Next Article in Journal
Preventive Effects of Medium-Chain Fatty Acid Intake on Muscle Atrophy
Previous Article in Journal
Exploring the Associations Between Dysphagia and Health-Related Outcomes in Older Adults: Results from the ilSirente Study
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Functional Foods in Modern Nutrition Science: Mechanisms, Evidence, and Public Health Implications

by
Mónika Fekete
1,2,3,*,†,
Andrea Lehoczki
1,2,3,†,
Agata Kryczyk-Poprawa
4,
Virág Zábó
1,3,
János Tamás Varga
5,
Madarász Bálint
1,2,
Vince Fazekas-Pongor
1,2,3,
Tamás Csípő
1,2,3,
Elżbieta Rząsa-Duran
6 and
Péter Varga
1,2,3
1
Institute of Preventive Medicine and Public Health, Faculty of Medicine, Semmelweis University, 1085 Budapest, Hungary
2
Health Sciences Division, Doctoral College, Semmelweis University, 1085 Budapest, Hungary
3
Fodor Center for Prevention and Healthy Aging, Semmelweis University, 1085 Budapest, Hungary
4
Department of Inorganic Chemistry and Pharmaceutical Analytics, Faculty of Pharmacy, Jagiellonian University Medical College, 31-008 Krakow, Poland
5
Department of Pulmonology, Semmelweis University, 1085 Budapest, Hungary
6
Branch in Krakow-Hospital Pharmacy, Maria Sklodowska-Curie National Research Institute of Oncology, Garncarska 11 Str., 31-115 Krakow, Poland
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Nutrients 2025, 17(13), 2153; https://doi.org/10.3390/nu17132153 (registering DOI)
Submission received: 4 June 2025 / Revised: 25 June 2025 / Accepted: 27 June 2025 / Published: 28 June 2025
(This article belongs to the Section Nutrition and Public Health)

Abstract

In recent years, functional foods have garnered increasing scientific and public health interest due to their potential to confer physiological benefits beyond basic nutritional value. International bodies such as EFSA, FDA, and WHO define functional foods as those containing bioactive components that may contribute to the prevention and management of chronic non-communicable diseases, including cardiovascular disease, type 2 diabetes, and certain cancers. The evolving paradigm of “food as medicine” reflects a broader shift in nutritional science towards proactive, health-oriented dietary strategies. This article provides a comprehensive, interdisciplinary overview of functional foods by examining their biological mechanisms, clinical evidence, public health significance, regulatory frameworks, and future prospects—particularly in the context of advances in personalized nutrition and nutrigenomics. A thorough literature review was conducted, drawing from recent peer-reviewed studies and guidelines from key health authorities. The review highlights the roles of specific compounds such as probiotics and prebiotics in modulating the gut microbiome, flavonoids and polyphenols in anti-inflammatory processes, omega-3 fatty acids in cardiometabolic regulation, and vitamins and minerals in supporting immune function. While an expanding body of clinical trials and meta-analyses supports the health benefits of these compounds—including reductions in LDL cholesterol, improved insulin sensitivity, and mitigation of oxidative stress—the integration of functional foods into everyday diets remains challenging. Socioeconomic disparities and limited health literacy often impede their accessibility and widespread adoption in public health practice. Functional foods represent a promising component of prevention-focused modern healthcare. To maximize their impact, a coordinated, evidence-based approach is essential, involving collaboration among healthcare professionals, nutrition scientists, policymakers, and the food industry. Looking forward, innovations in artificial intelligence, microbiome research, and genomic technologies may unlock novel opportunities for the targeted and effective application of functional foods in population health.

1. Introduction

In the 21st century, one of the most pressing global public health challenges is the rising burden of non-communicable diseases (NCDs), including cardiovascular diseases [1,2,3], type 2 diabetes [4], obesity [5,6], and certain cancers [7,8]. Lifestyle-related factors—chiefly poor nutrition—play a pivotal role in the development of these chronic conditions [9,10,11,12,13,14]. Against this backdrop, the focus of nutrition science has shifted beyond basic sustenance toward disease prevention and health optimization [15,16,17,18,19,20]. In this context, the concept of functional foods has emerged as a bridge between traditional dietary habits and targeted health promotion [21].
NCDs are not only lifestyle-related—they are also fundamentally age-related diseases. Their pathogenesis is closely intertwined with the biological processes of aging [22], including cellular senescence [23,24,25,26,27,28,29,30,31,32], oxidative stress [33,34], epigenetic regulation [35,36,37,38,39,40], mitochondrial dysfunction [41,42,43,44,45,46,47], loss of proteostasis [48], chronic low-grade inflammation (“inflammaging”) [49,50,51], genomic instability [52,53], and altered neuroendocrine signaling [54]. Emerging evidence suggests that dietary factors exert significant modulatory effects on these aging mechanisms [55,56,57,58,59]. Nutrients and bioactive food components can influence the activity of key molecular pathways such as sirtuins, mTOR, AMPK, and Nrf2, modulate epigenetic patterns and mitochondrial health [60], and affect endocrine function and systemic inflammation [61]. Therefore, diet is not only a determinant of metabolic health but also a modifiable factor in shaping healthy aging trajectories [62,63,64,65,66,67].
Functional foods represent a promising strategy in addressing the complex challenge of non-communicable diseases, as they offer the potential to improve health outcomes and reduce disease risk through dietary diversification [68]. These are foods that, beyond their basic nutritional value, contain bioactive compounds capable of modulating physiological functions and contributing to the prevention of chronic diseases [69]. According to regulatory and scientific bodies such as the European Food Safety Authority (EFSA), the U.S. Food and Drug Administration (FDA), and the World Health Organization (WHO), functional foods may be either naturally occurring or intentionally modified to enhance their health-promoting properties [70]. Naturally functional foods include unprocessed fruits, nuts [71], vegetables, and whole grains [18] rich in essential nutrients, fiber [72], antioxidants, and phytochemicals [73]. In contrast, processed functional foods undergo deliberate compositional adjustments—such as enrichment, fortification, or reduction in certain ingredients—in order to increase their bioactive potential and support specific health outcomes [74]. Such modifications may involve the addition of beneficial components (e.g., omega-3 fatty acids, plant sterols, or probiotics) or a reduction in undesirable ones (e.g., saturated fats, sugars, or sodium). Functional food products can thus contain higher-than-average levels of health-enhancing ingredients or lower amounts of compounds associated with negative health impacts. In some cases, functional foods overlap with nutraceuticals—a category of products situated between conventional foods and pharmaceuticals—including concentrated extracts, dietary supplements, antioxidant-rich formulations, and encapsulated compounds such as fish oil or chitosan.
The growing popularity of the “food as medicine” paradigm reflects a broader shift in public and scientific discourse, where food is not merely a source of energy and essential nutrients but also a potential vehicle for disease prevention and therapeutic intervention [75]. This perspective is gaining importance as healthcare systems face increasing pressure from chronic disease management, while the public shows a growing preference for prevention-oriented and self-directed strategies for health maintenance [76]. However, the integration of functional foods into mainstream dietary habits remains limited by several factors. These include insufficient public understanding of scientific evidence, inconsistencies in the regulation of health claims, socioeconomic barriers to access, and a lack of standardized definitions and criteria for efficacy [77]. The gap between traditional dietary practices and evidence-based nutritional interventions continues to be a significant challenge for public health strategies.
Consequently, functional food research is not limited to nutritional science—it is a highly interdisciplinary domain that encompasses biochemistry, clinical medicine, food technology, regulatory policy, public health, and behavioral science [78]. Clinical trials and epidemiological studies increasingly support the health-promoting effects of specific functional components—such as probiotics [79], prebiotics [80], polyphenols, omega-3 fatty acids [81,82], vitamins [83], and minerals—on inflammation, metabolic health, gut microbiota composition [84], and immune modulation [85,86].
This paper aims to provide a comprehensive and critical overview of functional foods from an interdisciplinary perspective. It explores their mechanisms of action, clinical evidence, regulatory landscape, and implications for public health nutrition. In addition, the study highlights emerging directions such as nutrigenomics, personalized nutrition, and the integration of artificial intelligence into food science and health research. Ultimately, the goal is to contribute to scientifically ground public communication strategies that support the responsible inclusion of functional foods in everyday dietary practices.

2. Methods

In line with the objectives of this study, a narrative literature review was conducted, focusing on the scientific foundations, clinical evidence, public health relevance, regulatory context, and future perspectives of functional foods and their health effects. The review was carried out between November 2024 and May 2025 and followed the methodological steps outlined below.

2.1. Database and Source Selection

Relevant scientific literature was identified using several major international databases, including PubMed, Scopus, Web of Science, and ScienceDirect, along with official publications and guidelines from the EFSA, FDA, and WHO. The search focused on peer-reviewed articles published in the last ten years (2014–2025), with particular attention to randomized controlled trials (RCTs), meta-analyses, systematic reviews, and public health recommendations.

2.2. Keywords and Search Terms

The main search terms included the following: functional foods, bioactive compounds, chronic disease prevention, probiotics, polyphenols, nutrigenomics, personalized nutrition, public health, health claims, and regulatory frameworks. Boolean operators (AND, OR) were used to construct complex search queries.

Inclusion and Exclusion Criteria

Studies were included in the review if they
  • were based on human clinical research (animal studies were excluded);
  • adhered to recognized methodological quality standards;
  • investigated the physiological effects of specific functional foods or components;
  • were published in English or Hungarian.
Studies were excluded if they
  • were speculative and lacked primary data;
  • relied solely on in vitro or animal model data.
Data extraction and analysis
The following data were extracted from the included studies:
  • the type of functional component investigated (e.g., probiotic, flavonoid);
  • the duration and dosage of the intervention;
  • characteristics of the study population;
  • outcome measures evaluated (e.g., cholesterol levels, insulin sensitivity, microbiome composition, cognitive function, inflammatory markers, glycemic control, antioxidant status, immune response);
  • statistical significance and effect sizes.
The extracted data were synthesized narratively, as the heterogeneity of the included studies (in terms of intervention types, populations, and methodologies) precluded the possibility of a quantitative meta-analysis. The results were interpreted within the broader contexts of public health, nutrition science, and regulatory policy.
The methodological foundation of this study is based on the structured presentation of the two main categories of functional foods. The analysis distinguishes between, on the one hand, natural functional foods, which inherently contain bioactive compounds beneficial to health (e.g., blueberries, broccoli, kefir), and, on the other hand, intentionally modified or functionalized foods, which acquire their functional properties through industrial processing and targeted enrichment (e.g., margarine fortified with omega-3 fatty acids, yogurts supplemented with pre- and probiotics).
A differentiated examination of these categories enables a clearer understanding that not all seemingly healthy foods qualify as functional, and not all functional foods are of natural origin. This distinction is based on classifications presented in the reviewed literature, ensuring that the categorization reflects the original research context and enhances transparency in the evaluation of functional foods. Such differentiation is essential for the objective assessment and scientific interpretation of functional food efficacy.
In accordance with the official definitions provided by the EFSA and the WHO, functional foods are defined as conventional food products that contain bioactive components capable of exerting beneficial effects on one or more target functions in the body beyond basic nutritional value, and are consumed as part of a regular diet—not in the form of pills or supplements [87,88,89]. This regulatory perspective helps delineate evidence-based functional foods from general health-promoting or fortified products.
Numerous natural functional foods—such as garlic, turmeric, green tea, and various berries—are well-documented for their positive health effects. Similarly, flaxseed, olive oil, nuts, fermented foods (e.g., natto, kimchi), pomegranate, and citrus fruits have been recognized for their roles in reducing inflammation, providing antioxidant protection, supporting cardiometabolic health, and maintaining gut microbiota balance. However, due to space constraints, this manuscript focuses on the most frequently studied intervention foods, rather than providing an exhaustive overview.

3. Physiological Effects and Preventive Potential

Functional foods contain biologically active components that, beyond providing essential nutrients, may exert targeted physiological effects that contribute to the maintenance of health and the prevention of chronic non-communicable diseases [87,88,89]. According to recent scientific evidence, the consumption of these foods can positively influence the human body through a variety of well-defined biological mechanisms [21,87,88,89,90].
One of the most frequently studied aspects of functional foods is their antioxidant capacity, which helps to neutralize reactive oxygen and nitrogen species, thereby reducing oxidative stress-induced cellular damage [91,92]. This mechanism is particularly important in the prevention of neurodegenerative diseases, cardiovascular conditions, and age-related physiological decline [93]. Compounds with antimutagenic properties—such as certain polyphenols, carotenoids, and folic acid—can mitigate the damaging effects of mutagenic agents on DNA, thus decreasing the risk of malignant transformation [94]. The anticarcinogenic potential of various bioactive substances has been confirmed in numerous in vitro and in vivo studies, particularly in the case of sulforaphane, isoflavones, and conjugated linoleic acid [95].
Functional foods containing probiotics and prebiotics can favorably modulate the composition of the gut microbiota, thereby supporting immune function, maintaining intestinal barrier integrity, and reducing the risk of inflammatory bowel diseases [96]. The prebiotic effect primarily enhances the growth of beneficial bacterial strains (e.g., Bifidobacterium, Lactobacillus), while probiotics contribute to immune modulation and pathogen exclusion through their direct microbiological activity [97].
Omega-3 fatty acids—especially eicosapentaenoic acid (EPA) and docosahexaenoic acid (DHA)—play a critical role in cardiovascular protection [98,99,100,101,102]. Their effects include lowering triglyceride levels, inhibiting the synthesis of inflammatory mediators, and improving endothelial function [98]. Similarly, soluble dietary fibers such as β-glucans, which are mainly found in oats and barley, help reduce LDL cholesterol levels, thereby lowering the risk of atherosclerosis [103,104,105,106].
Many functional foods also exhibit immunomodulatory effects, particularly those containing vitamins C, D [1,107,108,109], and E, as well as trace elements like zinc and selenium [100,110,111]. These compounds enhance immune cell activity, modulate inflammatory responses, and support effective host defense against infections. In addition to strengthening immune function, certain bioactive components possess neuroprotective and neuroregulatory properties [112]. Omega-3 fatty acids, B vitamins, and polyphenols can positively influence neurotransmission and reduce neuroinflammation, helping to preserve cognitive function and decrease the risk of neuropsychiatric disorders [113,114,115].
Phytochemicals—such as curcumin and sulforaphane—are also capable of exerting epigenetic effects by modulating gene expression through mechanisms such as DNA methylation, histone acetylation, and regulation of non-coding RNAs [116]. This opens new avenues in the personalized prevention of chronic diseases [117].
A distinct category within functional foods includes those aimed at the prevention or management of obesity [118]. These typically feature reduced energy content, such as low-sugar or low-fat formulations, which play a vital role in maintaining energy balance and preventing metabolic syndrome [119]. Moreover, sodium-reduced products are of particular importance in the population-level prevention of hypertension, especially in countries where daily salt intake exceeds the recommended levels [120].
In conclusion, functional foods exhibit a wide range of biological activities that support health maintenance and contribute to the prevention of lifestyle-related diseases. However, their use should be guided by scientifically validated evidence and applied within a well-defined nutritional context. This includes careful consideration of their mechanisms of action, effective dosages, bioavailability of active compounds, and the specific needs of the target population.

4. Clinical and Epidemiological Evidence on the Efficacy of Functional Foods

Over the past two decades, there has been a substantial increase in the number of randomized controlled trials and meta-analyses investigating the physiological effects and disease-preventive potential of functional foods. The growing body of clinical and epidemiological data convincingly supports the notion that foods containing specific bioactive components may play a significant role in the primary prevention of cardiometabolic diseases, type 2 diabetes mellitus (T2DM), metabolic syndrome, and certain types of cancer.
Functional foods are generally classified into three main categories:

4.1. Naturally Functional Foods

These are whole, minimally processed foods that inherently contain health-promoting, beneficial compounds. Examples include:
  • Blueberries, which may positively influence cardiovascular and cognitive functions;
  • Broccoli, which exhibits significant anti-carcinogenic and anti-inflammatory properties;
  • Kefir, which supports gut microbiota balance and immune function through its probiotic content.

4.2. Intentionally Modified Functional Foods

This category includes foods whose composition has been deliberately altered through industrial processing to enhance their content of specific bioactive components such as nutrients, antioxidants, prebiotics, or probiotics. Representative examples include:
  • Margarine enriched with omega-3 fatty acids (cardiovascular protection)
  • Yogurt containing added prebiotics (support for gut microbiota)
  • Micronutrient-fortified products (ensuring essential nutrient intake)
  • Formulations enriched with polyphenols (enhanced antioxidant activity)

4.3. Functionalized Foods

A third emerging category includes conventional foods that are fortified during processing with functional ingredients—often plant-derived compounds like polyphenols—to enhance their antioxidant capacity or gut health benefits. While many findings in this area stem from in vitro studies, the potential for broader health applications is increasingly recognized. Examples include:
  • Polyphenol-enriched baked products, showing increased antioxidant activity
  • Pasta fortified with carrot and olive leaf flour, resulting in higher polyphenol content and enhanced antioxidant capacity
  • Yogurt with added mango peel powder, demonstrating improved in vitro prebiotic effects
There is a growing body of scientific evidence supporting the preventive and therapeutic application of such foods. However, the objective evaluation of their efficacy requires rigorous and transparent research practices. In the following sections, we provide a detailed summary of relevant studies, including tabulated data for clearer comparison.

5. Clinical and Epidemiological Evidence

The health-promoting effects of functional foods are increasingly supported by scientific research. Randomized controlled trials and systematic reviews provide reliable insights into how specific food components affect human physiology [121,122,123,124]. These studies reveal the ways in which such foods influence inflammatory processes, metabolism, hormonal balance, and cardiovascular function. Long-term prospective cohort studies offer particularly strong evidence for the preventive role of functional foods in chronic, non-communicable diseases such as metabolic syndrome, type 2 diabetes, and certain types of cancer [125,126]. This growing body of data serves as a critical foundation for developing dietary guidelines and shaping public health strategies.
Natural functional foods are traditional foods that not only provide essential nutrients but are also proven to have beneficial effects on various physiological processes in the body, thereby contributing to the prevention and treatment of chronic, non-communicable diseases [127]. These foods naturally contain bioactive compounds such as antioxidants, polyphenols, flavonoids, phytochemicals, prebiotics, and probiotics, which together support metabolism, immune responses, and the regulation of inflammatory processes [128].
Natural functional foods include, among others, fresh fruits and vegetables, whole grains, legumes, and certain fermented dairy products [129]. Through their complex nutrient and phytochemical profiles, they can influence health at multiple levels. For example, certain fruits, such as berries, are particularly rich in polyphenols, known for their antioxidant and anti-inflammatory properties [130]. Vegetables, such as cruciferous vegetables, contain compounds [glucosinolates, isothiocyanates] that have cell-protective and potentially cancer-preventive effects [131].
Whole grains—such as oats, barley, buckwheat, or quinoa—are rich in dietary fiber, B vitamins, magnesium, and phytic acid, which together contribute to stabilizing blood sugar levels, lowering cholesterol, and maintaining the balance of the gut microbiome [132]. Additionally, fermented dairy products like kefir, due to their probiotic content, can promote gut health and optimize the immune response [133].
Numerous clinical and epidemiological studies confirm that the regular consumption of these foods can have a positive impact on cardiovascular health, metabolic functions, and insulin sensitivity, as well as cognitive and mental well-being [134,135]. Therefore, natural functional foods are not only part of daily nutrient intake but can actively contribute to health maintenance and the reduction of disease risk. The following tables illustrate the health effects of natural functional foods, with clinical research clearly supporting their positive impacts on various populations and health conditions (Table 1 and Table 2). However, further long-term RCT studies will help deepen the understanding of the mechanisms and enable the targeted application of functional foods in the prevention and treatment of chronic diseases.

