Next Article in Journal
The Role of Exchange Energy in Modeling Core-Electron Binding Energies of Strongly Polar Bonds
Previous Article in Journal
Revealing Serotonin Derivatives in Safflower Seed Meal as Potential Anti-Ulcerative Colitis Drugs: In Vitro and Computational Evidence
Previous Article in Special Issue
p-Coumaroyl Amides from the Plant Kingdom: A Comprehensive Review of Natural Sources, Biosynthesis, and Biological Activities
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Increasing Life Expectancy with Plant Polyphenols: Lessons from the Mediterranean and Japanese Diets

by
Marco Fiore
1,*,
Anton B. Tonchev
2,3,
Ruzha Z. Pancheva
3,4,
Tetsumori Yamashima
5,
Sabrina Venditti
6,
Giampiero Ferraguti
7,† and
Sergio Terracina
7,†
1
Institute of Biochemistry and Cell Biology (IBBC-CNR), Department of Sensory Organs, Sapienza University of Rome, 00185 Rome, Italy
2
Department of Anatomy and Cell Biology, Faculty of Medicine, Medical University, 9000 Varna, Bulgaria
3
NutriLect Study Group, Project No. BG-RRP-2.004-0009-C02, Medical University, 9000 Varna, Bulgaria
4
Group on Early Childhood Intervention, Department of Hygiene and Epidemiology, Research Institute, Faculty of Public Health, Medical University, 9002 Varna, Bulgaria
5
Department of Psychiatry and Behavioral Sciences, Kanazawa University Graduate School of Medical Science, Kanazawa 920-8641, Japan
6
Department of Biology and Biotechnologies “Charles Darwin”, Sapienza University of Rome, 00185 Rome, Italy
7
Department of Experimental Medicine, Sapienza University of Rome, 00185 Rome, Italy
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Molecules 2025, 30(13), 2888; https://doi.org/10.3390/molecules30132888
Submission received: 11 June 2025 / Revised: 27 June 2025 / Accepted: 30 June 2025 / Published: 7 July 2025
(This article belongs to the Special Issue Bioactive Phenolic and Polyphenolic Compounds, 3rd Edition)

Abstract

Plant polyphenols have emerged as potent bioactive molecules that can modulate key cellular pathways associated with aging and chronic disorders. The Mediterranean diet and the traditional Japanese style of life are rich in polyphenol-containing foods and beverages, and epidemiological evidence links these dietary patterns to increased longevity and reduced morbidity. This narrative review examines the chemical description of plant polyphenols, their mechanisms of action, including anti-inflammatory, antioxidant, and hormetic effects, and how supplementation or a diet rich in these compounds may provide further life extension. We discuss the major classes of polyphenols present in the Mediterranean dietary pattern (e.g., resveratrol and hydroxytyrosol) and in the Japanese diet (e.g., epigallocatechin gallate and soy isoflavones), comparing their biological behaviors and cooperative effects on metabolic, cardiovascular, and neurodegenerative conditions. We also examine a few preclinical and clinical studies that explain the beneficial impact of these chemicals on aging-associated biomarkers. Furthermore, both dietary habits are characterized by low consumption of processed foods and sugary carbonated drinks and reduced utilization of deep-frying with linoleic acid-rich oils, a practice that reduces the formation of harmful lipid peroxidation products, notably 4-hydroxynonenal, known to be implicated in accelerating the aging process. The Mediterranean dietary pattern is also characterized by a low/moderate daily consumption of wine, mainly red wine. This work debates emerging evidence addressing issues of bioavailability, dosage optimization, and formulation technologies for polyphenol supplementation, also comparing differences and similarities with the vegan and vegetarian diets. We also explore how these chemicals could modulate epigenetic modifications that affect gene expression patterns pertinent to health and aging. In conclusion, we aim to show a consolidated framework for the comprehension of how plant polyphenols could be utilized in nutritional strategies for potentiating life expectancy while stimulating further research on nutraceutical development.

1. Introduction

The contemporary search for increased life expectancy and enhanced quality of life has strengthened interest in nutritional interventions that could regulate aging processes at the molecular level. Aging is a multifactorial event influenced by genetic, environmental, and dietary factors [1,2,3]. A growing body of evidence suggests that chronic inflammation and oxidative stress elicit the decline of cellular function, finally leading to age-related disorders such as cardiovascular diseases, neurodegenerative conditions, and certain types of tumors [4,5,6,7,8,9,10]. Polyphenols—a diverse group of naturally occurring chemicals found in vegetables, fruits, wines, teas, and extra-virgin olive oil—show a compelling role in mitigating these deleterious processes [11,12,13,14,15].
The Mediterranean diet, characterized by an elevated intake of plant-based foods, nuts, extra-virgin olive oil, and moderate wine consumption, has recurrently been associated with enhanced cardiovascular health and increased lifespan [16,17,18,19,20,21,22]. Similarly, traditional Japanese dietary patterns, which accentuate soy products, vegetables, seaweed, and green tea, have been related to longevity and low occurrences of dementia and metabolic conditions [23,24,25,26,27,28,29]. Although the two dietary styles originate from divergent culinary and cultural traditions, a common feature is the profusion of polyphenolic chemicals that seem to confer protective effects.
In this narrative review, we discuss the hypothesis that supplementation with plant polyphenols, whether across selected dietary interventions or concentrated nutraceutical formulations, could positively impact life expectancy. We first aim to review the chemistry and biochemistry of polyphenols and describe their mechanistic functions, including free radical scavenging, the modulation of inflammatory paths, and the stimulation of cellular defense mechanisms. We then explore detailed analyses of the polyphenol-rich Mediterranean and Japanese diets to demonstrate how long-term adherence to these dietary patterns could correlate with reduced mortality rates. By integrating data from clinical trials, epidemiological studies, and preclinical research, we postulate an integrated assessment of the potential mechanisms underlying polyphenols’ longevity-promoting effects. We also aim to discuss future directions in this field, including approaches for potentiated bioavailability and innovation in supplementation procedures.
The objective of this review is twofold: to provide a comprehensive indication of the current scientific comprehension of plant polyphenols, their protein substrates that protect against oxidation injury, and their role in counteracting age-related decline, and to gain insights into how lessons from two of the world’s healthiest dietary lifestyles can be utilized to improve longevity. Indeed, a better comprehension of these mechanisms may incentivize the development of targeted nutritional approaches aimed at optimizing health extent and reducing the weight of chronic disorders linked to aging.

2. Materials and Methods

In April 2025, an accurate literature search was carried out to identify crucial studies across multiple databases, including Scopus, PubMed, and Web of Science, to perform this narrative review. Papers were recruited using keywords such as “health”, “polyphenols”, “Mediterranean diet”, “Japanese diet”, “inflammation”, “neurodegeneration”, “cardiovascular conditions”, “metabolic disorders”, and “oxidative stress” without a limit on publication year. Delimited inclusion criteria were as follows: (1) English-language articles, (2) original studies on the above-mentioned keywords, and among polyphenols we considered in the discussion resveratrol, epigallocatechin gallate (EGCG), quercetin, hydroxytyrosol/oleuropein/tyrosol (olive polyphenols). Letters, editorials, and case reports were not included. Studies following these criteria were further evaluated, and pertinent findings were extracted from each work.

3. Plant Polyphenols: Chemistry and Mechanisms of Action

3.1. Classification and Chemical Description

Plant polyphenols are a vast family of secondary metabolites and are generally divided according to their chemical structure into numerous major classes [30,31,32]:
-
Flavonoids [33,34]: This group includes flavanols (e.g., EGCG), flavones (e.g., luteolin), flavonols (e.g., quercetin and kaempferol), flavanones, neoflavonoids, isoflavones (e.g., Genistein and Daidzein), and anthocyanins.
-
Phenolic acids [35]: Such as ferulic acid and caffeic acid.
-
Stilbenoid [36,37]: With resveratrol being one of the most famous.
-
Lignans [38]: Discovered in whole grains and numerous seeds.
-
Tannins [39,40]: A class of astringent, polyphenolic biomolecules such as gallic acid.
These chemicals are characterized by one or several hydroxyl groups on aromatic (phenolic) rings and show noteworthy redox properties. Their ability to donate electrons permits them to be potent free radical scavengers, so rapidly acting against oxidative damage at the physiological level [11,41,42,43].

3.2. Antioxidant and Anti-Inflammatory Mechanisms

Plant polyphenols elicit their biological action mainly through three related mechanisms:
  • Reactive Oxygen Species (ROS) Direct Scavenging [44,45,46,47,48]: By counteracting free radicals such as hydroxyl radicals and superoxide anions, plant polyphenols may neutralize oxidative damage directly to DNA, lipids, and proteins.
  • Fine Regulation of Cellular Signaling: Many plant polyphenols modulate key intracellular pathways, involving the activation of the nuclear factor erythroid 2-related factor 2 (Nrf2), which has a crucial role in the expression of antioxidant peptides [49,50,51,52]. Furthermore, they reduce the nuclear factor kappa-light-chain-enhancer of the activated B cell (NF-κB) path, thus decreasing the activity of pro-inflammatory cytokines [11,53,54,55,56].
  • Hormetic Actions: Hormesis is a two-step dose–response association with an environmental agent, whereby low-dose amounts may have a positive effect and high-dose quantities could be functionally inhibitory or toxic [57,58]. Indeed, at low doses, polyphenols may elicit a minor stress response aimed at enhancing cellular resilience, which may modulate stress resistance and regulate endogenous repair mechanisms [59,60,61,62].
These processes represent the biochemical basis for the protective actions of polyphenols against the development of age-related conditions, supporting the hypothesis that regular polyphenol consumption might slow down aging progression.

3.3. Polyphenols’ Role in Cellular Metabolism and Longevity

At the biomolecular level, polyphenols also regulate pathways with important roles in mitochondrial biogenesis, autophagy, and apoptosis [63,64,65,66]. EGCG, a main catechin discovered in green tea, has been shown to modulate signaling pathways associated with inflammation and cell survival [67,68,69]. Other polyphenols impact the above pathways by means of epigenetic mechanisms. For example, resveratrol, a well-known stilbene, can modulate the actions of sirtuins [70,71,72], a class of chromatin-modifying enzymes that control cellular responses to stress and have been associated with potentiated lifespan in numerous organisms [73,74,75].
Moreover, polyphenols may induce epigenetic modifications with a synergistic action with microRNAs [76,77,78,79], thus regulating gene expression patterns related to aging. This multifaceted approach allows polyphenols to subtly remodel cellular homeostasis and participate in a healthier and longer lifespan (Figure 1).

4. The Mediterranean Diet: A Polyphenol-Rich Nutritional Paradigm

4.1. Dietary Pattern and Polyphenol Sources

The Mediterranean diet is renowned for its emphasis on vegetables, fresh fruits, whole grains, legumes, nuts, and, particularly, extra-virgin olive oil [3,17,80,81,82]. Moderate consumption of wine (mainly red wine)—rich in resveratrol and other polyphenols—is another distinct characteristic [15,83,84,85,86,87,88,89,90]. Indeed, a wide variety of plant foods deliver an extensive selection of polyphenols, each with distinctive biological activities and chemical properties.
Key polyphenolic compounds found in the Mediterranean diet include the following:
-
Hydroxytyrosol, Tyrosol, and Oleuropein: Mostly found in olive leaves and extra-virgin olive oil, these chemicals display robust antioxidant and anti-inflammatory capabilities [85,91,92,93,94].
-
Resveratrol: Found in the skin of grapes, blueberries, raspberries, mulberries, and peanuts [95,96,97], resveratrol has been associated with the activation of longevity-related pathways such as the Phosphoinositide 3-Kinase/Protein Kinase B (PI3K/Akt), Sirtuin 1 (SIRT1), and AMP-Activated Protein Kinase (AMPK) pathways [98,99,100,101] while inhibiting MTOR (mammalian target of rapamycin), a protein kinase that plays a crucial role in cell growth, proliferation, and metabolism [102].
-
Flavonoids: These are abundant in vegetables and fruits (e.g., quercetin in radish leaves, radicchio, tomatoes, and red onions; catechins in tea) that participate in the total antioxidative shape of the diet [103,104,105,106,107].

4.2. Health Outcomes Correlated with the Mediterranean Diet

Several epidemiological studies have associated the Mediterranean diet with lower percentages of certain tumors, decreased incidences of metabolic syndrome, and enhanced cardiovascular conditions [108,109,110,111,112]. For example, the PREDIMED (Prevención con Dieta Mediterránea) trial showed convincing evidence that a Mediterranean dietary pattern supplemented with encouraged low-fat food items, nuts, or extra-virgin olive oil lowers the risk of the main cardiovascular disorders [113,114]. These findings, combined with observed improved lipid profiles and reductions in inflammatory markers, strongly imply that a diet enriched in high plant polyphenol content may have a key role in potentiating protective actions [115].
The interaction between different polyphenols emerges as particularly significant. When assumed as part of a whole diet, these chemicals may have additive or even accumulating effects on health, due to their capability to concurrently target numerous signaling cascades. This multifaceted interplay of dietary elements configures the Mediterranean dietary pattern as a nutritional style that effectively counteracts inflammation and oxidative stress, both of which are key factors in age-associated diseases.

4.3. Further Elements from Mediterranean Diet Studies

Other biomolecular studies have shown that polyphenol-enriched extracts from Mediterranean foods can potentiate (i) the production of neurotrophins (namely brain-derived neurotrophic factor, BDNF, and nerve growth factor, NGF) [116,117,118] and (ii) endothelial function [119,120,121,122,123], and (iii) stimulate nitric oxide (NO) release—a crucial cause in preserving vascular health [124,125,126,127].
Moreover, these compounds support the cell transition from a pro-inflammatory up to an anti-inflammatory condition by regulating cytokine shapes, suppressing the expression of genes involved in pro-inflammatory molecules’ release, such as toll-like receptor (TLR), and decreasing the levels of C-reactive protein (CRP) [11,128,129,130]. It should be noted that several investigations have correlated such biomolecular improvements with decreased arterial rigidity and elevated overall cardiovascular functioning, which may in turn contribute to longer life expectancy [131,132].
By diminishing oxidative injury and reducing inflammatory responses at a systemic level, the observance of the Mediterranean dietary pattern could offer a molecular environment leading to better healthy aging. Elucidating the substrate of polyphenols and its protection would be crucial for both decreasing cell degeneration/death and increasing life expectancy. This dietary practice could not only delay the beginning of age-related conditions but also increase the life quality in the elderly—a dichotomy that emphasizes its potentiality as a prototype for nutritional interventions designed to extend the lifespan.

5. Japanese Dietary Patterns and the Role of Polyphenols in Longevity

5.1. Traditional Japanese Diet and Its Polyphenol Profile

Japan is recognized for having one of the world’s highest life expectancies [133]. This fact could be somewhat attributable to the traditional Japanese dietary practice, which is certainly elevated in marine/soy-based proteins, low in saturated fats, and rich in vegetables and beverages containing high amounts of polyphenols, mostly green tea [25,28,134]. Green tea, which is high in EGCG, represents a major contributor to the dietary polyphenol burden in Japan. Other important components encompass soy-based elements (e.g., miso and tofu, containing genistein and daidzein) that offer isoflavones and a wide plethora of sea vegetables that provide distinctive phenolic chemicals [135,136].

5.2. Epidemiological Proof of Longevity

The intake of green tea has been recurrently associated with decreased incidences of certain tumors, cardiovascular disorders, and neurodegenerative conditions. Epidemiological studies in Japan display that regular green tea consumers show lower mortality rates compared to non-drinkers—an advantage attributed not only to EGCG but also to the general phytochemical setting of Japanese dietary practice [137,138,139]. Isoflavones from soy, for instance, might contribute to enhanced bone density and decreased risk of hormone-dependent tumors, while seaweed-derived molecules have been related to lower blood cholesterol presence and potentiated thyroid function [140,141,142,143].
Several cohort investigations have clearly shown relationships between elevated urinary polyphenol concentrations, mirroring a dietary style abundant in these chemicals, and positive health outcomes. These findings suggest that the growing effects of daily assumption of modest amounts of different polyphenols can cause a biologically valuable “polyphenol signature” that could decelerate aging events and potentiate cellular defense mechanisms [144,145,146].

5.3. Biomolecular Mechanisms of the Japanese Dietary Schedule

Investigations on the biomolecular actions of green tea polyphenols have clarified some pathways pertinent to longevity. Indeed, EGCG has been demonstrated to facilitate mitochondrial function, activate critical metabolic controllers (e.g., AMPK), and decrease cellular aging [147,148,149,150]. EGCG also activates the Keap1–Nrf2/ARE axis and up-regulating phase II antioxidant enzymes and glutathione synthesis, restoring redox homeostasis in neurons and hepatocytes [147,148,149,150,151,152,153]. Furthermore, EGCG blocks NF-κB nuclear translocation, reducing pro-inflammatory cytokine (e.g., TNF-α and IL-6) production in the vascular endothelium and microglia [154,155,156,157,158,159].
The soy isoflavones (i.e., genistein and daidzein) of the Japanese dietary patterns possess slight estrogen-like properties that support the maintenance of skeletal and cardiovascular health, mostly in postmenopausal women [151,152,153]. Furthermore, soy isoflavones might (i) modulate PI3K/Akt/eNOS (phosphatidylinositol 3-kinase/protein kinase B/endothelial nitric oxide synthase) signaling in endothelial cells, eliciting vasodilation and inhibiting vascular smooth muscle cell proliferation [160,161]; (ii) inhibit NADPH (nicotinamide adenine dinucleotide phosphate) oxidase isoforms and down-regulate reactive oxygen species in cardiomyocytes [162]; and (iii) stimulate epigenetic actions by regulating DNA methyltransferase, histone deacetylase, and microRNA expression (e.g., miR-155 and miR-21) known to contribute to anti-inflammatory and anti-fibrotic gene expression profiles [163,164,165,166].
In addition to these straight outcomes, the traditional lifestyle of Japanese people—embracing communal eating patterns and mild physical activity, and placing a cultural importance on balance—underlines the beneficial influence of dietary polyphenols by decreasing stress and endorsing general well-being [167,168]. This multifactorial method not only reduces the inflammatory load but also elicits a complex metabolic setting that supports longevity.

