can be determined by assessing the regenerative ability of the damaged tissues or organs [12]. All dividing and differentiating cells are vulnerable to insults causing intrinsic ageing [13]. The visual traits of ageing start appearing in the early 40s. Most cells, tissues and organs steadily undergo ageing and become incompetent [14]. A significant effect can be observed on the skin, which turns loose, thin wrinkled and inelastic [15]. The face fat also reduces, leading to hollowed eye sockets and cheeks.
Furthermore, the hair starts thinning from the armpits, pubic area and scalp [16]. As melanin content decreases, the hair strands become thinner grey, and the nails become thinner as well [17]. At over 80 years old, more noticeable visual changes can be observed, such as the compression of spinal disks, vertebrae and joints. The hearing abilities also diminish depending on the severity of the ageing phenomenon [18].
Other than this, the elderly population gets presbyopia and may require reading glasses [19]. In comparison to healthy adults, they lack deep sleep and are unable to take sufficient rest as required by the body at this stage [20]. The bone density decreases and becomes weaker, increasing the risk of fracture [21]. Due to slow metabolism and hormonal changes, there is a reduction in muscle mass and an increase in body fat [22]. Besides this, older adults also suffer from lapses of memory and vagueness, preventing them from recalling names and memories [23]. The heart and lungs become less efficient with time, and kidney functions are abated [24]. The accumulated harmful metabolic waste later appears as dangerous diseases and allergies, causing significant discomfort to older people [25]. Moreover, females at menopause produce reduced amounts of estrogen, due to which they experience various changes, such as vaginal dryness, hot flashes, chills, night sweats, sleeping problems, mood swings, weight gain and slowed metabolism [26]. Besides this, an unhealthy diet and indolent lifestyle further increase the risk of occurrence of chronic diseases in elderly people, such as cancer, osteoarthritis, type 2 diabetes, obesity, coronary artery disease osteoporosis and high blood pressure [27].

3. Theories of Ageing

Several theories have been formulated to define the ageing phenomenon. These theories have been postulated based on certain assumptions, but none of them provide a satisfactory explanation [28]. There are three major theories for ageing, i.e., genetic theories, dysfunction of interlinked organs and physiological approaches [29]. Of these, three physiological theories have been extensively studied, which comprise the cross-linking theory, the waste material accumulation theory and the free radical theory [30].
In 1950, Denham Harman stated that ageing is the result of the massive production of free radicals [31]. In general, free radicals are those atoms or molecules that have unpaired electrons and possess the ability to form electronic couples [32]. This explains the short life and high reactiveness of these molecules. These free radicals are usually formed during the metabolic reactions under normal conditions [33]. Moreover, the generation of these free radicals also takes place during exposure to cigarette smoke, UV rays and toxic substances, as well as during emotional stress [34]. Even though free radicals are involved in normal metabolic processes, but they do not generally infiltrate the cells. Still, when they do, they have harmful and deleterious effects on various organs [35].
Free radicals released from food are essential for energy production within the cell [36]. Additionally, their production also protects the body from opportunistic infections and elicits the synthesis of hormones involved in effective communication within the body [37]. However, the excessive production of free radicals has detrimental effects on DNA, collagen, elastin and blood vessels [38]. Oxidative damage to different biomolecules, such as DNA, macromolecules and proteins, takes place over time [39]. It is considered a significant factor, but is not the only factor responsible for ageing [40]. Fundamentally, oxygen has a dual role in our body, i.e., it is necessary for life and is one of the chief components of harmful compounds like free radicals [41]. Free radicals are generated by the aerobic metabolism. They liberate different types of reactive oxygen species, such as singlet oxygen (1[O2]), superoxide anion radicals (O2), hydroxyl radicals (OH), hydroperoxyl radicals (HO2), peroxide radicals (R = lipid) (ROO) and hydrogen peroxide (H2O2) [42]. The various sources involved in the generation of free radicals are illustrated in Figure 2.
For example, if the free-radical-mediated DNA mutations are left uncorrected via repair mechanisms, this defect persists even after successive replication cycles, transcription and translation [43]. It is well-known that free radicals are formed by the aerobic metabolism for the synthesis of energy-rich molecules like ATP, which are synthesized in mitochondria (also known as cell factories) [44]. As humans start ageing, the efficacy of mitochondria in synthesizing ATP substantially decreases, thereby allowing the accumulation of free radicals in mitochondria as well as permitting the passage of free radicals through the mitochondrial membrane, thereby damaging other parts of the cell [45]. These alterations have helped to determine the key factors which favor ageing, i.e., increases in oxidative stress and a decrease in energy production [46]. Even the published literature has stated that a high degree of mutation is observed in mitochondrial DNA in contrast to nuclear DNA due to oxidative stress [47]. Therefore, calorie restriction (CR) impedes the process of ageing and increases the lifespans of flies, fish, spiders and mammals (mice and rats) [48]. This happens because CR decreases the oxidative load, which reduces the free radical formation in mitochondria [49]. The reduction in the free radical formation substantially reduces the number of oxidized proteins, lipids and mutated mitochondrial DNA [50]. Extensive studies have been conducted on rodent models to assess the effects of a diet enriched with minerals and vitamins in ageing [44]. As such, it is believed that calorie restriction and the consumption of food rich in antioxidants can considerably prolong the life span of individuals [51].
An important theory that explains the process of ageing is the shortening of the telomeres. Due to the end-replication problem, the telomeres are shortened in every generation of the cell till they reach a critical length in the crisis stage of ageing [52]. At this stage, the cell division slows down considerably, causing the cell to slowly die. This may be referred to as “replicative mortality”. Cells involved in growth, development and reproduction express high levels of the enzyme telomerase, which maintains the length of the telomeric DNA [53]. These cells include the stem cells and reproductive cells (eggs and sperms). However, most adult cells have low expressions or no expression of telomerase, which causes these cells to age and eventually die [54].

4. Plant-Based Supplements with Anti-Ageing Potential

Plants and their inherent components are well known to exhibit antioxidant potentials, sch as carotenoids, flavonoids and vitamins, that aid in the prevention and treatment of ROS-associated chronic conditions [55]. These supplements have antagonistic effects against the degenerative and inflammatory processes in the body and show beneficial effects on the immune and digestive system, hence improving the quality of life [56]. Some of the predominantly used plant-based supplements have been discussed below.

4.1. Adaptogens

Adaptogens are compounds obtained from herbal plants for maintaining homeostasis and stabilizing the physiological processes in humans [57]. These compounds reduce cellular sensitivity to stress and improve the ability of the body to resist the damage from other risk factors [58]. Moreover, they also help in restoring and promoting normal physiological function [59]. A few of the highly known adaptogens have been discussed below.

4.2. Bacopa monnieri

Bacopa monnieri, also known as Brahmi, is a perennial herb with small oblong leaves and purple flowers [60]. Highly valuable nootropic phytochemicals, such as bacosides, are found in this medicinal herb [61]. Brahmine and Herpestine are the two essential phytochemicals that are predominantly extracted from this herb [62]. The phytochemicals obtained from Brahmi aid in protecting the brain from the attack of free radicals and stimulating cognitive functioning and learning [63]. It has been comprehended that the regular consumption of Brahmi oil reduces the chance of various diseases like Alzheimer’s disease and amnesia [64]. Bhattacharya et al. (2000) found that extracts of Bacopa monnieri enhance the activity of reactive oxygen species-scavenging enzyme catalase (CAT), glutathione peroxidase (GPX) and superoxide dismutase (SOD), in a dose-dependent manner. This study was carried out in the brain regions of rats and investigated after 14 and 21 days [65]. Shinomol and colleagues conducted an in vitro and in vivo study using 3-nitropropionic acid (NPA) (fungal toxin responsible for causing neurotoxicity in humans and animals) and Bacopa monnieri extract. The result obtained showed that NPA was effective in inducing the oxidative stress in dopaminergic (N27) cells and mitochondria of the striatum of rats, whereas Bacopa monnieri extract was found to be effective in regulating the NPA-induced oxidative reactions and reducing the Glutathione (GSH) and thiol levels [66]. Kumar and his colleagues also conducted a six-week randomized placebo-controlled trial to assess the effect of Bacopa monnieri extract on the cognitive functions of students studying medicine. The result obtained from the study showed significant improvement in the cognitive functioning of the students [67].

4.3. Curcuma longa

Curcuma longa is a plant of the ginger family that produces a compound known as curcumin [68]. It is known for diverse biological activities, such as its anti-cancerous, anti-inflammatory and antioxidant properties [69]. Due to these natural properties, curcumin is a potential therapeutic agent for treating different types of cancers [70]. Many studies have revealed that curcumin can suppress the expression or activity of cyclooxygenase-2 (COX-2), prostaglandin E2 (PGE2), pro-inflammatory cytokines and tumor necrosis factor-α (TNF-α) [71]. The antioxidant properties of curcumin can aid in the reduction of ROS production, the scavenging of free oxygen radicals and obstructing lipid peroxidation [72]. The consumption of curcumin via the oral route in rodents has been shown to ameliorate cystic fibrosis and block tumor progression; still, the evaluation of humans is pending [70]. A study reported that curcumin induces a cellular stress response in human fibroblasts via redox signaling and the phosphatidylinositol 3-kinase/Akt (Protein Kinase B; PKB) pathway. This provides evidence that curcumin-triggered cellular antioxidant defenses can serve as an effective approach to anti-ageing intervention [73].
Moreover, it has been reported to increase the life span of fruit flies, mice and nematodes [74,75,76]. In fact, curcumin has been stated to improve and regulate the symptoms of age-related diseases such as atherosclerosis, cancer and diabetes [77,78]. Other than this, curcumin has been reported to show protective activity against chemotherapy-induced side effects and radiation-induced dermatitis in breast cancer patients [79,80]. Some studies have claimed that curcumin has anti-ageing potential because it can delay cellular senescence [81]. Cox et al. conducted a study to assess the effects of solid lipid curcumin on mood and cognition in healthy adults aged 60–85. In this study, subjects were examined for the effects of solid lipid curcumin formulation, i.e., 400 mg of Longvida® for acute (1 and 3 h after a single dose), chronic (4 weeks) and acute-on-chronic (1 and 3 h after a single dose following regular treatment) dosing. The results obtained showed significant improvements in the working memory for both acute and chronic dosing. Additionally, it also decreased physical fatigue (measured per Chalder Fatigue Scale) as well as total and LDL cholesterol [82].

4.4. Emblica officinalis

Emblica officinalis, also known as Amla, is a member of the Phyllanthaceae family [83]. The churn of Amla is known for reducing cholesterol level and improving memory potential [84]. The consumption of Amla in the diet is effective in lowering the cholesterol level in the brain as well as in the body [85]. It has also been stated as a beneficial functional food for treating Alzheimer’s disease [86]. Draelos and colleagues conducted a double-blind study to evaluate the skin-lightening potential of a topical formulation comprising E. officinalis extract, glycolic acid and kojic acid. The study revealed that the topical formulation was 4% better than hydroquinone, due to which researchers claimed that the topical formulation could be an effective natural alternative for mild to moderate facial dyschromia [87]. Accumulation of free radicals in different tissues is associated with various stress-induced conditions leading to the progression of the process of ageing [88]. Tannoids obtained from E. officinalis also show a protective effect because of their antioxidant potential against the tardive dyskinesia rat model [89]. Moreover, the extract of E. officinalis shows antidepressant properties by inhibiting the activity of Gamma Amino Butyric Acid (GABA) and Monoamine oxidase-A (MAO-A) in consort with antioxidant activity in mice models [90].

4.5. Ginkgo biloba

Ginkgo biloba, also known as Gingko, is a functional food which improves the availability of oxygen in the tissues [91]. The leaves of Ginkgo have been reported to play a significant role in maintaining the blood flow and glucose level in the brain [92]. Moreover, it also improves the mental functioning of the brain [93]. Ascorbic acid, catechin, shikimic acid, lactone derivatives (ginkgolides) and isorhamnetin are some of the flavone glycosides, which are active scavengers of free radicals and are obtained from the extract of ginkgo leaves [94]. Huang conducted a study to assess the effect of Gingko biloba extract on the liver of the aged rat. The result revealed that administration of Gingko biloba extract reduced the level of liver metalloproteinase as well as malondialdehyde, and improved the SOD activity to minimize the oxidative stress [95]. Another study has revealed that the administration of Ginkgo biloba extract improves the cognitive function in aged female rats [96]. Even clinical studies have been conducted to assess the effect of Gingko biloba extract in the treatment of Alzheimer’s disease and cognitive function. Extensive analysis has revealed that the consumption of Gingko biloba extract improves the cognitive functioning of individuals who have mild dementia [97].