6. Multifaceted Health Benefits of Kefir: Cardiovascular, Metabolic, Gut Microbiota, and Cognitive Perspectives

Recent studies have consistently shown that kefir, a probiotic-fermented dairy product, offers a wide range of health benefits, particularly in the areas of cardiovascular risk reduction, metabolic health improvement, modulation of gut microbiota, and enhancement of cognitive function. These findings highlight kefir as a promising functional food with therapeutic potential.
Cardiovascular risk and inflammation: Numerous studies have demonstrated that kefir consumption can positively influence cardiovascular risk factors. In a study by Bellikci-Koyu et al. [136], participants consuming kefir experienced significant reductions in LDL-C levels, systolic and diastolic blood pressure, and pro-inflammatory cytokines such as TNF-α and IL-6. Bellikci-Koyu et al. [137] reported similar improvements in fasting insulin levels and blood pressure, although no significant between-group differences were observed compared to control groups. Additionally, Yılmaz et al. [138] observed significant reductions in erythrocyte sedimentation rate and C-reactive protein (CRP) in patients with Crohn’s disease after 4 weeks of kefir consumption, along with improvements in subjective well-being and a reduction in bloating.
Metabolic syndrome and insulin resistance: Kefir also appear to have beneficial effects on metabolic health. Alihosseini et al. [139] demonstrated that kefir supplementation significantly reduced the Homeostatic Model Assessment for Insulin Resistance (HOMA-IR), indicating improved insulin sensitivity in patients with type 2 diabetes. Additionally, both kefir and conventional fermented milk groups showed reductions in homocysteine levels. Bellikci-Koyu et al. [137] also reported significant decreases in fasting insulin, HOMA-IR, and inflammatory markers, though no statistically significant differences were found between the kefir and control groups. In a study by Fathi et al. [140], kefir intake led to reductions in total cholesterol (TC), LDL-C, and non-HDL-C, indicating a potential benefit in improving lipid profiles in overweight or obese individuals.
Gut microbiota modulation: The influence of kefir on gut microbiota composition has been widely studied, with positive results. Öneş et al. [141] reported increased microbial diversity, specifically an increase in Akkermansia muciniphila and Faecalibacterium prausnitzii, which are associated with improved metabolic and inflammatory status. Wang et al. [142] found that kefir consumption significantly increased bifidobacteria abundance in male subjects and improved the gut microbiota composition in female participants, with increases in total anaerobes and total bacterial counts. Additionally, Pražnikar et al. [143] observed significant reductions in serum zonulin levels following kefir consumption, indicating improved gut barrier function.
Cognitive function: Kefir consumption has also been associated with improvements in cognitive function, particularly in neurodegenerative diseases. In Ton et al. [144], patients with Alzheimer’s disease showed significant improvements in memory, visual–spatial abilities, and executive/language functions after 90 days of kefir supplementation. This was accompanied by reductions in inflammatory cytokines and oxidative stress markers, suggesting that kefir’s neuroprotective effects may be mediated through its anti-inflammatory and antioxidant properties. Similarly, Noori et al. [145] reported improvements in depression and appetite regulation following the consumption of probiotic-fortified kefir (Lactobacillus helveticus and Bifidobacterium longum), though no significant changes in oxidative stress or inflammatory markers were observed. The evidence presented suggests that kefir, as a probiotic dairy product, offers a range of health benefits, including a reduction in cardiovascular risk, improvement of metabolic health, modulation of gut microbiota, and enhancement of cognitive function. These effects are likely attributable to kefir’s anti-inflammatory, antioxidant, and microbiome-modulating properties. Given its potential to improve multiple aspects of health, kefir may represent a valuable dietary intervention for the prevention and management of various chronic conditions. Further research, particularly long-term, large-scale studies, is necessary to fully understand the mechanisms behind these effects and to optimize the application of kefir in clinical practice. The table below (Table 1) summarizes the results of randomized controlled trials investigating the health effects of kefir and other fermented dairy products in various populations.

7. Effects of Fruits, Vegetables, and Plant-Based Functional Foods on Cognitive and Cardiometabolic Parameters

Numerous clinical studies have demonstrated that regular consumption of fruits [176], vegetables, and their bioactive components—primarily polyphenols, dietary fibers, and vitamins—exerts beneficial effects on both cognitive functions and cardiometabolic parameters. Nilsson et al. [150], in a 16-week intervention study among elderly individuals, confirmed that increased intake of fruits and vegetables led to a significant anti-inflammatory effect, as evidenced by reductions in inflammatory markers, while physical activity levels remained unchanged. Similarly, studies by Carrillo [151] and Arcusa [152] reported that polyphenol-rich fruit and vegetable extracts significantly improved executive functions, short-term memory, and attention. These cognitive benefits were accompanied by reductions in oxidative stress markers, including oxidized LDL and CRP levels, alongside increased dopamine concentrations, supporting the biochemical basis for enhanced neural function.
Regarding the role of the microbiome, Kopf and colleagues [153] observed in overweight subjects that a six-week intake of whole grain cereals and vegetables resulted in marked decreases in inflammatory cytokines IL-6 and TNF-α, as well as increased gut microbial diversity, a key factor in anti-inflammatory mechanisms. Consistent with these findings, postprandial studies by Yuan [154] demonstrated that consumption of fiber-rich functional bread favorably modulated blood glucose levels, contributing to metabolic balance without adverse effects.
The cognitive and emotional benefits of plant-based diets were further substantiated by Campbell et al. [155] who showed that an eight-week whole-food plant-based dietary intervention in women with metastatic breast cancer improved cognitive functions, reduced fatigue, and enhanced emotional well-being, with excellent adherence.
Berries and berry-derived juices, including grape, cherry, blueberry, and citrus fruits, have also been shown in multiple studies to confer cognitive advantages, particularly in memory, reaction time, and attention, while reducing blood pressure and inflammatory markers. Lamport and colleagues [156] reported improved cerebral blood flow and cognitive test performance following grape and citrus juice consumption, whereas Haskell-Ramsay’s study [157] confirmed rapid cognitive improvements after a single dose of grape juice. Whyte [172] documented preserved cognitive performance under fatigue conditions following a berry smoothie intake. Nilsson [158] highlighted that the combined presence of polyphenols and fibers favorably modified lipid profiles by reducing total and LDL cholesterol levels while improving working memory.
Cruciferous vegetables such as broccoli also exert positive effects on cardiovascular health: Connolly’s RCT [159] demonstrated reductions in systolic blood pressure, while Riso et al. [160] observed decreased CRP levels. Moreover, Navarro’s [161] investigations revealed that the anti-inflammatory effects were modulated by Glutathione S-transferase (GST) GSTM1/GSTT1 genotypes, underscoring the role of genetic factors in dietary responsiveness.
The significance of antioxidants and vitamins was emphasized in studies by Khan [162], where blackcurrant juice consumption significantly lowered oxidative stress markers and increased plasma vitamin C levels. Similarly, Duthie [163] reported elevated vitamin and carotenoid concentrations following 12 weeks of fruit and vegetable intake, although antioxidant capacity remained unchanged. Nadeem’s research [164] indicated a reduction in serum amyloid A [SAA] protein in the HDL fraction, potentially improving HDL’s anti-atherogenic function.
Functional fruit juices also show promise for cognitive maintenance. Siddarth [165] demonstrated that a 12-month pomegranate juice intervention preserved visuospatial learning in older adults, while Ahles’ study [166] with citrus- and pomegranate-based beverages improved handgrip strength and reduced oxidative stress markers. Kennedy’s research [167] on functional breakfast consumption revealed a lower insulin response, though no significant effects were noted on mood or memory.
Several clinical studies have confirmed the beneficial effects of plant-based foods rich in polyphenols and other bioactive compounds on cognitive performance, inflammatory markers, and metabolic parameters [58]. Kent et al. [168] investigated the effects of anthocyanin-rich cherry juice (200 mL/day for 12 weeks) in older adults (≥70 years) with mild to moderate dementia. The intervention led to significant improvements in verbal fluency (p = 0.014) and short- and long-term memory performance (p ≤ 0.014), along with a reduction in systolic blood pressure (p = 0.038). However, no significant changes were observed in CRP or IL-6 levels.
In an acute single-blind crossover study, Lamport et al. [169] examined the effect of a single 500 mL dose of flavanone-rich citrus juice (70.5 mg flavanones) in healthy young adults (n = 24). Two hours post-consumption, increased cerebral blood flow was observed in the right frontal gyrus, along with improved performance in the Digit Symbol Substitution Test (DSST), although no other cognitive improvements were reported.
Rosli et al. [170] conducted a 10-week intervention using polyphenol-rich tropical fruit juice in 31 middle-aged women with impaired cognitive function. Significant improvements were found in immediate verbal memory (RAVLT) and cognitive flexibility (CTMT Trail 4, p < 0.05). Additionally, urinary levels of thyroxine and 3-methyladenine—metabolites associated with cognitive performance—were elevated.
Saeidi et al. [171] assessed the combined effects of daily broccoli supplementation (10 g/day) and combined aerobic–resistance exercise training (CARET) over 12 weeks in men with type 2 diabetes. The intervention groups showed significant reductions in plasma dectin-1 levels (p < 0.05), along with improvements in insulin resistance and various cardiometabolic markers (p < 0.05).
Whyte et al. [172] evaluated the acute effects of a single 400 mL dose of a flavonoid-rich mixed-berry smoothie in healthy young adults. While the placebo group experienced a decline in cognitive performance due to fatigue, the berry group maintained accuracy and showed improved reaction times between 2 and 6 h post-consumption. Gouvarchinghaleh et al. [173] investigated the effects of a plant-based functional soup mixture in hospitalized COVID-19 patients. Daily consumption resulted in significant reductions in inflammatory cytokines (IL-1β, IL-6, IL-17, IL-10, TNF-α), D-dimer, blood urea nitrogen (BUN), and creatinine levels (p < 0.05). Notably, CRP and potassium levels improved more prominently in the control group.
Buckwheat-based products have also demonstrated beneficial health effects. Studies by Stokić [174] and Dinu [175] reported that regular consumption of buckwheat-containing foods significantly reduces blood lipid levels and oxidative stress markers, while concurrently enhancing antioxidant capacity.
Furthermore, regular intake of tomatoes and tomato-derived products, such as tomato juice, has been shown in clinical trials to exert a broad spectrum of physiological benefits. These include improved antioxidant status, attenuation of cardiometabolic risk factors, and favorable modulation of glucose metabolism. Based on these outcomes, tomato juice may be regarded as a functional food, a classification substantiated by a substantial body of clinical evidence [177,178,179,180,181,182,183,184,185,186].
In summary, the synergistic action of bioactive compounds found in fruits, vegetables, and other plant-based functional foods plays a pivotal role not only in preserving and enhancing cognitive performance, but also in reducing inflammatory and oxidative stress markers, lowering cardiometabolic risk, and optimizing key metabolic parameters. These findings reinforce the critical role of plant-based dietary patterns in health promotion and chronic disease prevention. The following table (Table 2) summarizes the findings of clinical studies on the health effects of fruits, vegetables, and other plant-based dietary interventions.

8. Health Benefits and Molecular Mechanisms of Blueberries

Scientific interest in the health benefits of blueberries (Vaccinium corymbosum, in particular) has markedly increased in recent years, largely due to the potential preventive and therapeutic roles of polyphenols found in the fruit—especially anthocyanins (ACNs) and other bioactive (poly)phenols [187,188,189]. These compounds contribute to cardiovascular health through various biological mechanisms [190]. A targeted search in medical databases using terms such as “blueberries and cognitive function,” “blueberries and inflammation/oxidative stress,” “blueberries and metabolic health,” and “blueberries and cardiovascular health” identified over 100 published clinical studies. Due to space limitations, this article does not provide a comprehensive review of these studies. Instead, we focus on the key molecular mechanisms through which blueberry (poly)phenols exert their effects on the pathophysiology of cardiovascular diseases. We specifically address their influence on inflammatory responses, oxidative stress, endothelial function, and glucose metabolism, as these are critical factors in reducing cardiometabolic risk.

8.1. Modulation of Inflammatory Processes

One of the most prominent mechanisms of action of blueberry polyphenols is the modulation of inflammatory responses [191]. A central player in inflammation is the nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB), a transcription factor activated by oxidative stress that promotes the expression of pro-inflammatory mediators, such as cytokines (TNF-α, IL-1α, IL-6), chemokines, and adhesion molecules [192]. Numerous in vitro and in vivo studies have demonstrated that anthocyanins found in blueberries can inhibit NF-κB activation and its translocation into the nucleus, thereby reducing the production of these inflammatory mediators [193,194,195]. Moreover, blueberry-derived compounds may increase the levels of anti-inflammatory molecules such as adiponectin and interleukin-10 (IL-10), further enhancing the anti-inflammatory response [196].

8.2. Reduction in Oxidative Stress

Oxidative stress plays a key role in the development of cardiovascular diseases [3,197,198,199,200]. Blueberry polyphenols exhibit potent antioxidant properties, partly through activation of the Nrf2/Keap1 signaling pathway [201]. Activation of nuclear factor erythroid 2–related factor 2 [202] promotes the expression of various antioxidant enzymes, including superoxide dismutase (SOD), glutathione peroxidase (GPX), and catalase, which are essential for neutralizing reactive oxygen species (ROS) [203]. In addition to upregulating endogenous antioxidant defense systems, blueberry bioactives are also capable of directly scavenging free radicals by donating electrons, thereby reducing their reactivity and harmful effects on cellular structures [204].

8.3. Improvement of Vascular Function

Blueberry polyphenols exert beneficial effects on vascular homeostasis, particularly through enhancement of endothelium-dependent vasodilation [205]. A key mechanism is the activation of endothelial nitric oxide synthase (eNOS), which increases the production of nitric oxide (NO) [206]. NO plays a crucial role in vascular tone regulation, endothelial health, and blood pressure control [207]. Furthermore, blueberry polyphenols may downregulate vasoconstrictive mediators, such as cyclooxygenase-2 (COX-2), and reduce oxidative stress, thereby supporting vascular function [193]. Recent findings also suggest that these compounds may stimulate glucagon-like peptide-1 (GLP-1) activity, which further enhances eNOS expression and NO synthesis, contributing to improved endothelial relaxation [193].

8.4. Regulation of Glucose Metabolism

Blueberry polyphenols also play a role in maintaining glucose homeostasis by improving insulin sensitivity and lowering blood glucose levels [208]. Both animal and human studies have shown that anthocyanins can improve glucose tolerance and promote cellular glucose uptake through upregulation of the glucose transporter GLUT-4 [209]. Additionally, these polyphenols may suppress hepatic gluconeogenesis by downregulating the expression of key enzymes such as glucose-6-phosphatase (G6Pase) and phosphoenolpyruvate carboxykinase (PEPCK) [210]. Blueberry compounds have also been shown to activate AMP-activated protein kinase (AMPK), a critical regulator of cellular energy balance and insulin sensitivity [211].

8.5. The Role of Blueberry Polyphenols in Neurodegenerative Disease Prevention

While the cardiometabolic benefits of blueberry polyphenols have been extensively studied, their potential neuroprotective effects in the context of neurodegenerative diseases have received increasing scientific attention. Neurodegenerative conditions are strongly associated with chronic inflammation, oxidative stress, and impaired neuronal signaling. Blueberry-derived polyphenols can cross the blood–brain barrier and exert direct effects on neural tissue, reducing oxidative damage and inflammatory signaling in the brain [212,213].
Clinical trials have shown that blueberry supplementation improves memory performance, working memory, and verbal learning in older adults [187,214,215]. These cognitive enhancements are thought to be mediated by increased brain-derived neurotrophic factor levels, synaptic plasticity, and neurogenesis [216]. In a randomized, double-blind, placebo-controlled trial, healthy older adults aged 60–75 years consumed one cup of freeze-dried blueberries [approximately 24 g] daily for 90 days, resulting in significant improvements in cognitive tests related to memory and executive function [187]. These improvements were associated with elevated levels of specific polyphenol metabolites in the bloodstream. In animal studies, blueberry extracts have been found to reduce β-amyloid deposition, a hallmark of Alzheimer’s pathology, and to improve cognitive performance and motor coordination [217,218].
Collectively, current findings support the hypothesis that polyphenol-rich foods such as blueberries may contribute to the prevention and adjunctive treatment of both neurodegenerative and cardiovascular diseases. The polyphenols present in blueberries exhibit complex biological activities, including the modulation of inflammation, oxidative stress, endothelial dysfunction, glucose metabolism, and neuronal signaling. These effects are particularly relevant in conditions such as Alzheimer’s disease, where blueberry-derived compounds can cross the blood–brain barrier and exert protective actions on neural tissue.

9. Intentionally Modified Functional Foods: Categories and Physiological Benefits

A key subgroup of functional foods comprises those products that are intentionally modified to enhance their nutritional value or to promote specific health-related outcomes. In the case of these so-called intentionally modified functional foods, the addition of bioactive components (e.g., phytosterols, omega-3 fatty acids, probiotics, micronutrients) serves not only to prevent deficiencies but is also aimed at specific preventive or therapeutic purposes—such as addressing cardiovascular diseases, dyslipidemia, or gut microbiota imbalances [219].
Dairy products, particularly yogurt, have received considerable attention in the food industry during the development of functional products. This is due to their widespread consumption, high biological value, ease of flavoring, and suitability as carriers for bioactive compounds. According to recent research, yogurts enriched with probiotics and micronutrients can serve not only to enhance nutrient content but also fulfill preventive and therapeutic roles [219]. Live microorganisms incorporated into functional yogurts—especially strains of Lactobacillus and Bifidobacterium—can restore and maintain gut microbiota balance, support the immune system, and positively influence digestion. Thanks to yogurt’s popularity, low cost, and adaptability to taste preferences, it represents an excellent base for the development of functional foods. In their study, Abdi-Moghadam et al. [219] highlighted that yogurts enriched with probiotics and micronutrients have been proven to reduce the incidence of diseases associated with nutritional deficiencies and exert beneficial physiological effects on the human body.
Prebiotic components, such as inulin, also play an important role in maintaining gut health. In a randomized, double-blind, crossover study, Russo et al. [220] demonstrated that pasta enriched with inulin significantly reduced intestinal permeability [measured by the lactulose-mannitol ratio] and zonulin levels, while increasing GLP-2 hormone levels. These findings suggest that prebiotics, including inulin, can effectively support the integrity of the intestinal epithelium, promote metabolic balance, and reduce the risk of inflammatory bowel diseases.
Fortified foods containing phytosterols (PS)—including plant sterols and stanols—are increasingly recognized for their LDL-C lowering effects in the context of cardiovascular disease prevention. A targeted meta-analysis by Fontané et al. [221], based on 125 randomized controlled trials, evaluated the LDL-C-lowering effects of these foods as well as influencing factors. The analysis showed an average LDL-C reduction of 0.55 mmol/L with phytosterol-enriched products, with a dose-dependent response. The most effective reduction occurred at daily intakes above 2.5 g PS. These results align with previous meta-analyses [222,223], which found LDL-C reductions between 0.34 and 0.41 mmol/L with similar doses.
The type of fortified food also plays a significant role: PS in “butter, margarine, and spreads” proved significantly more effective than in “bread, biscuits, and cereals,” likely due to lower doses and reduced bioavailability in solid, starchy matrices. In contrast, fat-based carriers such as margarine may improve PS absorption and thus enhance efficacy. The frequency (once vs. multiple times daily), timing with or without meals, and treatment duration did not significantly influence LDL-C changes. It is important to note, however, that LDL-C reduction appears within a few weeks and remains stable for at least three months. Clinically noteworthy is the finding that phytosterols further lowered LDL-C even when taken alongside statin therapy, making them especially beneficial for individuals who do not meet lipid targets with statins alone. As such, the European Society of Cardiology (ESC/EAS, 2019) guidelines recommend phytosterol supplementation as adjunct therapy for high- and very-high-risk patients [224]. The study also notes certain limitations: substantial heterogeneity among trials and often short follow-up durations (under three months). Nonetheless, the results support that phytosterol-enriched foods are safe, effective, and that daily intakes of 2–2.5 g are recommended as part of dietary management for dyslipidemia.
Polyunsaturated omega-3 fatty acids (PUFAs) have garnered increasing attention in nutritional science over recent decades [225] due to their numerous beneficial physiological effects, particularly in the prevention of cardiovascular diseases [102,115,226,227]. These fatty acids—especially alpha-linolenic acid, eicosapentaenoic acid, and docosahexaenoic acid —contain multiple double bonds, making them highly prone to oxidation. As a result, incorporating omega-3s into shelf-stable foods presents a technological challenge, since oxidation impairs both sensory properties and bioactive efficacy.
Microencapsulation offers a promising solution, enclosing the fatty acids in a polymer (typically polysaccharide) matrix, thereby increasing their stability and enabling controlled release. This technique also helps preserve the original fatty acid composition of the oils and improves their oxidative stability. Thus, microencapsulation presents a viable approach for developing functional foods capable of delivering omega-3 fatty acids efficiently to the body [228].
Dawczynski et al. [229] investigated the effects of consuming yogurt enriched with long-chain omega-3 PUFAs on cardiovascular health. In a 10-week randomized, placebo-controlled, double-blind trial, 53 individuals with mildly elevated triglyceride levels (TAG ≥ 1.7 mmol/L) consumed yogurt containing 0 g, 0.8 g, or 3 g of PUFA daily. The 3 g/day dose significantly increased EPA and DHA levels in plasma and red blood cell membranes, improved several lipid profile parameters [reduced triglycerides and AA/EPA ratio, increased HDL cholesterol and n-3 index), and incorporated the fatty acids into cell membranes.
These fatty acids were then converted into anti-inflammatory lipid mediators (e.g., PGE3, HEPE), while levels of pro-inflammatory mediators derived from arachidonic acid decreased. The 3 g/day dose also reduced markers of lipid peroxidation (e.g., 9-HETE, 9- and 13-HODE). Furthermore, the study showed that EPA derivatives are less pro-inflammatory than their arachidonic acid-derived counterparts, reinforcing the anti-inflammatory potential of n-3 PUFAs. While markers of immune response (e.g., IL-2-producing T cells) decreased, ex vivo stimulation showed no significant difference compared to the control group. Overall, these findings suggest that regular consumption of n-3 PUFA-enriched yogurt may be an effective strategy to reduce cardiovascular risk factors, particularly in older individuals or those with inflammatory conditions.
Micronutrient deficiencies—especially iron and zinc deficiency—remain serious global public health challenges. According to Kong et al. [230], around two billion people are affected, primarily in regions where rice (Oryza sativa] and wheat (Triticum aestivum] are staple foods. Biofortification of these crops with iron and zinc presents a promising and low-cost strategy to alleviate such deficiencies. The authors discuss both genetic and agronomic approaches to biofortification and factors affecting micronutrient bioavailability. Kodkany et al. [231] demonstrated promising results in practice: among two-year-old Indian children, consumption of biofortified pearl millet led to significantly increased iron and zinc absorption. The quantities consumed were sufficient to meet physiological requirements. The study also highlighted the inhibitory effects of phytates and tannins, and the role of traditional food processing methods (e.g., fermentation, cooking] in reducing these inhibitors. Overall, biofortification appears to be an effective and sustainable strategy for mitigating micronutrient deficiencies, particularly in undernourished populations in developing countries.