6. Relationship Between Mediterranean and Japanese Polyphenol Contents

6.1. Major Polyphenolic Presence Characterizing the Japanese/Mediterranean Diet Profiles

A qualitative evaluation of the two dietary schedules reveals different but complementary polyphenol paradigms. Table 1 highlights some of the key polyphenolic chemicals prevalent in each dietary pattern.
Table 1. Major polyphenol elements found in the Mediterranean and Japanese diets. This table shows that while each dietary paradigm provides a distinct composition, the essential similarity in the types of polyphenols—such as flavonoids and catechins—indicates a convergent evolution dealing with nutrition that boosts cellular health and antioxidant defenses.
Table 1. Major polyphenol elements found in the Mediterranean and Japanese diets. This table shows that while each dietary paradigm provides a distinct composition, the essential similarity in the types of polyphenols—such as flavonoids and catechins—indicates a convergent evolution dealing with nutrition that boosts cellular health and antioxidant defenses.
Polyphenolic ClassMediterranean DietJapanese Diet
StilbenesResveratrol (from red wine, grapes, and berries) [169]
Phenolic AlcoholsHydroxytyrosol, tyrosol, and oleuropein (from extra-virgin olive oil and olive leaves) [81]
FlavonoidsQuercetin and catechins (from fruits/vegetables) [170]Catechins (EGCG from green tea) [171]
IsoflavonesGenistein and daidzein (from soy products) [172]
Other Phenolic ChemicalsVarious minor polyphenols (nuts and legumes) [173]Unique compounds from seaweeds and mushrooms [174]

6.2. Cooperative Effects and Nutrient Connections

The holistic advantages derived from the Mediterranean and Japanese diets are rarely caused by a single chemical in isolation. Instead, their effectiveness seems to be established by combinatory interplay among several nutrients. For instance, the consumption of extra-virgin olive oil in the Mediterranean diet not only provides hydroxytyrosol/tyrosol/oleuropein but also supplies monounsaturated fats that contribute to effective energy metabolism and cell membrane integrity [175,176,177,178]. Likewise, the Japanese diet’s emphasis on soy products and green tea originates from a biochemical environment in which the distinct actions of isoflavones and EGCG strengthen each other to regulate pathways dealing with stress-responsive signaling [135,179] (Figure 2).
Previous investigations in nutritional biochemistry have shown that the collective antioxidant ability of a mixed polyphenol-enriched diet may be significantly superior to the sum of its components [154,155,156]. These data strongly sustain the hypothesis that a wide dietary paradigm, rather than single-compound supplementation, could be the ideal strategy for exploiting the full range of polyphenol benefits.

6.3. The Impact on Aging Critical Biomarkers

Both diets have been correlated with encouraging modifications in biomarkers associated with inflammatory conditions and oxidative stress. Individuals strictly adherent to the Mediterranean diet often display a lower plasma presence of oxidized low-density lipoprotein (ox-LDL), decreased inflammatory cytokine concentrations, and better endothelial functionality [157,158,159]. Comparably, regular drinkers of green tea in Japan show a lower presence of pro-inflammatory biomarkers, potentiated mitochondrial efficacy, and elevation in protective genes associated with longevity [180,181,182].
In both circumstances, the long-term consumption of polyphenol-rich foods seems to participate in reduced cellular senescence and enhanced resilience against metabolic problems. Such molecular biomarkers are progressively being documented in the scientific literature as prognosticators of increased quality of life and life expectancy in the elderly.

7. Preclinical and Clinical Findings

7.1. Preclinical Studies

Animal models [183,184,185,186,187,188] and in vitro studies [189,190,191,192,193,194] have shown significant information on the mechanisms by which polyphenol supplementation may potentiate life expectancy. In rodent studies, resveratrol administration improved mitochondrial function and induced a retardation at the beginning of age-related decline [195,196,197,198,199]. Likewise, supplementation of EGCG in different studies has revealed reductions in biomarkers of oxidative stress [200,201,202,203] and augmented neuronal survival in animal models of neurodegeneration [204,205,206,207,208].
Mouse studies also disclosed a role played by NGF and BDNF in modulating oxidative stress and brain functionality following olive polyphenol and/or resveratrol administration [82,116,117,118]. These data not only emphasize the critical role played by polyphenols as anti-aging components but also support the hypothesis describing how dietary patterns rich in these antioxidant chemicals could finely modulate cellular homeostasis.

7.2. Clinical Studies and Epidemiological Findings

Several human investigations have tried to translate these preclinical data into clinical evidence. For instance, randomized controlled trials (e.g., PREDIMED and CARDIOPREV) investigating the Mediterranean dietary pattern have constantly shown improvements in cardiovascular risk elements—such as arterial stiffness, circulating inflammatory biomarkers, and blood lipid presence—known to be crucial longevity determinants [22,209,210,211]. Observational studies carried out in Japan (e.g., Japan Public Health Center-based Prospective Study) have analogously discovered an inverse association between green tea drinking and all-cause morbidity and mortality [137,212,213].
Moreover, according to biomarker investigations, it has been shown that individuals with a greater baseline presence of blood polyphenols display a reduced incidence of neurodegenerative conditions and a decreased occurrence of metabolic syndrome [105,214,215,216]. Based on these considerations, the human findings indicate that plant polyphenol supplementation—whether via rigid dietary approaches or as supplemented nutraceuticals—can elicit significant effects on the biomolecular pathways that trigger aging and chronic conditions.
It should be noted, however, that unpredictability in polyphenol metabolism and bioavailability among individuals is a noteworthy challenge in nutrition. Clinical results are influenced by many factors, such as genetic polymorphisms regulating metabolic enzymes, the food substrate in which polyphenols are carried, and the gut microbiota configuration. Identifying these biases could be an essential tool for planning future studies and for the eventual personalization of polyphenol-based dietary patterns (Table 2).

8. The Impact of Sugary Carbonated Drinks, Processed Foods and Cooking Practices on Hydroxynonenal Formation on Aging Acceleration

An unacknowledged dietary issue contributing to aging is the presence of reactive aldehydes, particularly 4-hydroxynonenal (HNE) [229,230,231,232,233]. HNE originates during the deep-frying of linoleic acid-rich cooking oils; under high-temperature conditions, the unsaturated bonds in linoleic acid oxidize, leading to lipid peroxidation and HNE formation [229]. This toxic element can make adducts with nucleic acids, proteins, and other cellular constituents, thus eliciting oxidative stress, cellular abnormalities, neurodegeneration, and the progression of age-related conditions [229,230,231,232,233,234].
Intriguingly, after consecutive injections of synthetic HNE in monkeys in which the serum concentration reached that observed in humans in their 60s, significant brain, pancreas, and liver damage were observed. HNE elevation, combined with age-dependent ischemia in humans, may overactivate μ-calpain, which can cleave the lysosomal membrane stabilizer, Hsp70.1, especially after its specific oxidation injury, i.e., carbonylation by ROS. The ‘calpain–cathepsin hypothesis’ formulated by Yamashima et al. postulates calpain-mediated damage of lysosomal limiting membranes and subsequent cathepsin release [229,230,231,232,233]. This represents a central cascade for programmed cell death in diverse organs and leads to the occurrence of lifestyle-related diseases.
Another modern dietary concern is the extensive consumption of sugary carbonated or not beverages. These drinks are not only characterized by highly refined sugars but might also be highly processed. Indeed, regular consumption of drinks enriched in sugar can lead to severe pathological conditions such as obesity, type 2 diabetes mellitus, and cardiovascular disorders, and the formation of several glycation end products (AGEs) [235,236,237,238,239]. Indeed, AGEs, in turn, participate in the elevation of oxidative stress [240,241,242] and have been associated with the elevation of lipid peroxidation processes that may further increase HNE presence [243,244,245]. The metabolic abnormalities induced by high sugar intake, such as chronic inflammation and insulin resistance, can potentiate the harmful actions of HNE, increasing the risk of chronic degenerative disorders and accelerating cellular senescence.
A further modern dietary concern is the abuse of ultraprocessed and processed foods. They are characterized by their wide industrial modification, which dramatically modifies both their nutritional features and chemical structures compared to normal or marginally processed foods [246,247,248]. These edulis elements are generally overloaded with high amounts of refined sugars and unhealthy fats, mainly linoleic acid from specific seed oils, and a wide collection of artificial additives, emulsifiers, and preservatives. It should be noted that the industrial processes may consist of high-temperature cooking and chemical management, facilitating broad lipid peroxidation with the formation of HNE. Accordingly, the systematic consumption of ultraprocessed foods might compromise metabolic homeostasis, potentiating oxidative stress and stimulating unhealthy epigenetic modifications that further accelerate several degenerative processes associated with aging.
In this regard, both the Mediterranean diet and the Japanese style of life express a common culinary legacy: an inclination to consume fresh, minimally processed foods, limiting frequent deep-frying procedures. In these dietary paradigms, oil cooking techniques such as light sautéing, boiling, or steaming are preferred over deep-frying, which reduce the exposure to the high temperatures that stimulate linoleic acid oxidation. Furthermore, the selection of cooking oils further characterizes these dietary patterns. For example, extra-virgin olive oil, commonly utilized in Mediterranean cuisine, displays a higher oxidative solidity and an inferior linoleic acid presence compared to many industrial seed oils [249,250]. Likewise, traditional Japanese cuisine accentuates the utilization of fresh components and seasonal vegetables according to methods that do not produce great lipid peroxidation.
The take-home message of these practices is a decreased in vivo load of HNE and other damaging lipid peroxidation components. By limiting sugary carbonated drinks, processed foods, and deep-frying of linoleic acid-rich oils, these dietary practices might not only decrease the generation of toxic metabolites but also contribute to preserving cellular functions, reducing chronic inflammatory processes. Accordingly, the reduction in HNE accumulation may be a key point in the decreased incidence of age-related conditions enhancing longevity detected in individuals following these nutritional lifestyles.

9. The Dual Janus-Faced Role of Alcohol Consumption Within These Dietary Patterns

Alcohol drinking, in any form (e.g., wine, beer, and spirits), possesses a distinctive position in the context of dietary paradigms, including in the Mediterranean area due to early cultural motivations, exhibiting a dualistic nature that is extremely dependent on the type and quantity of intake. Indeed, in the Mediterranean diet, moderate consumption—mainly in the form of red wine—has been correlated with different cardiometabolic benefits [20,80,251,252]. However, chronic or excessive alcohol drinking is established to induce a wide spectrum of adverse health effects, from cancer to accelerated aging [253,254].

9.1. Toxic Effects of Alcohol Abuse

The abuse of alcohol is associated with several toxic outcomes [255,256]. At any dose, alcohol is metabolized into acetaldehyde, an extremely reactive chemical capable of triggering protein and DNA adducts [257,258,259,260,261,262,263]. This phenomenon not only contributes to liver disorders, such as alcoholic hepatitis, liver tumors, and cirrhosis, but also elevates the risk of cardiovascular diseases, certain types of cancer, and neurodegeneration. The chronic consumption of large amounts of alcohol affects cellular homeostasis, promoting systemic inflammatory processes and inducing cellular senescence—a plethora of effects that are well documented to decrease life expectancy, affecting the quality of life in the elderly.

9.2. Alcohol Drinking During Pregnancy: Fetal Alcohol Spectrum Disorders

Fetal alcohol spectrum disorders (FASDs) are a group of pathological conditions that can happen in an individual who was exposed to ethanol during pregnancy [264,265,266,267,268,269]. In Western countries, FASD affects between 1 and 4 in 20 newborns but is probably misdiagnosed and underdiagnosed [264,270]. The more severe form of FASD is fetal alcohol syndrome (FAS). The main mandatory signs for FAS diagnosis include growth deficiency or failure to grow, congenital malformation of the lips, short palpebral fissure lengths, and nervous system damage associated with functional impairments [271,272,273,274,275]. Quite surprisingly, in a mouse model of FASD carried out with red wine or alcohol per se supplementation, some of the deleterious effects of alcohol were counteracted when ethanol was administered as red wine, showing a sort of protection due to the natural presence of polyphenols in the wine [276,277,278,279]. Furthermore, resveratrol restored Nrf2 levels and prevented ethanol-induced toxic effects in the cerebellum of a rodent model of FASD [280], while curcumin did not lead to protective effects in growing zebrafish embryos [281]. It should also be noted that paternal alcohol exposure during mating may elicit outcomes in the offspring/newborns comparable to those observed for FASD [282,283,284].

9.3. Effects of Moderate Alcohol Consumption

Intriguingly, different investigations have shown that moderate consumption of alcohol—particularly wine in the Mediterranean area—seems to provide health benefits that could contribute to longevity [20,285,286]. Indeed, red wine is a natural source of polyphenolic chemicals such as resveratrol, anthocyanins, and quercetin [20,55,287], biomolecules known to induce antioxidant, cardioprotective, and anti-inflammatory effects. Epidemiological findings from Mediterranean countries, although debated, indicate that low-to-moderate red wine intake (circa one glass per day for non-gestating women and up to two for men) is associated with better endothelial function, a decrease in circulating inflammatory biomarkers, and potentiated lipid patterns [288,289,290,291]. In these conditions, the helpful actions of these polyphenols seem to counteract the potentially damaging metabolic effects of alcohol, assisting in the maintenance of a favorable oxidative and inflammatory equilibrium that could lead to healthy aging.

9.4. Balancing the Dual Effects: A Context-Dependent Paradigm

The “Janus face” of alcohol drinking emphasizes the crucial importance of the circumstance of its consumption. Within the frame of a dietary pattern characterized by low sugar intake and low ultraprocessed/processed food intake but abundant in protective natural polyphenols—as typified by the Mediterranean or Japanese diet—the moderate presence of good-quality wine could be integrated as a secondary element of a healthful lifestyle. Nevertheless, it is important to underscore that the health benefits identified in such populations rely upon a strict adherence to low/moderate consumption indications. According to the knowledge dealing with alcohol abuse, the toxic effects of consumption prevail, moving the balance toward inflammation, oxidative stress, and cellular injury, all of which contribute to accelerated aging and several disorders’ development.

10. The Close Relationship Between the Mediterranean/Japanese Diets and the Vegan/Vegetarian Diets

While the Mediterranean and Japanese diets both highlight polyphenol-rich whole foods, firm vegan and lacto-ovo vegetarian profiles also provide high total polyphenol content, often matching or exceeding those of other plant-related regimens, but with different characteristics [292,293,294,295,296,297,298,299,300]. For instance, a soy-centric vegetarian diet may provide more isoflavone content (genistein and daidzein) than the Japanese diet. Further, fruit-heavy vegan regimens could offer elevated levels of anthocyanins and flavonols.
A strict diet rich in nuts, whole grains, legumes, and a wide array of vegetables could strengthen both the vegan and vegetarian polyphenol assortment. However, the absence of wine stilbenes (resveratrol), extra-virgin olive oil polyphenols, and marine phlorotannins could limit their coverage of redox and anti-inflammatory properties.
The fat content further distinguishes these dietary patterns. Vegan and vegetarian diets are generally based on polyunsaturated-rich oils (e.g., walnut and flaxseed) and nut butters, which can modify micelle formation and the intestinal uptake of lipophilic polyphenols compared to the monounsaturated-rich extra virgin olive oil of the Mediterranean dietary pattern. Thus, different lipid transporters could modulate both the plasma content and tissue distribution of crucial antioxidant chemicals.
However, it should be noted that epidemiological data has established the health benefits among all four plant-based approaches. Several cohort investigations link vegan and vegetarian diets to lower all-cause mortality [292,293,294,295,296,297,298,299,300], including reduced incidence of cardiovascular disorders by improving the glycemic responses, findings comparable to those observed in Mediterranean and Japanese regimens. Thus, these parallel clinical outcomes, such as preserved bone density in soy consumers or the cardiometabolic advantages of Mediterranean dietary polyphenols, show that each regimen acts through sometimes non-overlapping arrangements of bioactive compounds.
It should also be considered that rigorous diets based only on vegetables could require subtle attention to micronutrients, particularly vitamin B12, iron, zinc, and iodine, chemicals known to play fine roles in the modulation of immune ability, mitochondrial function, and redox balance. Indeed, the unbalanced status of these co-factors could impair polyphenol action by disrupting antioxidant enzyme actions and eliciting low-grade inflammation.

11. Future Assessments and Proposals

11.1. Improving Polyphenol Administration

Investigations regarding the formulation of polyphenol supplements are promptly proceeding. Novel encapsulation methods, including liposomal carriage and nano-emulsion systems, are being established to better improve the bioavailability of these chemicals [301,302,303]. Indeed, many industrial plans have been developed to disclose effective dosages and combinations that optimize the biological effectiveness of polyphenols while minimizing metabolism-related outcomes.
The adoption of a unifying approach that blends whole-food dietary paradigms with scientifically established nutraceuticals could offer a double strategy for potentiating health span and life expectancy. Indeed, individuals incapable of strictly adhering to a Mediterranean or Japanese dietary pattern due to several reasons, including geographic, cultural, or socioeconomic explanations, might take advantage of directed supplementation formulas that resemble the polyphenol characterization of these diets.

11.2. Investigating Metabolic Contents and Bioavailability

The pharmacokinetic qualities of polyphenols change widely and features such as food processing, chemical organization, and interaction with other dietary elements severely influence polyphenols’ absorption and systemic spreading. Recent discoveries in gut microbiome research and metabolomics are beginning to show how these elements modulate the efficacy of polyphenols [304,305,306]. Forthcoming clinical studies should amalgamate these indications, utilizing the stratification of the microbiota composition and/or potential genetic markers to predict which individuals are expected to take advantage of targeted polyphenol supplementation.

11.3. Integration with Lifestyle and Cultural Practices

It is also important to distinguish that the benefits of the Japanese and Mediterranean dietary patterns go beyond their polyphenol concentration. Both lifestyles encourage social cohesion, physical activity, and stress decrease—all aspects that encourage healthy aging. Consequently, the adoption of polyphenol-rich dietary paradigms should be noticed as a small part of a holistic strategy to potentiate longevity. Politicians, legislators, and professionals in nutritional sciences should contribute to developing national and international guidelines that combine dietary recommendations with a salubrious style of life to optimize public health.