4.6. Glycyrrhiza glabra

Glycyrrhiza glabra, also known as licorice, is a member of the Fabaceae family [98]. The rhizomes, as well as roots of this plant, serve as a brain tonic which helps in regulating the blood sugar level [99]. Glycyrrhizin is the prime bioactive molecule obtained from this plant rich in antioxidants, which protects the brain from oxidative damage, maintains the normal functioning of the nervous system and improves the memory of the individual [100]. Glycyrrhiza glabra has a phenolic compound named “liquorice” which has antioxidant potential, due to which it is effective in the chelating of metal ions and the scavenging of free radicals [101]. It has been reported that G. glabra enhances the memory in the murine model of scopolamine-induced dementia [102]. Dhingra and colleagues also reported improvements in the memory of mice administered with Glycyrrhiza glabra. Three different doses, i.e., 75, 150 and 300 mg/kg p.o. of Glycyrrhiza glabra extracts, were administered for seven consecutive days. The result obtained showed that a dose of 150 mg/kg was effective in enhancing memory in the mice model [103].

4.7. Panax ginseng

Panax ginseng, also known as ginseng, is highly known for its medicinal value [104]. The bioactive molecule ginsenoside is obtained from the roots of this plant [105]. This bioactive molecule improves the resistance of the body against anxiety, fatigue, stress and trauma, and modulates the immune function [106]. Moreover, it also shows anti-stress properties and improves learning performance and memory [104]. A study reported an increase in the life span of juvenile mice with leukaemia upon the administration of ginseng [107]. Another study on Panax ginseng reported that it is able to decrease lipid peroxidation and improve antioxidant potential by reducing oxidative stress [108].
Moreover, double-blind clinical trials have confirmed that the consumption of ginseng improves the psychomotor performance of the individuals [109]. Panax ginseng has also been reported to have anti-melanogenic potential, and is associated with the activation of the foxo3a gene, also stated as the longevity gene [110]. Certain studies have reported that Panax ginseng prevents skin ageing. Furthermore, a randomized, placebo-controlled, double-blind study was conducted to assess the potential of both Panax ginseng and ginsenosides in preventing skin ageing. The result obtained from the study showed a significant reduction in wrinkle formation, and no participant showed an adverse reaction to the treatment [111].

5. Plant-Based Metabolites with Anti-Ageing and Medicinal Properties

5.1. Polyphenols

Plants are prime producers of secondary metabolites, especially polyphenolic compounds, and these are abundantly found in vegetables, fruits, cereals and beverages [112]. Polyphenols have intrigued researchers globally owing to their inherent properties, such as antioxidant potential, and their anticarcinogenic and anti-inflammatory action [113]. These characteristics enable polyphenolic compounds to be useful in the amelioration of various diseases, such as cancer, asthma, microbial infections, diabetes and cardiovascular diseases [114]. Studies have been conducted on numerous polyphenolic compounds, such as resveratrol, proanthocyanins and silymarin. They have been evaluated for their action on animal models subjected to DNA damage, oxidative stress and UV-induced skin irritation [115]. Moreover, these polyphenols, consolidated with sun protection cosmetic products, can effectively shield the skin from UV radiation-associated skin problems and aid in reducing the incidence of skin cancer [116]. Some polyphenols with therapeutic properties have been described below.
Resveratrol (Stilbenes) is a natural polyphenolic compound with antioxidant potential, and is present in the skin of peanuts and grapes [117]. In the last two decades, it has been a prime area of extensive research owing to its application as an anti-ageing ingredient [118]. Additionally, it exhibits anti-inflammatory action and radical scavenging properties, and can act as a chelating agent [119]. Studies have found it to be effective in the treatment of various diseases, including Alzheimer’s and cardiovascular disease [120]. Moreover, Bhat et al. stated that resveratrol possesses cancer chemo-preventive potential [121]. It also has a protective action against human skin, which was confirmed via the study conducted on HaCat cells exposed to nitric oxide free radical donor sodium nitroprusside [122]. Giardina and colleagues conducted an in vitro study on skin fibroblast to assess the efficacy of resveratrol on the proliferation and inhibition of collagen activity. The result obtained showed a dose-related increase in the proliferation rate of cells and substantial inhibition of collagenase activity [123]. Although it has been claimed that resveratrol has the potential to combat ageing at the cellular level and could be a breakthrough in anti-ageing and geriatric medicine, data supporting this claim in the human context are quite limited [124,125,126]. It has been well comprehended that resveratrol modulates mitochondrial biogenesis via stimulating Peroxisome proliferator-activated receptor gamma coactivator 1-alpha (PGC-1α), which further slows down the process of ageing and circumvents the chronic diseases [127,128].
Flavonoids (Phlorizin): Few plants have been found to synthesize phlorizin, a type of flavonoid [129]. It has been immensely exploited by pharmaceutical industries for more than a century, while also serving as a platform to evaluate physiological functioning [115]. Several studies have been conducted on the nutritional benefits of phlorizin. In a recent study, the anti-aging effects of phlorizin and phloretin were tested on murine senile osteoporosis models. The study revealed that phlorizin helped in the management of the ratio of receptor activator of nuclear factor kappa-Β ligand (RANKL) to osteoprotegerin (OPG), which is a biochemical marker of osteoporosis. Phlorizin also reduced the population of osteoclast cells expressing tartrate-resistant acid phosphatase (TRAP) [130]. Phlorizin is found at high concentrations in unripe apples. A preliminary study on human volunteers revealed the beneficial effects of unripe apples containing phlorizin in mitigating post-prandial hyperglycemia. The study was carried out on six healthy individuals and revealed that the consumption of unripe apples caused statistically significant reductions in post-prandial glucose response, as well as increased urinary glucose [131]. Mela and colleagues conducted a study to evaluate the effects of eight plant extracts as well as their combinations (apple (AE, 2.0 g), mulberry fruit (MFE, 1.5 g), elderberry (EE, 2.0 g), mulberry leaf (MLE, 1.0 g), turmeric (TE, 0.18 g), white bean (WBE, 3.0 g), EE  +  TE and AE  +  TE) on post-prandial insulin (PPI) and glucose (PPG) response. The results obtained from the study revealed that extracts of AE, MLE and MFE were effective in reducing PPI and PPG response [132]. Hyperglycemia has been reported to accelerate the aging process, which describes the potential of phlorizin in mitigating the effects of ageing, thereby improving the quality of life [133]. Many other plant extracts have emerged as potent sources of compounds with antioxidant potential [134]. Metabolites such as silymarin, genistein and apigenin have been found to impact the symptoms of skin ageing positively [91]. Still, no clinical or human trials have been conducted to unveil the real anti-ageing potential of phlorizin.
Apple Polyphenols: Apple is enriched with phytochemicals, especially polyphenols that exhibit immense antioxidant potential [135]. A wide range of polyphenolic compounds is found in apples, such as rutin, chlorogenic acid, catechin phloretin, epicatechin and proanthocyanidin B2 [136]. The daily consumption of apples has been portrayed to reduce the incidence of the occurrence of hypercholesterolemia and cardiovascular diseases [137]. Research studies have suggested that consuming apples can considerably lower the risk of lung cancer, especially in females [138]. Different studies have proven that apple is effective in impeding low-density lipoprotein (LDL) oxidation [137]. A study was conducted to evaluate the effects of apple polyphenols on the gene expression of CcO (cytochrome c oxidase) subunits III, CAT (catalase), Mth (methuselah), Rpn11, SOD and VIb. The result obtained from the study revealed that apple polyphenols increased the life span of fruit flies by 10%. Moreover, the downregulation of Mth, the upregulation of gene CAT, SOD1 and SOD2, and no significant change in the gene expression of CcO subunits, Rpn11 or VIb, were observed in the fruit flies [139]. Furthermore, concentrated apple juice has neuroprotective potential, confirmed via the studies conducted on normal aged mice and genetically compromised mice. Still, the anti-ageing potential of apple and its underlining mechanisms remain indefinable [51].
Blueberry Extract: Polyphenols are more abundantly found in blueberries than in other fruits and vegetables [140]. The high antioxidant potential of blueberry extracts has been associated with the amelioration of ageing symptoms [141]. Studies suggest that the regular consumption of blueberries can potentially enhance memory-related issues in elderly populations [142]. It has been stated that the consumption of blueberry extract slows down age-related functional and physiological deficits [143]. Galli and colleagues have found that supplementation with blueberry extract reversed the age-linked decline in the heat shock protein (HSP) of the hippocampal in rats [144]. Additionally, blueberries have been found to be effective in improving motor and cognitive behavior in aged rat models [145]. The life-prolonging potential of blueberry extracts has also been studied in fruit flies to understand the underlying mechanism. The results obtained from the study revealed that the incorporation of 5 mg/mL of blueberry extract into the diet significantly increased the lifespan of fruit flies by 10% [146].
Tea Catechins and Theaflavins: Tea has emerged as the most preferred beverage in the Asian subcontinent [147]. The beneficial aspects associated with the consumption of tea can be attributed to its inherent compounds, namely theaflavins and catechins [148]. Studies have shown the reduced oxidation of DNA molecules via regular intake of green or black tea [149]. Other in vivo studies on Drosophila have reported positive results concerning the increase in average life span by theaflavins and catechins [150]. Various published reports have stated that the consumption of oral tea polyphenols, as well as topical treatment with green tea, inhibits UV radiation- or chemical-induced skin tumorigenesis in various animal models [151]. Tea catechins and theaflavins possess both anti-inflammatory and anticarcinogenic properties [148]. Elmets and his team conducted a study to assess the effect of tea polyphenol extract on parameters linked with acute UV injury. For this, the skin of volunteers was first treated with green tea extract or its constituents, and treated sites were subjected to two minimal erythema doses of solar simulated radiation. Later, the skin was examined for the biochemical, clinical and histologic characteristics of UV-induced DNA damage. The results revealed that tea extract has a dose-dependent inhibitory effect on erythema response induced by UV irradiation. The histologic evaluation also showed a reduced number of Langerhans and sunburn cells [152].
Moreover, tea polyphenol extracts also reduced the DNA damage in the skin. Therefore, researchers stated that tea polyphenol extract could serve as a natural alternative for photoprotection [152]. Chiu and colleagues conducted a study to assess the effect of a combination therapy course of topical and oral green tea on the histological and clinical characteristics of photo-ageing. For this study, 40 women with rational photo-ageing were randomized either to a placebo regimen or a combination of 300 mg tea oral supplements (consumed twice daily) and 10% green tea cream for eight weeks. The results obtained from the study did not show any significant differences in the clinical characteristics of photo-ageing for the placebo or green tea-treated group. However, a histologic improvement in elastic tissue content was observed in the treated participants [153].
Black Rice Anthocyanins: Black rice is abundant in antioxidants, the supplementation of which has been proven to relieve symptoms in patients who have Alzheimer’s [10]. It also has an anticarcinogenic and anti-inflammatory effect [154]. It is also rich in anthocyanins, namely peonidin-3-glucoside and cyanidin-3-o glucoside [155]. Zuo and colleagues conducted a study of the potential of black rice in extending the lifespan of fruit flies. For determination, the effects on the gene expressions of CAT, Mth, Rpn11, SOD1 and SOD2 were evaluated. The result obtained from the study revealed that the consumption of 30 mg/dL of black rice anthocyanins prolonged the lifespan by 14% of the fruit flies. Moreover, the downregulated gene expression of Mth and the upregulated gene expression of CAT, Rpn11, SOD1 and SOD2 was recorded [156]. Huang et al. also conducted a study on a subacute ageing mice model to assess the effect of black rice anthocyanins, and found that black rice anthocyanins exhibit anti-ageing, anti-fatigue and anti-hypoxic properties [157].