10. Functionalized Foods: New Directions in Health-Promoting Nutrition

The term functional foods traditionally denote products that provide health benefits beyond basic nutrition, with effects supported by robust scientific evidence. In contrast, functionalized foods refer to products that have been enhanced with bioactive compounds or modified through technological processes to potentially confer additional physiological benefits. However, in many cases, these purported benefits have yet to be substantiated by sufficient clinical evidence [78].
This conceptual distinction is particularly relevant today, as the food industry continues to introduce and market new products, often labeling them as functional foods despite the lack of scientific validation. A common assumption is that the mere addition of a bioactive ingredient—such as a herbal extract, polyphenol, or prebiotic—is sufficient to establish the product’s functional status. Yet such modifications raise new concerns regarding the safety, mechanisms of action, and efficacy of these foods, highlighting the need for rigorous scientific investigation.
Laboratory studies have shown that bakery products enriched with polyphenols exhibit significantly increased antioxidant capacity and may reduce the formation of toxic compounds during heat processing. However, their bioavailability and sensory impact still require further evaluation [232]. Similarly, pasta fortified with carrot or olive leaf flour demonstrated higher polyphenol content and antioxidant activity than conventional pasta in vitro. These values largely persisted through simulated gastrointestinal digestion, particularly in the olive leaf–enriched variants [233]. Yogurt fortified with mango peel powder also exhibited favorable prebiotic effects under in vitro conditions, such as increased populations of beneficial gut bacteria (Lactobacillus and Bifidobacterium spp.) and enhanced production of short-chain fatty acids (SCFAs) during colonic fermentation [234]. While these findings are promising, clinical validation of the health benefits remains in the early stages.
A food product should only be considered functional if its effects are supported by randomized controlled trials and/or systematic reviews. In contrast, most newly introduced functionalized foods have not reached these scientific benchmarks: fewer than 0.1% of industry-developed dietary supplements and functional foods have been involved in at least one placebo-controlled clinical trial, and even fewer have shown statistically significant outcomes [235].
Distinguishing between functional and functionalized foods is crucial from regulatory, industrial, and consumer perspectives. In Japan, for example, the Foods for Specified Health Use (FOSHU) system requires scientific validation for functional claims, whereas in the European Union and the United States, the regulatory framework is less definitive [88]. Introducing a separate category for functionalized foods could help clarify the distinction between enriched products and truly functional foods, promoting transparent labeling and scientifically grounded health claims [88,236].
For industry stakeholders, functionalized foods present both a challenge and an opportunity. Although stricter scientific and regulatory standards may increase development costs, producers who transparently and scientifically validate their product claims can gain a competitive advantage. On the consumer side, trust significantly increases when functional claims are backed by independent clinical evidence [237]. Voluntary certification schemes—such as labels indicating “clinically proven”—could further assist in differentiating reliable products in the marketplace.
Future research priorities should include mapping the proportion of products marketed as functional without scientific substantiation, identifying optimal regulatory models, and developing labeling frameworks that clearly communicate the level of scientific evidence to consumers. In addition, long-term, longitudinal studies will be essential for determining the true health impacts of functionalized foods.

11. The Impact of Functional Foods on Public Health Nutrition

11.1. Implementation Strategies

One of the most significant strategies from a public health perspective is consumer education, aimed at scientifically influencing dietary habits. A prime example is the DASH program (Dietary Approaches to Stop Hypertension), which promotes a fiber-rich, low-sodium diet [238,239]. It has been shown to reduce hospitalizations related to hypertension by 18%, demonstrating that preventive nutrition interventions can be effective tools in mitigating the spread of chronic diseases [240,241].
Another successful example is mass food fortification, such as the addition of folic acid to flour [242]. This measure has contributed to a 50–70% reduction in neural tube defects across 78 countries [243]. Such interventions are highly cost-effective and have widespread societal impact, particularly when supported by governmental regulation and quality control [244,245].

11.2. Global Challenges

One of the major global challenges is access inequality [246]. Functional foods often cost three to five times more than conventional products, making them inaccessible for lower-income populations. This disparity exacerbates existing health inequalities [246]. Another concern is the risk of overdose and toxicity. While natural foods—like carrots—safely provide vitamin A, isolated high-dose vitamin A supplements can be potentially toxic [247]. This underscores the need for functional foods to be based on scientific evidence and rigorous safety testing.

11.3. Consumer Expectations and Sensitivity Toward Functional Foods

The success of functional foods depends not only on their health benefits but also on consumer perceptions and expectations [248]. Consumers expect such products to be tasty, practical, and sensory-equivalent to traditional foods, while also offering measurable health benefits [249].
Proper consumer information is essential: individuals must understand which health conditions a product may help prevent or manage, its limitations, and any associated risks [250]. Clear, reliable advice builds trust, which plays a key role—consumers must believe the product provides real value for them [251].
Quality assessment of functional foods involves several dimensions, including:
  • Sensory properties (taste, smell, texture);
  • Nutritional value;
  • Functional efficacy;
  • Food safety;
  • Environmental impact;
  • Intangible aspects (e.g., ethics, sustainability).
Food safety is of paramount importance [70]. Functional foods must meet the same safety standards as conventional products. However, the use of novel ingredients (e.g., traditional Eastern herbs), along with specific manufacturing, storage, and transportation conditions, may introduce new types of risks—such as the presence of toxic substances, drug interactions, or bioavailability issues during digestion [252].

11.4. Future Directions: Personalized Nutrition and Regulation of Functional Foods

One major future direction in nutrition is the development of personalized diets based on genetic profiles and gut microbiome analysis. Companies like Nutrigenomix already offer DNA tests to map individual differences in caffeine, lactose, or vitamin D metabolism [253].
At the same time, regulatory frameworks must evolve. One proposal is to establish a clear distinction between proven functional foods and “functionalized” products—i.e., foods whose functionality is not yet supported by human clinical trials [254]. This would help prevent misleading health claims. A model example is Japan’s FOSHU system, which mandates scientific substantiation for health claims [255].
Labeling systems may also evolve in the future: a “functionality scale” could be introduced, indicating the level of scientific evidence behind a product (e.g., in vitro, in vivo, randomized controlled trial). Independent third-party certification and transparent communication (e.g., using labels like “clinically proven”) could further enhance consumer confidence.
Functional foods possess genuine public health potential, but this can only be fully realized through robust scientific validation, clear regulatory frameworks, and informed consumers [21]. Moving forward, the development of a unified European or global regulatory system will be essential to ensure product quality, safety, and honest communication. The expansion of functional foods aimed at disease prevention could yield measurable benefits not only for public health, but also in economic and social terms [256].

11.5. Futures Studies—The Current State of Science

The future of functional foods is closely linked to scientific advances in the fields of nutrigenomics, artificial intelligence, next-generation probiotics, and biofortification [257]. Integrating these technologies enables the development of personalized nutrition strategies and more effective responses to public health challenges. Nutrigenomics investigates the interaction between genetic information and nutrition, with the goal of creating individualized dietary recommendations [258]. Recent research integrates genomic, proteomic, metabolomic, and transcriptomic data to understand how genes influence individual nutrient needs and disease susceptibility [259]. For example, polymorphisms in the MTHFR gene can affect folate metabolism, highlighting the need for personalized folate intake [260,261].
Artificial intelligence (AI) and machine learning are also revolutionizing nutritional science, particularly in functional food research [262]. AI allows for the analysis of complex nutritional data, the development of personalized dietary advice, and the identification of bioactive compounds in food [262]. For instance, Brightseed’s AI-powered platform Forager can detect previously unknown phytonutrients in plants that may offer health benefits [263].
Microbial fermentation and next-generation probiotics (NGPs) are key pillars of future functional food innovation [264]. NGPs such as Akkermansia muciniphila and Faecalibacterium prausnitzii show promising results in modulating the gut microbiome and in the prevention or treatment of inflammatory bowel diseases and metabolic disorders [265,266]. Advances in microbial fermentation technologies make it possible to cultivate these strains on an industrial scale while preserving their beneficial properties [267].
Finally, biofortification—especially CRISPR-Cas9–based genome editing—offers the opportunity to enhance the nutritional content of crops [268]. This includes, for example, the development of rice varieties with increased iron and folate content [269]. CRISPR can also be used to improve yield, nutrient density, and stress resistance, delivering long-term nutritional and food security benefits [270].

11.6. Legal, Economic, and Ethical Challenges

In recent years, emerging fields such as nutrigenomics, personalized nutrition, and the integration of artificial intelligence into food science and health research have gained increasing prominence. Nutrigenomics enables the molecular-level investigation of dietary effects, with particular emphasis on individual genetic variability that determines nutrient responses. Based on these insights, personalized nutrition can tailor dietary interventions according to individual health status and genetic profiles, thereby promoting the prevention and management of chronic diseases. In parallel, AI and machine learning algorithms have become powerful tools for analyzing large-scale biological and clinical datasets, facilitating the discovery of complex associations and the identification of novel predictive biomarkers. These developments contribute to advances in food safety, nutritional guidelines, and personalized therapeutic strategies—collectively exerting a significant impact on public health.
However, the rapidly expanding market of functional foods also presents a range of legal, economic, and ethical challenges that can affect both public health and consumer trust. One of the most critical concerns is the regulation and substantiation of health-related claims. In the European Union, such claims can only be approved by the European Food Safety Authority (EFSA) based on rigorous scientific evidence, including human clinical trials and a well-defined biological mechanism of action. In the United States, the Food and Drug Administration (FDA) similarly regulates health and structure/function claims. Nevertheless, in practice, certain claims may appear on the market without prior approval, provided they are supported by the scientific literature and are not misleading [271,272,273].
Despite these regulatory frameworks, the marketing of functional foods is often dominated by “nutri-marketing” strategies that promote scientifically unverified or misleading claims, particularly via social media and influencer-based content. This can lead to consumer misinformation and erosion of trust in evidence-based scientific findings. Consequently, experts increasingly call for a clear legal distinction between substantiated scientific claims and promotional content, along with supportive measures such as subsidies and educational programs to ensure equitable access. Transparent, evidence-based health communication remains essential for informed consumer decision-making and sustainable public health outcomes [274,275,276].

12. Limitations

While this study provides a comprehensive overview of the scientific and practical approaches to functional foods, several limitations should be acknowledged. First, the analysis relies primarily on accessible, peer-reviewed English-language sources, which may have resulted in the omission of certain local or less widely published research findings. Second, although numerous studies report positive health effects of functional foods, the methodological quality and strength of evidence vary considerably. Some findings have yet to be confirmed through large-scale, independent clinical trials. Third, this article does not explore in detail how access to and acceptance of functional foods may differ among populations with diverse socio-economic backgrounds, which could influence their real-world applicability. Lastly, the legal and regulatory frameworks surrounding functional foods differ significantly across countries, limiting the generalizability and global applicability of some of the approaches and conclusions presented in this review.

13. Conclusions

Functional foods are gaining increasing importance in today’s prevention-oriented healthcare paradigm, as they have the potential to go beyond basic nutritional value and contribute to the prevention or complementary management of chronic non-communicable diseases. However, realizing this potential requires close, multidisciplinary collaboration between nutrition science, medicine, and the food industry. Evidence-based and accessible communication to the general public is also essential—particularly in relation to substantiated health claims and the clear presentation of the true benefits of functional foods. While the body of scientific evidence continues to grow, practical implementation still faces several challenges, including unequal access and low levels of health literacy. In the future, advances in artificial intelligence, microbiome research, and nutrigenomics may open new pathways for the targeted and personalized use of functional foods in public health strategies.

Author Contributions

Conceptualization, M.F. and P.V.; methodology, writing—original draft preparation, V.Z. and E.R.-D.; software, writing—original draft preparation, V.F.-P.; validation, writing—original draft preparation, V.Z. and A.K.-P.; formal analysis, writing—original draft preparation A.K.-P. and J.T.V.; investigation, writing—original draft preparation, supervision, J.T.V. and E.R.-D.; resources, supervision, M.B.; data curation, supervision, writing—original draft preparation, V.F.-P., J.T.V. and E.R.-D.; writing—original draft preparation, supervision, V.F.-P., T.C. and E.R.-D.; writing—review and editing, M.F., A.K.-P., V.Z., M.B. and P.V.; visualization, supervision, A.K.-P. and M.B.; writing—review and editing, M.F., A.L. and T.C.; writing—original draft preparation, project administration, supervision, M.F. and A.L. All authors have read and agreed to the published version of the manuscript.

Funding

Open access funding was provided by Semmelweis University. This work was supported by TKP2021-NKTA-47, implemented with the support provided by the Ministry of Innovation and Technology of Hungary from the National Research, Development and Innovation Fund, and financed under the TKP2021-NKTA funding scheme. Funding through the National Cardiovascular Laboratory Program (RRF-2.3.1–21-2022–00003) was also provided by the Ministry of Innovation and Technology of Hungary from the National Research, Development and Innovation Fund. Additional support was provided by the European University for Well-Being (EuniWell) program (grant agreement number: 101004093/EUniWell/EAC-A02-2019/EAC-A02-2019–1). A.L. was supported by the EKÖP-2024-9 New National Excellence Program of the Ministry for Culture and Innovation from the source of the National Research, Development and Innovation Fund. The funding sources had no role in the study design, data collection, analysis, and interpretation, writing of the report, or the decision to submit the article for publication.