11.4. Limitations and Emerging Directions

Numerous critical research open questions remain. In particular, (i) large-scale, long-term controlled randomized trials with strong endpoints (i.e., age-related morbidity and mortality) are necessary to clearly indicate the causal correlation between polyphenol supplementation and potentiated life expectancy; (ii) the disclosure of consistent biomarkers of polyphenol efficiency, including, for example, HNE, could address early practices of intervention to shape personalized strategies; and (iii) since the field of nutrigenomics is in rapid progress, the investigation of putative gene–diet interactions may eventually expose new levels of information and opportunities to add other bricks in the regulation of aging by dietary polyphenols.

12. Discussion and Conclusions

The evidence presented in this work emphasizes the encouraging role of plant polyphenols and other plant-derived compounds such as phytosteroids (e.g., 20-Hydroxyecdysone, a natural substance found in spinach [307]) in promoting longevity. Both the traditional Japanese lifestyle and the Mediterranean diet offer convincing real-world examples of how a polyphenol-rich nutritional paradigm can regulate key biological pathways—such as inflammation, oxidative stress, and mitochondrial functionality—that play crucial roles in aging and chronic disorders. However, the exact molecular interactions continue to be an active area of research, but preclinical and clinical findings support the hypothesis that regular intake of diverse polyphenols may be a practicable approach for extending health span and enhancing life expectancy.
According to the intricacies of polyphenol bioavailability and metabolic interindividual disparities, future research should focus on integrating novel delivery systems with personalized nutrition methods. Similarly, a wider public health emphasis on whole-diet paradigms balancing bioactive chemicals with a non-stressing lifestyle and cultural practices seems necessary. These indications aim for a future in which the supplementation of plant polyphenols through a personalized dietary pattern or as optimized nutraceuticals should develop different preventive medicine milestones as a basic part of our strategy to counteract age-related disorders.
In conclusion, the convergence of findings from several disciplines, ranging from epidemiology to molecular biology, supports the hypothesis that plant polyphenols can be considered pivotal elements in the mission for longevity. It is conceivable that lifestyle-related diseases such Alzheimer’s disease, type 2 diabetes, and nonalcoholic steatohepatitis are parallel pathological phenomena induced by HNE accumulation. Therefore, the prevention of Hsp70.1 carbonylation by plant polyphenols may contribute to inhibiting the occurrence of diverse lifestyle-related conditions. The lessons emerging from Mediterranean and Japanese nutritional paradigms not only reinforce our comprehension of the aging process but could also disclose innovative interventions that might ultimately improve the duration and quality of human life.

Author Contributions

Conceptualization, M.F., A.B.T., R.Z.P., T.Y., S.V. and G.F.; methodology M.F., S.V., S.T. and G.F.; validation, M.F., S.T. and G.F.; investigation, M.F., S.T. and G.F.; resources, M.F., S.T. and G.F.; data curation, M.F., A.B.T., R.Z.P., T.Y., S.V., S.T. and G.F.; writing—original draft preparation, M.F.; writing—review and editing, M.F., A.B.T., R.Z.P., T.Y., S.V., S.T. and G.F.; visualization, M.F., A.B.T., R.Z.P., T.Y., S.V., S.T. and G.F.; supervision, M.F., A.B.T., R.Z.P., T.Y., S.V., S.T. and G.F. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Acknowledgments