5.2. Carotenoids

Carotenoids are vitamin A derivates, such as lycopene and β-carotene, which are known to possess high antioxidant potential as well as photoprotective characteristics [158]. β-carotene and lycopene can moderately improve skin texture [159].
β-Carotene is obtained from various plant sources, such as carrots, mangoes, papaya and pumpkins, among others [160]. It has emerged as a significant carotenoid owing to its characteristics, such as pro-vitamin-A activity, lipid radical scavenging activity and single oxygen quenching properties [161]. β-Carotene has been reported to avert erythema induced by UV rays and possess excellent photoprotection properties [162]. Reports have suggested the association of cellular ageing with low β-Carotene levels in plasma. A study conducted on 68 old-age subjects showed that β-carotene might modulate telomerase activity in older adults [163]. On the other hand, there are well-known ill effects of supplementary beta carotene for smokers, leading to the progression of lung cancer. A pioneering study in 1994 was published in the New England Journal of Medicine by the alpha tocopherol, beta carotene cancer prevention study group. This study reported that there was an unexpected observation of a greater incidence of lung cancer in men receiving supplementary beta-carotene, as opposed to those who did not [164].
Lycopene is a red carotene, carotenoid and phytochemical present in numerous fruits and vegetables such as papayas, watermelons, tomatoes, carrots and others [4]. It possesses a high single oxygen quenching potential, but lacks vitamin A activity [165]. Moreover, a study confirmed the role of lycopene in attenuating oxidative damage in tissues. Upon exposure to UV light, it was observed that more skin lycopene was destroyed in contrast to β-carotene [166]. Products of lycopene have also been reported to be effective against cancerous cells, in addition to their potential to significantly reduce MMP-1 activity, which is known to degrade collagen [167]. Both lycopene and β-carotene, dominant carotenoids found in human tissues and blood, are known to regulate skin properties [168]. In a very recently published paper, Cheng and co-workers reported that lycopene induces the base excision repair pathway in vitro in A549 cells. This study has opened a molecular pathway, which needs further investigation in vivo and in animal models [169].

5.3. Vitamins

Vitamin C is commonly known as ascorbic acid, and is a highly water-soluble vitamin [170]. This colorless compound has high antioxidant potential owing to its strong reducing nature [171]. The photosensitive ascorbic acid works best in a hydrophilic environment [172]. This crystalline compound is not synthesized in humans; therefore, it has to be taken in the regular diet [173]. Diets should be supplemented with vitamin C-rich sources, such as oranges, broccoli, brussels sprouts, green peppers, strawberries, kiwifruit and grapefruit, to avoid the vitamin C deficiency associated health problems like cardiovascular diseases, scurvy, and others [174]. Ascorbic acid has a high antioxidant potential and free radical-scavenging properties, which helps in preventing the oxidation of tissues, cell membranes and macromolecules (DNA and proteins) by free radicals [173].
Vitamin E is a fat-soluble membrane-bound compound which has high free radical-scavenging as well as antioxidant potential [175]. This nonenzymatic antioxidant is found in wheat germ oil, safflower oil, sunflower oil, vegetables, peanuts, corn, almonds, soy and meat [176]. A deficiency of vitamin E in the body may lead to the development of various health conditions in infants, such as dryness, papular erythema, depigmentation and oedema [177]. Vitamin E consumption helps in combating skin ageing symptoms due to its efficacy in preventing the peroxidation of lipids and the cross-connection of collagen fibers [4]. Vitamin E has been proven to relieve sunburn and UV-associated skin damage [178].
Both vitamins C and E work synergistically. For instance, when UV-induced molecules oxidize the cellular constituents, a chain reaction of lipid peroxidation starts in the membrane rich in polyunsaturated fatty acids. During this, d-α-tocopherol (antioxidant) gets oxidized to the tocopheroxyl radical and regenerates itself through ascorbic acid [179,180]. Different food sources such as corn, seeds, vegetable oils (sunflower oil and safflower oil) and soy are rich in tocopherol [4]. Moreover, the consumption of vitamin E from natural sources help against lipid peroxidation and collagen cross-linking, as both are associated with skin ageing. Additionally, topically applied vitamin E has also been reported to reduce chronic UVB-induced skin damage, erythema, sunburned cells and photocarcinogenesis [181,182]. A deficiency of vitamin E is also associated with a syndrome of edema with seborrheic changes, as well as depigmentation and dryness in premature infants [183]. Ekanayake-Mudiyanselage and Thiele, upon analyzing their study, stated that the level of vitamin E is dependent on the density of sebaceous glands in the skin. The oral supplementation of α-tocopherol for three weeks has been shown to cause a substantial increase in vitamin E levels in the sebaceous gland, especially on the face [184]. In a comparative study, the oral consumption of both vitamin C and E has been shown to improve the photoprotective effect in contrast to monotherapies [185]. Another study was conducted on 33 participants who received 100 or 180 mg vitamin C or placebo per day for four weeks. The result obtained from the study revealed that orally consumed vitamin C improved the radical scavenging activity of the skin by 22% (for 100 mg) and 37% (for 180 mg) from the baseline [87]. In the study by the alpha-tocopherol and beta carotene cancer prevention study group, it was found that vitamin E has insignificant effects on the prevention of lung cancer [164].
Nutraceuticals, functional foods and dietary supplements encompass a large group of compounds which are well known to improve health [186]. Functional foods have gained global attention owing to their impact on improving the symptoms of skin ageing [187]. Notably, fruits constitute an essential source of active metabolites used to curb skin ageing symptoms, as they are enriched with phenolic compounds, carotenoids and ascorbic acid, and possess high antioxidant potential [188]. The various plants and their components with anti-ageing potential are listed in Table 1.

6. Concluding Remarks

Ageing is a complex and progressive biological process, which gets affected by environmental and genetic factors. Nowadays, ageing is also linked with the consumption of an imbalanced diet deficient in many essential nutrients. Lately, nutraceuticals have gained appreciation and are being considered as a crucial element in improving life and providing antioxidant-containing molecules. Various vegetables and fruits contain antioxidant molecules with beneficial properties that can help in delaying the process of ageing. Moreover, these nutraceuticals do not show unwanted symptoms; instead, they have a beneficial impact on the digestive system. Therefore, nutraceuticals as food supplements have promising potential in combating as well as delaying the ageing process. The benefits associated with nutraceuticals prompts their incorporation into the diet for health benefits and long life. The current review meticulously summarizes the anti-ageing effects of plant-based supplements and plant-derived metabolites. Since most of the data have been obtained in vitro, caution is advised for inferring the clinical applicability of in vitro-tested molecules. Referencing, examining and confirming the human trial data is highly recommended.

Author Contributions

Conceptualization, R.S. (Reena Singh) and K.K.; Manuscript writing, D.S.D. and S.B.; Manuscript editing, D.S.D., S.B., R.S. (Ruchi Sharma), K.B., D.K., C.C. and E.N.; Critical revising, R.S. (Reena Singh) and K.K. All authors have read and agreed to the published version of the manuscript.

Funding

This research was funded by the University of Hradec Kralove (Faculty of Science VT2019-2021) and Ministry of Health (No. NV19-09-00578).