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Brandi, M.L.; Marini, F.; Parri, S.; Bandinelli, S.; Iantomasi, T.; Giusti, F.; Talluri, E.; Sini, G.; Nannipieri, F.; Battaglia, S.; et al. Association of vitamin D and bisphenol A levels with cardiovascular risk in an elderly Italian population: Results from the InCHIANTI study. Geroscience 2024, 46, 6141–6156. [Google Scholar] [CrossRef] [PubMed]
  2. Liu, D.; Aziz, N.A.; Pehlivan, G.; Breteler, M.M.B. Cardiovascular correlates of epigenetic aging across the adult lifespan: A population-based study. Geroscience 2023, 45, 1605–1618. [Google Scholar] [CrossRef] [PubMed]
  3. Nyul-Toth, A.; Patai, R.; Csiszar, A.; Ungvari, A.; Gulej, R.; Mukli, P.; Yabluchanskiy, A.; Benyo, Z.; Sotonyi, P.; Prodan, C.I.; et al. Linking peripheral atherosclerosis to blood-brain barrier disruption: Elucidating its role as a manifestation of cerebral small vessel disease in vascular cognitive impairment. Geroscience 2024, 46, 6511–6536. [Google Scholar] [CrossRef]
  4. Wikstrom Shemer, D.; Mostafaei, S.; Tang, B.; Pedersen, N.L.; Karlsson, I.K.; Fall, T.; Hagg, S. Associations between epigenetic aging and diabetes mellitus in a Swedish longitudinal study. Geroscience 2024, 46, 5003–5014. [Google Scholar] [CrossRef] [PubMed]
  5. Ungvari, Z.; Fekete, M.; Varga, P.; Lehoczki, A.; Fekete, J.T.; Ungvari, A.; Gyorffy, B. Overweight and obesity significantly increase colorectal cancer risk: A meta-analysis of 66 studies revealing a 25–57% elevation in risk. Geroscience 2024, 47, 3343–3364. [Google Scholar] [CrossRef]
  6. Cao, X.; Wang, M.; Zhou, M.; Mi, Y.; Fazekas-Pongor, V.; Major, D.; Lehoczki, A.; Guo, Y. Trends in prevalence, mortality, and risk factors of dementia among the oldest-old adults in the United States: The role of the obesity epidemic. Geroscience 2024, 46, 4761–4778. [Google Scholar] [CrossRef]
  7. Goswami, N. A dual burden dilemma: Navigating the global impact of communicable and non-communicable diseases and the way forward. Int. J. Med. Res. 2024, 12, 65–77. [Google Scholar] [CrossRef]
  8. Abdullahi, Z.M.; Kingsley, O.I.; Yusuf, M.; EO, U.; Otutu, M.; Nwose, E.U. Epidemiology, Prevention and Management of Non-Communicable Diseases (NCDS): A Review. J. Health Wellness Saf. Res. 2025, 7, 161–178. [Google Scholar]
  9. Khan, B. The Role of Lifestyle Interventions in Preventing and Managing Non-Communicable Diseases. Non-Commun. Dis. Care 2025, 2, 50–60. [Google Scholar]
  10. Fekete, M.; Szarvas, Z.; Fazekas-Pongor, V.; Feher, A.; Csipo, T.; Forrai, J.; Dosa, N.; Peterfi, A.; Lehoczki, A.; Tarantini, S.; et al. Nutrition Strategies Promoting Healthy Aging: From Improvement of Cardiovascular and Brain Health to Prevention of Age-Associated Diseases. Nutrients 2022, 15, 47. [Google Scholar] [CrossRef]
  11. Dai, Z.; Lee, S.Y.; Sharma, S.; Ullah, S.; Tan, E.C.; Brodaty, H.; Schutte, A.E.; Sachdev, P.S. A systematic review of diet and medication use among centenarians and near-centenarians worldwide. GeroScience 2024, 46, 6625–6639. [Google Scholar] [CrossRef] [PubMed]
  12. Fekete, M.; Varga, P.; Ungvari, Z.; Fekete, J.T.; Buda, A.; Szappanos, Á.; Lehoczki, A.; Mózes, N.; Grosso, G.; Godos, J. The role of the Mediterranean diet in reducing the risk of cognitive impairement, dementia, and Alzheimer’s disease: A meta-analysis. GeroScience 2025, 47, 3111–3130. [Google Scholar] [CrossRef]
  13. Šelb, J.; Cvetko, F.; Deutsch, L.; Bedrač, L.; Kuščer, E.; Maier, A.B. Personalization matters: The effect of sex in multivitamin-multimineral-based cancer prevention. GeroScience 2024, 46, 1351–1356. [Google Scholar] [CrossRef] [PubMed]
  14. Ng, T.P.; Nyunt, S.Z.; Gao, Q.; Gwee, X.; Chua, D.Q.L.; Yap, K.B. Curcumin-rich curry consumption and life expectancy: Singapore longitudinal ageing study. GeroScience 2024, 46, 969–980. [Google Scholar] [CrossRef]
  15. Ghosh, D.; Bogueva, D.; Smarta, R. Nutrition Science, Marketing Nutrition, Health Claims, and Public Policy; Academic Press: Cambridge, MA, USA, 2023. [Google Scholar]
  16. Zupo, R.; Donghia, R.; Castellana, F.; Bortone, I.; De Nucci, S.; Sila, A.; Tatoli, R.; Lampignano, L.; Sborgia, G.; Panza, F.; et al. Ultra-processed food consumption and nutritional frailty in older age. Geroscience 2023, 45, 2229–2243. [Google Scholar] [CrossRef]
  17. Chen, Y.; Zhang, Y.; Yang, H.; Li, H.; Zhou, L.; Zhang, M.; Wang, Y. Associations of sugar-sweetened, artificially sweetened, and naturally sweet juices with Alzheimer’s disease: A prospective cohort study. Geroscience 2024, 46, 1229–1240. [Google Scholar] [CrossRef]
  18. Dobreva, I.; Marston, L.; Mukadam, N. Which components of the Mediterranean diet are associated with dementia? A UK Biobank cohort study. Geroscience 2022, 44, 2541–2554. [Google Scholar] [CrossRef]
  19. Hetherington-Rauth, M.; Johnson, E.; Migliavacca, E.; Parimi, N.; Langsetmo, L.; Hepple, R.T.; Grzywinski, Y.; Corthesy, J.; Ryan, T.E.; Ferrucci, L.; et al. Nutrient Metabolites Associated With Low D3Cr Muscle Mass, Strength, and Physical Performance in Older Men. J. Gerontol. A Biol. Sci. Med. Sci. 2024, 79, glad217. [Google Scholar] [CrossRef] [PubMed]
  20. Thomas, A.; Belsky, D.W.; Gu, Y. Healthy Lifestyle Behaviors and Biological Aging in the U.S. National Health and Nutrition Examination Surveys 1999-2018. J. Gerontol. A Biol. Sci. Med. Sci. 2023, 78, 1535–1542. [Google Scholar] [CrossRef]
  21. Alongi, M.; Anese, M. Re-thinking functional food development through a holistic approach. J. Funct. Foods 2021, 81, 104466. [Google Scholar] [CrossRef]
  22. Teuliere, J.; Bernard, C.; Corel, E.; Lapointe, F.J.; Martens, J.; Lopez, P.; Bapteste, E. Network analyses unveil ageing-associated pathways evolutionarily conserved from fungi to animals. Geroscience 2023, 45, 1059–1080. [Google Scholar] [CrossRef] [PubMed]
  23. Marcozzi, S.; Bigossi, G.; Giuliani, M.E.; Giacconi, R.; Piacenza, F.; Cardelli, M.; Brunetti, D.; Segala, A.; Valerio, A.; Nisoli, E.; et al. Cellular senescence and frailty: A comprehensive insight into the causal links. Geroscience 2023, 45, 3267–3305. [Google Scholar] [CrossRef]
  24. Ting, K.K.; Coleman, P.; Kim, H.J.; Zhao, Y.; Mulangala, J.; Cheng, N.C.; Li, W.; Gunatilake, D.; Johnstone, D.M.; Loo, L.; et al. Vascular senescence and leak are features of the early breakdown of the blood-brain barrier in Alzheimer’s disease models. Geroscience 2023, 45, 3307–3331. [Google Scholar] [CrossRef]
  25. Cummings, S.R.; Lui, L.Y.; Zaira, A.; Mau, T.; Fielding, R.A.; Atkinson, E.J.; Patel, S.; LeBrasseur, N. Biomarkers of cellular senescence and major health outcomes in older adults. Geroscience 2024, 47, 3407–3415. [Google Scholar] [CrossRef] [PubMed]
  26. Ungvari, Z.; Ungvari, A.; Fekete, M.; Kiss, C.; Gyorffy, B. Senescence-related genes as prognostic indicators in breast cancer survival. Geroscience 2024, 47, 2995–3006. [Google Scholar] [CrossRef] [PubMed]
  27. Ungvari, Z.; Ungvari, A.; Bianchini, G.; Gyorffy, B. Prognostic significance of a signature based on senescence-related genes in colorectal cancer. Geroscience 2024, 46, 4495–4504. [Google Scholar] [CrossRef]
  28. Habiballa, L.; Hruby, A.; Granic, A.; Dodds, R.M.; Hillman, S.J.; Jurk, D.; Passos, J.F.; Sayer, A.A. Determining the feasibility of characterising cellular senescence in human skeletal muscle and exploring associations with muscle morphology and physical function at different ages: Findings from the MASS_Lifecourse Study. Geroscience 2024, 46, 1141–1158. [Google Scholar] [CrossRef]
  29. Fang, Y.; Peck, M.R.; Quinn, K.; Chapman, J.E.; Medina, D.; McFadden, S.A.; Bartke, A.; Hascup, E.R.; Hascup, K.N. Senolytic intervention improves cognition, metabolism, and adiposity in female APP(NL)(-F/NL-F) mice. Geroscience 2024, 47, 1123–1138. [Google Scholar] [CrossRef]
  30. Faakye, J.; Nyul-Toth, A.; Muranyi, M.; Gulej, R.; Csik, B.; Shanmugarama, S.; Tarantini, S.; Negri, S.; Prodan, C.; Mukli, P.; et al. Preventing spontaneous cerebral microhemorrhages in aging mice: A novel approach targeting cellular senescence with ABT263/navitoclax. Geroscience 2024, 46, 21–37. [Google Scholar] [CrossRef]
  31. Fielding, R.A.; Atkinson, E.J.; Aversa, Z.; White, T.A.; Heeren, A.A.; Mielke, M.M.; Cummings, S.R.; Pahor, M.; Leeuwenburgh, C.; LeBrasseur, N.K. Biomarkers of Cellular Senescence Predict the Onset of Mobility Disability and Are Reduced by Physical Activity in Older Adults. J. Gerontol. A Biol. Sci. Med. Sci. 2024, 79, glad257. [Google Scholar] [CrossRef]
  32. Zhu, Y.Z.; Liu, J.K.; Li, X.E.; Yu, Z.P.; Yang, L.Q.; Wan, Q.; Zhao, Y.; Saeed, M.; Wu, A.D.; Tian, X.L. Genome-Wide Search Links Senescence-Associated Secretory Proteins With Susceptibility for Coronary Artery Disease in Mouse and Human. J. Gerontol. A Biol. Sci. Med. Sci. 2024, 79, glae070. [Google Scholar] [CrossRef] [PubMed]
  33. Kravvariti, E.; Ntouros, P.A.; Vlachogiannis, N.I.; Pappa, M.; Souliotis, V.L.; Sfikakis, P.P. Geriatric Frailty Is Associated With Oxidative Stress, Accumulation, and Defective Repair of DNA Double-Strand Breaks Independently of Age and Comorbidities. J. Gerontol. A Biol. Sci. Med. Sci. 2023, 78, 603–610. [Google Scholar] [CrossRef]
  34. Liu, W.; Wang, J.; Wang, M.; Hou, H.; Ding, X.; Ma, L.; Liu, M. Oxidative Stress Factors Mediate the Association Between Life’s Essential 8 and Accelerated Phenotypic Aging: NHANES 2005-2018. J. Gerontol. A Biol. Sci. Med. Sci. 2024, 79, glad240. [Google Scholar] [CrossRef]
  35. Cribb, L.; Hodge, A.M.; Southey, M.C.; Giles, G.G.; Milne, R.L.; Dugue, P.A. Dietary factors and DNA methylation-based markers of ageing in 5310 middle-aged and older Australian adults. Geroscience 2024, 47, 1685–1698. [Google Scholar] [CrossRef] [PubMed]
  36. Jain, P.; Binder, A.; Chen, B.; Parada, H., Jr.; Gallo, L.C.; Alcaraz, J.; Horvath, S.; Bhatti, P.; Whitsel, E.A.; Jordahl, K.; et al. The Association of Epigenetic Age Acceleration and Multimorbidity at Age 90 in the Women’s Health Initiative. J. Gerontol. A Biol. Sci. Med. Sci. 2023, 78, 2274–2281. [Google Scholar] [CrossRef]
  37. Mak, J.K.L.; Karlsson, I.K.; Tang, B.; Wang, Y.; Pedersen, N.L.; Hagg, S.; Jylhava, J.; Reynolds, C.A. Temporal Dynamics of Epigenetic Aging and Frailty From Midlife to Old Age. J. Gerontol. A Biol. Sci. Med. Sci. 2024, 79, glad251. [Google Scholar] [CrossRef]
  38. Mutambudzi, M.; Brown, M.T.; Chen, N.W. Association of Epigenetic Age and Everyday Discrimination With Longitudinal Trajectories of Chronic Health Conditions in Older Adults. J. Gerontol. A Biol. Sci. Med. Sci. 2024, 79, glae005. [Google Scholar] [CrossRef]
  39. Gao, T.; Zheng, Y.; Joyce, B.T.; Kho, M.; Terry, J.G.; Wang, J.; Nannini, D.; Carr, J.J.; Nair, S.; Zhang, K.; et al. Epigenetic Aging Is Associated With Measures of Midlife Muscle Volume and Attenuation in CARDIA Study. J. Gerontol. A Biol. Sci. Med. Sci. 2024, 79, glad261. [Google Scholar] [CrossRef] [PubMed]
  40. Zavala, D.V.; Dzikowski, N.; Gopalan, S.; Harrington, K.D.; Pasquini, G.; Mogle, J.; Reid, K.; Sliwinski, M.; Graham-Engeland, J.E.; Engeland, C.G.; et al. Epigenetic Age Acceleration and Chronological Age: Associations With Cognitive Performance in Daily Life. J. Gerontol. A Biol. Sci. Med. Sci. 2024, 79, glad242. [Google Scholar] [CrossRef]
  41. Lopez, F.V.; O’Shea, A.; Huo, Z.; DeKosky, S.T.; Trouard, T.P.; Alexander, G.E.; Woods, A.J.; Bowers, D. Neurocognitive correlates of cerebral mitochondrial function and energy metabolism using phosphorus magnetic resonance spectroscopy in older adults. Geroscience 2024, 47, 2223–2234. [Google Scholar] [CrossRef]
  42. Tian, Q.; Zweibaum, D.A.; Qian, Y.; Oppong, R.F.; Pilling, L.C.; Casanova, F.; Atkins, J.L.; Melzer, D.; Ding, J.; Ferrucci, L. Mitochondrial DNA copy number associated dementia risk by somatic mutations and frailty. Geroscience 2024, 47, 825–835. [Google Scholar] [CrossRef] [PubMed]
  43. Seman, A.; Chandra, P.K.; Byrum, S.D.; Mackintosh, S.G.; Gies, A.J.; Busija, D.W.; Rutkai, I. Targeting mitochondria in the aged cerebral vasculature with SS-31, a proteomic study of brain microvessels. Geroscience 2023, 45, 2951–2965. [Google Scholar] [CrossRef]
  44. Qi, X.; Rusch, N.J.; Fan, J.; Mora, C.J.; Xie, L.; Mu, S.; Rabinovitch, P.S.; Zhang, H. Mitochondrial proton leak in cardiac aging. Geroscience 2023, 45, 2135–2143. [Google Scholar] [CrossRef] [PubMed]
  45. Pharaoh, G.; Kamat, V.; Kannan, S.; Stuppard, R.S.; Whitson, J.; Martin-Perez, M.; Qian, W.J.; MacCoss, M.J.; Villen, J.; Rabinovitch, P.; et al. The mitochondrially targeted peptide elamipretide (SS-31) improves ADP sensitivity in aged mitochondria by increasing uptake through the adenine nucleotide translocator (ANT). Geroscience 2023, 45, 3529–3548. [Google Scholar] [CrossRef] [PubMed]
  46. Campbell, M.D.; Samuelson, A.T.; Chiao, Y.A.; Sweetwyne, M.T.; Ladiges, W.C.; Rabinovitch, P.S.; Marcinek, D.J. Intermittent treatment with elamipretide preserves exercise tolerance in aged female mice. Geroscience 2023, 45, 2245–2255. [Google Scholar] [CrossRef]
  47. Mau, T.; Blackwell, T.L.; Cawthon, P.M.; Molina, A.J.A.; Coen, P.M.; Distefano, G.; Kramer, P.A.; Ramos, S.V.; Forman, D.E.; Goodpaster, B.H.; et al. Muscle Mitochondrial Bioenergetic Capacities Are Associated With Multimorbidity Burden in Older Adults: The Study of Muscle, Mobility and Aging. J. Gerontol. A Biol. Sci. Med. Sci. 2024, 79, glae101. [Google Scholar] [CrossRef]
  48. Norling, A.M.; Lipsitz, L.A. Exercise to Mitigate Cerebrovascular Aging: A Geroscience Perspective. J. Gerontol. A Biol. Sci. Med. Sci. 2024, 79, glae083. [Google Scholar] [CrossRef]
  49. Bencivenga, L.; Strumia, M.; Rolland, Y.; Martinez, L.; Cestac, P.; Guyonnet, S.; Andrieu, S.; Parini, A.; Lucas, A.; Vellas, B.; et al. Biomarkers of mitochondrial dysfunction and inflammaging in older adults and blood pressure variability. Geroscience 2023, 45, 797–809. [Google Scholar] [CrossRef]
  50. Andonian, B.J.; Hippensteel, J.A.; Abuabara, K.; Boyle, E.M.; Colbert, J.F.; Devinney, M.J.; Faye, A.S.; Kochar, B.; Lee, J.; Litke, R.; et al. Inflammation and aging-related disease: A transdisciplinary inflammaging framework. Geroscience 2024, 47, 515–542. [Google Scholar] [CrossRef]
  51. Pellegrino, R.; Paganelli, R.; Di Iorio, A.; Bandinelli, S.; Moretti, A.; Iolascon, G.; Sparvieri, E.; Tarantino, D.; Tanaka, T.; Ferrucci, L. Beyond Inflammaging: The Impact of Immune System Aging on Age-Related Muscle Decline, Results From the InCHIANTI Study. J. Gerontol. A Biol. Sci. Med. Sci. 2024, 79, glad238. [Google Scholar] [CrossRef]
  52. Ren, P.; Zhang, J.; Vijg, J. Somatic mutations in aging and disease. Geroscience 2024, 46, 5171–5189. [Google Scholar] [CrossRef] [PubMed]
  53. Andrawus, M.; David, G.B.; Terziyska, I.; Sharvit, L.; Bergman, A.; Barzilai, N.; Raj, S.M.; Govindaraju, D.R.; Atzmon, G. Genome integrity as a potential index of longevity in Ashkenazi Centenarian’s families. Geroscience 2024, 46, 4147–4162. [Google Scholar] [CrossRef]
  54. Stankovics, L.; Ungvari, A.; Fekete, M.; Nyul-Toth, A.; Mukli, P.; Patai, R.; Csik, B.; Gulej, R.; Conley, S.; Csiszar, A.; et al. The vasoprotective role of IGF-1 signaling in the cerebral microcirculation: Prevention of cerebral microhemorrhages in aging. Geroscience 2024, 47, 445–455. [Google Scholar] [CrossRef]
  55. Blanchard, T.; Eppe, J.; Mugnier, A.; Delfour, F.; Meynadier, A. Enhancing cognitive functions in aged dogs and cats: A systematic review of enriched diets and nutraceuticals. Geroscience 2025, 47, 2925–2947. [Google Scholar] [CrossRef]
  56. Fulton, T.L.; Wansbrough, M.R.; Mirth, C.K.; Piper, M.D.W. Short-term fasting of a single amino acid extends lifespan. Geroscience 2024, 46, 3607–3615. [Google Scholar] [CrossRef] [PubMed]
  57. Darrah, M.A.; Longtine, A.G.; Greenberg, N.T.; Mahoney, S.A.; Venkatasubramanian, R.; VanDongen, N.S.; Reisz, J.A.; D’Alessandro, A.; Seals, D.R.; de Quiros Miranda, Y.B.; et al. The influence of a human macronutrient-matched diet on phenotypes in old mice. Geroscience 2024, 47, 2293–2308. [Google Scholar] [CrossRef]
  58. Maroto-Rodriguez, J.; Delgado-Velandia, M.; Ortola, R.; Carballo-Casla, A.; Garcia-Esquinas, E.; Rodriguez-Artalejo, F.; Sotos-Prieto, M. Plant-based diets and risk of frailty in community-dwelling older adults: The Seniors-ENRICA-1 cohort. Geroscience 2023, 45, 221–232. [Google Scholar] [CrossRef]
  59. Bray, E.E.; Zheng, Z.; Tolbert, M.K.; McCoy, B.M.; Dog Aging Project, C.; Kaeberlein, M.; Kerr, K.F. Once-daily feeding is associated with better health in companion dogs: Results from the Dog Aging Project. Geroscience 2022, 44, 1779–1790. [Google Scholar] [CrossRef] [PubMed]
  60. Berry, B.J.; Mjelde, E.; Carreno, F.; Gilham, K.; Hanson, E.J.; Na, E.; Kaeberlein, M. Preservation of mitochondrial membrane potential is necessary for lifespan extension from dietary restriction. Geroscience 2023, 45, 1573–1581. [Google Scholar] [CrossRef]
  61. Orr, M.E.; Kotkowski, E.; Ramirez, P.; Bair-Kelps, D.; Liu, Q.; Brenner, C.; Schmidt, M.S.; Fox, P.T.; Larbi, A.; Tan, C.; et al. A randomized placebo-controlled trial of nicotinamide riboside in older adults with mild cognitive impairment. Geroscience 2024, 46, 665–682. [Google Scholar] [CrossRef]
  62. Ealey, K.N.; Togo, J.; Lee, J.H.; Patel, Y.; Kim, J.R.; Park, S.Y.; Sung, H.K. Intermittent fasting promotes rejuvenation of immunosenescent phenotypes in aged adipose tissue. Geroscience 2024, 46, 3457–3470. [Google Scholar] [CrossRef] [PubMed]
  63. Wang, L.; Derous, D.; Huang, X.; Mitchell, S.E.; Douglas, A.; Lusseau, D.; Wang, Y.; Speakman, J.R. The Effects of Graded Levels of Calorie Restriction: XIX. Impact of Graded Calorie Restriction on Protein Expression in the Liver. J. Gerontol. A Biol. Sci. Med. Sci. 2023, 78, 1125–1134. [Google Scholar] [CrossRef] [PubMed]
  64. Mitchell, S.E.; Togo, J.; Green, C.L.; Derous, D.; Hambly, C.; Speakman, J.R. The Effects of Graded Levels of Calorie Restriction: XX. Impact of Long-Term Graded Calorie Restriction on Survival and Body Mass Dynamics in Male C57BL/6J Mice. J. Gerontol. A Biol. Sci. Med. Sci. 2023, 78, 1953–1963. [Google Scholar] [CrossRef]
  65. Panda, S.; Maier, G.; Villareal, D.T. Targeting Energy Intake and Circadian Biology to Engage Mechanisms of Aging in Older Adults With Obesity: Calorie Restriction and Time-Restricted Eating. J. Gerontol. A Biol. Sci. Med. Sci. 2023, 78 (Suppl. 1), 79–85. [Google Scholar] [CrossRef] [PubMed]
  66. Xia, F.; Wu, Y.; Li, M.; Mitchell, S.E.; Speakman, J.R. The Effects of Graded Levels of Calorie Restriction XXI: Impact of short term graded restriction on gene expression profiles of stomach and skeletal muscle. J. Gerontol. A Biol. Sci. Med. Sci. 2024, glae299. [Google Scholar] [CrossRef]
  67. Lengele, L.; Rolland, Y.; Martinez, L.O.; Guyonnet, S.; Parini, A.; Lucas, A.; Vellas, B.; Barreto, P.S.; Group, M.D. Longitudinal Associations Between ATPase Inhibitory Factor 1, Growth Differentiation Factor-15, and Nutritional Status in Older Adults From the MAPT Study. J. Gerontol. A Biol. Sci. Med. Sci. 2024, 79, glad236. [Google Scholar] [CrossRef]
  68. Adefegha, S.A. Functional foods and nutraceuticals as dietary intervention in chronic diseases; novel perspectives for health promotion and disease prevention. J. Diet. Suppl. 2018, 15, 977–1009. [Google Scholar] [CrossRef]
  69. Hasler, C.M.; Brown, A.C. Position of the American Dietetic Association: Functional foods. J. Am. Diet. Assoc. 2009, 109, 735–746. [Google Scholar]
  70. Granato, D.; Barba, F.J.; Bursać Kovačević, D.; Lorenzo, J.M.; Cruz, A.G.; Putnik, P. Functional foods: Product development, technological trends, efficacy testing, and safety. Annu. Rev. Food Sci. Technol. 2020, 11, 93–118. [Google Scholar] [CrossRef]
  71. Bizzozero-Peroni, B.; Diaz-Goni, V.; Beneit, N.; Oliveira, A.; Jimenez-Lopez, E.; Martinez-Vizcaino, V.; Mesas, A.E. Nut consumption is associated with a lower risk of all-cause dementia in adults: A community-based cohort study from the UK Biobank. Geroscience 2024, 47, 1721–1733. [Google Scholar] [CrossRef]
  72. Kunutsor, S.K.; Jassal, D.S.; Ravandi, A.; Lehoczki, A. Dietary flaxseed: Cardiometabolic benefits and its role in promoting healthy aging. Geroscience 2025, 47, 2895–2923. [Google Scholar] [CrossRef]
  73. Vignesh, A.; Amal, T.C.; Sarvalingam, A.; Vasanth, K. A review on the influence of nutraceuticals and functional foods on health. Food Chem. Adv. 2024, 5, 100749. [Google Scholar] [CrossRef]
  74. Granato, D.; Nunes, D.S.; Barba, F.J. An integrated strategy between food chemistry, biology, nutrition, pharmacology, and statistics in the development of functional foods: A proposal. Trends Food Sci. Technol. 2017, 62, 13–22. [Google Scholar] [CrossRef]
  75. Bogan, R.R. Food-as-Medicine: An Everyday Strategy of Health. Ph.D. Thesis, City University of New York, New York, NY, USA, September 2021. [Google Scholar]
  76. Neufeld, L.M.; Ho, E.; Obeid, R.; Tzoulis, C.; Green, M.; Huber, L.G.; Stout, M.; Griffiths, J.C. Advancing nutrition science to meet evolving global health needs. Eur. J. Nutr. 2023, 62, 1–16. [Google Scholar] [CrossRef] [PubMed]
  77. Baker, M.T.; Lu, P.; Parrella, J.A.; Leggette, H.R. Consumer acceptance toward functional foods: A scoping review. Int. J. Environ. Res. Public Health 2022, 19, 1217. [Google Scholar] [CrossRef] [PubMed]
  78. Frumuzachi, O.; Flanagan, A.; Rohn, S.; Mocan, A. The dichotomy between functional and functionalized foods–A critical characterization of concepts. Food Res. Int. 2025, 208, 116173. [Google Scholar] [CrossRef]
  79. Tang, H.; Zhang, X.; Luo, N.; Huang, J.; Zhu, Y. Association of Dietary Live Microbes and Nondietary Prebiotic/Probiotic Intake With Cognitive Function in Older Adults: Evidence From NHANES. J. Gerontol. A Biol. Sci. Med. Sci. 2024, 79, glad175. [Google Scholar] [CrossRef]