The authors thank the IBBC-CNR and the Sapienza University of Rome in Rome, Italy.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Sharifi-Rad, M.; Anil Kumar, N.V.; Zucca, P.; Varoni, E.M.; Dini, L.; Panzarini, E.; Rajkovic, J.; Tsouh Fokou, P.V.; Azzini, E.; Peluso, I.; et al. Lifestyle, Oxidative Stress, and Antioxidants: Back and Forth in the Pathophysiology of Chronic Diseases. Front. Physiol. 2020, 11, 694. [Google Scholar] [CrossRef] [PubMed]
  2. Joubert, C.; Chainay, H. Aging brain: The effect of combined cognitive and physical training on cognition as compared to cognitive and physical training alone—A systematic review. Clin. Interv. Aging 2018, 13, 1267–1301. [Google Scholar] [CrossRef] [PubMed]
  3. Petrella, C.; Di Certo, M.G.; Gabanella, F.; Barbato, C.; Ceci, F.M.; Greco, A.; Ralli, M.; Polimeni, A.; Angeloni, A.; Severini1, C.; et al. Mediterranean Diet, Brain and Muscle: Olive Polyphenols and Resveratrol Protection in Neurodegenerative and Neuromuscular Disorders. Curr. Med. Chem. 2021, 28, 7595–7613. [Google Scholar] [CrossRef] [PubMed]
  4. Pandey, S.N.; Agrawal, N.; Moglad, E.; Priya, G.P.; Srivastava, M.; Chennakesavulu, K.; Mohanty, B.; Arya, R.; Kazmi, I.; Alzarea, S.I.; et al. CHIP and aging: A key regulator of proteostasis and cellular senescence. Biogerontology 2025, 26, 104. [Google Scholar] [CrossRef]
  5. Ungvari, A.; Nyúl-Tóth, Á.; Patai, R.; Csik, B.; Gulej, R.; Nagy, D.; Shanmugarama, S.; Benyó, Z.; Kiss, T.; Ungvari, Z.; et al. Cerebromicrovascular senescence in vascular cognitive impairment: Does accelerated microvascular aging accompany atherosclerosis? GeroScience 2025. ahead of print. [Google Scholar] [CrossRef]
  6. Beaver, L.M.; Jamieson, P.E.; Wong, C.P.; Hosseinikia, M.; Stevens, J.F.; Ho, E. Promotion of Healthy Aging Through the Nexus of Gut Microbiota and Dietary Phytochemicals. Adv. Nutr. 2025, 16, 100376. [Google Scholar] [CrossRef]
  7. Pandics, T.; Major, D.; Fazekas-Pongor, V.; Szarvas, Z.; Peterfi, A.; Mukli, P.; Gulej, R.; Ungvari, A.; Fekete, M.; Tompa, A.; et al. Exposome and unhealthy aging: Environmental drivers from air pollution to occupational exposures. GeroScience 2023, 45, 3381–3408. [Google Scholar] [CrossRef]
  8. Coradduzza, D.; Congiargiu, A.; Chen, Z.; Zinellu, A.; Carru, C.; Medici, S. Ferroptosis and Senescence: A Systematic Review. Int. J. Mol. Sci. 2023, 24, 3658. [Google Scholar] [CrossRef]
  9. Wang, K.; Dong, Y.; Liu, J.; Qian, L.; Wang, T.; Gao, X.; Wang, K.; Zhou, L. Effects of REDOX in Regulating and Treatment of Metabolic and Inflammatory Cardiovascular Diseases. Oxid. Med. Cell. Longev. 2020, 2020, 5860356. [Google Scholar] [CrossRef]
  10. Barnes, P.J. Mechanisms of development of multimorbidity in the elderly. Eur. Respir. J. 2015, 45, 790–806. [Google Scholar] [CrossRef]
  11. Yahfoufi, N.; Alsadi, N.; Jambi, M.; Matar, C. The immunomodulatory and anti-inflammatory role of polyphenols. Nutrients 2018, 10, 1618. [Google Scholar] [CrossRef]
  12. Truong, V.L.; Jun, M.; Jeong, W.S. Role of resveratrol in regulation of cellular defense systems against oxidative stress. BioFactors 2018, 44, 36–49. [Google Scholar] [CrossRef] [PubMed]
  13. Zhang, L.X.; Li, C.X.; Kakar, M.U.; Khan, M.S.; Wu, P.F.; Amir, R.M.; Dai, D.F.; Naveed, M.; Li, Q.Y.; Saeed, M.; et al. Resveratrol (RV): A pharmacological review and call for further research. Biomed. Pharmacother. 2021, 143, 112164. [Google Scholar] [CrossRef] [PubMed]
  14. Ashraf, M.V.; Khan, S.; Misri, S.; Gaira, K.S.; Rawat, S.; Rawat, B.; Khan, M.A.H.; Shah, A.A.; Asgher, M.; Ahmad, S. High-Altitude Medicinal Plants as Promising Source of Phytochemical Antioxidants to Combat Lifestyle-Associated Oxidative Stress-Induced Disorders. Pharmaceuticals 2024, 17, 975. [Google Scholar] [CrossRef]
  15. Fiore, M.; Messina, M.P.; Petrella, C.; D’Angelo, A.; Greco, A.; Ralli, M.; Ferraguti, G.; Tarani, L.; Vitali, M.; Ceccanti, M. Antioxidant properties of plant polyphenols in the counteraction of alcohol-abuse induced damage: Impact on the Mediterranean diet. J. Funct. Foods 2020, 71, 104012. [Google Scholar] [CrossRef]
  16. Raederstorff, D. Antioxidant activity of olive polyphenols in humans: A review. Int. J. Vitam. Nutr. Res. 2009, 79, 152–165. [Google Scholar] [CrossRef]
  17. Trichopoulou, A. From research to education: The Greek experience. Nutrition 2000, 16, 528–531. [Google Scholar] [CrossRef]
  18. Rajaram, S.; Jones, J.; Lee, G.J. Plant-based dietary patterns, plant foods, and age-related cognitive decline. Adv. Nutr. 2019, 10, 422–436. [Google Scholar] [CrossRef]
  19. Casamenti, F.; Stefani, M. Olive polyphenols: New promising agents to combat aging-associated neurodegeneration. Expert Rev. Neurother. 2017, 17, 345–358. [Google Scholar] [CrossRef]
  20. Giacosa, A.; Barale, R.; Bavaresco, L.; Faliva, M.A.; Gerbi, V.; La Vecchia, C.; Negri, E.; Opizzi, A.; Perna, S.; Pezzotti, M.; et al. Mediterranean Way of Drinking and Longevity. Crit. Rev. Food Sci. Nutr. 2016, 56, 635–640. [Google Scholar] [CrossRef]
  21. Estruch, R.; Martínez-González, M.A.; Corella, D.; Salas-Salvadó, J.; Ruiz-Gutiérrez, V.; Covas, M.I.; Fiol, M.; Gómez-Gracia, E.; López-Sabater, M.C.; Vinyoles, E.; et al. Effects of a Mediterranean-style diet on cardiovascular risk factors: A randomized trial. Ann. Intern. Med. 2006, 145, 1–11. [Google Scholar] [CrossRef]
  22. Finicelli, M.; Di Salle, A.; Galderisi, U.; Peluso, G. The Mediterranean Diet: An Update of the Clinical Trials. Nutrients 2022, 14, 2956. [Google Scholar] [CrossRef] [PubMed]
  23. Suzuki, N.; Goto, Y.; Ota, H.; Kito, K.; Mano, F.; Joo, E.; Ikeda, K.; Inagaki, N.; Nakayama, T. Characteristics of the japanese diet described in epidemiologic publications: A qualitative systematic review. J. Nutr. Sci. Vitaminol. 2018, 64, 129–137. [Google Scholar] [CrossRef] [PubMed]
  24. Shirota, M.; Watanabe, N.; Suzuki, M.; Kobori, M. Japanese-Style Diet and Cardiovascular Disease Mortality: A Systematic Review and Meta-Analysis of Prospective Cohort Studies. Nutrients 2022, 14, 2008. [Google Scholar] [CrossRef]
  25. Matsuyama, S.; Shimazu, T.; Tomata, Y.; Zhang, S.; Abe, S.; Lu, Y.; Tsuji, I. Japanese Diet and Mortality, Disability, and Dementia: Evidence from the Ohsaki Cohort Study. Nutrients 2022, 14, 2034. [Google Scholar] [CrossRef]
  26. Vitale, E. An Optimal Dietary Pattern for Healthy Longevity: Scoping Differencing Review between the Mediterranean and the Japanese Diet. Endocr. Metab. Immune Disord.—Drug Targets 2023, 24, 1711–1720. [Google Scholar] [CrossRef]
  27. Moriki, D.; Koumpagioti, D.; Francino, M.P.; Rufián-Henares, J.Á.; Kalogiannis, M.; Priftis, K.N.; Douros, K. How Different Are the Influences of Mediterranean and Japanese Diets on the Gut Microbiome? Endocr. Metab. Immune Disord.—Drug Targets 2024, 24, 1733–1745. [Google Scholar] [CrossRef]
  28. Damigou, E.; Kosti, R.I.; Downs, S.M.; Naumovski, N.; Panagiotakos, D. Comparing The Mediterranean and The Japanese Dietary Pattern in Relation to Longevity—A Narrative Review. Endocr. Metab. Immune Disord.—Drug Targets 2024, 24, 1746–1755. [Google Scholar] [CrossRef]
  29. Mensah, E.O.; Danyo, E.K.; Asase, R.V. Exploring the effect of different diet types on ageing and age-related diseases. Nutrition 2025, 129, 112596. [Google Scholar] [CrossRef]
  30. Quideau, S.; Deffieux, D.; Douat-Casassus, C.; Pouységu, L. Plant polyphenols: Chemical properties, biological activities, and synthesis. Angew. Chem.—Int. Ed. 2011, 50, 586–621. [Google Scholar] [CrossRef]
  31. Nonaka, G. Isolation and structure elucidation of tannins. Pure Appl. Chem. 1989, 61, 357–360. [Google Scholar] [CrossRef]
  32. Manach, C.; Scalbert, A.; Morand, C.; Rémésy, C.; Jiménez, L. Polyphenols: Food sources and bioavailability. Am. J. Clin. Nutr. 2004, 79, 727–747. [Google Scholar] [CrossRef] [PubMed]
  33. De Souza Farias, S.A.; Da Costa, K.S.; Martins, J.B.L. Analysis of Conformational, Structural, Magnetic, and Electronic Properties Related to Antioxidant Activity: Revisiting Flavan, Anthocyanidin, Flavanone, Flavonol, Isoflavone, Flavone, and Flavan-3-ol. ACS Omega 2021, 6, 8908–8918. [Google Scholar] [CrossRef] [PubMed]
  34. Ververidis, F.; Trantas, E.; Douglas, C.; Vollmer, G.; Kretzschmar, G.; Panopoulos, N. Biotechnology of flavonoids and other phenylpropanoid-derived natural products. Part I: Chemical diversity, impacts on plant biology and human health. Biotechnol. J. 2007, 2, 1214–1234. [Google Scholar] [CrossRef] [PubMed]
  35. Heleno, S.A.; Martins, A.; Queiroz, M.J.R.P.; Ferreira, I.C.F.R. Bioactivity of phenolic acids: Metabolites versus parent compounds: A review. Food Chem. 2015, 173, 501–513. [Google Scholar] [CrossRef]
  36. Valletta, A.; Iozia, L.M.; Leonelli, F. Impact of environmental factors on stilbene biosynthesis. Plants 2021, 10, 90. [Google Scholar] [CrossRef]
  37. Dubrovina, A.S.; Kiselev, K.V. Regulation of stilbene biosynthesis in plants. Planta 2017, 246, 597–623. [Google Scholar] [CrossRef]
  38. Saleem, M.; Hyoung, J.K.; Ali, M.S.; Yong, S.L. An update on bioactive plant lignans. Nat. Prod. Rep. 2005, 22, 696–716. [Google Scholar] [CrossRef]
  39. Hemingway, R.W.; Karchesy, J.J. Chemistry and Significance of Condensed Tannins; Springer Science & Business Media: Berlin/Heidelberg, Germany, 2012; ISBN 1468475118. [Google Scholar]
  40. Scalbert, A. Quantitative Methods for the Estimation of Tannins in Plant Tissues. In Plant Polyphenols; Springer: Berlin/Heidelberg, Germany, 1992; pp. 259–280. [Google Scholar]
  41. Rivas, F.; Poblete-Aro, C.; Pando, M.E.; Allel, M.J.; Fernandez, V.; Soto, A.; Nova, P.; Garcia-Diaz, D. Effects of Polyphenols in Aging and Neurodegeneration Associated with Oxidative Stress. Curr. Med. Chem. 2021, 29, 1045–1060. [Google Scholar] [CrossRef]
  42. Uddin, M.S.; Al Mamun, A.; Kabir, M.T.; Ahmad, J.; Jeandet, P.; Sarwar, M.S.; Ashraf, G.M.; Aleya, L. Neuroprotective role of polyphenols against oxidative stress-mediated neurodegeneration. Eur. J. Pharmacol. 2020, 886, 173412. [Google Scholar] [CrossRef]
  43. Hussain, T.; Tan, B.; Yin, Y.; Blachier, F.; Tossou, M.C.B.B.; Rahu, N. Oxidative Stress and Inflammation: What Polyphenols Can Do for Us? Oxid. Med. Cell. Longev. 2016, 2016, 7432797. [Google Scholar] [CrossRef]
  44. Ahmadinejad, F.; Møller, S.G.; Hashemzadeh-Chaleshtori, M.; Bidkhori, G.; Jami, M.S. Molecular mechanisms behind free radical scavengers function against oxidative stress. Antioxidants 2017, 6, 51. [Google Scholar] [CrossRef] [PubMed]
  45. Rolt, A.; Cox, L.S. Structural basis of the anti-ageing effects of polyphenolics: Mitigation of oxidative stress. BMC Chem. 2020, 14, 50. [Google Scholar] [CrossRef] [PubMed]
  46. Godos, J.; Romano, G.L.; Laudani, S.; Gozzo, L.; Guerrera, I.; Dominguez Azpíroz, I.; Martínez Diaz, R.; Quiles, J.L.; Battino, M.; Drago, F.; et al. Flavan-3-ols and Vascular Health: Clinical Evidence and Mechanisms of Action. Nutrients 2024, 16, 2471. [Google Scholar] [CrossRef] [PubMed]
  47. Boonyong, C.; Vardhanabhuti, N.; Jianmongkol, S. Natural polyphenols prevent indomethacin-induced and diclofenac-induced Caco-2 cell death by reducing endoplasmic reticulum stress regardless of their direct reactive oxygen species scavenging capacity. J. Pharm. Pharmacol. 2020, 72, 583–591. [Google Scholar] [CrossRef]
  48. Caruso, F.; Incerpi, S.; Pedersen, J.; Belli, S.; Kaur, S.; Rossi, M. Aromatic Polyphenol π-π Interactions with Superoxide Radicals Contribute to Radical Scavenging and Can Make Polyphenols Mimic Superoxide Dismutase Activity. Curr. Issues Mol. Biol. 2022, 44, 5209–5220. [Google Scholar] [CrossRef]
  49. Calabrese, V.; Cornelius, C.; Dinkova-Kostova, A.T.; Calabrese, E.J. Vitagenes, cellular stress response, and acetylcarnitine: Relevance to hormesis. BioFactors 2009, 35, 146–160. [Google Scholar] [CrossRef]
  50. Divyajanani, S.; Harithpriya, K.; Ganesan, K.; Ramkumar, K.M. Dietary Polyphenols Remodel DNA Methylation Patterns of NRF2 in Chronic Disease. Nutrients 2023, 15, 3347. [Google Scholar] [CrossRef]
  51. Bhullar, K.S.; Rupasinghe, H.P.V. Polyphenols: Multipotent therapeutic agents in neurodegenerative diseases. Oxid. Med. Cell. Longev. 2013, 2013, 891748. [Google Scholar] [CrossRef]
  52. Plauth, A.; Geikowski, A.; Cichon, S.; Wowro, S.J.; Liedgens, L.; Rousseau, M.; Weidner, C.; Fuhr, L.; Kliem, M.; Jenkins, G.; et al. Hormetic shifting of redox environment by pro-oxidative resveratrol protects cells against stress. Free Radic. Biol. Med. 2016, 99, 608–622. [Google Scholar] [CrossRef]
  53. Kurhaluk, N.; Kamiński, P.; Bilski, R.; Kołodziejska, R.; Woźniak, A.; Tkaczenko, H. Role of Antioxidants in Modulating the Microbiota–Gut–Brain Axis and Their Impact on Neurodegenerative Diseases. Int. J. Mol. Sci. 2025, 26, 3658. [Google Scholar] [CrossRef]
  54. Young, A.K.; Kim, G.Y.; Park, K.Y.; Yung, H.C. Resveratrol inhibits nitric oxide and prostaglandin E2 production by lipopolysaccharide-activated C6 microglia. J. Med. Food 2007, 10, 218–224. [Google Scholar] [CrossRef]
  55. Scoditti, E.; Calabriso, N.; Massaro, M.; Pellegrino, M.; Storelli, C.; Martines, G.; De Caterina, R.; Carluccio, M.A. Mediterranean diet polyphenols reduce inflammatory angiogenesis through MMP-9 and COX-2 inhibition in human vascular endothelial cells: A potentially protective mechanism in atherosclerotic vascular disease and cancer. Arch. Biochem. Biophys. 2012, 527, 81–89. [Google Scholar] [CrossRef] [PubMed]
  56. Sajadimajd, S.; Bahramsoltani, R.; Iranpanah, A.; Kumar Patra, J.; Das, G.; Gouda, S.; Rahimi, R.; Rezaeiamiri, E.; Cao, H.; Giampieri, F.; et al. Advances on Natural Polyphenols as Anticancer Agents for Skin Cancer. Pharmacol. Res. 2020, 151, 104584. [Google Scholar] [CrossRef]
  57. Bhakta-Guha, D.; Efferth, T. Hormesis: Decoding two sides of the same coin. Pharmaceuticals 2015, 8, 865–883. [Google Scholar] [CrossRef] [PubMed]
  58. Calabrese, E.J. Hormesis: A fundamental concept in biology. Microb. Cell 2014, 1, 145–149. [Google Scholar] [CrossRef]
  59. Murakami, A. Impact of hormesis to deepen our understanding of the mechanisms underlying the bioactivities of polyphenols. Curr. Opin. Biotechnol. 2024, 86, 103074. [Google Scholar] [CrossRef]
  60. Calabrese, V.; Wenzel, U.; Piccoli, T.; Jacob, U.M.; Nicolosi, L.; Fazzolari, G.; Failla, G.; Fritsch, T.; Osakabe, N.; Calabrese, E.J. Investigating hormesis, aging, and neurodegeneration: From bench to clinics. Open Med. 2024, 19, 20240986. [Google Scholar] [CrossRef]
  61. Calabrese, V.; Osakabe, N.; Siracusa, R.; Modafferi, S.; Di Paola, R.; Cuzzocrea, S.; Jacob, U.M.; Fritsch, T.; Abdelhameed, A.S.; Rashan, L.; et al. Transgenerational hormesis in healthy aging and antiaging medicine from bench to clinics: Role of food components. Mech. Ageing Dev. 2024, 220, 111960. [Google Scholar] [CrossRef]
  62. Chirumbolo, S. Hormesis, resveratrol and plant-derived polyphenols: Some comments. Hum. Exp. Toxicol. 2011, 30, 2027–2030. [Google Scholar] [CrossRef]
  63. de Oliveira, M.R.; Jardim, F.R.; Setzer, W.N.; Nabavi, S.M.; Nabavi, S.F. Curcumin, mitochondrial biogenesis, and mitophagy: Exploring recent data and indicating future needs. Biotechnol. Adv. 2016, 34, 813–826. [Google Scholar] [CrossRef]
  64. Meira Martins, L.A.; Vieira, M.Q.; Ilha, M.; de Vasconcelos, M.; Biehl, H.B.; Lima, D.B.; Schein, V.; Barbé-Tuana, F.; Borojevic, R.; Guma, F.C.R. The Interplay Between Apoptosis, Mitophagy and Mitochondrial Biogenesis Induced by Resveratrol Can Determine Activated Hepatic Stellate Cells Death or Survival. Cell Biochem. Biophys. 2015, 71, 657–672. [Google Scholar] [CrossRef] [PubMed]
  65. Kumar, S.; Chhabra, V.; Shenoy, S.; Daksh, R.; Ravichandiran, V.; Swamy, R.S.; Kumar, N. Role of Flavonoids in Modulation of Mitochondria Dynamics during Oxidative Stress. Mini-Rev. Med. Chem. 2023, 24, 908–919. [Google Scholar] [CrossRef] [PubMed]
  66. Naoi, M.; Wu, Y.; Shamoto-Nagai, M.; Maruyama, W. Mitochondria in Neuroprotection by Phytochemicals: Bioactive Polyphenols Modulate Mitochondrial Apoptosis System, Function and Structure. Int. J. Mol. Sci. 2019, 20, 2451. [Google Scholar] [CrossRef]
  67. Pervin, M.; Unno, K.; Ohishi, T.; Tanabe, H.; Miyoshi, N.; Nakamura, Y. Beneficial Effects of Green Tea Catechins on Neurodegenerative Diseases. Molecules 2018, 23, 1297. [Google Scholar] [CrossRef]