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Jin, K.; Rose, M.R. Modern Biological Theories of Aging. Aging Dis. 1988, 1, 220–221. [Google Scholar] [CrossRef]
  2. Zhang, S.; Duan, E. Fighting against Skin Aging: The Way from Bench to Bedside. Cell Transplant. 2018, 27, 729–738. [Google Scholar] [CrossRef] [PubMed]
  3. Bocheva, G.; Slominski, R.M.; Slominski, A.T. Neuroendocrine aspects of skin aging. Int. J. Mol. Sci. 2019, 20, 2798. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  4. Schagen, S.K.; Zampeli, V.A.; Makrantonaki, E.; Zouboulis, C.C. Discovering the link between nutrition and skin aging. Dermatoendocrinology 2012, 4, 298–307. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Lobo, V.; Patil, A.; Phatak, A.; Chandra, N. Free radicals, antioxidants and functional foods: Impact on human health. Pharmacogn. Rev. 2010, 4, 118–126. [Google Scholar] [CrossRef] [Green Version]
  6. Alam, I.; Almajwal, A.M.; Alam, W.; Alam, I.; Ullah, N.; Abulmeaaty, M.; Razak, S.; Khan, S.; Pawelec, G.; Paracha, P.I. The immune-nutrition interplay in aging-Facts and controversies. Nutr. Healthy Aging 2019, 5, 73–95. [Google Scholar] [CrossRef] [Green Version]
  7. Rathore, H.; Prasad, S.; Sharma, S. Mushroom nutraceuticals for improved nutrition and better human health: A review. PharmaNutrition 2017, 5, 35–46. [Google Scholar] [CrossRef]
  8. Da Costa, J.P. A current look at nutraceuticals–Key concepts and future prospects. Trends Food Sci. Technol. 2017, 62, 68–78. [Google Scholar] [CrossRef]
  9. Chauhan, B.; Kumar, G.; Kalam, N.; Ansari, S.H. Current concepts and prospects of herbal nutraceutical: A review. J. Adv. Pharm. Technol. Res. 2013, 4, 4–8. [Google Scholar]
  10. Liu, Z.; Ren, Z.; Zhang, J.; Chuang, C.C.; Kandaswamy, E.; Zhou, T.; Zuo, L. Role of ROS and nutritional antioxidants in human diseases. Front. Physiol. 2018, 9, 477. [Google Scholar] [CrossRef] [Green Version]
  11. Conlon, M.A.; Bird, A.R. The impact of diet and lifestyle on gut microbiota and human health. Nutrients 2015, 7, 17–44. [Google Scholar] [CrossRef] [PubMed]
  12. Eming, S.A.; Martin, P.; Tomic-Canic, M. Wound repair and regeneration: Mechanisms, signaling, and translation. Sci. Transl. Med. 2014, 6, 265sr6. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  13. Salam, N.; Rane, S.; Das, R.; Faulkner, M.; Gund, R.; Kandpal, U.; Lewis, V.; Mattoo, H.; Prabhu, S.; Ranganathan, V.; et al. T cell ageing: Effects of age on development, survival & function. Indian J. Med. Res. 2013, 138, 595–608. [Google Scholar] [PubMed]
  14. Amarya, S.; Singh, K.; Sabharwal, M. Ageing Process and Physiological Changes. In Gerontology; InTech: London, UK, 2018. [Google Scholar]
  15. Prohaska, T.R.; Keller, M.L.; Leventhal, E.A.; Leventhal, H. Impact of symptoms and aging attribution on emotions and coping. Health Psychol. 1987, 6, 495–514. [Google Scholar] [CrossRef]
  16. Heinemann, L.A.J.; Zimmermann, T.; Vermeulen, A.; Thiel, C.; Hummel, W. A new «aging males» symptoms’ rating scale. Aging Male 1999, 2, 105–114. [Google Scholar] [CrossRef]
  17. Maddy, A.J.; Tosti, A. Hair and nail diseases in the mature patient. Clin. Dermatol. 2018, 36, 159–166. [Google Scholar] [CrossRef]
  18. Sarbacher, C.A.; Halper, J.T. Connective tissue and age-related diseases. In Subcellular Biochemistry; Springer: New York, NY, USA, 2019; pp. 281–310. [Google Scholar]
  19. Patel, I.; West, S.K. Presbyopia: Prevalence, impact, and interventions. Community Eye Health J. 2007, 20, 40–41. [Google Scholar]
  20. Boostani, R.; Karimzadeh, F.; Nami, M. A comparative review on sleep stage classification methods in patients and healthy individuals. Comput. Methods Progr. Biomed. 2017, 140, 77–91. [Google Scholar] [CrossRef]
  21. Locantore, P.; Del Gatto, V.; Gelli, S.; Paragliola, R.M.; Pontecorvi, A. The Interplay between Immune System and Microbiota in Osteoporosis. Mediat. Inflamm. 2020, 2020, 3686749. [Google Scholar] [CrossRef]
  22. Trexler, E.T.; Smith-Ryan, A.E.; Norton, L.E. Metabolic adaptation to weight loss: Implications for the athlete. J. Int. Soc. Sports Nutr. 2014, 11, 7. [Google Scholar] [CrossRef] [Green Version]
  23. Wood, R.L. Accelerated cognitive aging following severe traumatic brain injury: A review. Brain Inj. 2017, 31, 1270–1278. [Google Scholar] [CrossRef] [PubMed]
  24. Sarnak, M.J. A patient with heart failure and worsening kidney function. Clin. J. Am. Soc. Nephrol. 2014, 9, 1790–1798. [Google Scholar] [CrossRef] [Green Version]
  25. De Martinis, M.; Sirufo, M.M.; Ginaldi, L. Allergy and aging: An Old/new emerging health issue. Aging Dis. 2017, 8, 162–175. [Google Scholar] [CrossRef] [Green Version]
  26. Santoro, N.; Epperson, C.N.; Mathews, S.B. Menopausal Symptoms and Their Management. Endocrinol. Metab. Clin. N. Am. 2015, 44, 497–515. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  27. Booth, F.W.; Roberts, C.K.; Laye, M.J. Lack of exercise is a major cause of chronic diseases. Compr. Physiol. 2012, 2, 1143–1211. [Google Scholar] [CrossRef] [Green Version]
  28. Da Costa, J.P.; Vitorino, R.; Silva, G.M.; Vogel, C.; Duarte, A.C.; Rocha-Santos, T. A synopsis on aging—Theories, mechanisms and future prospects. Ageing Res. Rev. 2016, 29, 90–112. [Google Scholar] [CrossRef] [PubMed]
  29. De, A.; Ghosh, C. Basics of aging theories and disease related aging-an overview. PharmaTutor 2017, 5, 16–23. [Google Scholar]
  30. Weinert, B.T.; Timiras, P.S. Invited review: Theories of aging. J. Appl. Physiol. 2003, 95, 1706–1716. [Google Scholar] [CrossRef]
  31. Gladyshev, V.N. The free radical theory of aging is dead. Long live the damage theory! Antioxid. Redox Signal. 2014, 20, 727–731. [Google Scholar] [CrossRef]
  32. Sailaja Rao, P.; Kalva, S.; Yerramilli, A.; Mamidi, S. Free Radicals and Tissue Damage: Role of Antioxidants. Free Radic. Antioxid. 2011, 1, 2–7. [Google Scholar] [CrossRef]
  33. Phaniendra, A.; Jestadi, D.B.; Periyasamy, L. Free Radicals: Properties, Sources, Targets, and Their Implication in Various Diseases. Indian J. Clin. Biochem. 2015, 30, 11–26. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  34. Di Meo, S.; Venditti, P. Evolution of the Knowledge of Free Radicals and Other Oxidants. Oxid. Med. Cell. Longev. 2020, 2020, 9829176. [Google Scholar] [CrossRef] [PubMed]
  35. Pham-Huy, L.A.; He, H.; Pham-Huy, C. Free radicals, antioxidants in disease and health. Int. J. Biomed. Sci. 2008, 4, 89–96. [Google Scholar] [PubMed]
  36. Aruoma, O.I. Nutrition and health aspects of free radicals and antioxidants. Food Chem. Toxicol. 1994, 32, 671–683. [Google Scholar] [CrossRef]
  37. Dröge, W. Free radicals in the physiological control of cell function. Physiol. Rev. 2002, 82, 47–95. [Google Scholar] [CrossRef]
  38. Floyd, R.A.; Carney, J.M. Free radical damage to protein and DNA: Mechanisms involved and relevant observations on brain undergoing oxidative stress. Ann. Neurol. 1992, 32, S22–S27. [Google Scholar] [CrossRef]
  39. Kumar, H.; Bhardwaj, K.; Nepovimova, E.; Kuča, K.; Singh Dhanjal, D.; Bhardwaj, S.; Bhatia, S.K.; Verma, R.; Kumar, D. Antioxidant Functionalized Nanoparticles: A Combat against Oxidative Stress. Nanomaterials 2020, 10, 1334. [Google Scholar] [CrossRef]
  40. Thanan, R.; Oikawa, S.; Hiraku, Y.; Ohnishi, S.; Ma, N.; Pinlaor, S.; Yongvanit, P.; Kawanishi, S.; Murata, M. Oxidative stress and its significant roles in neurodegenerative diseases and cancer. Int. J. Mol. Sci. 2014, 16, 193–217. [Google Scholar] [CrossRef] [Green Version]
  41. Sharma, P.; Jha, A.B.; Dubey, R.S.; Pessarakli, M. Reactive Oxygen Species, Oxidative Damage, and Antioxidative Defense Mechanism in Plants under Stressful Conditions. J. Bot. 2012, 2012, 217037. [Google Scholar] [CrossRef] [Green Version]
  42. Ozcan, A.; Ogun, M. Biochemistry of Reactive Oxygen and Nitrogen Species. In Basic Principles and Clinical Significance of Oxidative Stress; InTech: London, UK, 2015. [Google Scholar]
  43. Pourahmad, J.; Salimi, A.; Seydi, E. Role of Oxygen Free Radicals in Cancer Development and Treatment. In Free Radicals and Diseases; InTech: London, UK, 2016. [Google Scholar]
  44. Jamshidi-kia, F.; Wibowo, J.P.; Elachouri, M.; Masumi, R.; Salehifard-Jouneghani, A.; Abolhasanzadeh, Z.; Lorigooini, Z. Battle between plants as antioxidants with free radicals in human body. J. Herbmed Pharmacol. 2020, 9, 191–199. [Google Scholar] [CrossRef]
  45. Santo, A.; Zhu, H.; Li, Y.R. Free radicals: From health to disease. React. Oxyg. Species 2016, 2, 245–263. [Google Scholar] [CrossRef]
  46. Liguori, I.; Russo, G.; Curcio, F.; Bulli, G.; Aran, L.; Della-Morte, D.; Gargiulo, G.; Testa, G.; Cacciatore, F.; Bonaduce, D.; et al. Oxidative stress, aging, and diseases. Clin. Interv. Aging 2018, 13, 757–772. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  47. Nissanka, N.; Moraes, C.T. Mitochondrial DNA damage and reactive oxygen species in neurodegenerative disease. FEBS Lett. 2018, 592, 728–742. [Google Scholar] [CrossRef] [PubMed]
  48. Cantó, C.; Auwerx, J. Caloric restriction, SIRT1 and longevity. Trends Endocrinol. Metab. 2009, 20, 325–331. [Google Scholar] [CrossRef] [Green Version]
  49. Armandola, E. Caloric Restriction and Life Expectancy: Highlights of the 5th European Molecular Biology Organization Interdisciplinary Conference on Science and Society—Time & Aging: Mechanisms and Meanings; November 5–6, 2004; Heidelberg, Germany. Med. Gen. Med. 2004, 6, 16. [Google Scholar]
  50. Gutierrez, J.; Ballinger, S.W.; Darley-Usmar, V.M.; Landar, A. Free radicals, mitochondria, and oxidized lipids: The emerging role in signal transduction in vascular cells. Circ. Res. 2006, 99, 924–932. [Google Scholar] [CrossRef]
  51. Peng, C.; Wang, X.; Chen, J.; Jiao, R.; Wang, L.; Li, Y.M.; Zuo, Y.; Liu, Y.; Lei, L.; Ma, K.Y.; et al. Biology of ageing and role of dietary antioxidants. BioMed Res. Int. 2014, 2014, 831841. [Google Scholar] [CrossRef] [Green Version]
  52. Hornsby, P.J. Telomerase and the aging process. Exp. Gerontol. 2007, 42, 575–581. [Google Scholar] [CrossRef] [Green Version]
  53. Schmidt, J.C.; Cech, T.R. Human telomerase: Biogenesis, trafficking, recruitment, and activation. Genes Dev. 2015, 29, 1095–1105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Kalmbach, K.H.; Fontes Antunes, D.M.; Dracxler, R.C.; Knier, T.W.; Seth-Smith, M.L.; Wang, F.; Liu, L.; Keefe, D.L. Telomeres and human reproduction. Fertil. Steril. 2013, 99, 23–29. [Google Scholar] [CrossRef] [Green Version]
  55. Kasote, D.M.; Katyare, S.S.; Hegde, M.V.; Bae, H. Significance of antioxidant potential of plants and its relevance to therapeutic applications. Int. J. Biol. Sci. 2015, 11, 982–991. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Chen, L.; Deng, H.; Cui, H.; Fang, J.; Zuo, Z.; Deng, J.; Li, Y.; Wang, X.; Zhao, L. Inflammatory responses and inflammation-associated diseases in organs. Oncotarget 2018, 9, 7204–7218. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  57. Liao, L.Y.; He, Y.F.; Li, L.; Meng, H.; Dong, Y.M.; Yi, F.; Xiao, P.G. A preliminary review of studies on adaptogens: Comparison of their bioactivity in TCM with that of ginseng-like herbs used worldwide Milen Georgiev, Ruibing Wang. Chin. Med. 2018, 13, 57. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Bhatia, N.; Jaggi, A.S.; Singh, N.; Anand, P.; Dhawan, R. Adaptogenic potential of curcumin in experimental chronic stress and chronic unpredictable stress-induced memory deficits and alterations in functional homeostasis. J. Nat. Med. 2011, 65, 532–543. [Google Scholar] [CrossRef] [PubMed]
  59. Singh, M.K.; Jain, G.; Das, B.K.; Patil, U.K. Biomolecules from Plants as an Adaptogen. Med. Aromat. Plants 2017, 6, 307. [Google Scholar] [CrossRef]
  60. Aguiar, S.; Borowski, T. Neuropharmacological review of the nootropic herb Bacopa monnieri. Rejuvenation Res. 2013, 16, 313–326. [Google Scholar] [CrossRef] [Green Version]
  61. Jain, P.K.; Das, D.; Kumar Jain, P. Pharmacognostic Comparison of Bacopa Monnieri, Cyperus Rotundus and Emblica Officinalis. Innovare J. Ayurvedic Sci. 2016, 4, 16–26. [Google Scholar]
  62. Tewari, I.; Sharma, L.; Lal Gupta, G. Synergistic antioxidant activity of three medicinal plants Hypericum perforatum, Bacopa monnieri, and Camellia Sinensis. Indo Am. J. Pharm. Res. 2014, 4, 2563–2568. [Google Scholar]
  63. Vollala, V.R.; Upadhya, S.; Nayak, S. Effect of Bacopa monniera Linn. (brahmi) extract on learning and memory in rats: A behavioral study. J. Vet. Behav. Clin. Appl. Res. 2010, 5, 69–74. [Google Scholar] [CrossRef]
  64. Simpson, T.; Pase, M.; Stough, C. Bacopa monnieri as an Antioxidant Therapy to Reduce Oxidative Stress in the Aging Brain. Evid. Based Complement. Altern. Med. 2015, 2015, 615384. [Google Scholar] [CrossRef] [Green Version]
  65. Bhattacharya, S.K.; Bhattacharya, A.; Kumar, A.; Ghosal, S. Antioxidant activity of Bacopa monniera in rat frontal cortex, striatum and hippocampus. Phyther. Res. 2000, 14, 174–179. [Google Scholar] [CrossRef]
  66. Shinomol, G.K.; Srinivas Bharath, M.M. Muralidhara Neuromodulatory propensity of bacopa monnieri leaf extract against 3-nitropropionic acid-induced oxidative stress: In vitro and in vivo evidences. Neurotox. Res. 2012, 22, 102–114. [Google Scholar] [CrossRef]
  67. Kumar, N.; Abichandani, L.G.; Thawani, V.; Gharpure, K.J.; Naidu, M.U.R.; Venkat Ramana, G. Efficacy of Standardized Extract of Bacopa monnieri (Bacognize®) on Cognitive Functions of Medical Students: A Six-Week, Randomized Placebo-Controlled Trial. Evid. Based Complement. Altern. Med. 2016, 2016. [Google Scholar] [CrossRef] [Green Version]