  80. Ayyanar, M.P.; Vijayan, M. A review on gut microbiota and miRNA crosstalk: Implications for Alzheimer’s disease. Geroscience 2024, 47, 339–385. [Google Scholar] [CrossRef]
  81. Kim, J.; Westra, J.; Tintle, N.; Harris, W.S.; Park, Y. Association of Plasma n-3 Polyunsaturated Fatty Acid Levels and the Prevalence of Frailty in Older Adults: A Cross-Sectional Analysis of UK Biobank. J. Gerontol. A Biol. Sci. Med. Sci. 2024, 79, glae085. [Google Scholar] [CrossRef]
  82. Vyas, C.M.; Kang, J.H.; Mischoulon, D.; Cook, N.R.; Reynolds Iii, C.F.; Chang, G.; Mora, S.; De Vivo, I.; Manson, J.E.; Okereke, O.I. Apolipoprotein E and Its Association With Cognitive Change and Modification of Treatment Effects of Vitamin D3 and Omega-3s on Cognitive Change: Results From the In-Clinic Subset of a Randomized Clinical Trial. J. Gerontol. A Biol. Sci. Med. Sci. 2024, 79, glad260. [Google Scholar] [CrossRef]
  83. LeBoff, M.S.; Bischoff-Ferrari, H.A. The Effects of Vitamin D Supplementation on Musculoskeletal Health: The VITAL and DO-Health Trials. J. Gerontol. A Biol. Sci. Med. Sci. 2023, 78 (Suppl. 1), 73–78. [Google Scholar] [CrossRef]
  84. Ji, Y.; Sun, H.; Wang, Y.; Li, Y.; Piao, R.; Bu, L.; Xu, H. Characterizing the oral and gastrointestinal microbiome associated with healthy aging: Insights from long-lived populations in Northeastern China. Geroscience 2024, 47, 2275–2292. [Google Scholar] [CrossRef]
  85. Awuchi, C.G.; Okpala, C.O.R. Natural nutraceuticals, especially functional foods, their ma-jor bioactive components, formulation, and health benefits for disease prevention-An overview. J. Food Bioact. 2022, 19, 97–123. [Google Scholar] [CrossRef]
  86. Ghanbari, F.; Hasani, S.; Aghili, Z.S.; Asgary, S. The potential preventive effect of probiotics, prebiotics, and synbiotics on cardiovascular risk factors through modulation of gut microbiota: A review. Food Sci. Nutr. 2024, 12, 4569–4580. [Google Scholar] [CrossRef]
  87. Temple, N.J. A rational definition for functional foods: A perspective. Front. Nutr. 2022, 9, 957516. [Google Scholar] [CrossRef] [PubMed]
  88. Henry, C. Functional foods. Eur. J. Clin. Nutr. 2010, 64, 657–659. [Google Scholar] [CrossRef] [PubMed]
  89. Vettorazzi, A.; Lopez de Cerain, A.; Sanz-Serrano, J.; Gil, A.G.; Azqueta, A. European regulatory framework and safety assessment of food-related bioactive compounds. Nutrients 2020, 12, 613. [Google Scholar] [CrossRef] [PubMed]
  90. Granado-Lorencio, F.; Hernández-Alvarez, E. Functional foods and health effects: A nutritional biochemistry perspective. Curr. Med. Chem. 2016, 23, 2929–2957. [Google Scholar] [CrossRef]
  91. Diplock, A.; Charuleux, J.-L.; Crozier-Willi, G.; Kok, F.; Rice-Evans, C.; Roberfroid, M.; Stahl, W.; Vina-Ribes, J. Functional food science and defence against reactive oxidative species. Br. J. Nutr. 1998, 80, S77–S112. [Google Scholar] [CrossRef]
  92. Valenzuela, A.; Sanhueza, J.; Nieto, S. Natural antioxidants in functional foods: From food safety to health benefits. Grasas Y Aceites 2003, 54, 295–303. [Google Scholar] [CrossRef]
  93. Tan, B.L.; Norhaizan, M.E. The Role of Antioxidants in Longevity and Age-Related Diseases; Springer: Cham, Switzerland, 2021. [Google Scholar]
  94. Ağagündüz, D.; Şahin, D.O.; Yılmaz, B.; Ekenci, K.D.; Ozer, S.D.; Capasso, R. Cruciferous Vegetables and Their Bioactive Metabolites: From Prevention to Novel Therapies of Colorectal Cancer. Evid.-Based Complement. Altern. Med. 2022, 2022, 1534083. [Google Scholar] [CrossRef] [PubMed]
  95. Bhutta, Z.A.; Choi, K.C. Phytochemicals as Novel Therapeutics for Triple-Negative Breast Cancer: A Comprehensive Review of Current Knowledge. Phytother. Res. 2025, 39, 364–396. [Google Scholar] [CrossRef]
  96. Chandra, P.; Sharma, H.; Sachan, N. The Potential Role of Prebiotics, Probiotics, and Synbiotics in Cancer Prevention and Therapy. In Synbiotics in Metabolic Disorders; CRC Press: Boca Raton, FL, USA, 2024; pp. 191–213. [Google Scholar]
  97. Onoruoiza, M.; Ayodele, A.; David-Momoh, T. A review on the effects of functional food on humans and microorganisms. Int. J. Probiotics Diet. 2024, 4, 1–14. [Google Scholar]
  98. Borghi, C.; Cicero, A.F. Omega-3 polyunsaturated fatty acids: Their potential role in blood pressure prevention and management. Heart Int. 2006, 2, 182618680600200205. [Google Scholar] [CrossRef]
  99. Bischoff-Ferrari, H.A.; Vellas, B.; Rizzoli, R.; Kressig, R.W.; da Silva, J.A.P.; Blauth, M.; Felson, D.T.; McCloskey, E.V.; Watzl, B.; Hofbauer, L.C.; et al. Effect of Vitamin D Supplementation, Omega-3 Fatty Acid Supplementation, or a Strength-Training Exercise Program on Clinical Outcomes in Older Adults: The DO-HEALTH Randomized Clinical Trial. JAMA 2020, 324, 1855–1868. [Google Scholar] [CrossRef]
  100. Fekete, M.; Csípő, T.; Fazekas-Pongor, V.; Fehér, Á.; Szarvas, Z.; Kaposvári, C.; Horváth, K.; Lehoczki, A.; Tarantini, S.; Varga, J.T. The Effectiveness of Supplementation with Key Vitamins, Minerals, Antioxidants and Specific Nutritional Supplements in COPD-A Review. Nutrients 2023, 15, 2741. [Google Scholar] [CrossRef]
  101. Fekete, M.; Lehoczki, A.; Tarantini, S.; Fazekas-Pongor, V.; Csipo, T.; Csizmadia, Z.; Varga, J.T. Improving Cognitive Function with Nutritional Supplements in Aging: A Comprehensive Narrative Review of Clinical Studies Investigating the Effects of Vitamins, Minerals, Antioxidants, and Other Dietary Supplements. Nutrients 2023, 15, 5116. [Google Scholar] [CrossRef]
  102. Fekete, M.; Szőllősi, G.; Németh, A.N.; Varga, J.T. Clinical value of omega-3 polyunsaturated fatty acid supplementation in chronic obstructive pulmonary disease. Orvosi Hetil. 2021, 162, 23–30. [Google Scholar] [CrossRef]
  103. Immerstrand, T. Cholesterol-Lowering Properties of Oats: Effects of Processing and the Role of Oat Components. Ph.D. Thesis, Lund University, Lund, Sweden, 2010. [Google Scholar]
  104. Andersson, K.E.; Immerstrand, T.; Swärd, K.; Bergenståhl, B.; Lindholm, M.W.; Öste, R.; Hellstrand, P. Effects of oats on plasma cholesterol and lipoproteins in C57BL/6 mice are substrain specific. Br. J. Nutr. 2010, 103, 513–521. [Google Scholar] [CrossRef]
  105. Ungvari, Z.; Fekete, M.; Fekete, J.T.; Grosso, G.; Ungvari, A.; Győrffy, B. Adherence to the Mediterranean diet and its protective effects against colorectal cancer: A meta-analysis of 26 studies with 2,217,404 participants. GeroScience 2024, 47, 1105–1121. [Google Scholar] [CrossRef]
  106. Vince, F.-P.; Zoltán, U.; Mónika, F. Táplálkozási stratégiák az egészséges öregedésért: Krónikus, életkorral összefüggő betegségek. Sci. J. Hung. Assoc. Gerontol. Geriatr. 2024, 9, 4–12. [Google Scholar]
  107. McGee, K.C.; Sullivan, J.; Hazeldine, J.; Schmunk, L.J.; Martin-Herranz, D.E.; Jackson, T.; Lord, J.M. A combination nutritional supplement reduces DNA methylation age only in older adults with a raised epigenetic age. Geroscience 2024, 46, 4333–4347. [Google Scholar] [CrossRef] [PubMed]
  108. Bray, N.W.; Pieruccini-Faria, F.; Witt, S.T.; Bartha, R.; Doherty, T.J.; Nagamatsu, L.S.; Almeida, Q.J.; Liu-Ambrose, T.; Middleton, L.E.; Bherer, L.; et al. Combining exercise with cognitive training and vitamin D(3) to improve functional brain connectivity (FBC) in older adults with mild cognitive impairment (MCI). Results from the SYNERGIC trial. Geroscience 2023, 45, 1967–1985. [Google Scholar] [CrossRef]
  109. Vetter, V.M.; Sommerer, Y.; Kalies, C.H.; Spira, D.; Bertram, L.; Demuth, I. Vitamin D supplementation is associated with slower epigenetic aging. Geroscience 2022, 44, 1847–1859. [Google Scholar] [CrossRef] [PubMed]
  110. Wintergerst, E.S.; Maggini, S.; Hornig, D.H. Contribution of selected vitamins and trace elements to immune function. Ann. Nutr. Metab. 2007, 51, 301–323. [Google Scholar] [CrossRef]
  111. Banik, S.P.; Ghosh, R.B.; Downs, B.W.; Chakraborty, S.; Bagchi, M.; Chakraborty, T.R.; Bagchi, D. Functional foods beyond nutrition: Therapeutic interventions to combat COVID-19 and other viral diseases. J. Food Bioact. 2021, 15, 63–73. [Google Scholar] [CrossRef]
  112. Bakhru, A. Nutrition and Integrative Medicine for Clinicians: Volume Two; CRC Press: Boca Raton, FL, USA, 2023. [Google Scholar]
  113. Wilczyńska, A.; Singh, R.B.; De Meester, F. Nutritional modulators of neuropsychiatric dysfunction. Open Nutraceuticals J. 2011, 4, 52–60. [Google Scholar] [CrossRef]
  114. De Domenico, S.; Giudetti, A.M. Nutraceutical intervention in ageing brain. J. Gerontol. Geriatr. 2017, 65, 79–92. [Google Scholar]
  115. Fekete, M.; Major, D.; Feher, A.; Fazekas-Pongor, V.; Lehoczki, A. Geroscience and pathology: A new frontier in understanding age-related diseases. Pathol. Oncol. Res. 2024, 30, 1611623. [Google Scholar] [CrossRef]
  116. Pop, S.; Enciu, A.M.; Tarcomnicu, I.; Gille, E.; Tanase, C. Phytochemicals in cancer prevention: Modulating epigenetic alterations of DNA methylation. Phytochem. Rev. 2019, 18, 1005–1024. [Google Scholar] [CrossRef]
  117. Hsieh, H.-H.; Kuo, M.-Z.; Chen, I.-A.; Lin, C.-J.; Hsu, V.; HuangFu, W.-C.; Wu, T.-Y. Epigenetic Modifications as Novel Therapeutic Strategies of Cancer Chemoprevention by Phytochemicals. Pharm. Res. 2025, 42, 69–78. [Google Scholar] [CrossRef]
  118. Milner, J.A. Functional foods: The US perspective. Am. J. Clin. Nutr. 2000, 71, 1654S–1659S. [Google Scholar] [CrossRef] [PubMed]
  119. Cozzolino, F.; Lecce, L.; Frisullo, P.; Conte, A.; Alessandro Del Nobile, M. Functional food: Product development and health benefits. Recent. Pat. Eng. 2012, 6, 2–19. [Google Scholar] [CrossRef]
  120. Boon, C.S.; Taylor, C.L.; Henney, J.E. Strategies to Reduce Sodium Intake in the United States; National Academies Press: Washington, DC, USA, 2010. [Google Scholar]
  121. Flore, G.; Deledda, A.; Lombardo, M.; Armani, A.; Velluzzi, F. Effects of Functional and Nutraceutical Foods in the Context of the Mediterranean Diet in Patients Diagnosed with Breast Cancer. Antioxidants 2023, 12, 1845. [Google Scholar] [CrossRef] [PubMed]
  122. Wang, L.L.; Zhang, P.H.; Yan, H.H. Functional foods and dietary supplements in the management of non-alcoholic fatty liver disease: A systematic review and meta-analysis. Front. Nutr. 2023, 10, 1014010. [Google Scholar] [CrossRef]
  123. Venkatakrishnan, K.; Chiu, H.F.; Wang, C.K. Extensive review of popular functional foods and nutraceuticals against obesity and its related complications with a special focus on randomized clinical trials. Food Funct. 2019, 10, 2313–2329. [Google Scholar] [CrossRef]
  124. Willis, H.J.; Slavin, J.L. The Influence of Diet Interventions Using Whole, Plant Food on the Gut Microbiome: A Narrative Review. J. Acad. Nutr. Diet. 2020, 120, 608–623. [Google Scholar] [CrossRef] [PubMed]
  125. Singh, R.B. Functional Foods and Nutraceuticals in Metabolic and Non-Communicable Diseases; Academic Press: Cambridge, MA, USA, 2021. [Google Scholar]
  126. Boone-Villa, V.D.; Obregón-Sánchez, N.H.; Del Bosque-Moreno, J.; Aguirre-Joya, J.A. Trends in functional food in non-communicable diseases. In Handbook of Research on Food Science and Technology; Apple Academic Press: Palm Bay, FL, USA, 2019; pp. 1–32. [Google Scholar]
  127. Hossain, A.; Rahman, M.J. Safety, nutrition and functionality of the traditional foods. Tradit. Foods: Hist. Prep. Process. Saf. 2019, 219–238. [Google Scholar]
  128. Mondal, S.; Soumya, N.P.P.; Mini, S.; Sivan, S.K. Bioactive compounds in functional food and their role as therapeutics. Bioact. Compd. Health Dis. 2021, 4, 24–39. [Google Scholar] [CrossRef]
  129. Essa, M.M.; Bishir, M.; Bhat, A.; Chidambaram, S.B.; Al-Balushi, B.; Hamdan, H.; Govindarajan, N.; Freidland, R.P.; Qoronfleh, M.W. Functional foods and their impact on health. J. Food Sci. Technol. 2023, 60, 820–834. [Google Scholar] [CrossRef]
  130. Pap, N.; Fidelis, M.; Azevedo, L.; do Carmo, M.A.V.; Wang, D.; Mocan, A.; Pereira, E.P.R.; Xavier-Santos, D.; Sant’Ana, A.S.; Yang, B. Berry polyphenols and human health: Evidence of antioxidant, anti-inflammatory, microbiota modulation, and cell-protecting effects. Curr. Opin. Food Sci. 2021, 42, 167–186. [Google Scholar] [CrossRef]
  131. Cirignano, S.M.; Morgan, K.T. The Role of Carotenoid-and Glucosinolate-Containing Vegetables in Cancer Prevention and Their Promotion in Clinical Practice. Top. Clin. Nutr. 2014, 29, 33–46. [Google Scholar] [CrossRef]
  132. Joye, I.J. Dietary fibre from whole grains and their benefits on metabolic health. Nutrients 2020, 12, 3045. [Google Scholar] [CrossRef]
  133. Kaur, H.; Kaur, G.; Ali, S.A. Dairy-based probiotic-fermented functional foods: An update on their health-promoting properties. Fermentation 2022, 8, 425. [Google Scholar] [CrossRef]
  134. Aguiar, L.M.; Geraldi, M.V.; Cazarin, C.B.B.; Junior, M.R.M. Functional food consumption and its physiological effects. In Bioactive Compounds; Elsevier: Amsterdam, The Netherlands, 2019; pp. 205–225. [Google Scholar]
  135. Alkhatib, A.; Tsang, C.; Tiss, A.; Bahorun, T.; Arefanian, H.; Barake, R.; Khadir, A.; Tuomilehto, J. Functional foods and lifestyle approaches for diabetes prevention and management. Nutrients 2017, 9, 1310. [Google Scholar] [CrossRef]
  136. Bellikci-Koyu, E.; Sarer-Yurekli, B.P.; Karagozlu, C.; Aydin-Kose, F.; Ozgen, A.G.; Buyuktuncer, Z. Probiotic kefir consumption improves serum apolipoprotein A1 levels in metabolic syndrome patients: A randomized controlled clinical trial. Nutr. Res. 2022, 102, 59–70. [Google Scholar] [CrossRef] [PubMed]
  137. Bellikci-Koyu, E.; Sarer-Yurekli, B.P.; Akyon, Y.; Aydin-Kose, F.; Karagozlu, C.; Ozgen, A.G.; Brinkmann, A.; Nitsche, A.; Ergunay, K.; Yilmaz, E.; et al. Effects of Regular Kefir Consumption on Gut Microbiota in Patients with Metabolic Syndrome: A Parallel-Group, Randomized, Controlled Study. Nutrients 2019, 11, 2089. [Google Scholar] [CrossRef]
  138. Yılmaz, İ.; Dolar, M.E.; Özpınar, H. Effect of administering kefir on the changes in fecal microbiota and symptoms of inflammatory bowel disease: A randomized controlled trial. Turk. J. Gastroenterol. 2019, 30, 242–253. [Google Scholar] [CrossRef]
  139. Alihosseini, N.; Moahboob, S.A.; Farrin, N.; Mobasseri, M.; Taghizadeh, A.; Ostadrahimi, A.R. Effect of Probiotic Fermented Milk (Kefir) on Serum Level of Insulin and Homocysteine in Type 2 Diabetes Patients. Acta Endocrinol. 2017, 13, 431–436. [Google Scholar] [CrossRef]
  140. Fathi, Y.; Ghodrati, N.; Zibaeenezhad, M.J.; Faghih, S. Kefir drink causes a significant yet similar improvement in serum lipid profile, compared with low-fat milk, in a dairy-rich diet in overweight or obese premenopausal women: A randomized controlled trial. J. Clin. Lipidol. 2017, 11, 136–146. [Google Scholar] [CrossRef]
  141. Öneş, E.; Zavotçu, M.; Nisan, N.; Baş, M.; Sağlam, D. Effects of Kefir Consumption on Gut Microbiota and Athletic Performance in Professional Female Soccer Players: A Randomized Controlled Trial. Nutrients 2025, 17, 512. [Google Scholar] [CrossRef] [PubMed]
  142. Wang, M.C.; Zaydi, A.I.; Lin, W.H.; Lin, J.S.; Liong, M.T.; Wu, J.J. Putative Probiotic Strains Isolated from Kefir Improve Gastrointestinal Health Parameters in Adults: A Randomized, Single-Blind, Placebo-Controlled Study. Probiotics Antimicrob. Proteins 2020, 12, 840–850. [Google Scholar] [CrossRef]
  143. Pražnikar, Z.J.; Kenig, S.; Vardjan, T.; Bizjak, M.Č.; Petelin, A. Effects of kefir or milk supplementation on zonulin in overweight subjects. J. Dairy. Sci. 2020, 103, 3961–3970. [Google Scholar] [CrossRef]
  144. Ton, A.M.M.; Campagnaro, B.P.; Alves, G.A.; Aires, R.; Côco, L.Z.; Arpini, C.M.; Guerra, E.O.T.; Campos-Toimil, M.; Meyrelles, S.S.; Pereira, T.M.C.; et al. Oxidative Stress and Dementia in Alzheimer’s Patients: Effects of Synbiotic Supplementation. Oxid. Med. Cell Longev. 2020, 2020, 2638703. [Google Scholar] [CrossRef] [PubMed]
  145. Noori, M.; Shateri, Z.; Babajafari, S.; Eskandari, M.H.; Parastouei, K.; Ghasemi, M.; Afshari, H.; Samadi, M. The effect of probiotic-fortified kefir on depression, appetite, oxidative stress, and inflammatory parameters in Iranian overweight and obese elderly: A randomized, double-blind, placebo-controlled clinical trial. J. Health Popul. Nutr. 2025, 44, 30. [Google Scholar] [CrossRef] [PubMed]
  146. Tatullo, M.; Marrelli, B.; Benincasa, C.; Aiello, E.; Amantea, M.; Gentile, S.; Leonardi, N.; Balestrieri, M.L.; Campanile, G. Potential impact of functional biomolecules-enriched foods on human health: A randomized controlled clinical trial. Int. J. Med. Sci. 2022, 19, 563–571. [Google Scholar] [CrossRef]
  147. Ostadrahimi, A.; Taghizadeh, A.; Mobasseri, M.; Farrin, N.; Payahoo, L.; Beyramalipoor Gheshlaghi, Z.; Vahedjabbari, M. Effect of probiotic fermented milk (kefir) on glycemic control and lipid profile in type 2 diabetic patients: A randomized double-blind placebo-controlled clinical trial. Iran. J. Public. Health 2015, 44, 228–237. [Google Scholar]
  148. Bourrie, B.C.T.; Forgie, A.J.; Makarowski, A.; Cotter, P.D.; Richard, C.; Willing, B.P. Consumption of kefir made with traditional microorganisms resulted in greater improvements in LDL cholesterol and plasma markers of inflammation in males when compared to a commercial kefir: A randomized pilot study. Appl. Physiol. Nutr. Metab. 2023, 48, 668–677. [Google Scholar] [CrossRef]
  149. O’Brien, K.V.; Stewart, L.K.; Forney, L.A.; Aryana, K.J.; Prinyawiwatkul, W.; Boeneke, C.A. The effects of postexercise consumption of a kefir beverage on performance and recovery during intensive endurance training. J. Dairy. Sci. 2015, 98, 7446–7449. [Google Scholar] [CrossRef]
  150. Nilsson, A.; Cano, A.; Bergens, O.; Kadi, F. Randomized Controlled Trial for Promotion of Healthy Eating in Older Adults by Increasing Consumption of Plant-Based Foods: Effect on Inflammatory Biomarkers. Nutrients 2021, 13, 3753. [Google Scholar] [CrossRef]
  151. Carrillo, J.; Arcusa, R.; Zafrilla, M.P.; Marhuenda, J. Effects of Fruit and Vegetable-Based Nutraceutical on Cognitive Function in a Healthy Population: Placebo-Controlled, Double-Blind, and Randomized Clinical Trial. Antioxidants 2021, 10, 116. [Google Scholar] [CrossRef]
  152. Arcusa, R.; Carrillo, J.; Xandri-Martínez, R.; Cerdá, B.; Villaño, D.; Marhuenda, J.; Zafrilla, M.P. Effects of a Fruit and Vegetable-Based Nutraceutical on Biomarkers of Inflammation and Oxidative Status in the Plasma of a Healthy Population: A Placebo-Controlled, Double-Blind, and Randomized Clinical Trial. Molecules 2021, 26, 3604. [Google Scholar] [CrossRef] [PubMed]
  153. Kopf, J.C.; Suhr, M.J.; Clarke, J.; Eyun, S.I.; Riethoven, J.M.; Ramer-Tait, A.E.; Rose, D.J. Role of whole grains versus fruits and vegetables in reducing subclinical inflammation and promoting gastrointestinal health in individuals affected by overweight and obesity: A randomized controlled trial. Nutr. J. 2018, 17, 72. [Google Scholar] [CrossRef] [PubMed]
  154. Yuan, J.Y.; Smeele, R.J.; Harington, K.D.; van Loon, F.M.; Wanders, A.J.; Venn, B.J. The effects of functional fiber on postprandial glycemia, energy intake, satiety, palatability and gastrointestinal wellbeing: A randomized crossover trial. Nutr. J. 2014, 13, 76. [Google Scholar] [CrossRef] [PubMed]
  155. Campbell, E.K.; Campbell, T.M.; Culakova, E.; Blanchard, L.; Wixom, N.; Guido, J.J.; Fetten, J.; Huston, A.; Shayne, M.; Janelsins, M.C.; et al. A whole food, plant-based randomized controlled trial in metastatic breast cancer: Feasibility, nutrient, and patient-reported outcomes. Breast Cancer Res. Treat. 2024, 206, 273–283. [Google Scholar] [CrossRef]
  156. Lamport, D.J.; Lawton, C.L.; Merat, N.; Jamson, H.; Myrissa, K.; Hofman, D.; Chadwick, H.K.; Quadt, F.; Wightman, J.D.; Dye, L. Concord grape juice, cognitive function, and driving performance: A 12-wk, placebo-controlled, randomized crossover trial in mothers of preteen children. Am. J. Clin. Nutr. 2016, 103, 775–783. [Google Scholar] [CrossRef]
  157. Haskell-Ramsay, C.; Stuart, R.; Okello, E.; Watson, A. Cognitive and mood improvements following acute supplementation with purple grape juice in healthy young adults. Eur. J. Nutr. 2017, 56, 2621–2631. [Google Scholar] [CrossRef]
  158. Nilsson, A.; Salo, I.; Plaza, M.; Björck, I. Effects of a mixed berry beverage on cognitive functions and cardiometabolic risk markers; A randomized cross-over study in healthy older adults. PLoS ONE 2017, 12, e0188173. [Google Scholar] [CrossRef]
  159. Connolly, E.L.; Liu, A.H.; Radavelli-Bagatini, S.; Shafaei, A.; Boyce, M.C.; Wood, L.G.; McCahon, L.; Koch, H.; Sim, M.; Hill, C.R.; et al. Cruciferous vegetables lower blood pressure in adults with mildly elevated blood pressure in a randomized, controlled, crossover trial: The VEgetableS for vaScular hEaLth (VESSEL) study. BMC Med. 2024, 22, 353. [Google Scholar] [CrossRef]
  160. Riso, P.; Vendrame, S.; Del Bo, C.; Martini, D.; Martinetti, A.; Seregni, E.; Visioli, F.; Parolini, M.; Porrini, M. Effect of 10-day broccoli consumption on inflammatory status of young healthy smokers. Int. J. Food Sci. Nutr. 2014, 65, 106–111. [Google Scholar] [CrossRef]
  161. Navarro, S.L.; Schwarz, Y.; Song, X.; Wang, C.Y.; Chen, C.; Trudo, S.P.; Kristal, A.R.; Kratz, M.; Eaton, D.L.; Lampe, J.W. Cruciferous vegetables have variable effects on biomarkers of systemic inflammation in a randomized controlled trial in healthy young adults. J. Nutr. 2014, 144, 1850–1857. [Google Scholar] [CrossRef] [PubMed]
  162. Khan, F.; Ray, S.; Craigie, A.M.; Kennedy, G.; Hill, A.; Barton, K.L.; Broughton, J.; Belch, J.J. Lowering of oxidative stress improves endothelial function in healthy subjects with habitually low intake of fruit and vegetables: A randomized controlled trial of antioxidant- and polyphenol-rich blackcurrant juice. Free Radic. Biol. Med. 2014, 72, 232–237. [Google Scholar] [CrossRef] [PubMed]