  68. Sharma, R.; Bhate, L.; Agrawal, Y.; Aspatwar, A. Advanced nutraceutical approaches to Parkinson’s disease: Bridging nutrition and neuroprotection. Nutr. Neurosci. 2025, 1–17. [Google Scholar] [CrossRef]
  69. Zhang, S.; Zhu, Q.; Chen, J.Y.; OuYang, D.; Lu, J.H. The pharmacological activity of epigallocatechin-3-gallate (EGCG) on Alzheimer’s disease animal model: A systematic review. Phytomedicine 2020, 79, 153316. [Google Scholar] [CrossRef]
  70. Sinclair, D.A. Toward a unified theory of caloric restriction and longevity regulation. Mech. Ageing Dev. 2005, 126, 987–1002. [Google Scholar] [CrossRef]
  71. Zhang, P.; Li, Y.; Du, Y.; Li, G.; Wang, L.; Zhou, F. Resveratrol Ameliorated Vascular Calcification by Regulating Sirt-1 and Nrf2. Transplant. Proc. 2016, 48, 3378–3386. [Google Scholar] [CrossRef]
  72. Opie, L.H.; Lecour, S. The red wine hypothesis: From concepts to protective signalling molecules. Eur. Heart J. 2007, 28, 1683–1693. [Google Scholar] [CrossRef]
  73. Ji, Z.; Liu, G.H.; Qu, J. Mitochondrial sirtuins, metabolism, and aging. J. Genet. Genom. 2022, 49, 287–298. [Google Scholar] [CrossRef]
  74. Chang, H.C.; Guarente, L. SIRT1 and other sirtuins in metabolism. Trends Endocrinol. Metab. 2014, 25, 138–145. [Google Scholar] [CrossRef] [PubMed]
  75. Singh, C.K.; Chhabra, G.; Ndiaye, M.A.; Garcia-Peterson, L.M.; MacK, N.J.; Ahmad, N. The Role of Sirtuins in Antioxidant and Redox Signaling. Antioxid. Redox Signal. 2018, 28, 643–661. [Google Scholar] [CrossRef] [PubMed]
  76. Daimiel, L.; Micó, V.; Valls, R.M.; Pedret, A.; Motilva, M.J.; Rubió, L.; Fitó, M.; Farrás, M.; Covas, M.I.; Solá, R.; et al. Impact of Phenol-Enriched Virgin Olive Oils on the Postprandial Levels of Circulating microRNAs Related to Cardiovascular Disease. Mol. Nutr. Food Res. 2020, 64, e2000049. [Google Scholar] [CrossRef]
  77. Cuevas, A.; Saavedra, N.; Salazar, L.A.; Abdalla, D.S.P. Modulation of immune function by polyphenols: Possible contribution of epigenetic factors. Nutrients 2013, 5, 2314–2332. [Google Scholar] [CrossRef]
  78. Pan, M.-H.; Lai, C.-S.; Wu, J.-C.; Ho, C.-T. Epigenetic and Disease Targets by Polyphenols. Curr. Pharm. Des. 2013, 19, 6156–6185. [Google Scholar] [CrossRef]
  79. Izzotti, A.; Cartiglia, C.; Steele, V.E.; De Flora, S. MicroRNAs as targets for dietary and pharmacological inhibitors of mutagenesis and carcinogenesis. Mutat. Res. 2012, 751, 287–303. [Google Scholar] [CrossRef]
  80. Guasch-Ferré, M.; Merino, J.; Sun, Q.; Fitó, M.; Salas-Salvadó, J. Dietary Polyphenols, Mediterranean Diet, Prediabetes, and Type 2 Diabetes: A Narrative Review of the Evidence. Oxid. Med. Cell. Longev. 2017, 2017, 6723931. [Google Scholar] [CrossRef]
  81. Bucciantini, M.; Leri, M.; Nardiello, P.; Casamenti, F.; Stefani, M. Olive polyphenols: Antioxidant and anti-inflammatory properties. Antioxidants 2021, 10, 1044. [Google Scholar] [CrossRef]
  82. Carito, V.; Ceccanti, M.; Tarani, L.; Ferraguti, G.; Chaldakov, G.N.; Fiore, M. Neurotrophins’ Modulation by Olive Polyphenols. Curr. Med. Chem. 2016, 23, 3189–3197. [Google Scholar] [CrossRef]
  83. Solfrizzi, V.; Capurso, C.; D’Introno, A.; Colacicco, A.M.; Santamato, A.; Ranieri, M.; Fiore, P.; Capurso, A.; Panza, F. Lifestyle-related factors in predementia and dementia syndromes. Expert Rev. Neurother. 2008, 8, 133–158. [Google Scholar] [CrossRef]
  84. Leri, M.; Scuto, M.; Ontario, M.L.; Calabrese, V.; Calabrese, E.J.; Bucciantini, M.; Stefani, M. Healthy effects of plant polyphenols: Molecular mechanisms. Int. J. Mol. Sci. 2020, 21, 1250. [Google Scholar] [CrossRef] [PubMed]
  85. Carluccio, M.A.; Siculella, L.; Ancora, M.A.; Massaro, M.; Scoditti, E.; Storelli, C.; Visioli, F.; Distante, A.; De Caterina, R. Olive oil and red wine antioxidant polyphenols inhibit endothelial activation: Antiatherogenic properties of Mediterranean diet phytochemicals. Arterioscler. Thromb. Vasc. Biol. 2003, 23, 622–629. [Google Scholar] [CrossRef] [PubMed]
  86. Amor, S.; Châlons, P.; Aires, V.; Delmas, D. Polyphenol Extracts from Red Wine and Grapevine: Potential Effects on Cancers. Diseases 2018, 6, 106. [Google Scholar] [CrossRef] [PubMed]
  87. Prezioso, I.; Corcione, G.; Digiorgio, C.; Fioschi, G.; Paradiso, V.M. Ageing of Red Wine (cv. Negroamaro) in Mediterranean Areas: Impact of Different Barrels and Apulian Traditional Amphorae on Phenolic Indices, Volatile Composition and Sensory Analysis. Foods 2025, 14, 650. [Google Scholar] [CrossRef]
  88. Di Renzo, L.; Gualtieri, P.; Frank, G.; Cianci, R.; Raffaelli, G.; Peluso, D.; Bigioni, G.; De Lorenzo, A. Sex-Specific Adherence to the Mediterranean Diet in Obese Individuals. Nutrients 2024, 16, 3076. [Google Scholar] [CrossRef]
  89. Reytor-González, C.; Zambrano, A.K.; Montalvan, M.; Frias-Toral, E.; Simancas-Racines, A.; Simancas-Racines, D. Adherence to the Mediterranean Diet and its association with gastric cancer: Health benefits from a Planeterranean perspective. J. Transl. Med. 2024, 22, 483. [Google Scholar] [CrossRef]
  90. Godos, J.; Scazzina, F.; Paternò Castello, C.; Giampieri, F.; Quiles, J.L.; Briones Urbano, M.; Battino, M.; Galvano, F.; Iacoviello, L.; de Gaetano, G.; et al. Underrated aspects of a true Mediterranean diet: Understanding traditional features for worldwide application of a “Planeterranean” diet. J. Transl. Med. 2024, 22, 294. [Google Scholar] [CrossRef]
  91. Marković, A.K.; Torić, J.; Barbarić, M.; Brala, C.J. Hydroxytyrosol, tyrosol and derivatives and their potential effects on human health. Molecules 2019, 24, 2001. [Google Scholar] [CrossRef]
  92. Frumuzachi, O.; Kieserling, H.; Rohn, S.; Mocan, A. The impact of oleuropein, hydroxytyrosol, and tyrosol on cardiometabolic risk factors: A meta-analysis of randomized controlled trials. Crit. Rev. Food Sci. Nutr. 2025, 1–21. [Google Scholar] [CrossRef]
  93. Castillo-Luna, A.; Priego-Capote, F. Phenolic enrichment of foods curated in olive oil: Kinetics and chemical evaluation. Food Chem. X 2024, 22, 101398. [Google Scholar] [CrossRef]
  94. Uğuz, A.C.; Rocha-Pimienta, J.; Martillanes, S.; Garrido, M.; Espino, J.; Delgado-Adámez, J. Chlorophyll Pigments of Olive Leaves and Green Tea Extracts Differentially Affect Their Antioxidant and Anticancer Properties. Molecules 2023, 28, 2779. [Google Scholar] [CrossRef] [PubMed]
  95. Wang, H. Medical Benefits and Polymer Applications of Grapes. Polymers 2025, 17, 750. [Google Scholar] [CrossRef] [PubMed]
  96. Grabarczyk, M.; Justyńska, W.; Czpakowska, J.; Smolińska, E.; Bielenin, A.; Glabinski, A.; Szpakowski, P. Role of Plant Phytochemicals: Resveratrol, Curcumin, Luteolin and Quercetin in Demyelination, Neurodegeneration, and Epilepsy. Antioxidants 2024, 13, 1364. [Google Scholar] [CrossRef] [PubMed]
  97. Zebeaman, M.; Tadesse, M.G.; Bachheti, R.K.; Bachheti, A.; Gebeyhu, R.; Chaubey, K.K. Plants and Plant-Derived Molecules as Natural Immunomodulators. Biomed. Res. Int. 2023, 2023, 7711297. [Google Scholar] [CrossRef]
  98. Shuid, A.N.; Abdul Nasir, N.A.; Ab Azis, N.; Shuid, A.N.; Razali, N.; Ahmad Hairi, H.; Mohd Miswan, M.F.; Naina Mohamed, I. A Systematic Review on the Molecular Mechanisms of Resveratrol in Protecting Against Osteoporosis. Int. J. Mol. Sci. 2025, 26, 2893. [Google Scholar] [CrossRef]
  99. Doghish, A.S.; El-Dakroury, W.A.; Abulsoud, A.I.; Abdelmaksoud, N.M.; Aly, S.H.; Elbadry, A.M.M.; Mohammed, O.A.; Abdel-Reheim, M.A.; Zaki, M.B.; Rizk, N.I.; et al. Natural compounds as regulators of miRNAs: Exploring a new avenue for treating brain cancer. Naunyn Schmiedebergs Arch. Pharmacol. 2025. ahead of print. [Google Scholar] [CrossRef]
  100. Ruggiero, M.; Motti, M.L.; Meccariello, R.; Mazzeo, F. Resveratrol and Physical Activity: A Successful Combination for the Maintenance of Health and Wellbeing? Nutrients 2025, 17, 837. [Google Scholar] [CrossRef]
  101. Carollo, C.; Sorce, A.; Cirafici, E.; Mulè, G.; Caimi, G. Sirtuins and Resveratrol in Cardiorenal Diseases: A Narrative Review of Mechanisms and Therapeutic Potential. Nutrients 2025, 17, 1212. [Google Scholar] [CrossRef]
  102. Park, D.; Jeong, H.; Lee, M.N.; Koh, A.; Kwon, O.; Yang, Y.R.; Noh, J.; Suh, P.G.; Park, H.; Ryu, S.H. Resveratrol induces autophagy by directly inhibiting mTOR through ATP competition. Sci. Rep. 2016, 6, 21772. [Google Scholar] [CrossRef]
  103. Christimann, G.; Rocha, G.; Sattler, J.A.G. Bioactive compounds and dietary patterns in Alzheimer’s disease. J. Alzheimers Dis. 2025, 104, 597–610. [Google Scholar] [CrossRef]
  104. Sbai, O.; Torrisi, F.; Fabrizio, F.P.; Rabbeni, G.; Perrone, L. Effect of the Mediterranean Diet (MeDi) on the Progression of Retinal Disease: A Narrative Review. Nutrients 2024, 16, 3169. [Google Scholar] [CrossRef]
  105. Ross, F.C.; Mayer, D.E.; Horn, J.; Cryan, J.F.; Del Rio, D.; Randolph, E.; Gill, C.I.R.; Gupta, A.; Ross, R.P.; Stanton, C.; et al. Potential of dietary polyphenols for protection from age-related decline and neurodegeneration: A role for gut microbiota? Nutr. Neurosci. 2024, 27, 1058–1076. [Google Scholar] [CrossRef] [PubMed]
  106. Farias-Pereira, R.; Zuk, J.B.; Khavaran, H. Plant bioactive compounds from Mediterranean diet improve risk factors for metabolic syndrome. Int. J. Food Sci. Nutr. 2023, 74, 403–423. [Google Scholar] [CrossRef] [PubMed]
  107. Montano, L.; Maugeri, A.; Volpe, M.G.; Micali, S.; Mirone, V.; Mantovani, A.; Navarra, M.; Piscopo, M. Mediterranean Diet as a Shield against Male Infertility and Cancer Risk Induced by Environmental Pollutants: A Focus on Flavonoids. Int. J. Mol. Sci. 2022, 23, 1568. [Google Scholar] [CrossRef]
  108. Salvo, A.; Tuttolomondo, A. The Role of Olive Oil in Cardiometabolic Risk. Metabolites 2025, 15, 190. [Google Scholar] [CrossRef]
  109. Aznar de la Riera, M.d.C.; Ortolá, R.; Kales, S.N.; Graciani, A.; Diaz-Gutierrez, J.; Banegas, J.R.; Rodríguez-Artalejo, F.; Sotos-Prieto, M. Health and environmental dietary impact: Planetary health diet vs. Mediterranean diet. A nationwide cohort in Spain. Sci. Total Environ. 2025, 968, 178924. [Google Scholar] [CrossRef]
  110. Zalaquett, N.; Lidoriki, I.; Lampou, M.; Saab, J.; Hadkhale, K.; Christophi, C.; Kales, S.N. Adherence to the Mediterranean Diet and the Risk of Head and Neck Cancer: A Systematic Review and Meta-Analysis of Case–Control Studies. Nutrients 2025, 17, 287. [Google Scholar] [CrossRef]
  111. Veronese, N.; Ragusa, F.S.; Maggi, S.; Witard, O.C.; Smith, L.; Dominguez, L.J.; Barbagallo, M.; Isanejad, M.; Prokopidis, K. Effect of the Mediterranean diet on incidence of heart failure in European countries: A systematic review and meta-analysis of cohort studies. Eur. J. Clin. Nutr. 2024, 79, 195–199. [Google Scholar] [CrossRef]
  112. Bakis, H.; Chauveau, P.; Combe, C.; Pfirmann, P. Mediterranean Diet for Cardiovascular Risk Reduction in Chronic Kidney Disease. Adv. Kidney Dis. Health 2023, 30, 496–501. [Google Scholar] [CrossRef]
  113. Hernandez, A.V.; Marti, K.M.; Marti, K.E.; Weisman, N.; Cardona, M.; Biello, D.M.; Pasupuleti, V.; Benites-Zapata, V.A.; Roman, Y.M.; Piscoya, A. Effect of Mediterranean Diets on Cardiovascular Risk Factors and Disease in Overweight and Obese Adults: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. J. Am. Nutr. Assoc. 2025, 44, 387–404. [Google Scholar] [CrossRef]
  114. Fernández-Lázaro, C.I.; Ruiz-Canela, M.; Martínez-González, M.Á. Deep dive to the secrets of the PREDIMED trial. Curr. Opin. Lipidol. 2021, 32, 62–69. [Google Scholar] [CrossRef]
  115. Martínez-González, M.A.; Salas-Salvadó, J.; Estruch, R.; Corella, D.; Fitó, M.; Ros, E. Benefits of the Mediterranean Diet: Insights From the PREDIMED Study. Prog. Cardiovasc. Dis. 2015, 58, 50–60. [Google Scholar] [CrossRef] [PubMed]
  116. Petrella, C.; Carito, V.; Carere, C.; Ferraguti, G.; Ciafrè, S.; Natella, F.; Bello, C.; Greco, A.; Ralli, M.; Mancinelli, R.; et al. Oxidative stress inhibition by resveratrol in alcohol-dependent mice. Nutrition 2020, 79–80, 110783. [Google Scholar] [CrossRef] [PubMed]
  117. Carito, V.; Venditti, A.; Bianco, A.; Ceccanti, M.; Serrilli, A.M.; Chaldakov, G.; Tarani, L.; De Nicolò, S.; Fiore, M. Effects of olive leaf polyphenols on male mouse brain NGF, BDNF and their receptors TrkA, TrkB and p75. Nat. Prod. Res. 2014, 28, 1970–1984. [Google Scholar] [CrossRef]
  118. De Nicoló, S.; Tarani, L.; Ceccanti, M.; Maldini, M.; Natella, F.; Vania, A.; Chaldakov, G.N.; Fiore, M. Effects of olive polyphenols administration on nerve growth factor and brain-derived neurotrophic factor in the mouse brain. Nutrition 2013, 29, 681–687. [Google Scholar] [CrossRef]
  119. Caprifico, A.E.; Calabrese, G.; Tornese, R.; Montefusco, A.; Placì, R.; Semeraro, T.; Durante, M.; De Caroli, M.; Lenucci, M.S. Pomegranate Extracts as Dual Regulators of Angiogenesis: A Systematic Review of Preclinical Evidence in Cancer and Chronic Wound Healing. Mol. Nutr. Food Res. 2025, 69, e70060. [Google Scholar] [CrossRef]
  120. Xie, Y.; Wang, H.; He, Z. Recent advances in polyphenols improving vascular endothelial dysfunction induced by endogenous toxicity. J. Appl. Toxicol. 2021, 41, 701–712. [Google Scholar] [CrossRef]
  121. Otręba, M.; Kośmider, L.; Stojko, J.; Rzepecka-Stojko, A. Cardioprotective activity of selected polyphenols based on epithelial and aortic cell lines. A review. Molecules 2020, 25, 5343. [Google Scholar] [CrossRef]
  122. Akbari, M.; Tamtaji, O.R.; Lankarani, K.B.; Tabrizi, R.; Dadgostar, E.; Kolahdooz, F.; Jamilian, M.; Mirzaei, H.; Asemi, Z. The Effects of Resveratrol Supplementation on Endothelial Function and Blood Pressures Among Patients with Metabolic Syndrome and Related Disorders: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. High Blood Press. Cardiovasc. Prev. 2019, 26, 305–319. [Google Scholar] [CrossRef]
  123. Chong, M.F.F.; MacDonald, R.; Lovegrove, J.A. Fruit polyphenols and CVD risk: A review of human intervention studies. Br. J. Nutr. 2010, 104, S28–S39. [Google Scholar] [CrossRef]
  124. Milena, E.; Maurizio, M. Exploring the Cardiovascular Benefits of Extra Virgin Olive Oil: Insights into Mechanisms and Therapeutic Potential. Biomolecules 2025, 15, 284. [Google Scholar] [CrossRef]
  125. Yang, Y.; Zhang, H.; Liu, Z.; Zhao, Z. Protective effect and mechanism of resveratrol on vascular endothelial cells. Chin. Crit. Care Med. 2024, 36, 664–668. [Google Scholar] [CrossRef]
  126. Silva, H.; Bárbara, R. Exploring the Anti-Hypertensive Potential of Lemongrass—A Comprehensive Review. Biology 2022, 11, 1382. [Google Scholar] [CrossRef] [PubMed]
  127. Mollace, V.; Rosano, G.M.C.; Anker, S.D.; Coats, A.J.S.; Seferovic, P.; Mollace, R.; Tavernese, A.; Gliozzi, M.; Musolino, V.; Carresi, C.; et al. Pathophysiological basis for nutraceutical supplementation in heart failure: A comprehensive review. Nutrients 2021, 13, 257. [Google Scholar] [CrossRef] [PubMed]
  128. Zhang, T.T.; Hu, T.; Jiang, J.G.; Zhao, J.W.; Zhu, W. Antioxidant and anti-inflammatory effects of polyphenols extracted from: Ilex latifolia Thunb. RSC Adv. 2018, 8, 7134–7141. [Google Scholar] [CrossRef]
  129. Wisnuwardani, R.W.; De Henauw, S.; Ferrari, M.; Forsner, M.; Gottrand, F.; Huybrechts, I.; Kafatos, A.G.; Kersting, M.; Knaze, V.; Manios, Y.; et al. Total Polyphenol Intake Is Inversely Associated with a Pro/Anti-Inflammatory Biomarker Ratio in European Adolescents of the HELENA Study. J. Nutr. 2020, 150, 1610–1618. [Google Scholar] [CrossRef]
  130. Shakoor, H.; Feehan, J.; Apostolopoulos, V.; Platat, C.; Al Dhaheri, A.S.; Ali, H.I.; Ismail, L.C.; Bosevski, M.; Stojanovska, L. Immunomodulatory effects of dietary polyphenols. Nutrients 2021, 13, 728. [Google Scholar] [CrossRef]
  131. Iqbal, I.; Wilairatana, P.; Saqib, F.; Nasir, B.; Wahid, M.; Latif, M.F.; Iqbal, A.; Naz, R.; Mubarak, M.S. Plant Polyphenols and Their Potential Benefits on Cardiovascular Health: A Review. Molecules 2023, 28, 6403. [Google Scholar] [CrossRef]
  132. De Bruyne, T.; Steenput, B.; Roth, L.; De Meyer, G.R.Y.; Dos Santos, C.N.; Valentová, K.; Dambrova, M.; Hermans, N. Dietary polyphenols targeting arterial stiffness: Interplay of contributing mechanisms and gut microbiome-related Metabolism. Nutrients 2019, 11, 578. [Google Scholar] [CrossRef]
  133. World Health Organization (WHO). Life Expectancy and Healthy Life Expectancy Data by Country 2022. Available online: https://apps.who.int/gho/data/node.main.688 (accessed on 29 June 2025).