  68. Kocaadam, B.; Şanlier, N. Curcumin, an active component of turmeric (Curcuma longa), and its effects on health. Crit. Rev. Food Sci. Nutr. 2017, 57, 2889–2895. [Google Scholar] [CrossRef] [PubMed]
  69. Prasad, S.; Aggarwal, B.B. Turmeric, the golden spice: From traditional medicine to modern medicine. In Herbal Medicine: Biomolecular and Clinical Aspects: Second Edition; CRC Press: Boca Raton, FL, USA, 2011; pp. 263–288. ISBN 9781439807163. [Google Scholar]
  70. Tomeh, M.A.; Hadianamrei, R.; Zhao, X. A review of curcumin and its derivatives as anticancer agents. Int. J. Mol. Sci. 2019, 20, 1033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Desai, S.J.; Prickril, B.; Rasooly, A. Mechanisms of Phytonutrient Modulation of Cyclooxygenase-2 (COX-2) and Inflammation Related to Cancer. Nutr. Cancer 2018, 70, 350–375. [Google Scholar] [CrossRef] [PubMed]
  72. Engwa, G.A. Free Radicals and the Role of Plant Phytochemicals as Antioxidants Against Oxidative Stress-Related Diseases. In Phytochemicals-Source of Antioxidants and Role in Disease Prevention; InTech: London, UK, 2018. [Google Scholar]
  73. Lima, C.F.; Pereira-Wilson, C.; Rattan, S.I.S. Curcumin induces heme oxygenase-1 in normal human skin fibroblasts through redox signaling: Relevance for anti-aging intervention. Mol. Nutr. Food Res. 2011, 55, 430–442. [Google Scholar] [CrossRef] [Green Version]
  74. Soh, J.W.; Marowsky, N.; Nichols, T.J.; Rahman, A.M.; Miah, T.; Sarao, P.; Khasawneh, R.; Unnikrishnan, A.; Heydari, A.R.; Silver, R.B.; et al. Curcumin is an early-acting stage-specific inducer of extended functional longevity in Drosophila. Exp. Gerontol. 2013, 48, 229–239. [Google Scholar] [CrossRef]
  75. Shen, L.-R.; Parnell, L.D.; Ordovas, J.M.; Lai, C.-Q. Curcumin and aging. BioFactors 2013, 39, 133–140. [Google Scholar] [CrossRef]
  76. Lee, K.S.; Lee, B.S.; Semnani, S.; Avanesian, A.; Um, C.Y.; Jeon, H.J.; Seong, K.M.; Yu, K.; Min, K.J.; Jafari, M. Curcumin extends life span, improves health span, and modulates the expression of age-associated aging genes in drosophila melanogaster. Rejuvenation Res. 2010, 13, 561–570. [Google Scholar] [CrossRef]
  77. He, Y.; Yue, Y.; Zheng, X.; Zhang, K.; Chen, S.; Du, Z. Curcumin, inflammation, and chronic diseases: How are they linked? Molecules 2015, 20, 9183–9213. [Google Scholar] [CrossRef] [PubMed]
  78. Olszanecki, R.; Jawien, J.; Gajda, M.; Mateuszuk, L.; Gebska, A.; Korabiowska, M.; Chlopicki, S.; Korbut, R. Effect of curcumin on atherosclerosis in apoE-LDLR-double knockout mice. J. Physiol. Pharmacol. 2005, 4, 627–635. [Google Scholar]
  79. Swamy, A.V.; Gulliaya, S.; Thippeswamy, A.; Koti, B.C.; Manjula, D.V. Cardioprotective effect of curcumin against doxorubicin-induced myocardial toxicity in albino rats. Indian J. Pharmacol. 2012, 44, 73–77. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  80. Ryan, J.L.; Heckler, C.E.; Ling, M.; Katz, A.; Williams, J.P.; Pentland, A.P.; Morrow, G.R. Curcumin for radiation dermatitis: A randomized, double-blind, placebo-controlled clinical trial of thirty breast cancer patients. Radiat. Res. 2013, 180, 34–43. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  81. Ray Hamidie, R.D.; Yamada, T.; Ishizawa, R.; Saito, Y.; Masuda, K. Curcumin treatment enhances the effect of exercise on mitochondrial biogenesis in skeletal muscle by increasing cAMP levels. Metabolism 2015, 64, 1334–1347. [Google Scholar] [CrossRef] [PubMed]
  82. Cox, K.H.M.; Pipingas, A.; Scholey, A.B. Investigation of the effects of solid lipid curcumin on cognition and mood in a healthy older population. J. Psychopharmacol. 2015, 29, 642–651. [Google Scholar] [CrossRef]
  83. Yadav, S.S.; Singh, M.K.; Singh, P.K.; Kumar, V. Traditional knowledge to clinical trials: A review on therapeutic actions of Emblica officinalis. Biomed. Pharmacother. 2017, 93, 1292–1302. [Google Scholar] [CrossRef]
  84. Kapoor, M.P.; Suzuki, K.; Derek, T.; Ozeki, M.; Okubo, T. Clinical evaluation of Emblica Officinalis Gatertn (Amla) in healthy human subjects: Health benefits and safety results from a randomized, double-blind, crossover placebo-controlled study. Contemp. Clin. Trials Commun. 2020, 17, 100499. [Google Scholar] [CrossRef]
  85. Wilson, D.W.; Nash, P.; Singh, H.; Griffiths, K.; Singh, R.; De Meester, F.; Horiuchi, R.; Takahashi, T. The role of food antioxidants, benefits of functional foods, and influence of feeding habits on the health of the older person: An overview. Antioxidants 2017, 6, 81. [Google Scholar] [CrossRef] [Green Version]
  86. Hasan, M.R.; Islam, M.N.; Islam, M.R. Phytochemistry, pharmacological activities and traditional uses of Emblica officinalis: A review. Int. Curr. Pharm. J. 2016, 5, 14–21. [Google Scholar] [CrossRef] [Green Version]
  87. Lauer, A.C.; Groth, N.; Haag, S.F.; Darvin, M.E.; Lademann, J.; Meinke, M.C. Dose-dependent vitamin C uptake and radical scavenging activity in human skin measured with in vivo electron paramagnetic resonance spectroscopy. Skin Pharmacol. Physiol. 2013, 26, 147–154. [Google Scholar] [CrossRef] [PubMed]
  88. Bhattacharya, A.; Ghosal, S.; Bhattacharya, S.K. Antioxidant activity of tannoid principles of Emblica officinalis (amla) in chronic stress induced changes in rat brain. Indian J. Exp. Biol. 2000, 38, 877–880. [Google Scholar] [PubMed]
  89. Bhattachary, S.K.; Bhattacharya, D.; Muruganandam, A.V. Effect of Emblica officinalis tannoids on a rat model of tardive dyskinesia. Indian J. Exp. Biol. 2000, 38, 945–947. [Google Scholar] [PubMed]
  90. Dhingra, D.; Joshi, P.; Gupta, A.; Chhillar, R. Possible Involvement of Monoaminergic Neurotransmission in Antidepressant-like activity of Emblica officinalis Fruits in Mice. CNS Neurosci. Ther. 2012, 18, 419–425. [Google Scholar] [CrossRef] [PubMed]
  91. Isah, T. Rethinking Ginkgo biloba L.: Medicinal uses and conservation. Pharmacogn. Rev. 2015, 9, 140–148. [Google Scholar] [CrossRef] [Green Version]
  92. Mashayekh, A.; Pham, D.L.; Yousem, D.M.; Dizon, M.; Barker, P.B.; Lin, D.D.M. Effects of Ginkgo biloba on cerebral blood flow assessed by quantitative MR perfusion imaging: A pilot study. Neuroradiology 2011, 53, 185–191. [Google Scholar] [CrossRef] [Green Version]
  93. Zuo, W.; Yan, F.; Zhang, B.; Li, J.; Mei, D. Advances in the studies of Ginkgo biloba leaves extract on aging-related diseases. Aging Dis. 2017, 8, 812–826. [Google Scholar] [CrossRef] [Green Version]
  94. Van Beek, T.A. Chemical analysis of Ginkgo biloba leaves and extracts. J. Chromatogr. A 2002, 967, 21–55. [Google Scholar] [CrossRef]
  95. Huang, S.Z.; Luo, Y.J.; Wang, L.; Cai, K.Y. Effect of ginkgo biloba extract on livers in aged rats. World J. Gastroenterol. 2005, 11, 132–135. [Google Scholar] [CrossRef]
  96. Belviranli, M.; Okudan, N. The effects of Ginkgo biloba extract on cognitive functions in aged female rats: The role of oxidative stress and brain-derived neurotrophic factor. Behav. Brain Res. 2015, 278, 453–461. [Google Scholar] [CrossRef]
  97. Liu, H.; Ye, M.; Guo, H. An Updated Review of Randomized Clinical Trials Testing the Improvement of Cognitive Function of Ginkgo biloba Extract in Healthy People and Alzheimer’s Patients. Front. Pharmacol. 2020, 10, 1688. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Pastorino, G.; Cornara, L.; Soares, S.; Rodrigues, F.; Oliveira, M.B.P.P. Liquorice (Glycyrrhiza glabra): A phytochemical and pharmacological review. Phyther. Res. 2018, 32, 2323–2339. [Google Scholar] [CrossRef] [PubMed]
  99. Frattaruolo, L.; Carullo, G.; Brindisi, M.; Mazzotta, S.; Bellissimo, L.; Rago, V.; Curcio, R.; Dolce, V.; Aiello, F.; Cappello, A.R. Antioxidant and anti-inflammatory activities of flavanones from glycyrrhiza glabra L. (licorice) leaf phytocomplexes: Identification of licoflavanone as a modulator of NF-kB/MAPK pathway. Antioxidants 2019, 8, 186. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  100. Grodzicki, W.; Dziendzikowska, K. The role of selected bioactive compounds in the prevention of alzheimer’s disease. Antioxidants 2020, 9, 229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  101. Ciganović, P.; Jakimiuk, K.; Tomczyk, M.; Končić, M.Z. Glycerolic licorice extracts as active cosmeceutical ingredients: Extraction optimization, chemical characterization, and biological activity. Antioxidants 2019, 8, 445. [Google Scholar] [CrossRef] [Green Version]
  102. Balmus, I.M.; Ciobica, A. Main Plant Extracts’ Active Properties Effective on Scopolamine-Induced Memory Loss. Am. J. Alzheimers Dis. Other Demen. 2017, 32, 418–428. [Google Scholar] [CrossRef]
  103. Dhingra, D.; Parle, M.; Kulkarni, S.K. Memory enhancing activity of Glycyrrhiza glabra in mice. J. Ethnopharmacol. 2004, 91, 361–365. [Google Scholar] [CrossRef]
  104. Rokot, N.T.; Kairupan, T.S.; Cheng, K.C.; Runtuwene, J.; Kapantow, N.H.; Amitani, M.; Morinaga, A.; Amitani, H.; Asakawa, A.; Inui, A. A Role of Ginseng and Its Constituents in the Treatment of Central Nervous System Disorders. Evid. Based Complement. Altern. Med. 2016, 2016, 2614742. [Google Scholar] [CrossRef] [Green Version]
  105. Yu, H.; Zhao, J.; You, J.; Li, J.; Ma, H.; Chen, X. Factors influencing cultivated ginseng (Panax ginseng C. A. Meyer) bioactive compounds. PLoS ONE 2019, 14, e0223763. [Google Scholar] [CrossRef]
  106. Kumar, G.P.; Khanum, F. Neuroprotective potential of phytochemicals. Pharmacogn. Rev. 2012, 6, 81–90. [Google Scholar] [CrossRef] [Green Version]
  107. Wee, J.J.; Mee Park, K.; Chung, A.-S. Biological Activities of Ginseng and Its Application to Human Health. In Herbal Medicine: Biomolecular and Clinical Aspects; Benzie, I.F.F., Wachtel-Galor, S., Eds.; CRC Press/Taylor and Francis: Boca Raton, FL, USA, 2011; ISBN 978-1-4398-0713-2. [Google Scholar]
  108. Lee, Y.M.; Yoon, H.; Park, H.M.; Song, B.C.; Yeum, K.J. Implications of red Panax ginseng in oxidative stress associated chronic diseases. J. Ginseng Res. 2017, 41, 113–119. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  109. Caldwell, L.K.; Dupont, W.H.; Beeler, M.K.; Post, E.M.; Barnhart, E.C.; Hardesty, V.H.; Anders, J.P.; Borden, E.C.; Volek, J.S.; Kraemer, W.J. The effects of a Korean ginseng, GINST15, on perceptual effort, psychomotor performance, and physical performance in men and women. J. Sport. Sci. Med. 2018, 17, 92–100. [Google Scholar]
  110. Kim, J.; Cho, S.Y.; Kim, S.H.; Cho, D.; Kim, S.; Park, C.W.; Shimizu, T.; Cho, J.Y.; Seo, D.B.; Shin, S.S. Effects of Korean ginseng berry on skin antipigmentation and antiaging via FoxO3a activation. J. Ginseng Res. 2017, 41, 277–283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  111. Hwang, E.; Park, S.Y.; Jo, H.; Lee, D.G.; Kim, H.T.; Kim, Y.M.; Yin, C.S.; Yi, T.H. Efficacy and Safety of Enzyme-Modified Panax ginseng for Anti-Wrinkle Therapy in Healthy Skin: A Single-Center, Randomized, Double-Blind, Placebo-Controlled Study. Rejuvenation Res. 2015, 18, 449–457. [Google Scholar] [CrossRef]
  112. Shahidi, F.; Ambigaipalan, P. Phenolics and polyphenolics in foods, beverages and spices: Antioxidant activity and health effects-A review. J. Funct. Foods 2015, 18, 820–897. [Google Scholar] [CrossRef]
  113. Quero, J.; Mármol, I.; Cerrada, E.; Rodríguez-Yoldi, M.J. Insight into the potential application of polyphenol-rich dietary intervention in degenerative disease management. Food Funct. 2020, 11, 2805–2825. [Google Scholar] [CrossRef]
  114. Pandey, K.B.; Rizvi, S.I. Plant polyphenols as dietary antioxidants in human health and disease. Oxid. Med. Cell. Longev. 2009, 2, 270–278. [Google Scholar] [CrossRef] [Green Version]
  115. Dunaway, S.; Odin, R.; Zhou, L.; Ji, L.; Zhang, Y.; Kadekaro, A.L. Natural antioxidants: Multiple mechanisms to protect skin from solar radiation. Front. Pharmacol. 2018, 9, 392. [Google Scholar] [CrossRef] [Green Version]
  116. D’Orazio, J.; Jarrett, S.; Amaro-Ortiz, A.; Scott, T. UV radiation and the skin. Int. J. Mol. Sci. 2013, 14, 12222–12248. [Google Scholar] [CrossRef] [Green Version]
  117. Adhikari, B.; Dhungana, S.K.; Waqas Ali, M.; Adhikari, A.; Kim, I.D.; Shin, D.H. Antioxidant activities, polyphenol, flavonoid, and amino acid contents in peanut shell. J. Saudi Soc. Agric. Sci. 2019, 18, 437–442. [Google Scholar] [CrossRef]
  118. Camins, A.; Junyent, F.; Verdaguer, E.; Beas-Zarate, C.; Rojas-Mayorquín, A.; Ortuño-Sahagún, D.; Pallàs, M. Resveratrol: An Antiaging Drug with Potential Therapeutic Applications in Treating Diseases. Pharmaceuticals 2009, 2, 194–205. [Google Scholar] [CrossRef]
  119. Salehi, B.; Mishra, A.P.; Nigam, M.; Sener, B.; Kilic, M.; Sharifi-Rad, M.; Fokou, P.V.T.; Martins, N.; Sharifi-Rad, J. Resveratrol: A double-edged sword in health benefits. Biomedicines 2018, 6, 91. [Google Scholar] [CrossRef] [Green Version]
  120. Quadros Gomes, B.A.; Bastos Silva, J.P.; Rodrigues Romeiro, C.F.; dos Santos, S.M.; Rodrigues, C.A.; Gonçalves, P.R.; Sakai, J.T.; Santos Mendes, P.F.; Pompeu Varela, E.L.; Monteiro, M.C. Neuroprotective mechanisms of resveratrol in Alzheimer’s disease: Role of SIRT1. Oxid. Med. Cell. Longev. 2018, 2018, 8152373. [Google Scholar]
  121. Bhat, K.P.L.; Pezzuto, J.M. Cancer chemopreventive activity of resveratrol. Ann. N. Y. Acad. Sci. 2002, 957, 210–229. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Bastianetto, S.; Dumont, Y.; Duranton, A.; Vercauteren, F.; Breton, L.; Quirion, R. Protective Action of Resveratrol in Human Skin: Possible Involvement of Specific Receptor Binding Sites. PLoS ONE 2010, 5, e12935. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Giardina, S.; Michelotti, A.; Zavattini, G.; Finzi, S.; Ghisalberti, C.; Marzatico, F. Efficacy study in vitro: Assessment of the properties of resveratrol and resveratrol + N-acetyl-cysteine on proliferation and inhibition of collagen activity. Minerva Ginecol. 2010, 62, 195–201. [Google Scholar]