  163. Duthie, S.J.; Duthie, G.G.; Russell, W.R.; Kyle, J.A.M.; Macdiarmid, J.I.; Rungapamestry, V.; Stephen, S.; Megias-Baeza, C.; Kaniewska, J.J.; Shaw, L.; et al. Effect of increasing fruit and vegetable intake by dietary intervention on nutritional biomarkers and attitudes to dietary change: A randomised trial. Eur. J. Nutr. 2018, 57, 1855–1872. [Google Scholar] [CrossRef]
  164. Nadeem, N.; Woodside, J.V.; Neville, C.E.; McCall, D.O.; McCance, D.; Edgar, D.; Young, I.S.; McEneny, J. Serum amyloid A-related inflammation is lowered by increased fruit and vegetable intake, while high-sensitive C-reactive protein, IL-6 and E-selectin remain unresponsive. Br. J. Nutr. 2014, 112, 1129–1136. [Google Scholar] [CrossRef] [PubMed]
  165. Siddarth, P.; Li, Z.; Miller, K.J.; Ercoli, L.M.; Merril, D.A.; Henning, S.M.; Heber, D.; Small, G.W. Randomized placebo-controlled study of the memory effects of pomegranate juice in middle-aged and older adults. Am. J. Clin. Nutr. 2020, 111, 170–177. [Google Scholar] [CrossRef]
  166. Ahles, S.; Cuijpers, I.; Hartgens, F.; Troost, F.J. The Effect of a Citrus and Pomegranate Complex on Physical Fitness and Mental Well-Being in Healthy Elderly: A Randomized Placebo-Controlled Trial. J. Nutr. Health Aging 2022, 26, 839–846. [Google Scholar] [CrossRef]
  167. Kennedy, S.J.; Ryan, L.; Clegg, M.E. The Effects of a Functional Food Breakfast on Gluco-Regulation, Cognitive Performance, Mood, and Satiety in Adults. Nutrients 2020, 12, 2974. [Google Scholar] [CrossRef]
  168. Kent, K.; Charlton, K.; Roodenrys, S.; Batterham, M.; Potter, J.; Traynor, V.; Gilbert, H.; Morgan, O.; Richards, R. Consumption of anthocyanin-rich cherry juice for 12 weeks improves memory and cognition in older adults with mild-to-moderate dementia. Eur. J. Nutr. 2017, 56, 333–341. [Google Scholar] [CrossRef]
  169. Lamport, D.J.; Pal, D.; Macready, A.L.; Barbosa-Boucas, S.; Fletcher, J.M.; Williams, C.M.; Spencer, J.P.; Butler, L.T. The effects of flavanone-rich citrus juice on cognitive function and cerebral blood flow: An acute, randomised, placebo-controlled cross-over trial in healthy, young adults. Br. J. Nutr. 2016, 116, 2160–2168. [Google Scholar] [CrossRef]
  170. Rosli, H.; Shahar, S.; Rajab, N.F.; Che Din, N.; Haron, H. The effects of polyphenols-rich tropical fruit juice on cognitive function and metabolomics profile—A randomized controlled trial in middle-aged women. Nutr. Neurosci. 2022, 25, 1577–1593. [Google Scholar] [CrossRef]
  171. Saeidi, A.; Soltani, M.; Daraei, A.; Nohbaradar, H.; Haghighi, M.M.; Khosravi, N.; Johnson, K.E.; Laher, I.; Hackney, A.C.; VanDusseldorp, T.A.; et al. The Effects of Aerobic-Resistance Training and Broccoli Supplementation on Plasma Dectin-1 and Insulin Resistance in Males with Type 2 Diabetes. Nutrients 2021, 13, 3144. [Google Scholar] [CrossRef] [PubMed]
  172. Whyte, A.R.; Cheng, N.; Butler, L.T.; Lamport, D.J.; Williams, C.M. Flavonoid-Rich Mixed Berries Maintain and Improve Cognitive Function Over a 6 h Period in Young Healthy Adults. Nutrients 2019, 11, 2685. [Google Scholar] [CrossRef] [PubMed]
  173. Gouvarchinghaleh, H.E.; Kiany, F.; Parastouei, K.; Alishiri, G.; Jafari, N.J.; Fooladi, A.A.I.; Pargar, A.; Ghazvini, A.; Mirnejad, R.; Raei, M.; et al. The effects of functional foods mixture on inflammatory cytokines and biochemical findings in hospitalized patients with COVID-19: A randomized double-blind controlled trial. Trials 2023, 24, 442. [Google Scholar] [CrossRef]
  174. Stokić, E.; Mandić, A.; Sakač, M.; Mišan, A.; Pestorić, M.; Šimurina, O.; Jambrec, D.; Jovanov, P.; Nedeljković, N.; Milovanović, I. Quality of buckwheat-enriched wheat bread and its antihyperlipidemic effect in statin treated patients. LWT-Food Sci. Technol. 2015, 63, 556–561. [Google Scholar] [CrossRef]
  175. Dinu, M.; Ghiselli, L.; Whittaker, A.; Pagliai, G.; Cesari, F.; Fiorillo, C.; Becatti, M.; Marcucci, R.; Benedettelli, S.; Sofi, F. Consumption of buckwheat products and cardiovascular risk profile: A randomized, single-blinded crossover trial. Nutr. Metab. Cardiovasc. Dis. 2017, 27, e20–e21. [Google Scholar] [CrossRef]
  176. Smith, L.; Lopez Sanchez, G.F.; Veronese, N.; Soysal, P.; Oh, H.; Kostev, K.; Rahmati, M.; Butler, L.; Gibson, P.; Keyes, H.; et al. Association of Fruit and Vegetable Consumption With Mild Cognitive Impairment in Low- and Middle-Income Countries. J. Gerontol. A Biol. Sci. Med. Sci. 2023, 78, 1410–1416. [Google Scholar] [CrossRef]
  177. Ghavipour, M.; Sotoudeh, G.; Ghorbani, M. Tomato juice consumption improves blood antioxidative biomarkers in overweight and obese females. Clin. Nutr. 2015, 34, 805–809. [Google Scholar] [CrossRef]
  178. Saito, Y.; Nitta, A.; Imai, S.; Kajiyama, S.; Miyawaki, T.; Ozasa, N.; Kajiyama, S.; Hashimoto, Y.; Fukui, M. Tomato juice preload has a significant impact on postprandial glucose concentration in healthy women: A randomized cross-over trial. Asia Pac. J. Clin. Nutr. 2020, 29, 491–497. [Google Scholar] [CrossRef]
  179. Michaličková, D.; Belović, M.; Ilić, N.; Kotur-Stevuljević, J.; Slanař, O.; Šobajić, S. Comparison of Polyphenol-Enriched Tomato Juice and Standard Tomato Juice for Cardiovascular Benefits in Subjects with Stage 1 Hypertension: A Randomized Controlled Study. Plant Foods Hum. Nutr. 2019, 74, 122–127. [Google Scholar] [CrossRef]
  180. Upritchard, J.E.; Sutherland, W.H.; Mann, J.I. Effect of supplementation with tomato juice, vitamin E, and vitamin C on LDL oxidation and products of inflammatory activity in type 2 diabetes. Diabetes Care 2000, 23, 733–738. [Google Scholar] [CrossRef]
  181. Hirose, A.; Terauchi, M.; Tamura, M.; Akiyoshi, M.; Owa, Y.; Kato, K.; Kubota, T. Tomato juice intake increases resting energy expenditure and improves hypertriglyceridemia in middle-aged women: An open-label, single-arm study. Nutr. J. 2015, 14, 34. [Google Scholar] [CrossRef] [PubMed]
  182. Valderas-Martinez, P.; Chiva-Blanch, G.; Casas, R.; Arranz, S.; Martínez-Huélamo, M.; Urpi-Sarda, M.; Torrado, X.; Corella, D.; Lamuela-Raventós, R.M.; Estruch, R. Tomato Sauce Enriched with Olive Oil Exerts Greater Effects on Cardiovascular Disease Risk Factors than Raw Tomato and Tomato Sauce: A Randomized Trial. Nutrients 2016, 8, 170. [Google Scholar] [CrossRef]
  183. Burton-Freeman, B.; Talbot, J.; Park, E.; Krishnankutty, S.; Edirisinghe, I. Protective activity of processed tomato products on postprandial oxidation and inflammation: A clinical trial in healthy weight men and women. Mol. Nutr. Food Res. 2012, 56, 622–631. [Google Scholar] [CrossRef]
  184. Chen, C.Y.; Chien, Y.W. Fresh Tomato (Lycopersicon esculentum Mill.) in the Diet Improves the Features of the Metabolic Syndrome: A Randomized Study in Postmenopausal Women. Biology 2024, 13, 588. [Google Scholar] [CrossRef] [PubMed]
  185. Thies, F.; Masson, L.F.; Rudd, A.; Vaughan, N.; Tsang, C.; Brittenden, J.; Simpson, W.G.; Duthie, S.; Horgan, G.W.; Duthie, G. Effect of a tomato-rich diet on markers of cardiovascular disease risk in moderately overweight, disease-free, middle-aged adults: A randomized controlled trial. Am. J. Clin. Nutr. 2012, 95, 1013–1022. [Google Scholar] [CrossRef] [PubMed]
  186. Nishimura, M.; Tominaga, N.; Ishikawa-Takano, Y.; Maeda-Yamamoto, M.; Nishihira, J. Effect of 12-Week Daily Intake of the High-Lycopene Tomato (Solanum Lycopersicum), A Variety Named "PR-7", on Lipid Metabolism: A Randomized, Double-Blind, Placebo-Controlled, Parallel-Group Study. Nutrients 2019, 11, 1177. [Google Scholar] [CrossRef]
  187. Miller, M.G.; Hamilton, D.A.; Joseph, J.A.; Shukitt-Hale, B. Dietary blueberry improves cognition among older adults in a randomized, double-blind, placebo-controlled trial. Eur. J. Nutr. 2018, 57, 1169–1180. [Google Scholar] [CrossRef]
  188. Dodd, G.F.; Williams, C.M.; Butler, L.T.; Spencer, J.P. Acute effects of flavonoid-rich blueberry on cognitive and vascular function in healthy older adults. Nutr. Healthy Aging 2019, 5, 119–132. [Google Scholar] [CrossRef]
  189. Wood, E.; Hein, S.; Mesnage, R.; Fernandes, F.; Abhayaratne, N.; Xu, Y.; Zhang, Z.; Bell, L.; Williams, C.; Rodriguez-Mateos, A. Wild blueberry (poly)phenols can improve vascular function and cognitive performance in healthy older individuals: A double-blind randomized controlled trial. Am. J. Clin. Nutr. 2023, 117, 1306–1319. [Google Scholar] [CrossRef]
  190. Stull, A.J.; Cassidy, A.; Djousse, L.; Johnson, S.A.; Krikorian, R.; Lampe, J.W.; Mukamal, K.J.; Nieman, D.C.; Porter Starr, K.N.; Rasmussen, H.; et al. The state of the science on the health benefits of blueberries: A perspective. Front. Nutr. 2024, 11, 1415737. [Google Scholar] [CrossRef]
  191. Lau, F.C.; Bielinski, D.F.; Joseph, J.A. Inhibitory effects of blueberry extract on the production of inflammatory mediators in lipopolysaccharide-activated BV2 microglia. J. Neurosci. Res. 2007, 85, 1010–1017. [Google Scholar] [CrossRef]
  192. Dąbek, J.; Kułach, A.; Gąsior, Z. Nuclear factor kappa-light-chain-enhancer of activated B cells (NF-κB): A new potential therapeutic target in atherosclerosis? Pharmacol. Rep. 2010, 62, 778–783. [Google Scholar] [CrossRef] [PubMed]
  193. Martini, D.; Marino, M.; Venturi, S.; Tucci, M.; Klimis-Zacas, D.; Riso, P.; Porrini, M.; Del Bo, C. Blueberries and their bioactives in the modulation of oxidative stress, inflammation and cardio/vascular function markers: A systematic review of human intervention studies. J. Nutr. Biochem. 2023, 111, 109154. [Google Scholar] [CrossRef] [PubMed]
  194. Lee, S.G.; Kim, B.; Yang, Y.; Pham, T.X.; Park, Y.K.; Manatou, J.; Koo, S.I.; Chun, O.K.; Lee, J.Y. Berry anthocyanins suppress the expression and secretion of proinflammatory mediators in macrophages by inhibiting nuclear translocation of NF-κB independent of NRF2-mediated mechanism. J. Nutr. Biochem. 2014, 25, 404–411. [Google Scholar] [CrossRef] [PubMed]
  195. Henriques, J.F.; Serra, D.; Dinis, T.C.P.; Almeida, L.M. The Anti-Neuroinflammatory Role of Anthocyanins and Their Metabolites for the Prevention and Treatment of Brain Disorders. Int. J. Mol. Sci. 2020, 21, 8653. [Google Scholar] [CrossRef]
  196. Ma, L.; Sun, Z.; Zeng, Y.; Luo, M.; Yang, J. Molecular Mechanism and Health Role of Functional Ingredients in Blueberry for Chronic Disease in Human Beings. Int. J. Mol. Sci. 2018, 19, 2785. [Google Scholar] [CrossRef]
  197. Senoner, T.; Dichtl, W. Oxidative Stress in Cardiovascular Diseases: Still a Therapeutic Target? Nutrients 2019, 11, 2090. [Google Scholar] [CrossRef]
  198. 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]
  199. Janssens, G.E.; Grevendonk, L.; Schomakers, B.V.; Perez, R.Z.; van Weeghel, M.; Schrauwen, P.; Hoeks, J.; Houtkooper, R.H. A metabolomic signature of decelerated physiological aging in human plasma. Geroscience 2023, 45, 3147–3164. [Google Scholar] [CrossRef]
  200. Ungvari, A.; Gulej, R.; Patai, R.; Papp, Z.; Toth, A.; Szabo, A.A.; Podesser, B.K.; Sotonyi, P.; Benyo, Z.; Yabluchanskiy, A.; et al. Sex-specific mechanisms in vascular aging: Exploring cellular and molecular pathways in the pathogenesis of age-related cardiovascular and cerebrovascular diseases. Geroscience 2025, 2025 47, 301–337. [Google Scholar] [CrossRef]
  201. Zhou, Y.; Jiang, Z.; Lu, H.; Xu, Z.; Tong, R.; Shi, J.; Jia, G. Recent Advances of Natural Polyphenols Activators for Keap1-Nrf2 Signaling Pathway. Chem. Biodivers. 2019, 16, e1900400. [Google Scholar] [CrossRef] [PubMed]
  202. Riordan, R.; Rong, W.; Yu, Z.; Ross, G.; Valerio, J.; Dimas-Munoz, J.; Heredia, V.; Magnusson, K.; Galvan, V.; Perez, V.I. Effect of Nrf2 loss on senescence and cognition of tau-based P301S mice. Geroscience 2023, 45, 1451–1469. [Google Scholar] [CrossRef] [PubMed]
  203. Shaw, P.; Chattopadhyay, A. Nrf2-ARE signaling in cellular protection: Mechanism of action and the regulatory mechanisms. J. Cell Physiol. 2020, 235, 3119–3130. [Google Scholar] [CrossRef] [PubMed]
  204. Ketnawa, S.; Reginio, F.C., Jr.; Thuengtung, S.; Ogawa, Y. Changes in bioactive compounds and antioxidant activity of plant-based foods by gastrointestinal digestion: A review. Crit. Rev. Food Sci. Nutr. 2022, 62, 4684–4705. [Google Scholar] [CrossRef]
  205. Woolf, E.K.; Lee, S.Y.; Ghanem, N.; Vazquez, A.R.; Johnson, S.A. Protective effects of blueberries on vascular function: A narrative review of preclinical and clinical evidence. Nutr. Res. 2023, 120, 20–57. [Google Scholar] [CrossRef]
  206. Najjar, R.S.; Schwartz, A.M.; Wong, B.J.; Mehta, P.K.; Feresin, R.G. Berries and Their Polyphenols as a Potential Therapy for Coronary Microvascular Dysfunction: A Mini-Review. Int. J. Mol. Sci. 2021, 22, 3373. [Google Scholar] [CrossRef]
  207. Tousoulis, D.; Kampoli, A.M.; Tentolouris, C.; Papageorgiou, N.; Stefanadis, C. The role of nitric oxide on endothelial function. Curr. Vasc. Pharmacol. 2012, 10, 4–18. [Google Scholar] [CrossRef]
  208. Stull, A.J. Blueberries’ Impact on Insulin Resistance and Glucose Intolerance. Antioxidants 2016, 5, 44. [Google Scholar] [CrossRef]
  209. Li, Z.; Tian, J.; Cheng, Z.; Teng, W.; Zhang, W.; Bao, Y.; Wang, Y.; Song, B.; Chen, Y.; Li, B. Hypoglycemic bioactivity of anthocyanins: A review on proposed targets and potential signaling pathways. Crit. Rev. Food Sci. Nutr. 2023, 63, 7878–7895. [Google Scholar] [CrossRef]
  210. Kang, G.G.; Francis, N.; Hill, R.; Waters, D.; Blanchard, C.; Santhakumar, A.B. Dietary Polyphenols and Gene Expression in Molecular Pathways Associated with Type 2 Diabetes Mellitus: A Review. Int. J. Mol. Sci. 2019, 21, 140. [Google Scholar] [CrossRef]
  211. Momtaz, S.; Salek-Maghsoudi, A.; Abdolghaffari, A.H.; Jasemi, E.; Rezazadeh, S.; Hassani, S.; Ziaee, M.; Abdollahi, M.; Behzad, S.; Nabavi, S.M. Polyphenols targeting diabetes via the AMP-activated protein kinase pathway; future approach to drug discovery. Crit. Rev. Clin. Lab. Sci. 2019, 56, 472–492. [Google Scholar] [CrossRef]
  212. Ashique, S.; Mukherjee, T.; Mohanty, S.; Garg, A.; Mishra, N.; Kaushik, M.; Bhowmick, M.; Chattaraj, B.; Mohanto, S.; Srivastava, S. Blueberries in focus: Exploring the phytochemical potentials and therapeutic applications. J. Agric. Food Res. 2024, 18, 101300. [Google Scholar] [CrossRef]
  213. Giacalone, M.; Di Sacco, F.; Traupe, I.; Pagnucci, N.; Forfori, F.; Giunta, F. Blueberry polyphenols and neuroprotection. In Bioactive Nutraceuticals and Dietary Supplements in Neurological and Brain Disease; Elsevier: Amsterdam, The Netherlands, 2015; pp. 17–28. [Google Scholar]
  214. Krikorian, R.; Shidler, M.D.; Nash, T.A.; Kalt, W.; Vinqvist-Tymchuk, M.R.; Shukitt-Hale, B.; Joseph, J.A. Blueberry supplementation improves memory in older adults. J. Agric. Food Chem. 2010, 58, 3996–4000. [Google Scholar] [CrossRef]
  215. Whyte, A.R.; Cheng, N.; Fromentin, E.; Williams, C.M. A randomized, double-blinded, placebo-controlled study to compare the safety and efficacy of low dose enhanced wild blueberry powder and wild blueberry extract (ThinkBlue™) in maintenance of episodic and working memory in older adults. Nutrients 2018, 10, 660. [Google Scholar] [CrossRef] [PubMed]
  216. Cheng, N. The acute effects of wild blueberry extract on cognition in healthy older adults. Ph.D. Thesis, University of Reading, Reading, Berkshire, UK, 2024. [Google Scholar]
  217. Choi, Y.-H.; Kwon, H.-S.; Shin, S.-G.; Chung, C.-K. Vaccinium uliginosum L. improves amyloid β protein-induced learning and memory impairment in Alzheimer’s disease in mice. Prev. Nutr. Food Sci. 2014, 19, 343. [Google Scholar] [CrossRef] [PubMed]
  218. Tan, L.; Zhang, H.; Li, H.; Sun, S.; Lyu, Q.; Jiang, Y. Blueberry extracts antagonize Aβ25–35 neurotoxicity and exert a neuroprotective effect through MEK-ERK-BDNF/UCH-L1 signaling pathway in rat and mouse hippocampus. Nutr. Neurosci. 2024, 27, 745–760. [Google Scholar] [CrossRef]
  219. Abdi-Moghadam, Z.; Darroudi, M.; Mahmoudzadeh, M.; Mohtashami, M.; Jamal, A.M.; Shamloo, E.; Rezaei, Z. Functional yogurt, enriched and probiotic: A focus on human health. Clin. Nutr. ESPEN 2023, 57, 575–586. [Google Scholar] [CrossRef]
  220. Russo, F.; Linsalata, M.; Clemente, C.; Chiloiro, M.; Orlando, A.; Marconi, E.; Chimienti, G.; Riezzo, G. Inulin-enriched pasta improves intestinal permeability and modifies the circulating levels of zonulin and glucagon-like peptide 2 in healthy young volunteers. Nutr. Res. 2012, 32, 940–946. [Google Scholar] [CrossRef] [PubMed]
  221. Fontané, L.; Pedro-Botet, J.; Garcia-Ribera, S.; Climent, E.; Muns, M.D.; Ballesta, S.; Satorra, P.; Flores-Le Roux, J.A.; Benaiges, D. Use of phytosterol-fortified foods to improve LDL cholesterol levels: A systematic review and meta-analysis. Nutr. Metab. Cardiovasc. Dis. 2023, 33, 1472–1480. [Google Scholar] [CrossRef]
  222. Demonty, I.; Ras, R.T.; van der Knaap, H.C.; Duchateau, G.S.; Meijer, L.; Zock, P.L.; Geleijnse, J.M.; Trautwein, E.A. Continuous dose-response relationship of the LDL-cholesterol–lowering effect of phytosterol intake. J. Nutr. 2009, 139, 271–284. [Google Scholar] [CrossRef]
  223. Musa-Veloso, K.; Poon, T.H.; Elliot, J.A.; Chung, C. A comparison of the LDL-cholesterol lowering efficacy of plant stanols and plant sterols over a continuous dose range: Results of a meta-analysis of randomized, placebo-controlled trials. Prostaglandins Leukot. Essent. Fat. Acids 2011, 85, 9–28. [Google Scholar] [CrossRef]
  224. Mach, F.; Baigent, C.; Catapano, A.L.; Koskinas, K.C.; Casula, M.; Badimon, L.; Chapman, M.J.; De Backer, G.G.; Delgado, V.; Ference, B.A. 2019 ESC/EAS Guidelines for the management of dyslipidaemias: Lipid modification to reduce cardiovascular risk: The Task Force for the management of dyslipidaemias of the European Society of Cardiology (ESC) and European Atherosclerosis Society (EAS). Eur. Heart J. 2020, 41, 111–188. [Google Scholar] [CrossRef]
  225. He, Y.; Huang, S.Y.; Wang, H.F.; Zhang, W.; Deng, Y.T.; Zhang, Y.R.; Dong, Q.; Feng, J.F.; Cheng, W.; Yu, J.T. Circulating polyunsaturated fatty acids, fish oil supplementation, and risk of incident dementia: A prospective cohort study of 440,750 participants. Geroscience 2023, 45, 1997–2009. [Google Scholar] [CrossRef] [PubMed]
  226. Murugkar, D.A.; Zanwar, A.A. With Omega-3 Fatty Acids: A Promising Approach for Improved Nutritional. Omega-3 Fatty Acids: Keys to Nutritional Health and Disease; Springer: Cham, Switzerland, 2025; p. 313. [Google Scholar]
  227. Ungvari, Z.; Fekete, M.; Varga, P.; Fekete, J.T.; Buda, A.; Szappanos, Á.; Lehoczki, A.; Mózes, N.; Grosso, G.; Menyhart, O. Impact of adherence to the Mediterranean diet on stroke risk. GeroScience 2025, 47, 3565–3581. [Google Scholar] [CrossRef] [PubMed]
  228. Ruiz Ruiz, J.C.; Ortiz Vazquez, E.D.L.L.; Segura Campos, M.R. Encapsulation of vegetable oils as source of omega-3 fatty acids for enriched functional foods. Crit. Rev. Food Sci. Nutr. 2017, 57, 1423–1434. [Google Scholar] [CrossRef]
  229. Dawczynski, C.; Massey, K.A.; Ness, C.; Kiehntopf, M.; Stepanow, S.; Platzer, M.; Grün, M.; Nicolaou, A.; Jahreis, G. Randomized placebo-controlled intervention with n-3 LC-PUFA-supplemented yoghurt: Effects on circulating eicosanoids and cardiovascular risk factors. Clin. Nutr. 2013, 32, 686–696. [Google Scholar] [CrossRef] [PubMed]
  230. Kong, D.; Khan, S.A.; Wu, H.; Liu, Y.; Ling, H.Q. Biofortification of iron and zinc in rice and wheat. J. Integr. Plant Biol. 2022, 64, 1157–1167. [Google Scholar] [CrossRef]
  231. Kodkany, B.S.; Bellad, R.M.; Mahantshetti, N.S.; Westcott, J.E.; Krebs, N.F.; Kemp, J.F.; Hambidge, K.M. Biofortification of pearl millet with iron and zinc in a randomized controlled trial increases absorption of these minerals above physiologic requirements in young children. J. Nutr. 2013, 143, 1489–1493. [Google Scholar] [CrossRef]
  232. Ou, J.; Wang, M.; Zheng, J.; Ou, S. Positive and negative effects of polyphenol incorporation in baked foods. Food Chem. 2019, 284, 90–99. [Google Scholar] [CrossRef]
  233. Conti, V.; Piccini, C.; Romi, M.; Salusti, P.; Cai, G.; Cantini, C. Pasta enriched with carrot and olive leaf flour retains high levels of accessible bioactives after in vitro digestion. Foods 2023, 12, 3540. [Google Scholar] [CrossRef]
  234. Zahid, H.F.; Ali, A.; Legione, A.R.; Ranadheera, C.S.; Fang, Z.; Dunshea, F.R.; Ajlouni, S. Probiotic yoghurt enriched with mango peel powder: Biotransformation of phenolics and modulation of metabolomic outputs after in vitro digestion and colonic fermentation. Int. J. Mol. Sci. 2023, 24, 8560. [Google Scholar] [CrossRef] [PubMed]
  235. Almada, A.L. Nutraceuticals and functional foods: Innovation, insulation, evangelism, and evidence. In Nutraceutical and Functional Food Regulations in the United States and Around the World; Elsevier: Amsterdam, The Netherlands, 2019; pp. 3–11. [Google Scholar]
  236. Iwatani, S.; Yamamoto, N. Functional food products in Japan: A review. Food Sci. Hum. Wellness 2019, 8, 96–101. [Google Scholar] [CrossRef]
  237. Firoozzare, A.; Boccia, F.; Yousefian, N.; Ghazanfari, S.; Pakook, S. Understanding the role of awareness and trust in consumer purchase decisions for healthy food and products. Food Qual. Prefer. 2024, 121, 105275. [Google Scholar] [CrossRef]
  238. Siervo, M.; Lara, J.; Chowdhury, S.; Ashor, A.; Oggioni, C.; Mathers, J.C. Effects of the Dietary Approach to Stop Hypertension (DASH) diet on cardiovascular risk factors: A systematic review and meta-analysis. Br. J. Nutr. 2015, 113, 1–15. [Google Scholar] [CrossRef] [PubMed]
  239. Trabado-Fernandez, A.; Garcia-Colomo, A.; Cuadrado-Soto, E.; Peral-Suarez, A.; Salas-Gonzalez, M.D.; Lorenzo-Mora, A.M.; Aparicio, A.; Delgado-Losada, M.L.; Maestu-Unturbe, F.; Lopez-Sobaler, A.M. Association of a DASH diet and magnetoencephalography in dementia-free adults with different risk levels of Alzheimer’s disease. Geroscience 2024, 47, 1747–1759. [Google Scholar] [CrossRef] [PubMed]
  240. Challa, H.J.; Ameer, M.A.; Uppaluri, K.R. DASH diet to stop hypertension. In StatPearls [Internet]; StatPearls Publishing: St. Petersburg, FL, USA, 2022. [Google Scholar]