  134. Kwack, S.J.; Kim, K.B.; Kim, H.S.; Yoon, K.S.; Lee, B.M. Risk assessment of soybean-based phytoestrogens. J. Toxicol. Environ. Health—Part A Curr. Issues 2009, 72, 1254–1261. [Google Scholar] [CrossRef]
  135. Ohishi, T.; Miyoshi, N.; Mori, M.; Sagara, M.; Yamori, Y. Health Effects of Soy Isoflavones and Green Tea Catechins on Cancer and Cardiovascular Diseases Based on Urinary Biomarker Levels. Molecules 2022, 27, 8899. [Google Scholar] [CrossRef]
  136. Radeva-Ilieva, M.; Stoeva, S.; Hvarchanova, N.; Georgiev, K.D. Green Tea: Current Knowledge and Issues. Foods 2025, 14, 745. [Google Scholar] [CrossRef] [PubMed]
  137. Saito, E.; Inoue, M.; Sawada, N.; Shimazu, T.; Yamaji, T.; Iwasaki, M.; Sasazuki, S.; Noda, M.; Iso, H.; Tsugane, S.; et al. Association of green tea consumption with mortality due to all causes and major causes of death in a Japanese population: The Japan Public Health Center-based Prospective Study (JPHC Study). Ann. Epidemiol. 2015, 25, 512–518.e3. [Google Scholar] [CrossRef] [PubMed]
  138. Singh, B.N.; Shankar, S.; Srivastava, R.K. Green tea catechin, epigallocatechin-3-gallate (EGCG): Mechanisms, perspectives and clinical applications. Biochem. Pharmacol. 2011, 82, 1807–1821. [Google Scholar] [CrossRef] [PubMed]
  139. Abe, S.K.; Saito, E.; Sawada, N.; Tsugane, S.; Ito, H.; Lin, Y.; Tamakoshi, A.; Sado, J.; Kitamura, Y.; Sugawara, Y.; et al. Green tea consumption and mortality in Japanese men and women: A pooled analysis of eight population-based cohort studies in Japan. Eur. J. Epidemiol. 2019, 34, 917–926. [Google Scholar] [CrossRef]
  140. Zhang, X.; Shu, X.O.; Li, H.; Yang, G.; Li, Q.; Gao, Y.T.; Zheng, W. Prospective cohort study of soy food consumption and risk of bone fracture among postmenopausal women. Arch. Intern. Med. 2005, 165, 1890–1895. [Google Scholar] [CrossRef]
  141. Taku, K.; Umegaki, K.; Sato, Y.; Taki, Y.; Endoh, K.; Watanabe, S. Soy isoflavones lower serum total and LDL cholesterol in humans: A meta-analysis of 11 randomized controlled trials2. Am. J. Clin. Nutr. 2007, 85, 1148–1156. [Google Scholar] [CrossRef]
  142. Baglia, M.L.; Gu, K.; Zhang, X.; Zheng, Y.; Peng, P.; Cai, H.; Bao, P.P.; Zheng, W.; Lu, W.; Shu, X.O. Soy isoflavone intake and bone mineral density in breast cancer survivors. Cancer Causes Control 2015, 26, 571–580. [Google Scholar] [CrossRef]
  143. Gómez-Zorita, S.; González-Arceo, M.; Fernández-Quintela, A.; Eseberri, I.; Trepiana, J.; Portillo, M.P. Scientific evidence supporting the beneficial effects of isoflavones on human health. Nutrients 2020, 12, 3853. [Google Scholar] [CrossRef]
  144. Clarke, E.D.; Collins, C.E.; Rollo, M.E.; Kroon, P.A.; Philo, M.; Haslam, R.L. The relationship between urinary polyphenol metabolites and dietary polyphenol intakes in young adults. Br. J. Nutr. 2022, 127, 589–598. [Google Scholar] [CrossRef]
  145. Del Rio, D.; Rodriguez-Mateos, A.; Spencer, J.P.E.; Tognolini, M.; Borges, G.; Crozier, A. Dietary (poly)phenolics in human health: Structures, bioavailability, and evidence of protective effects against chronic diseases. Antioxid. Redox Signal. 2013, 18, 1818–1892. [Google Scholar] [CrossRef]
  146. Zamora-Ros, R.; Achaintre, D.; Rothwell, J.A.; Rinaldi, S.; Assi, N.; Ferrari, P.; Leitzmann, M.; Boutron-Ruault, M.C.; Fagherazzi, G.; Auffret, A.; et al. Urinary excretions of 34 dietary polyphenols and their associations with lifestyle factors in the EPIC cohort study. Sci. Rep. 2016, 6, 26905. [Google Scholar] [CrossRef] [PubMed]
  147. Sharma, R.; Kumar, R.; Sharma, A.; Goel, A.; Padwad, Y. Long-term consumption of green tea EGCG enhances murine health span by mitigating multiple aspects of cellular senescence in mitotic and post-mitotic tissues, gut dysbiosis, and immunosenescence. J. Nutr. Biochem. 2022, 107, 109068. [Google Scholar] [CrossRef] [PubMed]
  148. Yilmaz, Y. Green Tea Mitigates the Hallmarks of Aging and Age-Related Multisystem Deterioration. Aging Dis. 2025. ahead of print. [Google Scholar] [CrossRef]
  149. Zhang, Y.; Li, J.; Wang, Z.; Chen, J.; Zhao, M.; Guo, C.; Wang, T.; Li, R.; Zhang, H.; Ma, X.; et al. Preclinical evidence construction for epigallocatechin-3-gallate against non-alcoholic fatty liver disease: A meta-analysis and machine learning study. Phytomedicine 2025, 142, 156651. [Google Scholar] [CrossRef] [PubMed]
  150. Al-Regaiey, K. Crosstalk between adipogenesis and aging: Role of polyphenols in combating adipogenic-associated aging. Immun. Ageing 2024, 21, 76. [Google Scholar] [CrossRef]
  151. Messina, M.; Duncan, A.; Messina, V.; Lynch, H.; Kiel, J.; Erdman, J.W. The health effects of soy: A reference guide for health professionals. Front. Nutr. 2022, 9, 970364. [Google Scholar] [CrossRef]
  152. Messina, M. Soy and health update: Evaluation of the clinical and epidemiologic literature. Nutrients 2016, 8, 754. [Google Scholar] [CrossRef]
  153. Kenny, A.M.; Mangano, K.M.; Abourizk, R.H.; Bruno, R.S.; Anamani, D.E.; Kleppinger, A.; Walsh, S.J.; Prestwood, K.M.; Kerstetter, J.E. Soy proteins and isoflavones affect bone mineral density in older women: A randomized controlled trial. Am. J. Clin. Nutr. 2009, 90, 234–242. [Google Scholar] [CrossRef]
  154. 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]
  155. Tsao, R. Chemistry and biochemistry of dietary polyphenols. Nutrients 2010, 2, 1231–1246. [Google Scholar] [CrossRef]
  156. Cianciosi, D.; Forbes-Hernández, T.Y.; Regolo, L.; Alvarez-Suarez, J.M.; Navarro-Hortal, M.D.; Xiao, J.; Quiles, J.L.; Battino, M.; Giampieri, F. The reciprocal interaction between polyphenols and other dietary compounds: Impact on bioavailability, antioxidant capacity and other physico-chemical and nutritional parameters. Food Chem. 2022, 375, 131904. [Google Scholar] [CrossRef]
  157. Covas, M.I. Olive oil and the cardiovascular system. Pharmacol. Res. 2007, 55, 175–186. [Google Scholar] [CrossRef] [PubMed]
  158. Manna, C.; Della Ragione, F.; Cucciolla, V.; Borriello, A.; D’Angelo, S.; Galletti, P.; Zappia, V. Biological effects of hydroxytyrosol, a polyphenol from olive oil endowed with antioxidant activity. Adv. Nutr. Cancer 1999, 472, 115–130. [Google Scholar]
  159. Vivancos, M.; Moreno, J.J. Effect of resveratrol, tyrosol and beta-sitosterol on oxidised low-density lipoprotein-stimulated oxidative stress, arachidonic acid release and prostaglandin E2 synthesis by RAW 264.7 macrophages. Br. J. Nutr. 2008, 99, 1199–1207. [Google Scholar] [CrossRef] [PubMed]
  160. Tang, Y.; Li, S.; Zhang, P.; Zhu, J.; Meng, G.; Xie, L.; Yu, Y.; Ji, Y.; Han, Y. Soy Isoflavone Protects Myocardial Ischemia/Reperfusion Injury through Increasing Endothelial Nitric Oxide Synthase and Decreasing Oxidative Stress in Ovariectomized Rats. Oxid. Med. Cell. Longev. 2016, 2016, 5057405. [Google Scholar] [CrossRef] [PubMed]
  161. Zheng, Z.; Yu, S.; Zhang, W.; Peng, Y.; Pu, M.; Kang, T.; Zeng, J.; Yu, Y.; Li, G. Genistein attenuates monocrotaline-induced pulmonary arterial hypertension in rats by activating PI3K/Akt/eNOS signaling. Histol. Histopathol. 2017, 32, 35–41. [Google Scholar] [CrossRef] [PubMed]
  162. Sankar, P.; Zachariah, B.; Vickneshwaran, V.; Jacob, S.E.; Sridhar, M.G. Amelioration of oxidative stress and insulin resistance by soy isoflavones (from Glycine max) in ovariectomized Wistar rats fed with high fat diet: The molecular mechanisms. Exp. Gerontol. 2015, 63, 67–75. [Google Scholar] [CrossRef]
  163. Sharma, M.; Tollefsbol, T.O. Combinatorial epigenetic mechanisms of sulforaphane, genistein and sodium butyrate in breast cancer inhibition. Exp. Cell Res. 2022, 416, 113160. [Google Scholar] [CrossRef]
  164. De La Parra, C.; Castillo-Pichardo, L.; Cruz-Collazo, A.; Cubano, L.; Redis, R.; Calin, G.A.; Dharmawardhane, S. Soy isoflavone genistein-mediated downregulation of miR-155 contributes to the anticancer effects of genistein. Nutr. Cancer 2016, 68, 154–164. [Google Scholar] [CrossRef]
  165. Phillip, C.J.; Giardina, C.K.; Bilir, B.; Cutler, D.J.; Lai, Y.H.; Kucuk, O.; Moreno, C.S. Genistein cooperates with the histone deacetylase inhibitor vorinostat to induce cell death in prostate cancer cells. BMC Cancer 2012, 12, 145. [Google Scholar] [CrossRef]
  166. Xie, X.; Cong, L.; Liu, S.; Xiang, L.; Fu, X. Genistein alleviates chronic vascular inflammatory response via the miR-21/NF-κB p65 axis in lipopolysaccharide-treated mice. Mol. Med. Rep. 2021, 23, 192. [Google Scholar] [CrossRef]
  167. Lange, K.W.; Nakamura, Y. Japanese food culture and human health–what we can learn from Japan. J. Dis. Prev. Health Promot. 2024, 8, 9–12. [Google Scholar]
  168. Gabriel, A.S.; Ninomiya, K.; Uneyama, H. The role of the Japanese traditional diet in healthy and sustainable dietary patterns around the world. Nutrients 2018, 10, 173. [Google Scholar] [CrossRef] [PubMed]
  169. Phenol-Explorer. Show All Foods Which Polyphen Resveratrol is Found n.d. Available online: http://phenol-explorer.eu/contents/polyphenol/592 (accessed on 24 June 2025).
  170. Micronutrient Information Center; Linus Pauling Institute; Oregon State University; Corvallis OU. Flavonoids. Available online: https://lpi.oregonstate.edu/mic/dietary-factors/phytochemicals/flavonoids (accessed on 24 June 2025).
  171. Bhagwat, S.; Haytowitz, D.B.; Holden, J.M.; U.S. Department of Agriculture; Agricultural Research Service. USDA Database for the Flavonoid Content of Selected Foods, Release 3.0. 2011; pp. 2+98–103. Available online: https://www.ars.usda.gov/ARSUserFiles/80400525/Data/Flav/Flav_R03.pdf (accessed on 24 June 2025).
  172. Linus Pauling Institute; Micronutrients Information Center. Soy Isoflavones. Oregon State University: Corvallis, OR, USA, 2024; Available online: https://lpi.oregonstate.edu/mic/dietary-factors/phytochemicals/soy-isoflavones (accessed on 24 June 2025).
  173. Phenol-Explorer 3.6. Database on Polyphenol Content in Foods. Available online: http://phenol-explorer.eu/ (accessed on 24 June 2025).
  174. Van Doan, H.; Hoseinifar, S.H.; Esteban, M.Á.; Dadar, M.; Thu, T.T.N. Mushrooms, Seaweed, and Their Derivatives as Functional Feed Additives for Aquaculture: An Updated View. In Studies in Natural Products Chemistry; Atta-ur-Rahman, Ed.; Elsevier: Amsterdam, The Netherlands, 2019; Volume 62, pp. 41–90. ISBN 1572-5995. [Google Scholar] [CrossRef]
  175. Terracina, S.; Petrella, C.; Francati, S.; Lucarelli, M.; Barbato, C.; Minni, A.; Ralli, M.; Greco, A.; Tarani, L.; Fiore, M.; et al. Antioxidant Intervention to Improve Cognition in the Aging Brain: The Example of Hydroxytyrosol and Resveratrol. Int. J. Mol. Sci. 2022, 23, 15674. [Google Scholar] [CrossRef]
  176. Tosti, V.; Bertozzi, B.; Fontana, L. Health Benefits of the Mediterranean Diet: Metabolic and Molecular Mechanisms. J. Gerontol.—Ser. A Biol. Sci. Med. Sci. 2018, 73, 318–326. [Google Scholar] [CrossRef]
  177. Barbouti, A.; Goulas, V. Dietary antioxidants in the mediterranean diet. Antioxidants 2021, 10, 1213. [Google Scholar] [CrossRef]
  178. De Santis, S.; Clodoveo, M.L.; Corbo, F. Correlation between Chemical Characterization and Biological Activity: An Urgent Need for Human Studies Using Extra Virgin Olive Oil. Antioxidants 2022, 11, 258. [Google Scholar] [CrossRef]
  179. Hsu, A.; Bruno, R.S.; Löhr, C.V.; Taylor, A.W.; Dashwood, R.H.; Bray, T.M.; Ho, E. Dietary soy and tea mitigate chronic inflammation and prostate cancer via NFκB pathway in the Noble rat model. J. Nutr. Biochem. 2011, 22, 502–510. [Google Scholar] [CrossRef]
  180. Tsuboi, H.; Takahashi, M.; Minamida, Y.; Yoshida, N. Psychological well-being and green tea consumption are associated with lower pentosidine serum levels among elderly female residents in Japan. J. Psychosom. Res. 2019, 126, 109825. [Google Scholar] [CrossRef]
  181. Kim, H.S.; Quon, M.J.; Kim, J.A. New insights into the mechanisms of polyphenols beyond antioxidant properties; lessons from the green tea polyphenol, epigallocatechin 3-gallate. Redox Biol. 2014, 2, 187–195. [Google Scholar] [CrossRef]
  182. Luo, Q.; Luo, L.; Zhao, J.; Wang, Y.; Luo, H. Biological potential and mechanisms of Tea’s bioactive compounds: An Updated review. J. Adv. Res. 2023, 65, 345–363. [Google Scholar] [CrossRef]
  183. Hernández-Silva, D.; López-Abellán, M.D.; Martínez-Navarro, F.J.; García-Castillo, J.; Cayuela, M.L.; Alcaraz-Pérez, F. Development of a Short Telomere Zebrafish Model for Accelerated Aging Research and Antiaging Drug Screening. Aging Cell 2025, 24, e70007. [Google Scholar] [CrossRef] [PubMed]
  184. Sharma, A.K.; Mukherjee, M.; Kumar, A.; Sharma, G.; Tabassum, F.; Akhtar, M.S.; Imam, M.T.; Almalki, Z.S. Preliminary investigation on impact of intergenerational treatment of resveratrol endorses the development of ‘super-pups’. Life Sci. 2023, 314, 121322. [Google Scholar] [CrossRef] [PubMed]
  185. Ansari, M.Y.; Ahmad, N.; Haqqi, T.M. Oxidative stress and inflammation in osteoarthritis pathogenesis: Role of polyphenols. Biomed. Pharmacother. 2020, 129, 110452. [Google Scholar] [CrossRef]
  186. Xicota, L.; Rodriguez-Morato, J.; Dierssen, M.; de la Torre, R. Potential Role of (-)-Epigallocatechin-3-Gallate (EGCG) in the Secondary Prevention of Alzheimer Disease. Curr. Drug Targets 2015, 18, 174–195. [Google Scholar] [CrossRef]
  187. Porquet, D.; Casadesús, G.; Bayod, S.; Vicente, A.; Canudas, A.M.; Vilaplana, J.; Pelegrí, C.; Sanfeliu, C.; Camins, A.; Pallàs, M.; et al. Dietary resveratrol prevents Alzheimer’s markers and increases life span in SAMP8. Age 2013, 35, 1851–1865. [Google Scholar] [CrossRef]
  188. Hector, K.L.; Lagisz, M.; Nakagawa, S. The effect of resveratrol on longevity across species: A meta-analysis. Biol. Lett. 2012, 8, 790–793. [Google Scholar] [CrossRef]
  189. Fumarola, S.; Cianfruglia, L.; Cecati, M.; Giammarchi, C.; Vaiasicca, S.; Gasparrini, M. Polyphenol Intake in Elderly Patients: A Novel Approach to Counteract Colorectal Cancer Risk? Int. J. Mol. Sci. 2025, 26, 2497. [Google Scholar] [CrossRef]
  190. Shinoda-Ito, Y.; Omori, K.; Ito, T.; Nakayama, M.; Ikeda, A.; Ito, M.; Ohara, T.; Takashiba, S. Novel Iron Chelators, Super-Polyphenols, Show Antimicrobial Effects against Cariogenic Streptococcus mutans. Antibiotics 2023, 12, 1562. [Google Scholar] [CrossRef]
  191. Silva, J.; Alves, C.; Soledade, F.; Martins, A.; Pinteus, S.; Gaspar, H.; Alfonso, A.; Pedrosa, R. Marine-Derived Components: Can They Be a Potential Therapeutic Approach to Parkinson’s Disease? Mar. Drugs 2023, 21, 451. [Google Scholar] [CrossRef]
  192. Moratilla-Rivera, I.; Sánchez, M.; Valdés-González, J.A.; Gómez-Serranillos, M.P. Natural Products as Modulators of Nrf2 Signaling Pathway in Neuroprotection. Int. J. Mol. Sci. 2023, 24, 3748. [Google Scholar] [CrossRef]
  193. Kubczak, M.; Szustka, A.; Rogalińska, M. Molecular targets of natural compounds with anti-cancer properties. Int. J. Mol. Sci. 2021, 22, 13659. [Google Scholar] [CrossRef] [PubMed]
  194. Figueira, I.; Menezes, R.; Macedo, D.; Costa, I.; dos Santos, C.N. Polyphenols Beyond Barriers: A Glimpse into the Brain. Curr. Neuropharmacol. 2016, 15, 562–594. [Google Scholar] [CrossRef] [PubMed]
  195. Lv, J.; Zhang, W.; Hu, R.; Yang, X.; Gong, J.; Ma, S.; Xiang, H.; Yuan, X.; Zhang, H.; He, X.; et al. Dietary resveratrol improves semen quality by promoting mitochondrial function related genes expression in aging boars. J. Anim. Sci. 2025. ahead of print. [Google Scholar] [CrossRef]
  196. Liu, J.; Wang, Y.; Sun, H.; Lei, D.; Liu, J.; Fei, Y.; Wang, C.; Han, C. Resveratrol ameliorates postoperative cognitive dysfunction in aged mice by regulating microglial polarization through CX3CL1/CX3CR1 signaling axis. Neurosci. Lett. 2025, 847, 138089. [Google Scholar] [CrossRef] [PubMed]
  197. Boondam, Y.; Saefoong, C.; Niltup, N.; Monteil, A.; Kitphati, W. The Cognitive Restoration Effects of Resveratrol: Insight Molecular through Behavioral Studies in Various Cognitive Impairment Models. ACS Pharmacol. Transl. Sci. 2024, 7, 3334–3357. [Google Scholar] [CrossRef]
  198. Zheng, X.M.; Zhang, X.D.; Tan, L.L.; Zhang, J.; Wang, T.T.; Ling, Q.; Wang, H.; Ouyang, K.W.; Wang, K.W.; Chang, W.; et al. Sirt1 m6A modification-evoked Leydig cell senescence promotes Cd-induced testosterone decline. Ecotoxicol. Environ. Saf. 2024, 284, 116884. [Google Scholar] [CrossRef]
  199. Napoli, C.; Coscioni, E.; Trama, U.; Strozziero, M.G.; Benincasa, G. An evidence-based debate on epigenetics and immunosenescence in COVID-19. Curr. Res. Immunol. 2023, 4, 100069. [Google Scholar] [CrossRef]