  124. Demidenko, Z.N.; Blagosklonny, M.V. At concentrations that inhibit mTOR, resveratrol suppresses cellular senescence. Cell Cycle 2009, 8, 1901–1904. [Google Scholar] [CrossRef] [Green Version]
  125. Xia, L.; Wang, X.X.; Hu, X.S.; Guo, X.G.; Shang, Y.P.; Chen, H.J.; Zeng, C.L.; Zhang, F.R.; Chen, J.Z. Resveratrol reduces endothelial progenitor cells senescence through augmentation of telomerase activity by Akt-dependent mechanisms. Br. J. Pharmacol. 2008, 155, 387–394. [Google Scholar] [CrossRef] [Green Version]
  126. Giovannelli, L.; Pitozzi, V.; Jacomelli, M.; Mulinacci, N.; Laurenzana, A.; Dolara, P.; Mocali, A. Protective effects of resveratrol against senescence-associated changes in cultured human fibroblasts. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2011, 66 A, 9–18. [Google Scholar] [CrossRef] [Green Version]
  127. López-Lluch, G.; Irusta, P.M.; Navas, P.; de Cabo, R. Mitochondrial biogenesis and healthy aging. Exp. Gerontol. 2008, 43, 813–819. [Google Scholar] [CrossRef] [Green Version]
  128. Lagouge, M.; Argmann, C.; Gerhart-Hines, Z.; Meziane, H.; Lerin, C.; Daussin, F.; Messadeq, N.; Milne, J.; Lambert, P.; Elliott, P.; et al. Resveratrol Improves Mitochondrial Function and Protects against Metabolic Disease by Activating SIRT1 and PGC-1α. Cell 2006, 127, 1109–1122. [Google Scholar] [CrossRef] [PubMed]
  129. Wang, T.; Li, Q.; Bi, K. Bioactive flavonoids in medicinal plants: Structure, activity and biological fate. Asian J. Pharm. Sci. 2018, 13, 12–23. [Google Scholar] [CrossRef] [PubMed]
  130. Antika, L.D.; Lee, E.J.; Kim, Y.H.; Kang, M.K.; Park, S.H.; Kim, D.Y.; Oh, H.; Choi, Y.J.; Kang, Y.H. Dietary phlorizin enhances osteoblastogenic bone formation through enhancing β-catenin activity via GSK-3β inhibition in a model of senile osteoporosis. J. Nutr. Biochem. 2017, 49, 42–52. [Google Scholar] [CrossRef] [PubMed]
  131. Makarova, E.; Górnaś, P.; Konrade, I.; Tirzite, D.; Cirule, H.; Gulbe, A.; Pugajeva, I.; Seglina, D.; Dambrova, M. Acute anti-hyperglycaemic effects of an unripe apple preparation containing phlorizin in healthy volunteers: A preliminary study. J. Sci. Food Agric. 2014, 95, 560–568. [Google Scholar] [CrossRef]
  132. Mela, D.J.; Cao, X.Z.; Dobriyal, R.; Fowler, M.I.; Lin, L.; Joshi, M.; Mulder, T.J.P.; Murray, P.G.; Peters, H.P.F.; Vermeer, M.A.; et al. The effect of 8 plant extracts and combinations on post-prandial blood glucose and insulin responses in healthy adults: A randomized controlled trial. Nutr. Metab. 2020, 17, 51. [Google Scholar] [CrossRef]
  133. Laiteerapong, N.; Karter, A.J.; Liu, J.Y.; Moffet, H.H.; Sudore, R.; Schillinger, D.; John, P.M.; Huang, E.S. Correlates of quality of life in older adults with diabetes: The diabetes & aging study. Diabetes Care 2011, 34, 1749–1753. [Google Scholar] [CrossRef] [Green Version]
  134. Ayaz, M.; Sadiq, A.; Junaid, M.; Ullah, F.; Ovais, M.; Ullah, I.; Ahmed, J.; Shahid, M. Flavonoids as prospective neuroprotectants and their therapeutic propensity in aging associated neurological disorders. Front. Aging Neurosci. 2019, 11, 155. [Google Scholar] [CrossRef] [Green Version]
  135. Zhang, Y.J.; Gan, R.Y.; Li, S.; Zhou, Y.; Li, A.N.; Xu, D.P.; Li, H.B.; Kitts, D.D. Antioxidant phytochemicals for the prevention and treatment of chronic diseases. Molecules 2015, 20, 21138–21156. [Google Scholar] [CrossRef]
  136. Kschonsek, J.; Wolfram, T.; Stöckl, A.; Böhm, V. Polyphenolic compounds analysis of old and new apple cultivars and contribution of polyphenolic profile to the in vitro antioxidant capacity. Antioxidants 2018, 7, 20. [Google Scholar] [CrossRef] [Green Version]
  137. Boyer, J.; Liu, R.H. Apple phytochemicals and their health benefits. Nutr. J. 2004, 3, 5. [Google Scholar] [CrossRef] [Green Version]
  138. Vafa, M.R.; Haghighatjoo, E.; Shidfar, F.; Afshari, S.; Gohari, M.R.; Ziaee, A. Effects of apple consumption on lipid profile of hyperlipidemic and overweight men. Int. J. Prev. Med. 2011, 2, 94–100. [Google Scholar] [PubMed]
  139. Peng, C.; Chan, H.Y.E.; Huang, Y.; Yu, H.; Chen, Z.Y. Apple polyphenols extend the mean lifespan of Drosophila melanogaster. J. Agric. Food Chem. 2011, 59, 2097–2106. [Google Scholar] [CrossRef] [PubMed]
  140. Cory, H.; Passarelli, S.; Szeto, J.; Tamez, M.; Mattei, J. The Role of Polyphenols in Human Health and Food Systems: A Mini-Review. Front. Nutr. 2018, 5, 87. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  141. Kalt, W.; Cassidy, A.; Howard, L.R.; Krikorian, R.; Stull, A.J.; Tremblay, F.; Zamora-Ros, R. Recent Research on the Health Benefits of Blueberries and Their Anthocyanins. Adv. Nutr. 2020, 11, 224–236. [Google Scholar] [CrossRef]
  142. Shukitt-Hale, B.; Thangthaeng, N.; Miller, M.G.; Poulose, S.M.; Carey, A.N.; Fisher, D.R. Blueberries Improve Neuroinflammation and Cognition differentially Depending on Individual Cognitive baseline Status. J. Gerontol. Ser. A Biol. Sci. Med. Sci. 2019, 74, 977–983. [Google Scholar] [CrossRef]
  143. Joseph, J.A.; Shukitt-Hale, B.; Casadesus, G. Reversing the deleterious effects of aging on neuronal communication and behavior: Beneficial properties of fruit polyphenolic compounds. Am. J. Clin. Nutr. 2005, 81, 313S–316S. [Google Scholar] [CrossRef]
  144. Galli, R.L.; Bielinski, D.F.; Szprengiel, A.; Shukitt-Hale, B.; Joseph, J.A. Blueberry supplemented diet reverses age-related decline in hippocampal HSP70 neuroprotection. Neurobiol. Aging 2006, 27, 344–350. [Google Scholar] [CrossRef]
  145. Goyarzu, P.; Malin, D.H.; Lau, F.C.; Taglialatela, G.; Moon, W.D.; Jennings, R.; Moy, E.; Moy, D.; Lippold, S.; Shukitt-Hale, B.; et al. Blueberry supplemented diet: Effects on object recognition memory and nuclear factor-kappa B levels in aged rats. Nutr. Neurosci. 2004, 7, 75–83. [Google Scholar] [CrossRef]
  146. Peng, C.; Zuo, Y.; Kwan, K.M.; Liang, Y.; Ma, K.Y.; Chan, H.Y.E.; Huang, Y.; Yu, H.; Chen, Z.Y. Blueberry extract prolongs lifespan of Drosophila melanogaster. Exp. Gerontol. 2012, 47, 170–178. [Google Scholar] [CrossRef]
  147. Su, Y.L.; Leung, L.K.; Huang, Y.; Chen, Z.Y. Stability of tea theaflavins and catechins. Food Chem. 2003, 83, 189–195. [Google Scholar] [CrossRef]
  148. Musial, C.; Kuban-Jankowska, A.; Gorska-Ponikowska, M. Beneficial properties of green tea catechins. Int. J. Mol. Sci. 2020, 21, 1744. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  149. Yan, Z.; Zhong, Y.; Duan, Y.; Chen, Q.; Li, F. Antioxidant mechanism of tea polyphenols and its impact on health benefits. Anim. Nutr. 2020, 6, 115–123. [Google Scholar] [CrossRef] [PubMed]
  150. Li, Y.M.; Chan, H.Y.E.; Huang, Y.; Chen, Z.Y. Green tea catechins upregulate Superoxide dismutase and catalase in fruit flies. Mol. Nutr. Food Res. 2007, 51, 546–554. [Google Scholar] [CrossRef] [PubMed]
  151. Oyetakinwhite, P.; Tribout, H.; Baron, E. Protective mechanisms of green tea polyphenols in skin. Oxid. Med. Cell. Longev. 2012, 2012, 560682. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Elmets, C.A.; Singh, D.; Tubesing, K.; Matsui, M.; Katiyar, S.; Mukhtar, H. Cutaneous photoprotection from ultraviolet injury by green tea polyphenols. J. Am. Acad. Dermatol. 2001, 44, 425–432. [Google Scholar] [CrossRef] [PubMed]
  153. Chiu, A.E.; Chan, J.L.; Kern, D.G.; Kohler, S.; Rehmus, W.E.; Kimball, A.B. Double-blinded, placebo-controlled trial of green tea extracts in the clinical and histologic appearance of photoaging skin. Dermatol. Surg. 2005, 31, 855–860. [Google Scholar] [CrossRef]
  154. Shaikh, R.; Pund, M.; Dawane, A.; Iliyas, S. Evaluation of anticancer, antioxidant, and possible anti-inflammatory properties of selected medicinal plants used in indian traditional medication. J. Tradit. Complement. Med. 2014, 4, 253–257. [Google Scholar] [CrossRef] [Green Version]
  155. Azevedo, J.; Fernandes, I.; Faria, A.; Oliveira, J.; Fernandes, A.; de Freitas, V.; Mateus, N. Antioxidant properties of anthocyanidins, anthocyanidin-3-glucosides and respective portisins. Food Chem. 2010, 119, 518–523. [Google Scholar] [CrossRef]
  156. Zuo, Y.; Peng, C.; Liang, Y.; Ma, K.Y.; Yu, H.; Edwin Chan, H.Y.; Chen, Z.Y. Black rice extract extends the lifespan of fruit flies. Food Funct. 2012, 3, 1271–1279. [Google Scholar] [CrossRef]
  157. Huang, J.J.; Zhao, S.M.; Jin, L.; Huang, L.J.; He, X.; Wei, Q. Anti-aging effect of black rice in subacute aging model mice. Chin. J. Clin. Rehabil. 2006, 10, 82–84. [Google Scholar]
  158. Institute of Medicine (US). Panel on Dietary Antioxidants and Related Compounds β-Carotene and Other Carotenoids. In Dietary Reference Intakes for Vitamin C, Vitamin E, Selenium, and Carotenoids; National Academies Press (US): Washington, DC, USA, 2000. [Google Scholar]
  159. Stahl, W.; Sies, H. β-Carotene and other carotenoids in protection from sunlight. Am. J. Clin. Nutr. 2012, 96, 1179S–1184S. [Google Scholar] [CrossRef] [PubMed]
  160. Pritwani, R.; Mathur, P. β-carotene Content of Some Commonly Consumed Vegetables and Fruits Available in Delhi, India. J. Nutr. Food Sci. 2017, 7, 5. [Google Scholar] [CrossRef]
  161. Jaswir, I.; Noviendri, D.; Hasrini, R.F.; Octavianti, F. Carotenoids: Sources, medicinal properties and their application in food and nutraceutical industry. J. Med. Plant Res. 2011, 5, 7119–7131. [Google Scholar] [CrossRef]
  162. Parrado, C.; Philips, N.; Gilaberte, Y.; Juarranz, A.; González, S. Oral photoprotection: Effective agents and potential candidates. Front. Med. 2018, 5, 188. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  163. Boccardi, V.; Arosio, B.; Cari, L.; Bastiani, P.; Scamosci, M.; Casati, M.; Ferri, E.; Bertagnoli, L.; Ciccone, S.; Rossi, P.D.; et al. Beta-carotene, telomerase activity and Alzheimer’s disease in old age subjects. Eur. J. Nutr. 2020, 59, 119–126. [Google Scholar] [CrossRef] [PubMed]
  164. The Alpha-Tocopherol Beta Carotene Cancer Prevention Study Group. The effect of vitamin e and beta carotene on the incidence of lung cancer and other cancers in male smokers. N. Engl. J. Med. 1994, 330, 1029–1035. [Google Scholar] [CrossRef]
  165. Evans, J.A.; Johnson, E.J. The role of phytonutrients in skin health. Nutrients 2010, 2, 903–928. [Google Scholar] [CrossRef]
  166. Ascenso, A.; Pedrosa, T.; Pinho, S.; Pinho, F.; De Oliveira, J.M.P.F.; Marques, H.C.; Oliveira, H.; Simões, S.; Santos, C. The Effect of Lycopene Preexposure on UV-B-Irradiated Human Keratinocytes. Oxid. Med. Cell. Longev. 2016, 2016. [Google Scholar] [CrossRef]
  167. Przybylska, S. Lycopene–A bioactive carotenoid offering multiple health benefits: A review. Int. J. Food Sci. Technol. 2020, 55, 11–32. [Google Scholar] [CrossRef]
  168. Darvin, M.E.; Sterry, W.; Lademann, J.; Vergou, T. The role of carotenoids in human skin. Molecules 2011, 16, 10491–10506. [Google Scholar] [CrossRef] [Green Version]
  169. Cheng, J.; Miller, B.; Balbuena, E.; Eroglu, A. Lycopene protects against smoking-induced lung cancer by inducing base excision repair. Antioxidants 2020, 9, 643. [Google Scholar] [CrossRef] [PubMed]
  170. Singh, B.; Singh, J.P.; Kaur, A.; Singh, N. Phenolic composition, antioxidant potential and health benefits of citrus peel. Food Res. Int. 2020, 132, 109114. [Google Scholar] [CrossRef] [PubMed]
  171. Carr, A.; Maggini, S. Vitamin C and Immune Function. Nutrients 2017, 9, 1211. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  172. Hemilä, H. Vitamin C and Infections. Nutrients 2017, 9, 339. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  173. Souyoul, S.A.; Saussy, K.P.; Lupo, M.P. Nutraceuticals: A Review. Dermatol. Ther. 2018, 8, 5–16. [Google Scholar] [CrossRef] [Green Version]
  174. Brickley, M.; Ives, R. Vitamin C Deficiency Scurvy. In The Bioarchaeology of Metabolic Bone Disease; Elsevier: Amsterdam, The Netherlands, 2008; pp. 1–74. [Google Scholar]
  175. Galli, F.; Azzi, A.; Birringer, M.; Cook-Mills, J.M.; Eggersdorfer, M.; Frank, J.; Cruciani, G.; Lorkowski, S.; Özer, N.K. Vitamin E: Emerging aspects and new directions. Free Radic. Biol. Med. 2017, 102, 16–36. [Google Scholar] [CrossRef]
  176. Sivakanesan, R. Antioxidants for health and longevity. In Molecular Basis and Emerging Strategies for Anti-Aging Interventions; Springer: Singapore, 2018; pp. 323–341. ISBN 9789811316999. [Google Scholar]
  177. Leonard, P.J.; Losowsky, M.S.; Pulvertaft, C.N. Vitamin-E Deficiency. Br. Med. J. 1966, 1, 1301–1302. [Google Scholar] [CrossRef]
  178. Keen, M.; Hassan, I. Vitamin E in dermatology. Indian Dermatol. Online J. 2016, 7, 311. [Google Scholar] [CrossRef]
  179. Fryer, M.J. Evidence for the photoprotective effects of vitamin E. Photochem. Photobiol. 1993, 58, 304–312. [Google Scholar] [CrossRef]
  180. Chan, A.C.; Tran, K.; Raynor, T.; Ganz, P.R.; Chow, C.K. Regeneration of vitamin E in human platelets-PubMed. J. Biol. Chem. 1991, 266, 17290–17295. [Google Scholar]
  181. Makrantonaki, E.; Zouboulis, C.C. Skin alterations and diseases in advanced age. Drug Discov. Today Dis. Mech. 2008, 5, e153–e162. [Google Scholar] [CrossRef]
  182. McVean, M.; Liebler, D.C. Prevention of DNA photodamage by vitamin E compounds and sunscreens: Roles of ultraviolet absorbance and cellular uptake. Mol. Carcinog. 1999, 24, 169–176. [Google Scholar] [CrossRef]
  183. Passi, S.; Morrone, A.; De Luca, C.; Picardo, M.; Ippolito, F. Blood levels of vitamin E, polyunsaturated fatty acids of phospholipids, lipoperoxides and glutathione peroxidase in patients affected with seborrheic dermatitis. J. Dermatol. Sci. 1991, 2, 171–178. [Google Scholar] [CrossRef]