  241. Wickman, B.E.; Enkhmaa, B.; Ridberg, R.; Romero, E.; Cadeiras, M.; Meyers, F.; Steinberg, F. Dietary management of heart failure: DASH diet and precision nutrition perspectives. Nutrients 2021, 13, 4424. [Google Scholar] [CrossRef]
  242. Tablante, E.C.; Pachón, H.; Guetterman, H.M.; Finkelstein, J.L. Fortification of wheat and maize flour with folic acid for population health outcomes. Cochrane Database Syst. Rev. 2019, 7, CD012150. [Google Scholar] [CrossRef]
  243. Castillo-Lancellotti, C.; Tur, J.A.; Uauy, R. Impact of folic acid fortification of flour on neural tube defects: A systematic review. Public. Health Nutr. 2013, 16, 901–911. [Google Scholar] [CrossRef]
  244. De Wals, P.; Tairou, F.; Van Allen, M.I.; Uh, S.H.; Lowry, R.B.; Sibbald, B.; Evans, J.A.; Van den Hof, M.C.; Zimmer, P.; Crowley, M.; et al. Reduction in neural-tube defects after folic acid fortification in Canada. N. Engl. J. Med. 2007, 357, 135–142. [Google Scholar] [CrossRef]
  245. Mills, J.L.; Signore, C. Neural tube defect rates before and after food fortification with folic acid. Birth Defects Res. Part. A: Clin. Mol. Teratol. 2004, 70, 844–845. [Google Scholar] [CrossRef]
  246. Darmon, N.; Drewnowski, A. Contribution of food prices and diet cost to socioeconomic disparities in diet quality and health: A systematic review and analysis. Nutr. Rev. 2015, 73, 643–660. [Google Scholar] [CrossRef] [PubMed]
  247. Miller, D.; Hayes, K. Vitamin excess and toxicity. Nutr. Toxicol. 1982, 1, 81–133. [Google Scholar]
  248. Frewer, L.; Scholderer, J.; Lambert, N. Consumer acceptance of functional foods: Issues for the future. Br. Food J. 2003, 105, 714–731. [Google Scholar] [CrossRef]
  249. Smith, V.; Clement, J.; Møgelvang-Hansen, P.; Sørensen, H.S. Assessing in-store food-to-consumer communication from a fairness perspective: An integrated approach. Fachsprache 2011, 33, 84–106. [Google Scholar] [CrossRef]
  250. Hasler, C.M. Functional foods: Benefits, concerns and challenges—A position paper from the American Council on Science and Health. J. Nutr. 2002, 132, 3772–3781. [Google Scholar] [CrossRef]
  251. Siró, I.; Kápolna, E.; Kápolna, B.; Lugasi, A. Functional food. Product development, marketing and consumer acceptance—A review. Appetite 2008, 51, 456–467. [Google Scholar] [CrossRef]
  252. Coppens, P.; Da Silva, M.F.; Pettman, S. European regulations on nutraceuticals, dietary supplements and functional foods: A framework based on safety. Toxicology 2006, 221, 59–74. [Google Scholar] [CrossRef]
  253. Grimaldi, K.A. Nutrigenetics and personalized nutrition: Are we ready for DNA-based dietary advice? Pers. Med. 2014, 11, 297–307. [Google Scholar] [CrossRef]
  254. da Silva, B.V.; Barreira, J.C.; Oliveira, M.B.P. Natural phytochemicals and probiotics as bioactive ingredients for functional foods: Extraction, biochemistry and protected-delivery technologies. Trends Food Sci. Technol. 2016, 50, 144–158. [Google Scholar] [CrossRef]
  255. Yamada, K.; Sato-Mito, N.; Nagata, J.; Umegaki, K. Health claim evidence requirements in Japan. J. Nutr. 2008, 138, 1192S–1198S. [Google Scholar] [CrossRef]
  256. Heasman, M.; Mellentin, J. The Functional Foods Revolution: Healthy People, Healthy Profits? Earthscan: Barcelona, Spain, 2001. [Google Scholar]
  257. Otero, M.C.B.; Bernolo, L.F. General principles in nutrigenomics and nutrigenetics. In Role of Nutrigenomics in Modern-Day Healthcare and Drug Discovery; Elsevier: Amsterdam, The Netherlands, 2023; pp. 3–18. [Google Scholar]
  258. Pray, L. Nutrigenomics and the Future of Nutrition; The National Academic Press: Washington, DC, USA, 2018. [Google Scholar]
  259. Chaudhary, D.; Guleria, D.; Aggarwal, H.; Mishra, V.; Chauhan, A.; Dufossé, L.; Joshi, N.C. Nutrigenomics and Personalized Diets-Tailoring Nutrition for Optimal Health. Appl. Food Res. 2025, 5, 100980. [Google Scholar] [CrossRef]
  260. Hiraoka, M.; Kagawa, Y. Genetic polymorphisms and folate status. Congenit. Anom. 2017, 57, 142–149. [Google Scholar] [CrossRef]
  261. Bueno, O.; Molloy, A.M.; Fernandez-Ballart, J.D.; García-Minguillán, C.J.; Ceruelo, S.; Ríos, L.; Ueland, P.M.; Meyer, K.; Murphy, M.M. Common polymorphisms that affect folate transport or metabolism modify the effect of the MTHFR 677C> T polymorphism on folate status. J. Nutr. 2016, 146, 1–8. [Google Scholar] [CrossRef]
  262. Hong, J.Y.; Kim, Y.J. Application of Big Data and Artificial Intelligence in The RESEARCH of Health Functional Foods. Food Suppl. Biomater. Health 2024, 4, e19. [Google Scholar] [CrossRef]
  263. Joshi, T.; Sehgal, H.; Puri, S.; Mahapatra, T.; Joshi, M.; Deepa, P.; Sharma, P.K. ML-based technologies in sustainable agro-food production and beyond: Tapping the (semi) arid landscape for bioactives-based product development. J. Agric. Food Res. 2024, 18, 101350. [Google Scholar] [CrossRef]
  264. Moreno, C. Landscaping a Biofuture in Latin America; The Centre for Research and Documentation Chile-Latin America: Berlin, Germany, 2020; pp. 4–27. [Google Scholar]
  265. Verhoog, S.; Taneri, P.E.; Roa Díaz, Z.M.; Marques-Vidal, P.; Troup, J.P.; Bally, L.; Franco, O.H.; Glisic, M.; Muka, T. Dietary factors and modulation of bacteria strains of Akkermansia muciniphila and Faecalibacterium prausnitzii: A systematic review. Nutrients 2019, 11, 1565. [Google Scholar] [CrossRef] [PubMed]
  266. Effendi, R.M.R.A.; Anshory, M.; Kalim, H.; Dwiyana, R.F.; Suwarsa, O.; Pardo, L.M.; Nijsten, T.E.; Thio, H.B. Akkermansia muciniphila and Faecalibacterium prausnitzii in immune-related diseases. Microorganisms 2022, 10, 2382. [Google Scholar] [CrossRef]
  267. Teng, T.S.; Chin, Y.L.; Chai, K.F.; Chen, W.N. Fermentation for future food systems: Precision fermentation can complement the scope and applications of traditional fermentation. EMBO Rep. 2021, 22, e52680. [Google Scholar] [CrossRef]
  268. Mishra, M.; Afzal, S.; Singh, N.K. CRISPR/Cas9-Mediated Genome Engineering for Production of Biofortified Crops. In Phytoremediation and Biofortification; Apple Academic Press: Palm Bay, FL, USA, 2024; pp. 303–333. [Google Scholar]
  269. Khan, A.; Pudhuvai, B.; Shrestha, A.; Mishra, A.K.; Shah, M.P.; Koul, B.; Dey, N. CRISPR-mediated iron and folate biofortification in crops: Advances and perspectives. Biotechnol. Genet. Eng. Rev. 2024, 40, 4138–4168. [Google Scholar] [CrossRef]
  270. Kumar, D.; Yadav, A.; Ahmad, R.; Dwivedi, U.N.; Yadav, K. CRISPR-based genome editing for nutrient enrichment in crops: A promising approach toward global food security. Front. Genet. 2022, 13, 932859. [Google Scholar] [CrossRef]
  271. Ververis, E.; Ackerl, R.; Azzollini, D.; Colombo, P.A.; de Sesmaisons, A.; Dumas, C.; Fernandez-Dumont, A.; da Costa, L.F.; Germini, A.; Goumperis, T. Novel foods in the European Union: Scientific requirements and challenges of the risk assessment process by the European Food Safety Authority. Food Res. Int. 2020, 137, 109515. [Google Scholar] [CrossRef] [PubMed]
  272. Martirosyan, D.; Alvarado, A. Functional foods regulation system: Proposed regulatory paradigm by functional food center. Funct. Food Sci. 2023, 3, 275–287. [Google Scholar] [CrossRef]
  273. Martirosyan, D.; Lampert, T.; Ekblad, M. Classification and regulation of functional food proposed by the functional food center. Funct. Food Sci. 2022, 2, 25–46. [Google Scholar] [CrossRef]
  274. González-Díaz, C.; Vilaplana-Aparicio, M.J.; Iglesias-García, M. How is functional food advertising understood? An approximation in university students. Nutrients 2020, 12, 3312. [Google Scholar] [CrossRef] [PubMed]
  275. Sääksjärvi, M.; Holmlund, M.; Tanskanen, N. Consumer knowledge of functional foods. Int. Rev. Retail. Distrib. Consum. Res. 2009, 19, 135–156. [Google Scholar] [CrossRef]
  276. Fanzo, J.; McLaren, R.; Bellows, A.; Carducci, B. Challenges and opportunities for increasing the effectiveness of food reformulation and fortification to improve dietary and nutrition outcomes. Food Policy 2023, 119, 102515. [Google Scholar] [CrossRef]
Table 1. Summary of clinical trials on the health effects of kefir and fermented dairy products in various populations.
Table 1. Summary of clinical trials on the health effects of kefir and fermented dairy products in various populations.
Study [Ref.]InterventionDuration/DesignParticipantsOutcomes MeasuredKey Findings
Bellikci-Koyu et al. [136]Kefir12 weeks, 180 mL/dayMetabolic syndrome (n = 62)Anthropometrics, glycemic, lipids, BP, inflammationApoA1 ↑ 3.4%, LDL-C ↓ 7.6%, BP ↓, inflammatory markers ↓ (p < 0.05)
Bellikci-Koyu et al. [137]Kefir12 weeks, 180 mL/dayMetabolic syndrome (n = 22)Insulin, HOMA-IR, cytokines, BP, microbiotaWithin-group improvements; no between-group differences; Actinobacteria ↑ (p = 0.023)
Yılmaz et al. [138]Kefir4 weeks, 400 mL/dayIBD (Inflammatory Bowel Disease) patients (n = 45)Stool bacteria, symptoms, inflammationLactobacillus ↑; ESR, CRP ↓ in Crohn’s; bloating ↓ (p < 0.05)
Alihosseini et al. [139]Probiotic kefir8 weeks, 600 mL/dayType 2 diabetes (n = 60)Insulin, HOMA-IR, QUICKI, homocysteineReduced HOMA-IR in probiotic group; homocysteine decreased in both groups
Fathi et al. [140]Kefir (probiotic dairy)8 weeks, 2 servings/dayOverweight women (n = 75)Lipids (TC, LDL-C, HDL-C, TG, ratios)TC, LDL-C, Non-HDL-C decreased in kefir and milk groups (p < 0.05)
Öneş et al. [141]Probiotic kefir4 weeks, 200 mL/dayFemale soccer players (n = 21)Microbiome, body composition, athletic performanceMicrobial diversity ↑; Akkermansia and Faecalibacterium ↑; VO2max ↑ (p < 0.05)
Wang et al. [142]AB-kefir (Lactobacillus acidophilus (A), Bifidobacterium bifidum (B)3 weeks, 1 sachet/dayHealthy adults (n = 56)GI symptoms, microbiotaAbdominal pain and bloating decreased in men; bifidobacteria increase maintained; total anaerobes increased in women
Pražnikar et al. [143]Kefir3 weeksOverweight adults (n = 28)Zonulin, lipids, glucose, CRP, moodZonulin improved (η2 = 0.275); slight mood improvement; lipids and glucose similar to milk
Ton et al. [144]Kefir90 days, 2 mL/kg/dayAlzheimer’s patients (n = 13)Cognitive tests, cytokines, oxidative stressCognitive functions improved; inflammatory and oxidative markers decreased (~30%)
Noori et al. [145]Probiotic-fortified kefir8 weeks, 240 mL/dayElderly men (n = 67)Depression, appetite, oxidative stress, inflammationDepression improved (p = 0.001); antioxidant capacity ↑ (p = 0.009); no change in inflammation
Tatullo et al. [146]Mediterranean buffalo milk (bioactive)12 weeksAdults, high CV risk, BMI > 25 (n = 20)BP, glucose, BMI, weightBlood pressure improved (p < 0.05)
Ostadrahimi et al. [147]Kefir (L. casei, L. acidophilus, Bifido)8 weeks, 600 mL/dayType 2 diabetes patients (n = 60)Fasting glucose, HbA1c, lipidsGlucose and HbA1c decreased (p ≤ 0.02); lipids unchanged
Bourrie et al. [148]Commercial vs. pitched kefir4 weeks, 2 servings/dayMales with high LDL-C (n = 21)Lipids, endothelial markers, inflammationPitched kefir decreased LDL-C, ICAM-1, VCAM-1, IL-8, CRP, TNF-α vs. commercial (p < 0.05)
O’Brien et al. [149]Kefir beverage15 weeks, 2 servings/weekYoung adults (n = 67)Running time, body composition, CRPCRP increase attenuated by kefir
BP—Blood Pressure; BMI—Body Mass Index; CV—Cardiovascular; HOMA-IR—Homeostatic Model Assessment of Insulin Resistance; QUICKI—Quantitative Insulin Sensitivity Check Index; TC—Total Cholesterol; LDL-C—Low-Density Lipoprotein Cholesterol; HDL-C—High-Density Lipoprotein Cholesterol; TG—Triglycerides; CRP—C-Reactive Protein; ApoA1—Apolipoprotein A1; VO2max—Maximal Oxygen Uptake; ICAM-1—Intercellular Adhesion Molecule 1; VCAM-1—Vascular Cell Adhesion Molecule 1; ESR—Erythrocyte Sedimentation Rate; GI—Gastrointestinal; HbA1c—Glycated Hemoglobin; η2—Eta-squared [effect size]; TNF-α—Tumor Necrosis Factor alpha; ↑—increase, ↓—decrease.
Table 2. Studies on fruits, vegetables, and plant-based interventions.
Table 2. Studies on fruits, vegetables, and plant-based interventions.
Study [Ref.]InterventionDuration/DesignParticipantsOutcomes MeasuredKey Findings
Nilsson et al. [150]Natural fruits and vegetables16 weeks; intake increased from ~2.2 to ~4.2 servings/day66 older adults (65–70 y); randomized (FV vs. control)Inflammatory biomarkers: CRP, IL-6, IL-18, TNF-α, MIP-1α/β, TRAIL, TRANCE, CX3CL1↓ TRAIL, TRANCE, CX3CL1 in FV group (p < 0.05); no significant change in other markers; waist circumference and physical activity unchanged
Carrillo et al. [151]Polyphenol-rich micronized fruit and vegetable preparationTwo 16-week periods; crossover with 4-week washout108 healthy adults (53 intervention, 55 placebo); sex-stratifiedCognitive tests: Stroop, TESEN, RISTImproved executive function, memory, attention, and processing speed; p-values not statistically significant but trend favored intervention
Arcusa et al. [152]High-polyphenol nutraceutical (fruit and vegetable-based)Two 16-week periods; 4-week washout108 healthy adults (53 intervention, 55 placebo); 92 completersOxidative stress and inflammatory markers; catecholamines↓ OxLDL (78.98→69.52) and CRP (1.50→1.39) (p < 0.001); ↑ dopamine (15.43→19.61; p < 0.05)
Kopf et al. [153]Whole grains and fruits/vegetables6 weeks; 3 servings/day (WG, FV, or refined grains)49 overweight/obese adults with low baseline FV/WG intakeIL-6, TNF-α, LBP, hs-CRP, gut microbiota diversity↓ LBP (WG p = 0.02; FV p = 0.005), ↓ IL-6 (FV p = 0.006), ↓ TNF-α (WG p < 0.001); ↑ alpha diversity in FV group; no significant differences in microbiota composition between groups
Yuan et al. [154]Functional fibers (fruit fiber, FibreMax)Acute; single dose (10 g fiber per bread serving)80 healthy adults; crossover, double-blind designPostprandial glycemia, satiety, energy intake, gastrointestinal well-beingFruit fiber: ↓ glycemia by 35% (p = 0.004), ↓ energy intake by 368 kJ (p = 0.001); FibreMax: ↓ glycemia by 43% (p = 0.004); no reported gastrointestinal side effects
Campbell et al. [155]Whole food, plant-based diet (WFPB)8 weeks; 3 ad libitum meals/day provided32 women with metastatic breast cancer (21 intervention, 11 control)Cognitive function, emotional well-being, fatigue↑ FACT-Cog score (+16.1; p = 0.040), ↑ emotional well-being (+2.3; p = 0.016), ↓ fatigue severity (p = 0.047); high adherence rate (94.3%)
Lamport et al. [156]Concord grape juice (777 mg polyphenols)12 weeks; 355 mL/day; crossover with 4-week washout25 healthy middle-aged working women (40–50 y)Cognitive function, simulated driving, blood pressure, moodImproved immediate spatial memory and driving performance vs. placebo; sustained cognitive benefits reported
Haskell-Ramsay et al. [157]Berry phenolics (purple grape juice)Acute; single 230 mL dose vs. placebo20 healthy young adultsMemory, attention, moodImproved attention reaction time (p = 0.047), ↑ calmness ratings (p = 0.046); memory RT showed order effects (p = 0.018)
Nilsson et al. [158]Mixed-berry beverage (polyphenols + fiber)5 weeks; daily intake (berries and tomatoes)40 healthy adults (50–70 y)Cognitive function, cardiometabolic markers↓ total and LDL cholesterol (p < 0.05); control group ↑ glucose and insulin; ↑ working memory vs. control (p < 0.05); no other cognitive effects observed
Connolly et al. [159]Cruciferous vegetables (~300 g/day)2-week crossover with 2-week washout18 adults with mildly elevated BP (median age 68 y)24 h and daytime systolic BP, triglycerides↓ 24 h SBP by −2.5 mmHg (p = 0.002), ↓ daytime SBP by −3.6 mmHg (p < 0.001), ↓ triglycerides by −0.2 mmol/L (p = 0.047)
Riso et al.
[160]
Broccoli (whole food, 250 g/day)10 daysYoung male smokers (n = 17)Plasma folate, lutein, CRP, TNF-α, IL-6, adiponectin↓ CRP by 48% (p < 0.05), ↑ folate (+17%) and lutein (+39%); no significant changes in TNF-α, IL-6, adiponectin
Navarro et al. [161]Cruciferous and apiaceous vegetablesFour 14-day controlled diets with 21-day washouts63 healthy adults (20–40 y)Serum IL-6, IL-8, CRP, TNF-α; genotype–diet interaction↓ IL-6 (19–20%) in cruciferous-only and combined diets; ↑ IL-8 (+16%) in cruciferous + apiaceous group; genotype influenced IL-6, CRP, and IL-8 response
Khan et al. [162]Blackcurrant juice (vitamin C + polyphenols)6 weeks; 250 mL × 4/day66 healthy adults with low fruit/veg intake (<2 servings/day)Flow-mediated dilation (FMD), plasma F2-isoprostanes, vitamin C↑ FMD at high dose (p = 0.022), ↑ plasma vitamin C (p < 0.001), ↓ F2-isoprostanes (p = 0.002–0.003)
Duthie et al. [163] High fruit, vegetable, and juice intake12 weeks; +480 g fruits/veg + 300 mL juice/day45 adults (39–58 y) with low baseline intake (<3/day)Nutritional biomarkers, antioxidant capacity, DNA damage↑ plasma vitamin C (+35%), folate (+15%), carotenoids (+50–70%); no change in antioxidant capacity or vascular markers
Nadeem et al. [164]Fruit and vegetable intake (1, 3, or 6 servings/day)8–16 weeksHypertensive subjects; older adults (65–85 y; n = 192)hsCRP, IL-6, E-selectin, SAA, HDL subfractions, CETPNo effect on hsCRP, IL-6, E-selectin; ↓ HDL3-SAA (p = 0.049), ↓ HDL2/3-SAA (p = 0.035/0.032), ↓ HDL3/2-CETP (p = 0.010/0.030)
Siddarth et al. [165]Pomegranate juice (polyphenol antioxidant)12 months; 236.5 mL daily261 nondemented adults aged 50–75; 200 completersMemory tests (BVMT-R, SRT)Significant group × time interaction in BVMT-R learning (p = 0.003); small improvement in pomegranate group vs. significant decline in placebo
Ahles et al. [166]Citrus and pomegranate complex (polyphenol-rich)4 weeks daily supplementation36 healthy elderly adults (60–75 years)Handgrip strength, fitness, QOL, cognition, oxidative stress↑ Handgrip strength (p = 0.019), ↑ cognition (p = 0.042), ↓ malondialdehyde (p = 0.033); no changes in fitness or other QOL markers
Kennedy et al. [167]Polyphenol-rich functional breakfast (FB)Single meal crossover (FB, control, RTEC)16 healthy adultsTotal polyphenols, glucose and insulin response, mood, memoryFB had higher polyphenols (230 mg vs. 147 mg; p < 0.05), lowest insulin AUC (p < 0.05); no effect on glucose AUC, mood, or memory
Kent et al.
[168]
Anthocyanin-rich cherry juice12 weeks; 200 mL/day49 older adults (≥70 years) with mild/moderate dementiaCognitive function, BP, inflammatory markers (CRP, IL-6)Improved verbal fluency (p = 0.014), short-/long-term memory (p ≤ 0.014), ↓ systolic BP (p = 0.038); no CRP or IL-6 changes
Lamport et al. [169]Flavanone-rich citrus juice (70.5 mg flavanones)Single 500 mL dose; single-blind crossover24 healthy young adults (18–30 years)Cognitive tests (DSST), regional cerebral blood flow (CBF)↑ CBF in right frontal gyrus at 2 h; improved DSST at 2 h; no other cognitive changes
Rosli et al.
[170]
Polyphenol-rich tropical fruit juice10 weeks; 500 mL × 3/day × 3 days/week31 middle-aged women with impaired cognitive functionCognitive tests (RAVLT, CTMT), metabolomics↑ Immediate verbal recall (RAVLT) and cognitive flexibility (CTMT Trail 4) (p < 0.05); ↑ urinary thyroxine and 3-methyladenine, linked to cognition
Saeidi et al. [171] Broccoli + aerobic-resistance exercise12 weeks; 10 g/day broccoli; CARET 3×/week44 men with type 2 diabetes (4 groups, n = 11 each)Plasma dectin-1, insulin resistance, cardiometabolic markersGroups showed ↓ dectin-1 (p < 0.05); improved insulin resistance and cardiometabolic markers (p < 0.05)
Whyte et al. [172] Flavonoid-rich mixed-berry smoothieSingle 400 mL dose; 6-hour monitoring40 healthy young adults (20 berry, 20 placebo)Executive function (MANT, TST), mood Placebo group showed performance decline; berry group maintained accuracy and improved reaction times at 2–6 h; cognitive benefits observed during fatigue
Gouvarchinghaleh et al. [173]Functional food mixture (plant-based soup)Daily dosing; hospitalized COVID-19 patients60 COVID-19 patients (30 intervention, 30 control)Cytokines (IL-1β, IL-6, IL-17, IL-10, TNF-α), D-dimer, BUN, creatinine, CRP, potassiumSignificant reductions in cytokines, D-dimer, BUN, creatinine (p < 0.05); CRP and potassium improved more in control group
Stokić et al. [174]Buckwheat-enriched wheat bread (50% flour)1 month regular consumptionNormal weight patients on statin therapy (n = 80)Total cholesterol, LDL, LDL/HDL ratio, sensory preference, fiber, phenolics↓ total cholesterol, LDL, LDL/HDL ratio; 2.22× fiber, 4.29× phenolics; 71.88% consumer preference
Dinu et al. [175]Buckwheat-enriched semi-wholegrain products8-week intervention + 8-week washout; crossover21 high cardiovascular risk adults (mean age 51.3 y)Cholesterol, triglycerides, glucose, insulin, TBARs, plasma ORAC↓ total cholesterol (−4.7%), LDL-C (−8.5%), triglycerides (−15%), glucose (−5.8%), insulin (−17%), TBARs (−29.5%); ↑ plasma antioxidant capacity (ORAC) by 9.7%
AUC—Area Under the Curve; BP—Blood Pressure; BUN—Blood Urea Nitrogen; BVMT-R—Brief Visuospatial Memory Test-Revised; CARET—Combined Aerobic and Resistance Exercise Training; CETP—Cholesteryl Ester Transfer Protein; CRP—C-Reactive Protein; CX3CL1—C-X3-C Motif Chemokine Ligand 1; DSST—Digit Symbol Substitution Test; FMD—Flow-Mediated Dilation; FV—Fruits and Vegetables; hs-CRP—High-Sensitivity C-Reactive Protein; HDL—High-Density Lipoprotein; IL-—Interleukin; LDL—Low-Density Lipoprotein; LBP—Lipopolysaccharide Binding Protein; MANT—Multi-Tasking Test; MIP-1α/β—Macrophage Inflammatory Protein-1 alpha/beta; ORAC—Oxygen Radical Absorbance Capacity; OxLDL—Oxidized Low-Density Lipoprotein; QOL—Quality of Life; RAVLT—Rey Auditory Verbal Learning Test; RIST—Reynolds Intellectual Screening Test; RT—Reaction Time; RTEC—Ready-To-Eat Cereal; SBP—Systolic Blood Pressure; SAA—Serum Amyloid A; SRT—Selective Reminding Test; TBARs—Thiobarbituric Acid Reactive Substances; TESEN—Test of Executive and Selective Attention; TNF-α—Tumor Necrosis Factor alpha; TRAIL—TNF-Related Apoptosis-Inducing Ligand; TRANCE—TNF-Related Activation-Induced Cytokine; TST—Task Switching Test; WFPB—Whole Food Plant-Based Diet; WG—Whole Grains; ↑—increase, ↓—decrease.
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