  200. Tipoe, G.L.; Leung, T.M.; Liong, E.C.; Lau, T.Y.H.; Fung, M.L.; Nanji, A.A. Epigallocatechin-3-gallate (EGCG) reduces liver inflammation, oxidative stress and fibrosis in carbon tetrachloride (CCl4)-induced liver injury in mice. Toxicology 2010, 273, 45–52. [Google Scholar] [CrossRef]
  201. Pan, A.L.; Hasalliu, E.; Hasalliu, M.; Angulo, J.A. Epigallocatechin Gallate Mitigates the Methamphetamine-Induced Striatal Dopamine Terminal Toxicity by Preventing Oxidative Stress in the Mouse Brain. Neurotox. Res. 2020, 37, 883–892. [Google Scholar] [CrossRef]
  202. Rasheed, N.O.A.; Ahmed, L.A.; Abdallah, D.M.; El-Sayeh, B.M. Nephro-toxic effects of intraperitoneally injected EGCG in diabetic mice: Involvement of oxidative stress, inflammation and apoptosis. Sci. Rep. 2017, 7, 40617. [Google Scholar] [CrossRef]
  203. Xu, T.; Liu, R.; Zhu, H.; Zhou, Y.; Pei, T.; Yang, Z. The Inhibition of LPS-Induced Oxidative Stress and Inflammatory Responses Is Associated with the Protective Effect of (-)-Epigallocatechin-3-Gallate on Bovine Hepatocytes and Murine Liver. Antioxidants 2022, 11, 914. [Google Scholar] [CrossRef]
  204. Ortiz-López, L.; Márquez-Valadez, B.; Gómez-Sánchez, A.; Silva-Lucero, M.D.C.; Torres-Pérez, M.; Téllez-Ballesteros, R.I.; Ichwan, M.; Meraz-Ríos, M.A.; Kempermann, G.; Ramírez-Rodríguez, G.B. Green tea compound epigallo-catechin-3-gallate (EGCG) increases neuronal survival in adult hippocampal neurogenesis in vivo and in vitro. Neuroscience 2016, 322, 208–220. [Google Scholar] [CrossRef] [PubMed]
  205. Wang, L.; Dai, M.; Ge, Y.; Chen, J.; Wang, C.; Yao, C.; Lin, Y. EGCG protects the mouse brain against cerebral ischemia/reperfusion injury by suppressing autophagy via the AKT/AMPK/mTOR phosphorylation pathway. Front. Pharmacol. 2022, 13, 921394. [Google Scholar] [CrossRef] [PubMed]
  206. Wang, J.; Kuang, X.; Peng, Z.; Li, C.; Guo, C.; Fu, X.; Wu, J.; Luo, Y.; Rao, X.; Zhou, X.; et al. EGCG treats ICH via up-regulating miR-137-3p and inhibiting Parthanatos. Transl. Neurosci. 2020, 11, 371–379. [Google Scholar] [CrossRef] [PubMed]
  207. Koh, S.H.; Lee, S.M.; Kim, H.Y.; Lee, K.Y.; Lee, Y.J.; Kim, H.T.; Kim, J.; Kim, M.H.; Hwang, M.S.; Song, C.; et al. The effect of epigallocatechin gallate on suppressing disease progression of ALS model mice. Neurosci. Lett. 2006, 395, 103–107. [Google Scholar] [CrossRef] [PubMed]
  208. Lai, S.W.; Chen, J.H.; Lin, H.Y.; Liu, Y.S.; Tsai, C.F.; Chang, P.C.; Lu, D.Y.; Lin, C. Regulatory Effects of Neuroinflammatory Responses Through Brain-Derived Neurotrophic Factor Signaling in Microglial Cells. Mol. Neurobiol. 2018, 55, 7487–7499. [Google Scholar] [CrossRef]
  209. Zupo, R.; Castellana, F.; Lisco, G.; Corbo, F.; Crupi, P.; Sardone, R.; Catino, F.; Perna, S.; Gesualdo, L.; Lozupone, M.; et al. The Effect of a Mediterranean Diet on Arterial Stiffness: A Systematic Review. Nutrients 2025, 17, 1192. [Google Scholar] [CrossRef]
  210. Widmer, R.J.; Flammer, A.J.; Lerman, L.O.; Lerman, A. The Mediterranean diet, its components, and cardiovascular disease. Am. J. Med. 2015, 128, 229–238. [Google Scholar] [CrossRef]
  211. Martínez-González, M.A.; Gea, A.; Ruiz-Canela, M. The Mediterranean Diet and Cardiovascular Health: A Critical Review. Circ. Res. 2019, 124, 779–798. [Google Scholar] [CrossRef]
  212. Kuriyama, S.; Shimazu, T.; Ohmori, K.; Kikuchi, N.; Nakaya, N.; Nishino, Y.; Tsubono, Y.; Tsuji, I. Green tea consumption and mortality due to cardiovascular disease, cancer, and all causes in Japan: The Ohsaki study. JAMA 2006, 296, 1255–1265. [Google Scholar] [CrossRef]
  213. Suzuki, E.; Yorifuji, T.; Takao, S.; Komatsu, H.; Sugiyama, M.; Ohta, T.; Ishikawa-Takata, K.; Doi, H. Green Tea Consumption and Mortality among Japanese Elderly People: The Prospective Shizuoka Elderly Cohort. Ann. Epidemiol. 2009, 19, 732–739. [Google Scholar] [CrossRef]
  214. Vázquez-Ruiz, Z.; Alonso, A.; Alonso-Gómez, Á.; Romaguera, D.; Martínez-González, M.Á.; Li, L.; Berrade, I.; Tojal-Sierra, L.; Noris, M.; Lamuela-Raventós, R.M.; et al. Associations between Dietary Phenolic Compounds and Biomarkers of Atrial Fibrillation Risk in Adults with Metabolic Syndrome: A Longitudinal Analysis. J. Nutr. 2025, in press. [Google Scholar] [CrossRef]
  215. Di Daniele, N.D.; Noce, A.; Vidiri, M.F.; Moriconi, E.; Marrone, G.; Annicchiarico-Petruzzelli, M.; D’Urso, G.; Tesauro, M.; Rovella, V.; De Lorenzo, A.D. Impact of Mediterranean diet on metabolic syndrome, cancer and longevity. Oncotarget 2017, 8, 8947–8979. [Google Scholar] [CrossRef] [PubMed]
  216. Quetglas-Llabrés, M.M.; Monserrat-Mesquida, M.; Bouzas, C.; García, S.; Mateos, D.; Ugarriza, L.; Gómez, C.; Sureda, A.; Tur, J.A. Long-Term Impact of Nutritional Intervention with Increased Polyphenol Intake and Physical Activity Promotion on Oxidative and Inflammatory Profiles in Patients with Metabolic Syndrome. Nutrients 2024, 16, 2121. [Google Scholar] [CrossRef] [PubMed]
  217. Kantartzis, K.; Fritsche, L.; Bombrich, M.; Machann, J.; Schick, F.; Staiger, H.; Kunz, I.; Schoop, R.; Lehn-Stefan, A.; Heni, M.; et al. Effects of resveratrol supplementation on liver fat content in overweight and insulin-resistant subjects: A randomized, double-blind, placebo-controlled clinical trial. Diabetes Obes. Metab. 2018, 20, 1793–1797. [Google Scholar] [CrossRef] [PubMed]
  218. Timmers, S.; Konings, E.; Bilet, L.; Houtkooper, R.H.; Van De Weijer, T.; Goossens, G.H.; Hoeks, J.; Van Der Krieken, S.; Ryu, D.; Kersten, S.; et al. Calorie restriction-like effects of 30 days of resveratrol supplementation on energy metabolism and metabolic profile in obese humans. Cell Metab. 2011, 14, 612–622. [Google Scholar] [CrossRef]
  219. Izzo, C.; Annunziata, M.; Melara, G.; Sciorio, R.; Dallio, M.; Masarone, M.; Federico, A.; Persico, M. The role of resveratrol in liver disease: A comprehensive review from in vitro to clinical trials. Nutrients 2021, 13, 933. [Google Scholar] [CrossRef]
  220. Hosseini, H.; Koushki, M.; Khodabandehloo, H.; Fathi, M.; Panahi, G.; Teimouri, M.; Majidi, Z.; Meshkani, R. The effect of resveratrol supplementation on C-reactive protein (CRP) in type 2 diabetic patients: Results from a systematic review and meta-analysis of randomized controlled trials. Complement. Ther. Med. 2020, 49, 102251. [Google Scholar] [CrossRef]
  221. Forcano, L.; Fauria, K.; Soldevila-Domenech, N.; Minguillón, C.; Lorenzo, T.; Cuenca-Royo, A.; Menezes-Cabral, S.; Pizarro, N.; Boronat, A.; Molinuevo, J.L.; et al. Prevention of cognitive decline in subjective cognitive decline APOE ε4 carriers after EGCG and a multimodal intervention (PENSA): Study design. Alzheimer’s Dement. Transl. Res. Clin. Interv. 2021, 7, e12155. [Google Scholar] [CrossRef]
  222. Bazyar, H.; Hosseini, S.A.; Saradar, S.; Mombaini, D.; Allivand, M.; Labibzadeh, M.; Alipour, M. Effects of epigallocatechin-3-gallate of Camellia sinensis leaves on blood pressure, lipid profile, atherogenic index of plasma and some inflammatory and antioxidant markers in type 2 diabetes mellitus patients: A clinical trial. J. Complement. Integr. Med. 2021, 18, 405–411. [Google Scholar] [CrossRef]
  223. Settakorn, K.; Hantrakool, S.; Petiwathayakorn, T.; Hutachok, N.; Tantiworawit, A.; Charoenkwan, P.; Chalortham, N.; Chompupoung, A.; Paradee, N.; Koonyosying, P.; et al. A randomized placebo−controlled clinical trial of oral green tea epigallocatechin 3−gallate on erythropoiesis and oxidative stress in transfusion−dependent β−thalassemia patients. Front. Mol. Biosci. 2023, 10, 1248742. [Google Scholar] [CrossRef]
  224. Loftis, J.M.; Wilhelm, C.J.; Huckans, M. Effect of epigallocatechin gallate supplementation in schizophrenia and bipolar disorder: An 8-week, randomized, double-blind, placebo-controlled study. Ther. Adv. Psychopharmacol. 2013, 3, 21–27. [Google Scholar] [CrossRef]
  225. Cornali, K.; Di Lauro, M.; Marrone, G.; Masci, C.; Montalto, G.; Giovannelli, A.; Schievano, C.; Tesauro, M.; Pieri, M.; Bernardini, S.; et al. The Effects of a Food Supplement, Based on Co-Micronized Palmitoylethanolamide (PEA)–Rutin and Hydroxytyrosol, in Metabolic Syndrome Patients: Preliminary Results. Nutrients 2025, 17, 413. [Google Scholar] [CrossRef] [PubMed]
  226. de Bock, M.; Derraik, J.G.B.; Brennan, C.M.; Biggs, J.B.; Morgan, P.E.; Hodgkinson, S.C.; Hofman, P.L.; Cutfield, W.S. Olive (Olea europaea L.) Leaf Polyphenols Improve Insulin Sensitivity in Middle-Aged Overweight Men: A Randomized, Placebo-Controlled, Crossover Trial. PLoS ONE 2013, 8, e57622. [Google Scholar] [CrossRef] [PubMed]
  227. Arjmandi, B.H.; Lucas, E.A.; Khalil, D.A.; Devareddy, L.; Smith, B.J.; McDonald, J.; Arquitt, A.B.; Payton, M.E.; Mason, C. One year soy protein supplementation has positive effects on bone formation markers but not bone density in postmenopausal women. Nutr. J. 2005, 4, 8. [Google Scholar] [CrossRef] [PubMed]
  228. Anderson, J.J.B.; Chen, X.; Boass, A.; Symons, M.; Kohlmeier, M.; Renner, J.B.; Garner, S.C. Soy isoflavones: No effects on bone mineral content and bone mineral density in healthy, menstruating young adult women after one year. J. Am. Coll. Nutr. 2002, 21, 388–393. [Google Scholar] [CrossRef]
  229. Yamashima, T. 4-Hydroxynonenal from Mitochondrial and Dietary Sources Causes Lysosomal Cell Death for Lifestyle-Related Diseases. Nutrients 2024, 16, 4171. [Google Scholar] [CrossRef]
  230. Yamashima, T.; Seike, T.; Mochly-Rosen, D.; Chen, C.H.; Kikuchi, M.; Mizukoshi, E. Implication of the cooking oil-peroxidation product “hydroxynonenal” for Alzheimer’s disease. Front. Aging Neurosci. 2023, 15, 1211141. [Google Scholar] [CrossRef]
  231. Yamashima, T.; Seike, T.; Oikawa, S.; Kobayashi, H.; Kido, H.; Yanagi, M.; Yamamiya, D.; Li, S.; Boontem, P.; Mizukoshi, E. Hsp70.1 carbonylation induces lysosomal cell death for lifestyle-related diseases. Front. Mol. Biosci. 2023, 9, 1063632. [Google Scholar] [CrossRef]
  232. Seike, T.; Boontem, P.; Yanagi, M.; Li, S.; Kido, H.; Yamamiya, D.; Nakagawa, H.; Okada, H.; Yamashita, T.; Harada, K.; et al. Hydroxynonenal Causes Hepatocyte Death by Disrupting Lysosomal Integrity in Nonalcoholic Steatohepatitis. Cell. Mol. Gastroenterol. Hepatol. 2022, 14, 925–944. [Google Scholar] [CrossRef]
  233. Boontem, P.; Yamashima, T. Hydroxynonenal causes Langerhans cell degeneration in the pancreas of Japanese macaque monkeys. PLoS ONE 2021, 16, e0245702. [Google Scholar] [CrossRef]
  234. Zhuang, Y.; Dong, J.; He, X.; Wang, J.; Li, C.; Dong, L.; Zhang, Y.; Zhou, X.; Wang, H.; Yi, Y.; et al. Impact of Heating Temperature and Fatty Acid Type on the Formation of Lipid Oxidation Products During Thermal Processing. Front. Nutr. 2022, 9, 913297. [Google Scholar] [CrossRef]
  235. Tranchida, N.; Molinari, F.; Franco, G.A.; Cordaro, M.; Di Paola, R. Potential Role of Dietary Antioxidants During Skin Aging. Food Sci. Nutr. 2025, 13, e70231. [Google Scholar] [CrossRef] [PubMed]
  236. Zhang, Y.; Zhang, Z.; Tu, C.; Chen, X.; He, R. Advanced Glycation End Products in Disease Development and Potential Interventions. Antioxidants 2025, 14, 492. [Google Scholar] [CrossRef] [PubMed]
  237. Ma, Y.; Wang, X.; Lin, S.; King, L.; Liu, L. The Potential Role of Advanced Glycation End Products in the Development of Kidney Disease. Nutrients 2025, 17, 758. [Google Scholar] [CrossRef]
  238. Malik, V.S.; Popkin, B.M.; Bray, G.A.; Després, J.P.; Hu, F.B. Sugar-sweetened beverages, obesity, type 2 diabetes mellitus, and cardiovascular disease risk. Circulation 2010, 121, 1356–1364. [Google Scholar] [CrossRef]
  239. Popkin, B.M.; Hawkes, C. Sweetening of the global diet, particularly beverages: Patterns, trends, and policy responses. Lancet Diabetes Endocrinol. 2016, 4, 174–186. [Google Scholar] [CrossRef]
  240. Pascual-Morena, C.; Garrido-Miguel, M.; Martínez-García, I.; Lucerón-Lucas-Torres, M.; Rodríguez-Gutiérrez, E.; Berlanga-Macías, C.; Fernández-Bravo-Rodrigo, J.; Patiño-Cardona, S. Association of Dietary Advanced Glycation End Products with Overall and Site-Specific Cancer Risk and Mortality: A Systematic Review and Meta-Analysis. Nutrients 2025, 17, 1638. [Google Scholar] [CrossRef]
  241. Giraldo-Gonzalez, G.C.; Roman-Gonzalez, A.; Cañas, F.; Garcia, A. Molecular Mechanisms of Type 2 Diabetes-Related Heart Disease and Therapeutic Insights. Int. J. Mol. Sci. 2025, 26, 4548. [Google Scholar] [CrossRef]
  242. Vianello, E.; Beltrami, A.P.; Aleksova, A.; Janjusevic, M.; Fluca, A.L.; Corsi Romanelli, M.M.; La Sala, L.; Dozio, E. The Advanced Glycation End-Products (AGE)–Receptor for AGE System (RAGE): An Inflammatory Pathway Linking Obesity and Cardiovascular Diseases. Int. J. Mol. Sci. 2025, 26, 3707. [Google Scholar] [CrossRef]
  243. Habes, D.; Kestranek, J.; Stranik, J.; Kacerovsky, M.; Spacek, J. Is there an association between pelvic organ prolapse and oxidative stress? A systematic review. PLoS ONE 2022, 17, e0271467. [Google Scholar] [CrossRef]
  244. Plotegher, N.; Bubacco, L. Lysines, Achilles’ heel in alpha-synuclein conversion to a deadly neuronal endotoxin. Ageing Res. Rev. 2016, 26, 62–71. [Google Scholar] [CrossRef]
  245. Steinerová, A.; Racek, J.; Stožický, F.; Zima, T.; Fialová, L.; Lapin, A. Antibodies against oxidized LDL—Theory and clinical use. Physiol. Res. 2001, 50, 131–141. [Google Scholar] [CrossRef] [PubMed]
  246. Mallapaty, S. How much ultra-processed food do you eat? Blood and urine record it. Nature 2025. ahead of print. [Google Scholar] [CrossRef]
  247. Hafner, E.; Hribar, M.; Pravst, I. Ultra-Processed Foods in the Food Supply: Prevalence, Nutritional Composition and Use of Voluntary Labelling Schemes. Nutrients 2025, 17, 1731. [Google Scholar] [CrossRef] [PubMed]
  248. Fontalba-Navas, A.; Echeverria, R.; Larrea-Killinger, C.; Gracia-Arnaiz, M.; Soar, C.; Arrebola, J.P. Association Between the Healthy Eating Index and the Body Mass Index of Older Adults: An Analysis of Food Frequency and Preferences. Nutrients 2025, 17, 1717. [Google Scholar] [CrossRef]
  249. Han, I.H.; Csallany, A.S. Temperature dependence of HNE formation in vegetable oils and butter oil. J. Am. Oil Chem. Soc. 2008, 85, 777–782. [Google Scholar] [CrossRef]
  250. Yuan, J.; Shoeman, D.W.; Csallany, A.S. Formation of 4-Hydroxy-2-Trans-Nonenal, a Toxic Aldehyde, in Thermally Treated Olive and Sunflower Oils. J. Am. Oil Chem. Soc. 2018, 95, 813–823. [Google Scholar] [CrossRef]
  251. Golan, R.; Shai, I.; Gepner, Y.; Harman-Boehm, I.; Schwarzfuchs, D.; Spence, J.D.; Parraga, G.; Buchanan, D.; Witkow, S.; Friger, M.; et al. Effect of wine on carotid atherosclerosis in type 2 diabetes: A 2-year randomized controlled trial. Eur. J. Clin. Nutr. 2018, 72, 871–878. [Google Scholar] [CrossRef]
  252. Serio, F.; Imbriani, G.; Acito, M.; Moretti, M.; Fanizzi, F.P.; De Donno, A.; Valacchi, G. Moderate red wine intake and cardiovascular health protection: A literature review. Food Funct. 2023, 14, 6346–6362. [Google Scholar] [CrossRef]
  253. McNeill, K.A. Epidemiology of Brain Tumors. Neurol. Clin. 2016, 34, 981–998. [Google Scholar] [CrossRef]
  254. Johnson, D.E.; Burtness, B.; Leemans, C.R.; Lui, V.W.Y.; Bauman, J.E.; Grandis, J.R. Head and neck squamous cell carcinoma. Nat. Rev. Dis. Prim. 2020, 6, 92. [Google Scholar] [CrossRef]
  255. Varghese, J.; Dakhode, S. Effects of Alcohol Consumption on Various Systems of the Human Body: A Systematic Review. Cureus 2022, 14, e30057. [Google Scholar] [CrossRef]
  256. MacKillop, J.; Agabio, R.; Feldstein Ewing, S.W.; Heilig, M.; Kelly, J.F.; Leggio, L.; Lingford-Hughes, A.; Palmer, A.A.; Parry, C.D.; Ray, L.; et al. Hazardous drinking and alcohol use disorders. Nat. Rev. Dis. Prim. 2022, 8, 80. [Google Scholar] [CrossRef] [PubMed]
  257. Ceci, F.M.; Ferraguti, G.; Petrella, C.; Greco, A.; Ralli, M.; Iannitelli, A.; Carito, V.; Tirassa, P.; Chaldakov, G.N.; Messina, M.P.; et al. Nerve Growth Factor in Alcohol Use Disorders. Curr. Neuropharmacol. 2020, 19, 45–60. [Google Scholar] [CrossRef] [PubMed]
  258. Rodriguez, F.D.; Coveñas, R. Biochemical mechanisms associating alcohol use disorders with cancers. Cancers 2021, 13, 3548. [Google Scholar] [CrossRef] [PubMed]
  259. Zorumski, C.F.; Mennerick, S.; Izumi, Y. Acute and chronic effects of ethanol on learning-related synaptic plasticity. Alcohol 2014, 48, 1–17. [Google Scholar] [CrossRef]
  260. Cederbaum, A.I. Alcohol Metabolism. Clin. Liver Dis. 2012, 16, 667–685. [Google Scholar] [CrossRef]