  184. Ekanayake-Mudiyanselage, S.; Thiele, J. Sebaceous glands as transporters of vitamin E. Hautarzt 2006, 57, 291–296. [Google Scholar] [CrossRef]
  185. Eberlein-König, B.; Ring, J. Relevance of vitamins C and E in cutaneous photoprotection. J. Cosmet. Dermatol. 2005, 4, 4–9. [Google Scholar] [CrossRef]
  186. Shahidi, F. Nutraceuticals, functional foods and dietary supplements in health and disease. J. Food Drug Anal. 2012, 20, 226–230. [Google Scholar]
  187. Pem, D.; Jeewon, R. Fruit and vegetable intake: Benefits and progress of nutrition education interventions-narrative review article. Iran. J. Public Health 2015, 44, 1309–1321. [Google Scholar] [PubMed]
  188. Petruk, G.; Del Giudice, R.; Rigano, M.M.; Monti, D.M. Antioxidants from plants protect against skin photoaging. Oxid. Med. Cell. Longev. 2018, 2018, 1454936. [Google Scholar] [CrossRef] [Green Version]
  189. Cimino, F.; Cristani, M.; Saija, A.; Bonina, F.P.; Virgili, F. Protective effects of a red orange extract on UVB-induced damage in human keratinocytes. Biofactors 2007, 30, 129–138. [Google Scholar] [CrossRef] [PubMed]
  190. Fujii, T.; Wakaizumi, M.; Ikami, T.; Saito, M. Amla (Emblica officinalis Gaertn.) extract promotes procollagen production and inhibits matrix metalloproteinase-1 in human skin fibroblasts. J. Ethnopharmacol. 2008, 119, 53–57. [Google Scholar] [CrossRef] [PubMed]
  191. Adil, M.D.; Kaiser, P.; Satti, N.K.; Zargar, A.M.; Vishwakarma, R.A.; Tasduq, S.A. Effect of Emblica officinalis (fruit) against UVB-induced photo-aging in human skin fibroblasts. J. Ethnopharmacol. 2010, 132, 109–114. [Google Scholar] [CrossRef] [PubMed]
  192. Nema, N.K.; Maity, N.; Sarkar, B.; Mukherjee, P.K. Cucumis sativus fruit-potential antioxidant, anti-hyaluronidase, and anti-elastase agent. Arch. Dermatol. Res. 2011, 303, 247–252. [Google Scholar] [CrossRef] [PubMed]
  193. Cao, X.; Sun, Y.; Lin, Y.; Pan, Y.; Farooq, U.; Xiang, L.; Qi, J. Antiaging of cucurbitane glycosides from fruits of Momordica charantia L. Oxid. Med. Cell. Longev. 2018, 2018. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  194. Lourith, N.; Kanlayavattanakul, M.; Chaikul, P.; Chansriniyom, C.; Bunwatcharaphansakun, P. In vitro and cellular activities of the selected fruits residues for skin aging treatment. An. Acad. Bras. Cienc. 2017, 89, 577–589. [Google Scholar] [CrossRef] [Green Version]
  195. Apraj, V.D.; Pandita, N.S. Evaluation of skin anti-aging potential of Citrus reticulata blanco peel. Pharmacogn. Res. 2016, 8, 160–168. [Google Scholar] [CrossRef] [Green Version]
  196. Girsang, E.; Lister, I.N.E.; Ginting, C.N.; Khu, A.; Samin, B.; Widowati, W.; Wibowo, S.; Rizal, R. Chemical Constituents of Snake Fruit (Salacca zalacca (Gaert.) Voss) Peel and in silico Anti-aging Analysis. Mol. Cell. Biomed. Sci. 2019, 3, 122. [Google Scholar] [CrossRef]
  197. Kim, D.B.; Shin, G.H.; Kim, J.M.; Kim, Y.H.; Lee, J.H.; Lee, J.S.; Song, H.J.; Choe, S.Y.; Park, I.J.; Cho, J.H.; et al. Antioxidant and anti-ageing activities of citrus-based juice mixture. Food Chem. 2016, 194, 920–927. [Google Scholar] [CrossRef]
  198. Lee, M.J.; Jeong, N.H.; Jang, B.S. Antioxidative activity and antiaging effect of carrot glycoprotein. J. Ind. Eng. Chem. 2015, 25, 216–221. [Google Scholar] [CrossRef]
  199. Zemour, K.; Labdelli, A.; Adda, A.; Dellal, A.; Talou, T.; Merah, O. Phenol Content and Antioxidant and Antiaging Activity of Safflower Seed Oil (Carthamus Tinctorius L.). Cosmetics 2019, 6, 55. [Google Scholar] [CrossRef] [Green Version]
  200. Itoh, S.; Yamaguchi, M.; Shigeyama, K.; Sakaguchi, I. The Anti-Aging Potential of Extracts from Chaenomeles sinensis. Cosmetics 2019, 6, 21. [Google Scholar] [CrossRef] [Green Version]
  201. Foolad, N.; Vaughn, A.R.; Rybak, I.; Burney, W.A.; Chodur, G.M.; Newman, J.W.; Steinberg, F.M.; Sivamani, R.K. Prospective randomized controlled pilot study on the effects of almond consumption on skin lipids and wrinkles. Phyther. Res. 2019, 33, 3212–3217. [Google Scholar] [CrossRef] [PubMed]
  202. Wang, L.; Cui, J.; Jin, B.; Zhao, J.; Xu, H.; Lu, Z.; Li, W.; Li, X.; Li, L.; Liang, E.; et al. Multifeature analyses of vascular cambial cells reveal longevity mechanisms in old Ginkgo biloba trees. Proc. Natl. Acad. Sci. USA 2020, 117, 2201–2210. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  203. Shailaja, M.; Damodara Gowda, K.M.; Vishakh, K.; Suchetha Kumari, N. Anti-aging Role of Curcumin by Modulating the Inflammatory Markers in Albino Wistar Rats. J. Natl. Med. Assoc. 2017, 109, 9–13. [Google Scholar] [CrossRef] [PubMed]
  204. Shin, S.; Lee, J.A.; Son, D.; Park, D.; Jung, E. Anti-Skin-Aging Activity of a Standardized Extract from Panax ginseng Leaves In Vitro and In Human Volunteer. Cosmetics 2017, 4, 18. [Google Scholar] [CrossRef] [Green Version]
  205. Hwang, E.; Park, S.Y.; Yin, C.S.; Kim, H.T.; Kim, Y.M.; Yi, T.H. Antiaging effects of the mixture of Panax ginseng and Crataegus pinnatifida in human dermal fibroblasts and healthy human skin. J. Ginseng Res. 2017, 41, 69–77. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  206. Choi, H.R.; Nam, K.M.; Lee, H.S.; Yang, S.H.; Kim, Y.S.; Lee, J.; Date, A.; Toyama, K.; Park, K.C. Phlorizin, an active ingredient of eleutherococcus senticosus, increases proliferative potential of keratinocytes with inhibition of MiR135b and increased expression of type IV collagen. Oxid. Med. Cell. Longev. 2016, 2016. [Google Scholar] [CrossRef] [Green Version]
  207. Shoko, T.; Maharaj, V.J.; Naidoo, D.; Tselanyane, M.; Nthambeleni, R.; Khorombi, E.; Apostolides, Z. Anti-aging potential of extracts from Sclerocarya birrea (A. Rich.) Hochst and its chemical profiling by UPLC-Q-TOF-MS. BMC Complement. Altern. Med. 2018, 18, 54. [Google Scholar] [CrossRef]
  208. Shimizu, C.; Wakita, Y.; Inoue, T.; Hiramitsu, M.; Okada, M.; Mitani, Y.; Segawa, S.; Tsuchiya, Y.; Nabeshima, T. Effects of lifelong intake of lemon polyphenols on aging and intestinal microbiome in the senescence-accelerated mouse prone 1 (SAMP1). Sci. Rep. 2019, 9, 3671. [Google Scholar] [CrossRef] [Green Version]
  209. Lu, X.; Zhou, Y.; Wu, T.; Hao, L. Ameliorative effect of black rice anthocyanin on senescent mice induced by d-galactose. Food Funct. 2014, 5, 2892–2897. [Google Scholar] [CrossRef]
  210. Xiong, L.G.; Chen, Y.J.; Tong, J.W.; Gong, Y.S.; Huang, J.A.; Liu, Z.H. Epigallocatechin-3-gallate promotes healthy lifespan through mitohormesis during early-to-mid adulthood in Caenorhabditis elegans. Redox Biol. 2018, 14, 305–315. [Google Scholar] [CrossRef]
  211. Ratnasooriya, W.D.; Abeysekera, W.K.S.M.; Muthunayake, T.B.S.; Ratnasooriya, C.D.T. In vitro antiglycation and cross-link breaking activities of Sri Lankan low-grown orthodox orange pekoe grade black tea (Camellia sinensis L). Trop. J. Pharm. Res. 2014, 13, 567–571. [Google Scholar] [CrossRef] [Green Version]
  212. Yoo, D.S.; Min Jeon, J.; Jeong Choi, M.; Sang Lee, H.; Woo Cheon, J.; Hoi Kim, S.; Ryu Ju, S. Potential anti-wrinkle effects of m. spaientum l. leaves extract. BioEvolution 2015, 2, 56–61. [Google Scholar]
  213. Widowati, W.; Fauziah, N.; Herdiman, H.; Afni, M.; Afifah, E.; Kusuma, H.S.W.; Nufus, H.; Arumwardana, S.; Rihibiha, D.D. Antioxidant and anti aging assays of Oryza sativa extracts, vanillin and coumaric acid. J. Nat. Remedies 2016, 16, 88–99. [Google Scholar] [CrossRef] [Green Version]
Figure 1. Hallmarks contributing to ageing.
Figure 1. Hallmarks contributing to ageing.
Nutrients 12 03008 g001
Figure 2. Schematic representation of sources involved in the formation of free radicals and their association with the ageing process.
Figure 2. Schematic representation of sources involved in the formation of free radicals and their association with the ageing process.
Nutrients 12 03008 g002
Table 1. Fruits and vegetable extracts and their phytochemicals with antiageing effects.
Table 1. Fruits and vegetable extracts and their phytochemicals with antiageing effects.
Common NameScientific NameStudy Conducted RegionActive CompoundsBiological ActivitiesDose and DurationStudy TypeExperimental ModelsReferences
Sweet orangeCitrus sinensis L.ItalyAnthocyanins, flavanones, hydroxycinnamic acid and ascorbic acidNF-B and AP-1 translocation and procaspase-3 cleavage15 and 30 µg/mL for 7 hIn vitroHuman keratinocytes (HaCaT cell line)[189]
Indian gooseberryEmblica officinalis L.JapanAscorbic acid, gallic
Acid, elaeocarpusin
Inhibited type-I collagen collagenase, increase TIMP-1 level; Cellular proliferation inhibition and procollagen 1 protection against UVB-induced depletion by inhibition of UVB-induced MMP-1(0–40 g/mL) for 48 hIn vitroNB1RGB human skin fibroblasts[190]
Indian gooseberryEmblica officinalisIndiaAscorbic acidPromotion of procollagen content and inhibition of matrix metalloproteinase levels in skin fibroblast10–40 μg/mL for 24 h In vitroFibroblast cell line (HS68 cell)[191]
CucumberCucumis sativus L.IndiaAscorbic acidIn vitro inhibition of hyaluronidase, elastase and MMP-120.98 and 6.14 μg/mLIn vitro assayND[192]
Bitter gourdMomordica charantia L.ChinaResveratrolAnti-oxidative stress enhancement and UTH1, SKN7, SOD1 and SOD2 yeast gene expression regulation 1–3 μM for 12 hIn vitroYeast[193]
Litchi, Rambutan, TamarindLitchi chinensis; Nephelium lappaceum L.; Tamarindus indicaThailandFerulic acid, gallic acid, epigallocatechinSuppression of melanin production in B16F10 melanoma cells through inhibition of tyrosinase and TRP-2; effectiveness for elastase and collagenase inhibition0.05, 0.01 and 0.007 mg/mL for 72 hIn vitroHuman skin fibroblasts[194]
Mandarin orangeCitrus reticulata BlancoIndiaD-Limonene, n-Hexadecanoic acidCollagenase and elastase inhibition, anti-enzymatic activityNSIn vitro assayND[195]
Snake fruitSalacca zalacca (Gaert.) VossIndonesia Chlorogenic acidMMP-1 inhibitionNSIn silicoND[196]
Mandarin, GrapesCitrus sunki Hort. ex Tanaka, Citrus unshiu Marcov, Citrus sinensis Osbeck, Citrus reticulata Blanco and Vitis vinifera L.Republic of KoreaNarirutin, hesperidin, ascorbic acidIncrease in the expression levels of antioxidant enzymes; Reduction in skin thickness and wrinkle formation while elevating collagen level in an ultraviolet light B-exposed hairless mouse model33, 100, 300 mg/kg for 10 weeksIn vitro and in vivoCell culture and mice[197]
CarrotDaucus carota L.South KoreaCarrot glycoproteinNeutralization of reactive oxygen, cell membrane protection0.3, 0.5, 1 mg/mLIn vitroCell culture[198]
Safflower Seed OilCarthamus tinctoriusFrancePhenolInhibition in the collagenase assay, inhibition in the elastase assayNSIn vitro assayND[199]
Chinese quinceChaenomeles sinensisJapanβ-1,4-xyloglucanInhibition of the activity of dermal extracellular matrix proteases: Elastase and CollagenaseNSIn vitro assayND[200]
AlmondsPrunus dulcisCaliforniaα-tocopherolDecreased wrinkle severity in postmenopausal females340 kcal/day of almonds (58.9 g) for 16 weeksObservational studyHuman subjects[201]
Maidenhair treeGinkgo biloba L.Chinakaempferol 3-O-β-D-glucopyranoside, isorhamnetin-3-O-glucoside, myricetin, ginkgolide A, bilobalideInhibition of ROS and MMP-1 degradation in human dermal fibroblasts0.1, 0.2 mg/mL for 24 hIn vitroHuman dermal fibroblasts[202]
TurmericCurcuma longaIndiaCurcuminReduction in levels of C-reactive protein (CRP) an anti-ageing inflammatory marker200 mg and 400 mg of Curcumin/kg bodyweight for six monthsIn vivoRat[203]
Asian ginsengPanax ginsengKoreaGingenosidePromotion in collagen synthesis through the activation of transforming growth factor-β (TGF-β) in human skin fibroblast cells0.05% PGLE for eight weeksIn vitro and In vivoIn vitro and human volunteer[204]
Korean ginseng, mountain hawthornPanax ginseng Meyer and Crataegus pinnatifidaRepublic of KoreaGinsenosideProtective effect against UVB-exposed photo-ageing of the skin by regulating procollagen type 1 and MMP-1 expression in NHDFs100 μg/mL for 12 weeksIn vitro and Observational studyHuman dermal fibroblasts, healthy human skin [205]
LicoriceGlycyrrhiza glabra L.CroatiaGlabridin and isoliquiritigeninTyrosinase and elastase inhibitory activityNSIn vitro assayND[101]
Siberian ginseng, touch-me-notEleutherococcus senticosusRepublic of KoreaPhlorizinmiR135b suppression improves the microenvironment and increases the proliferative potential of basal epidermal cellsNSIn vitroHuman keratinocytes [206]
MarulaSclerocarya birreaSouth AfricaQuinic acid, catechin, epigallocatechin gallate and epicatechin gallateExhibited collagenase inhibition activities100, 200 μg/mLIn vitro assayND[207]
LemonCitrus limonJapan Eriocitrin (Polyphenols)Increase in ageing-related scores (e.g., periophthalmic lesions) and delay in locomotor atrophy 4 mL and 6 mL/day/mouseIn vivoMice[208]
Black riceZizania aqaticaChina Cyanidin -3-O-glucosideIncreases superoxide dismutase (SOD) and catalase (CAT), while decreases MDA and the activity of monoamine oxidase (MAO) 15, 30 and 60 mg/kgIn vivoMice[209]
Green teaCamellia sinensis L.China Epigallocatechin-3-gallateExtension of lifespan through mitohormesis50–300 μM for six daysIn vivoCaenorhabditis elegans [210]
Orange Pekoe black teaCamellia sinensis L.Sri LankaEpigallocatechin gallateInhibition of elastase activityNSIn vitro assayND[211]
BananaMusa spaientumKorea Corosolic acidInhibitory effects on MMPs activitiesNSIn vitro assayND[212]
RiceOryza sativaIndonesia Vanillin and coumaric acidElastase inhibitory activityNSIn vitro assayND[213]
NS: not specified; ND: not defined.