Fekete, M.; Lehoczki, A.; Kryczyk-Poprawa, A.; Zábó, V.; Varga, J.T.; Bálint, M.; Fazekas-Pongor, V.; Csípő, T.; Rząsa-Duran, E.; Varga, P. Functional Foods in Modern Nutrition Science: Mechanisms, Evidence, and Public Health Implications. Nutrients 2025, 17, 2153. https://doi.org/10.3390/nu17132153

AMA Style

Fekete M, Lehoczki A, Kryczyk-Poprawa A, Zábó V, Varga JT, Bálint M, Fazekas-Pongor V, Csípő T, Rząsa-Duran E, Varga P. Functional Foods in Modern Nutrition Science: Mechanisms, Evidence, and Public Health Implications. Nutrients. 2025; 17(13):2153. https://doi.org/10.3390/nu17132153

Chicago/Turabian Style

Fekete, Mónika, Andrea Lehoczki, Agata Kryczyk-Poprawa, Virág Zábó, János Tamás Varga, Madarász Bálint, Vince Fazekas-Pongor, Tamás Csípő, Elżbieta Rząsa-Duran, and Péter Varga. 2025. "Functional Foods in Modern Nutrition Science: Mechanisms, Evidence, and Public Health Implications" Nutrients 17, no. 13: 2153. https://doi.org/10.3390/nu17132153

APA Style

Fekete, M., Lehoczki, A., Kryczyk-Poprawa, A., Zábó, V., Varga, J. T., Bálint, M., Fazekas-Pongor, V., Csípő, T., Rząsa-Duran, E., & Varga, P. (2025). Functional Foods in Modern Nutrition Science: Mechanisms, Evidence, and Public Health Implications. Nutrients, 17(13), 2153. https://doi.org/10.3390/nu17132153

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

Article Metrics

Back to TopTop