  261. Jiang, Y.; Zhang, T.; Kusumanchi, P.; Han, S.; Yang, Z.; Liangpunsakul, S. Alcohol metabolizing enzymes, microsomal ethanol oxidizing system, cytochrome P450 2E1, catalase, and aldehyde dehydrogenase in alcohol-associated liver disease. Biomedicines 2020, 8, 50. [Google Scholar] [CrossRef]
  262. Millonig, G.; Wang, Y.; Homann, N.; Bernhardt, F.; Qin, H.; Mueller, S.; Bartsch, H.; Seitz, H.K. Ethanol-mediated carcinogenesis in the human esophagus implicates CYP2E1 induction and the generation of carcinogenic DNA-lesions. Int. J. Cancer 2011, 128, 533–540. [Google Scholar] [CrossRef]
  263. Cahill, P.A.; Redmond, E.M. Alcohol and cardiovascular disease--modulation of vascular cell function. Nutrients 2012, 4, 297–318. [Google Scholar] [CrossRef]
  264. Ceccanti, M.; Coriale, G.; Fiorentino, D.; Iannitelli, A.; Tarani, L.; Fiore, M. Italian Guidelines for the diagnosis and treatment of Fetal Alcohol Spectrum Disorders. Riv. Psichiatr. 2024, 59, 191–193. [Google Scholar] [CrossRef]
  265. Ferraguti, G.; Fanfarillo, F.; Nicotera, S.; Terracina, S.; Moschella, C.; Mattia, A.; David, M.C.; Pichini, S.; Coriale, G.; Fiorentino, D.; et al. Italian Guidelines for the diagnosis and treatment of Fetal Alcohol Spectrum Disorders: Detecting alcohol drinking during pregnancy. Riv. Psichiatr. 2024, 59, 241–249. [Google Scholar] [CrossRef]
  266. Popova, S.; Dozet, D.; Shield, K.; Rehm, J.; Burd, L. Alcohol’s impact on the fetus. Nutrients 2021, 13, 3452. [Google Scholar] [CrossRef] [PubMed]
  267. Carpita, B.; Migli, L.; Chiarantini, I.; Battaglini, S.; Montalbano, C.; Carmassi, C.; Cremone, I.M.; Dell’osso, L. Autism Spectrum Disorder and Fetal Alcohol Spectrum Disorder: A Literature Review. Brain Sci. 2022, 12, 792. [Google Scholar] [CrossRef] [PubMed]
  268. Wilhoit, L.F.; Scott, D.A.; Simecka, B.A. Fetal Alcohol Spectrum Disorders: Characteristics, Complications, and Treatment. Community Ment. Health J. 2017, 53, 711–718. [Google Scholar] [CrossRef] [PubMed]
  269. del Campo, M.; Jones, K.L. A review of the physical features of the fetal alcohol spectrum disorders. Eur. J. Med. Genet. 2017, 60, 55–64. [Google Scholar] [CrossRef]
  270. Ceccanti, M.; Coriale, G.; Fiorentino, D.; Tarani, L.; Messina, M.P.; Vitali, M.; Fiore, M.; May, P.A. Italian Guidelines for the diagnosis and treatment of Fetal Alcohol Spectrum Disorders: Epidemiology. Riv. Psichiatr. 2024, 59, 259–268. [Google Scholar] [CrossRef]
  271. Menghi, M.; Micangeli, G.; Paparella, R.; Ceccanti, M.; Coriale, G.; Ferraguti, G.; Fiore, M.; Fiorentino, D.; Piccioni, M.G.; Tarani, L. Italian Guidelines for the diagnosis and treatment of Fetal Alcohol Spectrum Disorders: Clinical hallmarks. Riv. Psichiatr. 2024, 59, 203–211. [Google Scholar] [CrossRef]
  272. Micangeli, G.; Menghi, M.; Paparella, R.; Ceccanti, M.; Coriale, G.; Fiorentino, D.; Ferraguti, G.; Fiore, M.; Tarani, L. Italian Guidelines for the diagnosis and treatment of Fetal Alcohol Spectrum Disorders: Diagnostic criteria. Riv. Psichiatr. 2024, 59, 195–202. [Google Scholar] [CrossRef]
  273. Carito, V.; Ceccanti, M.; Ferraguti, G.; Coccurello, R.; Ciafrè, S.; Tirassa, P.; Fiore, M. NGF and BDNF Alterations by Prenatal Alcohol Exposure. Curr. Neuropharmacol. 2019, 17, 308–317. [Google Scholar] [CrossRef]
  274. Chabenne, A.; Moon, C.; Ojo, C.; Khogali, A.; Nepal, B.; Sharma, S. Biomarkers in fetal alcohol syndrome. Biomark. Genom. Med. 2014, 6, 12–22. [Google Scholar] [CrossRef]
  275. Burd, L.; Klug, M.G.; Li, Q.; Kerbeshian, J.; Martsolf, J.T. Diagnosis of fetal alcohol spectrum disorders: A validity study of the fetal alcohol syndrome checklist. Alcohol 2010, 44, 605–614. [Google Scholar] [CrossRef]
  276. Ceccanti, M.; De Nicolò, S.; Mancinelli, R.; Chaldakov, G.; Carito, V.; Ceccanti, M.; Laviola, G.; Tirassa, P.; Fiore, M. NGF and BDNF long-term variations in the thyroid, testis and adrenal glands of a mouse model of fetal alcohol spectrum disorders. Ann. Ist. Super. Sanita 2013, 49, 383–390. [Google Scholar] [CrossRef] [PubMed]
  277. Fiore, M.; Laviola, G.; Aloe, L.; di Fausto, V.; Mancinelli, R.; Ceccanti, M. Early exposure to ethanol but not red wine at the same alcohol concentration induces behavioral and brain neurotrophin alterations in young and adult mice. Neurotoxicology 2009, 30, 59–71. [Google Scholar] [CrossRef] [PubMed]
  278. Fiore, M.; Mancinelli, R.; Aloe, L.; Laviola, G.; Sornelli, F.; Vitali, M.; Ceccanti, M. Hepatocyte growth factor, vascular endothelial growth factor, glial cell-derived neurotrophic factor and nerve growth factor are differentially affected by early chronic ethanol or red wine intake. Toxicol. Lett. 2009, 188, 208–213. [Google Scholar] [CrossRef]
  279. Ceccanti, M.; Mancinelli, R.; Tirassa, P.; Laviola, G.; Rossi, S.; Romeo, M.; Fiore, M. Early exposure to ethanol or red wine and long-lasting effects in aged mice. A study on nerve growth factor, brain-derived neurotrophic factor, hepatocyte growth factor, and vascular endothelial growth factor. Neurobiol. Aging 2012, 33, 359–367. [Google Scholar] [CrossRef]
  280. Kumar, A.; Singh, C.K.; LaVoie, H.A.; DiPette, D.J.; Singh, U.S. Resveratrol restores Nrf2 Level and prevents ethanol-induced toxic effects in the cerebellum of a rodent model of fetal alcohol spectrum disorders. Mol. Pharmacol. 2011, 80, 446–457. [Google Scholar] [CrossRef]
  281. Małkowska, A.; Makarowa, K.; Zawada, K.; Grzelak, M.; Zmysłowska, A. Effect of curcumin on the embryotoxic effect of ethanol in a zebrafish model. Toxicol. Vitr. 2024, 101, 105951. [Google Scholar] [CrossRef]
  282. Ceccanti, M.; Coccurello, R.; Carito, V.; Ciafrè, S.; Ferraguti, G.; Giacovazzo, G.; Mancinelli, R.; Tirassa, P.; Chaldakov, G.N.; Pascale, E.; et al. Paternal alcohol exposure in mice alters brain NGF and BDNF and increases ethanol-elicited preference in male offspring. Addict. Biol. 2016, 21, 776–787. [Google Scholar] [CrossRef]
  283. Ciafrè, S.; Ferraguti, G.; Greco, A.; Polimeni, A.; Ralli, M.; Ceci, F.M.; Ceccanti, M.; Fiore, M. Alcohol as an early life stressor: Epigenetics, metabolic, neuroendocrine and neurobehavioral implications. Neurosci. Biobehav. Rev. 2020, 118, 654–668. [Google Scholar] [CrossRef]
  284. Abel, E.L. Paternal contribution to fetal alcohol syndrome. Addict. Biol. 2004, 9, 127–133. [Google Scholar] [CrossRef]
  285. Bastianetto, S.; Ménard, C.; Quirion, R. Neuroprotective action of resveratrol. Biochim. Biophys. Acta—Mol. Basis Dis. 2015, 1852, 1195–1201. [Google Scholar] [CrossRef]
  286. Trichopoulou, A.; Critselis, E.; Vasilopoulou, E. Mediterranean diet and longevity. Eur. J. Cancer Prev. 2004, 13, 453–456. [Google Scholar] [CrossRef] [PubMed]
  287. Bastianetto, S.; Zheng, W.H.; Quirion, R. Neuroprotective abilities of resveratrol and other red wine constituents against nitric oxide-related toxicity in cultured hippocampal neurons. Br. J. Pharmacol. 2000, 131, 711–720. [Google Scholar] [CrossRef] [PubMed]
  288. Hrelia, S.; Di Renzo, L.; Bavaresco, L.; Bernardi, E.; Malaguti, M.; Giacosa, A. Moderate Wine Consumption and Health: A Narrative Review. Nutrients 2023, 15, 175. [Google Scholar] [CrossRef]
  289. Ditano-Vázquez, P.; Torres-Peña, J.D.; Galeano-Valle, F.; Pérez-Caballero, A.I.; Demelo-Rodríguez, P.; Lopez-Miranda, J.; Katsiki, N.; Delgado-Lista, J.; Alvarez-Sala-Walther, L.A. The fluid aspect of the mediterranean diet in the prevention and management of cardiovascular disease and diabetes: The role of polyphenol content in moderate consumption of wine and olive oil. Nutrients 2019, 11, 2833. [Google Scholar] [CrossRef]
  290. Lombardo, M.; Feraco, A.; Camajani, E.; Caprio, M.; Armani, A. Health Effects of Red Wine Consumption: A Narrative Review of an Issue That Still Deserves Debate. Nutrients 2023, 15, 1921. [Google Scholar] [CrossRef]
  291. Snopek, L.; Mlcek, J.; Sochorova, L.; Baron, M.; Hlavacova, I.; Jurikova, T.; Kizek, R.; Sedlackova, E.; Sochor, J. Contribution of red wine consumption to human health protection. Molecules 2018, 23, 1684. [Google Scholar] [CrossRef]
  292. Bakaloudi, D.R.; Halloran, A.; Rippin, H.L.; Oikonomidou, A.C.; Dardavesis, T.I.; Williams, J.; Wickramasinghe, K.; Breda, J.; Chourdakis, M. Intake and adequacy of the vegan diet. A systematic review of the evidence. Clin. Nutr. 2021, 40, 3503–3521. [Google Scholar] [CrossRef]
  293. Łuszczki, E.; Boakye, F.; Zielińska, M.; Dereń, K.; Bartosiewicz, A.; Oleksy, Ł.; Stolarczyk, A. Vegan diet: Nutritional components, implementation, and effects on adults’ health. Front. Nutr. 2023, 10, 1294497. [Google Scholar] [CrossRef]
  294. Bali, A.; Naik, R. The Impact of a Vegan Diet on Many Aspects of Health: The Overlooked Side of Veganism. Cureus 2023, 15, e35148. [Google Scholar] [CrossRef]
  295. Craig, W.J. Health effects of vegan diets. Am. J. Clin. Nutr. 2009, 89, 1627S–1633S. [Google Scholar] [CrossRef]
  296. Sebastiani, G.; Barbero, A.H.; Borrás-Novel, C.; Casanova, M.A.; Aldecoa-Bilbao, V.; Andreu-Fernández, V.; Tutusaus, M.P.; Martínez, S.F.; Roig, M.D.G.; García-Algar, O. The effects of vegetarian and vegan diet during pregnancy on the health of mothers and offspring. Nutrients 2019, 11, 557. [Google Scholar] [CrossRef] [PubMed]
  297. Hargreaves, S.M.; Raposo, A.; Saraiva, A.; Zandonadi, R.P. Vegetarian diet: An overview through the perspective of quality of life domains. Int. J. Environ. Res. Public Health 2021, 18, 4067. [Google Scholar] [CrossRef] [PubMed]
  298. Hargreaves, S.M.; Rosenfeld, D.L.; Moreira, A.V.B.; Zandonadi, R.P. Plant-based and vegetarian diets: An overview and definition of these dietary patterns. Eur. J. Nutr. 2023, 62, 1109–1121. [Google Scholar] [CrossRef] [PubMed]
  299. Craig, W.J. Nutrition concerns and health effects of vegetarian diets. Nutr. Clin. Pract. 2010, 25, 613–620. [Google Scholar] [CrossRef]
  300. Parker, H.W.; Vadiveloo, M.K. Diet quality of vegetarian diets compared with nonvegetarian diets: A systematic review. Nutr. Rev. 2019, 77, 144–160. [Google Scholar] [CrossRef]
  301. Ashfaq, R.; Rasul, A.; Asghar, S.; Kovács, A.; Berkó, S.; Budai-Szűcs, M. Lipid Nanoparticles: An Effective Tool to Improve the Bioavailability of Nutraceuticals. Int. J. Mol. Sci. 2023, 24, 15764. [Google Scholar] [CrossRef]
  302. Kamble, M.G.; Singh, A.; Singh, S.V.; Kamble, M.G.; Sagar, N.A.; Rani, N. Nanotechnology for encapsulation of bioactive components: A review. Discov. Food 2025, 5, 116. [Google Scholar] [CrossRef]
  303. Yang, B.; Dong, Y.; Wang, F.; Zhang, Y. Nanoformulations to enhance the bioavailability and physiological functions of polyphenols. Molecules 2020, 25, 4613. [Google Scholar] [CrossRef]
  304. Berding, K.; Vlckova, K.; Marx, W.; Schellekens, H.; Stanton, C.; Clarke, G.; Jacka, F.; Dinan, T.G.; Cryan, J.F. Diet and the Microbiota-Gut-Brain Axis: Sowing the Seeds of Good Mental Health. Adv. Nutr. 2021, 12, 1239–1285. [Google Scholar] [CrossRef]
  305. Pasinetti, G.M.; Singh, R.; Westfall, S.; Herman, F.; Faith, J.; Ho, L. The Role of the Gut Microbiota in the Metabolism of Polyphenols as Characterized by Gnotobiotic Mice. J. Alzheimer’s Dis. 2018, 63, 409–421. [Google Scholar] [CrossRef]
  306. Pratelli, G.; Tamburini, B.; Carlisi, D.; De Blasio, A.; D’Anneo, A.; Emanuele, S.; Notaro, A.; Affranchi, F.; Giuliano, M.; Seidita, A.; et al. Foodomics-Based Approaches Shed Light on the Potential Protective Effects of Polyphenols in Inflammatory Bowel Disease. Int. J. Mol. Sci. 2023, 24, 14619. [Google Scholar] [CrossRef]
  307. Todorova, V.; Ivanova, S.; Chakarov, D.; Kraev, K.; Ivanov, K. Ecdysterone and Turkesterone-Compounds with Prominent Potential in Sport and Healthy Nutrition. Nutrients 2024, 16, 1382. [Google Scholar] [CrossRef]
Figure 1. Polyphenol pathways and aging. NF-kB (nuclear factor kappa-light-chain-enhancer of activated B cells); mTOR (mammalian target of rapamycin); AMPK (5′ AMP-activated protein kinase); SIRT1 (sirtuin 1); Nrf2 (nuclear factor erythroid 2-related factor 2). Servier Medical Art by Servier is licensed under a Creative Commons Attribution 3.0 Unported License (https://creativecommons.org/licenses/by/3.0/, accessed on 29 June 2025). ↑ indicates elevation. ↓ indicates reduction.
Figure 1. Polyphenol pathways and aging. NF-kB (nuclear factor kappa-light-chain-enhancer of activated B cells); mTOR (mammalian target of rapamycin); AMPK (5′ AMP-activated protein kinase); SIRT1 (sirtuin 1); Nrf2 (nuclear factor erythroid 2-related factor 2). Servier Medical Art by Servier is licensed under a Creative Commons Attribution 3.0 Unported License (https://creativecommons.org/licenses/by/3.0/, accessed on 29 June 2025). ↑ indicates elevation. ↓ indicates reduction.
Molecules 30 02888 g001
Figure 2. Relationship between Mediterranean and Japanese polyphenol contents. The Mediterranean and Japanese diets are both associated with exceptional health benefits and longevity, and a key shared feature is their high content of polyphenols. However, these diets differ in polyphenol sources, types, and patterns of consumption, leading to distinct metabolic profiles and potentially complementary effects on aging and disease prevention. EGCG, epigallocatechin gallate. Parts of the figure were drawn by using pictures from Servier Medical Art and Microsoft PowerPoint 365 Version 2112 (https://www.microsoft.com/microsoft-365, accessed on 29 June 2025). Servier Medical Art by Servier is licensed under a Creative Commons Attribution 3.0 Unported License (https://creativecommons.org/licenses/by/3.0/, accessed on 29 June 2025).
Figure 2. Relationship between Mediterranean and Japanese polyphenol contents. The Mediterranean and Japanese diets are both associated with exceptional health benefits and longevity, and a key shared feature is their high content of polyphenols. However, these diets differ in polyphenol sources, types, and patterns of consumption, leading to distinct metabolic profiles and potentially complementary effects on aging and disease prevention. EGCG, epigallocatechin gallate. Parts of the figure were drawn by using pictures from Servier Medical Art and Microsoft PowerPoint 365 Version 2112 (https://www.microsoft.com/microsoft-365, accessed on 29 June 2025). Servier Medical Art by Servier is licensed under a Creative Commons Attribution 3.0 Unported License (https://creativecommons.org/licenses/by/3.0/, accessed on 29 June 2025).
Molecules 30 02888 g002
Table 2. Selected clinical trials of major polyphenols. ↑ indicates elevation. ↓ indicates reduction.
Table 2. Selected clinical trials of major polyphenols. ↑ indicates elevation. ↓ indicates reduction.
Polyphenol TypeFormulationDose (Daily)Population and InterventionPrimary OutcomeReferences
ResveratrolPurified capsule150 mgOverweight adults; several weeks↑ SIRT1 activity, ↓ CRP[217,218,219,220]
EGCGGreen tea extract100/600 mgMild cognitive impairment; thalassemics; diabetics; 6/12 months↑ Cognitive scores, ↓ oxidative markers[221,222,223,224]
HydroxytyrosolOlive oil phenolic extract10/15 mgMetabolic syndrome; 8/12 weeks↓ ox-LDL, ↓ IL-6[225,226]
Soy IsoflavonesSoy protein isolate60/90 mgPost-menopausal women; 1 year↑ Bone mineral density, ↓ LDL-cholesterol[227,228]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Fiore, M.; Tonchev, A.B.; Pancheva, R.Z.; Yamashima, T.; Venditti, S.; Ferraguti, G.; Terracina, S. Increasing Life Expectancy with Plant Polyphenols: Lessons from the Mediterranean and Japanese Diets. Molecules 2025, 30, 2888. https://doi.org/10.3390/molecules30132888

AMA Style

Fiore M, Tonchev AB, Pancheva RZ, Yamashima T, Venditti S, Ferraguti G, Terracina S. Increasing Life Expectancy with Plant Polyphenols: Lessons from the Mediterranean and Japanese Diets. Molecules. 2025; 30(13):2888. https://doi.org/10.3390/molecules30132888

Chicago/Turabian Style

Fiore, Marco, Anton B. Tonchev, Ruzha Z. Pancheva, Tetsumori Yamashima, Sabrina Venditti, Giampiero Ferraguti, and Sergio Terracina. 2025. "Increasing Life Expectancy with Plant Polyphenols: Lessons from the Mediterranean and Japanese Diets" Molecules 30, no. 13: 2888. https://doi.org/10.3390/molecules30132888

APA Style

Fiore, M., Tonchev, A. B., Pancheva, R. Z., Yamashima, T., Venditti, S., Ferraguti, G., & Terracina, S. (2025). Increasing Life Expectancy with Plant Polyphenols: Lessons from the Mediterranean and Japanese Diets. Molecules, 30(13), 2888. https://doi.org/10.3390/molecules30132888

Article Metrics

Back to TopTop