Share and Cite

MDPI and ACS Style

Dhanjal, D.S.; Bhardwaj, S.; Sharma, R.; Bhardwaj, K.; Kumar, D.; Chopra, C.; Nepovimova, E.; Singh, R.; Kuca, K. Plant Fortification of the Diet for Anti-Ageing Effects: A Review. Nutrients 2020, 12, 3008. https://0-doi-org.brum.beds.ac.uk/10.3390/nu12103008

AMA Style

Dhanjal DS, Bhardwaj S, Sharma R, Bhardwaj K, Kumar D, Chopra C, Nepovimova E, Singh R, Kuca K. Plant Fortification of the Diet for Anti-Ageing Effects: A Review. Nutrients. 2020; 12(10):3008. https://0-doi-org.brum.beds.ac.uk/10.3390/nu12103008

Chicago/Turabian Style

Dhanjal, Daljeet Singh, Sonali Bhardwaj, Ruchi Sharma, Kanchan Bhardwaj, Dinesh Kumar, Chirag Chopra, Eugenie Nepovimova, Reena Singh, and Kamil Kuca. 2020. "Plant Fortification of the Diet for Anti-Ageing Effects: A Review" Nutrients 12, no. 10: 3008. https://0-doi-org.brum.beds.ac.uk/10.3390/nu12103008

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop